Abstract
The Drosophila RNA-binding protein (RBP) Nab2 acts in neurons to regulate neurodevelopment and is orthologous to the human intellectual disability-linked RBP, ZC3H14. Nab2 governs axon projection in mushroom body neurons and limits dendritic arborization of class IV sensory neurons in part by regulating splicing events in ∼150 mRNAs. Analysis of the Sex-lethal (Sxl) mRNA revealed that Nab2 promotes an exon-skipping event and regulates m6A methylation on Sxl pre-mRNA by the Mettl3 methyltransferase. Mettl3 heterozygosity broadly rescues Nab2null phenotypes implying that Nab2 acts through similar mechanisms on other RNAs, including unidentified targets involved in neurodevelopment. Here, we show that Nab2 and Mettl3 regulate the removal of a 5′UTR (untranslated region) intron in the trio pre-mRNA. Trio utilizes two GEF domains to balance Rac and RhoGTPase activity. Intriguingly, an isoform of Trio containing only the RhoGEF domain, GEF2, is depleted in Nab2null nervous tissue. Expression of Trio-GEF2 rescues projection defects in Nab2null axons and dendrites, while the GEF1 Rac1-regulatory domain exacerbates these defects, suggesting Nab2-mediated regulation Trio-GEF activities. Collectively, these data indicate that Nab2-regulated processing of trio is critical for balancing Trio-GEF1 and -GEF2 activity and show that Nab2, Mettl3, and Trio function in a common pathway that shapes axon and dendrite morphology.
Drosophila Nab2, orthologue of the human RBP ZC3H14 lost in inherited intellectual disability, acts through unknown RNA targets to control axon and dendrite morphology.
This study shows that Nab2 and the Mettl3 methyltransferase guide splicing of trio mRNA, which encodes a conserved GEF-domain protein. Loss of Nab2 is associated with an imbalance in levels of Trio GEF domains in Nab2-deficient neurons. Restoring this balance partially rescues neuronal defects.
These findings suggest that Nab2 control of Trio levels is required to pattern axon and dendrite growth and suggests that ZC3H14 may play a similar role in the vertebrate brain.
INTRODUCTION
RNA-binding proteins (RBPs) associate with nascent RNA transcripts and govern expression via a multitude of mechanisms, including regulation of splicing, polyadenylation, nuclear export, translation, and stability (Maniatis and Reed, 2002; McKee and Silver, 2007; Corbett, 2018; Corley et al., 2020). These RNA–RBP interactions are particularly important in highly specialized cells such as neurons which require fine-tuned spatiotemporal control of gene expression to ensure proper development and function of the nervous system (Bardoni et al., 2012; Santoro et al., 2012; Conlon and Manley, 2017; Thelen and Kye, 2019; Gebauer et al., 2021). The importance of RBP function in neurons is highlighted by the prevalence of neurodevelopmental diseases that have been linked to defects in RBPs, leading to aberrant processing of RNAs encoding neurodevelopment factors (Cooper et al., 2009; Pak et al., 2011; Bardoni et al., 2012; Darnell and Richter, 2012; Gross et al., 2012; Santoro et al., 2012; Edens et al., 2015; Agrawal et al., 2019). Intriguingly, many of these RBPs are ubiquitously expressed and have roles in relatively common RNA processing mechanisms (Barbe et al., 1996; Franke et al., 1996; Brais, 2003; Lage et al., 2008; Kolb and Kissel, 2011; Pirozzi et al., 2011). Therefore, defining roles for these RBPs in neurons has become key to understanding why they are linked to neurological disease.
One important family of posttranscriptional regulatory proteins consists of polyadenosine binding proteins (Pabs) (Kelly et al., 2010). Conventional Pab family members bind polyadenosine RNA via RNA recognition motifs and modulate a multitude of RNA processing events such as splicing, export, polyadenylation, translation, and stability (Banerjee et al., 2013; Goss and Kleiman, 2013; Wigington et al., 2014). Another less well-studied group of Pabs utilize zinc finger (ZnF) domains to bind specific RNA motifs and modulate downstream processing events (Kelly et al., 2007; Kelly et al., 2010; Kelly et al., 2014). One such ZnF Pab termed zinc finger Cys-Cys-Cys-His-type containing 14 (ZC3H14; also termed MSUT2) is expressed ubiquitously and binds tracts of polyadenosine RNA with high affinity via tandem ZnF domains (Kelly et al., 2007; Leung et al., 2009; Kelly et al., 2010; Kelly et al., 2014; Wheeler et al., 2019). Despite ubiquitous expression, mutations in human ZC3H14 cause a form of inherited nonsyndromic autosomal recessive intellectual disability, which implies a specific requirement for ZC3H14 in the developing brain (Pak et al., 2011; Kelly et al., 2012).
The ZC3H14 protein is evolutionarily conserved among eukaryotes and has been studied in Mus musculus (Zc3h14) (Guthrie et al., 2011; Pak et al., 2011; Soucek et al., 2016), Caenorhabditis elegans (sut-2) (Guthrie et al., 2009), Sacchromyces cerevisiae (Nab2) (Anderson et al., 1993; Green et al., 2002; Hector et al., 2002; Marfatia et al., 2003; Kelly et al., 2007; Kelly et al., 2010; Schmid et al., 2015; Soucek et al., 2016), Sacchromyces pombe (Nab2) (Grenier St-Sauveur et al., 2013), and Drosophila melanogaster (Nab2) (Pak et al., 2011; Kelly et al., 2016; Fasken et al., 2019; Lee et al., 2020; Corgiat et al., 2021; Corgiat et al., 2022; Rounds et al., 2022; Jalloh et al., 2023). These studies have collectively uncovered molecular and neuronal functions for this conserved ZnF Pab. For example, Zc3h14 loss impairs working memory in mice where ZC3H14 protein localizes to synaptosomes in hippocampal neurons and regulates the abundance of synaptic proteins, including CaMK2α (Rha et al., 2017; Jones et al., 2020). Moreover, studies in C. elegans identified SUT-2 as a modulator of Tau-induced toxicity, as loss of sut-2 robustly rescues the toxic consequences of Tau overexpression in worms (Guthrie et al., 2009; Currey et al., 2023), a function of ZC3H14/MSUT2 that extends to mice (McMillan et al., 2021). On the other hand, yeast NAB2 is essential for viability (Anderson et al., 1993) and has critical functions in regulating transcription termination (Alpert et al., 2020), nuclear export (Hector et al., 2002), and transcript stability (Batisse et al., 2009; Schmid et al., 2015; Fasken et al., 2019; Alpert et al., 2020). Moreover, NAB2/Nab2 loss leads to increases in bulk poly(A) tail length in yeast, mice, and flies supporting a conserved function for Nab2 in restricting poly(A) tail length (Green et al., 2002; Hector et al., 2002; Kelly et al., 2010; Kelly et al., 2014). Taken together, these findings suggest that ZC3H14/Nab2 is involved in multiple aspects of posttranscriptional RNA metabolism and that these roles may be particularly significant in neurons.
D. melanogaster is a genetically tractable system in which to define molecular and developmental roles of the ZC3H14 invertebrate homologue, Nab2 (Pak et al., 2011). Our prior studies have determined that Nab2 function is necessary in neurons, as pan-neuronal expression of Drosophila Nab2 or human ZC3H14 is sufficient to rescue viability and locomotor defects associated with zygotic loss of Nab2 (Pak et al., 2011; Kelly et al., 2014). Moreover, Nab2 has a cell-autonomous role in Kenyon cells to pattern axonal projections from these cells into the mushroom bodies (Kelly et al., 2016), a twin neuropil structure that regulates Drosophila associative olfactory learning and memory (Heisenberg, 2003; Kahsai and Zars, 2011; Yagi et al., 2016; Takemura et al., 2017). Biochemical studies show that Nab2 interacts with Fmr1, the fly homolog of Fragile X Syndrome RBP, FMRP (Wan et al., 2000), and that these two RBPs coregulate mushroom body morphology and olfactory memory through a mechanism likely to involve translational repression of shared Nab2-Fmr1 target RNAs (Bienkowski et al., 2017). Beyond the brain, Nab2 limits dendritic branching of class IV dorsal dendritic arborization (ddaC) sensory neurons through a mechanism involving the planar cell polarity (PCP) pathway (Corgiat et al., 2022), suggesting that Nab2 controls RNA targets encoding regulators of the actin cytoskeleton. Our recent work studying the effect of Nab2 loss on the brain transcriptome revealed that Nab2 is required for proper splicing of ∼150 mRNAs (Jalloh and Lancaster et al., 2023). Furthermore, Nab2 limits methylation at the N-6 position of adenosine (m6A) on key mRNAs, including the alternatively spliced Sex-lethal (Sxl) transcript (Jalloh and Lancaster et al., 2023). However, Nab2-regulated transcripts encoding factors that guide axon and dendrite morphology have not been identified.
A recent study uncovered multiple Nab2-regulated candidate transcripts with key functions in neurodevelopment (Jalloh and Lancaster et al., 2023). Specifically, this work revealed significant retention of a 5′UTR intron in the trio transcript. Trio is a member of the Dbl homology (DH) family of guanine nucleotide exchange factor (GEF) proteins with well-conserved orthologues in C. elegans and mammals that control F-actin polymerization through the Rac and Rho small GTPases (Bellanger et al., 1998b; Awasaki et al., 2000; Bateman et al., 2000; Newsome et al., 2000; Bateman and Van Vactor, 2001; Briancon-Marjollet et al., 2008; Ba et al., 2016; Pengelly et al., 2016; Katrancha et al., 2017; Backer et al., 2018; Bircher and Koleske, 2021). As a result of these roles, Trio loss affects axon guidance and dendritic branching as well as synaptic transmission and plasticity (Briancon-Marjollet et al., 2008; Iyer et al., 2012; DeGeer et al., 2015; Ba et al., 2016; Katrancha et al., 2019). Notably, Drosophila Trio is enriched in the brain mushroom bodies where it controls axon projection and Trio also regulates arborization of sensory ddaC neurons in the larval peripheral nervous system (PNS) (Awasaki et al., 2000; Iyer et al., 2012; Shivalkar and Giniger, 2012). Moreover, several recent studies have identified loss- and gain-of-function mutations in the human TRIO gene and its paralogue KALRN that lead to genetically dominant forms of intellectual disability and neurodevelopmental disease (Ba et al., 2016; Pengelly et al., 2016; Katrancha et al., 2017; Paskus et al., 2020).
Trio contains two GEF domains, GEF1 and GEF2, that differentially activate Rac1 or RhoA/Rho1 GTPases, respectively (Debant et al., 1996; Bellanger et al., 1998a; Bellanger et al., 1998b). Trio-GEF1 activation of Rac1 regulates motor neuron axon guidance, cell migration, and axon outgrowth (Bateman et al., 2000; Newsome et al., 2000; Briancon-Marjollet et al., 2008; Peng et al., 2010; Song and Giniger, 2011). Comparatively, little is known about the function of Trio-GEF2; however, recent work suggests that it promotes growth cone collapse through RhoA/Rho1 (Backer et al., 2018). Supporting this model of opposing roles for Trio GEF1 and GEF2 function, studies in Drosophila ddaC neurons suggest that Trio promotes dendritic branching via GEF1 and restricts this process via GEF2 (Iyer et al., 2012). Despite insight into Trio GEF specificity for Rac and RhoA/Rho1, how these two opposing Trio activities are modulated within axons and dendrites remains unclear.
