Activating anaplastic lymphoma kinase (ALK) receptor tyrosine kinase (RTK) mutations occur in pediatric neuroblastoma and are associated with poor prognosis. To study ALK-activating mutations in a genetically controllable system, we employed CRIPSR/Cas9, incorporating orthologs of the human oncogenic mutations ALKF1174L and ALKY1278S in the Drosophila Alk locus. AlkF1251L and AlkY1355S mutant Drosophila exhibited enhanced Alk signaling phenotypes, but unexpectedly depended on the Jelly belly (Jeb) ligand for activation. Both AlkF1251L and AlkY1355S mutant larval brains displayed hyperplasia, represented by increased numbers of Alk-positive neurons. Despite this hyperplasic phenotype, no brain tumors were observed in mutant animals. We showed that hyperplasia in Alk mutants was not caused by significantly increased rates of proliferation, but rather by decreased levels of apoptosis in the larval brain. Using single-cell RNA sequencing, we identified perturbations during temporal fate specification in AlkY1355S mutant mushroom body lineages. These findings shed light on the role of Alk in neurodevelopmental processes and highlight the potential of Alk-activating mutations to perturb specification and promote survival in neuronal lineages.

This article has an associated First Person interview with the first author of the paper.

Neuroblastoma is the most common and deadly extracranial solid tumor in children (Maris, 2010; Tolbert and Matthay, 2018). Approximately 10% of neuroblastoma harbor mutations in the anaplastic lymphoma kinase (ALK) receptor tyrosine kinase (RTK), providing an important therapeutic target (Trigg and Turner, 2018). Failure of differentiation in the neural crest lineage during development is thought to result in aggressive neuroblastoma tumors with poor prognosis (Marshall et al., 2014; Tomolonis et al., 2018). Such tumors often harbor genetic aberrations such as MYCN amplification as well as amplification and/or gain-of-function mutations in ALK (Carén et al., 2008; Chen et al., 2008; George et al., 2008; Hallberg and Palmer, 2013; Janoueix-Lerosey et al., 2008; Mossé et al., 2008). Understanding the mechanisms underlying the contribution of ALK mutations to neuroblastoma development is important, and highly controlled genetic models to investigate their function in vivo provide critical insight.

ALK is mainly expressed in the central and peripheral nervous system (Iwahara et al., 1997; Vernersson et al., 2006), and Alk knockout mice are viable with mild phenotypes (Berry et al., 2012; Bilsland et al., 2008; Borenäs et al., 2021; Cazes et al., 2014; Ono et al., 2019; Orthofer et al., 2020; Witek et al., 2015). Knock-in mice harboring neuroblastoma-associated point mutations of Alk do not exhibit spontaneous tumor formation after birth but show neuronal hyperplasia (Berry et al., 2012; Borenäs et al., 2021; Cazes et al., 2014; Ono et al., 2019). In zebrafish and chicken models, Alk family RTKs are expressed in neural tissues and play a role during development of the neural crest (Fadeev et al., 2018; Mo et al., 2017; Vieceli and Bronner, 2018; Yao et al., 2013). However, the exact role of ALK in the nervous system and how it relates to development of neuroblastoma is poorly understood.

Drosophila Alk is indispensable for the embryonic development of the visceral muscles (Englund et al., 2003; Lee et al., 2003; Lorén et al., 2003; Stute et al., 2004). Alk signaling in response to the Jelly belly (Jeb) ligand engages the Ras–Raf–MAPK pathway, leading to specification of founder cell fate, which is critical for visceral muscle fusion (Englund et al., 2003; Lee et al., 2003; Lorén et al., 2003; Stute et al., 2004). During larval stages, Alk is robustly expressed in the central nervous system (CNS), where it performs multiple functions, including regulation of neuronal targeting and survival, synapse development and body size regulation, brain sparing, longevity, memory, circadian rhythm, ethanol response and learning (Bazigou et al., 2007; Cheng et al., 2011; Gouzi et al., 2018, 2011; Kumar et al., 2021; Lasek et al., 2011; Pecot et al., 2014; Rohrbough and Broadie, 2010; Rohrbough et al., 2013; Weiss et al., 2017, 2012; Woodling et al., 2020).

In the fruit fly, the process of neurogenesis, including the role of neuroblasts (NBs) and neuronal fate specification processes, is well-characterized, with each NB producing a distinct series of neurons (Yu et al., 2006). In the central brain, two types of NBs that are classified as Type I (NB I) and Type II (NB II) create distinct lineages (Doe, 2008; Knoblich, 2008; Urbach and Technau, 2004). Diversity of neurons in the Drosophila nervous system is achieved by highly regulated consecutive expression of transcription factors, which generate unique combinatorial transcription factor codes within NBs and their progeny (Liu et al., 2015; Syed et al., 2017a,b; Yang et al., 2016). The final complement of neurons is achieved through a binary, Notch-dependent manner via apoptosis, which occurs after division of the ganglion mother cell (GMC) (Pinto-Teixeira et al., 2016).

Alk has previously been described to function within the mushroom body (MB), where it regulates sleep behavior and long-term memory formation (Bai and Sehgal, 2015; Gouzi et al., 2011). The MB is a bilateral structure in the Drosophila brain that contains neurons that arise from four NBs per brain hemisphere, ultimately producing ∼2000 so-called Kenyon cells in the adult fly (Crittenden et al., 1998). It contains mainly three neuronal cell types: early-born γ-neurons, middle-born α′β′-neurons and late-born αβ-neurons, which form two dorsal lobes (α and α′) and three medial lobes (β, β′ and γ) (Crittenden et al., 1998; Ito et al., 1997; Lee et al., 1999). The γ-lobe undergoes remodeling during pupation in response to Ecdysone receptor B1 (EcR-B1; also referred to as EcR) activity (Lee et al., 2000), whereas the α′- and β′-lobe neurons are specified by expression of the Maternal gene required for meiosis (Mamo) BTB/POZ-containing C2H2-zinc finger transcription factor, which also maintains their cell fate (Liu et al., 2019).

Here, we exploited the genetically controlled Drosophila model system to investigate the effect of Alk mutation during neurogenesis. To do this, we generated two Alk alleles, AlkF1251L and AlkY1355S, which are orthologous to human ALKF1174L and ALKY1278S mutations in neuroblastoma patients. Remarkably, although AlkY1355S exhibits ectopic Alk signaling in the embryonic visceral mesoderm (VM), this mutant receptor is still ligand dependent. Although brains of both AlkF1251L and AlkY1355S mutants exhibited reduced apoptosis resulting in a mild hyperplasia, mutation of Alk alone was not sufficient to drive spontaneous tumor development in the Drosophila brain. Single-cell RNA-sequencing (scRNA-seq) analysis of AlkY1355S larval brains led the identification of differentially expressed genes, including mamo. Because both Mamo and Alk have described functions in the MB (Gouzi et al., 2011; Liu et al., 2019; Rossi and Desplan, 2020), we focused on Mamo in the MB lineage. Aberrant AlkY1355S signaling in the MB lineage leads to precocious Mamo expression in γ-neurons during wandering third-instar (wL3) larval stages, reflecting defects in neuronal fate specification in MB NB lineages that persist into adulthood. These results provide novel insight into the effect of oncogenic Alk mutations on neuronal specification and survival during development.

AlkF1251L and AlkY1355S mutant alleles exhibit gain-of-function activity

Overexpression of either Alk or Jeb ligand in the Drosophila CNS results in a reduced pupal size phenotype that provides a sensitive readout for Alk activity (Gouzi et al., 2011; Mendoza-García et al., 2017; Wolfstetter et al., 2017). To address whether the ability of oncogenic human ALK to drive the reduced pupal size phenotype is conserved, we employed the pan-neuronal C155-Gal4 driver to overexpress either (1) wild-type human ALK, (2) a human ALK-F1174L gain-of-function mutant (representing an ALK hotspot mutation in neuroblastoma) or (3) an additional ALK-Y1278S gain-of-function neuroblastoma mutant (Umapathy et al., 2019; Vasseur et al., 2019). Expression of either ALK-F1174L or ALK-Y1278S mutant variants, but not wild-type ALK, resulted in a reduced pupal size phenotype, suggesting that ALK receptor signaling output is conserved between human and Drosophila (Fig. 1A). However, although informative, overexpression results in substantial modification of receptor numbers and signaling dynamics that can potentially lead to non-specific phenotypes. We therefore generated mutations in the endogenous Alk locus that model neuroblastoma patient mutations in a controlled genetic background, selecting two representative mutations – human ALK-F1174L and ALK-Y1278S. These gain-of-function ALK mutations are reported as constitutively active ALK mutations found in human neuroblastoma patients, with ALK-F1174L being a more frequently occurring ‘hot-spot’ mutation (Fig. 1B,C) (Umapathy et al., 2019; Vasseur et al., 2019). Sequence alignment analysis identified the equivalent residues in Drosophila Alk as Alk-F1251 and Alk-Y1355 (Fig. 1C; Fig. S1A). A CRISPR/Cas9-mediated homology-directed repair (HDR) strategy was used to generate AlkF1251L (phenylalanine to leucine at position 1251) and AlkY1355S (tyrosine to serine at position 1355) mutations in the Drosophila Alk locus, allowing investigation of the effect of these patient-derived gain-of-function ALK mutations in the fly brain (Fig. S1B).

Fig. 1.

CRISPR/Cas9-mediated generation of neuroblastoma-associated Alk mutations in Drosophila. (A) Pupal size analysis of animals expressing human ALK transgenes pan-neuronally (C155/+>hALK/+, C155/+>hALKF1174L/+ or C155/+>hALKY1278S/+). n=number of animals analyzed. One-way ANOVA, Dunnett's multiple comparison test; ***P<0.001. ns, non-significant. Scale bar: 500 µm. (B) Structural model of the Drosophila Alk kinase domain indicating residues F1251 and Y1355. Blue, alpha-C helix; green, activation loop; red, amino acids F1251 and Y1355. (C) Amino acid alignments between the wild-type Alk sequence (bottom line in Y1355S and F1251L) and AlkF1251L or AlkY1355S, respectively. Asterisks mark amino acid changes. Boxed D indicates aspartic acid at residue 1347. (D) Pupal size analysis of homozygous AlkF1251L and AlkY1355S, as well as C155/+>jeb/+ and C155/+>Alk.EC/+, animals. One-way ANOVA, Kruskal–Wallis test; ***P<0.001 and *P<0.014. Scale bar: 500 µm. n=animals analyzed in three experimental replicates. In box plots, the box represents the 25-75th percentiles, and the line indicates the median; whiskers indicate minimum and maximum values. Representative examples are shown.

Fig. 1.

CRISPR/Cas9-mediated generation of neuroblastoma-associated Alk mutations in Drosophila. (A) Pupal size analysis of animals expressing human ALK transgenes pan-neuronally (C155/+>hALK/+, C155/+>hALKF1174L/+ or C155/+>hALKY1278S/+). n=number of animals analyzed. One-way ANOVA, Dunnett's multiple comparison test; ***P<0.001. ns, non-significant. Scale bar: 500 µm. (B) Structural model of the Drosophila Alk kinase domain indicating residues F1251 and Y1355. Blue, alpha-C helix; green, activation loop; red, amino acids F1251 and Y1355. (C) Amino acid alignments between the wild-type Alk sequence (bottom line in Y1355S and F1251L) and AlkF1251L or AlkY1355S, respectively. Asterisks mark amino acid changes. Boxed D indicates aspartic acid at residue 1347. (D) Pupal size analysis of homozygous AlkF1251L and AlkY1355S, as well as C155/+>jeb/+ and C155/+>Alk.EC/+, animals. One-way ANOVA, Kruskal–Wallis test; ***P<0.001 and *P<0.014. Scale bar: 500 µm. n=animals analyzed in three experimental replicates. In box plots, the box represents the 25-75th percentiles, and the line indicates the median; whiskers indicate minimum and maximum values. Representative examples are shown.

AlkF1251L and AlkY1355S flies were homozygous viable and fertile. We performed two independent assays to characterize gain-of-function Alk activity: (1) analysis of pupal size and (2) analysis of Alk signaling in the embryonic VM. As previously reported, pan-neuronal expression of the Alk ligand Jeb (C155>jeb) resulted in a reduced pupal size phenotype (Fig. 1D), whereas inhibition of Alk signaling by overexpression of a dominant-negative Alk variant (C155>Alk.EC) increased pupal size (Fig. 1D). Homozygous AlkF1251L and AlkY1355S animals displayed a reduced size phenotype, confirming them as Alk gain-of-function mutations (Fig. 1D). Notably, AlkY1355S exhibited a stronger phenotype than AlkF1251L, indicating that these mutations are not equally strong gain-of-function mutations, and heterozygous (e.g. AlkY1355S/+) pupae displayed a weaker phenotype than homozygous mutants, suggesting dosage sensitivity (Fig. S3A).

To further analyze Alk signaling in AlkF1251L and AlkY1355S mutants, we turned to the embryonic VM (controls in Fig. 2A,A′,F,F′) using phospho-ERK (pERK) staining or HandC-GFP reporter expression as read-out (Englund et al., 2003; Lee et al., 2003; Lorén et al., 2003; Stute et al., 2004). In contrast to the restricted founder cell activation observed in controls (Fig. 2A,A′,F,F′), all Alk-expressing cells in the VM responded with robust pERK staining and HandC-GFP reporter activation (Fig. 2B,G) when Jeb was ectopically expressed (bap>jeb). In contrast, no detectable pERK staining or HandC-GFP reporter expression was observed in the AlkKO kinase domain deletion mutant (Wolfstetter et al., 2017) (Fig. 2C,H). Ectopic Alk signaling was also observed in 21% of the homozygous AlkY1355S embryos (Fig. 2D,I; Fig. S2A,B). Surprisingly, no difference was seen between Alk signaling output in the VM of AlkF1251L mutants and control embryos (Fig. 2E,J). Taken together, these findings suggest that the AlkF1251L and AlkY1355S alleles generated here represent Drosophila Alk gain-of-function alleles of differing strength, with AlkY1355S representing a stronger gain-of-function allele than AlkF1251L.

Fig. 2.

Analysis of Alk signaling in the embryonic visceral mesoderm (VM) of AlkF1251L and AlkY1355S mutants. (A,A′) Phospho-ERK (pERK) staining in the VM founder cell (FC) row of control embryos. (A′) Magnification of boxed region in A. Scale bars: 50 µm (A) and 10 µm (A′). (B) Ectopic expression of Jeb with bap-Gal4 (bap/+>jeb/+) leads to ERK phosphorylation within the whole VM. (C) No pERK staining is observed in AlkKO loss-of-function homozygous mutants. (D) Homozygous AlkY1355S mutant embryos (21%) display ectopic pERK signals at stage 10/11 in VM cells adjacent to the FC row. n=155 embryos analyzed. (E) FC-specific pERK staining in homozygous AlkF1251L embryos. (F,F′) Control embryos display FC-specific expression of the HandC-GFP reporter in the VM. (F′) Magnification of F. (G) Ectopic expression of Jeb with bap-Gal4 (bap/+>jeb/+) results in HandC-GFP expression in all VM cells. (H) HandC-GFP expression is absent in the VM of homozygous AlkKO embryos. (I) Ectopic HandC-GFP reporter expression was observed in HandC-GFP, AlkY1355S embryos. (J) HandC-GFP expression in AlkF1251L homozygous embryos is similar to controls. (K) AlkY1355S, D1347A homozygous double mutants do not show HandC-GFP reporter expression in the VM. Scale bar: 20 µm. (L) HandC-GFP expression in heterozygous AlkY1355S, D1347A/CyO, wg-lacZ embryos. (M,N) HandC-GFP reporter expression in jebweli homozygous mutants (M) and jebweli/CyO, wg-lacZ heterozygous controls (N). (O,Q) HandC-GFP expression in AlkY1355S, jebweli (O) and AlkF1251L, jebweli (Q) homozygous mutants. (P,R) AlkY1355S, jebweli (P) and AlkF1251L, jebweli (R) heterozygous controls at stage 11. >150 embryos were analyzed. All embryos shown are in stage 10/11.

