Insulin signaling misregulation underlies circadian and cognitive deficits in a Drosophila fragile X model

R E Monyak, D Emerson, B P Schoenfeld, X Zheng, D B Chambers, C Rosenfelt, S Langer, P Hinchey, C H Choi, T V McDonald, F V Bolduc, A Sehgal, S M J McBride, T A Jongens, R E Monyak, D Emerson, B P Schoenfeld, X Zheng, D B Chambers, C Rosenfelt, S Langer, P Hinchey, C H Choi, T V McDonald, F V Bolduc, A Sehgal, S M J McBride, T A Jongens

Abstract

Fragile X syndrome (FXS) is an undertreated neurodevelopmental disorder characterized by low intelligence quotent and a wide range of other symptoms including disordered sleep and autism. Although FXS is the most prevalent inherited cause of intellectual disability, its mechanistic underpinnings are not well understood. Using Drosophila as a model of FXS, we showed that select expression of dfmr1 in the insulin-producing cells (IPCs) of the brain was sufficient to restore normal circadian behavior and to rescue the memory deficits in the fragile X mutant fly. Examination of the insulin signaling (IS) pathway revealed elevated levels of Drosophila insulin-like peptide 2 (Dilp2) in the IPCs and elevated IS in the dfmr1 mutant brain. Consistent with a causal role for elevated IS in dfmr1 mutant phenotypes, the expression of dfmr1 specifically in the IPCs reduced IS, and genetic reduction of the insulin pathway also led to amelioration of circadian and memory defects. Furthermore, we showed that treatment with the FDA-approved drug metformin also rescued memory. Finally, we showed that reduction of IS is required at different time points to rescue circadian behavior and memory. Our results indicate that insulin misregulation underlies the circadian and cognitive phenotypes displayed by the Drosophila fragile X model, and thus reveal a metabolic pathway that can be targeted by new and already approved drugs to treat fragile X patients.

Conflict of interest statement

Conflict of Interest: The authors declare no conflicts of interest.