Here, we exploit both genetic and molecular approaches to assess the role of Nab2 and the m6A machinery in regulating expression of the neuronally enriched protein Trio in the adult fly brain. Consistent with our previous findings that Nab2 limits m6A methylation on specific transcripts, reduced levels of either Drosophila m6A reader protein—the nuclear reader Yt521-B or the cytoplasmic reader Ythdf—is sufficient to rescue Nab2null viability and locomotion defects, indicating that m6A-mediated changes in RNA nuclear processing and cytoplasmic metabolism contribute to defects in Nab2 mutants. Focusing on the trio mRNA, we find that Nab2 and the m6A methyltransferase, Mettl3, each promote an intron-excision event within the 5′UTR of a trio mRNA species encoding only the GEF2 (RhoGEF) domain. Intriguingly, levels of the corresponding Trio-GEF2 protein drop in heads of Nab2null but not Mettl3null flies, consistent with a model in which Nab2 modulates both nuclear splicing and cytoplasmic metabolism of the GEF2-only variant of trio mRNA. Critically transgenic expression of Trio-GEF2 rescues axon projection defects in Nab2null mushroom body neurons and class IV ddaC neurons while Trio-GEF1 has the opposite effect of exacerbating Nab2null axon projection defects. Together, these data identify Nab2 and Mettl3 as key regulators of trio 5′UTR structure and provide evidence that altered expression of Trio-GEF2 contributes to axon and dendrite defects in Drosophila lacking Nab2.
RESULTS
Loss of m6A-reader proteins rescues Nab2null defects in viability and adult locomotion
Nab2 loss causes severe defects in Drosophila viability, adult locomotion, and lifespan (Pak et al., 2011). Building on the previous finding that Nab2 loss elevates m6A methylation on select mRNAs (Jalloh and Lancaster et al., 2023), we hypothesized that some Nab2null organismal phenotypes could result from ectopic recruitment of m6A reader proteins onto affected mRNAs. These m6A reader proteins recognize m6A-modified adenosines via a YTH domain (Xu et al., 2014) and act downstream of the methyltransferase machinery to bind and regulate the fate of methylated RNAs (Luo and Tong, 2014; Theler et al., 2014; Xu et al., 2014; Xu et al., 2015; Patil et al., 2018). Unlike more complex mammalian systems, Drosophila have a single nuclear m6A reader protein, YT-521-B (or Ythdc1) and a single cytoplasmic m6A reader protein, Ythdf (Haussmann et al., 2016; Kan et al., 2017).
To assess roles of nuclear Yt521-B and cytoplasmic Ythdf in Nab2 mutant phenotypes, the yt521-B∆N and ythdf∆YTH alleles (Lence et al., 2016; Worpenberg et al., 2021) were individually recombined with a zygotic Nab2null allele (also known as Nab2ex3; imprecise excision of EP3716) (Pak et al., 2011) and assessed for effects on viability, adult locomotion, and lifespan. Homozygous double mutant yt521-B∆N/∆N, Nab2null flies show increase viability compared with Nab2null flies indicating that the nuclear m6A reader is required for the effect of Nab2 loss of viability (Figure 1A). The ythdf∆YTH, Nab2null double mutants are inviable; furthermore, heterozygous reduction of cytoplasmic Ythdf (ythdf∆YTH/+, Nab2null) does not improve Nab2null viability (Figure 1A). In contrast, homozygous loss of nuclear Yt521-B has no effect on Nab2null locomotion, whereas heterozygous reduction of cytoplasmic Ythdf rescues Nab2null climbing rates by approximately 6-fold as assessed in a negative geotaxis assay (at the 30 s timepoint; Figure 1B). Despite the ability of reader alleles (e.g., yt521-B∆N/∆N or ythdf∆YTH/+) to rescue viability or adult locomotion, neither mutant alone rescues Nab2null lifespan defects (Figure 1C). Together, these genetic rescue data provide evidence that neurological effects of Nab2 loss require nuclear and cytoplasmic m6A readers, and suggest that each of these mechanisms may involve different mRNAs.
FIGURE 1:
Loss of m6A-reader proteins rescues Nab2null defects in viability and adult locomotion. (A) Percent of control, Nab2null, yt521-B∆N/∆NNab2null, yhdf∆YTH/+Nab2null, yt521-B∆N/df, and ythdf∆YTH/+ that eclose as viable adults (calculated as #observed/#expected). At least 100 larvae were analyzed for each biological replicate. (B) Negative geotaxis as a measure of locomotion of age-matched adult flies of indicated genotypes over time in seconds (s) taken to reach the top of a vial. The negative geotaxis was observed for at least 10 animals for each biological replicate. (C) Survival of age matched adult flies of the indicated genotypes over time in days. The survival of at least 10 flies was observed for each biological replicate. At least three biological replicates were tested for each genotype and experiment. Significance values are indicated.
Nab2 and Mettl3 regulate splicing of the trio 5′UTR in the Drosophila head
In light of the effects of Nab2 loss on axon and dendrite development (Kelly et al., 2016; Bienkowski et al., 2017; Rounds et al., 2021; Corgiat et al., 2022), we mined our high-throughput RNA sequencing (RNA-seq) analysis of adult heads from Nab2null mutants (zygotic null; imprecise excision of EP3761) and isogenic Controls (precise excision of EP3716) (Pak et al., 2011; Jalloh and Lancaster et al., 2023) to identify potential Nab2 target transcripts regulated by m6A with functions in neurodevelopment. One transcript identified in this analysis was trio, which encodes a Rho guanine nucleotide exchange factor (RhoGEF) that activates specific downstream Rho family GTPases (Bellanger et al., 1998b; Bircher and Koleske, 2021). There are multiple different variants of the trio transcript, two of which are readily detected in adult fly heads: hereafter referred to as trio Medium (trio M) and trio Long (trio L) (Figure 2A, top panel). Visualization of RNA-seq reads from Nab2null and Control heads using Integrative Genomics Viewer (IGV) (Robinson et al., 2017) reveals an increase in reads in introns within the 5′UTR of both trio M and trio L in Nab2null heads relative to Control (Figure 2A). Normal splicing patterns are detected across all other trio intron-exon junctions. Utilizing a publicly available me-RIP-Seq dataset from Drosophila heads (Kan et al., 2021), we bioinformatically identified three m6A sites in the trio M 5′UTR (Figure 2A, red lollipops), but none in the trio L 5′UTR. These data suggest that Nab2 is required for removal of 5′UTR introns in trio M and trio L and present the possibility that the removal of the trio M 5′UTR could also involve m6A.
FIGURE 2:
Nab2 and Mettl3 regulate splicing of the trio 5′UTR in the Drosophila head. (A) RNA-seq reads across the trio locus in Control and Nab2null fly heads (Jalloh and Lancaster et al., 2023). Boxed insets highlight the sequencing reads from the trio L (scale bar = 70 nt) and trio M (scale bar = 145 nt) 5′UTR. Red lollipops denote location of mapped m6A sites. (B) Diagram of the trio M 5′UTR annotated to show location of color-coded primer pairs. The position of the ATG is also indicated (C) RT-PCR analysis of trio M mRNA from Control, Nab2null, and Mettl3null heads. Properly spliced transcript and intron reattaining transcript bands are indicated. Rpl32 serves as a control. qRT-PCR analysis detecting levels of (D) intron retaining or (E) properly spliced trio M transcript from Control, Nab2null, and Mettl3null heads, where control is set to 1.0. (F) Diagram of the trio L 5′UTR annotated to show location of color-coded primer pairs. (G) RT-PCR analysis of trio L mRNA from Control, Nab2null, and Mettl3null heads. Properly spliced and intron retaining transcript bands are indicated. (H) qRT-PCR analysis detecting levels of intron retaining and (I) properly spliced trio L transcript in Control, Nab2null, and Mettl3null heads. RNA from 25 heads was extracted from each genotype for each biological replicate. At least three biological replicates were performed for each experiment. Significance values are indicated.
To experimentally test this prediction, we first analyzed the trio M transcript using reverse transcription PCR (RT-PCR) analysis with primers that detect the trio M 5′UTR intron (exon 1-intron 1 and intron 1-exon 2) (Figure 2B, blue and orange primer pairs). This analysis reveals that the trio M 5′UTR intron-retaining transcript is enriched in adult heads of Nab2null flies as well as in heads lacking Mettl3, the catalytic subunit of the methyltransferase complex (Figure 2C) with concomitant reduction or loss of properly spliced trio M 5′UTR (exon 1-exon 2) (Figure 2B, red primer pair; see Figure 2C). Primers that detect correctly spliced exon 1-exon 2 trio M transcript (Figure 2B, red primer pair) also amplify a ∼550 bp band in Nab2null heads that is an aberrantly spliced product corresponding to the trio M pre-mRNA transcript (Figure 2C). This RT-PCR analysis did not detect the ∼4 kb trio M 5′UTR intron-retaining transcript, possibly due to the large size of the expected product. To quantitate these results, we performed reverse transcription quantitative PCR (qRT-PCR) analysis, which confirms a significant increase in the levels of the trio M 5′UTR intron-retaining transcript in both Nab2null and Mettl3null heads (Figure 2D). Reciprocal qRT-PCR analysis to quantify the levels of properly spliced trio M confirms reduced levels of the properly spliced transcript in Nab2null heads and a complete loss of the properly spliced transcript in Mettl3null heads (Figure 2E).
Shifting the analysis to the trio L 5′UTR intron using primers to detect exon 1-exon 2 confirms the presence of the trio L 5′UTR intron-retaining transcript (exon 1-intron 1-exon 2) in Nab2null heads and reduced levels of the properly spliced transcript (exon 1-exon 2) (Figure 2F, green primer pair; Figure 2G). In contrast, only properly spliced trio L 5′ UTR is detected in heads of flies lacking Mettl3 (Figure 2G). qRT-PCR analysis confirms increased levels of the trio L 5′UTR intron-retaining transcript in Nab2null, but not Mettl3null heads, as compared with Control (Figure 2H). Reciprocally, qRT-PCR analysis using primers that detect levels of properly spliced trio L 5′UTR show reduced transcript levels in Nab2null heads and no change in Mettl3null heads compared with Control (Figure 2I). Collectively, these data confirm that splicing of the trio M and trio L 5′UTR introns are both Nab2-dependent, but that only splicing of the trio M 5′UTR and not the trio L 5′UTR is Mettl3 dependent.
Nab2 regulates levels of Trio M in the Drosophila head
The Drosophila Trio L protein is most similar to a form of human Trio protein (Trio A) that is enriched in the human brain and nervous system (Portales-Casamar et al., 2006). As illustrated in Figure 3A, Trio L contains a Sec14 domain, nine spectrin repeats, one Src homology 3 (SH3) domain, and two catalytic GEF domains comprised of tandem DH and pleckstrin homology (PH) domains, referred to as GEF1 and GEF2. The Drosophila Trio M protein corresponds to the C-terminal end of Trio L, including only the SH3 domain and the GEF2 catalytic domain (Figure 3A).
FIGURE 3:
Nab2 regulates Trio M protein level in the Drosophila head. (A) Schematic of the human (H.s.) Trio A and Drosophila melanogaster (D.m.) Trio L and Trio M proteins. Trio contains a Sec14 domain, nine spectrin repeats, one Src homology 3 (SH3) domain, and two catalytic GEF domains (GEF1 and GEF2), comprised of tandem DH and PH domains. Amino acid lengths are indicated for each Trio protein and the amino acids corresponding to Trio GEF1 and GEF2 are indicated in the schematic for Drosophila Trio L. (B) Immunoblotting analysis of Trio L and Trio M protein levels from Control, Nab2null, and Mettl3null heads. Lamin serves as a loading Control. Molecular weights in kDa are indicated to the left. (C) Quantification of Trio L (left) and Trio M (right) protein levels in B using Image Lab software. Protein levels are normalized to Control, with the value for Control set to 1.0. Protein from at least 25 heads was extracted from each genotype for each biological replicate. Three biological replicates were performed. Significance values are indicated.
