- Virág Vincze1,2,
- Zsombor Esküdt1,3,4,
- Erzsébet Fehér-Juhász1,
- Aishwarya Sanjay Chhatre1,2,
- András Jipa1,
- Martin Csordós1,5,
- Anna Rita Galambos1,
- Dalma Börcsök1,
- Gábor Juhász1,6,* and
- Áron Szabó1,*
- 1HUN-REN Biological Research Center, Institute of Genetics, H-6726 Szeged, Hungary
- 2Doctoral School of Biology, University of Szeged, H-6726 Szeged, Hungary
- 3National Academy of Scientist Education Program of the National Biomedical Foundation, Szeged, H-6721, Hungary
- 4Albert Szent-Györgyi Medical School, University of Szeged, Szeged, H-6720, Hungary
- 5Faculty of Science and Informatics, University of Szeged, Szeged, H-6726, Hungary
- 6Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, H-1117, Hungary
- ↵*Correspondence to: aszabo{at}brc.hu, juhasz.gabor{at}brc.hu
Abstract
Glial immunity plays a pivotal role in the maintenance of nervous system homeostasis and responses to stress conditions, including neural injuries. In Drosophila melanogaster, the transcription factor Stat92E is activated in glial cells following central nervous system injury, independently of the canonical JAK/STAT pathway, to shape glial reactivity towards degenerated axons. However, the upstream regulatory mechanisms governing Stat92E activation remain elusive. Here, we reveal a selective autophagy-mediated regulation of Stat92E in glia by the degradation of the Stat92E repressor Su(var)2-10, a member of the PIAS SUMO ligase family. Atg8a, a core autophagy factor co-localizes and interacts with Su(var)2-10. Su(var)2-10 elimination is required for efficient Stat92E-dependent transcription after injury. Furthermore, we demonstrate that autophagy is essential for the upregulation of immune pathways, exemplified by virus-induced RNA 1 (vir-1), in glial cells following axon injury. We propose that Stat92E function is gated both by activating phosphorylation and autophagic Su(var)2-10 breakdown to licence glial reactivity. These findings underscore the critical role of autophagy in glial immunity and its potential impact on neural injury responses.
Introduction
The brain tissue in homeostatic conditions is constantly monitored by glia to mount an enhanced response in face of a challenge. Microglia persistently sample the environment with elongated processes looking for signals emitted from dying, damaged or infected cells. These trigger morphological and signalling changes in them to induce a transformation into a reactive state. Astrocytes undergo a similar transition upon stimuli. Reactive glia have distinct transcriptional signatures as exemplified by disease-associated microglia in neurodegeneration1. They sense damage- and pathogen-associated molecular patterns (DAMPs and PAMPs), find-me and eat-me cues via dedicated receptors to mount an immune response. They also release various chemokines and cytokines to spread and control neuroinflammation2. Upon central nervous system (CNS) injury, these receptors trigger signalling events that include MAPK, JAK-STAT and NF-κB pathways2. The transcriptomic changes in reactive glia show a great contribution from these signalling networks3–7.
The JAK-STAT pathway is an important but less studied part of glial immunity, especially compared to the NF-κB pathway8. Most interleukin and interferon receptors signal through the JAK-STAT pathway in mammals and induce the expression of either pro- or anti-inflammatory cytokines depending on the STAT paralog, both in peripheral immune cells and in glia8,9. In neurodegenerative diseases, the JAK-STAT pathway contributes to neuroinflammation. Increased interferon-dependent microglial Stat1 activation is observed in models of Alzheimer’s disease, causing complement-dependent synapse elimination10.
Research on Drosophila has elucidated key early responses to axon injury in glia11–15 with the single STAT transcription factor, Stat92E driving upregulation of the draper (drpr), the main phagocytic receptor in glia and the matrix metalloproteinase Mmp1 to induce glial reactivity16,17. Interestingly, this response does not rely on canonical pathway components such as the JAK kinase (Hopscotch (Hop) in flies) or the upstream receptor Domeless (Dome)16. Instead, the Drpr receptor and its downstream signalling partners culminating in Rac1 activation are required for glial Stat92E activation after injury16. Insulin-like signalling has also been shown to promote glial Stat92E activity18. How Rac1 relays signalling to Stat92E is not known19. However, besides Stat92E, the transcription factor AP-1 has also been shown to be required for drpr upregulation12, and Draper/Stat92E/AP-1 activity contributes to proper Mmp1 expression after axon injury17.
Autophagy (more specifically macroautophagy) is a membrane-limited intracellular degradation pathway that removes cytoplasmic components including protein aggregates and organelles19. Autophagy initiation depends on the Atg1 (ULK1/2) complex. Upon induction by starvation or other stress conditions, nucleation process prepares membranes for vesicle formation. Along with Atg8a conjugation, a cup-shaped phagophore forms around the cargo and engulfs in by closing it into an autophagosome. Atg8a conjugation depends on two ubiquitin-like conjugation systems, Atg8a being lipidated by the Atg16/Atg5-Atg12 complex. Autophagosomes fuse with lysosomes where the cargo is broken down and recycled.
A targeted and precise variation on this theme is selective autophagy. During this process, cargo receptors recognize specific cellular components ranging from organelles to single molecules and recruit the autophagosome biogenesis machinery, irrespective of nutritional status20,21. Cargo receptors can bind to ubiquitylated material or are embedded in organelles, but labile molecules can also feature receptor-like motifs for direct degradation.
Stat92E activation being pivotal to glial adaptation to challenges, we need to gain a better insight into its regulatory mechanisms. In Drosophila glia, immediate upstream regulation of Stat92E activation upon injury is not well understood. Here we show that autophagy clears the direct Stat92E repressor PIAS/Su(var)2-10 in wing glia after wing transection thereby gating Stat92E-dependent transcriptional responses. Interestingly, drpr expression is not responsive to injury in these peripheral glial types, instead vir-1, another Stat92E target is induced after wing transection in an autophagy-dependent manner.
