Skip to main content
PLOS Genetics logoLink to PLOS Genetics
. 2023 Sep 25;19(9):e1010973. doi: 10.1371/journal.pgen.1010973

TDP-43 pathology in Drosophila induces glial-cell type specific toxicity that can be ameliorated by knock-down of SF2/SRSF1

Sarah Krupp 1, Isabel Hubbard 1, Oliver Tam 2,¤, Gale M Hammell 2,¤, Josh Dubnau 1,3,*
Editor: Fengwei Yu4
PMCID: PMC10553832  PMID: 37747929

Abstract

Accumulation of cytoplasmic inclusions of TAR-DNA binding protein 43 (TDP-43) is seen in both neurons and glia in a range of neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD) and Alzheimer’s disease (AD). Disease progression involves non-cell autonomous interactions among multiple cell types, including neurons, microglia and astrocytes. We investigated the effects in Drosophila of inducible, glial cell type-specific TDP-43 overexpression, a model that causes TDP-43 protein pathology including loss of nuclear TDP-43 and accumulation of cytoplasmic inclusions. We report that TDP-43 pathology in Drosophila is sufficient to cause progressive loss of each of the 5 glial sub-types. But the effects on organismal survival were most pronounced when TDP-43 pathology was induced in the perineural glia (PNG) or astrocytes. In the case of PNG, this effect is not attributable to loss of the glial population, because ablation of these glia by expression of pro-apoptotic reaper expression has relatively little impact on survival. To uncover underlying mechanisms, we used cell-type-specific nuclear RNA sequencing to characterize the transcriptional changes induced by pathological TDP-43 expression. We identified numerous glial cell-type specific transcriptional changes. Notably, SF2/SRSF1 levels were found to be decreased in both PNG and in astrocytes. We found that further knockdown of SF2/SRSF1 in either PNG or astrocytes lessens the detrimental effects of TDP-43 pathology on lifespan, but extends survival of the glial cells. Thus TDP-43 pathology in astrocytes or PNG causes systemic effects that shorten lifespan and SF2/SRSF1 knockdown rescues the loss of these glia, and also reduces their systemic toxicity to the organism.

Author summary

Neurodegenerative disorders such as amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD) and Alzheimer’s disease (AD) involve dysfunction in both neuronal and non-neuronal cells in the brain. Each of these disorders involves abnormal accumulation of a protein called TDP-43, which is thought to cause toxic effects that ultimately lead to cell death. We used cell type specific sequencing to characterize the effects of inducing pathological TDP-43 within each of the non-neuronal cell types in Drosophila. This identified changes in expression of a gene called SF2/SRSF1, which has previously been implicated in mediating toxic effects. We found that knocking down the expression of this gene within individual glial cell types was sufficient to prevent the loss of nearby neurons.

Introduction

Pathological cytoplasmic inclusions of TDP-43 are seen in post-mortem brain tissues from >90% of patients with ALS, ~45% of FTD cases, and nearly 50% of AD cases [18]. TDP-43 is an RNA and DNA binding protein that is normally found in the nucleus of healthy cells, where it performs a variety of cellular functions, including regulation of splicing, transcription, and silencing of retrotransposons (RTEs) and endogenous retroviruses (ERVs) [5,913]. Although a few percent of familial ALS cases are caused by amino acid substitutions in the TDP-43 protein coding region, the vast majority of cases involve inclusions with TDP-43 protein of normal amino acid sequence [3,14]. The upstream mechanisms that trigger TDP-43 aggregation are poorly understood, but TDP-43 protein levels are under tight regulatory control, including via an autoregulatory mechanism in which nuclear TDP-43 binds to its own transcript to inhibit protein expression [15].

Under pathological conditions, TDP-43 is lost from the nucleus and becomes sequestered into cytoplasmic inclusions, which removes this autoregulatory mechanism [1517]. This may further increase cytoplasmic concentrations of the protein. The most commonly used animal models rely on overexpression of transgenic TDP-43 to drive aggregation [1619]. Such over-expression largely sidesteps the role of upstream disease initiators. Although it is an imperfect representation of the spontaneous appearance of pathology in sporadic disease, overexpression can drive robust accumulation of hyperphosphorylated, insoluble cytoplasmic inclusions and loss of nuclear localization [3,5,20].

The loss of nuclear function is in fact a shared feature of over-expression and knock out models, but the over-expression approach also has the potential to model impacts from presence of cytoplasmic inclusions [21]. Over-expression models of TDP-43 pathology have revealed myriad cellular roles for TDP-43 as well as downstream effects of pathological TDP-43 that are found to be relevant to disease [5,2125]. The majority of animal model studies have focused on impacts of TDP-43 pathology in neurons. In this study, we investigated the mechanisms at play in glia, including specialized glial cell types that form the Drosophila blood brain barrier (BBB). There are several motivations for these experiments. First, pathological TDP-43 in patients is observed both in neurons and in glial cells [2,2429] and there are established roles in disease progression of both astrocytes and microglia [3034]. For example, primary astrocytes derived from post-mortem sporadic ALS patients are toxic to motor neurons as revealed either with co-culture or application of astrocyte conditioned media [35,36].

Such astrocyte toxicity has also been observed in in vivo rodent and fly models [3537]. Activated microglia also may stimulate astrocytes to adopt a pro-inflammatory state that drives non-cell-autonomous toxicity [34,38]. In addition to these glial impacts, there are clear effects of blood brain barrier dysfunction [3941]. Findings in a Drosophila TDP-43 model are convergent with this literature, implicating a variety of non-neuronal cells in toxicity to neurons [37,42]. We have previously demonstrated that initiating TDP-43 protein pathology by overexpression in Drosophila glial cells is sufficient to cause severe motor deficits, drive TDP-43 pathology in nearby neurons, cause neuronal cell death, and greatly reduce lifespan [20,37,42].

Drosophila have five major glial cell types in the adult central brain, whose functional roles and anatomical features bear many similarities to non-neuronal cells (including glia) in the mammalian brain [43,44]. The perineurial glia (PNG) and subperineurial glia (SPG) form an epithelial like BBB on the brain surface, held together by cell-cell junctions [45]. Although they have a glial origin, the SPG undergo a mesenchymal-to-epithelial transition [43]. In addition to these specialized BBB forming glial cells, the cortex glia (CG), astrocyte-like glia (ALG), and ensheathing glia (EG) provide many of the functional characteristics of mammalian astrocytes and microglia [4347]. We previously reported that TDP-43 pathology in astrocytes, PNG and SPG each contribute non-cell autonomous effects to nearby neurons [37,42].

Here, we used cell-type-specific nuclear RNA-seq both to better characterize the baseline transcriptional profiles of the 5 major glial cell types and to investigate mechanisms underlying glial cell type specific impacts of TDP-43 pathology. We identified numerous glial cell-type-specific transcriptional changes in response to induced TDP-43 expression. Notably, these included SF2/SRSF1, a known suppressor of C9orf72 ALS/FTD models [48] that we also recently identified in a screen for suppression of TDP-43 toxicity in motor neurons [49]. We observe that in response to TDP-43 pathology, SF2/SRSF1 is down-regulated in both the PNG and SPG specialized BBB glia. We demonstrate among the 5 glial cell types, TDP-43 toxicity in PNG is a primary driver of organismal toxicity. And we demonstrate that the systemic effects on the animal’s survival from TDP-43 pathology in PNG can be ameliorated by PNG specific knockdown of SF2/SRSF1.

Results

Organismal impacts from glial cell-type specific TDP-43 pathology reveals PNG as a primary driver of systemic toxicity

We have previously demonstrated that in Drosophila, pan-glial over-expression of TDP-43 causes loss of nuclear localization and accumulation of cytoplasmic hyperphosphorylated TDP-43 [20,42]. Such pan-glial TDP-43 toxicity reduces lifespan and causes non-cell autonomous toxicity to neurons that appears to involve some combination of astrocytes, PNG and SPG [37,42]. Importantly, the toxicity to neurons from glial-specific TDP-43 over-expression in a humanized Drosophila model also triggers pathological cytoplasmic accumulation of TDP-43 in nearby neurons where TDP-43 is not over-expressed [42]. We sought to distinguish the contribution of TDP-43 proteinopathy in each glial cell type to the animal’s survival. Using cell-type specific Gal4 lines (methods) and the temperature sensitive Gal80 (Gal-80ts) approach, we induced post development over-expression of TDP-43 in each of the 5 individual glial cell types. This temperature switch expression system allowed us to avoid effects on development from over-expressing TDP-43 during glial specification, and also permitted us to determine the time-course of lethality after induction on the first day of adult life (Fig 1A). In order to monitor effects on survival of each glial cell type, we co-expressed a nuclear-envelope tagged fluorescent reporter (5X-UAS-unc84-2X-GFP) [37,42]. We then monitored the effects over time of inducing glial-cell type specific TDP-43 over-expression on both organismal and cell type survival. We found induced TDP-43 expression in each of the five glial cell types was sufficient to significantly reduce lifespan (Fig 1B–1F) though we note this reduction is quite modest in size for the cases of the EG and CG and is intermediate in magnitude for the case of SPG. In contrast, we see more substantial reduction in lifespan for PNG (median survival reduced from 42 for control to 18 days) and ALG (median survival reduced from 36 vs. 28 days).

Fig 1. Effect on lifespan of post-developmental TDP-43 overexpression in individual glial cell types of the Drosophila CNS.

Fig 1

(A) To induce post development expression of hTDP-43 and 5XUAS-Unc84-2XGFP, flies were reared at 21°C and shifted to 29°C upon eclosion (day 0). (B-F) Lifespan analyses with expression in each individual glial cell type, compared to a control without TDP-43; Effects were significant (Log-Rank) for each glial cell type (PNG, P< 9.003e-28; SPG, P< 1.596e-7; ALG, P< 9.198e-24; CG, P< 0.00000197; EG, P< 0.000004475). (G) Lifespan analysis for pan-glial TDP-43 expression compared to a control without TDP-43 (Repo) P<2.231e-27. Gal4 lines and full genotypes used for each cell type are listed in methods.

It is challenging to make direct comparisons between the severity of lethality with TDP-43 over-expression in each of the 5 glial cell types because the levels of expression may vary across cell types. But it is noteworthy that the effects on lifespan with induced TDP-43 pathology in PNG (Fig 1B) were particularly severe, and approached the magnitude of effect on lifespan seen with pan-glial hTDP-43 expression (Fig 1G). Thus, inducing TDP-43 pathology within each of the 5 glial cell types does impact survival of the animals, but the magnitude of effect on lifespan from TDP-43 pathology is most severe with PNG.

