Enhanced insulin signalling ameliorates C9orf72 hexanucleotide repeat expansion toxicity in Drosophila

  1. Magda L Atilano
  2. Sebastian Grönke
  3. Teresa Niccoli
  4. Liam Kempthorne
  5. Oliver Hahn
  6. Javier Morón-Oset
  7. Oliver Hendrich
  8. Miranda Dyson
  9. Mirjam Lisette Adams
  10. Alexander Hull
  11. Marie-Therese Salcher-Konrad
  12. Amy Monaghan
  13. Magda Bictash
  14. Idoia Glaria
  15. Adrian M Isaacs  Is a corresponding author
  16. Linda Partridge  Is a corresponding author
  1. Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, United Kingdom
  2. UK Dementia Research Institute at UCL, United Kingdom
  3. Max Planck Institute for Biology of Ageing, Germany
  4. Department of Neurodegenerative Disease, UCL Institute of Neurology, United Kingdom
  5. Alzheimer's Research United Kingdom UCL Drug Discovery Institute, University College London, United Kingdom
6 figures, 1 table and 1 additional file

Figures

Figure 1 with 4 supplements
Insulin signalling is down-regulated in flies expressing C9orf72 repeats.

(A) Gene ontology enrichment of genes with altered expression when GR100 was expressed in neurons. In the top graph, bars represent enrichment of up- and down-regulated genes. In the bottom graph, upper bars represent only up-regulated genes, lower bars represent only down-regulated genes. Lengths of bars represent negative log-transformed, adjusted p-values for Fisher’s exact enrichment test. Bar colour indicates log2-fold changes between GR100 and control per gene. Neuropeptide/hormone activity genes were down-regulated. (B) Quantitative RT-PCR analysis of dilp 2, 3, and 5 normalized against tubulin in fly heads expressing GR100 in neurons. Data was assessed by t-test and presented as mean ± SD, n = 3; dilp2: p=0.0004; dilp3: p<0.0001, dilp5: p<0.0001. Genotypes: (A) w; UAS-GR100/+; ElavGS/+ (GR100), w; +; ElavGS/+ (ctrl) and (B) w; UAS-GR100/+; ElavGS/+.

Figure 1—figure supplement 1
Expression of GR100 in fly neurons induces strong perturbation of the transcriptome.

(A) MA-plot of DESeq2 output indicating genes that were significantly differentially expressed (red dots), non-significantly differentially expressed (grey circles), and significantly expressed outside the 4 to −4 log-fold change limit (red triangles) in GR100 over-expression flies relative to control flies. Dilp2, 3, and 5 are annotated (blue circles). (B) Graphs display the relative expression of all genes associated with the identified GO-term categories with altered expression. Log2-fold change of the genes in GO categories: DNA repair, proteasome complex, spliceosome complex, RISC complex, and small and large ribosomal subunits. (A and B) Genotypes: w; UAS-GR100/+; ElavGS/+ (GR100) and w; +; ElavGS/+ (ctrl).

Figure 1—figure supplement 2
Insulin-like peptides 2, 3, and 5 are down-regulated in flies expressing (G4C2)36.

Quantitative RT-PCR analysis of dilp 2, 3, and 5 normalized against tubulin in fly heads expressing (G4C2)36 in neurons. Blue circles show data obtained from uninduced flies, red squares data obtained from induced flies. Data was assessed by t-test and presented as mean ± SD, n = 3, *p=0.017, **p=0.008, ns p=0.0620. Genotype: w; UAS-(G4C2)36 /+; ElavGS/+.

Figure 1—figure supplement 3
Expression of GR100 in fly neurons does not induce loss of IPCs.

(A) Representative confocal images of dilp2 immunostaining (green) in the IPCs of adult female flies expressing neuronal GR100 (induced) or not (non-induced). (B) Quantified IPC number present in brains of non-induced and induced flies expressing GR100. No significant difference (p=0.694) was found. Data was assessed by t-test and presented as mean ± SD, n = 5. (C) Quantified dilp2 mean immunofluorescence intensities in IPCs. Flies expressing GR100 had significantly reduced dilp2 levels (Mann-Witney test, ***p<0.0001). Data are presented as mean ± SD, n = 24. (D) IPC immunostaining with anti-GR (red) and anti-GFP (green) antibodies. Local expression of poly-GR repeats in IPCs using dilp3-Gal4 driver showed that the IPCs were still able to produce GR100 (red). Flies expressing GFP in IPCs were used as control. Genotypes (A), (B), and (C) w; UAS-GR100/+; ElavGS/+. (D) w; UAS-GR100 /dilp3-Gal4 and w; UAS-mcd8-GFP /dilp3-Gal4.