Based on the finding that Nab2 regulates splicing of the trio L and trio M 5′UTR introns and that Mettl3 only regulates splicing of the trio M 5′UTR intron, we tested whether these intron retention events affect levels of Trio L or Trio M proteins in the fly head. Immunoblotting analysis reveals that Trio L and Trio M are the major isoforms of Trio in Control brains, and that Nab2 loss reduces levels of Trio M but has no apparent effect on Trio L protein levels relative to Control (Figure 3B). Densitometry analysis of the Trio L and Trio M protein levels demonstrates that the decrease in the steady-state level of Trio M protein is statistically significant (Figure 3C). Although loss of Mettl3 or Nab2 results in trio M 5′UTR intron retention (Figure 2, C–E), only loss of Nab2 causes a concomitant drop in Trio M protein levels (Figure 3, B and C). These results imply an independent effect of Nab2 on postsplicing metabolism of the trio M 5′UTR intron retaining mRNA, suggesting additional roles for Nab2 in the regulation of trio M translation, trafficking, or turnover.
Trio is altered in the Nab2null mushroom body
Previous studies demonstrated that Trio is enriched in the mushroom bodies (Awasaki et al., 2000), which are divided into five lobes per hemisphere (α/α’, β/β’, and γ) that project anteriorly from the dorsally located Kenyon cells (Figure 4A) (Heisenberg, 1998; Roman and Davis, 2001). The α/α’ lobes project dorsally, while the β/β’ and γ lobes project medially, toward the central ellipsoid body (EB) (Figure 4A). As demonstrated previously, loss of Nab2 causes defects in α and β lobe structures, specifically loss or thinning of the α lobes and midline fusion of the β lobe structures (Kelly et al., 2016; Corgiat et al., 2022; Rounds et al., 2022) (Figure 4B). To visualize Trio in these structures, we stained Control brains overexpressing membrane tethered GFP in α/β/γ lobes (201Y-Gal4, UAS-mcd8::GFP) with an α-Trio antibody, which recognizes both Trio L and Trio M protein (Awasaki et al., 2000) (Figure 4B). This analysis confirms that Trio is enriched in γ lobes, Kenyon cell bodies, and the calyx as shown by colocalization with GFP, but absent or below the level of detection in α and β lobes (Awasaki et al., 2000) (Figure 4B; Supplemental Figure S1). Intriguingly, Trio accumulates in dysplasic axons near the midline that are not labeled with the 201Y-Gal4 driver (which exclusively drives mcd8::GFP expression in α/β/γ lobes), suggesting these dysplasic axons belong to the β’ lobe (Figure 4B, middle panel, bottom row).
FIGURE 4:
Trio is altered in the Nab2null mushroom body. (A) Diagram of the adult Drosophila mushroom body lobes depicting axons of the medially projecting gamma (γ) neurons, the vertical alpha (α) and alpha prime (α’) neurons, the medially projecting beta (β) and beta prime (β’) neurons, and the EB. (B) Immunofluorescence images of Control (w-;UAS-mcd8::GFP/201Y-Gal4;;) and Nab2null (w-;UAS-mcd8::GFP/201Y-Gal4;Nab2null;) mushroom bodies driving UAS-mcd8::GFP under the α, β, γ lobe-specific mushroom body 201Y-Gal4 driver. (C) Immunofluorescence images of Control (w-;UAS-mcd8::GFP/C305a-Gal4;;) and Nab2null (w-;UAS-mcd8::GFP/C305a-Gal4;Nab2null;) mushroom bodies driving UAS-mcd8::GFP under the α’ and β’ lobe-specific mushroom body C305a-Gal4 driver. False colored panels show fluorescence corresponding to α-GFP (green, mcd8::GFP), α-trio (purple), and merges of the channels. At least 25 brains were analyzed for each genotype. Scale bar = 50 μm.
We extended this analysis to explore how loss of Nab2 impacts Trio localization. Trio is lost in the GFP-positive γ lobes of Nab2null brains (Figure 4B, bottom row). Given the strong reduction of Trio M protein detected by immunoblotting of Nab2null heads (see Figure 2B), this result suggests that Trio M may be the primary isoform of Trio present in mushroom body γ lobes.
To explore the localization of Trio specifically in the α’ and β’ lobe structures, we utilized the Gal4-UAS system to overexpress GFP (UAS-mcd8::GFP) using a prime lobe specific Gal4 driver (CkaC305a-Gal4; exclusively drives mcd8::GFP expression in α’and β’ lobes) (Figure 4C). As reported previously, Trio is enriched in α’ and β’ axons, and is also detected in γ lobe axons and EB of Control brains (Awasaki et al., 2000) (Figure 4C, top row). Nab2null brains show complete loss or thinning of the α’ lobe axons as well as a distinct defasciculation phenotype in the β’ lobe structures (Figure 4C). These morphological phenotypes are accompanied by Trio loss in Nab2nullγ lobe axons and EB, and Trio accumulation in the distal portion of β’ lobe axons closest to the brain midline (Figure 4C, middle panel, bottom row).
Expression of Trio GEF2 rescues α/α’ and β’ defects in Nab2null mushroom bodies
The reduced level of Trio M protein in Nab2null heads (Figure 3B) raises the possibility that an imbalance in the relative dose of Trio-GEF1 and Trio-GEF2 activities contributes to mushroom body morphology defects observed in Nab2null flies (Kelly et al., 2016). This model is based on the established role of the Trio protein in patterning of axons in the mushroom body (Awasaki et al., 2000), and predicts that loss of Trio M lowers Trio-GEF2 activity within specific mushroom body lobes.
To test this model, transgenes encoding specifically the Trio-GEF1 (UAS-Trio-GEF1; Figure 3A, AAs 1287–1583 tagged at the 3′end with eight copies of the tag MYC) or Trio-GEF2 domain (UAS-Trio-GEF2; Figure 3A, AAs 1945–2246 tagged at the 3′end with eight copies of the tag MYC) (Awasaki et al., 2000; Newsome et al., 2000), were expressed in all mushroom body lobes of Nab2null brains using the OK107-Gal4 driver. As previously reported (Kelly et al., 2016; Bienkowski et al., 2017; Corgiat et al., 2021; Corgiat et al., 2022; Rounds et al., 2022) and shown in Figures 5, A–C, Nab2null mutant brains have highly penetrant defects in the structure of the mushroom body α lobes (missing or thinned) and β lobes (missing, thinned, or midline crossed) as detected by α-FasII staining, which specifically recognizes the α, β and weakly the γ lobes (Crittenden et al., 1998). Transgenic expression of Trio-GEF2 alone has no effect on mushroom body structure in a control background. Intriguingly, transgenic expression of Trio-GEF2 does not rescue Nab2null β lobe defects, but significantly suppresses Nab2null α lobe defects (Figure 5, A–C). In contrast, mushroom body expression of Trio-GEF1 in control brains causes complete loss of axonal projection from the Kenyon cells (no lobe structures were detected in any of the brains analyzed; n = 25) and is completely lethal in a Nab2null background (Figure 5, A–C). Thus, Nab2 loss sensitizes mushroom body axons to the dose of Trio GEF domains such that expression of Trio-GEF2 rescues α lobe axons and expression of extra Trio-GEF1 is lethal to the animal; neither effect is observed in Control brains, indicative of a tight link between Nab2 and Trio GEF dosage in the developing mushroom body.
FIGURE 5:
Expression of Trio GEF2 rescues α/α’ and β’ defects in Nab2null mushroom bodies. (A) Top: Schematic of the Drosophila mushroom body with lobes stained by α-Fasciclin II (FasII) highlighted. Bottom: Representative max projections of mushroom bodies of indicated genotypes stained by α-FasII. (B) Quantification of frequency of α lobe defects in each indicated genotype. (C) Quantification of frequency of β lobe defects in indicated genotypes. (D) Top: Schematic of the Drosophila mushroom body with lobes overexpressing mcd8::GFP under the C305a-Gal4 driver highlighted. Bottom: Representative max projections of mushroom bodies of indicated genotypes stained with α-GFP. (E) Quantification of frequency of α’ lobe defects in each indicated genotype. (F) Quantification of frequency of β’ lobe defects in indicated genotypes. (G) Quantification of defasciculation phenotype severity in the indicated genotypes. At least 25 brains were analyzed for each genotype. Scale bar = 50 μm.
Given the enrichment of Trio protein in the α’ and β’ lobes (see Figure 4B), we also tested whether transgenic expression of the Trio GEF1 or GEF2 domain could autonomously rescue Nab2null α’ or β’ lobe morphology as visualized with the CkaC305a-Gal4 driver (with UAS-mcd8::GFP). As with Trio-GEF1 or Trio-GEF2 expression in the α, β, and γ lobes using the 201Y-Gal4 driver, expression of Trio-GEF2 alone has no effect on α’ and β’ lobe morphology in Control brains but rescues Nab2null defects in α’ lobe structure and strongly reduces β’ lobe defasciculation (Figure 5, D–G). Expression of Trio-GEF1 in α’ and β’ lobes has the inverse effect of late larval lethality in both Control and Nab2null animals (Figure 5D). These data are consistent with a model in which Nab2 acts through Trio-GEF2 to guide axon projection and fasciculation in the α’ and β’ lobes.
Expression of Trio GEF2 rescues Nab2null dendritic arborization defects in class IV ddaC sensory neurons
Nab2 normally restricts branching of sensory dendrites in larval class IV ddaC neurons in body wall neurons (Corgiat et al., 2022). Significantly, Trio-GEF1 promotes and Trio-GEF2 restricts branching of dendrites from these same class IV ddaC neurons (Figure 6, A and B) (Iyer et al., 2012). Thus, we reasoned that an imbalance of Trio-GEF1 and Trio-GEF2 activities in Nab2null ddaC neurons might contribute to dendritic defects. To test this model, we employed a pickpocket (ppk)-Gal4,UAS-mcd8::GFP system to visualize class IV ddaC cell bodies and dendritic trees. As observed previously, loss of Nab2 (Corgiat et al., 2022) or transgenic expression of Trio-GEF1 (Iyer et al., 2012) individually increases dendritic branch complexity, while transgenic expression of Trio-GEF2 reduces dendritic branch complexity (Iyer et al., 2012) (Figure 6, A and B). Significantly, combining Nab2 loss (Nab2null) with ddaC-specific expression of Trio-GEF2 rescues overarborization normally observed in Nab2null larvae (Figure 6, A and B). Intriguingly, transgenic expression of Trio-GEF1 in Nab2null larvae causes an increase the variance between arborization phenotypes among the neurons analyzed (Figure 6B). Together, these data indicate that loss of Trio-GEF2 in Nab2null larvae likely contributes to Nab2null ddaC overarborization defects.
FIGURE 6:
Expression of Trio GEF2 rescues Nab2null dendritic arborization defects in class IV ddaC sensory neurons. (A) Maximum intensity projections of Drosophila class IV ddaC neurons from Control, Nab2null, Nab2null UAS-Trio-GEF2, Nab2null UAS-Trio-GEF1, UAS-Trio GEF2, and UAS-Trio-GEF1 L3 larvae. Inset black boxes show high magnification views of dendritic arbors. (B) Quantification of total number of dendritic terminals for each genotype. At least eight neurons were measured for each genotype. Significance values are indicated.
Expression of Trio GEF2 rescues Nab2null defects in viability and locomotion
Given the genetic interactions between the UAS-Trio-GEF1 and UAS-Trio-GEF2 transgenes and Nab2, we assessed whether transgenic expression of these distinct Trio GEF domains in the mushroom body (OK107-Gal4, UAS-Trio-GEF1 or UAS-Trio-GEF2) affects the organismal phenotypes of viability, adult locomotion, and lifespan. Confirming our previous findings (Pak et al., 2011), we detected severe reductions in viability, adult locomotion, and lifespan in Nab2null flies compared with Control (Figure 7, A–C). As previously noted, expression of Trio-GEF1 in mushroom bodies is lethal at late larval stages (Figure 7A). In contrast, transgenic expression of Trio-GEF2 in mushroom bodies strongly suppresses Nab2null defects in viability and suppresses defects in adult locomotion but does not significantly alter lifespan (Figure 7, A–C), indicating that a deficit in Trio-GEF2-regulated cytoskeletal dynamics within OK107-expressing brain neurons contributes to developmental and postdevelopmental defects in Drosophila lacking Nab2.