Fig. 2.

Analysis of Alk signaling in the embryonic visceral mesoderm (VM) of AlkF1251L and AlkY1355S mutants. (A,A′) Phospho-ERK (pERK) staining in the VM founder cell (FC) row of control embryos. (A′) Magnification of boxed region in A. Scale bars: 50 µm (A) and 10 µm (A′). (B) Ectopic expression of Jeb with bap-Gal4 (bap/+>jeb/+) leads to ERK phosphorylation within the whole VM. (C) No pERK staining is observed in AlkKO loss-of-function homozygous mutants. (D) Homozygous AlkY1355S mutant embryos (21%) display ectopic pERK signals at stage 10/11 in VM cells adjacent to the FC row. n=155 embryos analyzed. (E) FC-specific pERK staining in homozygous AlkF1251L embryos. (F,F′) Control embryos display FC-specific expression of the HandC-GFP reporter in the VM. (F′) Magnification of F. (G) Ectopic expression of Jeb with bap-Gal4 (bap/+>jeb/+) results in HandC-GFP expression in all VM cells. (H) HandC-GFP expression is absent in the VM of homozygous AlkKO embryos. (I) Ectopic HandC-GFP reporter expression was observed in HandC-GFP, AlkY1355S embryos. (J) HandC-GFP expression in AlkF1251L homozygous embryos is similar to controls. (K) AlkY1355S, D1347A homozygous double mutants do not show HandC-GFP reporter expression in the VM. Scale bar: 20 µm. (L) HandC-GFP expression in heterozygous AlkY1355S, D1347A/CyO, wg-lacZ embryos. (M,N) HandC-GFP reporter expression in jebweli homozygous mutants (M) and jebweli/CyO, wg-lacZ heterozygous controls (N). (O,Q) HandC-GFP expression in AlkY1355S, jebweli (O) and AlkF1251L, jebweli (Q) homozygous mutants. (P,R) AlkY1355S, jebweli (P) and AlkF1251L, jebweli (R) heterozygous controls at stage 11. >150 embryos were analyzed. All embryos shown are in stage 10/11.

AlkY1355S and AlkF1251L mutants are ligand dependent

To ensure that the phenotypes observed were specific for modification of the Alk locus, we combined a kinase-dead mutation (AlkD1347A) with the AlkY1355S allele using CRISPR/Cas9-mediated HDR. AlkD1347A harbors a modification of the highly conserved DFG motif in the kinase domain, in which the ATP-γ-phosphate-binding aspartic acid 1347 is changed to alanine (Fig. 1C; Fig. S1A). As expected, homozygous AlkD1347A animals were lethal and failed to specify VM founder cells (Fig. S3B). Analysis of the AlkD1347A, Y1355S double mutant allele showed that the embryonic VM phenotype of AlkY1355S was abrogated by the D1347A kinase-dead mutation, resulting in an Alk loss-of-function phenotype (Fig. 2K,L). Further, the reduced size phenotype of AlkY1355S/+ was rescued in AlkD1347A, Y1355S/+ animals (Fig. S3A). Taken together, these data confirm that Alk kinase activity is required for signaling output in both wild-type and gain-of-function backgrounds, and that the phenotypes observed in the AlkY1355S mutant allele are specific effects of increased Alk signaling and not due to CRISPR/Cas9 off-target effects.

We next investigated whether the AlkF1251L and AlkY1355S alleles were ligand independent by testing their ability to rescue jeb loss-of-function mutants, which also fail to specify VM founder cells (Englund et al., 2003; Lee et al., 2003; Stute et al., 2004; Weiss et al., 2001) (Fig. 2M compared to control Fig. 2N). Remarkably, both AlkY1355S, jebweli and AlkF1251L, jebweli double mutants were lethal and exhibited the jebweli phenotype (Fig. 2O-R). These data clearly show that although AlkY1355S and AlkF1251L exhibit a gain-of-function Alk signaling pupal size phenotype, they remain ligand dependent in the context of embryonic VM founder cell specification.

Alk is expressed in mature neurons of the Drosophila larval brain

Previous studies have reported broad expression of Alk mRNA and protein in the Drosophila nervous system (Bazigou et al., 2007; Cheng et al., 2011; Gouzi et al., 2011; Lorén et al., 2003, 2001). We used fluorescent in situ hybridization chain reaction (HCR) to detect Alk and jeb mRNA in the CNS of control and AlkY1355S animals. Both Alk and jeb mRNA were robustly expressed at similar levels in both control and AlkY1355S third-instar (L3) larval brains (Fig. 3A). To further investigate the effect of the AlkY1355S mutation, we analyzed L3 larval brains of control and AlkY1355S alleles using 3′ scRNA-seq based on the 10x Genomics platform. In total, high-quality scRNA-seq data were collected for 3967 control and 4099 AlkY1355S CNS cells and further analyzed with R (Seurat) and Python-based (Scanpy) pipelines. This analysis initially defined 19 cell clusters (Fig. S4A), which were merged based on canonical markers (Fig. S4B,D) into eight distinct clusters displayed on a uniform manifold approximation and projection (UMAP)-based two-dimensional projection (Fig. 3B). These eight clusters were defined as: early NBs, NB-enriched cells, immature neurons, mature neurons, NB proliferating cells, optic lobe epithelium (OLE), Repo-positive cells and Wrapper-positive cells. Canonical markers for these subsets were expressed in these clusters (Fig. 3C; Fig. S4D), and we identified several other unique marker genes for each subtype (Ariss et al., 2020; Brunet Avalos et al., 2019; Cattenoz et al., 2016; Estacio-Gómez et al., 2020; Michki et al., 2021) (Fig. S4E). In addition to these clusters, we identified two additional clusters representing hemocytes (cluster 3 in Fig. S4A) and cuticle-associated cells (cluster 18 in Fig. S4A,C) (Cattenoz et al., 2020; Dong et al., 2020; Evans et al., 2014; Öztürk-Çolak et al., 2016).

Fig. 3.

AlkF1251L and AlkY1355S larval brains display hyperplasia. (A) Expression of Alk (red) and jeb (green) mRNAs in situ are shown; DAPI is in blue. n>20. Two independent replicates. Scale bar: 50 µm. (B) Uniform manifold approximation and projection (UMAP) displaying the cellular heterogeneity of the control single-cell RNA-sequencing (scRNA-seq) dataset as eight cell types [early neuroblast (NB), NB-enriched cells, immature neurons, mature neurons, NB proliferating cells, optic lobe epithelium (OLE), Repo-positive cells and Wrapper-positive cells]. (C) Matrix plot visualizing canonical markers (two per cluster) defining the cellular heterogeneity of the scRNA-seq dataset. Scale indicates mean expression values. (D) Violin plots showing the mRNA expression of Alk and jeb across larval brain scRNA-seq clusters (blue denotes control and red denotes AlkY1355S). (E) Feature plots visualizing mRNA expression of Alk, jeb, elav, dpn, repo and wrapper within the single-cell population. Color scale indicates expression level. (F,G) Alk protein expression in the central brain area [one wandering third-instar (wL3) larval brain hemisphere is shown], in control (F) and AlkΔRA mutant (G). Alk (red), Elav (green) and Dlg (blue) are shown. Scale bar: 20 µm. n>30 larval brains in three replicates. (H) Schematic outlining the genomic organization of the Alk locus (blue). Intron–exon structure of both Alk-RA and Alk-RB transcripts is shown below. Open reading frame is in pink. The sequence of the AlkΔRA mutant is shown below, and the region deleted in the previously described AlkΔRB mutant (Mendoza-Garcia et al., 2017) is indicated for reference (red line). Schematic created with biorender.com. (I) Alk mRNA expression levels in control, homozygous AlkY1355S and AlkΔRA third-instar larval brain bulk RNA-seq. Three biological replicates (45 brains per sample) per genotype are shown. Ordinary one-way ANOVA, Dunnett's multiple comparison test; ***P<0.001. (J) Immunoblot indicating Alk protein levels in control, AlkY1355S and AlkΔRA third-instar larval brain lysates. Tubulin was employed as loading control. One representative image of two experimental replicates is shown. (K) Violin plot indicating elav mRNA expression across third-instar larval brain scRNA-seq clusters. Red oval highlights expression of elav in the NB-enriched cluster in AlkY1355S. (L) Orthogonal projections of the wL3 central brain area showing Elav-positive perikarya at the NB level. Scale bars: 10 µm. (M) Nuclei count per region of interest (ROI) in homozygous AlkY1355S, homozygous AlkF1251L, C155/+>jeb/+, homozygous AlkΔRA and C155/+>AlkEC/+. In box plots, the box represents the 25-75th percentiles, and the line indicates the median; whiskers indicate minimum and maximum values. n=number of brain lobes analyzed in three independent replicates. One-way ANOVA, Dunnett's multiple comparison test; ***P<0.001. scRNA-seq data from 45 dissected and pooled wL3 larval brains were used for B-E and K. ns, not significant.

Fig. 3.

AlkF1251L and AlkY1355S larval brains display hyperplasia. (A) Expression of Alk (red) and jeb (green) mRNAs in situ are shown; DAPI is in blue. n>20. Two independent replicates. Scale bar: 50 µm. (B) Uniform manifold approximation and projection (UMAP) displaying the cellular heterogeneity of the control single-cell RNA-sequencing (scRNA-seq) dataset as eight cell types [early neuroblast (NB), NB-enriched cells, immature neurons, mature neurons, NB proliferating cells, optic lobe epithelium (OLE), Repo-positive cells and Wrapper-positive cells]. (C) Matrix plot visualizing canonical markers (two per cluster) defining the cellular heterogeneity of the scRNA-seq dataset. Scale indicates mean expression values. (D) Violin plots showing the mRNA expression of Alk and jeb across larval brain scRNA-seq clusters (blue denotes control and red denotes AlkY1355S). (E) Feature plots visualizing mRNA expression of Alk, jeb, elav, dpn, repo and wrapper within the single-cell population. Color scale indicates expression level. (F,G) Alk protein expression in the central brain area [one wandering third-instar (wL3) larval brain hemisphere is shown], in control (F) and AlkΔRA mutant (G). Alk (red), Elav (green) and Dlg (blue) are shown. Scale bar: 20 µm. n>30 larval brains in three replicates. (H) Schematic outlining the genomic organization of the Alk locus (blue). Intron–exon structure of both Alk-RA and Alk-RB transcripts is shown below. Open reading frame is in pink. The sequence of the AlkΔRA mutant is shown below, and the region deleted in the previously described AlkΔRB mutant (Mendoza-Garcia et al., 2017) is indicated for reference (red line). Schematic created with biorender.com. (I) Alk mRNA expression levels in control, homozygous AlkY1355S and AlkΔRA third-instar larval brain bulk RNA-seq. Three biological replicates (45 brains per sample) per genotype are shown. Ordinary one-way ANOVA, Dunnett's multiple comparison test; ***P<0.001. (J) Immunoblot indicating Alk protein levels in control, AlkY1355S and AlkΔRA third-instar larval brain lysates. Tubulin was employed as loading control. One representative image of two experimental replicates is shown. (K) Violin plot indicating elav mRNA expression across third-instar larval brain scRNA-seq clusters. Red oval highlights expression of elav in the NB-enriched cluster in AlkY1355S. (L) Orthogonal projections of the wL3 central brain area showing Elav-positive perikarya at the NB level. Scale bars: 10 µm. (M) Nuclei count per region of interest (ROI) in homozygous AlkY1355S, homozygous AlkF1251L, C155/+>jeb/+, homozygous AlkΔRA and C155/+>AlkEC/+. In box plots, the box represents the 25-75th percentiles, and the line indicates the median; whiskers indicate minimum and maximum values. n=number of brain lobes analyzed in three independent replicates. One-way ANOVA, Dunnett's multiple comparison test; ***P<0.001. scRNA-seq data from 45 dissected and pooled wL3 larval brains were used for B-E and K. ns, not significant.

Alk and jeb were robustly expressed in the scRNA-seq dataset of both control and AlkY1355S larval brains. Both Alk and jeb were predominantly expressed in mature neuronal lineages (Fig. 3D,E), including cholinergic, glutamatergic and GABAergic neurons (Fig. S4F). This is in keeping with previously described expression of Alk in the medulla of the optic lobe (Bazigou et al., 2007) and of Jeb in cholinergic neurons (Okamoto and Nishimura, 2015). Interestingly, we detected very little Alk or jeb at the mRNA level in either glial or NB-enriched cell populations (Fig. 3D,E).

We next investigated Alk protein expression in the L3 larval brain, confirming that Alk was strongly expressed in Elav-positive progeny of both Type I and Type II NBs (Fig. 3F). To examine the role of Alk further, we generated an Alk mutant allele (AlkΔRA), specifically removing Alk expression in the larval brain (Fig. 3G,H). In previous work, transcripts of the Alk-RB isoform were specifically detected in the Drosophila embryonic VM (Mendoza-García et al., 2017). Employing isoform-specific probes for Alk-RA and Alk-RB, we confirmed the previously described VM-expression of Alk-RB (Mendoza-García et al., 2017) and revealed that the Alk-RA isoform is specifically expressed in the developing nervous system (Fig. S5A,B). Based on this, we employed CRISPR/Cas9-mediated targeted deletion to remove the Alk-RA transcript generating the AlkΔRA mutant allele (Fig. 3H). Indeed, expression of Alk protein in the L3 larval central brain was significantly reduced, which was further confirmed by RNA-sequencing (RNA-seq) analysis (Fig. 3G,I) and immunoblotting (Fig. 3J). AlkΔRA mutants were viable with no obvious defects during development, providing an excellent tool with which to further investigate Alk function in the larval brain.

