Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 May 22.
Published in final edited form as: Curr Biol. 2023 May 11;33(10):2063–2074.e4. doi: 10.1016/j.cub.2023.04.049

Neurobeachin controls the asymmetric subcellular distribution of electrical synapse proteins

E Anne Martin 1,*, Jennifer Carlisle Michel 1, Jane S Kissinger 1, Fabio A Echeverry 2, Ya-Ping Lin 3, John O’Brien 3, Alberto E Pereda 2, Adam C Miller 1,*
PMCID: PMC10266475  NIHMSID: NIHMS1899928  PMID: 37172585

Summary

The subcellular positioning of synapses and their specialized molecular compositions form the fundamental basis of neural circuits. Like chemical synapses, electrical synapses are constructed from an assortment of adhesion, scaffolding, and regulatory molecules, yet little is known about how these molecules localize to specific neuronal compartments. Here we investigated the relationship between the autism- and epilepsy-associated gene Neurobeachin, the neuronal gap junction channel-forming Connexins, and the electrical synapse scaffold ZO1. Using the zebrafish Mauthner circuit we found Neurobeachin localizes to the electrical synapse independently of ZO1 and Connexins. By contrast, we show Neurobeachin is required postsynaptically for the robust localization of ZO1 and Connexins. We demonstrate Neurobeachin binds ZO1 but not Connexins. Finally, we find Neurobeachin is required to restrict electrical postsynaptic proteins to dendrites, but not electrical presynaptic proteins to axons. Together, the results reveal an expanded understanding of electrical synapse molecular complexity and the hierarchical interactions required to build neuronal gap junctions. Further, these findings provide novel insight into the mechanisms by which neurons compartmentalize the localization of electrical synapse proteins and provide a cell biological mechanism for the subcellular specificity of electrical synapse formation and function.

Keywords: Electrical synapse, Neuronal gap junction, Neurobeachin, Zebrafish, Mauthner, Connexin, ZO1

Graphical Abstract

graphic file with name nihms-1899928-f0001.jpg

eTOC_Blurb

Martin et al. show that the autism- and epilepsy-linked Neurobeachin functions in electrical synapse formation to compartmentalize postsynaptic proteins to neuronal dendrites. The findings reveal a mechanism by which neurons selectively assembly electrical synapses with subcellular specificity.

Introduction

Neuronal synapses can be separated into two main classes, chemical and electrical, each of which is required for neural circuit development and function1,2. Chemical synapses are well-studied structures with molecularly diverse asymmetrical junctions, which are known to rely upon highly specialized proteins that orchestrate their localization, assembly, and function3,4 By contrast, electrical synaptic transmission is achieved via neuronal gap junction channels, which in vertebrates are created between neurons by apposed hemichannels of Connexin hexamers that allow for the bidirectional flow of ions and other small molecules5. In addition to channels, electrical synapses contain a diverse proteome including cell adhesion, scaffold, and regulatory molecules, yet the mechanisms required to construct neuronal gap junctions remain poorly understood6-8. Electrical synapses form between all neuronal compartments with functional effects on circuit activity dependent upon each unique coupling arrangement. For example, dendro-dendritic electrical synapses allow for lateral excitation via the spread of local synaptic potentials between neighboring neurons9. Alternatively, axo-axonic electrical synapses can promote strong and highly synchronized firing, and such coupling may contribute to fast ripples such as those aberrantly present in patients with epilepsy1,10-13. Thus, at a cell biological level, the mechanisms specifying electrical synapse subcellular specifity are critical for appropriate neural circuit formation and function.

While neurons control the subcellular locations of neuronal gap junctions, the mechanisms by which neurons compartmentalize electrical synapse proteins are not well understood. Recent work in Caenorhabditis elegans, which use Innexins to form gap junction channels, identified a cAMP-dependent signaling pathway regulating the trafficking of Innexins to distal synaptic sites14. The findings support the notion of regulatory mechanisms controlling the trafficking of electrical synapse proteins to distinct subcellular locations. Yet because Innexins are evolutionarily distinct from Connexins15, it is not clear if vertebrates share related pathways. For chemical synapses, the mechanisms controlling the polarized distribution of synaptic proteins to axons and dendrites are well studied and require the coordination of motor proteins, the cytoskeleton, and numerous adaptors and scaffolds to ensure the fidelity of compartmentalized delivery16-19. Whether there are distinct, or shared, mechanisms to guide the polarized transport of electrical synapse proteins remains unknown.

The formation of electrical and chemical synapses are generally thought to be cell biologically and biochemically distinct processes. Yet in recent years molecules have emerged that suggest links between the development and function of these structures1,2,8,20. One of these proteins, Neurobeachin, was found to be required for both electrical and chemical synapse development20-22. Neurobeachin is a large (~330 kDa) protein that is highly conserved in vertebrates and contains various protein-binding domains including a BEACH domain, AKAP domain, and multiple WD40 domains21. More than 20 de novo variants of Neurobeachin have been identified in patients with autism, intellectual disability, and epilepsy23-26. Previous work suggests Neurobeachin regulates the localization of chemical synapse proteins including glutamate receptors and the scaffolds SAP102 and PSD9521,27-30. Yet its mechanistic role in electrical synapse development is unknown.

Here we explore the contributions of Neurobeachin to electrical synapse formation and examine its biochemical and cell biological functions. We used the stereotyped and identifiable synaptic contacts of the Mauthner cell of larval zebrafish as they are exquisitely accessible to in vivo cell biological analysis. We show that Neurobeachin is localized to the electrical synapse and functions postsynaptically to promote the robust localization of the electrical synapse scaffold Zonula Occludens 1 (ZO1) and neuronal Connexins. We find that Neurobeachin interacts with ZO1 but not the Connexin proteins, supporting a hierarchical model of electrical synapse formation in which Neurobeachin recruits ZO1 which in turn recruits Connexin proteins. Finally, we find that Neurobeachin functions to restrict postsynaptic electrical synapse proteins to dendritic synapses, such that in its absence postsynaptic proteins are inappropriately localized and stabilized at presynaptic locations in the axon. The results presented provide new insight into the molecular complexity and hierarchical interactions that build electrical synapses. Moreoever, the results reveal a mechanism for the asymmetric synaptic localization of electrical synapse components providing a basis for the subcellular specialization of neuronal gap junctions.

Results

Neurobeachin is required for the localization of electrical synapse proteins

To investigate how Neurobeachin regulates electrical synapse development we used the zebrafish Mauthner cell circuit, which uses a combination of electrical and chemical synapses to control a fast escape response31-33. This circuit is composed of two Mauthner cells per fish that receive sensory input from several modalities, including the auditory system, and send output onto motorneurons and interneurons in the spinal cord, including Commissural Local (CoLo) interneurons (Figure 1A,B). Electrical synapses are prominent throughout the circuit, including at mixed electrical/glutamatergic inputs from auditory afferents onto the lateral Mauthner dendrite, called Club Ending (CE) synapses (Figure 1B)34. Additionally, the Mauthner cell uses electrical synapses between its axon and CoLos, called M/CoLo synapses (Figure 1B). These M/CoLo synapses repeat down the length of the spinal cord, with CoLo being present as a single cell in each hemisegement of the ~30 segments of the trunk. Mauthner and CoLo cell morphology can be observed using the transgenic line Et(Tol-056:GFP)35. The CE and M/CoLo electrical synapses are molecularly asymmetric composed of presynaptic Connexin 35.5 hemichannels (Cx35.5 – encoded by the gjd2a gene) that pair with postsynaptic Cx34.1 hemichannels (encoded by the gjd1a gene)36. In addition, the electrical synapse scaffold ZO1b (encoded by the tjp1b gene) localizes exclusively postsynaptically where it binds Cx34.1 (Figure 1B)37-38. At the CE synapses, the Mauthner cell is the postsynaptic partner and uses ZO1b and Cx34.1 to build electrical synapses, while at the M/CoLo synapses, the Mauthner cell is presynaptic and uses Cx35.5 (Figure 1B). The asymmetry of Mauthner cell electrical synapses highlights the cell biological requirement of the neuron to deliver unique electrical synaptic proteins to distinct neuronal compartments.

Figure 1: Neurobeachin is required for the localization of electrical synapse proteins.