FIGURE 7:
Expression of Trio GEF2 rescues Nab2null defects in viability and locomotion. (A) Percent of Control, Nab2null, Nab2null UAS-Trio-GEF2, UAS-Trio GEF2, or UAS-Trio-GEF1 that eclose as viable adults (calculated as #observed/#expected) using the OK107-Gal4 mushroom body driver. (B) Negative geotaxis of age-matched adult flies of indicated genotypes over time in seconds (s). (C) Survival of age matched adult flies of the indicated genotypes over time in days. At least three biological replicated were tested for each genotype. Significance values are indicated.
DISCUSSION
Here, we identify a role for the Drosophila Nab2 RBP and Mettl3 m6A methyltransferase in regulating the trio mRNA, which encodes a conserved RhoGEF protein that is altered in human intellectual disability and regulates axon guidance and dendritic arborization through two GEF domains that individually control the cytoskeletal regulators Rac and RhoA/Rho1 (Debant et al., 1996; Bellanger et al., 1998a; Bellanger et al., 1998b; Bateman and Van Vactor, 2001; Jaffe and Hall, 2005; Briancon-Marjollet et al., 2008; Iyer et al., 2012; Bircher and Koleske, 2021). The data presented provide strong evidence that the trio transcript is a key downstream target of Nab2 in neurons based on an m6A- and Nab2-dependent splicing event and identifies specific effects of each Trio GEF domain within axons and dendrites that develop from neurons lacking Nab2. The results of this study combine with our previous work (Corgiat et al., 2022; Jalloh and Lancaster et al., 2023) to support a model in which Nab2 regulates transcripts that encode key regulators of neurodevelopment, including the conserved GEF, Trio. In the broader context, the phenotypic consequences of loss of an RBP result from the collective changes to numerous target transcripts, and defining the mechanistic basis of these phenotypes requires systematic analysis of how individual targets, such as trio, are impacted. In the case of Nab2, evidence now supports both m6A-dependent and independent roles in trio mRNA splicing as well as potential effects on downstream processing such as cytoplasmic metabolism of trio mRNA. Taken together, these findings support a model where RBPs such as Nab2/ZC3H14 regulate a collection of target transcripts, potentially through multiple mechanisms, that all contribute to downstream phenotypes.
Previous work illustrating broad rescue of Nab2null phenotypes by Mettl3 heterozygosity (Jalloh and Lancaster et al., 2023) suggested that other regulators of m6A-modified transcripts could also contribute to Nab2null defects in viability, adult locomotion, and lifespan. Here, we demonstrate that loss of nuclear or cytoplasmic m6A reader function rescues some, but not all, organismal phenotypes associated with loss of Nab2 (Figure 1). Homozygous loss of the nuclear m6A reader YT521-B suppresses Nab2null defects in viability, but not locomotion or lifespan, whereas heterozygous loss of the cytoplasmic m6A reader Ythdf dominantly suppresses Nab2null defects in locomotion, but not viability or lifespan. Collectively, these data suggest that mRNA targets of Nab2 responsible for these behavioral phenotypes may be differentially regulated between cell types and in an m6A-dependent manner. For instance, Nab2-regulated transcripts encoding proteins that govern Drosophila viability could rely more heavily on nuclear m6A regulatory mechanisms, such as splicing or export. On the other hand, Nab2-regulated transcripts encoding proteins that govern Drosophila negative geotaxis could more heavily require cytoplasmic m6A regulatory mechanisms, such as translation or stability. Moreover, the inability of YT521-B or Ythdf loss to rescue Nab2null defects in lifespan suggests that Nab2-regulated transcripts that govern lifespan may not by modified by m6A, or that Nab2 plays m6A-independent roles in regulating transcripts critical to control lifespan.
Previous studies demonstrated that loss of m6A regulatory proteins disrupts axon projection in the Drosophila mushroom body (Kan et al., 2021; Worpenberg et al., 2021). Although Mettl3 heterozygosity broadly rescues Nab2null behavioral defects (Jalloh and Lancaster et al., 2023), a decrease in Mettl3 does not dominantly suppress mushroom body morphology defects (Supplemental Figure S2). This finding suggests that Mettl3 heterozygosity is insufficient to reduce m6A methylation on Nab2-target transcripts to a degree necessary for rescue of axonal projection defects. Given the ability of m6A writer and reader alleles to broadly rescue Nab2null phenotypes, future studies will aim to further define the relationship between m6A machinery and Nab2 in relation to regulation of Drosophila mushroom body morphology. Moreover, a genome-wide approach to assess transcriptomic changes in m6A in Nab2null flies will help delineate the neuronal, Nab2-regulated transcripts that exhibit changes in m6A methylation.
A number of trio transcript variants exist in the Drosophila brain (Awasaki et al., 2000). Here, we demonstrate that both Nab2 and Mettl3 are required for proper splicing of the 5′UTR intron of trio M. On the other hand, splicing of the 5′UTR intron of trio L is dependent on Nab2 but not Mettl3. These data align with previously published RNA-seq data from Nab2null Drosophila heads (Jalloh and Lancaster et al., 2023), as well as a publicly available mi-CLIP-seq dataset that mapped m6A sites in the trio M 5′UTR intron, but not in the trio L 5′UTR intron (Kan et al., 2021). Notably, our data reveal that disruptions in splicing of the trio 5′UTR due to loss of Nab2 or Mettl3 do not correspond with perturbations in steady-state levels of Trio L and Trio M proteins. These results show that the intron retention event in the 5′UTR of trio M is associated with a significant reduction in the level of Trio M protein in the Nab2null fly head. Surprisingly, the steady-state level of Trio M protein is unaffected in Mettl3null heads even though trio 5′UTR intron retention is comparable to the levels observed in Nab2null heads. These data imply that Nab2 may regulate trio M protein levels in a manner independent of trio M 5′UTR splicing. However, given previously defined roles for Nab2 as a negative regulator of m6A methylation (Jalloh and Lancaster et al., 2023), this observation more likely suggests that excess m6A on the trio M pre-mRNA upon loss of Nab2 may disrupt subsequent translation, trafficking, or stability. In contrast, retention of the trio L 5′UTR intron that occurs upon loss of Nab2 does not affect the steady-state level of the Trio L protein, suggesting this intron retention event may not disrupt translation. Alternatively, the remaining level of the properly spliced trio L in Nab2null heads may be sufficient to maintain the steady-state level of Trio L protein. Further studies expanding to additional coregulated Nab2 and Mettl3 targets will be required to define how these factors regulate posttranscriptional events.
Given that Nab2 regulates splicing and downstream processing of numerous neuronally-enriched mRNAs (Jalloh and Lancaster et al., 2023), morphological defects observed in Nab2null mushroom bodies are likely due to collective processing defects that affect multiple RNAs critical for axon development. Our results support a model where loss of Trio M, and therefore GEF2 function, contributes to morphological defects in mushroom bodies of Nab2null flies. Previous studies demonstrated that Trio is a critical regulator of mushroom body morphology and Trio is enriched in the α’, β’, and γ lobes, but is virtually absent in the α and β lobes (Awasaki et al., 2000). We confirm these findings and further demonstrate that upon loss of Nab2, Trio levels are depleted in the γ lobes. Despite the established functions of Trio in regulating γ lobe formation (Awasaki et al., 2000), γ lobe defects are not detected upon loss of Nab2 (Kelly et al., 2016). Given that Trio M is the only isoform of Trio depleted in Nab2null brains (Figure 3B) and γ lobes show no defects (Figure 5A), this finding suggests that Trio L, and therefore GEF1 function, is responsible for patterning γ lobe axons in the developing brain.
Studies of axon pathfinding mechanisms in the mushroom body demonstrate that the α’ and β’ lobes guide development of the α and β lobes (Fushima and Tsujimura, 2007). Given that Trio is enriched in the α’/β’/γ lobes of the mushroom body, our data suggest that loss of Trio M, and therefore GEF2 levels, in α’/β’ lobes contributes to Nab2null α/β lobe defects. We demonstrate that transgenic expression of Trio-GEF2 in α’/β’ lobes (C305a-Gal4) of Nab2null mushroom bodies rescues α’ lobe defects and β’ lobe defasciculation phenotypes in a cell autonomous manner (Figure 5). Moreover, expression of Trio-GEF2 in all mushroom body lobes (OK107-Gal4) rescues Nab2null α lobe defects; however, whether this rescue occurs in a cell autonomous manner remains unknown (Figure 5). Collectively, these data demonstrate that Trio-GEF2 rescue is limited to some mushroom body lobes (α, α’, β’) and not others (β), implying that Trio-GEF2 is required for projection of only some Nab2null axons. In this regard, misprojection defects in Nab2null β axons are not rescued by multiple genetic manipulations that rescue α axon defects (e.g., by transgenic expression of Trio-GEF2 [this study] or by single copy alleles of fmr1, Atx2, or PCP components [Kelly et al., 2016; Bienkowski et al., 2017; Rounds et al., 2021; Corgiat et al., 2022]). These findings imply specific roles for Nab2 in these two types of Kenyon cell projections and suggest that Nab2 regulates different mRNAs to govern development of distinct mushroom body lobes.
Previous studies established that the Trio-GEF1 domain acts primarily through activation of Rac1 to promote axon outgrowth and pathfinding, while Trio-GEF2 acts primarily through RhoA/Rho1 to restrict neurite outgrowth (Leeuwen et al., 1997; Iyer et al., 2012; Bircher and Koleske, 2021). Given that loss of Nab2 disrupts the ratio of GEF1 and GEF2 in Drosophila heads by decreasing the level of Trio M but not Trio L, we hypothesized that expression of the GEF1 effector, Rac1, in mushroom body neurons would exacerbate Nab2null phenotypic and morphological defects, whereas expression of the GEF2 effector, RhoA/Rho1, would rescue these same defects. Intriguingly, we observed that expression of either Rac1 or RhoA/Rho1 in the absence of Nab2 in Trio-enriched mushroom body neurons is lethal (Supplemental Figure S3). These data suggest that further expression of Rac1 in Nab2null flies in which GEF1 levels, and therefore likely Rac1 activation, dominates is detrimental to nervous system development and the lethality induced by expression of RhoA/Rho1 upon loss of Nab2, indicates that Trio-GEF2 may act via other unknown effectors to govern mushroom body development. On the other hand, alternative RhoGEFs may compensate for the loss of Trio M protein in the mushroom body thereby leading to the lethality of Nab2null animals transgenically expressing Rac1 or RhoA/Rho1. In sum, further studies are required to elucidate how loss of Nab2 alters Rac1 and RhoA/Rho1 activity in the Drosophila brain.
Nab2 and Trio have established roles in sculpting dendritic arborization of class IV ddaC neurons in the Drosophila PNS (Iyer et al., 2012; Corgiat et al., 2022). Here, we demonstrate that transgenic expression of the Trio-GEF2 domain rescues overarborization defects in Nab2null class IV ddaC neurons. In line with previous studies (Iyer et al., 2012), we validate that transgenic expression of Trio-GEF1 in class IV ddaC neurons causes dramatic overarborization defects, while transgenic expression of Trio-GEF2 causes underarborization defects compared with control animals. Interestingly, we also demonstrate that expression of Trio-GEF1 in class IV ddaC neurons of Nab2null flies results in a wide range of arborization phenotypes. Very few of these animals survive to the wandering third instar larval stage and no animals survive to adulthood. Given these observations, disruption of Nab2-regulated mRNAs in these neurons as well as overactivation of Rac1 by GEF1 may severely disrupt ddaC development such that arborization defects are highly variable from animal-to-animal. Overall, these data support a role for Trio M, and therefore GEF2 loss, in contributing to the established overarborization defects in Nab2null class IV ddaC neurons (Corgiat et al., 2022).