Figure 1—figure supplement 4
Insulin pathway activity is down-regulated in flies expressing (G4C2)36.

(A) Insulin signalling pathway. Insulin binds to insulin receptor, activating insulin receptor substrates and subsequently PI3K-Akt-mTOR pathway. SF1670 is a PTEN inhibitor and MK2206 is a pan-Akt inhibitor. (B) Western blot analysis of the phosphorylated and non-phosphorylated 4E-BP1. Flies expressing (G4C2)36 had a significantly decreased ratio of phosphorylated 4E-BP1 to the non-phosphorylated form compared with flies expressing both (G4C2)36 and InRActive (*p=0.03, unpaired t-test). Data are presented as mean ± SD, n = 3. Genotype (B) w; UAS-(G4C2)36/+; ElavGS/+ and w; UAS-(G4C2)36 /UAS-InRActive; ElavGS/+.

Figure 2 with 1 supplement
Activation of insulin signalling reduces G4C2 repeat toxicity in vivo.

(A) Lifespan of flies (n = 150) expressing (G4C2)36 or co-expressing InR constructs (InRActive, InRDN) in neurons. Lifespan was significantly extended in (G4C2)36 disease flies co-expressing InRActive compared with (G4C2)36 expressing flies (***p=2.8×10−21 – log-rank test) and decreased when co-expressed with InRDN (*p=0.027). (B) Lifespan of wild-type flies (n = 150) expressing InRActive or InRDN in neurons. Lifespan was significantly reduced in flies expressing InRActive compared with control flies (***p=2.64×10−6 – log-rank test) and increased in flies expressing InRDN (**p=0.0035). (C) Total activity of flies expressing (G4C2)36 in neurons was significantly reduced compared with uninduced control flies (***p=0.0003). (G4C2)36 flies co-expressing InRActive showed increased activity (*p=0.018) compared with flies expressing (G4C2)36 alone (two-away ANOVA followed by Holm-Sidak’s comparison test). Data are presented as mean with SD (n = 30 per genotype). (D) Flies expressing (G4C2)36 alone had significantly increased levels of p53 compared with flies expressing InRactive (**p=0.0014, t-test). Data are presented as mean ±95% confidence intervals, n = 3. Genotypes (A) w; UAS-(G4C2)36/+; ElavGS/+, w; UAS-(G4C2)36/UAS-InRActive; ElavGS/+, w; UAS-(G4C2)36/UAS-InRDN; ElavGS/+. (B) w; ElavGS/+, w; +/UAS-InRActive; ElavGS/+, w; +/UAS-InRDN; ElavGS/+. (C and D) w; UAS-(G4C2)36/+; ElavGS/+, w; UAS-(G4C2)36/UAS-InRActive; ElavGS/+.

Figure 2—figure supplement 1
Expression of InR construct does not affect fly feeding or ElavGS expression system.

