Skip to main content

Advertisement

Log in

Targeted Downregulation of dMyc Suppresses Pathogenesis of Human Neuronal Tauopathies in Drosophila by Limiting Heterochromatin Relaxation and Tau Hyperphosphorylation

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Human tauopathies such as Alzheimer’s Disease (AD), frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17), Pick’s disease etc., are a group of neurodegenerative diseases which are characterized by abnormal hyperphosphorylation of tau that leads to formation of neurofibrillary tangles. Recapitulating several features of human neurodegenerative disorders, the Drosophila tauopathy model displays compromised lifespan, locomotor function impairment, and brain vacuolization in adult brain which is progressive and age dependent. Here, we demonstrate that tissue-specific downregulation of the Drosophila homolog of human c-myc proto-oncogene (dMyc) suppresses tau-mediated morphological and functional deficits by reducing abnormal tau hyperphosphorylation and restoring the heterochromatin loss. Our studies show for the first time that the inherent chromatin remodeling ability of myc proto-oncogenes could be exploited to limit the pathogenesis of human neuronal tauopathies in the Drosophila disease model. Interestingly, recent reports on successful uses of some anti-cancer drugs against Alzheimer's and Parkinson's diseases in clinical trials and animal models strongly support our findings and proposed possibility.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Lee VM, Goedert M, Trojanowski JQ (2001) Neurodegenerative tauopathies. Annu Rev Neurosci 24:1121–1159

    Article  CAS  PubMed  Google Scholar 

  2. Iqbal K, Liu F, Gong CX (2015) Tau and neurodegenerative disease: the story so far. Nat Rev Neurol. doi:10.1038/nrneurol.2015.225

    PubMed  Google Scholar 

  3. Rademakers R, Cruts M, van Broeckhoven C (2004) The role of tau (MAPT) in frontotemporal dementia and related tauopathies. Hum Mutat 24:277–295

    Article  CAS  PubMed  Google Scholar 

  4. Wang Y, Mandelkow E (2015) Tau in physiology and pathology. Nat Rev Neurosci. doi:10.1038/nrn.2015.1

    PubMed  Google Scholar 

  5. Lee VM, Kenyon TK, Trojanowski JQ (2005) Transgenic animal models of tauopathies. Biochim Biophys Acta 1739:251–259

    Article  CAS  PubMed  Google Scholar 

  6. Lewis J, McGowan E, Rockwood J, Melrose H, Nacharaju P, Van Slegtenhorst M, Gwinn-Hardy K, Paul Murphy M et al (2000) Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat Genet 25:402–405

    Article  CAS  PubMed  Google Scholar 

  7. Wittmann CW, Wszolek MF, Shulman JM, Salvaterra PM, Lewis J, Hutton M, Feany MB (2001) Tauopathy in Drosophila: neurodegeneration without neurofibrillary tangles. Science 293:711–714

    Article  CAS  PubMed  Google Scholar 

  8. Jackson GR, Wiedau-Pazos M, Sang TK, Wagle N, Brown CA, Massachi S, Geschwind DH (2002) Human wild-type tau interacts with wingless pathway components and produces neurofibrillary pathology in Drosophila. Neuron 34:509–519

    Article  CAS  PubMed  Google Scholar 

  9. Wu TH, Lu YN, Chuang CL, Wu CL, Chiang AS, Krantz DE, Chang HY (2013) Loss of vesicular dopamine release precedes tauopathy in degenerative dopaminergic neurons in a Drosophila model expressing human tau. Acta Neuropathol 125:711–725

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Frost B, Hemberg M, Lewis J, Feany MB (2014) Tau promotes neurodegeneration through global chromatin relaxation. Nat Neurosci 17:357–366

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Cohen-Carmon D, Meshorer E (2012) Polyglutamine (polyQ) disorders: the chromatin connection. Nucleus 3:433–441

    Article  PubMed  PubMed Central  Google Scholar 

  12. Singh MD, Raj K, Sarkar S (2014) Drosophila Myc, a novel modifier suppresses the poly(Q) toxicity by modulating the level of CREB binding protein and histone acetylation. Neurobiol Dis 63:48–61

    Article  CAS  PubMed  Google Scholar 

  13. Hay BA, Wolff T, Rubin GM (1994) Expression of baculovirus P35 prevents cell death in Drosophila. Development 120:2121–2129

    CAS  PubMed  Google Scholar 

  14. Benassayag C, Montero L, Colombié N, Gallant P, Cribbs D, Morello D (2005) Human c-Myc isoforms differentially regulate cell growth and apoptosis in Drosophila melanogaster. Mol Cell Biol 25:9897–9909

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ali YO, Ruan K, Zhai RG (2012) NMNAT suppresses tau-induced neurodegeneration by promoting clearance of hyperphosphorylated tau oligomers in a Drosophila model of tauopathy. Hum Mol Genet 21:237–250