Similar intracellular toxicity of induced TDP-43 across glial cell types

The observed differences in magnitude of effects on lifespan seen above could result from (1) differential intracellular sensitivity of a given cell type to TDP-43 toxicity, (2) differences in severity of effects on brain homeostasis from loss of each glial cell type, or (3) from cell-type specificity in non-cell autonomous toxic effects from TDP-43 proteinopathy [37,42]. We first tested whether there were differences in sensitivity to the toxicity of induced TDP-43 proteinopathy within each glial cell type. This could derive from either biological differences in the resistance of each cell type, from differences in expression levels within that cell type, or from a combination of the two. To distinguish these possibilities, we took advantage of the co-induced 5X-UAS-unc84-2X-GFP fluorescent reporter, to monitor cell number over a time-course after induction of TDP-43. With each genotype, we dissected brains at 2, 5, and 10-days post-induction of both TDP-43 and used confocal microscopy to quantify the numbers of nuclei labeled with GFP.

With each cell type, we observed a progressive loss in the numbers of labeled nuclei over time. With PNG, ALG, EG and CG, we see a similar time-course of cell loss (Fig 2). With each of those four glial cell types, there is a near complete loss of labeled nuclei by 10 days post induction. With SPG, the rate of loss appears to be somewhat slower (2C and 2D). Despite the fact that PNG, ALG, EG and CG appear to be lost at similar rates, TDP-43 over-expression in PNG and ALG caused a palpably more severe shortening in lifespan compared to the other cell types, with the PNG expression causing the most severe effect (Fig 1). The SPG exhibit an intermediate severity of lifespan impact and a slower loss of cell number, making comparisons more difficult. But comparing PNG with EG and CG, there is a starkly higher organismal impact that cannot be explained by differences in rate of loss of these glial subtypes.

Fig 2. TDP-43 overexpression leads to age-dependent loss of each glial cell type.

Fig 2

Representative confocal images (Scale bar = 10μm.) of equivalent anterior section of the Drosophila brain on days 2,5, and 10 post eclosion for flies control (top) vs flies that express TDP-43 in PNGts (A), SPGts (C), CG (E) ALG (G), EG (I). Quantification of the number of each glial cell type in an equivalent anterior section was determined by counting nuclei labeled by Unc84-2X-GFP, which was co-expressed (B,D,F,H,J). Mean and SD are shown. Students T-test used to compare genotypes within each timepoint. * p<0.05, ***p<0.001. Gal4 lines and full genotypes used for each cell type are listed in methods.

Lifespan deficit observed with PNG cell-type-specific TDP-43 induction is not recapitulated by PNG ablation

Given the severe impact on lifespan that we observe with PNG specific TDP-43 induction, we wondered if this reflected loss of an essential function that is specific to this specialized group of BBB glial cells. To test this idea, we compared the effects on lifespan of inducing expression in PNG of TDP-43 vs the pro-apoptotic gene reaper (rpr). In order to quantify effects on PNG cell loss, we co-expressed a UAS-nuclear-GFP. In each case, we used the same Gal-80ts approach to induce post development expression of GFP and either TDP-43 or rpr. As above, animals were reared at 21°C and then shifted to 29°C after eclosion (Day 0) in order to induce expression. We then quantified the effects on lifespan and PNG cell loss (Fig 3).

Fig 3. Lifespan deficit observed with TDP-43 expression in PNG is not recapitulated by PNG ablation.

Fig 3

(A) confocal images (Scale bar = 10μm) of an anterior section of the Drosophila brain on days 2,5, and 10 for flies expressing of PNGts + TDP-43 (top) or PNGts + rpr (bottom). (B) quantification of PNG cells in equivalent anterior sections was performed on days 2,5, and 10 post induction by counting the number of nuclei labeled with GFP, which was co-expressed using the UAS-WM reporter (Methods). Mean and SD shown for each. * p<0.05, T-test. (C) Lifespan analysis of flies expressing rpr (PNGts + rpr) or TDP-43 (PNGts + TDP-43). TDP-43 expression in PNG yields a significantly shorter lifespan than does rpr expression (Log rank, P<1.085e-20). Gal4 lines and full genotypes are listed in methods.

As with induction of TDP-43 pathology, expression of rpr causes progressive loss of the PNG cell population. This occurs with similar kinetics across both groups (Fig 3A and 3B). Despite the indistinguishable effects on PNG cell survival, we find that lifespan of the animals after inducing TDP-43 pathology in PNG is greatly reduced compared to lifespan after induction of rpr (Fig 3C, p<0.0001). Thus, the lifespan deficit resulting from TDP-43 pathology in PNG cannot be explained merely by a loss of this population of glial cells. Instead, this supports the interpretation that TDP-43 pathology in PNG causes systemic effects that reduce survival42 of the animals.

Glial-cell type specific RNA-sequencing reveals differential effects of TDP-43

In order to investigate glial cell-type specific impacts, we profiled both baseline expression in each glial cell type, and differential expression in response to TDP-43 induction. To accomplish this, we used RNA sequencing of nuclei purified from individual glial cell types using tandem affinity purification of INTACT nuclei (TAPIN) [50,51]. Here too, we used the Gal80ts approach to induce the UAS-unc84-2X-GFP nuclear tag either alone or with TDP-43. The 5X-UAS-unc84-2X-GFP reporter is tethered to the inner nuclear membrane, but has the GFP molecule on the outer surface of the nucleus. This provides the means for affinity purification of labeled nuclei from heterogeneous tissue (S1 Fig, S1 Table). In this TAPIN method, GFP-expressing nuclei are purified via two rounds of affinity purification, and then nuclear RNA is extracted and used to produce barcoded Illumina sequencing libraries.

Using this method, we generated nuclear RNA sequencing libraries from each of the 5 cell types and compared expression 2 days after induction of either the GFP tag alone (baseline), or the GFP tag plus TDP-43 (N = 3 libraries per group). Because each of the cell types are comprised of different numbers of cells per brain (S1 Table), we adjusted the number of brains to yield approximately similar numbers of cells per library. Of these 30 samples, 28 yielded high read depth and passed quality control metrics (Table 1, Methods). One CG baseline library and one ALG baseline library were removed from analysis due to low sequencing depth (<5 million reads for one CG sample) and poor correlation with other samples (for one ALG sample). Among the remaining 28 libraries the total number of reads ranged from ~62.5 million to ~103.5 million, with an average of 85% of reads mapped to the reference genome (Table 1). This corresponded to detection of ~15,000 features [i.e. genes, transposable elements (TEs), structural RNAs] on average.

Table 1. Nuclear RNA-seq from each of five glial cell types, with and without TDP-43 induction.

TAPIN RNA-seq library metrics at day 2 across each glial cell type with and without TDP-43.

Library Metric PNGts+TDP43/PNGts Control (N = 3) SPGts+TDP43/SPGts Control (N = 3) CGts+TDP43/CGts Control (N = 3) ALGts+TDP43/ALGts Control (N = 2/3) EGts+TDP43/EGts Control (N = 3)
Total# of reads 70,421,315/62,494,049 81,368,497/77,664,506 64,108,924/103,515,309 48,552,166/62,506,607 71,281,357/74,018,004
% mapped (avg) 86.7/85.9 83.4/84.5 84.8/84.7 82.3/84.2 85.2/85.0
% iniquely aligned (avg) 53.3/58.2 52.7/54.7 48.8/64.3 65.4/70.0 58.7/65.9
Features deteted 15 315 15 009 14 301 14 354 14 954
# of upreg features (P<0.05 1 289 1 012 2 931 1 483 2 498
# of downreg features (P<0.05) 748 782 2 316 1 025 1 772

When comparing the baseline expression profiles across each of the five glial cell types, several things are of note. First, we found that the CG and EG show higher correlation in gene expression with each other than with the other cell types (S2A and S2B Fig). Similarly, the PNG and SPG, which constitute the BBB, are more similar to each other in gene expression profiles than they are with the other cell types (S2B and S2C Fig). Within each cell type, we also found that induction of TDP-43 over-expression resulted in differential expression in numerous genes, including ones that were cell-type specific. Overall, differential expression analysis with DESeq2 revealed upregulation of ~1,000 to ~3,000 features and downregulation of ~750 to ~2,300 features across the various cell types. The majority of the differentially expressed features were cellular genes (Fig 4A,4C,4E,4G and 4I, S2 Table), although we also detected many changes in expression of TEs, including RTEs and ERVs as expected (Fig 4B,4D,4F,4H and 4J; S3 Table) [20,5257].

Fig 4. Differentially expressed genes and TEs in response to TDP-43 overexpression in each glial cell type.

Fig 4

Significantly (p < .05, see methods) upregulated (Red) and downregulated (Blue) genes (A,C,E,G,I) and TEs (B,D,F,H,J) are shown for TDP43 expressing (Green bar, left) vs control (Black bar, right) for each cell type. Genes or TEs that are significantly (p < .05) upregulated and downregulated are shown for PNG (A, B), SPG (C,D), CG (E,F), ALG (G,H) or and EG (I,J). Full genotypes and Gal4 lines are listed in methods.

The transcriptional responses to TDP-43 pathology within each glial cell type included significant differential expression of many genes from cellular pathways that previously have been implicated in TDP-43 related neurodegeneration. These included genes involved in neuroinflammation [52,53], DNA damage/repair [5456], chromatin organization [49,57] and nucleocytoplasmic transport [58], and many transposable elements [20,5257], each of which have been extensively implicated in TDP-43 related neurodegeneration (S3S7 Table). Although GO analyses do not identify enrichment for effects within these pathways, this may reflect differences in cell-type-specific transcriptomes and/or the fact that we profiled an early timepoint (Day 2 post TDP43 induction), in order to identify early effects. By contrast, expression profiling from human brain samples are at a disease endpoint. Given how soon after induction our expression profiling was done, it was of note that we identified many statistically significant changes in expression of TEs (including RTEs and ERVs), which was true across cell types. Indeed, we found a total of 299 upregulated TEs and 131 downregulated TEs across all five cell types (Fig 4B,4D,4F,4H and 4J; S3 Table). This is convergent with an emerging literature demonstrating that RTEs/ERVs become expressed in response to TDP-43 proteinopathy and may contribute to ALS progression [20,5257]. In this fly model, we have previously demonstrated that RTEs/ERVs play a key role in mediating the toxicity of TDP-43 in glia, as well as in the intercellular toxicity to neurons [20,37,42].