Results
Selective autophagy is necessary for Stat92E but not for JNK pathway activation
In an effort to understand nervous system adaptation to altered physiology after injury, we investigated whether autophagy impacts signalling pathways in glia. Two well-characterized immune pathways in glial responses are the JAK-STAT and the JNK pathways. We used a Drosophila wing injury model22,23 (Fig. S1A) to measure transgenic reporter upregulation for both pathways. As in the antennal lobe of the brain after antennal ablation11,12,16, the Stat92E reporter 10xStat92E-GFP24 and the AP-1 reporter TRE-EGFP25 both showed robust activation after injury in the wing nerve (Fig. 1A,B, S1B,C). This expression pattern coincided with glially driven myr::tdTomato signal (Fig. S1B,C), similarly to the glial upregulation of Stat92E and AP-1 reporters in the CNS. In order to establish the dynamics of 10xStat92E-GFP upregulation in the wing nerve, we tested the GFP reporter intensity at different time points after injury (Fig. S1D,E). As 10xStat92E-GFP signal intensity reached its peak 3 days post wing injury (3 dpi), we measured the reporter activation levels 3 dpi hereafter. We then depleted Atg16, a core autophagy factor, specifically in glia using repo-Gal4. While Atg16 silencing did not affect TRE-EGFP expression, it abrogated 10xStat92E-GFP activation after injury (Fig. 1A-C). We also measured the Stat92E reporter activity in Atg5 (Atg55cc5) and Atg8a (Atg8aΔ12) null-mutant (Fig. S2A) backgrounds, observing a comparable effect to the Atg16 knockdown (KD) (Fig. 1D-F). This lack of activation could be rescued by an endogenous promoter-driven Atg5 transgene (Atg5::3xHA) in the Atg5-deficient background (Fig. 1D,F).
Atg5, Atg8 and Atg16 are not only involved in autophagy but also in autophagy-related processes including LC3-associated phagocytosis (LAP), which we characterized in glia during axon debris degradation in glia26. Therefore, we set out to distinguish the contribution of these two processes to Stat92E signalling. Atg13 is a specific component of the autophagic machinery as part of the autophagy initiation complex which does not participate in LAP. Atg13 silencing in glia abrogated Stat92E reporter activation (Fig. 2A,C). Unlike Atg13, Rubicon is not involved in autophagy, but it is required for LAPosome formation during LC3-associated phagocytosis. In contrast to Atg13 knockdown, Rubicon RNA interference (RNAi) did not lead to loss of Stat92E activation (Fig. 2B,D). We conclude that canonical autophagy but not LAP sustains Stat92E signalling in glia.
We aimed to further investigate how autophagy specifically impacts Stat92E activation. Through their LC3-interacting region (LIR), selective autophagy receptors (SARs) bind the LIR motif docking site (LDS) on the core autophagy protein Atg8a. SARs directly interact with to-be-degraded cargos or with ubiquitin (Ub) chains on cargos. LIRs can be also present in cargos themselves leading to degradation via Atg8a. We evaluated a mutant that specifically removes the Atg8a LDS, for Stat92E-dependent gene activation27. Atg8aK48A/Y49A (Atg8aLDS) prevented Stat92E reporter upregulation following injury (Fig. 2E,F). Although silencing ref(2)p/p62, the best known Ub chain-binding SAR in Drosophila also showed a mild decrease in GFP reporter intensity, it eventually had no significant effect on 10xStat92E-GFP activation (Fig. S2B,C). Interestingly, bulk autophagic degradation in glia does not change in response to injury when we assay mCherry+ (lysosomal) versus mCherry+ GFP+ (non-lysosomal) puncta abundance of the tandem tag autophagic flux reporter mCherry-GFP-Atg8a directed into wing glia28 (Fig. S2D). Considering the degradative function of autophagy, this suggests the removal of a negative Stat92E signalling regulator by selective autophagy, mediated mainly by a factor other than Ref(2)P/p62.
Su(var)2-10 depletion enhances Stat92E activation in glia after wing injury
We screened Stat92E repressors and upstream kinases for a potential to repress Stat92E in glia. As the upstream components of the JAK-STAT pathway, hop and Dome are not involved in glial injury responses in flies16, we focused on interactors of Stat92E itself29. A direct repressor of Stat92E is Su(var)2-10 / Protein inhibitor of activated STAT (PIAS)30, a SUMO ligase whose orthologues (PIAS1-4) in mammals SUMOylate and thereby inactivate STAT proteins31. We drove two independent Su(var)2-10 RNAi constructs in glia using repo-GeneSwitch (repo-GS) where RNAi expression was induced only in adults to circumvent lethality. It also gave us opportunity to reduce the native Su(var)2-10 level directly before and after injury, thus preventing adverse effects caused by developmental defects. Unexpectedly, Su(var)2-10 RNAi relieved Stat92E reporter repression predominantly following injury (Fig. 3A-B). This suggests that Su(var)2-10 elimination is required for efficient Stat92E-dependent activation after injury. In contrast to the effect caused by the knockdown or abrogation of an autophagy related component, Su(var)2-10 depletion enhanced glial Stat92E activity. From this, we can draw the conclusion that autophagy plays an activator role, while Su(var)2-10 inhibits Stat92E. Therefore, we wondered whether Su(var)2-10 relays the repressive effect of autophagy disruption onto Stat92E reporter activation. While knockdown of Atg16 abrogated 10xStat92E-GFP activation, depletion of Su(var)2-10 in this background restored the normal magnitude of Stat92E response to injury (Fig. 3C,D). In contrast, introducing a second inert UAS construct into the context of Atg16 RNAi did not restore normal activation (Fig. S3A,B). This indicates that Su(var)2-10 can mediate the effect of autophagy on Stat92E-dependent transcription.