AlkF1251L and AlkY1355S exhibit neuronal hyperplasia at larval stages

Because our scRNA-seq and Alk antibody staining highlighted robust expression of Alk in central brain neurons, we focused on this area for further investigation. A closer examination of the central brain area of AlkY1355S and AlkF1251L identified increased numbers of neurons relative to controls (Fig. 3L). This increase in Elav-positive neurons was also observed on pan-neuronal expression of Jeb with C155-Gal4 (Fig. 3L). Loss of Alk activity, either by overexpression of dominant-negative Alk (C155>Alk.EC) or in the AlkΔRA mutant (Fig. 3L), did not significantly affect the numbers of Elav-positive neurons. These results were further strengthened by a slight increase in Elav-expressing cells in the AlkY1355S scRNA-seq dataset (Fig. 3K). To quantify this phenotype, L3 larval brains were stained with 4′,6-diamidino-2-phenylindole (DAPI), and perikarya within a region of interest (ROI) at the NB level were counted. Significantly increased nuclei numbers were observed in both AlkY1355S and AlkF1251L mutant alleles as well as on Jeb overexpression (C155>Jeb) (Fig. 3M). Notably, these observations are in keeping with observations of hyperplasia in the nervous system of Alk gain-of-function mouse models (Borenäs et al., 2021; Cazes et al., 2014). No significant change in neuron number was observed in L3 larval brains lacking Alk activity, either employing an Alk dominant-negative transgene (C155>Alk.EC) or the AlkΔRA mutant allele, suggesting that Alk function is not critically required for differentiation. Thus, our data suggest that the larval brains of Alk gain-of-function alleles contain increased numbers of mature neurons.

scRNA-seq analysis identifies perturbed neuronal identity in AlkY1355S larval brains

Integration of AlkY1355S and control scRNA-seq datasets was performed to identify shared and distinct cell identities (Fig. 4A), highlighting increased numbers of cells in the ‘NB-enriched’ cluster (Fig. 4B), and further suggesting that increased Alk activation may result in a perturbation of NB-derived lineages and populations. We therefore further interrogated this cell cluster in our scRNA-seq datasets. Re-analysis of the NB-enriched cell cluster resulted in five cell clusters (Fig. 4C,D). Although cluster 1 was enriched in AlkY1355S compared to the control scRNA-seq dataset, similar numbers of miranda (mira)-positive NBs (clusters 3 and 4) were observed, and antibodies to Mira confirmed similar numbers of Mira-positive NBs in AlkY1355S and control larval brains (Fig. 4E). We also counted NBs per brain hemisphere in AlkY1355S, AlkF1251L and AlkΔRA, but were unable to observe any change in overall NB number in Alk mutant brains, indicating that the additional neurons observed in AlkY1355S and AlkF1251L brains were not a result of elevated NB numbers (Fig. 4F).

Fig. 4.

Single-cell analysis identifies perturbed NB lineages in AlkY1355S mutants. (A) UMAP of integrated scRNA-seq control (blue) and AlkY1355S (red) datasets. (B) Bar chart indicating the number of cells in each cluster across the whole brain cell population in both control (blue) and AlkY1355S (red) scRNA-seq datasets. (C) Bar chart displaying cell distribution in a re-analysis of the NB-enriched cell population in B. Increased numbers of cells are observed in cluster 1 (green rectangle) of AlkY1355S (red), compared to control (blue). (D) UMAPs showing the five sub-clusters (shown in C) identified in control and AlkY1355S from the NB-enriched population. (E) UMAPs indicating miranda (mira) mRNA expression in both control and AlkY1355S scRNA-seq datasets. Violin plots show mira mRNA expression and distribution in the subclusters of the NB-enriched cell population of control and AlkY1355S, together with Mira expression in wL3 larval brains. Scale bar: 20 µm. (F) Quantification of NBs per brain hemisphere in homozygous AlkY1355S, AlkF1251L and AlkΔRA compared to controls. One-way ANOVA, Dunnett's multiple comparisons test. n=brain lobes analyzed. (G) GFP-positive clones derived from Type I and Type II NBs in either control or homozygous AlkY1355S mutant backgrounds. Scale bar: 5 µm. Quantification shown on the right. Unpaired two-tailed Student's t-test; ***P<0.001. (H) Total number of Type II NBs per brain hemisphere in control and homozygous AlkY1355S. Unpaired two-tailed Student's t-test, Kolmogorov–Smirnov test. n=brain lobes analyzed. (I) Quantification of EdU-positive cells in Type II NB lineages as identified by anti-Asense staining for control and homozygous AlkY1355S wL3 larval brains. Unpaired two-tailed Student's t-test. Scale bars: 20 µm (left) and 5 µm (right). n=brain lobes analyzed. (J) EdU pulse-chase experiments show no significant difference in EdU-positive cells arising from NB Type I lineages in homozygous AlkY1355S larval brains compared with control. EdU incorporation was quantified for 3 h pulse and 3 h pulse plus 24 h chase conditions. Unpaired two-tailed Student's t-test, Mann–Whitney test. Scale bar: 5 µm. n=brain lobes analyzed. scRNA-seq data from 45 dissected and pooled wL3 larval brains were used for A-E. In box plots, the box represents the 25-75th percentiles, and the line indicates the median; whiskers indicate minimum and maximum values. Representative examples are shown. ns, not significant.

Fig. 4.

Single-cell analysis identifies perturbed NB lineages in AlkY1355S mutants. (A) UMAP of integrated scRNA-seq control (blue) and AlkY1355S (red) datasets. (B) Bar chart indicating the number of cells in each cluster across the whole brain cell population in both control (blue) and AlkY1355S (red) scRNA-seq datasets. (C) Bar chart displaying cell distribution in a re-analysis of the NB-enriched cell population in B. Increased numbers of cells are observed in cluster 1 (green rectangle) of AlkY1355S (red), compared to control (blue). (D) UMAPs showing the five sub-clusters (shown in C) identified in control and AlkY1355S from the NB-enriched population. (E) UMAPs indicating miranda (mira) mRNA expression in both control and AlkY1355S scRNA-seq datasets. Violin plots show mira mRNA expression and distribution in the subclusters of the NB-enriched cell population of control and AlkY1355S, together with Mira expression in wL3 larval brains. Scale bar: 20 µm. (F) Quantification of NBs per brain hemisphere in homozygous AlkY1355S, AlkF1251L and AlkΔRA compared to controls. One-way ANOVA, Dunnett's multiple comparisons test. n=brain lobes analyzed. (G) GFP-positive clones derived from Type I and Type II NBs in either control or homozygous AlkY1355S mutant backgrounds. Scale bar: 5 µm. Quantification shown on the right. Unpaired two-tailed Student's t-test; ***P<0.001. (H) Total number of Type II NBs per brain hemisphere in control and homozygous AlkY1355S. Unpaired two-tailed Student's t-test, Kolmogorov–Smirnov test. n=brain lobes analyzed. (I) Quantification of EdU-positive cells in Type II NB lineages as identified by anti-Asense staining for control and homozygous AlkY1355S wL3 larval brains. Unpaired two-tailed Student's t-test. Scale bars: 20 µm (left) and 5 µm (right). n=brain lobes analyzed. (J) EdU pulse-chase experiments show no significant difference in EdU-positive cells arising from NB Type I lineages in homozygous AlkY1355S larval brains compared with control. EdU incorporation was quantified for 3 h pulse and 3 h pulse plus 24 h chase conditions. Unpaired two-tailed Student's t-test, Mann–Whitney test. Scale bar: 5 µm. n=brain lobes analyzed. scRNA-seq data from 45 dissected and pooled wL3 larval brains were used for A-E. In box plots, the box represents the 25-75th percentiles, and the line indicates the median; whiskers indicate minimum and maximum values. Representative examples are shown. ns, not significant.

To address the dynamics of NB lineages, GFP-marked clones were generated in both control and AlkY1355S backgrounds (Fig. 4G). Analysis of clones descended from Type I NBs revealed significant increase in volume in AlkY1355S (Fig. 4G), in agreement with our earlier observation of increased numbers of Elav-positive perikarya in AlkY1355S larval brains. Analysis of Type II NB clone size showed a non-significant trend towards increased volume (Fig. 4G), which was also reflected by similar numbers of Type II NBs per brain lobe (Fig. 4H). We further employed 5-ethynyl-2′-deoxyuridine (EdU) pulse-chase analysis, subjecting AlkY1355S and control larvae to either (1) EdU pulse (1.5 h for NB II and 3 h for NB I) followed by immediate dissection or (2) EdU pulse followed by 24 h chase. EdU pulse-chase analyses did not reveal any significant differences in proliferation of Type I or Type II NBs when comparing AlkY1355S larval brains with controls (Fig. 4I,J), indicating that the AlkY1355S mutation does not increase proliferation rates in larval brains. Moreover, we employed the fluorescence ubiquitination cell cycle indicator (FUCCI) system in both wL3 larvae after eclosion and wL3 larvae, which enabled us to identify cells in different phases of the cell cycle (Zielke et al., 2014) (Figs S6 and S7). No differences in cell cycle phases were detected in AlkY1355S compared to the control, indicating that proliferation is not enhanced at the examined time points. Thus, our scRNA-seq and subsequent experimental validations suggest that the hyperplasia observed in AlkY1355S larval brains is not due to increased NB numbers or proliferation, implying that other mechanisms are responsible.

NB quiescence proceeds as normal in AlkY1355S mutants

In Drosophila, NBs enter a quiescent state during late embryogenesis and remain quiescent until larvae begin feeding (∼4 h after larval hatching) (Curt et al., 2019). Exceptions are the four MB NBs that do not undergo quiescence and continue dividing during late embryo and L1 stages (Ito and Hotta, 1992). To better understand the cause of the observed hyperplasia in AlkY1355S larval brains, we therefore examined NB quiescence in these mutants. Newly hatched AlkY1355S and control L1 larvae brains were dissected and stained for pH3 (also known as phospho-His3) (Fig. 5A-B′). Similar to controls, AlkY1355S mutants exhibited four pH3-positive MB NBs, indicating that there are no detectable defects in NB quiescence. Later, during early pupal development, NBs shrink, exit the cell cycle and terminally differentiate (Homem et al., 2014), and in adult flies there is no detectable NB proliferation. To exclude the possibility that NBs continue to proliferate at later stages in AlkY1355S mutants, we measured NB size in pre-pupae of control and AlkY1355S in comparison to wL3 larvae. Similar to control, AlkY1355S NBs undergo a progressive size reduction in pre-pupal stages indicating proper terminal differentiation (Fig. S8A,B). Finally, we did not detect excessive proliferation in adult brains of AlkY1355S mutants using anti-pH3 staining (Fig. S8C). Taken together, no perturbations in NB quiescence were observed in AlkY1355S mutants in first instar (L1), excluding this mechanism as the source of the observed hyperplasia. In addition, no change in proliferation dynamics could be detected at adult stages.

Fig. 5.

AlkY1355S larval brains exhibit decreased levels of apoptosis. (A,B) Four pH3-positive MB NBs are detected in both homozygous control (A) and AlkY1355S (B) first-instar larval brains. Scale bar: 20 µm. n>5 brains. (C) Gene Ontology (GO) analysis of genes enriched in sub-cluster 1 of the reanalyzed NB-enriched cells cluster in AlkY1355S, highlighting an enrichment in cell death-defective components in the AlkY1355S scRNA-seq dataset. (D) Expression of Diap1 in bulk RNA-seq from both control and homozygous AlkY1355S larval brains. Three biological replicates (45 brains per sample) per genotype are shown. (E) Violin plot indicating Diap1 expression in the NB-enriched cell population in homozygous AlkY1355S and controls. (F) Quantification of Cleaved caspase-1 staining (Dcp-1) in the central brain area of homozygous AlkY1355S, AlkF1251L and AlkΔRA compared to controls. One-way ANOVA, Dunnett's multiple comparison test; ***P<0.001 and **P<0.005. n=number of brain lobes analyzed. (G) TUNEL analysis visualizing apoptotic cells in homozygous AlkY1355S and controls. Unpaired two-tailed Student's t-test; ***P<0.001. n=number of brain lobes analyzed. (H) PARP1 reporter assay in control and homozygous AlkY1355S larval brains. Unpaired two-tailed Student's t-test; ***P<0.001. (I,I′) Volume analysis of the Engrailed-positive MC sub-lineage in homozygous AlkY1355S, AlkΔRA and controls. Scale bar: 20 µm. One-way ANOVA, Kruskal–Wallis test. n=number of analyzed brain lobes. AC, anterior cluster; MC, medial cluster; PC, posterior cluster. (J,J′) Analysis of Dcp-1-positive neurons adjacent to the Engrailed sub-lineages AC, PC and MC in homozygous AlkY1355S, AlkΔRA and controls. Scale bar: 5 µm. One-way ANOVA, Kruskal–Wallis test. AC, **P<0.007; MC1, P>0.05; MC2, P>0.05; PC, *P<0.05, **P<0.005. n=number of brain lobes analyzed. scRNA-seq data from 45 dissected and pooled wL3 larval brains were used for C-E. In box plots, the box represents the 25-75th percentiles, and the line indicates the median; whiskers indicate minimum and maximum values. Representative examples are shown. ns, not significant.

Fig. 5.

AlkY1355S larval brains exhibit decreased levels of apoptosis. (A,B) Four pH3-positive MB NBs are detected in both homozygous control (A) and AlkY1355S (B) first-instar larval brains. Scale bar: 20 µm. n>5 brains. (C) Gene Ontology (GO) analysis of genes enriched in sub-cluster 1 of the reanalyzed NB-enriched cells cluster in AlkY1355S, highlighting an enrichment in cell death-defective components in the AlkY1355S scRNA-seq dataset. (D) Expression of Diap1 in bulk RNA-seq from both control and homozygous AlkY1355S larval brains. Three biological replicates (45 brains per sample) per genotype are shown. (E) Violin plot indicating Diap1 expression in the NB-enriched cell population in homozygous AlkY1355S and controls. (F) Quantification of Cleaved caspase-1 staining (Dcp-1) in the central brain area of homozygous AlkY1355S, AlkF1251L and AlkΔRA compared to controls. One-way ANOVA, Dunnett's multiple comparison test; ***P<0.001 and **P<0.005. n=number of brain lobes analyzed. (G) TUNEL analysis visualizing apoptotic cells in homozygous AlkY1355S and controls. Unpaired two-tailed Student's t-test; ***P<0.001. n=number of brain lobes analyzed. (H) PARP1 reporter assay in control and homozygous AlkY1355S larval brains. Unpaired two-tailed Student's t-test; ***P<0.001. (I,I′) Volume analysis of the Engrailed-positive MC sub-lineage in homozygous AlkY1355S, AlkΔRA and controls. Scale bar: 20 µm. One-way ANOVA, Kruskal–Wallis test. n=number of analyzed brain lobes. AC, anterior cluster; MC, medial cluster; PC, posterior cluster. (J,J′) Analysis of Dcp-1-positive neurons adjacent to the Engrailed sub-lineages AC, PC and MC in homozygous AlkY1355S, AlkΔRA and controls. Scale bar: 5 µm. One-way ANOVA, Kruskal–Wallis test. AC, **P<0.007; MC1, P>0.05; MC2, P>0.05; PC, *P<0.05, **P<0.005. n=number of brain lobes analyzed. scRNA-seq data from 45 dissected and pooled wL3 larval brains were used for C-E. In box plots, the box represents the 25-75th percentiles, and the line indicates the median; whiskers indicate minimum and maximum values. Representative examples are shown. ns, not significant.