Knockdown of SF2/SRSF1 in astrocytes or PNG ameliorates the toxicity of TDP-43

The differential expression analysis identified numerous cell type specific responses to TDP-43, as well as responses that were found to be in common across several cell types (S2C Fig). Among those that were cell type specific, we made particular note of the differential expression of SF2/SRSF1 (S8 Table) because we and others have recently identified knock-down of this gene as a potent suppressor of TDP-43 and C9orf72 pathology [48,49,58]. We previously reported that knockdown of SF2/SRSF1 in animals that overexpress TDP-43 in a subset of motor neurons is sufficient to largely prevent the age-dependent decline in escape locomotion behavior [49]. We also reported that SF2/SRSF1 knock down in glia was sufficient to extend lifespan of animals in which we had induced glial TDP-43 pathology. In that study, we did not examine cell type specificity, but instead used pan-glial manipulations that included all five of the major glial cell types. Our differential expression analysis identified SF2/SRSF1 as being down regulated in response to TDP-43 expression in PNG (-1.31 log2 fold change, p<0.0001) and SPG (-0.85 log2 fold change, p < .001), but not in CG, ALG, or EG (S8 Table).

The fact that SF2/SRSF1 levels are reduced in SPG and PNG, but not other glial cell types, is intriguing given that pan-glial knockdown of SF2/SRSF1 can ameliorate the lifespan effects of pan-glial TDP-43 pathology (Fig 5A) [55]. We observe a substantial impact on lifespan from pathological TDP-43 induction in PNG and ALG (Fig 1), but only see down regulation of SF2/SRSF1 in PNG (S8 Table). We therefore wondered whether cell-type specific knockdown of SF2/SRSF1 would impact the lifespan and cell survival from TDP-43 induction in PNG vs Astrocytes. We used the PNG or Astrocyte Gal4 lines, together with the Gal80ts method, to induce TDP-43 together with either a control shRNA against mCherry or the shRNA that targets the SF2/SRSF1 gene55. We used the same Gal80ts induction conditions described in previous experiments to turn on expression after development (reared at 21°C, shifted to 29°C on adult day 0). We found that SF2/SRSF1 knockdown significantly suppressed the lifespan defects caused by TDP-43 induction in either PNG or ALG (Fig 5B and 5C). Thus, despite the fact that TDP-43 induction itself causes down-regulation of SF2/SRSF1 in PNG, further knockdown in PNG has a substantial protective effect that extends organismal survival. And although TDP-43 induction in astrocytes does not on its own cause a decrease in SF2/SRSF1 expression, knock-down in astrocytes of SF2/SRSF1 by RNAi does ameliorate the effects on lifespan from astrocytic induction of TDP-43 pathology.

Fig 5. SF2/SRSF1 knockdown prevents glial cell loss and extends lifespan after induced TDP-43 overexpression.

Fig 5

(A-C) Lifespan analysis of (A) Repots + TDP-43 + SF2-IR vs Repots + TDP-43 + mCherry-IR, (B) PNGts + TDP-43 + SF2-IR vs PNGts + TDP-43 + mCherry-IR and (C) ALGts + TDP-43 + SF2-IR vs ALGts + TDP-43 + mCherry-IR. For each case, SF2/SRSF1 knock down provides a significant extension of lifespan (Log rank test. Repo, P<7.193e-19; PNG, P<1.062e-16; ALG, P<1.081e-22). (D) confocal images (Scale bar = 10μm) of an anterior section of the Drosophila brain on days 2,5, and 10 for flies expressing of Repots + TDP-43 + SF2-IR (top) or Repots + TDP-43 + mCherry-IR (bottom). (E) quantification of Repo+ glial cells on days 2,5, and 10 per anterior brain section. Mean and SD are shown. T-tests, * p<0.05, ***p<0.001. Full genotypes and Gal4 lines used are listed in methods.

Inducing TDP-43 in PNG causes loss of most of this population of specialized BBB glia within about 10 days (Fig 2), but the effects on lifespan (Fig 1) are not explained by loss of essential functions of PNG because ablating these cells with rpr expression (Fig 2) does not recapitulate the severe lifespan defect seen with TDP-43 induction. This is consistent with the interpretation that in the presence of TDP-43 pathology, these glia become neurotoxic before they die. Indeed, we previously demonstrated that TDP-43 induction in either SPG, PNG or astrocytes results in non-cell autonomous toxicity that causes the appearance of TDP-43 pathology and DNA damage in neurons, leading to neuronal cell death [37,42]. And in that context, blocking apoptosis of glia was sufficient to prevent glial loss, but this increased toxicity to neurons [37]. In light of these findings, two possibilities could explain the amelioration of the systemic effects on lifespan from knocking down SF2/SRSF1 in PNG or astrocytes. First, it is possible that knock down of SF2/SRSF1 in glia prevents release of a toxic factor that kills neurons. Alternatively, knocking down SF2/SRSF1 may cause glia to die more quickly, thereby preventing their release of a toxic substance. But this second hypothesis can be ruled out because we find that knockdown of SF2/SRSF1 actually delays the loss of glia in response to TDP-43 induction (Fig 5D and 5E). Taken together, these findings support the hypothesis that the protective effects from SF2/SRSF1 are mediated by both an amelioration of the lethality of TDP-43 pathology to these glial cell types, as well as a reduction in the non-cell autonomous toxicity of the glia to surrounding neurons.

Discussion

We use glial cell-type-specific TDP-43 overexpression in Drosophila, which triggers pathological cytoplasmic inclusions, to identify the relative contribution of each cell type to systemic effects on organismal lifespan, cell autonomous effects on glial cell loss, and transcriptional changes within each cell type. The PNG, which constitute a specialized glial cell type that contributes to BBB function [43,44,59], emerged as a primary driver of the effects of TDP-43 induction on lifespan despite similar kinetics of TDP-43 induced cellular loss of each of the glial cell types. It is unlikely that this dramatic effect on lifespan from PNG driven TDP-43 pathology could be explained by loss of essential functions from these specialized BBB cells because ablation of PNG by induced expression of the pro-apoptotic rpr gene, does not phenocopy the shortened lifespan seen with TDP-43 expression despite a similar rate of loss of the PNG cell population.

The PNG and SPG, the ‘surface glia’, are thought to function analogously to the vertebrate BBB. PNG, which comprise the outer most layer of surface glia, far outnumber SPG (~2246 PNG vs ~300 SPG per brain) [43], and lie directly in contact with the hemolymph (analog of blood). Together the PNG and SPG isolate the CNS from hemolymph components by blocking paracellular diffusion [43,45,59,60]. Although BBB function is vital for many aspects of normal physiology [60,61], we demonstrate that loss of these BBB glia is not sufficient to explain the lifespan defects seen when TDP-43 pathology is driven in PNG. Our data argue rather that induced expression of TDP-43 in these glia triggers systemic effects with more severe impact on survival than is seen with ablation of the cell type. This is consistent with the hypothesis that PNG are induced to release toxic factors that contribute to systemic effects that cause shortened lifespan. And our findings also indicate that SF2/SRSF1 plays a key role in mediating the cell autonomous toxicity from TDP-43 over-expression to PNG and astrocytes, but also the systemic toxicity to the animal from such cell-type specific expression.

A sizeable literature supports the idea that release of factors from disease-activated glial cells is sufficient to promote toxicity to other cells including neurons [31,31,62]. This phenomenology is seen across a variety of models, including vertebrate systems and ALS patient-derived glial cells. In mammalian systems, there is evidence for signaling from neurons to microglia, from microglia to astrocytes, and from astrocytes to neurons. In response to TDP-43 pathology, both microglia and astrocytes are thought to enter activation states that are toxic to neurons. Although Drosophila do not have a clear microglial cell type, they do have an astrocyte like cell type. And expression of pathological TDP-43 in Drosophila astrocytes drives non-cell autonomous toxicity to neurons [37]. The findings reported here highlight the impact the PNG, which provide the outermost layer of the BBB. PNG are in direct contact with the hemolymph as well as with the underlying SPG. Thus, the systemic effects of induced TDP-43 within the PNG could involve release of toxic factors into hemolymph. Dysfunction of the BBB from loss of PNG and increased permeability could also synergize with other systemic effects driven by TDP-43 proteinopathy. BBB defects have been noted in patients across a range of neurodegenerative diseases including ALS, FTD and AD. On the other hand, PNG are also in close contact with the underlying SPG, which in turn are in contact with neuronal somata that lie near the surface of the brain.

Our prior work has demonstrated that TDP-43 proteinopathy in Drosophila SPG, PNG and astrocytes is sufficient to trigger loss of nearby neurons. Although mechanisms that mediate non-cell autonomous toxic effects in TDP-43 related and other neurodegenerative disorders are not fully understood, there is evidence implicating inflammatory effects [63], movement of pathological aggregates which seed prion-like propagation [6466] and effects of endogenous retroviruses (ERVs) [42,67]. We have previously shown in the context of this Drosophila model that TDP-43 pathology drives expression of the mdg4-ERV, and that mdg4 contributes to the non-cell autonomous effects on neuronal survival [37,42]. Indeed, there is evidence that intercellular movement of ERV generated viral particles may underlie the propagation of toxicity from cell-to-cell [67]. But it is not at all clear whether different mechanisms underlie inter-cellular signaling effects among each glial and neuronal cell type.

The cell-type specific expression profiling reported here provides some insight into the functional differences between glial cell types in Drosophila, and potentially to the non-cell autonomous effects of TDP-43. Although several prior publications have reported baseline expression profiling of several glial-cell-types during larval development, and adult stages in Drosophila, this is the first direct comparison of RNA expression profiles across all five major glial subtypes in the adult, using a single platform. This dataset supports the idea that CG and EG are functionally most similar, and that PNG and SPG also are more similar to each other than to the other glial cell types (S2 Fig, S2 Table). We anticipate this dataset will serve as a valuable resource to investigate functional properties of each of these glial cell types in Drosophila. Differential gene expression, in response to induction of TDP-43 over-expression within each glial cell type identifies a number of genes and signaling pathways that have previously been implicated in neurodegeneration, with some core similarities across cell types, but also many cell type specific effects (Fig 4, S2S7 Tables). This included numerous differentially expressed TEs (Fig 4 and S3 Table), consistent with previous reports [20,52,6872]. Notably, the SF2/SRSF1 gene exhibits decreased expression in PNG and SPG, but not other glial cell types (S8 Table).

Several previous reports identified SF2/SRSF1 as a suppressor of both TDP-43 and C9orf72 pathology [48,49,58]. As we previously reported, knockdown of SF2/SRSF1 in animals that overexpress TDP-43 in either motor neurons or all glia is sufficient to largely prevent the systemic effects on locomotion or lifespan, respectively [49]. Findings reported here knockdown of SF2/SRSF1 in either PNG or ALG partially rescued effects of TDP-43 pathology on lifespan (Fig 5), but importantly, this also extended cellular survival of the glial cells. This is in contrast with the effects of expressing caspase-3 inhibitors in glial cells that over-express TDP-4342. Like knockdown of SF2/SRSF1, inhibition of apoptotic cell death prevents glial cell death, but this exacerbates neuronal cell death and further shortens lifespan [37], the opposite sign of effect seen with SF2/SRSF1 knockdown (Fig 5). We therefore hypothesize that SF2 decreases both the cell autonomous and non-autonomous toxicity of TDP-43 over-expression, either by interacting directly with TDP-43 or through some other mechanism.