Figures

Figure 1
Figure 1
Expression of dFMR1 in the IPCs of the brain rescues defects in circadian behavior. (a to d) Panels show the percent rhythmic (black) and relative FFT values (white) for genetic combinations testing the spatial requirement of dFMR1 expression in dfmr1 mutants for normal circadian behavior. Relative FFT represents how the average FFT of the depicted genotype compares to the average FFT of a wild-type control. (a)Dfmr1 mutants expressing dfmr1 pan-neuronally display an increased percentage of rhythmic flies and more robust circadian rhythmicity than dfmr1 mutants with either transgene alone, p<0.001. (b) Circadian behavior of dfmr1 mutants with both pdf-Gal4 or cry-Gal4 and UAS-dfmr1 is not significantly different from dfmr1 mutants with any of the relevant transgenes alone. (c) Circadian behavior of dfmr1 mutants with tim-Gal4 or per-Gal4 and UAS-dfmr1 is not significantly improved compared to dfmr1 mutants with any of the relevant transgenes alone. (d) Circadian behavior of dfmr1 mutants with both dilp2R-Gal4 or dilp2W-Gal4 and UAS-dfmr1is significantly improved relative to dfmr1 mutants with any of the relevant transgenes alone, p<0.001. Statistically significant levels of rescue are denoted with asterisks (*p<0.05, ** p<0.01, ***p<0.001). Error bars represent s.e.m.
Figure 2
Figure 2
The IS pathway is upregulated in dfmr1 mutant brains. (a) Dilp2 protein levels in the IPC cell bodies of dfmr1 mutant brains are higher than in controls (dfmr1 mutants containing a WTrescue transgene which expresses dfmr1 at wild-type levels). DE-cadherin was used as a staining control. (b) Quantification reveals Dilp2 is significantly increased in dfmr1 mutants, p<0.001. (c) The GFP-PH reporter is more localized to the membrane in the cells of dfmr1 mutant than in controls. Brains were imaged on their posterior side in the mushroom body calyx region. (d) Quantification of reporter distribution shown as a ratio of membrane/cytoplasm fluorescence. Dfmr1 mutants show a significantly higher ratio, p<0.05. The membrane/fluorescence ratio was also calculated for the DE-cadherin staining control and was found to be the same in both genotypes (not shown). (e) A notable decrease in p-S505-Akt levels is observed in dfmr1 mutants that have dfmr1 expressed in the IPCs. (f) Quantification of p-S505-Akt fluorescence reveals that dfmr1 mutants expressing dfmr1 in the IPCs show significantly lower p-S505-Akt fluorescence than either dfmr1 mutant control with the driver or UAS-construct alone, p<0.001 and p<0.05. p-S505-Akt fluorescence was normalized to DE-cadherin fluorescence. All images in this figure are representative of quantification.
Figure 3
Figure 3
Genetic reduction of the insulin pathway rescues the circadian defect observed in dfmr1 mutants. (a to d) Panels show the percentage of rhythmic flies (black) and relative FFT values (white) for genetic combinations testing the effect of reducing IS in dfmr1 mutants on circadian behavior. (a) Circadian behavior (as indicated by increased percentage of rhythmic flies and increased relative FFT) of dfmr1 mutants with the WTrescue transgene or with one copy of a null allele of dilp2 (dilp2/+; dfmr1-) is significantly improved relative to dfmr1 mutant controls, p<0.001. (b) Circadian behavior of dfmr1 mutants with the WTrescue transgene or with one copy of a mutant allele of the insulin receptor (InR/+; dfmr1-) is significantly improved relative to dfmr1 mutant controls, p<0.001. (c) Circadian behavior of dfmr1 mutants with both elav-Gal4 and UAS-DP110DN (elav>DP110DN; dfmr1-) is significantly improved relative to dfmr1 mutants with either transgene alone (elav-Gal4; dfmr1-) and (UAS-DP110DN; dfmr1-), p<0.01. (d) Circadian behavior of dfmr1 mutants with both elav-Gal4 and UAS-PTEN (elav>PTEN; dfmr1-) is significantly improved relative to dfmr1 mutants with either transgene alone (elav-Gal4; dfmr1-) and (UAS-PTEN; dfmr1), p<0.001 and p<0.05 respectively. Significance denoted as described in Fig. 1. Error bars represent s.e.m.
Figure 4
Figure 4
Expression of dFMR1 in the IPCs and genetic reduction of IS rescue memory in dfmr1 mutants. (a to d) STM in the courtship paradigm is presented as a memory index (left) which conveys the difference between the CIs of trained and untrained flies in a single values, and as separate CIs (right). (a) Expression of dfmr1 in the IPCs of dfmr1 mutants rescues STM, p<0.0001, N=16-20 (b) STM is rescued by reduction of dilp2 in dfmr1 mutants, p<0.05. FSrescue represents a frame-shifted version of the dfmr1 open reading frame. N=25-31. (c) STM is rescued by pan-neural expression of DP110DN, p<0.01. N=22-91 or (D) PTEN, p<0.001. N=28-91. (e to h) Performance index (PI) represents the percent of flies which avoid the shock-conditioned odor. (e)Dfmr1 mutants expressing dFMR1 within the IPCs show rescue of learning, (N=4, p=0.0016) and (f) memory (N=8, p=0.0002). (g)Dfmr1 mutants expressing DP110DN pan-neuronally show rescue of learning (N=4, p=0.0235) and (h) memory, (N=8, p=0.0005). Graphs depict mean ± s.e.m.
Figure 5
Figure 5
Metformin treatment rescues memory in dfmr1 mutants. (a) Metformin treatment restores STM to dfmr1 mutants, p<.001 N=36-86. (b) Metformin improves learning (p<0.0001). (N= 6) and (c) memory in dfmr1 mutants.(N=8, p <0.00018). Graphs depict mean ± s.e.m.