Alk mutants display reduced levels of apoptosis

In models of childhood neuroblastoma, expression of the MYCN oncogene in the developing neural crest drives tumorigenesis. In a zebrafish neuroblastoma model, the additional transgenic expression of the AlkF1174L activated variant overcomes excessive apoptosis observed in MYCN-only tumors, decreasing cell death and promoting tumorigenesis (Zhu et al., 2012), suggesting an important role for Alk signaling in promotion of survival. Moreover, the zebrafish Alk family RTK Leukocyte tyrosine kinase (Ltk) that functions in neural crest-derived iridiophore development has been reported to promote survival when carrying a neuroblastoma-associated mutation (Fadeev et al., 2016). Further, in the fly visual system, Alk has been shown to be important for the survival of L3 neurons (Pecot et al., 2014), prompting us to address apoptosis in more detail. Indeed, Gene Ontology (GO) analysis of cluster 1 in our scRNA-seq dataset identified an enrichment of ‘cell-death-defective’ markers in postembryonic AlkY1355S mutant brains (Fig. 5C). These included the Diap1 suppressor of apoptosis, which was also upregulated in bulk RNA-seq of AlkY1355S brains (Fig. 5D,E). This prompted us to test whether decreased apoptosis in AlkY1355S and AlkF1251L mutant brains may lead to the observed increased numbers of neurons. To investigate this, Cleaved caspase-1 (Dcp-1; also known as DCP1) antibody staining of AlkY1355S, AlkF1251L and AlkΔRA wL3 brains was performed, and Dcp-1-positive cells in the central brain area were counted. Significantly reduced numbers of Dcp-1-positive cells were observed in both AlkY1355S and AlkF1251L mutants, although we did not observe increased apoptosis in AlkΔRA mutant larval brains (Fig. 5F). The Dcp-1 analysis was complemented with two additional apoptosis assays – first, a terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay and, second, a PARP1 reporter – which both confirmed reduced levels of apoptosis in AlkY1355S mutants (Fig. 5G,H). Finally, we analyzed apoptosis in the controlled setting of the Engrailed-positive anterior cluster (AC), posterior cluster (PC), medial cluster (MC)1 and MC2 hemi-lineages, in which immediate progeny of specific GMCs are removed via apoptosis (Kumar et al., 2009). In the MC2 lineage, all Engrailed-positive neurons are removed via apoptosis, while Engrailed-positive or -negative neurons in the MC1 lineage do not undergo programmed cell death (Kumar et al., 2009). In the AC and PC lineage, the Engrailed-negative GMC progeny are removed via apoptosis (Kumar et al., 2009). Analysis of the overall volume of the MC lineage via Engrailed staining, while simultaneously staining for Dcp-1, failed to identify any differences in MC lineage size in either AlkY1355S or AlkΔRA compared with control (Fig. 5I). However, analysis of Dcp-1-positive cells within and in proximity to the other hemi-lineages identified significantly more Dcp-1-positive cells around the AC lineage in AlkΔRA compared to control and AlkY1355S. This finding was supported by the presence of significantly fewer Dcp-1-positive neurons around the PC lineage in AlkY1355S compared to control and AlkΔRA (Fig. 5J). Taken together, these four independent assays support decreased levels of apoptosis in AlkY1355S mutants as a contributory factor to the increased numbers of surviving neurons observed.

Perturbed neuronal fate in the MB lineage in AlkY1355S mutant brains

Interestingly, scRNA-seq analysis highlighted increased proportions of cells that exhibited high levels of expression of neuronal lineage markers such as mamo, br, Tlk, nkd, DnaJ-1, Hr4 and Hsp68 in cluster 1 of AlkY1355S larval brains (Fig. 6A,B). To visualize differentially expressed markers, we analyzed the expression of the BTB/POZ-containing C2H2 zinc finger transcription factor Mamo via antibody staining in control and AlkY1355S. Mamo has been reported to be critical for temporal specification of MB α′β′-neurons (Liu et al., 2019; Rossi and Desplan, 2020). Initial investigation confirmed that Mamo is expressed in Alk-positive larval neurons in the MB lineage (Fig. 6C). As our scRNA-seq datasets detected significantly more mamo expression in the AlkY1355S mutant allele (Fig. 6A,B), we measured the overall volume of Mamo-positive cell clusters within the MB lineage, observing an increase in the total volume of Mamo-positive cells in AlkY1355S (Fig. 6D) compared to the control, validating our scRNA-seq data.

Fig. 6.

Ectopic Mamo expression in the mushroom body (MB) γ-lineage in AlkY1355S indicates perturbed α′β′- and γ-neuronal differentiation. (A) Violin plots indicating mRNA expression of mamo, br, Tlk, nkd, DnaJ-1, Hr4 and Hsp68 in cluster 1 of the AlkY1355S scRNA-seq dataset. (B) Feature plots projecting mamo mRNA expression in both wild-type and AlkY1355S scRNA-seq datasets. (C) Mamo (green) and Alk (red) are co-expressed in neurons. Scale bar: 5 µm. n>10 larval brains. (D) Quantification of Mamo-positive cell area in homozygous AlkY1355S compared to controls. In box plots, the box represents the 25-75th percentiles, and the line indicates the median; whiskers indicate minimum and maximum values. Representative examples are shown. Unpaired two-tailed Student's t-test; ***P<0.001. n=number of brain lobes analyzed. Scale bar: 20 µm. (E) Schematic showing the generation of the three different major MB lineages during development (adapted from Lee et al., 1999; Liu et al., 2019). Schematic created with biorender.com. This image is not published under the terms of the CC-BY license of this article. For permission to reuse, please see Lee et al. (1999). GMC, ganglion mother cell. (F) Mamo is detected in the MB γ-neuron lineage (EcR-B1 positive) in homozygous wL3 AlkY1355S larvae, but not in controls or the homozygous AlkΔRA mutant. Overexpression of Jeb ligand (C155/+>jeb/+) phenocopies the ectopic Mamo expression in the γ-lineage observed in AlkY1355S. Scale bar: 20 µm. n>brain lobes analyzed per genotype in Fig. S9B. (G) MB-specific OK107-Gal4 driver overexpressing Jeb (OK107/+>jeb/+) phenocopies the AlkY1355S phenotype. Scale bar: 20 µm. n>brain lobes analyzed per genotype in Fig. S9B. (H) Schematic summarizing the ectopic Mamo expression observed in homozygous AlkY1355S and C155/+>jeb/+ compared to control MB lineages. Schematic created with biorender.com. scRNA-seq data from 45 dissected wL3 larval brains were used for A and B.

Fig. 6.

Ectopic Mamo expression in the mushroom body (MB) γ-lineage in AlkY1355S indicates perturbed α′β′- and γ-neuronal differentiation. (A) Violin plots indicating mRNA expression of mamo, br, Tlk, nkd, DnaJ-1, Hr4 and Hsp68 in cluster 1 of the AlkY1355S scRNA-seq dataset. (B) Feature plots projecting mamo mRNA expression in both wild-type and AlkY1355S scRNA-seq datasets. (C) Mamo (green) and Alk (red) are co-expressed in neurons. Scale bar: 5 µm. n>10 larval brains. (D) Quantification of Mamo-positive cell area in homozygous AlkY1355S compared to controls. In box plots, the box represents the 25-75th percentiles, and the line indicates the median; whiskers indicate minimum and maximum values. Representative examples are shown. Unpaired two-tailed Student's t-test; ***P<0.001. n=number of brain lobes analyzed. Scale bar: 20 µm. (E) Schematic showing the generation of the three different major MB lineages during development (adapted from Lee et al., 1999; Liu et al., 2019). Schematic created with biorender.com. This image is not published under the terms of the CC-BY license of this article. For permission to reuse, please see Lee et al. (1999). GMC, ganglion mother cell. (F) Mamo is detected in the MB γ-neuron lineage (EcR-B1 positive) in homozygous wL3 AlkY1355S larvae, but not in controls or the homozygous AlkΔRA mutant. Overexpression of Jeb ligand (C155/+>jeb/+) phenocopies the ectopic Mamo expression in the γ-lineage observed in AlkY1355S. Scale bar: 20 µm. n>brain lobes analyzed per genotype in Fig. S9B. (G) MB-specific OK107-Gal4 driver overexpressing Jeb (OK107/+>jeb/+) phenocopies the AlkY1355S phenotype. Scale bar: 20 µm. n>brain lobes analyzed per genotype in Fig. S9B. (H) Schematic summarizing the ectopic Mamo expression observed in homozygous AlkY1355S and C155/+>jeb/+ compared to control MB lineages. Schematic created with biorender.com. scRNA-seq data from 45 dissected wL3 larval brains were used for A and B.

To examine Mamo expression in AlkY1355S larval brains further, we focused on the MB NB lineage in wL3. The MB lineage comprises three major cell types: early-born γ-neurons, middle-born α′β′-neurons and late-born αβ-neurons (Fig. 6E) (Lee et al., 1999). Mamo expression is restricted to α′β′-neurons, whereas EcR-B1 is exclusively expressed within γ-neurons in wL3 (Fig. 6E) (Lee et al., 2000; Liu et al., 2019; Rossi and Desplan, 2020). Remarkably, we observed ectopic expression of Mamo in EcR-B1-positive γ-neurons in AlkY1355S mutant wL3 brains (Fig. 6F,H). To confirm that excessive Alk activity results in improper Mamo expression, we ectopically overexpressed Jeb with C155-Gal4, which also resulted in ectopic expression of Mamo in EcR-B1-positive MB γ-neurons (Fig. 6F,H). Analysis of Mamo in AlkΔRA mutants showed that although increased Alk activation leads to Mamo expression, Alk is not critically required for normal Mamo expression in the α′- and β′-lineage, as Mamo expression in AlkΔRA mutants MB α′β′-neurons is unchanged (Fig. 6F). These findings were confirmed by expression of Jeb using the MB-specific OK107-Gal4 and 201γ-Gal4 drivers, which also led to Mamo expression in wL3 EcR-B1-positive MB γ-neurons (Fig. 6G; Fig. S9, quantification in Fig. S9B).

We next asked whether ectopic Mamo expression leads to a neuronal fate change in γ-neurons in AlkY1355S larval brains that is maintained in adults, comparing axonal morphology and the MB cell bodies (MBCs) in wild-type and AlkY1355S with antibodies to the Rho guanine exchange factor (RhoGEF) Trio. Trio is important for directional extension of neurites in the MB and is strongly expressed in the cytoplasm and plasma membrane of α′β′-cell bodies, whereas in γ-cell bodies expression is enriched at plasma membranes but largely absent from the cytoplasm (Awasaki et al., 2000). We also examined the Abrupt zinc finger BTB domain-containing transcription factor that functions in motoneuron guidance and connectivity (Hu et al., 1995).

In adult flies, the γ-, α′β′- and αβ-MBCs branch anteriorly through the peduncle into three main lobes: the bifurcating α′β′- and αβ-lobes, and a single γ-lobe (Fig. 7A) (Crittenden et al., 1998). No gross defects in overall MB morphology were observed in AlkY1355S adult brains (Fig. S10); however, examination of Trio and Abrupt expression in MBCs identified more cell bodies with enhanced and cytoplasmic Trio expression (Fig. 7B), whereas expression of Abrupt was decreased in AlkY1355S. These results were strengthened by increased cytoplasmic Trio-positive cell bodies in OK107>jeb adults (Fig. 7C), further indicating that increased Alk signaling shifts γ identity to a more α′β′ neuronal identity in larval stages, which is maintained through metamorphosis into adulthood.

Fig. 7.

Perturbed neuronal differentiation in AlkY1355S is maintained to adulthood. (A) Schematic displaying the three main MB neuronal types (αβ, α′β′ and γ) and the mushroom body cell bodies (MBC) in adults. (B) Analysis of Trio and Abrupt in the MBC of homozygous AlkY1355S and controls. Scale bars: 20 µm. (C) Trio expression in MBC of OK107/+>jeb/+ and OK107/+ controls. Scale bars: 10 µm.

Fig. 7.

Perturbed neuronal differentiation in AlkY1355S is maintained to adulthood. (A) Schematic displaying the three main MB neuronal types (αβ, α′β′ and γ) and the mushroom body cell bodies (MBC) in adults. (B) Analysis of Trio and Abrupt in the MBC of homozygous AlkY1355S and controls. Scale bars: 20 µm. (C) Trio expression in MBC of OK107/+>jeb/+ and OK107/+ controls. Scale bars: 10 µm.

Taken together, our data show that neuroblastoma-associated Alk point mutations do not result in changes in NB number, or in gross increases in proliferation, but rather perturb neuronal fate specification, which leads to precocious Mamo expression in the γ-cell lineage of AlkY1355S mutant wL3 brains that persists until adult stages.

This work set out to address mechanisms by which Alk mutations affect neural development. This is particularly relevant in pediatric neuroblastoma, where patients with ALK mutations exhibit poor prognosis, and tumors are considered to arise due to defective differentiation during development of the nervous system. For this reason, we generated and characterized two independent Alk mutations (AlkY1355S and AlkF1251L) in Drosophila, for which the human orthologs (ALKY1278S and ALKF1174L) have previously been characterized as ‘gain-of-function’ mutations in neuroblastoma patients (Chand et al., 2013; Guan et al., 2017).

Flies harboring the in locus modifications AlkY1355S or AlkF1251L are homozygous viable, which is in line with observations in vertebrate model systems in which Alk gain-of-function mutations have been investigated (Borenäs et al., 2021; Cazes et al., 2014; Fadeev et al., 2016). Employing Drosophila allowed us to investigate neurogenesis in a highly controlled environment, revealing, first, that AlkY1355S and AlkF1251L mutant brains exhibit mild neuronal hyperplasia and, second, no increase in NB numbers. This is in keeping with reports of hyperplasia in peripheral nervous system ganglia of Alk ‘gain-of-function’ mice (Borenäs et al., 2021; Cazes et al., 2014; Ono et al., 2019). Interestingly, although the mechanisms underlying this neuronal hyperplasia are not well understood, it is clear that mutations in the endogenous Alk locus collaborate with the MYCN oncogene to drive highly penetrant and aggressive neuroblastoma in mice (Borenäs et al., 2021; Cazes et al., 2014; Ono et al., 2019).

Interestingly, reduced apoptosis leads to hyperplasia in the nervous system of transgenic zebrafish expressing Alk gain-of-function variants (Zhu et al., 2012), and regulation of apoptosis during development is important to establish final neuron numbers in the CNS in Drosophila (Pinto-Teixeira et al., 2016). Indeed, our results reveal decreased apoptosis as a contributing mechanism underlying the hyperplastic phenotype observed in AlkY1355S and AlkF1251L brains. Additional reports suggest that Alk signaling leads to advantages in growth and cell competition (Wolfstetter et al., 2020), and Alk has been shown to promote L3 neuronal survival in the Drosophila optic lobe (Pecot et al., 2014). Further, knockdown or inhibition of Alk in zebrafish induces apoptosis in the fish hindbrain (Yao et al., 2013).

One important aspect is the expression of Alk itself. Our scRNA-seq datasets identified strong expression of Alk mRNA in neuronal lineages particularly in mature neurons, but not in NBs, and this was confirmed at the protein level by immunostaining. This Alk expression profile is in line with an anti-apoptotic effect in neurons later during the differentiation process. Whether AlkY1355S mutants are protected from apoptosis throughout development and in the adult brain is not known. However, in zebrafish, aberrant signaling by the Alk family RTK, Ltk, which carries a neuroblastoma-related mutation (Ltkmoonstone), promotes survival of neural crest-derived iridophores and increases their final number (Fadeev et al., 2016). In addition, our findings that AlkY1355S and AlkF1251L do not drive uncontrolled proliferation in the Drosophila brain support earlier reports in mouse and zebrafish models (Borenäs et al., 2021; Cazes et al., 2014; Fadeev et al., 2016; Ono et al., 2019), where mutation of Alk alone is insufficient to drive spontaneous tumor development.

An important tool generated in this study is the AlkΔRA mutant in which Alk mRNA and protein expression in the larval CNS is strongly reduced. Analysis of AlkΔRA suggests that Alk activity is not critical for neuronal differentiation in the MBs. Our observation of increased apoptosis in this AlkΔRA mutant, although slight, is in keeping with the finding of decreased apoptosis in AlkY1355S and AlkF1251L brains. Whether higher levels of apoptosis might be observed in conditions of stress or injury in the AlkΔRA mutant will be interesting to study in the future.