Methods

Drosophila strains

Flies were reared at 21°C and maintained on standard propionic acid food for all experiments in this study. A list of transgenic lines used in this study can be found in the table below. All were backcrossed to Canton-S derivative w1118 (isoCJ1), our laboratory wild-type strain, for at least five generations. Female flies were utilized throughout the study. Strains used are shown in Table 2.

Table 2. Drosophila Strains used.

Canton-S derivative w1118 (isoCJ1)
R85G01-Gal4 (PNG)
R54C07-Gal4 (SPG)
R54H02-Gal4 (CG)
R86E01-Gal4 (ALG)
R56F03-Gal4 (EG)
tub-Gal80ts (2nd chromosome)
5X-UAS-unc84-2X-GFP (INTACT)
UAS-hTDP-43 (3rd chromosome)
UAS-hTDP-43 (2nd chromosome)
UAS-rpr
UAS-WM
UAS-SF2-IR
UAS-mcherry-IR
Repo-Gal4
UAS-nlsGFP
tub-Gal80ts (3rd chromosome)

Lifespan analysis

All flies were reared at 21°C and shifted to 29°C upon eclosion, in order to relieve the Gal80ts repression, allowing Gal4 mediated expression. Each lifespan experiments included 20 female flies per vial of a single genotype, with at least 60 flies total per genotype in each experiment. Flies were counted and flipped onto fresh food every other day. The Log-Rank (Mantel-Cox) test and the Gehan-Breslow-Wilcoxon test were used to compare the survival curves.

Genotypes used in Figs 15 are delineated in Table 3.

Table 3. Genotypes used in each figure.

Figure Drosophila Strains
Figs 1B; 2A, B; 4A, 4B Tubulin-Gal80 ts /5XUAS-Unc84-2XGFP; UAS-TDP-43/ R85G01-Gal4
Tubulin-Gal80 ts /5XUAS-Unc84-2XGFP; R85G01-Gal4/+
Figs 1C; 2C, 2D; 4C, 4D Tubulin-Gal80 ts /5XUAS-Unc84-2XGFP; UAS-TDP-43/ R54C07-Gal4
Tubulin-Gal80 ts /5XUAS-Unc84-2XGFP; R54C07-Gal4/+
Figs 1D; 2E, 2F; 4E, 4F Tubulin-Gal80 ts /5XUAS-Unc84-2XGFP; UAS-TDP-43/ R54H02-Gal4
Tubulin-Gal80 ts /5XUAS-Unc84-2XGFP; R54H02-Gal4/+
Figs 1E; 2G, 2H; 4G, 4H Tubulin-Gal80 ts /5XUAS-Unc84-2XGFP; UAS-TDP-43/ R86E01-Gal4
Tubulin-Gal80 ts /5XUAS-Unc84-2XGFP; R86E01-Gal4/+
Figs 1F; 2I, 2J; 4I, 4J Tubulin-Gal80 ts /5XUAS-Unc84-2XGFP; UAS-TDP-43/ R56F03-Gal4
Tubulin-Gal80 ts /5XUAS-Unc84-2XGFP; R56F03-Gal4/+
Fig 1G Tubulin-Gal80 ts /5XUAS-Unc84-2XGFP; UAS-TDP-43/ Repo-Gal4
Fig 3A–3C Tubulin-Gal80 ts /UAS-TDP-43; UAS-WM/ R85G01-Gal4
Tubulin-Gal80 ts /UAS-rpr; UAS-WM/ R85G01-Gal4
Fig 5A, 5D, 5E Tubulin-Gal80 ts / UAS-SF2-IR; UAS-TDP-43/ Repo-Gal4
Tubulin-Gal80 ts / UAS-mcherry-IR; UAS-TDP-43/ Repo-Gal4
Fig 5B Tubulin-Gal80 ts / UAS-SF2-IR; UAS-TDP-43/ R85G01-Gal4
Tubulin-Gal80 ts / UAS-mcherry-IR; UAS-TDP-43/ R85G01-Gal4
Fig 5C Tubulin-Gal80 ts / UAS-SF2-IR; UAS-TDP-43/ R86E01-Gal4
Tubulin-Gal80 ts / UAS-mcherry-IR; UAS-TDP-43/ R86E01-Gal4

Immunohistochemistry

Brains from adult drosophila were dissected in phosphate-buffered-saline (PBS) on days 2, 5, and 10. After dissection flies were transferred to 4% paraformaldehyde (PFA) solution (1XPBS, 4% PFA, and 0.2% Triton-X-100) and fixed for one hour in a vacuum to remove air from trachea. Brains were then washed 3 times in 1XPBST for 10 minutes each prior to either (1) being mounted directly for imaging; or (2) transferred to a blocking solution (10% normal goat serum in 1X PBST; ThermoFisher: 31872) overnight at 4°C on a nutator in preparation for staining. After blocking brains were washed once in 1X PBST for 10 minutes and transferred to primary antibodies against Repo (1:10, Developmental Studies Hybridoma Bank 8D12) and Elav (1:10, Developmental Studies Hybridoma Bank 7E8A10) in 1XPBST and incubated overnight at 4°C on a nutator. Following primary antibody incubation brains were washed 4 times for 15 minutes each in a 3% salt solution (in 1X PBST) and transferred to secondary antibodies (Abcam: ab175673; ThermoFisher: A-21236) in 1XPBST and incubated overnight at 4°C on a nutator. After secondary incubation brains were washed 4 times for 15 minutes each in a 3% salt solution (in 1X PBST) and mounted in FocusClear (CelExplorer).

Confocal imaging and quantification

Brains were imaged within 24 hours of mounting using a Zeiss LSM 800 confocal microscope at 20X. All images were processed by Zeiss Zen software package. A single standard anterior section, defined by anatomy, was chosen for analysis for all cell quantification. Images were analyzed using FIJI (ImageJ). Mixed-effect analysis and two-way ANOVA were used to compare cell counts.

TAPIN

Methods are described in detail in prior studies [50,51]. Briefly, 2 day old female flies were anesthetized with CO2 and flash frozen in liquid nitrogen before being stored at -80°C until an appropriate number of flies were collected for each group (>1,000,000 cells for all groups except SPG, for which cell number was >300,000). Fly heads were separated, homogenized, and incubated with an antibody against GFP (ThermoFisher: G10362) before being bound to protein A beads, cleaved, and rebound to protein G beads. Twice purified TAPIN-tagged nuclei were then burst and nuclear RNA was collected and stored at -80°C.

Library preparation and sequencing

TAPIN-generated nuclear RNA was treated with DNase and purified by the Arcturus PicoPure system. Purified RNA was amplified to cDNA via Nugen Ovation v2 system (Nugen: 7102–32) and was then fragmented with a Covaris s-series to 200bp. Fragmented cDNA was repaired and underwent second strand synthesis before being ligated to barcoded linkers (Nugen: 0319–32, 0320–32). Libraries were quantified via KAPA qPCR before being sequenced on Illumina NextSeq to 76bp read length.

Computational analysis of RNA-seq libraries

Reads were aligned to the dm6 genome using STAR v2.7.6a [73], allowing for 4% mismatch and up to 100 alignments per read to ensure capture of young transposon sequences. Abundance of gene (UCSC curated Refseq, March 2018) and transposon (UCSC RepeatMasker) sequences was calculated with TEtranscripts v2.2.1 [74]. Differential analysis was performed using DESeq2 v1.28.198 using the DESeq normalization strategy and negative binomial modeling. B-H corrected FDR P-value threshold of p< 0.05 was used to determine significance. For correlation plots, Pearson correlation was calculated using normalized count values from DESeq2. For heatmap visualization, counts were normalized using a variance stabilizing transformation in DESeq2

Statistical analysis

With the exception of the TAPINseq expression analysis, all statistical analyses were performed in GraphPad Prism v9.2. Specific statistical analyses are listed under each methods description and include mixed-effect analysis, two-way ANOVA, Log-rank (Mantel-Cox) test, and Gehan-Breslow-Wilcoxon test.

Supporting information

S1 Fig. Nuclear RNA-seq Schematic.

Flies expressing UAS-TDP-43 and the INTACT reporter (5XUAS-unc84-2XGFP) under control of a temperatures sensitive Gal80 plus a glial cell-type-specific Gal4 (methods) were (A) reared at 21°C and shifted to 29°C upon eclosion (day 0). Heads were collected for sequencing on day 2 and stored at -80°C until use. (B) schematic of the INTACT nuclear tag used to purify nuclei and schematic of the tandem affinity purification process.

(PDF)

S2 Fig. Gene expression profiles; overlap, PCA, and correlation plots.

Gene expression profiles generated from (A) Pearson correlation analysis demonstrating EG and CG more closely resemble each other at baseline than they do their own cell type with TDP-43 expression (B) principle component analysis plot demonstrating PNG and SPG separate from other glial cell types. (C) upregulated and downregulated genes (p < .05) in a given glial cell type and their overlap with other glial cell types.

(PDF)

S3 Fig. Pathological expression of human TDP-43 in PNG causes loss of neurons.

The PNG-Gal4 and Gal80ts were used to induce post development expression of hTDP-43 and UAS- Watermelon (WM), a dual reporter module that encodes both a myristylated GFP that tethers to the membrane (myr-GFP-V5) and nuclear localized mCherry (H2B-mCherry-HA). As in other figures, flies were reared at 21°C and shifted to 29°C upon eclosion (day 0). To quantify effects on neuron number, brains were dissected at day 10 (D10) post temperature shift. PNG glia were visualized using the mCherry nuclear reporter, and neuronal nulei were visualized using the Elav antibody. The numbers of Elav-labeled neuronal nuclei were quantified in A 4.68 μm stack (10 sections, 0.52 μm intervals) of ventral, posterior brain region as depicted with a yellow box in the image at left. This reveals a statistically significant (N = 15; Student’s T-test; P = 0.0319) reduction in numbers of neurons in TDP-43 expressing (PNG-Gal4ts:UAS-TDP-43/UAS-WM) vs controls (PNG-Gal4ts>UAS-WM). Means and SEM are shown.

(PDF)

S1 Table. Numbers of fly heads used for TAPIN purification and sequencing based on number of glial cells of each type per brain.

(PDF)

S2 Table. Statistically significant differentially expressed features.

(XLSX)

S3 Table. Statistically significant differentially expressed TEs.

(XLSX)

S4 Table. DNA damage/repair pathways.

(XLSX)

S5 Table. Chromatin organization pathway.