Su(var)2-10 mediates the autophagic modulation of Stat92E activity upon injury by being subject to degradation
Next, we wanted to validate the hypothesis that Su(var)2-10 autophagic degradation leads to Stat92E activation. We depleted Atg16 in glia and examined Su(var)2-10 levels in the adult brain following antennal ablation. Removal of antennae results in axon fragmentation in the antennal lobe (AL) that triggers reactivity in surrounding glia15. Su(var)2-10 is mainly nuclear due to its transcriptional repressor and chromatin organizer functions30,32. Presumably, higher overall Su(var)2-10 levels result in more nuclear abundance and increased nuclear repressor activity. Therefore, we coexpressed nuclear GFP in glia and examined the co-localizing Su(var)2-10 pool. Su(var)2-10 levels were elevated upon Atg16 silencing in both uninjured and injured conditions in glia nuclei adjacent to the AL (Fig. 4A-C). We confirmed the specificity of the Su(var)2-10 antibody on Western blots of Su(var)2-10::EGFP ectopically expressed in Schneider 2 R+ (S2R+) Drosophila cell culture (Fig. S3C). These results suggest that Su(var)2-10 could be an autophagic cargo and explain repression of Stat92E after injury in Atg16 deficiency by higher inhibitory Su(var)2-10 levels in the nucleus.
Interestingly, we found that nuclear Su(var)2-10 colocalizes with Atg8a in glia in many cases (Fig. 4D). Although we did not find a consensus LIR motif in Su(var)2-10, the interaction with Atg8a can be mediated by other labels21. Su(var)2-10 is known to autoSUMOylate itself33,34. We found that SUMO colocalizes with Su(var)2-10+ and Atg8a+ structures inside nuclei (Fig. 4D). However, Su(var)2-10 degradation is supposed to occur in the cytoplasm if it is an autophagy cargo21. Accordingly, we also found cytoplasmic structures which were double positive for Su(var)2-10 and Atg8a in glia in the Drosophila brain around the antennal lobe (Fig. 4E). Therefore, Su(var)2-10 colocalizes with Atg8a and is likely degraded in the cytoplasm by autophagy.
We also tested Su(var)2-10 degradation in a heterologous system. We transfected S2R+ cells with Su(var)2-10::EGFP and treated cells with Bafilomycin A1 (BafA1), a V-ATPase proton pump inhibitor that prevents lysosomal acidification. Su(var)2-10 levels increased upon BafA1 treatment indicating lysosomal degradation of this protein (Fig. 4F,G). Next, we hypothesized that autoSUMOylation of Su(var)2-10 might be a lysosomal degradation signal. The C-terminal domain (CTD) of Su(var)2-10 interacts with SUMO. Mutating two residues in the CTD2 region (CTD2mut) disrupts SUMO binding and autoSUMOylation of Su(var)2-1034. This leads to a decreased nuclear : cytoplasmic Su(var)2-10 ratio in vivo. Using the same CTD2 mutant construct, we found that baseline levels of CTD2mut are increased relative to the wild-type in S2 cells hinting to a degradation defect (Fig. 4F,G). Interestingly, CTD2mut protein levels were insensitive to BafA1 treatment that suggests that SUMOylation-deficient Su(var)2-10 cannot undergo lysosomal degradation.
Finally, we aimed to reveal whether Su(var)2-10 physically interacts with Atg8a in vivo. Atg8a serves as a platform for binding of a selection of autophagy cargos and cargo receptors. After precipitating mCherry::Atg8a from whole fly extracts, we found interaction between certain Su(var)2-10 isoforms and Atg8a (Fig. 4H). We also identified a Su(var)2-10 band that was specific to the eluate showing that it is enriched in the Atg8a interactome and may constitute a modified version of a lower isoform. Collectively, these results led us conclude that interaction of Su(var)2-10 with Atg8a in the nucleus and in the cytoplasm will trigger its autophagic breakdown possibly via its conjugation to SUMO.
vir-1 but not drpr is upregulated by autophagy-activated Stat92E in glia following wing transection
What are the relevant targets of this autophagy-regulated Su(var)2-10/Stat92E axis in glial injury responses? It has been shown that following antennal ablation, baseline drpr expression in the brain increases via Stat92E-dependent transcription under the control of Drpr signalling, creating a positive feedback loop16. This is required for the efficient degradation of the significant amount of debris generated during injury. We decided to monitor drpr transcription after injury to see whether it is under the influence of autophagic regulation. We converted a MiMIC insertion in drpr (drprMI07659) by introducing a Trojan Gal4 cassette to fully capture the scope of drpr-dependent transcription (Fig. S4A). We expressed the recently developed transcriptional reporter UAS-TransTimer35 with drpr-Gal4. TransTimer relies on the ratio of destabilised nuclear GFP versus a stable nuclear RFP to monitor dynamic changes in transcription. drpr-MiMIC-Gal4 flies did not show TransTimer upregulation after wing injury (Fig. S4B,C). Next, we asked whether the complexity of drpr enhancer regulation in the wing hinders the detection of the increase in drpr levels. To this end, we drove UAS-TransTimer with an intronic drpr enhancer-Gal4 (dee7-Gal4) that harbors a functional Stat92E binding site (Fig. S4A) and has been shown to upregulate transcription after injury16. First, we verified that dee7 is activated in the brain after antennal injury as described previously (Fig. S5A)16. Despite this activation, we failed to observe an increase in dee7-Gal4>TransTimer flies in the number of RFP puncta 3 days after wing injury (Fig. S5B,C). We conclude that drpr upregulation by injury is absent in the wing injury model and it is apparently not a prerequisite for efficient debris processing in the wing nerve.