References

    1. Santoro MR, Bray SM, Warren ST. Molecular mechanisms of fragile X syndrome: a twenty-year perspective. Annual review of pathology. 2012;7:219–45.
    1. Dolen G, Carpenter RL, Ocain TD, Bear MF. Mechanism-based approaches to treating fragile X. Pharmacology & therapeutics. 2010;127(1):78–93.
    1. Jacquemont S, Hagerman RJ, Hagerman PJ, Leehey MA. Fragile-X syndrome and fragile X-associated tremor/ataxia syndrome: two faces of FMR1. Lancet neurology. 2007;6(1):45–55.
    1. O'Donnell WT, Warren ST. A decade of molecular studies of fragile X syndrome. Annual review of neuroscience. 2002;25:315–38.
    1. Turk J. Fragile X syndrome: lifespan developmental implications for those without as well as with intellectual disability. Current opinion in psychiatry. 2011;24(5):387–97.
    1. Cross J, Yang JC, Johnson FR, Quiroz J, Dunn J, Raspa M, et al. Caregiver Preferences for the Treatment of Males with Fragile X Syndrome. Journal of developmental and behavioral pediatrics : JDBP. 2016;37(1):71–9.
    1. Schaefer TL, Davenport MH, Erickson CA. Emerging pharmacologic treatment options for fragile X syndrome. The application of clinical genetics. 2015;8:75–93.
    1. Zhang YQ, Bailey AM, Matthies HJ, Renden RB, Smith MA, Speese SD, et al. Drosophila fragile X-related gene regulates the MAP1B homolog Futsch to control synaptic structure and function. Cell. 2001;107(5):591–603.
    1. Dockendorff TC, Su HS, McBride SM, Yang Z, Choi CH, Siwicki KK, et al. Drosophila lacking dfmr1 activity show defects in circadian output and fail to maintain courtship interest. Neuron. 2002;34(6):973–84.
    1. McBride SM, Choi CH, Wang Y, Liebelt D, Braunstein E, Ferreiro D, et al. Pharmacological rescue of synaptic plasticity, courtship behavior, and mushroom body defects in a Drosophila model of fragile X syndrome. Neuron. 2005;45(5):753–64.
    1. Bolduc FV, Valente D, Nguyen AT, Mitra PP, Tully T. An assay for social interaction in Drosophila Fragile X mutants. Fly. 2010;4(3):216–25.
    1. Bolduc FV, Bell K, Cox H, Broadie KS, Tully T. Excess protein synthesis in Drosophila fragile X mutants impairs long-term memory. Nature neuroscience. 2008;11(10):1143–5.
    1. Michalon A, Sidorov M, Ballard TM, Ozmen L, Spooren W, Wettstein JG, et al. Chronic pharmacological mGlu5 inhibition corrects fragile X in adult mice. Neuron. 2012;74(1):49–56.
    1. Renn SC, Park JH, Rosbash M, Hall JC, Taghert PH. A pdf neuropeptide gene mutation and ablation of PDF neurons each cause severe abnormalities of behavioral circadian rhythms in Drosophila. Cell. 1999;99(7):791–802.
    1. Emery P, Stanewsky R, Helfrich-Forster C, Emery-Le M, Hall JC, Rosbash M. Drosophila CRY is a deep brain circadian photoreceptor. Neuron. 2000;26(2):493–504.
    1. Kaneko M, Hall JC. Neuroanatomy of cells expressing clock genes in Drosophila: transgenic manipulation of the period and timeless genes to mark the perikarya of circadian pacemaker neurons and their projections. J Comp Neurol. 2000;422(1):66–94.
    1. Glossop NR, Houl JH, Zheng H, Ng FS, Dudek SM, Hardin PE. VRILLE feeds back to control circadian transcription of Clock in the Drosophila circadian oscillator. Neuron. 2003;37(2):249–61.
    1. Rulifson EJ, Kim SK, Nusse R. Ablation of insulin-producing neurons in flies: growth and diabetic phenotypes. Science. 2002;296(5570):1118–20.
    1. Wu Q, Zhang Y, Xu J, Shen P. Regulation of hunger-driven behaviors by neural ribosomal S6 kinase in Drosophila. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(37):13289–94.
    1. Gronke S, Clarke DF, Broughton S, Andrews TD, Partridge L. Molecular evolution and functional characterization of Drosophila insulin-like peptides. PLoS genetics. 2010;6(2):e1000857.
    1. Fernandez R, Tabarini D, Azpiazu N, Frasch M, Schlessinger J. The Drosophila insulin receptor homolog: a gene essential for embryonic development encodes two receptor isoforms with different signaling potential. The EMBO journal. 1995;14(14):3373–84.