(XLSX)

S6 Table. Nucleocytoplasmic transport pathway.

(XLSX)

S7 Table. Inflammatory response pathways.

(XLSX)

S8 Table. SF2 expression across glial cell types. SF2 expression was significantly reduced at day 2 post induction of TDP-43 in SPG and PNG day 2.

(PDF)

Acknowledgments

We are grateful to Roger Sher, Lillian Talbot, Shreevidya Korada, Enas Gad El-Karim, Richard Keegan, Jorge Azpurua, Meng-Fu Shih, and Yung-Heng Chang for many helpful discussions. We also thank Yung-Heng Chang for comments on the manuscript, and Gilbert L Henry for technical help and advice with the nuclear sequencing methods.

Data Availability

All of the raw sequencing data are available on GEO (GSE242209;https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE242209) All other data are included in the main figures and tables and in the supplemental figures and tables.

Funding Statement

This work was supported by grants RF1AG057338, R01AG078788 and RF1AG076493 from the NIA to JD. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Forman MS, Trojanowski JQ, Lee VM-Y. TDP-43: a novel neurodegenerative proteinopathy. Curr Opin Neurobiol. 2007;17: 548–555. doi: 10.1016/j.conb.2007.08.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314: 130–133. doi: 10.1126/science.1134108 [DOI] [PubMed] [Google Scholar]
  • 3.Ling S-C, Polymenidou M, Cleveland DW. Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis. Neuron. 2013;79: 416–438. doi: 10.1016/j.neuron.2013.07.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Saberi S, Stauffer JE, Jiang J, Garcia SD, Taylor AE, Schulte D, et al. Sense-encoded poly-GR dipeptide repeat proteins correlate to neurodegeneration and uniquely co-localize with TDP-43 in dendrites of repeat-expanded C9orf72 amyotrophic lateral sclerosis. Acta Neuropathol. 2018;135: 459–474. doi: 10.1007/s00401-017-1793-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Prasad A, Bharathi V, Sivalingam V, Girdhar A, Patel BK. Molecular Mechanisms of TDP-43 Misfolding and Pathology in Amyotrophic Lateral Sclerosis. Front Mol Neurosci. 2019;12: 25. doi: 10.3389/fnmol.2019.00025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Neumann M, Kwong LK, Sampathu DM, Trojanowski JQ, Lee VM-Y. TDP-43 proteinopathy in frontotemporal lobar degeneration and amyotrophic lateral sclerosis: protein misfolding diseases without amyloidosis. Arch Neurol. 2007;64: 1388–1394. doi: 10.1001/archneur.64.10.1388 [DOI] [PubMed] [Google Scholar]
  • 7.Amador-Ortiz C, Lin W-L, Ahmed Z, Personett D, Davies P, Duara R, et al. TDP-43 immunoreactivity in hippocampal sclerosis and Alzheimer’s disease. Ann Neurol. 2007;61: 435–445. doi: 10.1002/ana.21154 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Uryu K, Nakashima-Yasuda H, Forman MS, Kwong LK, Clark CM, Grossman M, et al. Concomitant TAR-DNA-binding protein 43 pathology is present in Alzheimer disease and corticobasal degeneration but not in other tauopathies. J Neuropathol Exp Neurol. 2008;67: 555–564. doi: 10.1097/NEN.0b013e31817713b5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Copley KE, Shorter J. Repetitive elements in aging and neurodegeneration. Trends Genet. 2023;39: 381–400. doi: 10.1016/j.tig.2023.02.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dubnau J. The Retrotransposon storm and the dangers of a Collyer’s genome. Curr Opin Genet Dev. 2018;49: 95–105. doi: 10.1016/j.gde.2018.04.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Donde A, Sun M, Ling JP, Braunstein KE, Pang B, Wen X, et al. Splicing repression is a major function of TDP-43 in motor neurons. Acta Neuropathol. 2019;138: 813–826. doi: 10.1007/s00401-019-02042-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bjork RT, Mortimore NP, Loganathan S, Zarnescu DC. Dysregulation of Translation in TDP-43 Proteinopathies: Deficits in the RNA Supply Chain and Local Protein Production. Front Neurosci. 2022;16: 840357. doi: 10.3389/fnins.2022.840357 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Buratti E, Baralle FE. Multiple roles of TDP-43 in gene expression, splicing regulation, and human disease. Front Biosci. 2008;13: 867–878. doi: 10.2741/2727 [DOI] [PubMed] [Google Scholar]
  • 14.Sreedharan J, Blair IP, Tripathi VB, Hu X, Vance C, Rogelj B, et al. TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis. Science. 2008;319: 1668–1672. doi: 10.1126/science.1154584 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ayala YM, De Conti L, Avendaño-Vázquez SE, Dhir A, Romano M, D’Ambrogio A, et al. TDP-43 regulates its mRNA levels through a negative feedback loop. EMBO J. 2011;30: 277–288. doi: 10.1038/emboj.2010.310 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Ash PEA, Zhang Y-J, Roberts CM, Saldi T, Hutter H, Buratti E, et al. Neurotoxic effects of TDP-43 overexpression in C. elegans. Hum Mol Genet. 2010;19: 3206–3218. doi: 10.1093/hmg/ddq230 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Liachko NF, Guthrie CR, Kraemer BC. Phosphorylation promotes neurotoxicity in a Caenorhabditis elegans model of TDP-43 proteinopathy. J Neurosci. 2010;30: 16208–16219. doi: 10.1523/JNEUROSCI.2911-10.2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wils H, Kleinberger G, Janssens J, Pereson S, Joris G, Cuijt I, et al. TDP-43 transgenic mice develop spastic paralysis and neuronal inclusions characteristic of ALS and frontotemporal lobar degeneration. Proc Natl Acad Sci U S A. 2010;107: 3858–3863. doi: 10.1073/pnas.0912417107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Xu Y-F, Gendron TF, Zhang Y-J, Lin W-L, D’Alton S, Sheng H, et al. Wild-type human TDP-43 expression causes TDP-43 phosphorylation, mitochondrial aggregation, motor deficits, and early mortality in transgenic mice. J Neurosci. 2010;30: 10851–10859. doi: 10.1523/JNEUROSCI.1630-10.2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Krug L, Chatterjee N, Borges-Monroy R, Hearn S, Liao W-W, Morrill K, et al. Retrotransposon activation contributes to neurodegeneration in a Drosophila TDP-43 model of ALS. PLoS Genet. 2017;13: e1006635. doi: 10.1371/journal.pgen.1006635 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wegorzewska I, Baloh RH. TDP-43-based animal models of neurodegeneration: new insights into ALS pathology and pathophysiology. Neurodegener Dis. 2011;8: 262–274. doi: 10.1159/000321547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Alrafiah AR. From Mouse Models to Human Disease: An Approach for Amyotrophic Lateral Sclerosis. In Vivo. 2018;32: 983–998. doi: 10.21873/invivo.11339 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Halliday G, Bigio EH, Cairns NJ, Neumann M, Mackenzie IRA, Mann DMA. Mechanisms of disease in frontotemporal lobar degeneration: gain of function versus loss of function effects. Acta Neuropathol. 2012;124: 373–382. doi: 10.1007/s00401-012-1030-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gendron TF, Petrucelli L. Rodent models of TDP-43 proteinopathy: investigating the mechanisms of TDP-43-mediated neurodegeneration. J Mol Neurosci. 2011;45: 486–499. doi: 10.1007/s12031-011-9610-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gendron TF, Rademakers R, Petrucelli L. TARDBP mutation analysis in TDP-43 proteinopathies and deciphering the toxicity of mutant TDP-43. J Alzheimers Dis. 2013;33 Suppl 1: S35–45. doi: 10.3233/JAD-2012-129036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Arai T, Hasegawa M, Akiyama H, Ikeda K, Nonaka T, Mori H, et al. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem Biophys Res Commun. 2006;351: 602–611. doi: 10.1016/j.bbrc.2006.10.093 [DOI] [PubMed] [Google Scholar]
  • 27.Igaz LM, Kwong LK, Xu Y, Truax AC, Uryu K, Neumann M, et al. Enrichment of C-terminal fragments in TAR DNA-binding protein-43 cytoplasmic inclusions in brain but not in spinal cord of frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Am J Pathol. 2008;173: 182–194. doi: 10.2353/ajpath.2008.080003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhang H, Tan C-F, Mori F, Tanji K, Kakita A, Takahashi H, et al. TDP-43-immunoreactive neuronal and glial inclusions in the neostriatum in amyotrophic lateral sclerosis with and without dementia. Acta Neuropathol. 2008;115: 115–122. doi: 10.1007/s00401-007-0285-7 [DOI] [PubMed] [Google Scholar]
  • 29.Hasegawa M, Arai T, Nonaka T, Kametani F, Yoshida M, Hashizume Y, et al. Phosphorylated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Ann Neurol. 2008;64: 60–70. doi: 10.1002/ana.21425 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yamashita H, Komine O, Fujimori-Tonou N, Yamanaka K. Comprehensive expression analysis with cell-type-specific transcriptome in ALS-linked mutant SOD1 mice: Revisiting the active role of glial cells in disease. Front Cell Neurosci. 2022;16: 1045647. doi: 10.3389/fncel.2022.1045647 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Naito M, Hideyama T, Teramoto S, Saito T, Kato H, Terashi H, et al. Pathological features of glial cells and motor neurons in the anterior horn of the spinal cord in sporadic ALS using ADAR2 conditional knockout mice. J Neurol Sci. 2023;444: 120520. doi: 10.1016/j.jns.2022.120520 [DOI] [PubMed] [Google Scholar]
  • 32.Vahsen BF, Gray E, Thompson AG, Ansorge O, Anthony DC, Cowley SA, et al. Non-neuronal cells in amyotrophic lateral sclerosis—from pathogenesis to biomarkers. Nat Rev Neurol. 2021;17: 333–348. doi: 10.1038/s41582-021-00487-8 [DOI] [PubMed] [Google Scholar]