Figure 1:

(A) Dorsal view of the larval zebrafish illustrating location of the two Mauthner cells (green), anterior left. Boxed region indicates the region shown in B. (B) Simplified illustration of the Mauthner circuit, anterior up, highlighting the Club Ending (CE) and M/CoLo synapses and the known protein localization of electrical synapse components Cx35.5 (orange), Cx34.1 (cyan), and ZO1b (magenta). The center dashed-circle diagram depicts the standard synapse organization for each Mauthner synapse which includes gap junction hemichannels composed of Cx35.5 in the presynapse, and Cx34.1 in the postsynapse where it binds scaffolding protein ZO1b. Arrows in Mauthner soma indicate the compartmentalization of the indicated electrical synapse proteins. (C-H) Confocal images of Mauthner electrical synapses in 5 days post fertilization Et(Tol-056:GFP) transgenic wildtype (WT) or nbeaa−/− mutants. Panels show staining for GFP labeling the Mauthner cell (green), Cx35.5 (orange), Cx34.1 (cyan), V5-ZO1b (magenta) at (C-E) CE synapses and (F-H) M/CoLo synapses. Dashed circles identify locations of Mauthner and CoLo contact. Images in this figure and other figures are maximum intensity projections of ~12μm at CE and ~20μm at M/CoLo synapses. (I,J) Quantification of indicated electrical synapse protein fluorescence intensities, normalized to WT, at (I) CE synapses and (J) M/CoLo synapses in WT and nbeaa−/− mutant animals. Mean ± SEM are shown, all comparisons were made between genotyped WT and mutant siblings. For I, n = 5, 5, 5, 5, 10, and 12 fish for each bar shown, left to right. Two Mauthner cells were analyzed per fish and averaged to represent a single value for each fish. For graph in J, n = 6, 6, 6, 6, 10, and 10 fish, for each bar shown, left to right. 20-24 M/Colo synapses were analyzed and averaged for each fish. ** indicates p < 0.01, *** indicates p < 0.001, **** indicates p < 0.0001 by unpaired t-test. Scale bars are as indicated. See also Figure S1.

We first examined the role of Neurobeachin (encoded by the nbeaa gene) in localizing the presynaptic Cx35.5 and the postsynaptic Cx34.1 and ZO1b to Mauthner cell electrical synapses (Figure 1C-J). We previously found that Neurobeachin is required for Connexin localization to Mauthner cell electrical synapses20. However, the molecular identity of the Connexins (Cx35.5/gjd2a and Cx34.1/gjd1a), the ZO1b/tjp1b scaffold, and the molecularly asymmetric distribution of these proteins at the synapse, were all unknown at the time. Therefore, we examined how the loss of Neurobeachin impacted the localization of all three of these electrical synapse components by comparing 5 days post fertilization (dpf) wildtype and nbeaafh364/fh364 mutants (hereafter called nbeaa−/−20). First, we tested Cx35.5 and Cx34.1 localization at CE (Figure 1C,D,I) and M/CoLo synapses (Figure 1F,G,J). In nbeaa−/− mutants, we observed similar decreases of both pre- and postsynaptic Connexin proteins at electrical synapses. Next, using a transgenic line that marks endogenous ZO1b with a V5 epitope (tjp1bb1406, hereafter called Pt(V5-ZO1b)38), we examined 5 dpf wildtype and nbeaa−/− mutant fish for ZO1b at CE (Figure 1E,I) and M/CoLo synapses (Figure 1H,J). We observe a similar reduction of ZO1b at Mauthner dendritic and axonal electrical synapses in nbeaa−/− mutant fish. While nbeaa−/− mutant Mauthner cell dendrites have reduced complexity20, we observed no gross morphological defects in the primary lateral or ventral dendrites that emerge from the soma, nor in the axons that course down the spinal cord (Figure S1A), suggesting that overall neuronal development and compartmentalization remains intact. Further, we observe that CE and M/CoLo synapses are still present in their stereotypical locations (Figure 1C-H), yet the amount of Connexins and ZO1 is significantly reduced (Figure 1I,J), suggesting that appropriate interneuronal contacts are made in mutants, yet there is a defect in robustly localizing synaptic proteins. The diminished synaptic localization is not due to decreased overall Connexin and ZO1 protein abundance, as similar protein levels are present in wildtype and nbeaa−/− mutants (Figure S1B-D). Together these results support the notion that nbeaa−/− mutants have defects in the localization of electrical synapse proteins. We conclude that loss of Neurobeachin function affects localization of pre- and postsynaptic Connexins and the postsynaptic scaffold ZO1.

Neurobeachin localizes to electrical synapses independent of ZO1 and Connexins

In rat cerebellar neurons, Neurobeachin localizes to the trans side of the Golgi, on tubulovesicular vesicles in dendrites, and at the postsynapse of glutamatergic chemical synapses21. Thus, we next examined Neurobeachin localization using the Mauthner cell circuit. At 5 dpf, we observe Neurobeachin localization in the Mauthner cell soma, yet within this compartment it showed little colocalization with electrical synapse proteins (Figure S2A,B). To examine Neurobeachin’s somatic distribution, we stained for GM130/Golga2, a cis-Golgi apparatus marker, and observed the Mauthner cell of wildtype and nbeaa−/− mutants. Golgi staining within the Mauthner soma was found throughout the compartment but asymmetrically biased towards the axon; this distribution was not altered in nbeaa−/− mutants (Figure S2C,D). In addition, we noticed a high degree of close proximity localization between Neurobeachin and GM130 as expected for colocalized proteins. This pattern is similar to what has been observed in rodents21,29.

We next examined Neurobeachin localization at Mauthner electrical synapses. At the CE synapses on Mauthner’s lateral dendrite, we see Neurobeachin localization at distinct foci adjacent to the electrical synapse proteins ZO1b and Cx34.1, as well as a diffuse staining colocalized across the extent of the observed gap junction staining (Figure 2A). This pattern of discrete localization surrounding and at times overlapping the Connexin/ZO1 staining is reminiscent of what has been described for Neurobeachin with chemical postsynaptic scaffolds in mammalian cultured neurons29. We also observe Neurobeachin staining at M/CoLo synapses, with staining appearing adjacent to ZO1 and Connexins, and we never observe staining within the prominent Mauthner axon between each synapse (Figure 2B). We note that at each Mauthner associated synapse (CE or M/CoLo) the pre- or postsynaptic localization of Neurobeachin cannot be resolved due to the limits of light microscopy — we address where the protein functions below. Neurobeachin staining is eliminated in nbeaa−/− mutants supporting the specificity of the antibody (Figure 2C-E, Figure S2B). We conclude that Neurobeachin can localize to electrical synapses, yet does so in a pattern that is distinct from the ZO1 scaffold and neural Connexins.

Figure 2: Neurobeachin localization to electrical synapses is independent of ZO1b and Connexins.

Figure 2:

(A-D) Confocal images of Mauthner electrical synapses in 5 days post fertilization Et(Tol-056:GFP) transgenic wildtype (WT, A,B) and nbeaa−/− (C,D) zebrafish. Panels show staining for GFP labeling the Mauthner cell (green), Cx34.1 (cyan), ZO1 (magenta), and Neurobeachin (yellow) at (A,C) CE synapses, and (B,D) M/CoLo synapses. Dashed circles identify locations of Mauthner and CoLo contact. BI, BII and DI, DII represent two independent M/CoLo synapses for each genotype. (E) Quantification of Neurobeachin fluorescence intensities at CE synapses comparing wildtype (WT, n=5) and nbeaa−/− mutants (n=6). ** indicates p < 0.01 by Mann Whitney. (F-G) Neurobeachin localization at Mauthner CE synapses in 5 dpf Et(Tol-056:GFP) transgenic zebrafish from (F) wildtype (WT, top)_and ZO1b−/− mutant animals (bottom), and (G) wildtype (WT, top) and Cx34.1−/− mutant animals (bottom) with markers and scale bars as indicated. Quantification of Neurobeachin fluorescence intensities at CE synapses in (F) compares wildtype (WT, n=5) and ZO1b−/− mutants (n=5). ns indicicates not significant by t-test. Quantification of Neurobeachin fluorescence intensities at CE synapses in (G) compares wildtype (WT, n=5) and Cx34.1−/− mutants (n=5). ns indicicates not significant by t-test.For all quantification, two Mauthner cells were analyzed per fish and averaged to represent a single value for each fish. Mean ± SEM are shown and values are normalized to WT sibling controls for each experiment. See also Figure S2.

Next we examined whether Neurobeachin localization to the electrical synapse was dependent upon the function of other electrical synapse proteins. The intracellular scaffold ZO1b is required for Connexin localization, but ZO1b largely localizes to electrical synapses independently of the Connexins38. We therefore examined Neurobeachin localization in ZO1b and Connexin mutants. We examined ZO1b/tjp1bb1370/b1370 37 and Cx34.1/gjd1abfh436/fh436 36 mutants and found that Neurobeachin localizes to electrical synapses in a manner similar to their wildtype siblings (Figure 2F-G). We conclude that Neurobeachin localizes to the electrical synapse independent of ZO1b and Connexins.