(A) There were no significant differences in food intake between flies expressing (G4C2 alone or co-expressing InR constructs, or between flies induced or uninduced with RU486, one-way ANOVA, followed by Tukey’s multiple comparisons test). Data is presented as mean with standard errors of the mean. (B) Dot blot analysis of (G4C2)36 transcript levels in flies ubiquitously expressing (G4C2)36 repeats alone or co-expressing InRActive and InRDN transgenes showed no significant differences across the genotypes (one-way ANOVA, followed by Dunnett’s multiple comparisons test). (C) GFP expression levels in UAS-GFP; ElavGS flies co-expressing InRActive or InRDN determined by western blot analysis. There were no significant differences (p=0.4) in GFP levels between genotypes (one-way ANOVA, followed by Tukey’s multiple comparisons test). Data is presented as mean with ±95% confidence intervals, n = 3. (D) Expression of (G4C2)36 in neurons increased day sleep of flies (***p<0.0001, two-away ANOVA followed by Holm-Sidak’s comparison test), while this was abolished when (G4C2)36 was co-expressed with InRActive (ns; p=0.87). Data are mean ± SD (n = 32 per genotype). (E) Expression of (G4C2)36 in neurons increased night sleep of flies (**p=0.0007, two-away ANOVA followed by Holm-Sidak’s comparison test), while co-expression of InRActive abolished this phenotype (ns; p=0.81). Data are mean ± SD (n = 32 per genotype). Genotype (A) w; UAS-(G4C2)36/+; ElavGS/+, w; UAS-(G4C2)36/UAS-InRActive; ElavGS/+ and w; UAS-(G4C2)36 /UAS-InRDN; ElavGS/+. (B) w; da-GS, UAS-(G4C2)36/+, w; da-GS, UAS-(G4C2)36/UAS-InRActive and w; da-GS, UAS-(G4C2)36/UAS-InRDN. (C) w; UAS-GFP/+; ElavGS/+, w; UAS-GFP/UAS-InRActive; ElavGS/+ and w; UAS-GFP/UAS-InRDN; ElavGS/+. (D and E) w; UAS-(G4C2)36/+; ElavGS/+, w; UAS-(G4C2)36/UAS-InRActive; ElavGS/+.

Activation of insulin signalling reduces poly-GR toxicity via InR/PI3K/Akt.

(A) Stereomicroscopy images of representative 2-day-old adult Drosophila eyes expressing InRActive or InRDN using the GMR-GAL4 driver (bottom panel) or co-expressing both GR36 and InR constructs (top panel). (B) Eye size of flies (n = 10 per genotype) normalized to the mean of the control eye size. Expression of InRActive in a wild-type background with GMR driver caused eye overgrowth, while InRDN decreased eye size (p<0.001). Co-expression of the GR36 with InRDN greatly decreased eye size (***p<0.0001), while with InRActive substantially increased it (two-way ANOVA followed by Holm-Sidak’s multiple comparison test). Two-way ANOVA showed a significant interaction between InR genotype and expression of the repeats (p<0.0001). Data is presented as mean ± 95% confidence intervals. (C) Eye size (n = 20) of 2-day-old adult Drosophila eyes expressing dilp2, PI3KCA, or AktCA using the GMR-GAL4 driver. Co-expression of PI3KCA or AktCA with GR36 repeats yielded a partial rescue of the size of the eye (***p<0.0001 and *p=0.036 respectively, two-way ANOVA, followed by Holm-Sidak’s multiple comparison test). Data are presented as mean ± SD. Genotypes: (A and B) w; GMR-Gal4/+, w; GMR-GAL4/UAS-InRActive, w; GMR-GAL4/UAS-InRDN, w; GMR-Gal4, UAS-GR36/+, w; GMR-Gal4, UAS-GR36/UAS-InRActive, w; GMR-Gal4, UAS-GR36/UAS-InRDN. (C) w; GMR-Gal4, UAS-GR36/+, w; GMR-Gal4, UAS-GR36/+;UAS-dilp2/+, w/PI3KCA; GMR-Gal4, UAS-GR36/+, w; GMR-Gal4, UAS-GR36/AktCA, w; GMR-Gal4/+, w; GMR-Gal4/+; UAS-dilp2/+, w/PI3KCA; GMR-Gal4/+, w; GMR-Gal4/AktCA.

Activation of insulin signalling reduces poly-GR levels in flies.

(A) GR dipeptide levels determined by Meso Scale Discovery (MSD) immunoassay were reduced in (G4C2)36 flies expressing InRActive compared to (G4C2)36 alone flies (***p=0.0001, one-way ANOVA, followed by Tukey’s multiple comparisons test). Levels of GR were normalized to the mean GR levels of control (G4C2)36 flies. Data is presented as mean ± SD, n = 8. (B) Expression of poly-GR determined by MSD immunoassay was also reduced in flies expressing both GR100 and InRActive compared to flies expressing GR100 alone (**p=0.0025, one-way ANOVA followed by Tukey’s multiple comparison test). Co-expression of InRDN slightly reduced poly-GR levels (*p=0.044). Levels of GR were normalized to the mean GR levels of control (G4C2)36. Data are presented as mean ± SD, n = 5. (C) Lifespan was significantly extended in flies (expressing ATG driven GR100 with over-expression of InRActive compared to flies only expressing GR100; ***p=1.62×10−11 – log rank test). Genotypes (A) w; UAS-(G4C2)36/+; ElavGS/+, w; UAS-(G4C2)36/UAS-InRActive; ElavGS/+, w; UAS-(G4C2)36/UAS-InRDN; ElavGS/+. (B, C) w; UAS-GR100/+; ElavGS/+, w; UAS-GR100/UAS-InRActive; ElavGS/+, w; UAS-GR100/UAS-InRDN; ElavGS/+.