  • 33.Szebényi K, Wenger LMD, Sun Y, Dunn AWE, Limegrover CA, Gibbons GM, et al. Human ALS/FTD brain organoid slice cultures display distinct early astrocyte and targetable neuronal pathology. Nat Neurosci. 2021;24: 1542–1554. doi: 10.1038/s41593-021-00923-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Guttenplan KA, Weigel MK, Adler DI, Couthouis J, Liddelow SA, Gitler AD, et al. Knockout of reactive astrocyte activating factors slows disease progression in an ALS mouse model. Nat Commun. 2020;11: 3753. doi: 10.1038/s41467-020-17514-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Tong J, Huang C, Bi F, Wu Q, Huang B, Liu X, et al. Expression of ALS-linked TDP-43 mutant in astrocytes causes non-cell-autonomous motor neuron death in rats. EMBO J. 2013;32: 1917–1926. doi: 10.1038/emboj.2013.122 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ditsworth D, Maldonado M, McAlonis-Downes M, Sun S, Seelman A, Drenner K, et al. Mutant TDP-43 within motor neurons drives disease onset but not progression in amyotrophic lateral sclerosis. Acta Neuropathol. 2017;133: 907–922. doi: 10.1007/s00401-017-1698-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Chang Y-H, Dubnau J. The Gypsy Endogenous Retrovirus Drives Non-Cell-Autonomous Propagation in a Drosophila TDP-43 Model of Neurodegeneration. Curr Biol. 2019;29: 3135–3152.e4. doi: 10.1016/j.cub.2019.07.071 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541: 481–487. doi: 10.1038/nature21029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Miyazaki K, Ohta Y, Nagai M, Morimoto N, Kurata T, Takehisa Y, et al. Disruption of neurovascular unit prior to motor neuron degeneration in amyotrophic lateral sclerosis. J Neurosci Res. 2011;89: 718–728. doi: 10.1002/jnr.22594 [DOI] [PubMed] [Google Scholar]
  • 40.Garbuzova-Davis S, Hernandez-Ontiveros DG, Rodrigues MCO, Haller E, Frisina-Deyo A, Mirtyl S, et al. Impaired blood-brain/spinal cord barrier in ALS patients. Brain Res. 2012;1469: 114–128. doi: 10.1016/j.brainres.2012.05.056 [DOI] [PubMed] [Google Scholar]
  • 41.Winkler EA, Sengillo JD, Sagare AP, Zhao Z, Ma Q, Zuniga E, et al. Blood-spinal cord barrier disruption contributes to early motor-neuron degeneration in ALS-model mice. Proc Natl Acad Sci U S A. 2014;111: E1035–1042. doi: 10.1073/pnas.1401595111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chang Y-H, Dubnau J. Endogenous retroviruses and TDP-43 proteinopathy form a sustaining feedback driving intercellular spread of Drosophila neurodegeneration. Nat Commun. 2023;14: 966. doi: 10.1038/s41467-023-36649-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kremer MC, Jung C, Batelli S, Rubin GM, Gaul U. The glia of the adult Drosophila nervous system. Glia. 2017;65: 606–638. doi: 10.1002/glia.23115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Stork T, Bernardos R, Freeman MR. Analysis of glial cell development and function in Drosophila. Cold Spring Harb Protoc. 2012;2012: 1–17. doi: 10.1101/pdb.top067587 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Limmer S, Weiler A, Volkenhoff A, Babatz F, Klämbt C. The Drosophila blood-brain barrier: development and function of a glial endothelium. Front Neurosci. 2014;8: 365. doi: 10.3389/fnins.2014.00365 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yildirim K, Petri J, Kottmeier R, Klämbt C. Drosophila glia: Few cell types and many conserved functions. Glia. 2019;67: 5–26. doi: 10.1002/glia.23459 [DOI] [PubMed] [Google Scholar]
  • 47.Kim T, Song B, Lee I-S. Drosophila Glia: Models for Human Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci. 2020;21: 4859. doi: 10.3390/ijms21144859 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hautbergue GM, Castelli LM, Ferraiuolo L, Sanchez-Martinez A, Cooper-Knock J, Higginbottom A, et al. SRSF1-dependent nuclear export inhibition of C9ORF72 repeat transcripts prevents neurodegeneration and associated motor deficits. Nat Commun. 2017;8. doi: 10.1038/ncomms16063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Azpurua J, El-Karim EG, Tranquille M, Dubnau J. A behavioral screen for mediators of age-dependent TDP-43 neurodegeneration identifies SF2/SRSF1 among a group of potent suppressors in both neurons and glia. PLoS Genet. 2021;17: e1009882. doi: 10.1371/journal.pgen.1009882 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Henry GL, Davis FP, Picard S, Eddy SR. Cell type-specific genomics of Drosophila neurons. Nucleic Acids Res. 2012;40: 9691–9704. doi: 10.1093/nar/gks671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Davis FP, Nern A, Picard S, Reiser MB, Rubin GM, Eddy SR, et al. A genetic, genomic, and computational resource for exploring neural circuit function. Elife. 2020;9: e50901. doi: 10.7554/eLife.50901 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Tam OH, Rozhkov NV, Shaw R, Kim D, Hubbard I, Fennessey S, et al. Postmortem Cortex Samples Identify Distinct Molecular Subtypes of ALS: Retrotransposon Activation, Oxidative Stress, and Activated Glia. Cell Rep. 2019;29: 1164–1177.e5. doi: 10.1016/j.celrep.2019.09.066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhan L, Xie Q, Tibbetts RS. Opposing roles of p38 and JNK in a Drosophila model of TDP-43 proteinopathy reveal oxidative stress and innate immunity as pathogenic components of neurodegeneration. Hum Mol Genet. 2015;24: 757–772. doi: 10.1093/hmg/ddu493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Giannini M, Bayona-Feliu A, Sproviero D, Barroso SI, Cereda C, Aguilera A. TDP-43 mutations link Amyotrophic Lateral Sclerosis with R-loop homeostasis and R loop-mediated DNA damage. PLoS Genet. 2020;16: e1009260. doi: 10.1371/journal.pgen.1009260 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kawaguchi T, Rollins MG, Moinpour M, Morera AA, Ebmeier CC, Old WM, et al. Changes to the TDP-43 and FUS Interactomes Induced by DNA Damage. J Proteome Res. 2020;19: 360–370. doi: 10.1021/acs.jproteome.9b00575 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Mitra J, Hegde ML. A Commentary on TDP-43 and DNA Damage Response in Amyotrophic Lateral Sclerosis. J Exp Neurosci. 2019;13: 1179069519880166. doi: 10.1177/1179069519880166 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Berson A, Sartoris A, Nativio R, Van Deerlin V, Toledo JB, Porta S, et al. TDP-43 Promotes Neurodegeneration by Impairing Chromatin Remodeling. Curr Biol. 2017;27: 3579–3590.e6. doi: 10.1016/j.cub.2017.10.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Castelli LM, Cutillo L, Souza CDS, Sanchez-Martinez A, Granata I, Lin Y-H, et al. SRSF1-dependent inhibition of C9ORF72-repeat RNA nuclear export: genome-wide mechanisms for neuroprotection in amyotrophic lateral sclerosis. Mol Neurodegener. 2021;16: 53. doi: 10.1186/s13024-021-00475-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Schwabe T, Li X, Gaul U. Dynamic analysis of the mesenchymal-epithelial transition of blood-brain barrier forming glia in Drosophila. Biol Open. 2017;6: 232–243. doi: 10.1242/bio.020669 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hindle SJ, Bainton RJ. Barrier mechanisms in the Drosophila blood-brain barrier. Front Neurosci. 2014;8: 414. doi: 10.3389/fnins.2014.00414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zhao Z, Nelson AR, Betsholtz C, Zlokovic BV. Establishment and Dysfunction of the Blood-Brain Barrier. Cell. 2015;163: 1064–1078. doi: 10.1016/j.cell.2015.10.067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Philips T, Rothstein JD. Glial cells in amyotrophic lateral sclerosis. Exp Neurol. 2014;262 Pt B: 111–120. doi: 10.1016/j.expneurol.2014.05.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Nichols MR, St-Pierre M-K, Wendeln A-C, Makoni NJ, Gouwens LK, Garrad EC, et al. Inflammatory mechanisms in neurodegeneration. Journal of Neurochemistry. 2019;149: 562–581. doi: 10.1111/jnc.14674 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Polymenidou M, Cleveland DW. The seeds of neurodegeneration: prion-like spreading in ALS. Cell. 2011;147: 498–508. doi: 10.1016/j.cell.2011.10.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Colin M, Dujardin S, Schraen-Maschke S, Meno-Tetang G, Duyckaerts C, Courade J-P, et al. From the prion-like propagation hypothesis to therapeutic strategies of anti-tau immunotherapy. Acta Neuropathol. 2020;139: 3–25. doi: 10.1007/s00401-019-02087-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Nonaka T, Hasegawa M. Prion-like properties of assembled TDP-43. Curr Opin Neurobiol. 2020;61: 23–28. doi: 10.1016/j.conb.2019.11.018 [DOI] [PubMed] [Google Scholar]
  • 67.Liu S, Heumüller S-E, Hossinger A, Müller SA, Buravlova O, Lichtenthaler SF, et al. Reactivated endogenous retroviruses promote protein aggregate spreading. Nat Commun. 2023;14: 5034. doi: 10.1038/s41467-023-40632-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Liu EY, Russ J, Cali CP, Phan JM, Amlie-Wolf A, Lee EB. Loss of Nuclear TDP-43 Is Associated with Decondensation of LINE Retrotransposons. Cell Rep. 2019;27: 1409–1421.e6. doi: 10.1016/j.celrep.2019.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Prudencio M, Gonzales PK, Cook CN, Gendron TF, Daughrity LM, Song Y, et al. Repetitive element transcripts are elevated in the brain of C9orf72 ALS/FTLD patients. Hum Mol Genet. 2017;26: 3421–3431. doi: 10.1093/hmg/ddx233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Li W, Prazak L, Chatterjee N, Grüninger S, Krug L, Theodorou D, et al. Activation of transposable elements during aging and neuronal decline in Drosophila. Nat Neurosci. 2013;16: 529–531. doi: 10.1038/nn.3368 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Douville R, Liu J, Rothstein J, Nath A. Identification of active loci of a human endogenous retrovirus in neurons of patients with amyotrophic lateral sclerosis. Ann Neurol. 2011;69: 141–151. doi: 10.1002/ana.22149 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Li W, Lee M-H, Henderson L, Tyagi R, Bachani M, Steiner J, et al. Human endogenous retrovirus-K contributes to motor neuron disease. Sci Transl Med. 2015;7: 307ra153. doi: 10.1126/scitranslmed.aac8201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Dobin A, Gingeras TR. Optimizing RNA-Seq Mapping with STAR. Methods Mol Biol. 2016;1415: 245–262. doi: 10.1007/978-1-4939-3572-7_13 [DOI] [PubMed] [Google Scholar]
  • 74.Jin Y, Tam OH, Paniagua E, Hammell M. TEtranscripts: a package for including transposable elements in differential expression analysis of RNA-seq datasets. Bioinformatics. 2015;31: 3593–3599. doi: 10.1093/bioinformatics/btv422 [DOI] [PMC free article] [PubMed] [Google Scholar]

Decision Letter 0

Gregory P Copenhaver, Fengwei Yu

29 May 2023

Dear Dr Dubnau,

Thank you very much for submitting your Research Article entitled 'TDP-43 pathology in Drosophila induces glial-cell type specific toxicity that can be ameliorated by knock-down of SF2/SRSF1.' to PLOS Genetics.