In aggregate, these data reveal a role for Nab2 and Mettl3 in regulating splicing and protein levels of the RhoGEF Trio to support proper nervous system development. Genetic interactions between the m6A machinery and Nab2 support a role for Nab2 in the regulation of m6A methylation. We show for the first time that loss of Trio M, and therefore GEF2 levels, in Nab2null flies contributes to several Nab2null defects, including neuronal defects, such as mushroom body morphology and class IV ddaC arborization. Moreover, we demonstrate that transgenic expression of Trio-GEF2 broadly rescues Nab2null viability and adult locomotion. This regulatory relationship between Nab2 and Trio-GEF2 could be cell autonomous, but further experiments are required determine the nature of this interaction. Given that mutations in human ZC3H14 and TRIO are both linked to intellectual disabilities (Pak et al., 2011; Ba et al., 2016), dysregulation of Trio function in neurons is one potential mechanism to explain axonal and dendritic phenotypes observed in Nab2null Drosophila (Pak et al., 2011; Kelly et al., 2016; Bienkowski et al., 2017; Corgiat et al., 2021; Corgiat et al., 2022; Jalloh and Lancaster et al., 2023) and Zc3h14 mutant mice (Jones et al., 2020), as well as the cognitive defect observed in human patients lacking ZC3H14 (Pak et al., 2011).
MATERIALS AND METHODS
Request a protocol through Bio-protocol.
Resources table
Reagent or resource | Source | Identifier |
---|---|---|
Antibodies | ||
Mouse monoclonal 9.4A anti-Trio | University of Iowa Developmental Studies Hybridoma Bank | RRID:AB_528494 |
Mouse monoclonal ADL101 anti-Lamin | University of Iowa Developmental Studies Hybridoma Bank | RRID:AB_528332 |
Mouse monoclonal 1D4 anti-Fasciclin II | University of Iowa Developmental Studies Hybridoma Bank | RRID: AB_528235 |
Rabbit Polyclonal Anti-Green Fluorescent Protein (GFP) | Thermo Fisher Scientific | Catalogue no. A-11122 |
Alexa Fluor 488 AffiniPure Polyclonal Goat Anti-rabbit IgG | Jackson ImmunoResearch Laboratories | RRID: AB_2338046 |
Cy3 AffiniPure Polyclonal Goat Anti-Mouse IgG | Jackson ImmunoResearch Laboratories | RRID: AB_2338690 |
Chemicals, peptides, and recombinant proteins | ||
TRIzol reagent | Invitrogen | Catalogue no. 15596018 |
1-Bromo-3-chloropropane | Scientific Laboratory Supplies | Catalogue no. B9673 |
2-propanol/isopropanol | Thermo Fisher Scientific | Catalogue no. A416-1 |
Ethanol 200 proof | Thermo Fisher Scientific | Catalogue no. 04-355-233 |
Diethyl pyrocarbonate (DEPC) | Sigma-Aldrich | Catalogue no. D5758 |
Agarose LE, Quick Dissolve | Genesee Scientific | Catalogue no. 20-102QD |
Red Safe | iNtRON Biotechnology | Catalogue no. 21141 |
NaCl | Sigma-Aldrich | Catalogue no. S7653 |
KCl | Sigma-Aldrich | Catalogue no. P3911 |
ZnCl2 | Sigma-Aldrich | Catalogue no. 208086 |
Polysorbate 20 (Tween 20) | Fisher Bioreagents | Catalogue no. BP-337-500 |
Triton X-100 | Sigma-Aldrich | Catalogue no. T8787 |
Tris Base Ultrapure | USBiological Life Sciences | Catalogue no. T8600 |
Sodium Dodecyl Sulfate | Fisher Bioreagents | Catalogue no.YBP166500 |
Sodium deoxycholate | Thermo Fisher Scientific | Catalogue no. 89905 |
NP-40 | Thermo Fisher Scientific | Catalogue no. 85124 |
Dithiothreitol (DTT) | Thermo Fisher Scientific | Catalogue no. R0861 |
Bromophenol Blue | Thermo Fisher Scientific Chemicals | Catalogue no. A18469-18 |
Glycerol | USBiological Life Sciences | Catalogue no. G8145 |
RNaseOUT | Invitrogen | Catalogue no. 10777019 |
p-Coumaric acid | Sigma-Aldrich | Catalogue no. C9008 |
Luminol | Sigma-Aldrich | Catalogue no.A8511 |
Acetic Acid, Glacial | Thermo Fisher Scientific | Catalogue no. A38-212 |
EDTA | USBiological Life Sciences | Catalogue no. E2210 |
Paraformaldehyde | Electron Microscopy Sciences | Catalogue no. 15713 |
VECTASHEILD mounting medium | Vector Laboratories Inc. | REF#H-1000 |
Normal Goat Serum | Jackson ImmunoResearch Laboratories | RRID:AB_2336990 |
Diethyl Ether Anhydrous (stabilized with BHT) | Tokyo Chemical Industry (TCI) America | Catalogue no. D3497 |
Halocarbon oil 27 | Sigma-Aldrich | Catalogue no. H8773 |
Critical commercial assays | ||
DNase I, Amplification Grade | Invitrogen | Catalogue no. 18068015 |
M-MLV Reverse Transcriptase | Invitrogen | Catalogue no. 28025013 |
Taq DNA Polymerase (1000U) | Qiagen | Catalogue no. 201205 |
QuantiTect SYBR Green PCR Kit | Qiagen | Catalogue no. 204145 |
MicroAmp Fast Optical 96-Well Reaction Plate with Barcode, 0.1 ml | Applied Biosystems | Catalogue no. 4346906 |
MicroAmp Optical Adhesive Film | Applied Biosystems | Catalogue no. 4311971 |
4–20% Mini-PROTEAN TGX Stain-Free™ Protein Gels | Bio-Rad | Catalogue no. 4568093 |
Nitrocellulose Membrane, 0.2 µm | Bio-Rad | Catalogue no. 1620112 |
iScript Reverse Transcriptase Supermix | Bio-Rad | Catalogue no. 1708841 |
Phusion High Fidelity PCR | Thermo Fisher Scientific | Catalogue no. F530S |
Pierce BCA Protein Assay Kit | Thermo Fisher Scientific | Catalogue no. 23225 |
SuperFrost Plus slides | Thermo Fisher Scientific | Catalogue no. 12-550-15 |
Il90p-Experimental models: Organisms/Strains | ||
D. melanogaster: w-;;Nab2pex41 (Control); | (Pak et al., 2011) | N/A |
D. melanogaster: w-;;Nab2ex3 (Nab2null); (Zygotic loss of Nab2) | (Pak et al., 2011) | N/A |
D. melanogaster: w-;;Mettl3null; | (Lence et al., 2016) | N/A |
D. melanogaster:YT521-B∆N | (Lence et al., 2016) | N/A |
D. melanogaster: Ythdf∆YTH (Ythdf0) | (Worpenberg et al., 2021) | N/A |
D. melanogaster:w1118;;Df(3R)BSC655 (ythdf deficiency) | Bloomington Drosophila Stock Company | BDSC:26507 |
D. melanogaster: y1w67c23;UAS-mcd8::GFP, 201Y-Gal4;; | Bloomington Drosophila Stock Company | BDSC:64296 |
D. melanogaster:w*;Ckac305a-Gal4 | Bloomington Drosophila Stock Company | BDSC:30829 |
D. melanogaster: y1w*;UAS-mcd8::GFP;; | Bloomington Drosophila Stock Company | BDSC:5137 |
D. melanogaster: w*;;;OK107-Gal4 | Bloomington Drosophila Stock Company | BDSC:854 |
D. melanogaster:y1w*:UAS-trio-GEF1-myc(Trio-GEF1 domain; AAs 1287-1583 tagged at the 3′end with eight copies of the tag MYC) | Bloomington Drosophila Stock Company | BDSC:9133 |
D. melanogaster: y1w*:UAS-trio-GEF2-myc(Trio-GEF2 domain; AAs 1945-2246 tagged at the 3′end with eight copies of the tag MYC) | Bloomington Drosophila Stock Company | BDSC:9134 |
D. melanogaster:w*;;ppk-Gal4 | Bloomington Drosophila Stock Company | BSDC:32079 |
D. melanogaster:w*;UAS-Rac1 | Bloomington Drosophila Stock Company | BSDC:6293 |
D. melanogaster:w*;UAS-Rho1 | Bloomington Drosophila Stock Company | BSDC:58819 |
Oligonucleotides | ||
RT-PCR trio L Forward: AACAAAACAGAGAGCGCCC | This study | N/A |
RT-PCR trio L Reverse: GATGGGCACTGCAGCATAA | This study | N/A |
qRT-PCR trio L Intron Retention Forward: 5′-TTAGCCCGCGTCAAGTC-3′ | This study | N/A |
qRT-PCR trio L Intron Retention Reverse: 5′-CTGCTTGTGCCACCAAAT-3′ | This study | N/A |
qRT-PCR trio L Properly Spliced Forward: 5′-GTTGTGTTGACAAAAGAGTG-3′ | This study | N/A |
qRT-PCR trio L Properly Spliced Reverse: 5′-GATGGGCACTGCAGCATAA-3′ | This study | N/A |
RT-PCR trio M Intron Retention (1) Forward: 5′-CAGCAGTCTCTTCTTCACTAA-3′ | This study | N/A |
RT-PCR trio M Intron Retention (1) Reverse: 5′-ACTCGGATTGTTGTTTCACTTT-3′ | This study | N/A |
RT-PCR trio M Intron Retention (2) Forward: 5′-GACTGCGCAAACATAGCATTA-3′ | This study | N/A |
RT-PCR trio M Intron Retention (2) Reverse: 5′-ATCCGCTCGTTGAGAAACT-3′ | This study | N/A |
RT-PCR trio M Properly Spliced Forward: 5′-CGCTAAAGAGGAGCGCAATA-3′ | This study | N/A |
RT-PCR trio M Properly Spliced Reverse: 5′-CTTCTTAACACTCTTCATGATTCG-3′ | This study | N/A |
qRT-PCR trio M Intron Retention Forward: 5′-TTGAGTGAACCCGCTAAAG-3′ | This study | N/A |
qRT-PCR trio M Intron Retention Reverse: 5′-CTTTGGAGTGCTTGTTCTTATC-3′ | This study | N/A |
qRT-PCR trio M Properly Spliced Forward: CGTCCATAAATTGAGTCGGAGAAC | This study | N/A |
qRT-PCR trio M Properly Spliced Reverse: 5′-CTTCTTAACACTCTTCATGATTCG-3′ | This study | N/A |
RT-PCR and qRT-PCR rpl32 Forward: AAGATGACCATCCGCCCAGCATAC | This study | N/A |
RT-PCR and qRT-PCR rpl32 Reverse: ACGCACTCTGTTGTCGATACCCTT | This study | N/A |
Software and algorithms | ||
Fiji/ImageJ | (Schindelin et al., 2012; Rueden et al., 2017) | https://imagej.net/ |
Integrated Genome Viewer (IGV) | (Robinson et al., 2011) | https://igv.org/ |
Image Lab | Bio-Rad | https://image-lab-4-0.software.informer.com/ |
GraphPad (Prism) | ||
Other | ||
ChemiDoc | Bio-Rad | Catalogue no. 12003153 |
Humidified Incubators | Shel Lab | SRI20PF |
#5 Dumont Fine Forceps | Ted Pella Inc. | Prod#5622 |
Motorized Pestle | Argos Technologies | Catalogue no. A0001 |
NanodropOne | Thermo Fisher Scientific | N/A |
Resource availability
Lead contact.
Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Ken Moberg (kmoberg@emory.edu).
Materials availability
The D. melanogaster lines generated in this study are available by contacting the Lead Contact.
Data and code availability
This study did not generate any dataset or codes.