Our results also produced several surprising findings. One of these was that neither AlkY1355S nor AlkF1251L was able to rescue founder cell specification in the developing VM in the absence of Jeb, suggesting that these activating Alk alleles are still ligand dependent. The issue of ligand dependency has not been tested in either mouse, zebrafish or human systems, in which ALK is activated by the ALKAL ligands (Fadeev et al., 2018; Guan et al., 2015; Mo et al., 2017; Reshetnyak et al., 2015). Although it is clear that both AlkY1355S and AlkF1251L are ligand dependent in the developing fly VM, we do not have the tools to test this in the Drosophila brain and therefore cannot be sure if the observed ligand dependence is tissue specific. Nonetheless, this finding has interesting and important implications for neuroblastoma. Although human ALK-F1174L and ALK-Y1278S (orthologous to Drosophila AlkF1251L and AlkY1355S) are classified as ligand-independent mutations (Chand et al., 2013), we cannot exclude that these human ALK mutations still require ligands to initiate activation at endogenous levels of expression. Thus, ALK ligands might still be important for tumor formation and progression, even in the case of activating ALK mutations.

The use of scRNA-seq analysis, in addition to bulk RNA-seq, was critical to identify apoptotic and neuronal differentiation defects during development in AlkY1355S mutants, revealing potential Alk signaling targets including mamo, a BTB transcription factor that defines terminal identity in certain MB neurons. In Drosophila, the MB is composed of ∼2000 neurons of the three main neuronal subtypes: γ, α′β′ and αβ (Cognigni et al., 2018; Ito et al., 1997; Lee et al., 1999). γ-, α′β′- and αβ-neurons are produced sequentially during defined developmental periods in response to Imp and Syp gradients (Liu et al., 2015; Ren et al., 2017; Syed et al., 2017a; Yang et al., 2017). Elegant work has shown that terminal identity and maintenance of MB α′β′-neurons is established by Mamo expression (Liu et al., 2019), which depends on extrinsic receptor signaling via the activin receptor Babo (Rossi and Desplan, 2020). Although we did not identify neuronal differentiation defects in AlkΔRA MBs, the γ-neuron lineage was Mamo positive upon pan-neuronal expression of Jeb and in AlkY1355S wL3 brains. Thus, aberrant Alk activity leads to inappropriate Mamo expression in the γ-lineage at this stage, and perturbation of MB neuronal fate that persists to adult stages, where MB neuron cell bodies exhibit expanded Trio expression. Interestingly, Alk signaling has previously been linked to learning and memory (Gouzi et al., 2011; Weiss et al., 2017, 2012), which could be caused by fate perturbations in MB neuron lineages.

Together, the results presented here identify neuronal hyperplasia caused by decreased apoptosis in AlkY1355S and AlkF1251L mutant brains. scRNA-seq identified molecular neuronal differentiation defects that were validated in the well-characterized process of MB neuron development. To our knowledge, this is the first time that molecular defects in neuronal differentiation have been identified in response to Alk-activating mutations. These findings have potentially important implications for pediatric neuroblastoma, where mutation of human ALK at orthologous sites (ALK-Y1278S and ALK-F1174L) is associated with tumor progression in the developing peripheral nervous system.

Drosophila husbandry

Standard Drosophila husbandry procedures were followed (Ashburner, 1989). Drosophila stocks were reared on standard diet at room temperature. Crosses were performed at 25°C, 60% humidity and 12 h/12 h day–night cycle.

Fly stocks

Fly stocks used were obtained from the Bloomington Drosophila Stock Center (NIH P40OD018537): P{GawB}elavC155 (C155-Gal4) (BL-458), y1, {Mvas-Cas9}ZH-2A, w1118 (BL-51323; used as wild-type control), w1118; P{70FLP}10 (BL-6938), w1118; P{GAL4-Act5C(FRT.CD2).P}S (BL-4780), P{UAS-RFP.W}3/TM3, Sb1 (Bl-30558), w*; P{Ubi-p63E(FRT.STOP)Stinger}15F2 (BL-32251), y1 *; P{UAS-CD8::PARP1-Venus}3 (BL-65609), w*; P{GawB}OK107 eyOK107/In(4)ciD, ciD panciD svspa-pol (BL-854), y1 w67c23; P{UAS-mCD8::GFP.L}LL5 P{GawB}Tab2201Y (BL-64296). Other lines used were as follows: P{UAS-Alk.EC.MYC} (Bazigou et al., 2007), P{UAS-jeb} (Varshney and Palmer, 2006), jebweli (Stute et al., 2004), UAS-hALK.F1174L and UAS-hALK (Martinsson et al., 2011), UAS-hALK.Y1278S (Guan et al., 2017), UAS-GFP.caax (Finley et al., 1998), TI{TI}AlkKO (Wolfstetter et al., 2017).

Generation of Alk mutations employing CRISPR/Cas9-mediated genome editing

The AlkF1251L and AlkY1355S alleles were generated using CRISPR/Cas9-induced HDR (Gratz et al., 2013). sgRNA target sites were identified using the flyCRISPR target finder (https://flycrispr.org/) (Gratz et al., 2014). Two sgRNAs for each approach (sgRNA1 Y1355S: TCGCCGATTTTGGCATGTCCCGG; sgRNA2 Y1355S: TCGGACTACTATCGCAAGGGAGG; sgRNA1 F1251L: CCTGAAGGAGGCGGCCATAATGG; sgRNA2 F1251L: CAAGTTCAATCACCCGAATATGG) were cloned into pBFv-U6.2 expression vector (Genome Engineering Production Group at Harvard Medical School). HDR donor vectors were generated by Genescript. Design of the AlkY1355S and AlkF1251L donor constructs was as follows: Y1355S: containing silent mutations to prevent annealing and destruction of the donor by Cas9: TtGCtGAcTTcGGtATGTCaCG and TCaGAtTAtTAcCGtAAaGGtGG; homology arms: 507 bp upstream, 506 bp downstream of the desired mutation site (Fig. S11). F1251L: silent mutations plus integrated nucleotide exchange to create the desired mutation: CTaAAaGAaGCaGCaATcATGGCaAAGcTtAAcCAtCCaAAcATG; homology arms: 1052 bp upstream, 902 bp downstream (Fig. S12). sgRNAs and donor constructs were injected into y1, {Mvas-Cas9}ZH-2A, w1118 embryos by BestGene Inc. Positive CRISPR/Cas9 mutants were identified by PCR screening of single males using primers directed against the region of HDR-integrated silent mutations (forward primer, 5′-ATCCTAATGATCTCGCTTGCCGTG-3′; Y1355S reverse primer, 5′-CTcCCGATCaGAtTAtTAcCGtAAaG-3′; F1251L reverse primer, 5′-TTtGGaTGgTTaAgcTTtGCCATgAT-3′) and subsequent sequencing (Eurofins Genomics).

To generate the kinase-dead AlkD1347A mutant allele and the AlkD1347A, Y1355S double mutant, the AlkY1355S HDR donor construct was modified accordingly using a Q5® site-directed mutagenesis kit (NEB, E0554S). Owing to the proximity of both codons within the sequence, the guide RNA target sequences (sgRNA Y1355S nr.1 and nr.2, cloned into pU6-BbsI-chiRNA) (Gratz et al., 2013) were used for injection into y1, {Mvas-Cas9}ZH-2A, w1118 embryos. Positive candidates were identified by single-fly PCR screening with Y1355S forward and reverse primers (AlkD1347A, Y1355S) or lethality screening (AlkD1347A) and further sequenced (Eurofins Genomics) (Fig. S13). All positive candidates obtained failed to complement the AlkKO kinase domain deletion mutation.

The AlkΔRA mutant allele was created by CRISPR/Cas9-induced non-homologous end joining in a dual-guide approach (sgRNA10_RA, 5′-CCGTCTATCCGCGATTCTGAGGG-3′; sgRNA20_RA, 5′-GCGACAGTGGCGCACTCTGGCGG-3′) targeting the 5′UTR of the Alk-RA isoform. sgRNA sequences were cloned into the pBFv-U6.2 expression vector (Genome Engineering Production Group at Harvard Medical School) and the subsequent constructs injected into y1, {Mvas-Cas9}ZH-2A, w1118 embryos by BestGene Inc. Screening of deletion events was performed on F2 single males by PCR using primers flanking the intended deletion (forward primer, Alk-RA Fw1, 5′-cgaaatttttcctgcagctc-3′; reverse primer, Alk-RA Rv1, 5′-atggggtccttaatgcactg-3′), and deletions were further characterized by sequencing (Eurofins Genomics) (Fig. S14).

Pupal size measurement

Six virgin females and four males of each genotype were reared for 2 days on a standard diet at 25°C. The flies were removed, and the vials were kept at 25°C. Shortly before hatching, female pupae were scored and collected on double-sided sticky tape on a glass slide, briefly frozen at −20°C and analyzed with a ZEISS AxioZoom V16 stereo microscope.

Fixation and immunohistochemistry staining of embryos

Embryo staining was carried out as described previously (Muller, 2008). For pERK staining, the protocol from Gabay et al. (1997) was followed.

Fixation and immunohistochemistry staining of larval and adult brains

Brains of all stages were dissected in PBS, collected on ice in PBS containing 3.7% formaldehyde, and subsequently fixed in the same solution for 30 min at room temperature (RT), followed by permeabilization in PBS containing 1% Triton X-100 for 10 min and three washes in PBS containing 0.5% Triton X-100 (PBT). Samples were blocked in 5% goat serum (Jackson ImmunoResearch) in PBT, then primary antibody was added overnight at 4°C. Samples were washed four times in PBT and incubated in PBT containing the secondary antibody and DAPI for 2 h at RT followed by four washes in PBT. Samples were mounted in Fluoromount G and analyzed with a ZEISS Axio Imager.72 microscope. A ZEISS LSM800 confocal microscope and ZEN 3.5 Blue edition software were used to acquire images. Acquired images were analyzed with ImageJ version 1.53c and assembled using Affinity Photo and Designer version 1.10.0.1127.

Antibodies

Primary antibodies used were as follows: guinea pig (gp)-anti-Alk (1:1000) and rabbit (rb)-anti-Alk (1:1000) (Lorén et al., 2001), gp-anti-Jeb (1:1500) (Englund et al., 2003), mouse (m)-anti-activated MAPK/dephosphorylated ERK1/2 (1:200, Sigma-Aldrich, M8159), rb-anti-pH3 (1:500, Millipore, 0657C), rb-anti-Miranda (1:1000, Abcam, ab197788), chicken (ch)-anti-GFP (1:500, Abcam, ab13970), rb-anti-RFP (1:1000, Abcam, ab62341), rb-anti-cleaved PARP1 (1:50, Abcam, ab2317, anti-β-Galactosidase (1:150, Abcam, ab616), rb-anti-Tubulin (1:5000; Cell Signaling Technology, 11H10) and rb-anti-Dcp-1 (1:50; Cell Signaling Technology, 9578), as well as m-anti-Dlg (1:500; 4F3), rat-anti-Elav (1:100; 9F8A9), m-anti-EcR-B1 (1:100; ADA4.4), m-anti-Prospero (1:100; MR1A), m-anti-Trio (1:100; 9.4A), m-anti-Abrupt (1:50) and m-anti-Engrailed (1:50; 4D9) from Developmental Studies Hybridoma Bank. Guinea pig-anti-Mamo (1:100) was a gift from Claude Desplan’s laboratory (Rossi and Desplan, 2020); rb-anti-Ase (1:400) was a gift from Cheng-Yu Lee’s laboratory (Weng et al., 2010). Secondary antibody goat anti-rb HRP (Invitrogen, 11859140) was used for immunoblotting; other secondary antibodies were purchased from Jackson ImmunoResearch. DAPI (1 mg/ml) (Sigma-Aldrich, D9564-10MG) was used at 1:1000.

In situ hybridization

Whole-mount in situ hybridization was carried out as described previously (Lécuyer et al., 2008) with adaptations according to Pfeifer et al. (2014).

HCR v.3

The HCR probe sets were generated by Molecular Instruments (www.molecularinstruments.com) against full-length Alk and jeb mRNA sequences from the NCBI database (accession numbers NM_001274098.1 and NM_136882, respectively). HCR amplifiers were as follows: jeb, B3 with the A488 amplifier fluorophore; Alk, B2 with the 546 amplifier fluorophore. Fluorescent in situ hybridization was carried out according to the protocol for Drosophila embryos for HCR v.3 from Molecular Instruments with the following modifications: Proteinase K digestion was carried out with 0.01 mg/ml proteinase K in PBT for 10 min.

Immunoblotting

Brains from 30 wL3 larvae were dissected, collected on ice and subsequently lysed in RIPA buffer. Protein concentration was measured using a BCA Protein Assay Kit (Thermo Fisher Scientific). Laemmli buffer (final concentration 1×) was added prior to loading on a 7.5% SDS-PAGE gel. After transfer to PVDF (Millipore, IPVH00010), membranes were immunoblotted with primary antibodies overnight at 4°C. Primary antibodies used were gp-anti-Alk (1:3000) (Lorén et al., 2001) and rb-anti-tubulin (1:5000; Cell Signaling Technology, 11H10). After washing, membranes were incubated with rb-anti-HRP (Invitrogen, 11859140) secondary antibodies at RT for 1 h. ECL prime (Thermo Fisher Scientific, GERPN2236) was used for detection.

Clonal analysis

To generate RFP or GFP clones, hs-flp females were crossed to males carrying Act5c-Gal4>FRT.CD2>UAS-RFP or Ubi-p68E>FRT.stop>stinger in either control or AlkY1355S backgrounds. L1 larvae were collected from 2 h collection plates after hatching and grown at 25°C for 72 h. Larvae were subsequently heat shocked at 37°C for 10 min and reared at 25°C for a further 24 h.

EdU pulse and chase experiments

The Click-iT™ EdU Alexa Fluor™ 488 Imaging Kit from Thermo Fisher Scientific (C10329) was employed according to the manufacturer's instructions. L3 larvae were reared on 0.2 M EdU in potato-based fly food for 3 h (3 h pulse) for analysis of NB I, 1.5 h (1.5 h pulse) for NB II and dissected immediately afterwards, or reared for 24 h at 25°C (24 h chase, for NB I) and then dissected and fixed. Staining was carried out according to the manufacturer's instructions.

TUNEL assay

ApopTag® Fluorescein In Situ Apoptosis Detection Kit (Sigma-Aldrich, S7110) was employed according to the manufacturer's instructions.

In vitro brain culture

wL3 larvae were washed twice in PBS and once in 70% ethanol before collection and dissection in Schneider's medium. Dissected brains were cultured in Schneider's medium containing 10% heat-activated fetal bovine serum (FBS; Sigma-Aldrich, F7524), 0.01% insulin solution (Sigma-Aldrich, I0516-5ML), 1% penicillin/streptavidin (HyClone, SV30010), 1 µg/ml 20-hydroxyecdysone (Sigma-Aldrich, H5142-5MG) for 18 h, 36 h or 120 h. Prior to fixation, brains were washed three times in PBS and then fixed in 4% formaldehyde in PBS for 20 min at RT. Staining was carried out as described above in the ‘Fixation and immunohistochemistry staining of larval and adult brains’ staining section.