    1. Leevers SJ, Weinkove D, MacDougall LK, Hafen E, Waterfield MD. The Drosophila phosphoinositide 3-kinase Dp110 promotes cell growth. The EMBO journal. 1996;15(23):6584–94.
    1. Huang H, Potter CJ, Tao W, Li DM, Brogiolo W, Hafen E, et al. PTEN affects cell size, cell proliferation and apoptosis during Drosophila eye development. Development. 1999;126(23):5365–72.
    1. Siegel S. Nonparametric Statistics. Am Stat. 1957;11(3):13–9.
    1. Keleman K, Kruttner S, Alenius M, Dickson BJ. Function of the Drosophila CPEB protein Orb2 in long-term courtship memory. Nature neuroscience. 2007;10(12):1587–93.
    1. Pepper AS, Beerman RW, Bhogal B, Jongens TA. Argonaute2 suppresses Drosophila fragile X expression preventing neurogenesis and oogenesis defects. PloS one. 2009;4(10):e7618.
    1. Pinal N, Goberdhan DC, Collinson L, Fujita Y, Cox IM, Wilson C, et al. Regulated and polarized PtdIns(3,4,5)P3 accumulation is essential for apical membrane morphogenesis in photoreceptor epithelial cells. Current biology : CB. 2006;16(2):140–9.
    1. Vandesompele J, De Preter K, Pattyn F, Poppe B, Van Roy N, De Paepe A, et al. Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes. Genome biology. 2002;3(7) RESEARCH0034.
    1. Ling D, Salvaterra PM. Robust RT-qPCR data normalization: validation and selection of internal reference genes during post-experimental data analysis. PloS one. 2011;6(3):e17762.
    1. Morales J, Hiesinger PR, Schroeder AJ, Kume K, Verstreken P, Jackson FR, et al. Drosophila fragile X protein, DFXR, regulates neuronal morphology and function in the brain. Neuron. 2002;34(6):961–72.
    1. Brand AH, Perrimon N. Targeted gene expression as a means of altering cell fates and generating dominant phenotypes. Development. 1993;118(2):401–15.
    1. Nitabach MN, Taghert PH. Organization of the Drosophila circadian control circuit. Current biology : CB. 2008;18(2):R84–93.
    1. Helfrich-Forster C, Homberg U. Pigment-dispersing hormone-immunoreactive neurons in the nervous system of wild-type Drosophila melanogaster and of several mutants with altered circadian rhythmicity. J Comp Neurol. 1993;337(2):177–90.
    1. Reeve SP, Bassetto L, Genova GK, Kleyner Y, Leyssen M, Jackson FR, et al. The Drosophila fragile X mental retardation protein controls actin dynamics by directly regulating profilin in the brain. Current biology : CB. 2005;15(12):1156–63.
    1. Helfrich-Forster C. Robust circadian rhythmicity of Drosophila melanogaster requires the presence of lateral neurons: a brain-behavioral study of disconnected mutants. Journal of comparative physiology A, Sensory, neural, and behavioral physiology. 1998;182(4):435–53.
    1. Helfrich-Forster C, Stengl M, Homberg U. Organization of the circadian system in insects. Chronobiology international. 1998;15(6):567–94.
    1. Britton JS, Lockwood WK, Li L, Cohen SM, Edgar BA. Drosophila's insulin/PI3-kinase pathway coordinates cellular metabolism with nutritional conditions. Developmental cell. 2002;2(2):239–49.
    1. Sharma A, Hoeffer CA, Takayasu Y, Miyawaki T, McBride SM, Klann E, et al. Dysregulation of mTOR signaling in fragile X syndrome. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2010;30(2):694–702.
    1. Gross C, Nakamoto M, Yao X, Chan CB, Yim SY, Ye K, et al. Excess phosphoinositide 3-kinase subunit synthesis and activity as a novel therapeutic target in fragile X syndrome. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2010;30(32):10624–38.
    1. Kim SA, Choi HC. Metformin inhibits inflammatory response via AMPK-PTEN pathway in vascular smooth muscle cells. Biochemical and biophysical research communications. 2012;425(4):866–72.
    1. Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, et al. Role of AMP-activated protein kinase in mechanism of metformin action. The Journal of clinical investigation. 2001;108(8):1167–74.
    1. Lee JH, Budanov AV, Park EJ, Birse R, Kim TE, Perkins GA, et al. Sestrin as a feedback inhibitor of TOR that prevents age-related pathologies. Science. 2010;327(5970):1223–8.