Experimental model and subject details
Flies (D. melanogaster) were raised on standard cornmeal agar medium and maintained in an incubator set at 25°C with a 12-h light/dark cycle. Crosses were reared under the same conditions, and standard medium was supplemented with dry yeast. The GAL4-UAS binary transgenic system was used to express transgenes of interest. Details of genotypes can be found in the Key Resources Table. One- to 5-day-old flies were used for experiments in this study. An equal number of males and females was used for all experiments.
Method details
D. melanogaster stocks and genetics.
D. melanogaster stocks were raised on standard cornmeal agar and maintained in humidified incubators (SRI20PF, Shel Lab) at 25°C with 12-h light/dark cycles. Crosses were reared under the same conditions and supplemented with dry yeast. The strains used in this study are described in the Key Resources Table.
Viability and lifespan analysis.
Viability at 25°C was measured by assessing eclosion rates of 100 wandering L3 larvae collected for each genotype, and then reared in a single vial. Hatching was recorded for 5–6 d. At least three independent biological replicates per genotype were tested for significance and calculated using group analysis on GraphPad (Prism). Lifespan was assessed at 25°C as described previously (Morton et al., 2020). In brief, newly eclosed flies were collected, placed in vials (10 flies per vial), and then transferred to fresh vials weekly, or as needed. Survivorship was scored daily. At least three independent biological replicates were tested for each genotype, and significance was calculated using group analysis on GraphPad (Prism).
Locomotion assays.
Negative geotaxis was tested as previously described (Morton et al., 2020). Briefly, newly eclosed flies (day 0) were collected, and kept in vials for 2–5 d. Cohorts of 10 age-matched flies were transferred to a 25 ml graduated cylinder for analysis. Flies in graduated cylinders were tapped to bring flies to the bottom of the vial and the rate at which the flies traveled to the top of the vial (25 ml mark) was measured at 5, 10, 15, and 30 s). At least three biological replicates per genotype were analyzed and significance was calculated using grouped analysis on GraphPad (Prism).
Drosophila decapitation.
CO2-anesthetized flies were collected and frozen at −80°C for approximately five minutes. Frozen flies were then placed on a metal plate over dry ice. Gently, #5 Dumont fine forceps (Ted Pella, Inc.) were placed between the Drosophila head and thorax to remove the head from the remainder of the body. Heads were carefully placed in Eppendorf tubes, on ice, for subsequent processing.
RNA isolation for RT-PCR and real-time qPCR.
Total RNA was isolated from ∼25 adult fly heads using the TRIzol (Invitrogen) method. Briefly, Drosophila heads were homogenized in 0.1 ml TRIzol using a motorized pestle (Argos Technologies) on ice. TRIzol was added to samples to bring to a total volume of 0.5 ml and 0.1 ml of 1-Bromo-3-chloropropane (Scientific Laboratory Supplies) was added. Samples were vortexed on high speed for 10 s and incubated at room temperature for 15 min. Next, samples were centrifuged for 15 min at 13,000 × g at 4°C. The top, aqueous layer was removed and placed into a clean Eppendorf tube. An equal volume of 2-propanol (∼250 μL) was added. Samples were inverted 10 times and incubated at room temperature for 10 min. Next, samples were centrifuged for 15 min at 13,000 × g at 4°C. The supernatant was removed, and 0.5 ml of 75% ethanol was added. Samples were centrifuged a final time for 15 min at 13,000 × g at 4°C. The supernatant was removed, and the samples were allowed to air dry until the remaining ethanol evaporated (∼5 min). The pellet was resuspended in 10–20 μL of DEPC water. RNA concentration and purity was assessed using a Spectrophotometer (Thermo Fisher Scientific). Total RNA (1 μg) was treated with DNaseI (Qiagen). cDNA was generated using M-MLV Reverse Transcriptase (Invitrogen). Qiagen Taq polymerase (Qiagen) was used for PCR amplification of target transcripts and products were resolved and imaged on 1–2% agarose gels (Chemi-Doc). qRT-PCR reactions were carried out in technical triplicate with QuantiTect SYBR Green Master Mix using Applied Biosystems StepOne Plus real-time machine (ABI). Results were analyzed using ∆∆CT method, normalized to loading Control (e.g., rpl32), and plotted as relative levels normalized to Control. At least three independent biological replicates were performed for each genotype, and significance was calculated using group analysis on GraphPad (Prism). Primers used for all PCR reactions are listed in the Key Resources Table.
Immunoblotting.
For analysis of Trio protein levels, ∼25 adult fly heads (1–5 d old) were decapitated and collected on dry ice. Protein lysates were prepared by homogenizing heads in 0.5 ml of RIPA-2 Buffer (50 mM Tris-HCl, pH 8; 150 mM NaCl; 0.5% sodium deoxycholate; 1% Igepal CA-630 0.1% SDS) supplemented with protease inhibitors (1 mM PMSF; Pierce Protease Inhibitors; Thermo Fisher Scientific) and 1% SDS. Samples were sonicated 3 × 10 s with 1 min on ice between repetitions, and then centrifuged at 13,000 × g for 15 min at 4°C. Protein lysate concentration was determined by Pierce BCA Protein Assay Kit (Life Technologies). Head lysate protein samples (40–60 μg) in reducing sample buffer (50 mM Tris-HCl, pH 6.8; 100 mM DTT; 2% SDS; 0.1% Bromophenol Blue; 10% glycerol) were resolved on 4–20% Criterion TGX Stain-Free Precast Polyacrylamide Gels (Bio-Rad), transferred to nitrocellulose membranes (Bio-Rad), and incubated for 1 h in blocking buffer (5% nonfat dry milk in 0.1% TBS-Tween) followed by overnight incubation with anti-Trio mAb (1:1000; DHSB #9.4A) diluted in blocking buffer. Primary antibody was detected using species-specific horseradish peroxidase conjugated secondary antibody (Jackson ImmunoResearch) with enhanced chemiluminescence (ECL, Sigma). Densitometry analysis was performed using Image Lab software (Bio-Rad). At least three independent biological replicates were performed for each genotype, and significance was calculated using group analysis on GraphPad (Prism).
Drosophila brain dissection, immunohistochemistry, visualization, and statistical analysis.
For Drosophila mushroom body morphology imaging, brains were dissected using #5 Dumont fine forceps (Ted Pella, Inc.) in PBS supplemented with 0.1% Triton X-100 (0.1% PBS-T). The proboscis was removed to provide a forceps grip point, and the remaining cuticle and trachea were peeled away from the brain. Brains were submerged in 1X PBS on ice until all brains were dissected. Dissected brains were fixed in 4% paraformaldehyde for 30 min and then permeabilized in 0.3% PBS-Triton X-100 (0.3% PBS-T) for 30 min, on ice. Brains were carefully transferred to 0.5 ml Eppendorf tubes in 0.1% PBS-T. For both primary and secondary antibody incubations, brains were left rocking at 4°C for 24–72 h (see list of dilutions and incubation times below) in 0.1% PBS-T supplemented with normal goat serum (Jackson ImmunoResearch) at a 1:20 dilution. Immunostained brains were mounted on SuperFrost Plus slides in Vectasheild (Vector Laboratories) using a coverslip bridge. Brains were imaged on a Nikon A1R confocal microscope. Approximately 25 brains were analyzed for each genotype, for each experiment. Maximum intensity projections were generated using Fiji ImageJ software.
Antibody: Mouse monoclonal 9.4A anti-Trio, Incubation time: 48–72 h, Dilution: 1:50
Antibody: Mouse monoclonal 1D4 anti-Fasciclin II, Incubation time: 48–72 h, Dilution: 1:50
Antibody: Rabbit Polyclonal Anti-Green Fluorescent Protein (GFP), Incubation time: Overnight-24 h, Dilution: 0.125:100
Antibody: Alexa Fluor 488 AffiniPure Polyclonal Goat Anti-rabbit IgG, Incubation time: Overnight-24 h, Dilution: 1:100
Antibody: Cy3 AffiniPure Polyclonal Goat Anti-Mouse IgG, Incubation time: Overnight-24 h, Dilution: 1:100
Drosophila neuron live imaging confocal microscopy, neuronal reconstruction, data analysis, and statistical analysis.
Live imaging of class IV ddaC neurons was performed as described previously (Iyer et al., 2012). Briefly, ∼10 wondering third instar ppk-Gal4,UAS-mcd8::GFP-labeled larvae were mounted in 1:5 (vol/vol) diethyl ether: halocarbon oil under an imaging bridge of 22 × 22 mm glass coverslips topped with a 22 × 55 mm glass coverslip. The ddaC images were captured on a Nikon A1R inverted Confocal microscope. Maximum intensity projections were generated using Fiji ImageJ software. Quantitative morphological data were compiled using the Simple Neurite tracer (SNT) plugin for Fiji (Ferreira et al., 2014; Arshadi et al., 2021). Batch processing was completed using a custom Fiji macro and Rstudio script created and gifted by Dr. Atit A. Patel (Dr. Dan Cox Lab, Georgia State University) (Lottes et al., 2024) and the resulting data were exported to Excel (Microsoft).
Statistical analysis.
Group analysis on biological triplicate experiments was performed using one-way or two-way ANOVA (Tukey’s multiple-comparison test) on GraphPad (Prism). Sample sizes (n) and p-values are denoted in text, figures, and/or figure legends and indicated by asterisks (e.g., *, p < 0.05).
Supplementary Material
Acknowledgments
This work was supported by the Emory University Emory Integrated Cellular Imaging Core Facility (RRID:SCR_023534). The authors thank Dr. Dan Cox, GA State Neuroscience Institute, and his lab members Drs. Erin Lottes and Atit Patel for training, reagents, and discussions. We also thank the members of the Moberg and Corbett laboratories, especially Dr. Milo Fasken for insightful discussions.
Abbreviations used:
- GEF
guanine nucleotide exchange factor.
- Nab2
nuclear polyadenosine RNA-binding protein 2
- Pab
polyadenosine binding protein
- RBP
RNA-binding protein
- ZC3H14
zinc finger Cys-Cys-Cys His type containing 14.
Footnotes
This article was published online ahead of print in MBoC in Press (http://www.molbiolcell.org/cgi/doi/10.1091/mbc.E24-04-0150) on July 10, 2024.