CNS dissociation for cell sorting and scRNA-seq analysis

Forty-five wL3 larvae per genotype were collected and washed in PBS. CNSs were dissected and collected in ice-cold PBS for a maximum of 1 h. After rinsing twice in ice-cold PBS, CNSs were incubated in 200 µl dissociation solution (1 mg/ml collagenase I, Merck, C9891) for 1 h with continuous agitation, after which the reaction was stopped by addition of 10 ml PBS [0.04% bovine serum albumin (BSA)] and left for 10 min to precipitate larger debris. The resulting dissociated cell solution was sieved through 70 µm and 40 µm cell strainers to remove cell clumps. The filtered suspension was centrifuged at 860 g for 5 min at 4°C. Supernatant was discarded, and cells were resuspended at 108 cells/ml (or at least in 100 μl) PBS, 2% FBS and 1 mM CaCl2. Dead cells or debris from the dissociated samples were removed using an EasySep Dead Cell Removal (Annexin V) Kit (STEMCELL, 17899) according to the manufacturer's guidelines. The remaining cells were respectively labeled with aqua-fluorescent reactive dye (dying cells) and Calcein Violet AM (living cells) using a LIVE/DEAD Violet Viability/Vitality Kit (Molecular Probes, L34958) according to the manufacturer's guidelines. Finally, each sample was washed twice in PBS, 2% FBS and resuspended in 500 µl PBS, 2% FBS. Living cells were enriched using a FACSAria III cell sorter (BD Biosciences) based on LIVE/DEAD staining. Cells were sorted using an 85 µm nozzle into Eppendorf tubes that had been pre-coated with PBS containing 2% BSA.

Single-cell library preparation, sequencing and scRNA-seq analysis

Approximately 7000 sorted cells were directly loaded in sheath fluid onto one lane of a Chromium 10x chip (10x Genomics), and libraries were prepared using the normal workflow for Single Cell 3´ v3 libraries (10x Genomics) and sequenced on the NextSeq 500 platform (Illumina). Initial preprocessing of scRNA-seq datasets began with demultiplexing, read QC, mapping (STAR aligner with the Drosophila melanogaster genome), and quantification of barcodes and unique molecular identifiers using the Cell Ranger (5.0) software. The wild-type dataset resulted in a total of 4081 cells, and the AlkY1355S dataset resulted in a total of 4222 cells. Downstream analysis including cell preprocessing, count normalization, feature selection, integration, clustering, dimensionality reduction/projection, trajectory inference and differential expression testing was performed using the R-based pipeline Seurat (4.0.3) (Stuart et al., 2019) and the Python-based pipeline Scanpy (Wolf et al., 2018). Cell quality was assessed by the proportion of cells with unique feature counts between 200 and 5000 (removing poor-quality cells), and transcripts between 500 and 50,000 (removing low-quality transcripts) and with less than 20% mitochondrial genes. After preprocessing, 3967 and 4099 cells remained, with a total of 10,411 and 10,316 RNA features for the control and AlkY1355S datasets, respectively. Normalization of scRNA-seq datasets and integration analysis were performed with the SCTransform approach (Hafemeister and Satija, 2019). The top 3000 highly variable genes were selected to determine the true dimensionality of the dataset. Principal component analysis was used for clustering, and, based on elbow plots, the numbers of principal components were determined. Non-linear dimensional reduction using UMAP, and neighborhood identification with k=30 was used. Clusters were identified using the Louvain algorithm (Blondel et al., 2008), with 0.56 resolution. To determine clusters, marker genes of each cluster were identified by FindAllMarkers function (Seurat) and logistic regression analysis (Scanpy). Based on canonical markers and gene expression profiles, genes in similar clusters were merged and annotated based on existing knowledge and literature (Ariss et al., 2020; Brunet Avalos et al., 2019; Cattenoz et al., 2016; Estacio-Gómez et al., 2020; Michki et al., 2021).

To identify differentially expressed genes, expression profiles of each cluster across AlkY1355S and wild-type conditions were compared by the MAST approach (Finak et al., 2015), with a threshold of log fold change of average expression between the two groups ≥0.2 and P-value≤0.05. Cluster correlation was performed using Pearson correlation algorithms (Scanpy), and relationship between clusters was determined by unrooted-phylogenetic tree (Seurat). GO analysis was performed using the R package EnrichR (employing FlyEnrichr as reference) (Kuleshov et al., 2016) on cluster 1 marker genes identified from NB-enriched cells (AlkY1355S).

RNA-seq and analysis

L3 larvae (n=45) were collected and quickly washed in water to remove food and yeast. Larvae were placed in ice-cold PBS in a depression well. Larval CNSs were fine dissected and transferred into microcentrifuge tubes containing 250 μl of an ice-cold 1:1 solution of RNAse-free PBS and RNALater (Invitrogen, AM7020). CNS samples were centrifuged at 382 g for 5 min at 4°C, and the supernatant was removed. Then, 500 μl fresh RNALater solution was added, and samples were stored at −80°C. Once CNS collection for three biological replicates per genotype had been performed, RNA extraction was carried out according to the manufacturer's protocol (Promega ReliaPrepTM RNA Tissue Miniprep System, Z6111). RNA concentration was measured using NanoDrop OneC (Thermo Fisher Scientific), and RNA integrity was checked by agarose gel electrophoresis. Four to six micrograms of total RNA per biological replicate were sequenced (Novogene Co. Ltd). Samples were assessed for quality with an Agilent 2100 Bioanalyzer system, and paired-end sequencing was performed on an Illumina platform. Over 40 million reads/genotype were generated and mapped to the genome at a rate of 94-96%. Drosophila melanogaster (ensemble bdgp6_gca_000001215_4 genome assembly) was used. HISAT2 algorithm for alignment and DESeq2 R package (Anders and Huber, 2010) for differential gene expression were used.

Alk domain sequence and structure analysis

Pairwise alignment of Drosophila melanogaster Alk and human ALK was performed using the EMBOSS Needleman-Wunsch algorithm (Madeira et al., 2019). The three-dimensional structure of the Drosophila Alk kinase domain was modeled based on the human ALK crystal structure dataset template (NCBI Protein Domain and Macromolecular Structures database, PDB ID 4TT7) and the annotated Drosophila Alk-PA sequence (NP_001261027.1) using PyMol 1.8.6.0 software.

Statistical analysis

Data acquirements and volume calculation was carried out with Microsoft Office Excel software. Statistical analysis was performed with GraphPad Prism version 9.3.1 software.

We thank Cheng-Yu Lee and Claude Desplan for sharing fly stocks and reagents. We acknowledge Bloomington Drosophila Stock Center (NIH P40OD018537) for fly stocks used in this study. The anti-Elav, anti-Trio, anti-Abrupt, anti-Dlg, anti-En, anti-Pros and anti-EcR-B1 antibodies developed by G. Rubin, C. Hama, S. Crews, C. Goodman (Dlg/En), C. Doe and C. Thummel, respectively, were obtained from the Developmental Studies Hybridoma Bank, created by the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health and maintained at The University of Iowa, Department of Biology, Iowa City, IA, USA. We thank Bengt Hallberg, Ezgi Uçkun, Linnea Molander and Sanjay Kumar Sukumar for critical reading of the manuscript.

Author contributions

Conceptualization: K.P., G.W., R.H.P.; Methodology: K.P., G.W., V.A., T.M., B.A., M.B., P.M.-G.; Software: M.B.; Validation: K.P., G.W., V.A., T.M., B.A., P.M.-G., R.H.P.; Formal analysis: K.P., G.W., V.A., T.M., B.A., P.M.-G., R.H.P.; Investigation: K.P., V.A., T.M., B.A., P.M.-G., R.H.P.; Resources: V.A., M.B., R.H.P.; Data curation: K.P., G.W., V.A., M.B., P.M.-G.; Writing - original draft: K.P., V.A., R.H.P.; Writing - review & editing: K.P., G.W., B.A., P.M.-G., R.H.P.; Visualization: K.P., G.W., V.A., T.M., B.A., P.M.-G.; Supervision: R.H.P.; Project administration: R.H.P.; Funding acquisition: K.P., G.W., B.A., M.B., R.H.P.

Funding

This work was supported by grants from the Swedish Cancer Foundation (RHP CAN18/0729; BA CAN200270P), Barncancerfonden (RHP 2019-0078; KP TJ2019-0071), Vetenskapsrådet (RHP 2019-03914; MB 2019-01708), Stiftelsen för Strategisk Forskning (RB13-0204), Göran Gustafssons Stiftelser (RHP2016), Åke Wiberg Stiftelse (GW M19-0561) and Knut och Alice Wallenbergs Stiftelse (KAW 2015.0144). Open Access funding provided by Göteborgs Universitet. Deposited in PMC for immediate release.

Data availability

scRNA-seq and bulk RNA-seq datasets for this study have been deposited in Gene Expression Omnibus (GEO) under accession numbers GSE198850 and GSE198812, respectively. To facilitate access to our scRNA-seq data, we employed the ShinyCell R for interactive interfaces package (Ouyang et al., 2021), available at https://ruthpalmerlab.se/flyr_larval_brain_2022.