The JAK-STAT pathway underlies many immune pathways including cytokine signalling and viral defense36. We screened immunity-related gene reporters and found that vir-1-GFP37 (Fig. 5A) is robustly upregulated after wing injury (Fig. 5B-E). vir-1-produced GFP colocalized with glially driven myr::tdTomato in the wing nerve indicating glial vir-1 induction (Fig. S6A). Stat92E signalling-driven vir-1 is one of the most strongly induced transcripts after viral infection in Drosophila37. However, besides its supposed systemic antiviral immunity role, vir-1 has been found to be expressed in glia. vir-1 is a target of Gcm, a master regulator of glial cell fate38 and is also a longitudinal glial cell marker in the embryo39. Curiously, vir-1 was described to be markedly upregulated in glia specifically one day after traumatic brain injury but not later40. We verified these data by using the vir-1-GFP reporter in uninjured and injured fly brains. 1 day post antennal ablation showed a significant increase in the reporter GFP levels in glia in and around the AL (Fig. 5F, G). This alludes to an important role for vir-1 in glial adaptation to injury.
vir-1 has seven transcript isoforms. RE and RD are shorter and encompass only the 3’ half of the longer isoforms (RA, RB, RG). Therefore, their transcription initiation is offset relative to these longer variants. We found that after wing injury, isoforms RA and RB are upregulated while RE is not (Fig. S6B). This indicates that an injury-responsive enhancer resides in the proximity of the long isoform transcriptional start site.
To corroborate that vir-1 is a glial Stat92E target, we silenced stat92e acutely in adult glia. This abrogated vir-1-GFP induction in the wing nerve in response to injury (Fig. 5B,C). This prompted us to investigate whether autophagy impacts vir-1 expression similar to the 10xStat92E-GFP reporter. We expressed Atg16 RNAi in glia and followed vir-1 induction after injury. While vir-1 still showed some upregulation upon Atg16 depletion, this change was not significant when compared to uninjured wings (Fig. 5D,E). Accordingly, we propose that autophagy may modify glial reactive states by acting on this important innate immune locus in glia via Stat92E.
Discussion
We have uncovered a mechanism of Stat92E transcription factor activation based on autophagy-mediated clearance of a direct repressor. This switch ensures proper glial Stat92E-dependent responses after nerve injury that may contribute to glial reactivity.
The vital role of autophagy in glia biology has just begun to be unravelled41,42,43. The aging brain features neuroprotective microglia that show elevated levels of autophagy44. Autophagy limits neuroinflammation as it degrades the inflammasome component NLRP3 in microglia to prevent cytokine release and proinflammatory responses45. We find that in addition to these roles, autophagy can also positively regulate glial reactivity by impacting a major immune signalling pathway. The autophagic elimination of Su(var)2-10 provides an alternative to canonical JAK-dependent Stat92E regulation that is apparently not employed in glial injury signalling16. LC3-associated phagocytosis is triggered in glia in response to injury via Drpr26. We excluded the role of LAP in Stat92E-dependent transcription using LAP-disrupting glial silencing of a crucial LAP effector, Rubicon and autophagy-specific genes. How and if injury boosts selective autophagy to remove Su(var)2-10 is an open question. Bulk autophagy in glia, as measured by the mCherry-GFP-Atg8a flux reporter, is not elevated upon wing transection. We can see significant increase in Su(var)2-10 levels upon Atg16 knockdown in glia and in S2 cells both in injured and uninjured conditions. 10xStat92E-GFP reporter data yield contrasting results since if Su(var)2-10 is eliminated from glia via silencing, Stat92E activity only increases when flies are simultaneously injured. However, it is not unexpected that Su(var)2-10 removal alone would be insufficient to trigger Stat92E activation. Stat92E activation still depends on tyrosine phosphorylation16,46 for its proper function in reactive glia. Therefore, we believe that Stat92E operates as a coincidence detector integrating signals from autophagic removal of Su(var)2-10 and activating phosphorylation for full activation.
In support of selective autophagic elimination of Su(var)2-10, we discovered that it is lysosomally degraded, and an autoSUMOylation-defective form of Su(var)2-10 is not subject to this clearance. SUMOylation regulates many developmental and critical cellular processes and enables the quick responsiveness to environmental and homeostatic changes. There is indeed evidence that autophagy could contribute to the elimination of SUMOylated Ataxin-347. Ubiquitylation of SUMOylated proteins by SUMO-targeted ubiquitin ligases (STUbLs) can lead to subsequent degradation48,49. We detected Su(var)2-10 colocalization with Atg8a in the cytoplasm in brain glia. Nuclear Su(var)2-10 also colocalizes with both SUMO and Atg8a. Given that the colocalizing structures are larger than single molecules and vesicles are excluded from the nucleus, these are likely liquid-liquid phase-separated (LLPS) multimolecular aggregates. SUMOylation regulates stress granule assembly50 and other types of phase separation51, and the Su(var)2-10 ortholog PIAS1 is predicted to undergo LLPS52. The lack of a LIR motif suggests that Su(var)2-10 does not interact directly with Atg8a but requires labelling, perhaps by Ub family proteins, for elimination by autophagy. Stat92E activation defects in the Atg8aLDS mutant, SUMO-dependent lysosomal degradation of Su(var)2-10 and its colocalization and interaction with Atg8a all point to selective autophagic clearance of Su(var)2-10.