Neurobeachin binds the electrical synapse scaffold ZO1b but not the Connexins

Neurobeachin is thought to orchestrate the delivery of neurotransmitter receptors to the glutamatergic chemical synapse through binding with chemical synapse scaffolds such as membrane associated guanylate kinase (MAGUK) family members PSD95 and SAP10230,39. Given that the electrical synapse scaffold ZO1b is also a member of the MAGUK family, we next examined Neurobeachin/ZO1b interactions. We attempted to clone full-length nbeaa sequences from fish, but repeated and varied attempts prevented the recovery of the entire coding sequence. We were able to generate N- and C-terminal fragments of Neurobeachin, each tagged with mVenus (Figure 3A), and used heterologous expression in HEK293T cells to test for interactions with previously cloned zebrafish ZO1b and neural Connexins38. We first co-transfected ZO1b with mVenus-tagged Neurobeachin fragments and performed coimmunoprecipitation (coIP) assays. We found that the C-terminal fragment of Neurobeachin pulls down ZO1 (Figure 3B-C). We next performed the reciprocal coIP experiment and co-transfected Flag-tagged-ZO1b with the mVenus-tagged Neurobeachin fragments and found that Flag-tagged ZO1b robustly pulls down the C-terminal fragment of Neurobeachin (Figure 3D,E). We also observe a faint, yet consistent, band in both IP experiments for ZO1b and N-terminal Neurobeachin fragment interactions (Figure 3C-E). Finally, we tested whether Neurobeachin could bind the pre- or postsynaptic Connexins. We co-transfected each Connexin with N-terminal and C-terminal Neurobeachin and find no evidence of pull down with Neurobeachin in immunoprecipitates (Figure 3F-I). By contrast, we find robust interaction between ZO1b and both Connexins, as previously shown38. We conclude that the C-terminal fragment of Neurobeachin can bind the postsynaptic electrical synapse scaffold ZO1b in vitro.

Figure 3: Neurobeachin binds the postsynaptic electrical synapse scaffold ZO1b, but not the neural Connexins, in vitro.

Figure 3:

(A) Schematic of Neurobeachin protein indicating domains included in mVenus-tagged N- and C-terminal fragments. (B) HEK293T/17 cells were transfected with plasmids to express ZO1b and either mVenus (lane 1), mVenus-tagged N-terminal-Neurobeachin (lane 2), or mVenus-tagged C-terminal-Neurobeachin (lane 3). Lysates were immunoprecipitated with anti-GFP antibody, which recognizes mVenus, and analyzed by immunoblot for the presence of ZO1b using ZO1 antibody (upper), or mVenus-containing proteins using GFP antibody (middle). Total extracts (bottom) were blotted for ZO1 to demonstrate expression and antibody recognition of expressed proteins. (C) Quantification of three representative blots in B, normalized to the strongest band. (D) HEK293T/17 cells were transfected with plasmids to express Flag-ZO1b and either mVenus (lane 1), mVenus-tagged N-terminal-Neurobeachin (lane 2), or mVenus-tagged C-terminal-Neurobeachin (lane 3). Lysates were immunoprecipitated with anti-Flag antibody and analyzed by immunoblot for the presence of mVenus-containing proteins using GFP antibody (upper), or ZO1b using ZO1 antibody (middle). Total extracts (bottom) were blotted for GFP to demonstrate expression and antibody recognition of expressed proteins. (E) Quantification of three representative blots in D, normalized to the strongest band. (F-I) HEK293T/17 cells were transfected with plasmids to express Cx35.5 (F) or Cx34.1 (H) and either mVenus-tagged N-terminal Neurobeachin (lane 1), mVenus-tagged C-terminal-Neurobeachin (lane 2), or mVenus-tagged ZO1b (lane 3). Lysates were immunoprecipitated with anti-GFP antibody and analyzed by immunoblot (F) for the presence of Cx35.5 using a Cx35.5 specific antibody or (H) Cx34.1 using a Cx34.1 specific antibody (upper), mVenus-containing proteins using GFP antibody (middle). Total extracts (bottom) were blotted for (F) Cx35.5 or (H) Cx34.1 to demonstrate expression and antibody recognition of expressed proteins. (G) Quantification of three representative blots in F, normalized to the strongest band. (I) Quantification of three representative blots in H, normalized to the strongest band. Mean ± SEM are shown and n = 3 for all blots.

Neurobeachin is required postsynaptically for electrical synapse protein localization

Observing that Neurobeachin binds ZO1b, and given that ZO1b is a postsynaptic scaffold38, we next wanted to examine whether Neurobeachin functioned postsynaptically in vivo. To achieve this, we generated chimeric animals via blastula transplantation40 to create larvae with Mauthner circuit neurons derived from distinct genotypes (Figure 4A). We first examined the localization of ZO1b within the Mauthner cell by using donor embryos that were Et(Tol-056:GFP); Pt(V5-ZO1b) double transgenics, with the former transgene labeling Mauthner and CoLo cells with GFP, and the latter tagging endogenous ZO1b protein with a V5 epitope tag. The host embryos were non-transgenic wildtypes while the Et(Tol-056:GFP); Pt(V5-ZO1b) donor embryos were either homozygous wildype at the nbeaa locus (nbeaa+/+) or homozygous mutant (nbeaa−/−). This allows us to examine the effects of the loss of Neurobeachin from the Mauthner cell. We focused on chimeric animals in which individual Mauthner cells were derived from the donor (GFP+) and found V5-ZO1b present at CE synapses in wildtype control transplants (nbeaa+/+ to WT), while in nbeaa−/− mutant Mauthner cells in a wildtype host (nbeaa−/− to WT), V5-ZO1b staining was reduced (Figure 4B).

Figure 4: Neurobeachin is required postsynaptically for the proper dendritic compartmentalization of ZO1b and Cx34.1.

Figure 4:

(A) Diagram illustrating 5 days post fertilization chimera in which a Mauthner cell is derived from the GFP+ transgenic donor (green) while the other neurons of the circuit are derived from the GFP-host (gray). Boxed outline 1: Location of CE synapses shown by representative confocal images in B and C. Boxed outline 2: Location of M/CoLo synapses shown by representative confocal images in D and E. (B-E) Confocal images of GFP+ Mauthner cells in chimeric animals. (B,D) Chimeras generated by transplanting cells from Et(Tol-056:GFP); Pt(V5-ZO1b) double transgenic donors into a wildtype (WT) host. (C,E) Chimeras were generated by transplanting cells from Tg(Cx34.1-GFPAE37) transgenic donors into a WT host. Left images show transplants from a nbeaa+/+ animal into a WT host while right images show transplants from a nbeaa−/− mutant into a WT host. Mauthner CE synapses (B,C) and M/CoLo synapses (D,E) are identified at stereotypical positions in the hindbrain and spinal cord of the GFP+ Mauthner cell, respectively. Panels show staining for GFP labeling the Mauthner cell (green in B,D) or Cx34.1-oxGFP (cyan in C,E), V5-ZO1b (magenta), and Cx35.5 (orange). Outline of Mauthner cell lateral dendrite shown by green dashed line in (B). M/CoLo sites of contact shown by white dashed circles in (D,E). White arrowheads denote aberrant presynaptic V5-ZO1b or Cx34.1 staining in transplanted nbeaa−/− Mauthner cells. (F-K) Quantification of synaptic protein localization at M/CoLo synapses in control nbeaa+/+ to WT (WT) and in nbeaa−/− to WT (nbeaa−/−) transplants for V5-ZO1b (F-H) or Cx34.1-GFP (I-K). Fluorescence intensity is quantified and graphed as fold change comparing protein localization associated with the transplanted Mauthner cell to that of the host synapses within the same animal. (F,I) Each dot represents the average fold change for all synapses in an animal. (G,J) Each indicator represents the fold change for anterior segments (first eight segments imaged) and posterior segments (next eight segments imaged) with a line connecting values for an individual animal. (H,K) Each dot represents the average fold change for anterior or posterior segment synapses in an animal. Mean ± SEM are shown. For (F-H) wt, n = 5, nbeaa−/−, n = 5. For (I-K) wt, n = 5, nbeaa−/−, n = 5. In (F,I) ** indicates p < 0.01 by unpaired t-test. In (H,K) * indicates p < 0.05, ** indicates p < 0.01, and *** indicates p < 0.001 by ANOVA. ant = anterior synapses, pos = posterior synapses. Scale bars are as indicated. See also Figure S3.

In the next set of transplants, we examined the effect of removing Neurobeachin from Mauthner on Connexin localization. For the transplant donor cells, we used the Tg(Cx34.1-GFPAE37) transgenic line that labels Cx34.1 with GFP. At the dendritic CEs of nbeaa−/− mutant Mauthner neurons, we observe that the localization of postsynaptic Cx34.1-GFP and presynaptic Cx35.5 (labeled by antibody) are both reduced as compared to wildtype transplants (Figure 4C). These data show that the loss of Neurobeachin from the transplanted postsynaptic neuron affects the localization of the dendritically localized ZO1b and Cx34.1. In addition, the removal of Neurobeachin from Mauthner affects presynaptic Cx35.5 localization in the neighboring auditory afferent neuron. We conclude Neurobeachin is required cell autonomously in the postsynaptic neuron to localize ZO1b and Cx34.1, and is also required cell non-automously for presynaptic Cx35.5 localization.