Anders
,
S.
and
Huber
,
W.
(
2010
).
Differential expression analysis for sequence count data
.
Genome Biol.
11
,
R106
.
Ariss
,
M. M.
,
Terry
,
A. R.
,
Islam
,
A. B. M. M. K.
,
Hay
,
N.
and
Frolov
,
M. V.
(
2020
).
Amalgam regulates the receptor tyrosine kinase pathway through Sprouty in glial cell development in the Drosophila larval brain
.
J. Cell Sci.
133
,
jcs250837
.
Ashburner
,
M.
(
1989
).
Drosophila: A Laboratory Handbook and Manual
. Cold Spring Harbor Laboratory Press.
Awasaki
,
T.
,
Saito
,
M.
,
Sone
,
M.
,
Suzuki
,
E.
,
Sakai
,
R.
,
Ito
,
K.
and
Hama
,
C.
(
2000
).
The Drosophila trio plays an essential role in patterning of axons by regulating their directional extension
.
Neuron
26
,
119
-
131
.
Bai
,
L.
and
Sehgal
,
A.
(
2015
).
Anaplastic lymphoma kinase acts in the Drosophila mushroom body to negatively regulate sleep
.
PLoS Genet.
11
,
e1005611
.
Bazigou
,
E.
,
Apitz
,
H.
,
Johansson
,
J.
,
Loren
,
C. E.
,
Hirst
,
E. M. A.
,
Chen
,
P.-L.
,
Palmer
,
R. H.
and
Salecker
,
I.
(
2007
).
Anterograde Jelly belly and Alk receptor tyrosine kinase signaling mediates retinal axon targeting in Drosophila
.
Cell
128
,
961
-
975
.
Berry
,
T.
,
Luther
,
W.
,
Bhatnagar
,
N.
,
Jamin
,
Y.
,
Poon
,
E.
,
Sanda
,
T.
,
Pei
,
D.
,
Sharma
,
B.
,
Vetharoy
,
W. R.
,
Hallsworth
,
A.
et al.
(
2012
).
The ALK(F1174L) mutation potentiates the oncogenic activity of MYCN in neuroblastoma
.
Cancer Cell
22
,
117
-
130
.
Bilsland
,
J. G.
,
Wheeldon
,
A.
,
Mead
,
A.
,
Znamenskiy
,
P.
,
Almond
,
S.
,
Waters
,
K. A.
,
Thakur
,
M.
,
Beaumont
,
V.
,
Bonnert
,
T. P.
,
Heavens
,
R.
et al.
(
2008
).
Behavioral and neurochemical alterations in mice deficient in anaplastic lymphoma kinase suggest therapeutic potential for psychiatric indications
.
Neuropsychopharmacology
33
,
685
-
700
.
Blondel
,
V. D.
,
Guillaume
,
J.-L.
,
Lambiotte
,
R.
and
Lefebvre
,
E.
(
2008
).
Fast unfolding of communities in large networks
.
J. Stat. Mech. Theory Exp.
2008
,
P10008
.
Borenäs
,
M.
,
Umapathy
,
G.
,
Lai
,
W. Y.
,
Lind
,
D. E.
,
Witek
,
B.
,
Guan
,
J.
,
Mendoza-Garcia
,
P.
,
Masudi
,
T.
,
Claeys
,
A.
,
Chuang
,
T. P.
et al.
(
2021
).
ALK ligand ALKAL2 potentiates MYCN-driven neuroblastoma in the absence of ALK mutation
.
EMBO J.
40
,
e105784
.
Brunet Avalos
,
C.
,
Maier
,
G. L.
,
Bruggmann
,
R.
and
Sprecher
,
S. G.
(
2019
).
Single cell transcriptome atlas of the Drosophila larval brain
.
eLife
8
,
e50354
.
Carén
,
H.
,
Abel
,
F.
,
Kogner
,
P.
and
Martinsson
,
T.
(
2008
).
High incidence of DNA mutations and gene amplifications of the ALK gene in advanced sporadic neuroblastoma tumours
.
Biochem. J.
416
,
153
-
159
.
Cattenoz
,
P. B.
,
Popkova
,
A.
,
Southall
,
T. D.
,
Aiello
,
G.
,
Brand
,
A. H.
and
Giangrande
,
A.
(
2016
).
Functional conservation of the Glide/Gcm regulatory network controlling glia, hemocyte, and tendon cell differentiation in Drosophila
.
Genetics
202
,
191
-
219
.
Cattenoz
,
P. B.
,
Sakr
,
R.
,
Pavlidaki
,
A.
,
Delaporte
,
C.
,
Riba
,
A.
,
Molina
,
N.
,
Hariharan
,
N.
,
Mukherjee
,
T.
and
Giangrande
,
A.
(
2020
).
Temporal specificity and heterogeneity of Drosophila immune cells
.
EMBO J.
39
,
e104486
.
Cazes
,
A.
,
Lopez-Delisle
,
L.
,
Tsarovina
,
K.
,
Pierre-Eugène
,
C.
,
De Preter
,
K.
,
Peuchmaur
,
M.
,
Nicolas
,
A.
,
Provost
,
C.
,
Louis-Brennetot
,
C.
,
Daveau
,
R.
et al.
(
2014
).
Activated Alk triggers prolonged neurogenesis and Ret upregulation providing a therapeutic target in ALK-mutated neuroblastoma
.
Oncotarget
5
,
2688
-
2702
.
Chand
,
D.
,
Yamazaki
,
Y.
,
Ruuth
,
K.
,
Schönherr
,
C.
,
Martinsson
,
T.
,
Kogner
,
P.
,
Attiyeh
,
E. F.
,
Maris
,
J.
,
Morozova
,
O.
,
Marra
,
M. A.
et al.
(
2013
).
Cell culture and Drosophila model systems define three classes of anaplastic lymphoma kinase mutations in neuroblastoma
.
Dis. Model. Mech.
6
,
373
-
382
.
Chen
,
Y.
,
Takita
,
J.
,
Choi
,
Y. L.
,
Kato
,
M.
,
Ohira
,
M.
,
Sanada
,
M.
,
Wang
,
L.
,
Soda
,
M.
,
Kikuchi
,
A.
,
Igarashi
,
T.
et al.
(
2008
).
Oncogenic mutations of ALK kinase in neuroblastoma
.
Nature
455
,
971
-
974
.
Cheng
,
L. Y.
,
Bailey
,
A. P.
,
Leevers
,
S. J.
,
Ragan
,
T. J.
,
Driscoll
,
P. C.
and
Gould
,
A. P.
(
2011
).
Anaplastic lymphoma kinase spares organ growth during nutrient restriction in Drosophila
.
Cell
146
,
435
-
447
.
Cognigni
,
P.
,
Felsenberg
,
J.
and
Waddell
,
S.
(
2018
).
Do the right thing: neural network mechanisms of memory formation, expression and update in Drosophila
.
Curr. Opin. Neurobiol.
49
,
51
-
58
.
Crittenden
,
J. R.
,
Skoulakis
,
E. M. C.
,
Han
,
K.-A.
,
Kalderon
,
D.
and
Davis
,
R. L.
(
1998
).
Tripartite mushroom body architecture revealed by antigenic markers
.
Learn. Mem.
5
,
38
-
51
.
Curt
,
J. R.
,
Yaghmaeian Salmani
,
B.
and
Thor
,
S.
(
2019
).
Anterior CNS expansion driven by brain transcription factors
.
eLife
8
,
e45274
.
Doe
,
C. Q.
(
2008
).
Neural stem cells: balancing self-renewal with differentiation
.
Development
135
,
1575
-
1587
.
Dong
,
W.
,
Gao
,
Y. H.
,
Zhang
,
X. B.
,
Moussian
,
B.
and
Zhang
,
J. Z.
(
2020
).
Chitinase 10 controls chitin amounts and organization in the wing cuticle of Drosophila
.
Insect Sci
27
,
1198
-
1207
.
Englund
,
C.
,
Lorén
,
C. E.
,
Grabbe
,
C.
,
Varshney
,
G. K.
,
Deleuil
,
F.
,
Hallberg
,
B.
and
Palmer
,
R. H.
(
2003
).
Jeb signals through the Alk receptor tyrosine kinase to drive visceral muscle fusion
.
Nature
425
,
512
-
516
.
Estacio-Gómez
,
A.
,
Hassan
,
A.
,
Walmsley
,
E.
,
Le
,
L. W.
and
Southall
,
T. D.
(
2020
).
Dynamic neurotransmitter specific transcription factor expression profiles during Drosophila development
.
Biol. Open
9
,
bio052928
.
Evans
,
C. J.
,
Liu
,
T.
and
Banerjee
,
U.
(
2014
).
Drosophila hematopoiesis: Markers and methods for molecular genetic analysis
.
Methods
68
,
242
-
251
.
Fadeev
,
A.
,
Krauss
,
J.
,
Singh
,
A. P.
and
Nüsslein-Volhard
,
C.
(
2016
).
Zebrafish Leucocyte tyrosine kinase controls iridophore establishment, proliferation and survival
.
Pigment Cell Melanoma Res.
29
,
284
-
296
.
Fadeev
,
A.
,
Mendoza-Garcia
,
P.
,
Irion
,
U.
,
Guan
,
J.
,
Pfeifer
,
K.
,
Wiessner
,
S.
,
Serluca
,
F.
,
Singh
,
A. P.
,
Nusslein-Volhard
,
C.
and
Palmer
,
R. H.
(
2018
).
ALKALs are in vivo ligands for ALK family receptor tyrosine kinases in the neural crest and derived cells
.
Proc. Natl. Acad. Sci. USA
.
115
,
E630
-
E638
.
Finak
,
G.
,
McDavid
,
A.
,
Yajima
,
M.
,
Deng
,
J.
,
Gersuk
,
V.
,
Shalek
,
A. K.
,
Slichter
,
C. K.
,
Miller
,
H. W.
,
McElrath
,
M. J.
,
Prlic
,
M.
et al.
(
2015
).
MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data
.
Genome Biol.
16
,
278
.
Finley
,
K. D.
,
Edeen
,
P. T.
,
Foss
,
M.
,
Gross
,
E.
,
Ghbeish
,
N.
,
Palmer
,
R. H.
,
Taylor
,
B. J.
and
McKeown
,
M.
(
1998
).
Dissatisfaction encodes a tailless-like nuclear receptor expressed in a subset of CNS neurons controlling Drosophila sexual behavior
.
Neuron
21
,
1363
-
1374
.
Gabay
,
L.
,
Seger
,
R.
and
Shilo
,
B. Z.
(
1997
).
MAP kinase in situ activation atlas during Drosophila embryogenesis
.
Development
124
,
3535
-
3541
.
George
,
R. E.
,
Sanda
,
T.
,
Hanna
,
M.
,
Fröhling
,
S.
,
Luther
,
W.
, II
,
Zhang
,
J.
,
Ahn
,
Y.
,
Zhou
,
W.
,
London
,
W. B.
,
McGrady
,
P.
et al.
(
2008
).
Activating mutations in ALK provide a therapeutic target in neuroblastoma
.
Nature
455
,
975
-
978
.
Gouzi
,
J. Y.
,
Moressis
,
A.
,
Walker
,
J. A.
,
Apostolopoulou
,
A. A.
,
Palmer
,
R. H.
,
Bernards
,
A.
and
Skoulakis
,
E. M. C.
(
2011
).
The receptor tyrosine kinase Alk controls neurofibromin functions in Drosophila growth and learning
.
PLoS Genet.
7
,
e1002281
.
Gouzi
,
J. Y.
,
Bouraimi
,
M.
,
Roussou
,
I. G.
,
Moressis
,
A.
and
Skoulakis
,
E. M. C.
(
2018
).
The Drosophila receptor tyrosine kinase Alk constrains long-term memory formation
.
J. Neurosci.
38
,
7701
-
7712
.
Gratz
,
S. J.
,
Cummings
,
A. M.
,
Nguyen
,
J. N.
,
Hamm
,
D. C.
,
Donohue
,
L. K.
,
Harrison
,
M. M.
,
Wildonger
,
J.
and
O'Connor-Giles
,
K. M.
(
2013
).
Genome engineering of Drosophila with the CRISPR RNA-guided Cas9 nuclease
.
Genetics
194
,
1029
-
1035
.
Gratz
,
S. J.
,
Ukken
,
F. P.
,
Rubinstein
,
C. D.
,
Thiede
,
G.
,
Donohue
,
L. K.
,
Cummings
,
A. M.
and
O'Connor-Giles
,
K. M.
(
2014
).
Highly specific and efficient CRISPR/Cas9-catalyzed homology-directed repair in Drosophila
.
Genetics
196
,
961
-
971
.
Guan
,
J.
,
Umapathy
,
G.
,
Yamazaki
,
Y.
,
Wolfstetter
,
G.
,
Mendoza
,
P.
,
Pfeifer
,
K.
,
Mohammed
,
A.
,
Hugosson
,
F.
,
Zhang
,
H.
,
Hsu
,
A. W.
et al.
(
2015
).
FAM150A and FAM150B are activating ligands for anaplastic lymphoma kinase
.
eLife
4
,
e09811
.
Guan
,
J.
,
Yamazaki
,
Y.
,
Chand
,
D.
,
van Dijk
,
J. R.
,
Ruuth
,
K.
,
Palmer
,
R. H.
and
Hallberg
,
B.
(
2017
).
Novel Mechanisms of ALK Activation Revealed by Analysis of the Y1278S Neuroblastoma Mutation
.
Cancers
9
,
149
.
Hafemeister
,
C.
and
Satija
,
R.
(
2019
).
Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression
.
Genome Biol.
20
,
296
.
Hallberg
,
B.
and
Palmer
,
R. H.
(
2013
).
Mechanistic insight into ALK receptor tyrosine kinase in human cancer biology
.
Nat. Rev. Cancer
13
,
685
-
700
.
Homem
,
C. C. F.
,
Steinmann
,
V.
,
Burkard
,
T. R.
,
Jais
,
A.
,
Esterbauer
,
H.
and
Knoblich
,
J. A.
(
2014
).
Ecdysone and mediator change energy metabolism to terminate proliferation in Drosophila neural stem cells
.
Cell
158
,
874
-
888
.
Hu
,
S.
,
Fambrough
,
D.
,
Atashi
,
J. R.
,
Goodman
,
C. S.
and
Crews
,
S. T.
(
1995
).
The Drosophila abrupt gene encodes a BTB-zinc finger regulatory protein that controls the specificity of neuromuscular connections
.
Genes Dev.
9
,
2936
-
2948
.
Ito
,
K.
and
Hotta
,
Y.
(
1992
).
Proliferation pattern of postembryonic neuroblasts in the brain of Drosophila melanogaster
.
Dev. Biol.
149
,
134
-
148
.
Ito
,
K.
,
Awano
,
W.
,
Suzuki
,
K.
,
Hiromi
,
Y.
and
Yamamoto
,
D.
(
1997
).
The Drosophila mushroom body is a quadruple structure of clonal units each of which contains a virtually identical set of neurones and glial cells
.
Development
124
,
761
-
771
.
Iwahara
,
T.
,
Fujimoto
,
J.
,
Wen
,
D.
,
Cupples
,
R.
,
Bucay
,
N.
,
Arakawa
,
T.
,
Mori
,
S.
,
Ratzkin
,
B.
and
Yamamoto
,
T.
(
1997
).
Molecular characterization of ALK, a receptor tyrosine kinase expressed specifically in the nervous system
.
Oncogene
14
,
439
-
449
.
Janoueix-Lerosey
,
I.
,
Lequin
,
D.
,
Brugières
,
L.
,
Ribeiro
,
A.
,
de Pontual
,
L.
,
Combaret
,
V.
,
Raynal
,
V.
,
Puisieux
,
A.
,
Schleiermacher
,
G.
,
Pierron
,
G.
et al.
(
2008
).
Somatic and germline activating mutations of the ALK kinase receptor in neuroblastoma
.
Nature
455
,
967
-
970
.
Knoblich
,
J. A.
(
2008
).
Mechanisms of asymmetric stem cell division
.
Cell
132
,
583
-
597
.
Kuleshov
,
M. V.
,
Jones
,
M. R.
,
Rouillard
,
A. D.
,
Fernandez
,
N. F.
,
Duan
,
Q.
,
Wang
,
Z.
,
Koplev
,
S.
,
Jenkins
,
S. L.
,
Jagodnik
,
K. M.
,
Lachmann
,
A.
et al.
(
2016
).
Enrichr: a comprehensive gene set enrichment analysis web server 2016 update
.
Nucleic Acids Res.
44
,
W90
-
W97
.
Kumar
,
A.
,
Bello
,
B.
and
Reichert
,
H.
(
2009
).
Lineage-specific cell death in postembryonic brain development of Drosophila
.
Development
136
,
3433
-
3442
.
Kumar
,
S.
,
Tunc
,
I.
,
Tansey
,
T. R.
,
Pirooznia
,
M.
and
Harbison
,
S. T.
(
2021
).
Identification of genes contributing to a long circadian period in Drosophila melanogaster
.
J. Biol. Rhythms
36
,
239
-
253
.
Lasek
,
A. W.
,
Lim
,
J.
,
Kliethermes
,
C. L.
,
Berger
,
K. H.
,
Joslyn
,
G.
,
Brush
,
G.
,
Xue
,
L.
,
Robertson
,
M.
,
Moore
,
M. S.
,
Vranizan
,
K.
et al.
(
2011
).
An evolutionary conserved role for anaplastic lymphoma kinase in behavioral responses to ethanol
.
PLoS ONE
6
,
e22636
.
Lécuyer
,
E.
,
Parthasarathy
,
N.
and
Krause
,
H. M.
(
2008
).
Fluorescent in situ hybridization protocols in Drosophila embryos and tissues
.
Methods Mol. Biol.
420
,
289
-
302
.
Lee
,
T.
,
Lee
,
A.
and
Luo
,
L.
(
1999
).
Development of the Drosophila mushroom bodies: sequential generation of three distinct types of neurons from a neuroblast
.
Development
126
,
4065
-
4076
.
Lee
,
T.
,
Marticke
,
S.
,
Sung
,
C.
,
Robinow
,
S.
and
Luo
,
L.
(
2000
).
Cell-autonomous requirement of the USP/EcR-B ecdysone receptor for mushroom body neuronal remodeling in Drosophila
.
Neuron
28
,
807
-
818
.
Lee
,
H.-H.
,
Norris
,
A.
,
Weiss
,
J. B.
and
Frasch
,
M.
(
2003
).
Jelly belly protein activates the receptor tyrosine kinase Alk to specify visceral muscle pioneers
.
Nature
425
,
507
-
512
.
Liu
,
Z.
,
Yang
,
C.-P.
,
Sugino
,
K.
,
Fu
,
C.-C.
,