We make the surprising finding that vir-1, a supposed antiviral molecule 37, is strongly induced in glia after axon injury by Stat92E both in wing and CNS. In a recent study, it was also shown that vir-1 is strongly upregulated in glia similarly to other antimicrobial peptides after closed head traumatic brain injury40. vir-1 expression may be differentially regulated among glial classes since vir-1 expression defines a distinct subset of plasmatocytes53. vir-1-GFP is induced in ensheathing glia and possibly astrocytes in the brain after injury (Fig. 5F). The cellular role of Vir-1 is not known. However, it appears that Vir-1 features a signal peptide and in fact localizes to the extracellular space in the foregut54. Therefore, we hypothesize that it can perform signalling or antimicrobial functions.
drpr upregulation after injury has been shown to be an adaptive response for enhanced debris phagocytosis55. We find that drpr is highly expressed in wing nerve glia in uninjured conditions and it is not induced further by wing transection. This would indicate high phagocytic activity in wing glia, however, axon debris clearance takes substantially longer in the wing as compared to antennal lobe glomeruli in the brain15,56. Therefore, we propose that Drpr levels do not readily predict phagocytic capacity in glia.
Glial reactivity is expected to rely heavily on Stat92E signalling. Phosphorylated Stat3 serves as a marker for reactive astrocytes in mice57. Stat3-dependent transcription counteracts traumatic brain injury, motor neuron injury, spinal cord injury and neonatal white matter injury, however, it contributes to degeneration in AD models57. Glial Stat92E activation is also observed in an amyloid-beta overproducing Drosophila model58. Accordingly, autophagy activation may promote Stat92E signalling and thereby assist functional recovery after injury. In line with this, autophagy deficiency in microglia predisposes to more neurodegeneration and demyelination in a multiple sclerosis (MS) model44. Based on our findings, we expect that the benefits of autophagy stimulation in the nervous system will partly be manifested through STAT family-driven glial reactivity.
Supporting information
Funding
This research was supported by:
the Momentum/Lendület Grant of Hungarian Academy of Sciences (LP2023-6) (GJ)
the Young Researchers’ Excellence Programme of the National Research, Development and Innovation Office (NRDIO) (FK132183) (AS)
the Janos Bolyai Research Scholarship of the Hungarian Academy of Sciences (BO/00078/18) (AS)
the NRDIO New National Excellence Programme (ÚNKP-20-5, ÚNKP-19-4, ÚNKP-18-4) (AS)
the NRDIO grant KKP129797 (GJ)
the NRDIO grant PD145868 (AJ)
the NRDIO grant GINOP-2.3.2-15-2016-00032 (GJ)
and the Biotechnology National Laboratory program of the National Research, Development and Innovation Office (NKFIH-871-3/2020) (GJ).
the National Talent Programme - Scholarship for the Young Talents of the Nation (NTP-NFTÖ-22-B-0055) (VV),
the New National Excellence Programme (ÚNKP-23-3 -SZTE-537) (VV)
and the National Academy of Scientist Education Program of the National Biomedical Foundation under the sponsorship of the Hungarian Ministry of Culture and Innovation (ZE, AS and GJ).
We would like to give a special thanks to László Tirián and Julius Brennecke for the mouse anti-Su(var)2-10 antibody, Giacomo Cavalli for the rabbit anti-SUMO antibody, Marc Freeman for the dee7-Gal4 and other stocks, Jean-Luc Imler for the vir-1-GFP, Melinda Bence and Miklós Erdélyi for Su(var)2-10 RNAi stocks and further support, Zoltán Hegedűs from the Laboratory of Bioinformatics for bioinformatic analyses, the Bloomington Stock Center and the Vienna Drosophila Resource Center for the ordered fly stocks, the Developmental Studies Hybridoma Bank for antibodies, Szilvia Bozsó and Ildikó Kresákné Erdődi for technical assistance. We also acknowledge the provided microscopy support of the Cellular Imaging Laboratory (HUN-REN Biological Research Centre). We are grateful to Gábor Csordás, Arindam Bhattacherjee, András Blastyák and Tamás Maruzs for fruitful discussions. Fig. 5A, 6, S1A and S2A were created by BioRender.com.
Author contributions
Conceptualization: V.V., A.S. and G.J.; Investigation: A.S., V.V., Z.E., E.F.J., A.S.C., A.J., M.C., D.F.B., A.R.G. and G.J. Resources: A.J., Z.E., D.F.B., A. R. G.; Results interpretation: A.S., V.V., Z.E., E.F.J., A.S.C., A.J., M.C., D.F.B., A.R.G. and G.J. Writing: V.V., Z.E., A.S. and G.J.; Manuscript revision: V.V., Z.E., A.S., A.S.C., and G.J.; Supervision: A.S. and G.J.; Funding acquisition: A.S. and G.J.
Competing interests
The authors declare no competing interests.
Data availability
All data needed to evaluate the conclusions in this paper are present in the paper and its Supplementary Information. Other data associated with the article such as raw data are available upon request.
Materials availability
All materials, Drosophila stocks and related information are available from the corresponding authors upon reasonable request.
Ethics statement
The research presented here uses the species Drosophila melanogaster for which no ethical approval is required in the HUN-REN Biological Research Center, Institute of Genetics or by Hungarian authorities. Maintenance of transgenic Drosophila melanogaster at the Institute is regulated by license No. SF/66/2014 of the Gene Technology Authority Registry, Ministry of Agriculture of the Hungarian Government.
Materials and methods
Drosophila stocks and maintenance
The flies were kept at 25°C on a medium containing cornmeal, yeast, agar and dextrose with Nipagin as a preservative in reusable glass vials and bottles. For our experiments, we injured male Drosophila melanogaster that were 2–7 days old after eclosion. During our experiments, we favored male flies to avoid technical inconvenience related to egg laying and to prevent larvae from making the medium too liquid and sticky. The animals were anesthetized by carbon dioxide. Progeny of repo-GS crossed with RNAi lines were raised on normal food during development, then males were kept for 5-7 days on food supplemented with 25 μg/ml RU486/mifepristone (Acros Organics) before axotomy and thereafter during the course of the experiment. Injury was carried out on 7-12 day old flies. The supplemented medium used for experiments using vir-1-GFP contained 125 μg/ml RU486.