Neurobeachin is required to restrict postsynaptic electrical synapse proteins to the dendritic compartment

We next took advantage of the fact that the Mauthner cell is both a postsynaptic partner at the CEs as well as the presynaptic partner at the M/CoLo electrical synapses. This configuration, in conjunction with the chimeric transplant experiments, allows us to visualize the compartmentalized localization of components of the electrical synapse (Figure 1A). For these experiments, as above, we focused on chimeric animals in which individual Mauthner cells were derived from the GFP+ donor. In control transplant experiments from Et(Tol-056:GFP); Pt(V5-ZO1b) to non-transgenic wildtypes (nbeaa+/+ to WT), the V5-tagged ZO1b is exclusively localized at CE synapses and is not present at M/CoLo synapses (Figure 4B,D, left panels). Thus, within the Mauthner cell, ZO1b is compartmentalized exclusively postsynaptically to the dendritic synapses and excluded from localizing presynaptically within the axon. Strikingly, in transplanted nbeaa−/− mutant Mauthner cells (nbeaa−/− to WT), we observed localization of V5-ZO1b presynaptically at M/CoLo synapses (Figure 4D, right panels, quantified in 4F). The Mauthner cell axon extends the length of the spinal cord and it makes en passant electrical synapses with CoLo neurons found in each hemi-segment. The mislocalized V5-ZO1b appears strongly at M/CoLo synapses found in the anterior half of the spinal cord, and diminishes at synapses found posteriorly (Figure 4G,H). We conclude that Neurobeachin is required to restrict ZO1b localization to the dendritic compartment and prevent it from localizing at axonal electrical synapses.

We next questioned whether the loss of Neurobeachin from the Mauthner cell would affect Connexins. In control Tg(Cx34.1-GFPAE37) transplants to non-transgenic wildtypes (nbeaa+/+ to WT), where Mauthner cells were derived from the donor, Cx34.1-GFP is exclusively found compartmentalized to the CE synapses and is excluded from M/CoLo synapses (Figure 4C,E, left panels). By contrast, we observe that transplanted nbeaa−/− mutant Mauthner cells (nbeaa−/− to WT) mislocalize Cx34.1 presynaptically at axonal M/CoLo synapses (Figure 4E, right panels, quantified in 4I). In addition, analogous to our V5-ZO1b transplant experiments, this mislocalization of Cx34.1 appears strongly at anterior M/CoLo synapses and diminishes posteriorly (Figure 4J,K). In the transplants in which Mauthner cells are nbeaa−/− mutant, and the CoLo neurons are derived from the wildtype host (i.e. they are GFP- and therefore nbea+/+), we observed that the Cx35.5 staining at M/CoLo synapses is not affected (Figure 4D,E, right panels). This result suggests that Neurobeachin does not function presynaptically to localize Connexins. To test this directly, we examined whether presynaptic Cx35.5 would mislocalize to dendritic postsynaptic CE synapses in nbeaa−/− mutants (Figure S3A-D). We used CRISPR to V5-tag endogenous Cx35.5 protein and examined injected mosaic F0 animals for the localization of Cx35.5-V5 in wildtype and nbeaa−/− mutants. In wildtype, we identified injected animals in which all M/CoLo synapses have V5 staining, as expected for the presynaptic usage of Cx35.5 in the Mauthner axon; in such cases, no Cx35.5-V5 staining was observed at CE synapses (Figure S3A,B). In nbeaa−/− mutants, we found animals with the same axonal distribution of Cx35.5-V5 present at all M/CoLo synapses (Figure S3D), however the amount localized was reduced as expected in nbeaa−/− mutants (compare to Figure 1F). Critically, in nbeaa−/− mutants, no Cx35.5-V5 staining is observed at CE synapses (Figure S3C). We conclude that Neurobeachin is not required for axonal Cx35.5 compartmentalization, but is required to restrict Cx34.1 to the dendrite.

Taken together, we find that the selectively removing Neurobeachin from the Mauthner cell leads to a reduction of ZO1b and Cx34.1 at the dendritic CEs, and that these proteins are mislocalized to the axonal M/CoLo presynapses. By contrast, presynaptic Cx35.5 in the Mauthner cell is unaffected by the removal of Neurobeachin. Thus, we conclude that Neurobeachin functions specifically to constrain dendritically localized synaptic components to their appropriate compartment, thereby supporting the subcellular specificity of electrical synapse formation (Figure 5).

Figure 5: Model of Neurobeachin function in electrical synapse formation.

Figure 5:

Diagram showing proposed Neurobeachin (yellow) roles in regulating the hierarchical assembly and compartmentalization of Mauthner neuron electrical synapse proteins.

Discussion

The experiments described here investigating Neurobeachin provide new biochemical and cell biological insight into the development of electrical synapse molecular organization. First, the findings expand the understanding of the hierarchical construction of the electrical synapse beyond the simplistic view of electrical synapses composed solely of Connexin gap junction channels. We find that Neurobeachin localizes to the electrical synapse independent of ZO1b or Connexins, yet Neurobeachin is necessary for these proteins to robustly localize to the developing synapse. Furthermore, we have identified a biochemical connection between Neurobeachin and ZO1b, and previous work found that ZO1b in turn binds Cx34.138. Thus, our emerging model is that Neurobeachin sits at the top of an electrical synapse hierarchy, directing ZO1b to the synapse, which in turn localizes Connexin to build the neuronal gap junction. This new model expands the molecular ‘borders’ of an electrical synapse and suggests that neuronal gap junction channels are the output of complex molecular interactions, including Neurobeachin, that regulate electrical synapse formation, analogous to chemical synaptogenesis. We further show that Neurobeachin functions postsynaptically in building the electrical synapse, and so why are presynaptic Cx35.5 proteins affected when Neurobeachin is mutant in all cells of the animal? We have observed that the removal of Cx34.1 from only the postsynaptic neuron results in a decrease in presynaptic Cx35.5 localization in the adjacent, coupled neuron36. Further, removing ZO1b from only the postsynaptic neuron cell autonomously reduces the localization of postsynaptic Cx34.1, and non-cell autonoumously affects presynaptic Cx35.5 localization38. Thus, we hypothesize that eliminating Neurobeachin function in all cells results first in the reduction of postsynaptic ZO1b/Cx34.1 localization, which then in turn results in the reduction of Cx35.5 presynaptically. This model is supported by the transplant experiments in which Neurobeachin function is specifically removed from the Mauthner neuron. These results add further support to the notion that electrical synapses, like chemical synapses, are molecularly complex synaptic compartments, that can have asymmetric biochemical compositions with molecular functions that have both cell autonomous and non-cell autonomous effects on building the neuronal gap junction channels.

While clear evidence of electrical synapse subcellular specificity exists in invertebrate and vertebrate neural circuits9-13,41, the mechanisms to achieve this are largely unknown. The results presented here establish that Neurobeachin is required for the compartmentalized synaptic localization of postsynaptic, but not presynaptic, electrical synapse proteins. These findings identify Neurobeachin as the first molecular scaffold to regulate the subcellular specificity of vertebrate electrical synapses. Given that Neurobeachin localizes on vesicular structures near the Golgi, in the dendrites, and also at electrical synapses, it is tantalizing to suggest that it directs synaptic components, such as a vesicular-associated ZO1b/Cx34.1 co-transported complex, to the postsynaptic site. We note that in nbeaa−/− mutant transplants the mislocalized postsynaptic proteins are most strongly concentrated at axonal synapses near the cell soma, and diminish at more distal synapstic sites. We suspect that in nbeaa−/− mutant cell axons, ZO1b/Cx34.1 that is found mislocalized to the axon may be in limiting quantities insufficient to localize to all M/CoLo synapses down the length of the spinal cord. Alternatively, these normally dendritically restricted proteins may not competently engage with the axonal transport machinery. We are currently unable to observe electrical synapse proteins while they are being trafficked within dendrites and axons. Instead, at this time, we can only view these proteins once they have accumulated at synaptic contacts. Therefore, we cannot exclude the possibility that in wildtype neurons, electrical synapse proteins are trafficked into both compartments and postsynaptic proteins are then stabilized at their appropriate synaptic contacts by interactions with Neurobeachin. Future technical improvements in detection and imaging will be required to assess the trafficking pathways taken by these, and other, electrical synapse proteins. Despite this, our results clearly demonstrate that Neurobeachin is required to restrict postsynaptic ZO1b and Cx34.1 to their normal dendritic locations, but it does not have a role in localizing presynaptic Cx35.5 to its axonal location. Taken together, we propose a model in which Neurobeachin is necessary for the trafficking and/or stabilization of postsynaptic proteins to the electrical synapse (Figure 5).

How might Neurobeachin direct electrical synapse proteins to specific compartments? Neurobeachin contains an A-Kinase Anchoring Protein (AKAP) domain which could promote the formation of a synaptic PKA signaling complex to phosphorylate electrical synapse proteins upon cAMP stimulation21,42. Phosphorylation can alter gap junction processes such as Connexin trafficking, assembly, degradation, and channel function43. Additionally, recent findings in C. elegans reveal that cAMP signaling is necessary for the trafficking of Innexins to specific subcellular regions within neurons14. Thus, it is enticing to speculate that Neurobeachin may be the cell biological link between cAMP signaling and the compartmentalized trafficking of electrical synapse components, and such experiments await further testing in both vertebrate and invertebrate neurons. Overall, these emerging pieces of evidence imply that vertebrate and invertebrate neurons, despite differences in the Connexin and Innexin building blocks of gap junctions, may share mechanisms in building and compartmentalizing electrical synapses within neural circuits.