Poly-GR levels are increased by Akt inhibition and decreased by PTEN inhibition in mammalian cells.

Poly-GR levels were measured using a NLuc reporter assay following a 48 hr treatment with either 1 µM MK2206 (AKT inhibitor) or SF1670 (PTEN inhibitor). (A) MK2206 significantly increases poly-GR levels (*p=0.0168). (B) SF1670 significantly decreases poly-GR levels (*p=0.0401). Each NLuc reading was normalized to FLuc for each well and further normalized to DMSO control treatment. Data given as mean ± SD of 4 biological replicates with 3–4 technical replicates per biological replicate. Data analyzed via two-tailed, unpaired Student’s t-test on the mean of each biological repeat.

Figure 6 with 1 supplement
Systemic injection of insulin rescues (G4C2)36 toxicity in Drosophila.

Injection of 0.03 mg/ml insulin (INS) significantly extended lifespan of flies (n = 120) expressing (G4C2)36 when compared with flies injected with PBS (**p=0.00034, log-rank test), while it slightly shortened lifespan in non-induced flies (*p=0.043). Genotype: w; UAS-(G4C2)36 /+; ElavGS/+.

Figure 6—figure supplement 1
Systemic injection of insulin reduces (G4C2)36 toxicity in Drosophila.

(A) Injection of different concentrations of insulin 0.03, 0.3, and 3 mg/ml into (G4C2)36 flies haemolymph (n = 120). Injection of 3 mg/ml insulin into (G4C2)36-induced flies greatly shortened their lifespan (p<0.0001, log-rank test), while 0.3 mg/ml had no significant effect (p=0.083), and 0.03 mg/ml modestly increased lifespan (p=0.038, log-rank test, PBS induced vs. 0.03 mg/ml INS induced). In uninduced (G4C2)36 flies, injection of insulin at 0.3 or 0.03 mg/ml shortened lifespan (p<0.0001, log-rank test, PBS uninduced flies vs. 0.3 mg/ml INS uninduced; p=0.039, log-rank test, PBS uninduced flies vs 0.03 mg/ml INS uninduced). (B) Injection of insulin (INS) at 0.03 mg/ml significantly extended lifespan of flies (n = 80) expressing (G4C2)36 when compared with flies injected with PBS (*p=0.037, log-rank test) in induced flies, while it shortened lifespan in non-induced flies (*p=0.007). Genotype (A, B) w; UAS-(G4C2)36/+; ElavGS/+.