The manuscript was fully evaluated at the editorial level and by independent peer reviewers. The reviewers appreciated the attention to an important topic but identified some concerns that we ask you address in a revised manuscript.

We therefore ask you to modify the manuscript according to the review recommendations. Your revisions should address the specific points made by each reviewer.

In addition we ask that you:

1) Provide a detailed list of your responses to the review comments and a description of the changes you have made in the manuscript.

2) Upload a Striking Image with a corresponding caption to accompany your manuscript if one is available (either a new image or an existing one from within your manuscript). If this image is judged to be suitable, it may be featured on our website. Images should ideally be high resolution, eye-catching, single panel square images. For examples, please browse our archive. If your image is from someone other than yourself, please ensure that the artist has read and agreed to the terms and conditions of the Creative Commons Attribution License. Note: we cannot publish copyrighted images.

We hope to receive your revised manuscript within the next 30 days. If you anticipate any delay in its return, we would ask you to let us know the expected resubmission date by email to plosgenetics@plos.org.

If present, accompanying reviewer attachments should be included with this email; please notify the journal office if any appear to be missing. They will also be available for download from the link below. You can use this link to log into the system when you are ready to submit a revised version, having first consulted our Submission Checklist.

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool. PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email us at figures@plos.org.

Please be aware that our data availability policy requires that all numerical data underlying graphs or summary statistics are included with the submission, and you will need to provide this upon resubmission if not already present. In addition, we do not permit the inclusion of phrases such as "data not shown" or "unpublished results" in manuscripts. All points should be backed up by data provided with the submission.

To enhance the reproducibility of your results, we recommend that you deposit your laboratory protocols in protocols.io, where a protocol can be assigned its own identifier (DOI) such that it can be cited independently in the future. Additionally, PLOS ONE offers an option to publish peer-reviewed clinical study protocols. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols

Please review your reference list to ensure that it is complete and correct. If you have cited papers that have been retracted, please include the rationale for doing so in the manuscript text, or remove these references and replace them with relevant current references. Any changes to the reference list should be mentioned in the rebuttal letter that accompanies your revised manuscript. If you need to cite a retracted article, indicate the article’s retracted status in the References list and also include a citation and full reference for the retraction notice.

PLOS has incorporated Similarity Check, powered by iThenticate, into its journal-wide submission system in order to screen submitted content for originality before publication. Each PLOS journal undertakes screening on a proportion of submitted articles. You will be contacted if needed following the screening process.

To resubmit, you will need to go to the link below and 'Revise Submission' in the 'Submissions Needing Revision' folder.

Please let us know if you have any questions while making these revisions.

Yours sincerely,

Fengwei Yu

Academic Editor

PLOS Genetics

Gregory P. Copenhaver

Editor-in-Chief

PLOS Genetics

Reviewer's Responses to Questions

Comments to the Authors:

Please note here if the review is uploaded as an attachment.

Reviewer #1: Krupp et al. described the effect of SF2/SRSF1 knockdown in glia-induced TDP-43 pathology in Drosophila. Overall the study is interesting, of interest, and well conducted. I have some major concerns prior to publication:

1. How physiologically relevant is glial-specific TDP-43 overexpression? TDP-43 accumulation occurs in both glia and neurons, but is TDP-43 expressed in both cell types? If it’s dominantly expressed in neurons, overexpressing TDP-43 in glia does not recapitulate the situation under normal physiological condition.

2. For all experiments in the study, does the control have similar GAL4:UAS ratio as the experimental group? As titration effect of gene expression might occur when using one GAL4 for two UAS, it will be best to compare all effects with similar number of UAS transgenes for the same GAL4.

3. Did the authors try overexpressing SF2/SRSF1 in the presence of TDP-43 in glia? It is a bit counter-intuitive to learn that knockdown of SF2/SRSF1 ameliorate TDP-43 pathology in glia, as RNA-sequencing results suggest that this gene is downregulated when TDP-43 is overexpressed. One would imagine that by overexpressing SF2/SRSF1 TDP-43 pathology will be rescued.

4. The discussion on the non-cell-autonomous toxicity from glia to neurons is a bit hard to understand. It will help if the author can revise and add in more discussion to make things more clear.

5. There is a typo in Figure 1A, the genotype is unc8402xGFP?

Reviewer #2: Understanding how TDP-43 related pathology and aggregation can underlie neurodegenerative diseases like ALS, FTD, and AD is a critical current goal for the field. Important recent work highlighted that not only neurons play a role in the pathological process, but that non-cell autonomous interactions with glia are important. Krupp and colleagues examine such interactions in more depth in this manuscript, to determine the effects of TDP-43 overexpression in specific subpopulations of glia on TDP-43 pathology. They show that organismal survival is most severely impacted following TDP-43 overexpression in perineurial glia or astrocytes; for perineurial glia, this is not due to cell ablation, but likely non-cell autonomous defects. Further, they observe that TDP-43 overexpression in each glial cell type leads to varying degrees of age-dependent cell loss. The authors conduct TAPIN-seq to determine how TDP-43 overexpression affects the transcriptome of each subpopulation of glial cells and show that SF2/SRSF1 is reduced in perineurial glia and astrocytes, consistent with previous broad cell-type findings in ALS models. Further knockdown of SF2/SRSF1 in either perineurial glia or astrocytes reduces TDP-43 pathology including cell loss and lifespan.

This paper represents a strong and important contribution to the field for two main reasons: 1) it demonstrates a fascinating biological finding that glial subtypes are not equally susceptible / causative in TDP-43 pathology and 2) it establishes a valuable resource in the nuclear RNA sequencing databases of different glial subtypes under normal conditions and TDP-43 overexpression conditions. Both aspects are notable advances in the field. Moreover, the paper is well-written, with the experimental logic clearly laid out and the results straightforward and easy to follow. My criticisms are minor and largely restricted to issues of data presentation and the need for additional discussion on the roles of glial subtypes and the mechanism of SF2/SRSF1 function. With these minor additions, the paper will be suitable for publication.

Experimental Point - Though consistent with data from the field, the knockdown of SF2/SRSF1 that rescues TDP-43 pathology when TDP-43 pathology already results in a reduction of SF2/SRSF1 is counterintuitive. If the two were causative / genetically linked, I expect SF2/SRSF1 overexpression would rescue the pathology. Can you instead overexpress SF2/SRSF1 and see altered TDP-43 pathology? Further, the authors posit that SF2/SRSF1 “rescue” results in “a reduction in the non-cell autonomous toxicity of the glia to surrounding neurons.” Can this be demonstrated and quantified? It would help strengthen the existing data. It would also be helpful for the authors to include further discussion of the role / mechanism of SF2/SRSF1.

Discussion Point - There is little discussion of the role that astrocytes are playing in TDP-43 overexpression pathology. The roles of subperineurial glia and perineurial glia are well discussed, but additional discussion / speculation on the role of astrocytes would be very helpful here.

Data Presentation Points - My biggest issue with this paper is in data presentation. In its current state, it is very challenging to discern labels and interpret the results of figures and graphs throughout the paper. Though the issue is present in all figures, it is most distinctly so with Figure 2. For all figures, can the authors adjust font size and clarity? In Figure 2, even at very high magnification, it’s still challenging to tell what the labels are showing. Additionally, larger graphs (i.e., 2B, D, F, H, J) would be very helpful in conveying the important points as with the rest of the figures. In all figures, larger image labels would be helpful.

In Figure 1, it is also difficult to tell the difference in the curve genotypes due to the size of the shapes. Can you color the TDP-43 containing curves so it’s easier to see? Also, can you denote significance on the graph? There is no significance in the figure despite noting it in the text and a p value in the figure legend.

Finally, in Figure 4, the letter labeling is inconsistent with the rest of the paper. Also, the pink bar denoting the Tg_TDP43 genotype is difficult to see. Can it be changed to aid in visibility?

Reviewer #3: TDP43 pathology plays a central role in the pathogenesis of ALS and FTD, and may be also involved in AD as well. Thus, it has been under intense investigation in the field. Over the years, Dr. Josh Dubnau and his colleagues have investigated the effects of TDP43 pathology in Drosophila by overexpressing it in different cell types and have made a series of important discoveries (such as DNA damage and retrotransposon activation). In the current study, the authors made an interesting observation that TDP43 expression in the perineural glia (PNG) and astrocytes had most pronounced effect on organismal survival compared with other types of glia. Because genetic ablation of PNG did not have the same effect, these findings suggest that some unknown factors released by TDP43-expressing PNG cause systematic toxicity and neurodegeneration. This is important, because they are now in an excellent position to perform genetic studies to identify the nature of these factors released by PNG. Of course, their work here would be much more significant and novel if they could report what these factors are. Then they went on to perform cell-type specific nuclear RNA-seq and found that SF2/SRSF1 is downregulated in PNG expressing TDP-43. Further KD of SF2/SRSF1 in PNG rescued loss of these glia and reduced systematic toxicity of TDP43. Although the beneficial effects of SF2/SRSF1 KD in TDP-43 or C9 models are already known, their RNA-seq datasets will be useful for the field.

The authors can consider the following suggestions for minor revisions to further improve their manuscript.

1. The strength of different glial subtype-specific Gal4 drivers may be different. Is it possible different levels of TDP43 are expressed in different glial subtypes to cause different effects on organismal survival? Western blot analysis will not help address this question because the numbers and location of glia in different subtypes are different. Or immunostaining of different flies is good enough to tell the difference. At a minimal, the authors can discuss more in detail the number and location of glial subtypes, and potentially differential strengths of different Gal4 drivers.

2. As mentioned above, SF2/SRSF1 is already known to be a modifier in TDP43 and C9 models (e.g., PLoS Genet. 2021). If not too much work, the authors can include another novel modifier identified from their RNA-seq analysis to enhance the novelty of the study. Alternatively, they can perform a few more mechanistic studies to understand further how SF2/SRSF1 modifies TDP-43 toxicity. For instance, is SF2/SRSF1 required to export TDP43 mRNA from the nucleus to the cytoplasm as it does for C9 repeats as reported? Does SF2/SRSF1 overexpression enhance the phenotype? And so on.

3. Maybe the authors published this in their earlier papers. In several places, the authors talk about toxicity of glial TDP43 on neurons, but they never showed any data. Is that possible to include neuron survival data at least in one figure?

4. In Figure 1 legend, it is stated that p<0.0001 for all panels. Is that true? This reviewer is a little bit surprised, because in Panel C, the small difference is largely due to the second data point and death occurred only few days after the experiment started. So the authors should carefully perform statistical analyses again and state in the legend what statistical methods were used. The number of flies for each panel should be stated and it is better to list p values in each panel of the figure as well.