Why would a neuron compartmentalize electrical synapse components? The localization of electrical synapses to distinct subcellular compartments has critical impacts on neuronal function, such as lateral excitation via dendro-dendritic electrical synapses9 or strong synchronization via axo-axonic electrical synapses10-13. These configurations require the compartmentalized delivery or capture of electrical synapse components at distinct subcellular sites. In the Mauthner cell, such compartmentalization creates molecularly asymmetric electrical synapses, with unique pre- and postsynaptic components36-37. Molecular asymmetry is an important feature of electrical synapses which can determine functional properties, for instance it is often associated with electrical rectification44-45. In nbeaa−/− neurons, postsynaptic proteins are inappropriately localized presynaptically, co-mingling with the normal presynaptic proteins at the axonal presynapse. The resulting synapse is therefore more symmetric, with ZO1 and Cx34.1 localized on both sides of the synapse, which could alter the functional properties of the circuit. Thus, Neurobeachin represents a cell biological mechanism by which neurons can direct electrical synapse components to appropriate locations, building unique synaptic structures at subcellular localizations to articulate specific circuit functions.

While here we have focused on the electrical synapse, it is intriguing to note that Neurobeachin is necessary for both electrical and chemical synapse development20-22. Here we show that Neurobeachin binds to the electrical synapse scaffold ZO1, while previous work showed that it can bind to chemical synapses scaffolds including PSD95 and SAP10230,39. Indeed, electrical and chemical synapse formation are well recognized to be intimately coordinated during development1-2,8. Neurobeachin therefore presents itself as an intriguing biochemical bridge uniting synapse formation and invites an essential question of whether or not this protein uses similar or distinct mechanisms to regulate these molecularly diverse structures. Previous work shows that Neurobeachin co-localizes with chemical synapse scaffolding molecules like SAP102 along dendrites as well as at the synapse39. Upon Neurobeachin loss, chemical synapse receptors do not localize to the cell surface, but instead accumulate within the cell soma29. Yet whether Neurobeachin functions to restrict postsynaptic chemical synapse proteins to the dendrite, as we have shown here for electrical synapse proteins, remains to be examined. Additionally, Neurobeachin mutations have been identified in patients with autism, intellectual disability, and epilepsy, with more than 20 variants occurring broadly across the recognized domains of the protein including throughout the C-terminal domain23-26, and our results here found this region is required to bind the electrical synapse scaffold ZO1b. Thus, understanding the structure/function relationship of how Neurobeachin coordinates electrical and chemical synapse formation will reveal insight into these neurodevelopmental disorders and provide potential therapeutic targets. Altogether, this work illuminates a biochemical and cell biological mechanism at the nexus of balancing electrical and chemical synaptogenesis and opens a window for future investigations underlying these fundamental processes.

STAR Methods

RESOURCE AVAILABILITY

Lead contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Adam C. Miller (acmiller@uoregon.edu).

Materials availability

Plasmids generated in this study are available upon request.

Data and code availability

  • Microscopy and other data reported in this paper will be shared by the lead contact upon request.

  • No original code is reported in this study.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS

Zebrafish

Zebrafish (Danio rerio) were bred and maintained with approval from the Institutional Animal Care and Use Committee (IACUC AUP 21-42) within the University of Oregon fish facility. Fish were kept at 28.5°C on a 14 hr on and 10 hr off light cycle and the developmental timepoints used originated from standard developmental staging31. Animals were housed in groups according to genotype with a limit of 30 animals per tank. All fish for this project originated from the ABC background and most fish had the enhancer trap transgene Et(Tol-056:GFP) in the background, which labels the Mauthner and CoLo neurons with GFP35, unless otherwise noted. Mutant lines were genotyped for all experiments. At this stage of development, zebrafish sex is not yet determined46.

Generation of Cx34.1-oxGFP fish

To generate the fish in which Cx34.1 is tagged with a fluorescent protein, Cx34.1 was tagged with oxGFP, which is optimized for use in oxidizing environments and biological membranes47. A Cx34.1 coding region expression plasmid was made using zebrafish genomic BAC clone CH211-87N9. The fluorescent protein coding region was inserted 21 amino acids before end of the C-terminus at a location that was previously found not to disrupt regulation of coupling by Connexin phosphorylation and not to block the C-terminal PDZ-domain interaction motif48. Promoter elements of the Connexin gene were derived from the same BAC clone. The transgene construct also includes a mCherry cardiac reporter for ease of identification in injected embryos. The transgenic fish line (Cx34.1oxGFPAE37) was generated at the Zebrafish Core of the Albert Einstein College of Medicine using the Tol2 transposon system. The localization of Cx34.1-oxGFP was found at the expected electrical synapses throughout the brain, and particularly at CE and M/CoLo synapses of the Mauthner circuit as shown in this manuscript, using double-labeling with anti-Cx34.1 and anti-GFP. Moreover, electrical transmission at Mauthner CE synapses was found to be indistinguishable from that of WT fish during electrophysiological recordings (data not shown).

Cas9-mediated genome engineering of Cx35.5-V5

To generate fish at which endogenous Cx35.5 is tagged with V5 epitope tag, a single guide RNA (sgRNA) targeting exon 2 of the endogenous gjd2a coding sequence was designed using the CRISPRscan algorithm49 and synthesized as previously described50. The sgRNA was generated using the T7 megascript kit (ThermoFisher, AMB13345). The target site, including PAM, is: 5’-CCCAATTTCGGCCGCACTCAGTC-3’. The gjd2a-V5 single stranded donor oligo (ssODN) was designed to repair into the endogenous gjd2a locus 42 bp prior to the stop codon and was synthesized complementary to the coding strand. This location was previously found not to disrupt Connexin localization and C-terminal PDZ-domain interactions48. The ssODN contained a 5’ 40 bp and 3’ 50 bp homology arms flanking a 5x glycine linker, with a V5 epitope tag in the center. The sequence of this repair construct is: 5’-CCGCAGGAAGTCCATCTATGAGATCAGGAACAAGGATTTGGGAGGTGGAGGCGGAGGTAAGCCTATCCCTAACCCTCTCCTCGGTCTCGATTCTACGGGAGGTGGAGGCGGACCGCGGATGAGTATGCCCAACTTTGGACGAACCCAGTCCAGTGACTCGGC-3’. Upon correct repair, the inserted sequence was designed to disrupt the endogenous recognition site of the CRISPR target sequence by creating four coding-silent mutations to prevent further double stranded breaks after repair. Injection mixes were prepared in a pH 7.5 buffer solution of 300 mM KCl and 4 mM HEPES and contained a final concentration of 200 pg/nL ssODN, 200 pg/nL gRNA, and 1600 pg/nL Cas9 protein (IDT, 1081058). Injection mixes were incubated at 37 °C for 5 min immediately prior to injection to promote formation of the Cas9 and sgRNA complex. Finally, 1 nL of solution was injected into embryos at the one-cell stage. F0 injected animals were used for analysis and signal verified by coimmunostaining with V5 and Cx35.5 antibody labeling.

Cell culture

HEK293T/17 verified cells were purchased from ATCC (CRL-11268; STR profile, amelogenin: X). Cells were passaged and maintained in Dulbecco's Modified Eagle's Medium (DMEM, ATCC) plus 10% fetal bovine serum (FBS, Gibco) at 37°C in a humidified incubator in the presence of 5% CO2. Low passage aliquots were cryopreserved in liquid nitrogen according to manufacturer’s instructions. Cells from each thawed cryovial were monitored for mycoplasma contamination using the Universal Mycoplasma Detection Kit (ATCC, 30–1012K).

METHOD DETAILS

Blastula cell transplantation

Cell transplantation was performed at the high stage approximately 3.3 hr into zebrafish development using standard techniques40. Embryos were chemically de-chorionated with protease type XIV (Sigma Aldrich, 9036-06-0) prior to transplantation. Cells were transplanted using a 50 μm wide glass capillary needle attached to an oil hydraulic. For nbeaa+/+ or nbeaa−/− ; Et(Tol-056:GFP); Pt(V5-ZO1b) to wildtype transplants, and nbeaa+/+ or nbeaa−/−; Tg(Cx34.1-GFPAE37) to wildtype transplants, cells from genotyped donor animals were transplanted into non-transgenic ABC hosts. Approximately 20 cells were deposited ~10–15 cell diameters away from the margin, with a single donor embryo supplying cells to 1-3 hosts. At 5 dpf, larvae were fixed in 2% trichloroacetic acid (TCA) in PBS51, donor animals genotyped, and host animals were sorted based on the genotype of the donor and processed for immunohistochemistry. As more than a single cell is transplanted to the host embryo, using confocal microscopy, immunostained larvae were examined for GFP+ Mauthner and GFP+ CoLo cells in the chimeras. Animals with successful GFP+ Mauthner cell transplants were imaged and analyzed, with any synapses resulting at the junction of both a GFP+ Mauthner cell and a GFP+ CoLo cell disregarded.