The following stocks were ordered from Bloomington Drosophila Center: repo-Gal4 (7415), w1118, daughterless-Gal4 (95282), vasa>Cas9 (51323), TRE-EGFP (59010), firefly luciferaseJF01355, Atg8aTKO.GS04635 (80828), Su(var)2-10JF03384 (29448), Su(var)2-10HMS00750 (32956), UASp-mCherry::Atg8a (37750), UASp-GFP-mCherry-Atg8a (37749), UAS-TransTimer (93411), drprMI07659 (843909), UAS-2xEYFP (60291), vas-int, w[*], hs-Cre (60299), pP{lox(Trojan-GAL4)x3} (60310)59, stat92EGL00437, 10XStat92E-ΕGFP (26197), UAS-NLS::GFP (4775) and 10XUAS-IVS-myr::tdTomato (32221).
The following stocks were sent by Vienna Drosophila Resource Center: RubiconKK108247, Atg13KK100340, Atg16KK10232 and ref(2)PKK105338. The Atg55cc5 60, Atg8aLDS 27, Atg55cc5; Atg5::3xHA 24 stocks were formerly created and published. We generated the 10xStat92E-GFP/CyO; repo-Gal4/Tm6, TRE-GFP/CyO; repo-Gal4/Tm6, NLS-GFP/CyO; repo-Gal4/Tm6, vir-1-GFP/CyO; repo-GS/Tm6 by standard crossing schemes. The following stocks received as gifts were much appreciated: repo-GeneSwitch was kindly shared by Véronique Monnier (Université de Paris, BFA Unit of Functional and Adaptative Biology, UMR 8251, France). dee7-Gal4 (drpr enhancer-Gal4) was kindly sent by Marc Freeman (Vollum Institute, Oregon Health & Science University, USA) and vir-1-GFP.2.6 (vir-1-GFP) by Jean-Luc Imler (IBMC, University of Strasbourg, France).
The Atg8anull frameshift mutant was generated by CRISPR-Cas9 editing. To generate this null allele (Atg8aΔ12), we used the Atg8aTKO.GS04635 gRNA line and crossed it with vasa>Cas9 transgenic flies. We screened candidate mutant lines by PCR and Sanger sequencing and identified the Atg8aΔ12 allele that carries an 8 bp deletion based on sequencing data (Fig. S2A), which deletion causes frameshift in all isoforms of the Atg8a gene.
The 2xmApple::Atg8a construct was cloned into the pACU vector (Addgene #58373). mApple sequences were amplified from the pCDH-TMEM106b-mApple vector (Addgene #179385). The PCR products which were used for the construct assembly with the NEBuilder® HiFi DNA Assembly Master Mix (New England Biolabs) are listed in the Supplementary Table. We validated the constructs by Sanger sequencing and we were able to detect mApple fluorescence in induced transfected S2 cells cotransfected with pMTGal4. The transgenic Drosophila line was established by the Drosophila Injection Service in the Biological Research Center, Szeged (Hungary). The transgene was inserted into the attP40 (2L) landing site.
A MiMIC transgene (MI07659) located in intron 6 of drpr was converted into a drpr-Gal4 fusion gene using a Gal4 donor, so that Gal4 is produced instead of most of the drpr coding sequence. We followed the protocol described in Diao et al. 201559 by crossing in recombinase and donor transgenes. A donor cassette containing a splice acceptor (SA), the Gal4 to be encoded in the correct frame and finally a stop codon and polyadenylation site between two attB sites was mobilized and circularized by heat shock-inducible Cre. Integration into MI07659 was achieved by vas-dΦC31. Candidate progeny were screened for drpr expression by crossing to UAS-2xEYFP. Integration into drpr was validated by PCR. drpr-Gal4 is expressed in glia in the CNS and PNS in addition to other tissues.
Rearing stocks with low bacterial content for RT-qPCR
We placed approximately 100 animals from the used strains into a cylinder containing black egg-laying agar coated with yeast for one night. Embryos were dechorionated in 4-5% hypochlorite on a filter for 1 minute followed by washing with flowing distilled water. Finally, we removed the filter fabric, placed it on a paper towel, and transferred the eggs into new vials containing a nutrient medium mixed with antibiotics already described61. We aimed at creating a fly flood that would partially kill the Drosophila microbiome to prevent the contamination coming from bacteria during the RNA purification. First, we autoclaved the commonly used cornmeal-yeast-agar-dextrose–Nipagin medium. Then, we mixed the food with 0.25 mg/ml ampicillin, tetracycline, streptomycin, and 1 mg/ml kanamycin61. Flies were kept for at least 3 generations on antibiotics. The concentration of the antibiotics were reduced to half 20 days before the sample collection to increase the amount of progeny. The food cocktail preparation and fly transfer took place in a laminar box using only autoclaved and sterile tools.
Wing injury
In all experiments we carried out in Drosophila, male flies were used. In the wing injury assay, we used the axotomy model developed by Fang et al.22 and Neukomm et al23. In accordance with the model (S1A), we removed half of the wing of flies from the distal end with a straight cut across using microsurgical scissors. For each individual, we only cut one wing, leaving the other intact as an uninjured control. The injured animals were then kept in vials at 25 °C. Three days after the injury (3 dpi), a strong 10xStat92E-GFP signal was already observable (Fig. S1D,E).
Antennal ablation
The antennae were ablated 1 day prior to brain dissection by pulling on with a forceps15, and the injured flies were kept in vials at 25°C.