    1. Davis RL. Olfactory memory formation in Drosophila: from molecular to systems neuroscience. Annual review of neuroscience. 2005;28:275–302.
    1. Griffith LC, Ejima A. Multimodal sensory integration of courtship stimulating cues in Drosophila melanogaster. Annals of the New York Academy of Sciences. 2009;1170:394–8.
    1. Joiner Ml A, Griffith LC. CaM kinase II and visual input modulate memory formation in the neuronal circuit controlling courtship conditioning. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1997;17(23):9384–91.
    1. McGuire SE, Le PT, Osborn AJ, Matsumoto K, Davis RL. Spatiotemporal rescue of memory dysfunction in Drosophila. Science. 2003;302(5651):1765–8.
    1. Nassel DR, Kubrak OI, Liu Y, Luo J, Lushchak OV. Factors that regulate insulin producing cells and their output in Drosophila. Frontiers in physiology. 2013;4:252.
    1. Milochau A, Lagree V, Benassy MN, Chaignepain S, Papin J, Garcia-Arcos I, et al. Synaptotagmin 11 interacts with components of the RNA-induced silencing complex RISC in clonal pancreatic beta-cells. FEBS letters. 2014;588(14):2217–22.
    1. Fernandez AM, Torres-Aleman I. The many faces of insulin-like peptide signalling in the brain. Nature reviews Neuroscience. 2012;13(4):225–39.
    1. Song J, Wu L, Chen Z, Kohanski RA, Pick L. Axons guided by insulin receptor in Drosophila visual system. Science. 2003;300(5618):502–5.
    1. Callan MA, Clements N, Ahrendt N, Zarnescu DC. Fragile X Protein is required for inhibition of insulin signaling and regulates glial-dependent neuroblast reactivation in the developing brain. Brain research. 2012;1462:151–61.
    1. Dolen G, Osterweil E, Rao BS, Smith GB, Auerbach BD, Chattarji S, et al. Correction of fragile X syndrome in mice. Neuron. 2007;56(6):955–62.
    1. Nowicki ST, Tassone F, Ono MY, Ferranti J, Croquette MF, Goodlin-Jones B, et al. The Prader-Willi phenotype of fragile X syndrome. Journal of developmental and behavioral pediatrics : JDBP. 2007;28(2):133–8.
    1. Hoeffer CA, Sanchez E, Hagerman RJ, Mu Y, Nguyen DV, Wong H, et al. Altered mTOR signaling and enhanced CYFIP2 expression levels in subjects with fragile X syndrome. Genes, brain, and behavior. 2012;11(3):332–41.
    1. Yurgel ME, Masek P, DiAngelo J, Keene AC. Genetic dissection of sleep-metabolism interactions in the fruit fly. Journal of comparative physiology A, Neuroethology, sensory, neural, and behavioral physiology. 2015;201(9):869–77.
    1. Itoh TQ, Tanimura T, Matsumoto A. Membrane-bound transporter controls the circadian transcription of clock genes in Drosophila. Genes to cells : devoted to molecular & cellular mechanisms. 2011;16(12):1159–67.
    1. Colombani J, Bianchini L, Layalle S, Pondeville E, Dauphin-Villemant C, Antoniewski C, et al. Antagonistic actions of ecdysone and insulins determine final size in Drosophila. Science. 2005;310(5748):667–70.
    1. Okamoto N, Yamanaka N, Yagi Y, Nishida Y, Kataoka H, O'Connor MB, et al. A fat body-derived IGF-like peptide regulates postfeeding growth in Drosophila. Dev Cell. 2009;17(6):885–91.
    1. Slaidina M, Delanoue R, Gronke S, Partridge L, Leopold P. A Drosophila insulin-like peptide promotes growth during nonfeeding states. Dev Cell. 2009;17(6):874–84.
    1. Kumar S, Chen D, Jang C, Nall A, Zheng X, Sehgal A. An ecdysone-responsive nuclear receptor regulates circadian rhythms in Drosophila. Nature communications. 2014;5:5697.
    1. Slack C, Foley A, Partridge L. Activation of AMPK by the putative dietary restriction mimetic metformin is insufficient to extend lifespan in Drosophila. PloS one. 2012;7(10):e47699.
    1. Miller RA, Chu Q, Xie J, Foretz M, Viollet B, Birnbaum MJ. Biguanides suppress hepatic glucagon signalling by decreasing production of cyclic AMP. Nature. 2013;494(7436):256–60.
    1. Hur KY, Lee MS. New mechanisms of metformin action: Focusing on mitochondria and the gut. Journal of diabetes investigation. 2015;6(6):600–9.

Source: PubMed

3
Se inscrever