Immunohistochemistry and confocal imaging

Anesthetized, 5 days post fertilization (dpf) larvae were fixed for 4 hr in 2% TCA in PBS51. Fixed tissue was washed in PBS + 0.5% Triton X-100, followed by standard blocking and antibody incubations. Primary antibody mixes included combinations of the following: rabbit anti-Cx35.5 (Fred Hutch Antibody Technology Facility, clone 12H5, 1:200), mouse IgG2A anti-Cx34.1 (Fred Hutch Antibody Technology Facility, clone 5C10A, 1:200), mouse IgG1 anti-ZO1 (Invitrogen, 33–9100, 1:350), mouse IgG2a anti-V5 peptide (Invitrogen, R960-25, 1:500), rabbit anti-Neurobeachin (Invitrogen, PA5-58903, 1:100), mouse anti-GM130, BD Biosciences, 610823, 1:100), mouse anti-RMO44** Neurofilament-M (Invitrogen, 13-0500, 1:50), and chicken anti-GFP (abcam, ab13970, 1:500). All secondary antibodies were raised in goat (Invitrogen, conjugated with Alexa-405,–488, –555, or –633 fluorophores, 1:500). Tissue was then cleared stepwise in a 25%, 50%, 75% glycerol series, dissected, and mounted in ProLong Gold antifade reagent (ThermoFisher, P36930). Images were acquired on a Leica SP8 Confocal using a 405-diode laser and a white light laser set to 499, 553/554/557 (consistent within experiments), and 631 nm, depending on the fluorescent dye imaged. Each laser line’s data was collected sequentially using custom detection filters based on the dye. Quantitative images of the Club Endings (CEs) and M/CoLo transplants were collected using a 63x, 1.40 numerical aperture (NA) oil immersion lens, and other images of M/Colo synapses were collected using a 40x, 1.20 NA water immersion lens. For each set of images, the optimal optical section thickness was used as calculated by the Leica software based on the pinhole, emission wavelengths and NA of the lens. Within each experiment where fluorescence intensity was to be quantified, all animals (including 5 or more wildtype controls) were stained together with the same antibody mix, processed at the same time, and all confocal settings (laser power, scan speed, gain, offset, objective, and zoom) were identical. Multiple animals per genotype were analyzed to account for biological variation. To account for technical variation, fluorescence intensity values for each region of each animal were an average across multiple synapses.

Analysis of confocal imaging

For fluorescence intensity quantitation, confocal images were processed and analyzed using FiJi software52. To quantify staining at M/Colo synapses, a standard region of interest (ROI) surrounding each M/CoLo site of contact was drawn, and the mean fluorescence intensity was measured. For the quantification of staining at the club endings, confocal z-stacks of the Mauthner soma and lateral dendrite were cropped to 200 x 200 pixels centered around the lateral dendritic bifurcation. A FIJIscript was generated to clear the outside of the Mauthner cell based on GFP staining, and a standard threshold was set within each experiment to remove background staining. The image was then transformed into a max intensity projection, synapses thresholded to WT, and the integrated density of each stain within the club ending synapses was extracted. Standard deviations and errors were computed using Prism (GraphPad) or Excel (Microsoft) software. Figure images were created using FiJi, Photoshop (Adobe), and Illustrator (Adobe). Statistical analyses were performed using Prism (GraphPad). For all experiments, unless otherwise listed in the figure legend, values were normalized to wildtype control animals, and n represents the number of fish used.

Cell transfection and co-immunoprecipitation

Full-length Cx34.1, Cx35.5, and ZO1b were cloned into the pCMV expression vectors as previously described38. A Flag-tag was added to the N-terminus of the ZO1b construct to facilitate Flag-ZO1b pull down experiments. N-terminal and C-terminal Neurobeachin constructs were cloned into the pCMV expression vector with an mVenus tag. Low passage HEK293T/17 cells were seeded 24 hr prior to transfection (1 × 106 cells/well of a six-well dish), and the indicated plasmids were co-transfected using 2ug DNA and Lipofectamine 3000 (Invitrogen) following the manufacturer’s instructions. Cells were collected 24 hr post-transfection and lysed in 500 mL solubilization buffer (50 mM Tris [pH7.4], 100 mM NaCl, 5 mM EDTA, 1.5 mM MgCl2, 1 mM DTT and 1% Triton X-100) plus a protease inhibitor cocktail (Pierce). Lysates were centrifuged at 20,000 x g for 30 min at 4°C, and equal amounts of extract were immunoprecipitated with 0.5 ug rabbit anti-GFP (Abcam, Ab290) or 0.5 ug mouse anti-Flag (Sigma, F3165) overnight with rocking at 4°C. Immunocomplexes were captured with 40 μl prewashed Protein A/G agarose beads for 1 hr with rocking at 4°C. Beads were washed three times with lysis buffer, and bound proteins were boiled for 5 min in the presence of LDS-PAGE loading dye containing 200 mM DTT. Samples were resolved by SDS-PAGE using a 4–20% or 8-16% gradient gel and analyzed by Western blot using the following primary antibodies: chick anti-GFP (Abcam Ab13970), rabbit anti-Cx34.1 3A4-conjugated-680LT, and rabbit anti-Cx35.5 12H5-conjugated-680LT, mouse IgG1 anti-ZO1 (Invitrogen, 33–9100, 1:1000). Compatible near-infrared secondary antibodies were used for visualization with the Odyssey system (LI-COR). Quantification was performed using Image Lab (Bio-Rad Laboratories, Inc.). To account for variable expression, values for immunopreciptated proteins were divided by the values detected in extracts, and values reported in the graphs are ratios created of co-immunopreciptated proteins to the immunopreciptated proteins. Resulting values were normalized to the strongest IP band53. Statistical analyses were performed using Prism (GraphPad).

Immunoprecipitation and western blotting of fish brain homogenates

Brains from wildtype, nbeaa−/−, Cx34.1−/−, and Cx35.5−/− euthanized adult fish (4–15 months old) were removed, snap frozen in liquid nitrogen and stored at −80°C until use. To test comparable levels of connexins, two brains of each condition were homogenized in 250uL HSE buffer (20 mM Hepes [pH7.5], 150 mM NaCl, 5 mM EDTA, 5 mM EGTA, and 1 mM DTT) plus a protease inhibitor cocktail using a glass homogenizer. Detergent was added to the homogenate (final 2% octyl ß-Dglucopyranoside, Anatrace) and solubilized overnight with rocking at 4°C. Solubilized homogenate was cleared by centrifugation at 20,000 x g for 30 min at 4°C and Bradford-equalized amounts of extract were immunoprecipitated with 1.5 ug mouse anti-Cx34.1 or mouse anti-Cx35.5 overnight with rocking at 4°C (For details regarding the specificity of antibodies see Miller et al., 2017). Immuno-complexes were captured with 40 μl prewashed Protein A/G agarose beads for 1 hr with rocking at 4°C. Beads were washed three times with lysis buffer, and bound proteins were boiled for 5 min in the presence of LDS-PAGE loading dye containing 200 mM DTT. Samples were resolved by SDS-PAGE using a 4–20% gradient gel and analyzed by Western blot using rabbit anti-Cx34.1 3A4-conjugated-680LT or rabbit antiCx35.5 12H5-conjugated-680LT. Quantification was performed using Image Lab. For the ZO1 assay, three brains of each condition (nbeaa+/+ and nbeaa−/−) were homogenized in 300uL of HSE buffer (20 mM Hepes [pH7.5], 150 mM NaCl, 5 mM EDTA, 5 mM EGTA, and 1 mM DTT) plus a protease inhibitor cocktail using a glass homogenizer. Detergent was added to the homogenate (final 2% octyl ß-Dglucopyranoside, Anatrace) and solubilized overnight with rocking at 4°C. Solubilized homogenate was cleared by centrifugation at 20,000 x g for 30 min at 4°C. Samples were boiled for 5 min in the presence of LDS-PAGE loading dye containing 200 mM DTT. Proteins were examined by western analysis using mouse anti-ZO1 (1:1000), and rabbit anti-beta tubulin (abcam, ab6046, 1:10,000). Compatible near-infrared secondary antibodies were used for visualization. Quantification was performed using Image Lab (Bio-Rad Laboratories, Inc.). Resulting values were normalized to the wildtype values53. Statistical analyses were performed using Prism (GraphPad).

QUANTIFICATION AND STATISTICAL ANALYSIS

Quantification and statistical details including analysis methods and materials used are included above in the Methods details as well as in the figure legends for each figure.