REFERENCES
- Agrawal S, Kuo PH, Chu LY, Golzarroshan B, Jain M, Yuan HS (2019). RNA recognition motifs of disease-linked RNA-binding proteins contribute to amyloid formation. Sci Rep 9, 6171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alpert T, Straube K, Oesterreich FC, Herzel L, Neugebauer KM (2020). Widespread transcriptional readthrough caused by Nab2 depletion leads to chimeric transcripts with retained introns. Cell Rep 33, 108496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anderson JT, Wilson SM, Datar KV, Swanson MS (1993). NAB2: a yeast nuclear polyadenylated RNA-binding protein essential for cell viability. Mol Cell Biol 13, 2730–2741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arshadi C, Gunther U, Eddison M, Harrington KIS, Ferreira TA (2021). SNT: a unifying toolbox for quantification of neuronal anatomy. Nat Methods 18, 374–377. [DOI] [PubMed] [Google Scholar]
- Awasaki T, Saito M, Sone M, Suzuki E, Sakai R, Ito K, Hama C (2000). The Drosophila trio plays an essential role in patterning of axons by regulating their directional extension. Neuron 26, 119–131. [DOI] [PubMed] [Google Scholar]
- Ba W, Yan Y, Reijnders MR, Schuurs-Hoeijmakers JH, Feenstra I, Bongers EM, Bosch DG, De Leeuw N, Pfundt R, Gilissen C, et al. (2016). TRIO loss of function is associated with mild intellectual disability and affects dendritic branching and synapse function. Hum Mol Genet 25, 892–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Backer S, Lokmane L, Landragin C, Deck M, Garel S, Bloch-Gallego E (2018). Trio GEF mediates RhoA activation downstream of Slit2 and coordinates telencephalic wiring. Development 145, dev153692. [DOI] [PubMed] [Google Scholar]
- Banerjee A, Apponi LH, Pavlath GK, Corbett AH (2013). PABPN1: molecular function and muscle disease. FEBS J 280, 4230–4250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barbe B, Franke P, Maier W, Leboyer M (1996). Fragile X syndrome. I. An overview on its genetic mechanism. Eur Psychiatry 11, 227–232. [DOI] [PubMed] [Google Scholar]
- Bardoni B, Abekhoukh S, Zongaro S, Melko M (2012). Intellectual disabilities, neuronal posttranscriptional RNA metabolism, and RNA-binding proteins: three actors for a complex scenario. Prog Brain Res 197, 29–51. [DOI] [PubMed] [Google Scholar]
- Bateman J, Shu H, Van Vactor D (2000). The guanine nucleotide exchange factor trio mediates axonal development in the Drosophila embryo. Neuron 26, 93–106. [DOI] [PubMed] [Google Scholar]
- Bateman J, Van Vactor D (2001). The Trio family of guanine-nucleotide-exchange factors: regulators of axon guidance. J Cell Sci 114, 1973–1980. [DOI] [PubMed] [Google Scholar]
- Batisse J, Batisse C, Budd A, Bottcher B, Hurt E (2009). Purification of nuclear poly(A)-binding protein Nab2 reveals association with the yeast transcriptome and a messenger ribonucleoprotein core structure. J Biol Chem 284, 34911–34917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bellanger JM, Lazaro JB, Diriong S, Fernandez A, Lamb N, Debant A (1998a). The two guanine nucleotide exchange factor domains of Trio link the Rac1 and the RhoA pathways in vivo. Oncogene 16, 147–152. [DOI] [PubMed] [Google Scholar]
- Bellanger JM, Zugasti O, Lazaro JB, Diriong S, Lamb N, Sardet C, Debant A (1998b). [Role of the multifunctional Trio protein in the control of the Rac1 and RhoA gtpase signaling pathways]. C R Seances Soc Biol Fil 192, 367–374. [PubMed] [Google Scholar]
- Bienkowski RS, Banerjee A, Rounds JC, Rha J, Omotade OF, Gross C, Morris KJ, Leung SW, Pak C, Jones SK, et al. (2017). The conserved, disease-associated RNA binding protein dNab2 interacts with the fragile X protein ortholog in Drosophila neurons. Cell Rep 20, 1372–1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bircher JE, Koleske AJ (2021). Trio family proteins as regulators of cell migration and morphogenesis in development and disease - mechanisms and cellular contexts. J Cell Sci 134, jcs248393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brais B (2003). Oculopharyngeal muscular dystrophy: a late-onset polyalanine disease. Cytogenet Genome Res 100, 252–260. [DOI] [PubMed] [Google Scholar]
- Briancon-Marjollet A, Ghogha A, Nawabi H, Triki I, Auziol C, Fromont S, Piche C, Enslen H, Chebli K, Cloutier JF, et al. (2008). Trio mediates netrin-1-induced Rac1 activation in axon outgrowth and guidance. Mol Cell Biol 28, 2314–2323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conlon EG, Manley JL (2017). RNA-binding proteins in neurodegeneration: mechanisms in aggregate. Genes Dev 31, 1509–1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cooper TA, Wan L, Dreyfuss G (2009). RNA and disease. Cell 136, 777–793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corbett AH (2018). Post-transcriptional regulation of gene expression and human disease. Curr Opin Cell Biol 52, 96–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corgiat EB, List SM, Rounds JC, Corbett AH, Moberg KH (2021). The RNA-binding protein Nab2 regulates the proteome of the developing Drosophila brain. J Biol Chem 297, 100877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corgiat EB, List SM, Rounds JC, Yu D, Chen P, Corbett AH, Moberg KH (2022). The Nab2 RNA-binding protein patterns dendritic and axonal projections through a planar cell polarity-sensitive mechanism. G3 (Bethesda) 12, jkac100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corley M, Burns MC, Yeo GW (2020). How RNA-binding proteins interact with RNA: Molecules and mechanisms. Mol Cell 78, 9–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crittenden JR, Skoulakis EM, Han KA, Kalderon D, Davis RL (1998). Tripartite mushroom body architecture revealed by antigenic markers. Learn Mem 5, 38–51. [PMC free article] [PubMed] [Google Scholar]
- Currey HN, Kraemer BC, Liachko NF (2023). sut-2 loss of function mutants protect against tau-driven shortened lifespan and hyperactive pharyngeal pumping in a C. elegans model of tau toxicity. MicroPubl Biol 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darnell JC, Richter JD (2012). Cytoplasmic RNA-binding proteins and the control of complex brain function. Cold Spring Harb Perspect Biol 4, a012344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Debant A, Serra-Pages C, Seipel K, O’Brien S, Tang M, Park SH, Streuli M (1996). The multidomain protein Trio binds the LAR transmembrane tyrosine phosphatase, contains a protein kinase domain, and has separate rac-specific and rho-specific guanine nucleotide exchange factor domains. Proc Natl Acad Sci USA 93, 5466–5471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeGeer J, Kaplan A, Mattar P, Morabito M, Stochaj U, Kennedy TE, Debant A, Cayouette M, Fournier AE, Lamarche-Vane N (2015). Hsc70 chaperone activity underlies Trio GEF function in axon growth and guidance induced by netrin-1. J Cell Biol 210, 817–832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Edens BM, Ajroud-Driss S, Ma L, Ma YC (2015). Molecular mechanisms and animal models of spinal muscular atrophy. Biochim Biophys Acta 1852, 685–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fasken MB, Corbett AH, Stewart M (2019). Structure-function relationships in the Nab2 polyadenosine-RNA binding Zn finger protein family. Protein Sci 28, 513–523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ferreira TA, Blackman AV, Oyrer J, Jayabal S, Chung AJ, Watt AJ, Sjostrom PJ, van Meyel DJ (2014). Neuronal morphometry directly from bitmap images. Nat Methods 11, 982–984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Franke P, Barbe B, Leboyer M, Maier W (1996). Fragile X syndrome. II. Cognitive and behavioral correlates of mutations of the FMR-1 gene. Eur Psychiatry 11, 233–243. [DOI] [PubMed] [Google Scholar]
- Fushima K, Tsujimura H (2007). Precise control of fasciclin II expression is required for adult mushroom body development in Drosophila. Dev Growth Differ 49, 215–227. [DOI] [PubMed] [Google Scholar]
- Gebauer F, Schwarzl T, Valcarcel J, Hentze MW (2021). RNA-binding proteins in human genetic disease. Nat Rev Genet 22, 185–198. [DOI] [PubMed] [Google Scholar]
- Goss DJ, Kleiman FE (2013). Poly(A) binding proteins: Are they all created equal? Wiley Interdiscip Rev RNA 4, 167–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Green DM, Marfatia KA, Crafton EB, Zhang X, Cheng X, Corbett AH (2002). Nab2p is required for poly(A) RNA export in Saccharomyces cerevisiae and is regulated by arginine methylation via Hmt1p. J Biol Chem 277, 7752–7760. [DOI] [PubMed] [Google Scholar]
- Grenier St-Sauveur V, Soucek S, Corbett AH, Bachand F (2013). Poly(A) tail-mediated gene regulation by opposing roles of Nab2 and Pab2 nuclear poly(A)-binding proteins in pre-mRNA decay. Mol Cell Biol 33, 4718–4731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gross C, Berry-Kravis EM, Bassell GJ (2012). Therapeutic strategies in fragile X syndrome: Dysregulated mGluR signaling and beyond. Neuropsychopharmacology 37, 178–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guthrie CR, Greenup L, Leverenz JB, Kraemer BC (2011). MSUT2 is a determinant of susceptibility to tau neurotoxicity. Hum Mol Genet 20, 1989–1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guthrie CR, Schellenberg GD, Kraemer BC (2009). SUT-2 potentiates tau-induced neurotoxicity in Caenorhabditis elegans. Hum Mol Genet 18, 1825–1838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haussmann IU, Bodi Z, Sanchez-Moran E, Mongan NP, Archer N, Fray RG, Soller M (2016). m(6)A potentiates Sxl alternative pre-mRNA splicing for robust Drosophila sex determination. Nature 540, 301–304. [DOI] [PubMed] [Google Scholar]
- Hector RE, Nykamp KR, Dheur S, Anderson JT, Non PJ, Urbinati CR, Wilson SM, Minvielle-Sebastia L, Swanson MS (2002). Dual requirement for yeast hnRNP Nab2p in mRNA poly(A) tail length control and nuclear export. EMBO J 21, 1800–1810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heisenberg M (1998). What do the mushroom bodies do for the insect brain? An introduction. Learn Mem 5, 1–10. [PMC free article] [PubMed] [Google Scholar]
- Heisenberg M (2003). Mushroom body memoir: from maps to models. Nat Rev Neurosci 4, 266–275. [DOI] [PubMed] [Google Scholar]
- Iyer SC, Wang D, Iyer EP, Trunnell SA, Meduri R, Shinwari R, Sulkowski MJ, Cox DN (2012). The RhoGEF trio functions in sculpting class specific dendrite morphogenesis in Drosophila sensory neurons. PLoS One 7, e33634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaffe AB, Hall A (2005). Rho GTPases: Biochemistry and biology. Annu Rev Cell Dev Biol 21, 247–269. [DOI] [PubMed] [Google Scholar]
- Jalloh B, Lancaster CL, Rounds JC, Brown BE, Leung SW, Banerjee A, Morton DJ, Bienkowski RS, Fasken MB, Kremsky IJ, et al. (2023). The Drosophila Nab2 RNA binding protein inhibits m(6)A methylation and male-specific splicing of Sex lethal transcript in female neuronal tissue. Elife 12, e64904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones SK, Rha J, Kim S, Morris KJ, Omotade OF, Moberg KH, Myers KR, Corbett AH (2020). The polyadenosine RNA binding protein ZC3H14 is required in mice for proper dendritic spine density. bioRxiv. [Google Scholar]
- Kahsai L, Zars T (2011). Learning and memory in Drosophila: Behavior, genetics, and neural systems. Int Rev Neurobiol 99, 139–167. [DOI] [PubMed] [Google Scholar]