Imaging
While imaging the reporter signals in the the wing, we focused on a specific region inside the L1 vein. L1 vein curves from the wing margin towards the inner area of the wing, containing axons and glial cells. We analyzed the samples at room temperature (RT) using the Axio Imager.M2 microscope (Zeiss) and the ORCAFlash4.0LT CMOS camera (Hamamatsu) using a Zeiss PlanApochromat 63x/1.40 NA objective. Optical slices were created with a Zeiss ApoTome.2 device which was connected to the microscope. The illumination was provided by the CoolLED pE-4000 system. We captured the images using the Zeiss Zen program.
For measuring the Su(var)2-10 signal in Su(var)2-10 and NLS-GFP co-immunostained brains, the Visitron VisiScope Spinning Disk Confocal Microscope (Visitron Systems GmbH, Puchheim, Germany) was used. The optical slices were created with an Olympus LUCPlanFl 40x NA 0.60 objective for quantification, while magnified images shown in the article were captured with an Olympus PlanApo 60x NA 1.42 (oil) objective. Detection was based on an Andor Zyla 4.2 PLUS camera.
For the three channel fluorescence images 405 nm excitation with 405/488/561 tri band emission filter, 488 nm excitation with 525/50 emission filter, and 561 nm excitation with 605/70 emission filter were used with a triple band 405/488/561 dichroic mirror. For the four channel fluorescence images 405 nm excitation with 405/488/561 tri band emission filter, 488 nm excitation with 525/50 emission filter, 561 nm excitation with 605/70 emission filter, and 640 nm excitation with 700/75 emission filter were used with a quad band 405/488/561/640 dichroic mirror. Channels were acquired in sequential mode, after the performed Z sectioning. We used a disk with 50 um pinholes.
For vir-1-GFP signal detection in brain, we used an LSM800 (Zeiss) inverted laser scanning confocal microscope. The brains were imaged at RT with a Zeiss Plan-Apochromat 40x/1.3 NA oil immersion objective with a Z-step of 2.00 μm.
The same microscope settings (illumination, exposure time, laser intensity) were used for all conditions and genotypes within one experiment.
Immunostaining of adult brains
Immunostaining of adult brains was performed as described with some modifications62. Adult brains were dissected from living flies in ice-cold phosphate buffered saline (PBS) and placed immediately in 4% paraformaldehyde in PBS with 0.3% Triton X-100 (PBT) on ice. Brains were fixed for 1 h at 25 °C. After two quick rinses with PBT, brains were washed three times 20 min each. Following blocking in 5% fetal bovine serum (FBS) in PBT for 1 h at RT (RT), brains were incubated with the primary antibody (mouse anti-Su(var)2-1063 1:100, rabbit anti-GFP 1:1000 Thermo Fisher Scientific A-11122, rabbit anti-SUMO64 1:200, rabbit anti-mCherry NBP2-25157, Novis Biologicals 1:500) for 3 days at 4 °C in 5% FBS in PBT. The SUMO antibody was gently provided by Giacomo Cavalli (Institute of Human Genetics (IGH), Montpellier, France) and the Su(var)2-10 antibody by Julius Brennecke (IMBA, Vienna, Austria). Washes were done as before and brains were incubated with the fluorescently labelled secondary antibody (goat anti-rabbit Alexa Fluor 488, A-11034, goat anti-mouse Alexa Fluor 568, A-11031, from Thermo Fisher Scientific, all 1:1000 diluted) in 5% FBS in PBT 2 days at 4 °C in darkness. The secondary antibody was removed by two quick rinses, and DAPI (1:15000) was put into the following first wash with PBT. Brains were washed three times 20 min each. After the washes, the brains were mounted in Vectashield (Vector Laboratories, H-1000-10), and the samples were kept in darkness at 4°C until imaging.
RNA purification and RT-qPCR
For RNA isolation, we collected around 150-200 wings per sample using only autoclaved tools with autoclaved wiping papers placed on the fly sorting pad previously washed with 70% ethanol. The wings were swept in TRI Reagent (Zymo Research)-containing microcentrifuge tubes, using a funnel. We homogenized the samples directly after putting them into TRI Reagent. Total RNA was purified with the Direct-zol RNA Microprep (Zymo Research). DNAse I digestion was also applied. 76 ng total RNA was reverse transcribed in 20 μl reaction volume with the RevertAid First Strand cDNA Synthesis Kit (Thermo Scientific). qRT-PCR was performed in 20 μl reactions in technical triplicates using the PerfeCTa SYBR Green FastMix (Quantabio) with 0.5 μl cDNA and cycled on a Rotor-Gene Q qPCR machine (Qiagen) running Rotor-Gene software 2.3.1.49 (Qiagen), with the following program: 95 °C, 3 min; 45 cycles of 95 °C, 20 s, 60 °C, 20 s and 72 °C, 20 s followed by melting curve analysis. The ΔΔCt method was employed to normalize the data, with Ribosomal protein L32 (RpL32, also known as rp49) serving as the internal control. All primers were designed with Primer-BLAST (https://www.ncbi.nlm.nih.gov/tools/primer-blast) with amplicon length set to 70–150 bp. The used melting temperature was 60 °C. One primer spanned an exon-exon junction in all cases. Primers used for RT-qPCR are listed in the Supplementary Table. We normalized the qPCR datasets to RpL32. The ΔCt-derived expression values were adjusted by a common scaling factor to ensure that the average of control values equaled 1 or 100.