Liu
,
L.-Y.
,
Yao
,
X.
,
Lee
,
L. P.
and
Lee
,
T.
(
2015
).
Opposing intrinsic temporal gradients guide neural stem cell production of varied neuronal fates
.
Science
350
,
317
-
320
.
Liu
,
L.-Y.
,
Long
,
X.
,
Yang
,
C.-P.
,
Miyares
,
R. L.
,
Sugino
,
K.
,
Singer
,
R. H.
and
Lee
,
T.
(
2019
).
Mamo decodes hierarchical temporal gradients into terminal neuronal fate
.
eLife
8
,
e48056
.
Lorén
,
C. E.
,
Scully
,
A.
,
Grabbe
,
C.
,
Edeen
,
P. T.
,
Thomas
,
J.
,
McKeown
,
M.
,
Hunter
,
T.
and
Palmer
,
R. H.
(
2001
).
Identification and characterization of DAlk: a novel Drosophila melanogaster RTK which drives ERK activation in vivo
.
Genes Cells
6
,
531
-
544
.
Lorén
,
C. E.
,
Englund
,
C.
,
Grabbe
,
C.
,
Hallberg
,
B.
,
Hunter
,
T.
and
Palmer
,
R. H.
(
2003
).
A crucial role for the Anaplastic lymphoma kinase receptor tyrosine kinase in gut development in Drosophila melanogaster
.
EMBO Rep.
4
,
781
-
786
.
Madeira
,
F.
,
Park
,
Y. M.
,
Lee
,
J.
,
Buso
,
N.
,
Gur
,
T.
,
Madhusoodanan
,
N.
,
Basutkar
,
P.
,
Tivey
,
A. R. N.
,
Potter
,
S. C.
,
Finn
,
R. D.
et al.
(
2019
).
The EMBL-EBI search and sequence analysis tools APIs in 2019
.
Nucleic Acids Res.
47
,
W636
-
W641
.
Maris
,
J. M.
(
2010
).
Recent advances in neuroblastoma
.
N. Engl. J. Med.
362
,
2202
-
2211
.
Marshall
,
G. M.
,
Carter
,
D. R.
,
Cheung
,
B. B.
,
Liu
,
T.
,
Mateos
,
M. K.
,
Meyerowitz
,
J. G.
and
Weiss
,
W. A.
(
2014
).
The prenatal origins of cancer
.
Nat. Rev. Cancer
14
,
277
-
289
.
Martinsson
,
T.
,
Eriksson
,
T.
,
Abrahamsson
,
J.
,
Caren
,
H.
,
Hansson
,
M.
,
Kogner
,
P.
,
Kamaraj
,
S.
,
Schönherr
,
C.
,
Weinmar
,
J.
,
Ruuth
,
K.
et al.
(
2011
).
Appearance of the novel activating F1174S ALK mutation in neuroblastoma correlates with aggressive tumor progression and unresponsiveness to therapy
.
Cancer Res.
71
,
98
-
105
.
Mendoza-García
,
P.
,
Hugosson
,
F.
,
Fallah
,
M.
,
Higgins
,
M. L.
,
Iwasaki
,
Y.
,
Pfeifer
,
K.
,
Wolfstetter
,
G.
,
Varshney
,
G.
,
Popichenko
,
D.
,
Gergen
,
J. P.
et al.
(
2017
).
The Zic family homologue Odd-paired regulates Alk expression in Drosophila
.
PLoS Genet.
13
,
e1006617
.
Michki
,
N. S.
,
Li
,
Y.
,
Sanjasaz
,
K.
,
Zhao
,
Y.
,
Shen
,
F. Y.
,
Walker
,
L. A.
,
Cao
,
W.
,
Lee
,
C.-Y.
and
Cai
,
D.
(
2021
).
The molecular landscape of neural differentiation in the developing Drosophila brain revealed by targeted scRNA-seq and multi-informatic analysis
.
Cell Rep.
35
,
109039
.
Mo
,
E. S.
,
Cheng
,
Q.
,
Reshetnyak
,
A. V.
,
Schlessinger
,
J.
and
Nicoli
,
S.
(
2017
).
Alk and Ltk ligands are essential for iridophore development in zebrafish mediated by the receptor tyrosine kinase Ltk
.
Proc. Natl. Acad. Sci. USA
114
,
12027
-
12032
.
Mossé
,
Y. P.
,
Laudenslager
,
M.
,
Longo
,
L.
,
Cole
,
K. A.
,
Wood
,
A.
,
Attiyeh
,
E. F.
,
Laquaglia
,
M. J.
,
Sennett
,
R.
,
Lynch
,
J. E.
,
Perri
,
P.
et al.
(
2008
).
Identification of ALK as a major familial neuroblastoma predisposition gene
.
Nature
455
,
930
-
935
.
Muller
,
H.-A. J.
(
2008
).
Immunolabeling of embryos
.
Methods Mol. Biol.
420
,
207
-
218
.
Okamoto
,
N.
and
Nishimura
,
T.
(
2015
).
Signaling from Glia and cholinergic neurons controls nutrient-dependent production of an insulin-like peptide for Drosophila body growth
.
Dev. Cell
35
,
295
-
310
.
Ono
,
S.
,
Saito
,
T.
,
Terui
,
K.
,
Yoshida
,
H.
and
Enomoto
,
H.
(
2019
).
Generation of conditional ALK F1174L mutant mouse models for the study of neuroblastoma pathogenesis
.
Genesis
57
,
e23323
.
Orthofer
,
M.
,
Valsesia
,
A.
,
Mägi
,
R.
,
Wang
,
Q.-P.
,
Kaczanowska
,
J.
,
Kozieradzki
,
I.
,
Leopoldi
,
A.
,
Cikes
,
D.
,
Zopf
,
L. M.
,
Tretiakov
,
E. O.
et al.
(
2020
).
Identification of ALK in Thinness
.
Cell
181
,
1246
-
1262.e22
.
Ouyang
,
J. F.
,
Kamaraj
,
U. S.
,
Cao
,
E. Y.
and
Rackham
,
O. J. L.
(
2021
).
ShinyCell: Simple and sharable visualisation of single-cell gene expression data
.
Bioinformatics
28
,
btab209
.
Öztürk-Çolak
,
A.
,
Moussian
,
B.
,
Araújo
,
S. J.
and
Casanova
,
J.
(
2016
).
A feedback mechanism converts individual cell features into a supracellular ECM structure in Drosophila trachea
.
Elife
5
,
e09373
.
Pecot
,
M. Y.
,
Chen
,
Y.
,
Akin
,
O.
,
Chen
,
Z.
,
Tsui
,
C. Y. K.
and
Zipursky
,
S. L.
(
2014
).
Sequential axon-derived signals couple target survival and layer specificity in the Drosophila visual system
.
Neuron
82
,
320
-
333
.
Pfeifer
,
K.
,
Schaub
,
C.
,
Domsch
,
K.
,
Dorresteijn
,
A.
and
Wolfstetter
,
G.
(
2014
).
Maternal inheritance of twist and analysis of MAPK activation in embryos of the polychaete annelid Platynereis dumerilii
.
PLoS ONE
9
,
e96702
.
Pinto-Teixeira
,
F.
,
Konstantinides
,
N.
and
Desplan
,
C.
(
2016
).
Programmed cell death acts at different stages of Drosophila neurodevelopment to shape the central nervous system
.
FEBS Lett.
590
,
2435
-
2453
.
Ren
,
Q.
,
Yang
,
C.-P.
,
Liu
,
Z.
,
Sugino
,
K.
,
Mok
,
K.
,
He
,
Y.
,
Ito
,
M.
,
Nern
,
A.
,
Otsuna
,
H.
and
Lee
,
T.
(
2017
).
Stem cell-intrinsic, seven-up-triggered temporal factor gradients diversify intermediate neural progenitors
.
Curr. Biol.
27
,
1303
-
1313
.
Reshetnyak
,
A. V.
,
Murray
,
P. B.
,
Shi
,
X.
,
Mo
,
E. S.
,
Mohanty
,
J.
,
Tome
,
F.
,
Bai
,
H.
,
Gunel
,
M.
,
Lax
,
I.
and
Schlessinger
,
J.
(
2015
).
Augmentor alpha and beta (FAM150) are ligands of the receptor tyrosine kinases ALK and LTK: hierarchy and specificity of ligand-receptor interactions
.
Proc. Natl. Acad. Sci. USA
112
,
15862
-
15867
.
Rohrbough
,
J.
and
Broadie
,
K.
(
2010
).
Anterograde Jelly belly ligand to Alk receptor signaling at developing synapses is regulated by Mind the gap
.
Development
137
,
3523
-
3533
.
Rohrbough
,
J.
,
Kent
,
K. S.
,
Broadie
,
K.
and
Weiss
,
J. B.
(
2013
).
Jelly Belly trans-synaptic signaling to anaplastic lymphoma kinase regulates neurotransmission strength and synapse architecture
.
Dev. Neurobiol.
73
,
189
-
208
.
Rossi
,
A. M.
and
Desplan
,
C.
(
2020
).
Extrinsic activin signaling cooperates with an intrinsic temporal program to increase mushroom body neuronal diversity
.
eLife
9
,
e58880
.
Stuart
,
T.
,
Butler
,
A.
,
Hoffman
,
P.
,
Hafemeister
,
C.
,
Papalexi
,
E.
,
Mauck
,
W. M.
, III
,
Hao
,
Y.
,
Stoeckius
,
M.
,
Smibert
,
P.
and
Satija
,
R.
(
2019
).
Comprehensive integration of single-cell data
.
Cell
177
,
1888
-
1902.e21
.
Stute
,
C.
,
Schimmelpfeng
,
K.
,
Renkawitz-Pohl
,
R.
,
Palmer
,
R. H.
and
Holz
,
A.
(
2004
).
Myoblast determination in the somatic and visceral mesoderm depends on Notch signalling as well as on milliways(mili(Alk)) as receptor for Jeb signalling
.
Development
131
,
743
-
754
.
Syed
,
M. H.
,
Mark
,
B.
and
Doe
,
C. Q.
(
2017a
).
Playing well with others: extrinsic cues regulate neural progenitor temporal identity to generate neuronal diversity
.
Trends Genet.
33
,
933
-
942
.
Syed
,
M. H.
,
Mark
,
B.
and
Doe
,
C. Q.
(
2017b
).
Steroid hormone induction of temporal gene expression in Drosophila brain neuroblasts generates neuronal and glial diversity
.
eLife
6
,
e26287
.
Tolbert
,
V. P.
and
Matthay
,
K. K.
(
2018
).
Neuroblastoma: clinical and biological approach to risk stratification and treatment
.
Cell Tissue Res.
372
,
195
-
209
.
Tomolonis
,
J. A.
,
Agarwal
,
S.
and
Shohet
,
J. M.
(
2018
).
Neuroblastoma pathogenesis: deregulation of embryonic neural crest development
.
Cell Tissue Res.
372
,
245
-
262
.
Trigg
,
R. M.
and
Turner
,
S. D.
(
2018
).
ALK in neuroblastoma: biological and therapeutic implications
.
Cancers
10
,
113
.
Umapathy
,
G.
,
Mendoza-Garcia
,
P.
,
Hallberg
,
B.
and
Palmer
,
R. H.
(
2019
).
Targeting anaplastic lymphoma kinase in neuroblastoma
.
APMIS
127
,
288
-
302
.
Urbach
,
R.
and
Technau
,
G. M.
(
2004
).
Neuroblast formation and patterning during early brain development in Drosophila
.
BioEssays
26
,
739
-
751
.
Varshney
,
G. K.
and
Palmer
,
R. H.
(
2006
).
The bHLH transcription factor Hand is regulated by Alk in the Drosophila embryonic gut
.
Biochem. Biophys. Res. Commun.
351
,
839
-
846
.
Vasseur
,
A.
,
Cabel
,
L.
,
Geiss
,
R.
,
Schleiermacher
,
G.
,
Pierron
,
G.
,
Kamal
,
M.
,
Jehanno
,
N.
,
Bataillon
,
G.
,
Guinebretiere
,
J.-M.
and
Bozec
,
L.
(
2019
).
Efficacy of lorlatinib in primary crizotinib-resistant adult neuroblastoma harboring ALK Y1278S mutation
.
JCO Precis Oncol.
3
,
1
-
5
.
Vernersson
,
E.
,
Khoo
,
N. K. S.
,
Henriksson
,
M. L.
,
Roos
,
G.
,
Palmer
,
R. H.
and
Hallberg
,
B.
(
2006
).
Characterization of the expression of the ALK receptor tyrosine kinase in mice
.
Gene Expr. Patterns
6
,
448
-
461
.
Vieceli
,
F. M.
and
Bronner
,
M. E.
(
2018
).
Leukocyte receptor tyrosine kinase interacts with secreted midkine to promote survival of migrating neural crest cells
.
Development
145
,
dev164046
.
Weiss
,
J. B.
,
Suyama
,
K. L.
,
Lee
,
H.-H.
and
Scott
,
M. P.
(
2001
).
Jelly belly: a Drosophila LDL receptor repeat-containing signal required for mesoderm migration and differentiation
.
Cell
107
,
387
-
398
.
Weiss
,
J. B.
,
Xue
,
C.
,
Benice
,
T.
,
Xue
,
L.
,
Morris
,
S. W.
and
Raber
,
J.
(
2012
).
Anaplastic lymphoma kinase and leukocyte tyrosine kinase: functions and genetic interactions in learning, memory and adult neurogenesis
.
Pharmacol. Biochem. Behav.
100
,
566
-
574
.
Weiss
,
J. B.
,
Weber
,
S.
,
Marzulla
,
T.
and
Raber
,
J.
(
2017
).
Pharmacological inhibition of Anaplastic Lymphoma Kinase rescues spatial memory impairments in Neurofibromatosis 1 mutant mice
.
Behav. Brain Res.
332
,
337
-
342
.
Weng
,
M.
,
Golden
,
K. L.
and
Lee
,
C.-Y.
(
2010
).
dFezf/Earmuff maintains the restricted developmental potential of intermediate neural progenitors in Drosophila
.
Dev. Cell
18
,
126
-
135
.
Witek
,
B.
,
El Wakil
,
A.
,
Nord
,
C.
,
Ahlgren
,
U.
,
Eriksson
,
M.
,
Vernersson-Lindahl
,
E.
,
Helland
,
A.
,
Alexeyev
,
O. A.
,
Hallberg
,
B.
and
Palmer
,
R. H.
(
2015
).
Targeted disruption of ALK reveals a potential role in hypogonadotropic hypogonadism
.
PLoS ONE
10
,
e0123542
.
Wolf
,
F. A.
,
Angerer
,
P.
and
Theis
,
F. J.
(
2018
).
SCANPY: large-scale single-cell gene expression data analysis
.
Genome Biol.
19
,
15
.
Wolfstetter
,
G.
,
Pfeifer
,
K.
,
van Dijk
,
J. R.
,
Hugosson
,
F.
,
Lu
,
X.
and
Palmer
,
R. H.
(
2017
).
The scaffolding protein Cnk binds to the receptor tyrosine kinase Alk to promote visceral founder cell specification in Drosophila
.
Sci. Signal.
10
,
eaan0804
.
Wolfstetter
,
G.
,
Pfeifer
,
K.
,
Backman
,
M.
,
Masudi
,
T. A.
,
Mendoza-García
,
P.
,
Chen
,
S.
,
Sonnenberg
,
H.
,
Sukumar
,
S. K.
,
Uçkun
,
E.
,
Varshney
,
G. K.
et al.
(
2020
).
Identification of the Wallenda JNKKK as an Alk suppressor reveals increased competitiveness of Alk-expressing cells
.
Sci. Rep.
10
,
14954
.
Woodling
,
N. S.
,
Aleyakpo
,
B.
,
Dyson
,
M. C.
,
Minkley
,
L. J.
,
Rajasingam
,
A.
,
Dobson
,
A. J.
,
Leung
,
K. H. C.
,
Pomposova
,
S.
,
Fuentealba
,
M.
,
Alic
,
N.
et al.
(
2020
).
The neuronal receptor tyrosine kinase Alk is a target for longevity
.
Aging Cell
19
,
e13137
.
Yang
,
C.-P.
,
Fu
,
C.-C.
,
Sugino
,
K.
,
Liu
,
Z.
,
Ren
,
Q.
,
Liu
,
L.-Y.
,
Yao
,
X.
,
Lee
,
L. P.
and
Lee
,
T.
(
2016
).
Transcriptomes of lineage-specific Drosophila neuroblasts profiled by genetic targeting and robotic sorting
.
Development
143
,
411
-
421
.
Yang
,
C.-P.
,
Samuels
,
T. J.
,
Huang
,
Y.
,
Yang
,
L.
,
Ish-Horowicz
,
D.
,
Davis
,
I.
and
Lee
,
T.
(
2017
).
Imp and Syp RNA-binding proteins govern decommissioning of Drosophila neural stem cells
.
Development
144
,
3454
-
3464
.
Yao
,
S.
,
Cheng
,
M.
,
Zhang
,
Q.
,
Wasik
,
M.
,
Kelsh
,
R.
and
Winkler
,
C.
(
2013
).
Anaplastic lymphoma kinase is required for neurogenesis in the developing central nervous system of zebrafish
.
PLoS ONE
8
,
e63757
.
Yu
,
F.
,
Kuo
,
C. T.
and
Jan
,
Y. N.
(
2006
).
Drosophila neuroblast asymmetric cell division: recent advances and implications for stem cell biology
.
Neuron
51
,
13
-
20
.
Zhu
,
S.
,
Lee
,
J.-S.
,
Guo
,
F.
,
Shin
,
J.
,
Perez-Atayde
,
A. R.
,
Kutok
,
J. L.
,
Rodig
,
S. J.
,
Neuberg
,
D. S.
,
Helman
,
D.
,
Feng
,
H.
et al.
(
2012
).
Activated ALK collaborates with MYCN in neuroblastoma pathogenesis
.
Cancer Cell
21
,
362
-
373
.
Zielke
,
N.
,
Korzelius
,
J.
,
van Straaten
,
M.
,
Bender
,
K.
,
Schuhknecht
,
G. F. P.
,
Dutta
,
D.
,
Xiang
,
J.
and
Edgar
,
B. A.
(
2014
).
Fly-FUCCI: a versatile tool for studying cell proliferation in complex tissues
.
Cell Rep
7
,
588
-
598
.

Competing interests

The authors declare no competing or financial interests.

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.

Supplementary information