Supplementary Material

1

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
rabbit anti-Cx35.5 Fred Hutch Antibody Technology Facility clone 12H5
mouse IgG2A anti-Cx34.1 Fred Hutch Antibody Technology Facility clone 5C10A
mouse IgG1 anti-ZO1 Invitrogen 33–9100
mouse IgG2a anti-V5 peptide Invitrogen R960-25
rabbit anti-Neurobeachin Invitrogen PA5-58903
mouse anti-GM130 BD Biosciences 610823
mouse anti-RMO44** Neurofilament-M Invitrogen 13-0500
chicken anti-GFP abcam ab13970
goat anti- Alexa405/488/555/633 Invitrogen
rabbit anti-GFP abcam ab290
mouse anti-Flag Sigma F3165
rabbit anti-beta tubulin abcam ab6046
Bacterial and virus strains
Biological samples
Chemicals, peptides, and recombinant proteins
Cas9 protein IDT 1081058
protease type XIV Sigma Aldrich 9036-06-0
ProLong Gold antifade reagent ThermoFisher P36930
Critical commercial assays
Universal Mycoplasma Detection Kit ATCC 30–1012K
T7 megascript kit ThermoFisher AMB13345
Deposited data
Experimental models: Cell lines
HEK293T/17; STR profile, amelogenin: X ATCC CRL-11268
Experimental models: Organisms/strains
Et(Tol-056:GFP) zebrafish Satou et al. 200935 zf206Et
tjp1bΔ16bp zebrafish Marsh et al., 201737 b1370
gjd1aΔ8bp zebrafish Miller et al., 201736 fh436
gjd2aΔ5bp zebrafish Miller et al., 201736 fh437
Pt(V5-ZO1b) zebrafish Lasseigne et al., 202138 b1406
nbeaaQ906* zebrafish Miller et al., 201520 fh364
Tg(Cx34.1-oxGFP) zebrafish This publication. AE37
Oligonucleotides
Cx35.5 target site: 5’-CCCAATTTCGGCCGCACTCAGTC-3 This publication. n/a
 Cx35.5 repair oligo: 5’-CCGCAGGAAGTCCATCTATGAGATCAGGAACAAGGATTTGGGAGGTGGAGGCGGAGGTAAGCCTATCCCTAACCCTCTCCTCGGTCTCGATTCTACGGGAGGTGGAGGCGGACCGCGGATGAGTATGCCCAACTTTGGACGAACCCAGTCCAGTGACTCGGC-3’ This publication. n/a
Recombinant DNA
Plasmid: pCMV:Cx34.1 Lasseigne et al., 202138 n/a
Plasmid: pCMV:Cx35.5 Lasseigne et al., 202138 n/a
Plasmid: pCMV:ZO1b Lasseigne et al., 202138 n/a
Plasmid: pCMV:Flag-ZO1b This paper. n/a
Plasmid: pCMV: mVenus N-term Nbea This paper. n/a
Plasmid: pCMV: mVenus C-term Nbea This paper. n/a
Plasmid: pCMV: mVenus This paper. n/a
Plasmid: pCMV: mVenus ZO1b This paper. n/a
Software and algorithms
FIJI Fiji.sc
Graphpad Prism Graphpad.com
ImageLab Bio-rad.com
Other

Highlights.

  • Neurobeachin instructs hierarchical electrical synapse assembly.

  • Electrical synapse proteins require Neurobeachin for robust synaptic localization.

  • Neurobeachin localizes to electrical synapses and interacts with the scaffold ZO1.

  • Neurobeachin restricts postsynaptic electrical synapse proteins to the dendrite.

Acknowledgements:

Research reported in this publication was supported by the Eunice Kennedy Shriver National Institute of Child Health and Human Development of the National Institutes of Health under Award Number F32HD102182. We acknowledge NIH grants RF1MH120016 to A.E.P, J.O.B., and A.C.M., R21NS085772 to A.E.P. and J.O.B., R01DC011099 to A.E.P., R01EY012857 to J.O.B., R21NS117967 and R01NS105758 to A.C.M. We would like to thank and acknowledge the Zebrafish facility at the University of Oregon who have given our fish the best possible care, especially through the challenges of the global pandemic. We would also like to thank and acknowledge Ali Eggling and Angela Loczi-Storm for contributions to data quantification.