5. In Figure 2 and Figure 3 legends, *p<0.5 must be an error. Is it *p<0.05? Also, please state the values are S.D. or S.E.M., what are n numbers, and by what statistical analysis in all figure legends.

6. In Figure 3B, the number of PNG cells at day 2 is >200, but the image does not show that many cells as in Figure 2. Please explain.

7. In Figure 4 legend, they say “upregulated (blue) and downregulated (red) genes”. But in the figure, upregulated genes are in red and downregulated genes are in blue.

8. Figure 5 can be reorganized to better utilize the empty spaces. Panels D and E can be new Panels A and B, and Panels A-C can be new Panels C-E.

9. Please increase font size in all figures.

**********

Have all data underlying the figures and results presented in the manuscript been provided?

Large-scale datasets should be made available via a public repository as described in the PLOS Genetics data availability policy, and numerical data that underlies graphs or summary statistics should be provided in spreadsheet form as supporting information.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: None

**********

PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

Decision Letter 1

Gregory P Copenhaver, Fengwei Yu

13 Sep 2023

Dear Dr Dubnau,

We are pleased to inform you that your manuscript entitled "TDP-43 pathology in Drosophila induces glial-cell type specific toxicity that can be ameliorated by knock-down of SF2/SRSF1." has been editorially accepted for publication in PLOS Genetics. Congratulations!

Before your submission can be formally accepted and sent to production you will need to complete our formatting changes, which you will receive in a follow up email. Please be aware that it may take several days for you to receive this email; during this time no action is required by you. Please note: the accept date on your published article will reflect the date of this provisional acceptance, but your manuscript will not be scheduled for publication until the required changes have been made.

Once your paper is formally accepted, an uncorrected proof of your manuscript will be published online ahead of the final version, unless you’ve already opted out via the online submission form. If, for any reason, you do not want an earlier version of your manuscript published online or are unsure if you have already indicated as such, please let the journal staff know immediately at plosgenetics@plos.org.

In the meantime, please log into Editorial Manager at https://www.editorialmanager.com/pgenetics/, click the "Update My Information" link at the top of the page, and update your user information to ensure an efficient production and billing process. Note that PLOS requires an ORCID iD for all corresponding authors. Therefore, please ensure that you have an ORCID iD and that it is validated in Editorial Manager. To do this, go to ‘Update my Information’ (in the upper left-hand corner of the main menu), and click on the Fetch/Validate link next to the ORCID field.  This will take you to the ORCID site and allow you to create a new iD or authenticate a pre-existing iD in Editorial Manager.

If you have a press-related query, or would like to know about making your underlying data available (as you will be aware, this is required for publication), please see the end of this email. If your institution or institutions have a press office, please notify them about your upcoming article at this point, to enable them to help maximise its impact. Inform journal staff as soon as possible if you are preparing a press release for your article and need a publication date.

Thank you again for supporting open-access publishing; we are looking forward to publishing your work in PLOS Genetics!

Yours sincerely,

Fengwei Yu

Academic Editor

PLOS Genetics

Gregory P. Copenhaver

Editor-in-Chief

PLOS Genetics

www.plosgenetics.org

Twitter: @PLOSGenetics

----------------------------------------------------

Comments from the reviewers (if applicable):

Reviewer's Responses to Questions

Comments to the Authors:

Please note here if the review is uploaded as an attachment.

Reviewer #1: The reviewer has no further questions and the manuscript is ready for publication in its current form

Reviewer #2: I thank the authors for their thoughtful replies to my comments, for the addition of new data in Figure S3, and especially the changes to data presentation which greatly improved the figures. This was very helpful. The authors have appropriately addressed all of my comments and I can support publication. Thank you!

Though I'd like to cast my vote (since all 3 reviewers asked for this) for overexpressing SF2/SRSF1 in the next paper!

Reviewer #3: The authors have satisfactorily addressed most of my comments. Some suggested experiments were not performed, such as SF2/SRSF1 overexpression, which was also proposed by another reviewer. I understand their reasoning why it is more important to focus on down-regulation.

**********

Have all data underlying the figures and results presented in the manuscript been provided?

Large-scale datasets should be made available via a public repository as described in the PLOS Genetics data availability policy, and numerical data that underlies graphs or summary statistics should be provided in spreadsheet form as supporting information.

Reviewer #1: None

Reviewer #2: Yes

Reviewer #3: Yes

**********

PLOS authors have the option to publish the peer review history of their article (what does this mean?). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy.

Reviewer #1: No

Reviewer #2: No

Reviewer #3: Yes: Fen-Biao Gao

----------------------------------------------------

Data Deposition

If you have submitted a Research Article or Front Matter that has associated data that are not suitable for deposition in a subject-specific public repository (such as GenBank or ArrayExpress), one way to make that data available is to deposit it in the Dryad Digital Repository. As you may recall, we ask all authors to agree to make data available; this is one way to achieve that. A full list of recommended repositories can be found on our website.

The following link will take you to the Dryad record for your article, so you won't have to re‐enter its bibliographic information, and can upload your files directly: 

http://datadryad.org/submit?journalID=pgenetics&manu=PGENETICS-D-23-00514R1

More information about depositing data in Dryad is available at http://www.datadryad.org/depositing. If you experience any difficulties in submitting your data, please contact help@datadryad.org for support.

Additionally, please be aware that our data availability policy requires that all numerical data underlying display items are included with the submission, and you will need to provide this before we can formally accept your manuscript, if not already present.

----------------------------------------------------

Press Queries

If you or your institution will be preparing press materials for this manuscript, or if you need to know your paper's publication date for media purposes, please inform the journal staff as soon as possible so that your submission can be scheduled accordingly. Your manuscript will remain under a strict press embargo until the publication date and time. This means an early version of your manuscript will not be published ahead of your final version. PLOS Genetics may also choose to issue a press release for your article. If there's anything the journal should know or you'd like more information, please get in touch via plosgenetics@plos.org.

Acceptance letter

Gregory P Copenhaver, Fengwei Yu

20 Sep 2023

PGENETICS-D-23-00514R1

TDP-43 pathology in Drosophila induces glial-cell type specific toxicity that can be ameliorated by knock-down of SF2/SRSF1.

Dear Dr Dubnau,

We are pleased to inform you that your manuscript entitled "TDP-43 pathology in Drosophila induces glial-cell type specific toxicity that can be ameliorated by knock-down of SF2/SRSF1." has been formally accepted for publication in PLOS Genetics! Your manuscript is now with our production department and you will be notified of the publication date in due course.

The corresponding author will soon be receiving a typeset proof for review, to ensure errors have not been introduced during production. Please review the PDF proof of your manuscript carefully, as this is the last chance to correct any errors. Please note that major changes, or those which affect the scientific understanding of the work, will likely cause delays to the publication date of your manuscript.

Soon after your final files are uploaded, unless you have opted out or your manuscript is a front-matter piece, the early version of your manuscript will be published online. The date of the early version will be your article's publication date. The final article will be published to the same URL, and all versions of the paper will be accessible to readers.

Thank you again for supporting PLOS Genetics and open-access publishing. We are looking forward to publishing your work!

With kind regards,

Zsofi Zombor

PLOS Genetics

On behalf of:

The PLOS Genetics Team

Carlyle House, Carlyle Road, Cambridge CB4 3DN | United Kingdom

plosgenetics@plos.org | +44 (0) 1223-442823

plosgenetics.org | Twitter: @PLOSGenetics

Associated Data

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

    Supplementary Materials

    S1 Fig. Nuclear RNA-seq Schematic.

    Flies expressing UAS-TDP-43 and the INTACT reporter (5XUAS-unc84-2XGFP) under control of a temperatures sensitive Gal80 plus a glial cell-type-specific Gal4 (methods) were (A) reared at 21°C and shifted to 29°C upon eclosion (day 0). Heads were collected for sequencing on day 2 and stored at -80°C until use. (B) schematic of the INTACT nuclear tag used to purify nuclei and schematic of the tandem affinity purification process.

    (PDF)

    S2 Fig. Gene expression profiles; overlap, PCA, and correlation plots.

    Gene expression profiles generated from (A) Pearson correlation analysis demonstrating EG and CG more closely resemble each other at baseline than they do their own cell type with TDP-43 expression (B) principle component analysis plot demonstrating PNG and SPG separate from other glial cell types. (C) upregulated and downregulated genes (p < .05) in a given glial cell type and their overlap with other glial cell types.

    (PDF)

    S3 Fig. Pathological expression of human TDP-43 in PNG causes loss of neurons.

    The PNG-Gal4 and Gal80ts were used to induce post development expression of hTDP-43 and UAS- Watermelon (WM), a dual reporter module that encodes both a myristylated GFP that tethers to the membrane (myr-GFP-V5) and nuclear localized mCherry (H2B-mCherry-HA). As in other figures, flies were reared at 21°C and shifted to 29°C upon eclosion (day 0). To quantify effects on neuron number, brains were dissected at day 10 (D10) post temperature shift. PNG glia were visualized using the mCherry nuclear reporter, and neuronal nulei were visualized using the Elav antibody. The numbers of Elav-labeled neuronal nuclei were quantified in A 4.68 μm stack (10 sections, 0.52 μm intervals) of ventral, posterior brain region as depicted with a yellow box in the image at left. This reveals a statistically significant (N = 15; Student’s T-test; P = 0.0319) reduction in numbers of neurons in TDP-43 expressing (PNG-Gal4ts:UAS-TDP-43/UAS-WM) vs controls (PNG-Gal4ts>UAS-WM). Means and SEM are shown.

    (PDF)

    S1 Table. Numbers of fly heads used for TAPIN purification and sequencing based on number of glial cells of each type per brain.

    (PDF)

    S2 Table. Statistically significant differentially expressed features.

    (XLSX)

    S3 Table. Statistically significant differentially expressed TEs.

    (XLSX)

    S4 Table. DNA damage/repair pathways.

    (XLSX)

    S5 Table. Chromatin organization pathway.

    (XLSX)

    S6 Table. Nucleocytoplasmic transport pathway.

    (XLSX)

    S7 Table. Inflammatory response pathways.

    (XLSX)

    S8 Table. SF2 expression across glial cell types. SF2 expression was significantly reduced at day 2 post induction of TDP-43 in SPG and PNG day 2.

    (PDF)

    Attachment

    Submitted filename: responses to reviewers.pdf

    Data Availability Statement

    All of the raw sequencing data are available on GEO (GSE242209;https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE242209) All other data are included in the main figures and tables and in the supplemental figures and tables.


    Articles from PLOS Genetics are provided here courtesy of PLOS

    RESOURCES