    Article  CAS  PubMed  Google Scholar 

  16. Prober DA, Edgar BA (2000) Ras1 promotes cellular growth in the Drosophila wing. Cell 100:435–446

    Article  CAS  PubMed  Google Scholar 

  17. Lilja T, Qi D, Stabell M, Mannervik M (2003) The CBP coactivator functions both upstream and downstream of Dpp/Screw signaling in the early Drosophila embryo. Dev Biol 262:294–302

    Article  CAS  PubMed  Google Scholar 

  18. Owusu-Ansah E, Yavari A, Banerjee U (2008) A protocol for in vivo detection of reactive oxygen species. Protocol Exch. doi:10.1038/nprot.2008.1023

    Google Scholar 

  19. Sun M, Chen L (2015) Studying tauopathies in Drosophila: a fruitful model. Exp Neurol 274:52–57

    Article  CAS  PubMed  Google Scholar 

  20. Brand AH, Perrimon N (1993) Targeted gene expression as a means of altering cell fates and generating dominant phenotypes. Development 118:401–415

    CAS  PubMed  Google Scholar 

  21. Gallant P (2013) Myc function in Drosophila. Cold Spring Harb Perspect Med 3:a014324

    Article  PubMed  PubMed Central  Google Scholar 

  22. Bonini NM (1999) A genetic model for human polyglutamine-repeat disease in Drosophila melanogaster. Philos Trans R Soc Lond B Biol Sci 354:1057–1060

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fahrbach SE (2006) Structure of the mushroom bodies of the insect brain. Annu Rev Entomol 51:209–232

    Article  CAS  PubMed  Google Scholar 

  24. Fushima K, Tsujimura H (2007) Precise control of fasciclin II expression is required for adult mushroom body development in Drosophila. Dev Growth Differ 49:215–227

    Article  CAS  PubMed  Google Scholar 

  25. Lee G, Leugers CJ (2012) Tau and tauopathies. Prog Mol Biol Transl Sci 107:263–293

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Augustinack JC, Schneider A, Mandelkow EM, Hyman BT (2002) Specific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer's disease. Acta Neuropathol 103:26–35

    Article  CAS  PubMed  Google Scholar 

  27. Feng X, Bai Z, Wang J, Xie B, Sun J, Han G, Song F, Crack PJ et al (2014) Robust gene dysregulation in Alzheimer's disease brains. J Alzheimers Dis 41:587–597

    CAS  PubMed  Google Scholar 

  28. Knoepfler PS, Zhang XY, Cheng PF, Gafken PR, McMahon SB, Eisenman RN (2006) Myc influences global chromatin structure. EMBO J 25:2723–2734

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Vervoorts J, Lüscher-Firzlaff JM, Rottmann S, Lilischkis R, Walsemann G, Dohmann K, Austen M, Lüscher B (2003) Stimulation of c-MYC transcriptional activity and acetylation by recruitment of the cofactor CBP. EMBO Rep 4:484–890

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Filion GJ, van Bemmel JG, Braunschweig U, Talhout W, Kind J, Ward LD, Brugman W, de Castro IJ et al (2010) Systematic protein location mapping reveals five principal chromatin types in Drosophila cells. Cell 143:212–224

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kraemer BC, Burgess JK, Chen JH, Thomas JH, Schellenberg GD (2006) Molecular pathways that influence human tau-induced pathology in Caenorhabditis elegans. Hum Mol Genet 15:1483–1496

    Article  CAS  PubMed  Google Scholar 

  32. Blard O, Feuillette S, Bou J, Chaumette B, Frébourg T, Campion D, Lecourtois M (2007) Cytoskeleton proteins are modulators of mutant tau-induced neurodegeneration in Drosophila. Hum Mol Genet 16:555–566

    Article  CAS  PubMed  Google Scholar 

  33. Citron BA, Dennis JS, Zeitlin RS, Echeverria V (2008) Transcription factor Sp1 dysregulation in Alzheimer's disease. J Neurosci Res 86:2499–2504

    Article  CAS  PubMed  Google Scholar 

  34. Müller M, Cárdenas C, Mei L, Cheung KH, Foskett JK (2011) Constitutive cAMP response element binding protein (CREB) activation by Alzheimer's disease presenilin-driven inositol trisphosphate receptor (InsP3R) Ca2+ signaling. Proc Natl Acad Sci U S A 108:13293–13298

    Article  PubMed  PubMed Central  Google Scholar 

  35. Marambaud P, Wen PH, Dutt A, Shioi J, Takashima A, Siman R, Robakis NK (2003) A CBP binding transcriptional repressor produced by the PS1/epsilon-cleavage of N-cadherin is inhibited by PS1 FAD mutations. Cell 114:635–645

    Article  CAS  PubMed  Google Scholar 

  36. Li Z, Van Calcar S, Qu C, Cavenee WK, Zhang MQ, Ren B (2003) A global transcriptional regulatory role for c-Myc in Burkitt's lymphoma cells. Proc Natl Acad Sci U S A 100:8164–8169