Footnotes

Declaration of Interests: The authors declare no competing interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Pereda AE (2014). Electrical synapses and their functional interactions with chemical synapses. Nat Rev Neurosci 15, 250–263. 10.1038/nrn3708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Jabeen S, and Thirumalai V (2018). The interplay between electrical and chemical synaptogenesis. J Neurophysiol 120, 1914–1922. 10.1152/jn.00398.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.O’Rourke NA, Weiler NC, Micheva KD, and Smith SJ (2012). Deep molecular diversity of mammalian synapses: why it matters and how to measure it. Nat Rev Neurosci 13, 365–379. 10.1038/nrn3170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Südhof TC (2012). The presynaptic active zone. Neuron 75, 11–25. 10.1016/j.neuron.2012.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Connors BW, and Long MA (2004). Electrical Synapses in the Mammalian Brain. Annual Review of Neuroscience 27, 393–418. 10.1146/annurev.neuro.26.041002.131128. [DOI] [PubMed] [Google Scholar]
  • 6.Nagy JI, and Lynn BD (2018). Structural and Intermolecular Associations Between Connexin36 and Protein Components of the Adherens Junction–Neuronal Gap Junction Complex. Neuroscience 384, 241–261. 10.1016/j.neuroscience.2018.05.026. [DOI] [PubMed] [Google Scholar]
  • 7.Nagy JI, Pereda AE, and Rash JE (2018). Electrical synapses in mammalian CNS: past eras, present focus and future directions. Biochim Biophys Acta 1860, 102–123. 10.1016/j.bbamem.2017.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Martin EA, Lasseigne AM, and Miller AC (2020). Understanding the Molecular and Cell Biological Mechanisms of Electrical Synapse Formation. Front Neuroanat 14, 12. 10.3389/fnana.2020.00012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Vervaeke K, Lőrincz A, Nusser Z, and Silver RA (2012). Gap Junctions Compensate for Sublinear Dendritic Integration in an Inhibitory Network. Science 335, 1624–1628. 10.1126/science.1215101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Draguhn A, Traub RD, Schmitz D, and Jefferys JGR (1998). Electrical coupling underlies high-frequency oscillations in the hippocampus in vitro. Nature 394, 189–192. 10.1038/28184. [DOI] [PubMed] [Google Scholar]
  • 11.Roopun AK, Simonotto JD, Pierce ML, Jenkins A, Nicholson C, Schofield IS, Whittaker RG, Kaiser M, Whittington MA, Traub RD, et al. (2010). A nonsynaptic mechanism underlying interictal discharges in human epileptic neocortex. Proceedings of the National Academy of Sciences 107, 338–343. 10.1073/pnas.0912652107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Curti S, Hoge G, Nagy JI, and Pereda AE (2012). Synergy between Electrical Coupling and Membrane Properties Promotes Strong Synchronization of Neurons of the Mesencephalic Trigeminal Nucleus. J. Neurosci 32, 4341–4359. 10.1523/JNEUROSCI.6216-11.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Simon A, Traub RD, Vladimirov N, Jenkins A, Nicholson C, Whittaker RG, Schofield I, Clowry GJ, Cunningham MO, and Whittington MA (2014). Gap junction networks can generate both ripple-like and fast ripple-like oscillations. Eur J Neurosci 39, 46–60. 10.1111/ejn.12386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Palumbos SD, Skelton R, McWhirter R, Mitchell A, Swann I, Heifner S, Von Stetina S, and Miller DM (2021). cAMP controls a trafficking mechanism that maintains the neuron specificity and subcellular placement of electrical synapses. Developmental Cell 56, 3235–3249.e4. 10.1016/j.devcel.2021.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Welzel G, and Schuster S (2022). Connexins evolved after early chordates lost innexin diversity. eLife 11, e74422. 10.7554/eLife.74422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ou C-Y, Poon VY, Maeder CI, Watanabe S, Lehrman EK, Fu AKY, Park M, Fu W-Y, Jorgensen EM, Ip NY, et al. (2010). Two cyclin-dependent kinase pathways are essential for polarized trafficking of presynaptic components. Cell 141, 846–858. 10.1016/j.cell.2010.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Easley-Neal C, Fierro J, Buchanan J, and Washbourne P (2013). Late Recruitment of Synapsin to Nascent Synapses Is Regulated by Cdk5. Cell Reports 3, 1199–1212. 10.1016/j.celrep.2013.03.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Maeder CI, Shen K, and Hoogenraad CC (2014). Axon and dendritic trafficking. Current Opinion in Neurobiology 27, 165–170. 10.1016/j.conb.2014.03.015. [DOI] [PubMed] [Google Scholar]
  • 19.Mignogna ML, and D’Adamo P (2017). Critical importance of RAB proteins for synaptic function. Small GTPases 9, 145–157. 10.1080/21541248.2016.1277001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Miller AC, Voelker LH, Shah AN, and Moens CB (2015). Neurobeachin is required postsynaptically for electrical and chemical synapse formation. Curr Biol 25, 16–28. 10.1016/j.cub.2014.10.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wang X, Herberg FW, Laue MM, Wullner C, Hu B, Petrasch-Parwez E, and Kilimann MW (2000). Neurobeachin: A protein kinase A-anchoring, beige/Chediak-higashi protein homolog implicated in neuronal membrane traffic. J Neurosci 20, 8551–8565. 10.1523/JNEUROSCI.20-23-08551.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Su Y, Balice-Gordon RJ, Hess DM, Landsman DS, Minarcik J, Golden J, Hurwitz I, Liebhaber SA, and Cooke NE (2004). Neurobeachin Is Essential for Neuromuscular Synaptic Transmission. J Neurosci 24, 3627–3636. 10.1523/JNEUROSCI.4644-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.De Rubeis S, He X, Goldberg AP, Poultney CS, Samocha K, Ercument Cicek A, Kou Y, Liu L, Fromer M, Walker S, et al. (2014). Synaptic, transcriptional and chromatin genes disrupted in autism. Nature 515, 209–215. 10.1038/nature13772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Iossifov I, O’Roak BJ, Sanders SJ, Ronemus M, Krumm N, Levy D, Stessman HA, Witherspoon KT, Vives L, Patterson KE, et al. (2014). The contribution of de novo coding mutations to autism spectrum disorder. Nature 515, 216–221. 10.1038/nature13908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bowling KM, Thompson ML, Amaral MD, Finnila CR, Hiatt SM, Engel KL, Cochran JN, Brothers KB, East KM, Gray DE, et al. (2017). Genomic diagnosis for children with intellectual disability and/or developmental delay. Genome Med 9, 43. 10.1186/s13073-017-0433-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Mulhern MS, Stumpel C, Stong N, Brunner HG, Bier L, Lippa N, Riviello J, Rouhl RPW, Kempers M, Pfundt R, et al. (2018). NBEA: developmental disease gene with early generalized epilepsy phenotypes. Ann Neurol 84, 788–795. 10.1002/ana.25350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Medrihan L, Rohlmann A, Fairless R, Andrae J, Döring M, Missler M, Zhang W, and Kilimann MW (2009). Neurobeachin, a protein implicated in membrane protein traffic and autism, is required for the formation and functioning of central synapses. J Physiol 587, 5095–5106. 10.1113/jphysiol.2009.178236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Niesmann K, Breuer D, Brockhaus J, Born G, Wolff I, Reissner C, Kilimann MW, Rohlmann A, and Missler M (2011). Dendritic spine formation and synaptic function require neurobeachin. Nat Commun 2, 557. 10.1038/ncomms1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Nair R, Lauks J, Jung S, Cooke NE, de Wit H, Brose N, Kilimann MW, Verhage M, and Rhee J (2013). Neurobeachin regulates neurotransmitter receptor trafficking to synapses. J Cell Biol 200, 61–80. 10.1083/jcb.201207113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Farzana F, Zalm R, Chen N, Li KW, Grant SGN, Smit AB, Toonen RF, and Verhage M (2016). Neurobeachin Regulates Glutamate- and GABA-Receptor Targeting to Synapses via Distinct Pathways. Mol Neurobiol 53, 2112–2123. 10.1007/s12035-015-9164-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kimmel CB, Ballard WW, Kimmel SR, Ullmann B, and Schilling TF (1995). Stages of embryonic development of the zebrafish. Developmental Dynamics 203, 253–310. 10.1002/aja.1002030302. [DOI] [PubMed] [Google Scholar]
  • 32.Liu KS, and Fetcho JR (1999). Laser ablations reveal functional relationships of segmental hindbrain neurons in zebrafish. Neuron 23, 325–335. 10.1016/s0896-6273(00180783-7. [DOI] [PubMed] [Google Scholar]
  • 33.Sillar KT (2009). Mauthner cells. Current Biology 19, R353–R355. 10.1016/j.cub.2009.02.025. [DOI] [PubMed] [Google Scholar]
  • 34.Yao C, Vanderpool KG, Delfiner M, Eddy V, Lucaci AG, Soto-Riveros C, Yasumura T, Rash JE, and Pereda AE (2014). Electrical synaptic transmission in developing zebrafish: properties and molecular composition of gap junctions at a central auditory synapse. J Neurophysiol 112, 2102–2113. 10.1152/jn.00397.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Satou C, Kimura Y, Kohashi T, Horikawa K, Takeda H, Oda Y, and Higashijima S (2009). Functional Role of a Specialized Class of Spinal Commissural Inhibitory Neurons during Fast Escapes in Zebrafish. J. Neurosci. 29, 6780–6793. 10.1523/JNEUROSCI.0801-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Miller AC, Whitebirch AC, Shah AN, Marsden KC, Granato M, O’Brien J, and Moens CB A genetic basis for molecular asymmetry at vertebrate electrical synapses. eLife 6, e25364. 10.7554/eLife.25364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Marsh AJ, Michel JC, Adke AP, Heckman EL, and Miller AC (2017). Asymmetry of an Intracellular Scaffold at Vertebrate Electrical Synapses. Curr Biol 27, 3561–3567.e4. 10.1016/j.cub.2017.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lasseigne AM, Echeverry FA, Ijaz S, Michel JC, Martin EA, Marsh AJ, Trujillo E, Marsden KC, Pereda AE, and Miller AC (2021). Electrical synaptic transmission requires a postsynaptic scaffolding protein. eLife 10, e66898. 10.7554/eLife.66898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lauks J, Klemmer P, Farzana F, Karupothula R, Zalm R, Cooke NE, Li KW, Smit AB, Toonen R, and Verhage M (2012). Synapse Associated Protein 102 (SAP102) Binds the C-Terminal Part of the Scaffolding Protein Neurobeachin. PLOS ONE 7, e39420. 10.1371/journal.pone.0039420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kemp HA, Carmany-Rampey A, and Moens C (2009). Generating Chimeric Zebrafish Embryos by Transplantation. J Vis Exp, 1394. 10.3791/1394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Alcamí P, and Pereda AE (2019). Beyond plasticity: the dynamic impact of electrical synapses on neural circuits. Nat Rev Neurosci 20, 253–271. 10.1038/s41583-019-0133-5. [DOI] [PubMed] [Google Scholar]
  • 42.Autenrieth K, Bendzunas NG, Bertinetti D, Herberg FW, and Kennedy EJ (2016). Defining A-Kinase Anchoring Protein (AKAP) Specificity for Protein Kinase A Subunit RI (PKA-RI). Chembiochem 17, 693–697. 10.1002/cbic.201500632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lampe PD, and Lau AF. (2000). Regulation of Gap Junctions by Phosphorylation of Connexins. Archives of Biochemistry and Biophysics 384, 205–215. 10.1006/abbi.2000.2131. [DOI] [PubMed] [Google Scholar]
  • 44.Phelan P, Goulding LA, Tam JLY, Allen MJ, Dawber RJ, Davies JA, and Bacon JP (2008). Molecular mechanism of rectification at identified electrical synapses in the Drosophila giant fiber system. Curr Biol 18, 1955–1960. 10.1016/j.cub.2008.10.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Rash JR, Curti S, Vanderpool KGV, Kamasawa N, Nannapaneni S, Palacios-Prado N, Flores CE, Yasumura T, O’Brien J, Lynn BD, et al. (2013). Molecular and functional asymmetry at a vertebrate electrical synapse. Neuron 79, 957–969. 10.1016/j.neuron.2013.06.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wilson CA, High SK, McCluskey BM, Amores A, Yan Y, Titus TA, Anderson JL, Batzel P, Carvan MJ III, Schartl M, et al. (2014). Wild Sex in Zebrafish: Loss of the Natural Sex Determinant in Domesticated Strains. Genetics 198, 1291–1308. 10.1534/genetics.114.169284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Costantini LM, Baloban M, Markwardt ML, Rizzo MA, Guo F, Verkhusha VV, and Snapp EL (2015). A palette of fluorescent proteins optimized for diverse cellular environments. Nat Commun 6, 7670. 10.1038/ncomms8670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wang HY, Lin Y-P, Mitchell CK, Ram S, and O’Brien J (2015). Two-color fluorescent analysis of connexin 36 turnover: relationship to functional plasticity. Journal of Cell Science 128, 3888–3897. 10.1242/jcs.162586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Moreno-Mateos MA, Vejnar CE, Beaudoin J-D, Fernandez JP, Mis EK, Khokha MK, and Giraldez AJ (2015). CRISPRscan: designing highly efficient sgRNAs for CRISPR-Cas9 targeting in vivo. Nat Methods 12, 982–988. 10.1038/nmeth.3543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Shah AN, Davey CF, Whitebirch AC, Miller AC, and Moens CB (2015). Rapid reverse genetic screening using CRISPR in zebrafish. Nat Methods 12, 535–540. 10.1038/nmeth.3360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Martin EA, Ijaz S, Pereda AE, and Miller AC (2022). Trichloroacetic Acid Fixation and Antibody Staining of Zebrafish Larvae. Bio Protoc 12, e4289. 10.21769/BioProtoc.4289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.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. 10.1038/nmeth.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Pillai-Kastoori L, Schutz-Geschwender AR, and Harford JA (2020). A systematic approach to quantitative Western blot analysis. Analytical Biochemistry 593, 113608. 10.1016/j.ab.2020.113608 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

Data Availability Statement

  • Microscopy and other data reported in this paper will be shared by the lead contact upon request.

  • No original code is reported in this study.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

RESOURCES