Drosophila S2R+ cell culture
Drosophila S2R + cells were cultured in 1x Schneider’s Drosophila media (GIBCO) supplemented with 10 % fetal bovine serum (GIBCO), penicillin-streptomycin solution (HyClone) in 25-cm2 T-flasks (Thermo Fisher Scientific) at RT. The cells (∼8×105) were seeded in a six well-culture plate and 24 h later were transiently co-transfected with pUASp-Su(var)2-10-WT::GFP, pUASp-Su(var)2-10-CTD2mut::GFP and pMT-Gal4 using the MirusIT-Insect transfection reagent based on the manufacturer’s recommendations. S2R+ cells were transfected with 2500 ng of each plasmid. Twenty-four hours after transfection, 0.5 mM of CuSO4 was added to the cells to induce Gal4 expression from the metallothionein promoter. 24h after induction, cells were treated with 100 nM bafilomycin A1 (Invitrogen), and incubated for 4 h. Cell pellets were stored at -80 °C.
Immunoprecipitation
Extracts were prepared from 0.5 ml of 3-7 days old whole flies from da-Gal4>UAS-myr-tdTomato and da-Gal4>UAS-mCherry::Atg8a genotypes. Flies were homogenized in 0.5 ml extraction buffer (50 mM Tris pH = 7.4, 150 mM NaCl, 2 mM EDTA, 0.5% Nonidet P-40, 5% glycerol, Pierce Protease Inhibitor Tablet (Thermo Fisher Scientific) and Halt Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific) on ice in a Potter homogenizer. Following centrifugation (15000 g, 15 min, 4 °C), the protein concentration in the supernatant was measured with Pierce Bradford Plus reagent (Thermo Fisher Scientific). 5 mg extract was precleared by rotating with 50 μl of Dynabeads M-280 Streptavidin (Thermo Fisher Scientific) for 1h at 4 °C to absorb bead-binding nonspecific proteins and immuoprecipitated using 50 μl of RFP-Trap magnetic Agarose (ChromoTek) for another 1h at 4 °C. Subsequently, beads were washed with extraction buffer 3 times for 10 min each. Beads were eluted in 20 μl of 2x Laemmli sample buffer without DTT for 5 min at 100 °C and diluted and supplemented with DTT. Half of the eluted sample was loaded on gels.
Western blotting
S2 cell pellets were lysed on ice for 30 min in three times packed volume RIPA buffer (50 mM Tris pH = 8, 150 mM NaCl, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, Pierce Protease Inhibitor Tablet (Thermo Fisher Scientific), Halt Protease and Phosphatase Inhibitor Cocktail (Thermo Fisher Scientific)) and cleared by centrifugation at 4 C. Protein concentration in the supernatant was measured with Pierce Bradford Plus reagent (Thermo Fisher Scientific). 5 μg total protein was loaded onto 8% polyacrylamide gels. After overnight blotting on Immobilon®-FL PVDF Transfer Membrane. Membranes were blocked with Intercept (TBS) Blocking Buffer (LI-COR) for 1 h at RT. The following primary antibodies were used: rabbit anti-GFP (Thermo Fisher Scientific A-11122), rabbit anti-mCherry (NBP2-25157, Novis Biologicals), mouse anti-Su(var)2-1063, mouse anti-α-tubulin (Developmental Studies Hybridoma Bank AA4.3) all 1:1000, diluted in Blocking Buffer and incubated for 1 h at RT. Secondary antibodies were the following: anti-mouse IRDye 800CW (926-32210, LI-COR) and anti-rabbit IRDye 680RD (926-68071, LI-COR) 1:15,000, diluted in Blocking Buffer supplemented with 0.02% SDS and 0.2% Tween-20 and incubated for 1h at RT. Dried blots were imaged and quantified in an Odyssey Clx instrument (LI-COR).
Image analysis
The signal intensity coming from the fluorescent images were quantified with Fiji (ImageJ 2.9.0, v1.54h). For assessing TRE-EGFP, 10xStat92E-GFP and vir-1-GFP intensity levels in wings, we measured a 174 x 54 px area in each case. When quantifying the intensity of the vir-1-GFP signal in the brain, intensity data were generated from both optic lobes within a 225 x 37 px area. To determine the level of Su(var)2-10 within the nuclei, we randomly selected 10 glial cells marked by repo-Gal4 > NLS-GFP from each brain around the AL (except in cases where fewer cells were found). The nuclei were outlined using freehand selection. To measure drpr activity in the wing by using drpr-Gal4 > UAS-TransTimer flies, we selected an area of 800 x 200 px and measured all the nuclei intensities inside the rectangles. The nuclei were outlined by freehand selection. In dee7-Gal4 > UAS-TransTimer brains, we used a 1300 x 1300 px area to count red puncta. In dee7-Gal4 > UAS-TransTimer wings, a 600 x 140 px sized rectangular area was selected for counting red puncta. All fluorescent signals detected in the area of interest were normalized to the background with identical area sizes. Western blot bands were quantified by the Image Studio 5.2 software (LI-COR). Su(var)2-10 signals were normalized to α-tubulin levels.
Statistical analysis
Experiments were independently repeated at least two times with different distinctive biological samples, with similar results. The quantified results are represented by truncated violin plots with median and quartiles containing all data points; no data were excluded. The statistical analyses were performed with GraphPad Prism 8.0.1 and 10. We first checked the normal distribution within each data group running the Shapiro-Wilk normality test (α = 0.05). Based on these results, we selected the appropriate statistical test to determine the difference between two datasets. When conducting pairwise comparisons on normally distributed data, we performed unpaired, two-tailed Student’s t-test, while for non-normally distributed data, the unpaired, two-tailed Mann-Whitney test was applied. For the comparison of multiple datasets of normal distribution, one-way ANOVA with Holm-Šídák test for multiple comparison correction was used as in Fig. 4G. No power calculations were performed. For the experiments, we always used a sample size that gave reproducible results, similarly to a relevant article11, except for Western blot data where n = 3 or 4.
Acknowledgements
- Received August 28, 2024.
- Revision received August 28, 2024.
- Accepted August 29, 2024.
- © 2024, Posted by Cold Spring Harbor Laboratory
This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at http://creativecommons.org/licenses/by-nc-nd/4.0/