Tables

Key resources table
Reagent type
(species) or resource
DesignationSource or referenceIdentifiersAdditional information
Genetic reagent (Drosophila melanogaster)Elav-GSOsterwalder et al., 2001
Genetic reagent (D. melanogaster)Da-GSTricoire et al., 2009
Genetic reagent (D. melanogaster)GMR-GAL4Bloomington Drosophila Stock CenterBL#9146
RRID:BDSC_9146
Genetic reagent (D. melanogaster)Dilp3-GAL4Bloomington Drosophila Stock CenterBL#52660
RRID:BDSC_52660
Genetic reagent (D. melanogaster)UAS-(G4C2)36Mizielinska et al., 2014
Genetic reagent (D. melanogaster)UAS-GR100Mizielinska et al., 2014
Genetic reagent (D. melanogaster)UAS-InRActiveBloomington Drosophila Stock CenterBL#8263
RRID:BDSC_8263
Genetic reagent (D. melanogaster)UAS-InRDNBloomington Drosophila Stock CenterBL#8252
RRID:BDSC_8252
Genetic reagent (D. melanogaster)UAS-PI3KCABloomington Drosophila Stock CenterBL#25908
RRID:BDSC_25908
Genetic reagent (D. melanogaster)UAS-AktCABloomington Drosophila Stock CenterBL#8194
RRID:BDSC_8194
Genetic reagent (D. melanogaster)UAS-mCD8::GFPLee and Luo, 1999
Cell line
(Homo sapiens)
HEK293T cellsUCL Drug Discovery InstituteMycoplasma negative HEK cells
Recombinant DNA reagentpGL4.53[luc2/PGK] VectorPromega#E5011Firefly luciferase reporter plasmid
Transfected construct
(H. sapiens)
92 repeat G4C2nanoluciferase reporterUCL Dementia Research Institute
AntibodyAnti-GFP (mouse, mix of two monoclonals)MerckCat#11814460001
RRID:AB_390913
WB (1:10.000)
AntibodyAnti-GR
(rabbit)
Moens et al., 2018MSD
Capture: 2 µg/ml
Detection: 12 µg/ml
AntibodyAnti-GR
(rat, monoclonal)
Mori et al., 20135H9IF (1:50)
AntibodyAnti-dilp2
(rabbit, polyclonal)
Okamoto et al., 2012IF (1:500)
AntibodyAnti-non-P 4E-BP1(rabbit monoclonal)Cell SignallingCat#4923:
RRID:AB_659944
WB (1:1000)
AntibodyAnti-P 4E-BP1 (rabbit monoclonal)Cell SignallingCat#2855
RRID:AB_560835
WB (1:1000)
AntibodyAnti-p53 (mouse monoclonal)DSHBDmp53-H3
RRID:AB_10804170
WB
(1:200)
AntibodyAnti-actin
(mouse monoclonal)
AbcamCat#Ab8224
RRID:AB_449644
WB (1:10.0000)
AntibodyAnti-tubulin
(mouse monoclonal)
Sigma- AldrichCat#T6199
RRID:AB_477583
WB (1:2000)
AntibodyAnti-rat IgG-Alexa fluor 647
(goat polyclonal)
ThermoFisherCat#A21247
RRID:AB_141778
IF (1:1000)
AntibodyAnti-rabbit IgG-Alexa fluor 488 (goat polyclonal)ThermoFisherCat#A32731
RRID:AB_2633280
IF (1:1000)
AntibodyHRP-conjugated anti-mouse (goat polyclonal)AbcamCat#Ab6789
RRID:AB_955439
WB (1:10.000)
AntibodyHRP-conjugated anti-rabbit (goat polyclonal)AbcamCat#Ab6721
RRID:AB_955447
WB (1:10.000)
Sequence-based reagentDilp2_forwardBroughton et al., 2008PCR primersATGAGCAAGCCTTTGTCCTTC
Sequence-based reagentDilp2_reverseBroughton et al., 2008PCR primersGACCACGGAGCAGTACTCCC
Sequence-based reagentDilp3_forwardThis studyPCR primersAGAGAACTTTGGACCCCGTGAA
Sequence-based reagentDilp3_reverseThis studyPCR primersTGAACCGAACTATCACTCAACAGTCT
Sequence-based reagentDilp5_forwardThis studyPCR primersGAGGCACCTTGGGCCTATTC
Sequence-based reagentDilp5_reverseThis studyPCR primersCATGTGGTGAGATTCGGAGCTA
Sequence-based reagentTubulin_forwardMoens et al., 2019PCR primersTGGGCCCGTCTGGACCACAA
Sequence-based reagentTubulin_reverseMoens et al., 2019PCR primersTCGCCGTCACCGGAGTCCAT

Additional files

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Magda L Atilano
  2. Sebastian Grönke
  3. Teresa Niccoli
  4. Liam Kempthorne
  5. Oliver Hahn
  6. Javier Morón-Oset
  7. Oliver Hendrich
  8. Miranda Dyson
  9. Mirjam Lisette Adams
  10. Alexander Hull
  11. Marie-Therese Salcher-Konrad
  12. Amy Monaghan
  13. Magda Bictash
  14. Idoia Glaria
  15. Adrian M Isaacs
  16. Linda Partridge
(2021)
Enhanced insulin signalling ameliorates C9orf72 hexanucleotide repeat expansion toxicity in Drosophila
eLife 10:e58565.
https://doi.org/10.7554/eLife.58565