- Kan L, Grozhik AV, Vedanayagam J, Patil DP, Pang N, Lim KS, Huang YC, Joseph B, Lin CJ, Despic V, et al. (2017). The m(6)A pathway facilitates sex determination in Drosophila. Nat Commun 8, 15737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kan L, Ott S, Joseph B, Park ES, Dai W, Kleiner RE, Claridge-Chang A, Lai EC (2021). A neural m(6)A/Ythdf pathway is required for learning and memory in Drosophila. Nat Commun 12, 1458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katrancha SM, Shaw JE, Zhao AY, Myers SA, Cocco AR, Jeng AT, Zhu M, Pittenger C, Greer CA, Carr SA, et al. (2019). Trio haploinsufficiency causes neurodevelopmental disease-associated deficits. Cell Rep 26, 2805–2817.e2809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katrancha SM, Wu Y, Zhu M, Eipper BA, Koleske AJ, Mains RE (2017). Neurodevelopmental disease-associated de novo mutations and rare sequence variants affect TRIO GDP/GTP exchange factor activity. Hum Mol Genet 26, 4728–4740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly S, Pak C, Garshasbi M, Kuss A, Corbett AH, Moberg K (2012). New kid on the ID block: Neural functions of the Nab2/ZC3H14 class of Cys(3)His tandem zinc-finger polyadenosine RNA binding proteins. RNA Biol 9, 555–562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly SM, Bienkowski R, Banerjee A, Melicharek DJ, Brewer ZA, Marenda DR, Corbett AH, Moberg KH (2016). The Drosophila ortholog of the Zc3h14 RNA binding protein acts within neurons to pattern axon projection in the developing brain. Dev Neurobiol 76, 93–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly SM, Leung SW, Apponi LH, Bramley AM, Tran EJ, Chekanova JA, Wente SR, Corbett AH (2010). Recognition of polyadenosine RNA by the zinc finger domain of nuclear poly(A) RNA-binding protein 2 (Nab2) is required for correct mRNA 3’-end formation. J Biol Chem 285, 26022–26032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly SM, Leung SW, Pak C, Banerjee A, Moberg KH, Corbett AH (2014). A conserved role for the zinc finger polyadenosine RNA binding protein, ZC3H14, in control of poly(A) tail length. RNA 20, 681–688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly SM, Pabit SA, Kitchen CM, Guo P, Marfatia KA, Murphy TJ, Corbett AH, Berland KM (2007). Recognition of polyadenosine RNA by zinc finger proteins. Proc Natl Acad Sci USA 104, 12306–12311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kolb SJ, Kissel JT (2011). Spinal muscular atrophy: A timely review. Arch Neurol 68, 979–984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lage K, Hansen NT, Karlberg EO, Eklund AC, Roque FS, Donahoe PK, Szallasi Z, Jensen TS, Brunak S (2008). A large-scale analysis of tissue-specific pathology and gene expression of human disease genes and complexes. Proc Natl Acad Sci USA 105, 20870–20875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee WH, Corgiat E, Rounds JC, Shepherd Z, Corbett AH, Moberg KH (2020). A genetic screen links the disease-associated Nab2 RNA-binding protein to the planar cell polarity pathway in Drosophila melanogaster. G3 (Bethesda) 10, 3575–3583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leeuwen FN, Kain HE, Kammen RA, Michiels F, Kranenburg OW, Collard JG (1997). The guanine nucleotide exchange factor Tiam1 affects neuronal morphology; opposing roles for the small GTPases Rac and Rho. J Cell Biol 139, 797–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lence T, Akhtar J, Bayer M, Schmid K, Spindler L, Ho CH, Kreim N, Andrade-Navarro MA, Poeck B, Helm M, Roignant JY (2016). m(6)A modulates neuronal functions and sex determination in Drosophila. Nature 540, 242–247. [DOI] [PubMed] [Google Scholar]
- Leung SW, Apponi LH, Cornejo OE, Kitchen CM, Valentini SR, Pavlath GK, Dunham CM, Corbett AH (2009). Splice variants of the human ZC3H14 gene generate multiple isoforms of a zinc finger polyadenosine RNA binding protein. Gene 439, 71–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lottes EN, Ciger F, Bhattacharjee S, Timmins EA, Tete B, Tran T, Matta J, Patel AA, Cox DN (2024). CCT and Cullin1 regulate the TORC1 pathway to promote dendritic arborization in health and disease. Cells 13, 1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo S, Tong L (2014). Molecular basis for the recognition of methylated adenines in RNA by the eukaryotic YTH domain. Proc Natl Acad Sci USA 111, 13834–13839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maniatis T, Reed R (2002). An extensive network of coupling among gene expression machines. Nature 416, 499–506. [DOI] [PubMed] [Google Scholar]
- Marfatia KA, Crafton EB, Green DM, Corbett AH (2003). Domain analysis of the Saccharomyces cerevisiae heterogeneous nuclear ribonucleoprotein, Nab2p. Dissecting the requirements for Nab2p-facilitated poly(A) RNA export. J Biol Chem 278, 6731–6740. [DOI] [PubMed] [Google Scholar]
- McKee AE, Silver PA (2007). Systems perspectives on mRNA processing. Cell Res 17, 581–590. [DOI] [PubMed] [Google Scholar]
- McMillan PJ, Strovas TJ, Baum M, Mitchell BK, Eck RJ, Hendricks N, Wheeler JM, Latimer CS, Keene CD, Kraemer BC (2021). Pathological tau drives ectopic nuclear speckle scaffold protein SRRM2 accumulation in neuron cytoplasm in Alzheimer’s disease. Acta Neuropathol Commun 9, 117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morton DJ, Jalloh B, Kim L, Kremsky I, Nair RJ, Nguyen KB, Rounds JC, Sterrett MC, Brown B, Le T, et al. (2020). A Drosophila model of Pontocerebellar Hypoplasia reveals a critical role for the RNA exosome in neurons. PLoS Genet 16, e1008901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Newsome TP, Schmidt S, Dietzl G, Keleman K, Asling B, Debant A, Dickson BJ (2000). Trio combines with dock to regulate Pak activity during photoreceptor axon pathfinding in Drosophila. Cell 101, 283–294. [DOI] [PubMed] [Google Scholar]
- Pak C, Garshasbi M, Kahrizi K, Gross C, Apponi LH, Noto JJ, Kelly SM, Leung SW, Tzschach A, Behjati F, et al. (2011). Mutation of the conserved polyadenosine RNA binding protein, ZC3H14/dNab2, impairs neural function in Drosophila and humans. Proc Natl Acad Sci USA 108, 12390–12395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paskus JD, Herring BE, Roche KW (2020). Kalirin and Trio: RhoGEFs in synaptic transmission, plasticity, and complex brain disorders. Trends Neurosci 43, 505–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patil DP, Pickering BF, Jaffrey SR (2018). Reading m(6)A in the transcriptome: m(6)A-binding proteins. Trends Cell Biol 28, 113–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peng YJ, He WQ, Tang J, Tao T, Chen C, Gao YQ, Zhang WC, He XY, Dai YY, Zhu NC, et al. (2010). Trio is a key guanine nucleotide exchange factor coordinating regulation of the migration and morphogenesis of granule cells in the developing cerebellum. J Biol Chem 285, 24834–24844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pengelly RJ, Greville-Heygate S, Schmidt S, Seaby EG, Jabalameli MR, Mehta SG, Parker MJ, Goudie D, Fagotto-Kaufmann C, Mercer C, et al. (2016). Mutations specific to the Rac-GEF domain of TRIO cause intellectual disability and microcephaly. J Med Genet 53, 735–742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pirozzi F, Tabolacci E, Neri G (2011). The FRAXopathies: Definition, overview, and update. Am J Med Genet A 155A, 1803–1816. [DOI] [PubMed] [Google Scholar]
- Portales-Casamar E, Briancon-Marjollet A, Fromont S, Triboulet R, Debant A (2006). Identification of novel neuronal isoforms of the Rho-GEF Trio. Biol Cell 98, 183–193. [DOI] [PubMed] [Google Scholar]
- Rha J, Jones SK, Fidler J, Banerjee A, Leung SW, Morris KJ, Wong JC, Inglis GAS, Shapiro L, Deng Q, et al. (2017). The RNA-binding protein, ZC3H14, is required for proper poly(A) tail length control, expression of synaptic proteins, and brain function in mice. Hum Mol Genet 26, 3663–3681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson JT, Thorvaldsdottir H, Wenger AM, Zehir A, Mesirov JP (2017). Variant review with the Integrative Genomics Viewer. Cancer Res 77, e31–e34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robinson JT, Thorvaldsdottir H, Winckler W, Guttman M, Lander ES, Getz G, Mesirov JP (2011). Integrative Genomics Viewer. Nat Biotechnol 29, 24–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roman G, Davis RL (2001). Molecular biology and anatomy of Drosophila olfactory associative learning. Bioessays 23, 571–581. [DOI] [PubMed] [Google Scholar]
- Rounds JC, Corgiat EB, Ye C, Behnke JA, Kelly SM, Corbett AH, Moberg KH (2021). The disease-associated proteins Drosophila Nab2 and Ataxin-2 interact with shared RNAs and coregulate neuronal norphology. bioRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rounds JC, Corgiat EB, Ye C, Behnke JA, Kelly SM, Corbett AH, Moberg KH (2022). The disease-associated proteins Drosophila Nab2 and Ataxin-2 interact with shared RNAs and coregulate neuronal morphology. Genetics 220, iyab175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rueden CT, Schindelin J, Hiner MC, DeZonia BE, Walter AE, Arena ET, Eliceiri KW (2017). ImageJ2: ImageJ for the next generation of scientific image data. BMC Bioinformatics 18, 529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santoro MR, Bray SM, Warren ST (2012). Molecular mechanisms of fragile X syndrome: a twenty-year perspective. Annu Rev Pathol 7, 219–245. [DOI] [PubMed] [Google Scholar]
- Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, Preibisch S, Rueden C, Saalfeld S, Schmid B, et al. (2012). Fiji: An open-source platform for biological-image analysis. Nat Methods 9, 676–682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmid M, Olszewski P, Pelechano V, Gupta I, Steinmetz LM, Jensen TH (2015). The nuclear PolyA-binding protein Nab2p is essential for mRNA production. Cell Rep 12, 128–139. [DOI] [PubMed] [Google Scholar]
- Shivalkar M, Giniger E (2012). Control of dendritic morphogenesis by Trio in Drosophila melanogaster. PLoS One 7, e33737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song JK, Giniger E (2011). Noncanonical Notch function in motor axon guidance is mediated by Rac GTPase and the GEF1 domain of Trio. Dev Dyn 240, 324–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soucek S, Zeng Y, Bellur DL, Bergkessel M, Morris KJ, Deng Q, Duong D, Seyfried NT, Guthrie C, Staley JP, et al. (2016). The evolutionarily-conserved polyadenosine RNA binding protein, Nab2, cooperates with splicing machinery to regulate the fate of pre-mRNA. Mol Cell Biol 36, 2697–2714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takemura SY, Aso Y, Hige T, Wong A, Lu Z, Xu CS, Rivlin PK, Hess H, Zhao T, Parag T, et al. (2017). A connectome of a learning and memory center in the adult Drosophila brain. Elife 6, e26975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thelen MP, Kye MJ (2019). The role of RNA binding proteins for local mRNA translation: Implications in neurological disorders. Front Mol Biosci 6, 161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Theler D, Dominguez C, Blatter M, Boudet J, Allain FH (2014). Solution structure of the YTH domain in complex with N6-methyladenosine RNA: A reader of methylated RNA. Nucleic Acids Res 42, 13911–13919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wan L, Dockendorff TC, Jongens TA, Dreyfuss G (2000). Characterization of dFMR1, a Drosophila melanogaster homolog of the fragile X mental retardation protein. Mol Cell Biol 20, 8536–8547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wheeler JM, McMillan P, Strovas TJ, Liachko NF, Amlie-Wolf A, Kow RL, Klein RL, Szot P, Robinson L, Guthrie C, et al. (2019). Activity of the poly(A) binding protein MSUT2 determines susceptibility to pathological tau in the mammalian brain. Sci Transl Med 11, eaao6545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wigington CP, Williams KR, Meers MP, Bassell GJ, Corbett AH (2014). Poly(A) RNA-binding proteins and polyadenosine RNA: New members and novel functions. Wiley Interdiscip Rev RNA 5, 601–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Worpenberg L, Paolantoni C, Longhi S, Mulorz MM, Lence T, Wessels HH, Dassi E, Aiello G, Sutandy FXR, Scheibe M, et al. (2021). Ythdf is a N6-methyladenosine reader that modulates Fmr1 target mRNA selection and restricts axonal growth in Drosophila. EMBO J 40, e104975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu C, Liu K, Ahmed H, Loppnau P, Schapira M, Min J (2015). Structural basis for the discriminative recognition of N6-methyladenosine RNA by the human YT521-B homology domain family of proteins. J Biol Chem 290, 24902–24913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu C, Wang X, Liu K, Roundtree IA, Tempel W, Li Y, Lu Z, He C, Min J (2014). Structural basis for selective binding of m6A RNA by the YTHDC1 YTH domain. Nat Chem Biol 10, 927–929. [DOI] [PubMed] [Google Scholar]
- Yagi R, Mabuchi Y, Mizunami M, Tanaka NK (2016). Convergence of multimodal sensory pathways to the mushroom body calyx in Drosophila melanogaster. Sci Rep 6, 29481. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
This study did not generate any dataset or codes.