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hann SR (2014) MYC cofactors: molecular switches controlling diverse biological outcomes. Cold Spring Harb Perspect Med 4:a014399

    Article  PubMed  PubMed Central  Google Scholar 

  38. Martinato F, Cesaroni M, Amati B, Guccione E (2008) Analysis of Myc-induced histone modifications on target chromatin. PLoS ONE 3, e3650

    Article  PubMed  PubMed Central  Google Scholar 

  39. Nesbit CE, Tersak JM, Prochownik EV (1999) MYC oncogenes and human neoplastic disease. Oncogene 18:3004–3016

    Article  CAS  PubMed  Google Scholar 

  40. Chen BJ, Wu YL, Tanaka Y, Zhang W (2014) Small molecules targeting c-Myc oncogene: promising anti-cancer therapeutics. Int J Biol Sci 10:1084–1096

    Article  PubMed  PubMed Central  Google Scholar 

  41. Bieszczad KM, Bechay K, Rusche JR, Jacques V, Kudugunti S, Miao W, Weinberger NM, McGaugh JL et al (2015) Histone Deacetylase Inhibition via RGFP966 Releases the Brakes on Sensory Cortical Plasticity and the Specificity of Memory Formation. J Neurosci 35:13124–13132

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kaufman AC, Salazar SV, Haas LT, Yang J, Kostylev MA, Jeng AT, Robinson SA, Gunther EC et al (2015) Fyn inhibition rescues established memory and synapse loss in Alzheimer mice. Ann Neurol 77:953–971

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

We are thankful to Prof. Mel Feany (Harvard Medical School, USA) and the Bloomington Stock Center for providing different fly stocks used in this study. We gratefully thank Prof. Bob Eisenman (Fred Hutchinson Cancer Research Center, USA) for anti-dMyc and Prof. T. Lilja (Stockholm University, USA) for anti-CBP antibodies. This work was supported by research grants from the Department of Biotechnology (DBT), Government of India, New Delhi, India, to S.S. SIC is supported by the Senior Research Fellowship (SRF) from the University Grant Commission (UGC), New Delhi, India. We also thank Delhi University for financial support under R&D scheme, DST-FIST(L2) support to the Department, and Central Instrument Facility (CIF) at South Campus. We are grateful to Ms. Nabanita Sarkar and Ms. Nisha for technical support.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Surajit Sarkar.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Fig. S1

Targeted downregulation of dMyc utilizing second independent RNAi line (UAS-dmyc JF01762) also suppresses human tau mediated rough eye phenotypes and degeneration of retinal tissues (a, b). Induced expression of human c-myc isoform (c-myc2) significantly aggravates tau mediated neurodegenerative phenotype in Drosophila as revealed by bright field and scanning electron microscopy (c, d). Targeted downregulation of dMyc suppresses neurodegenerative phenotypes induced by mutant human tauV337M or tauR406W isoforms (e-n). Compared to GMR-Gal4/+ flies (e, f), eye specific expression of tauV337M (g, h) and tauR406W (k, l) resulted in mild and severe degeneration respectively, which was suppressed upon targeted downregulation of dMyc in tauV337M (i, j) or tauR406W (m, n) expressing cells. In contrast, increased expression of dMyc in tauV337M expressing eye cells resulted in notable aggravation in neurodegenerative phenotype (o, p). (Scale bars: b, d, f, h, j, l, n, p = 100 μm). (GIF 299 kb)

High Resolution Image (TIF 15321 kb)

Fig. S2

Expression of tauWT induces neurodegenerative phenotypes in Drosophila pupal eyes. 55 h old pupal eye disc stained with anti-armadillo (a-c’) and anti-Disc large (d-f’) antibodies and counterstained with DAPI. With normal development and arrangement of primary, secondary, tertiary and cone cells in control pupae (a, a’; d, d’), targeted expression of tauWT deteriorates the gross morphology of the pupal eyes associated with fusion of the surrounding primary, secondary and tertiary cells (b, b’; e, e’) and these morphological impairments were dominantly suppressed upon downregulation of dMyc (c, c’; f, f’). (Scale bars: a-f’ = 10 μm). (GIF 215 kb)

High Resolution Image (TIF 4418 kb)

Fig. S3

Human tauWT mediated neurodegeneration and vacuolization was dominantly suppressed by tissue specific downregulation of dMyc. Paraffin sections of adult head across the midbrain stained with DAPI (a-d) revealed normal internal cellular architecture in GMR-Gal4/+ (a) and degeneration of optic lobes and retina with prominent vacuoles in tauWT expressing neurons (b). Targeted downregulation of dMyc in tauWT expressing photoreceptors suppressed neuronal degeneration in rescue flies (c) or aggravated further upon upregulation of dMyc (d). (Scale bar: a-d = 100 μm). (GIF 191 kb)

High Resolution Image (TIF 3129 kb)

Fig. S4

Graph representing relative level of dMyc expression (±SD) in eye imaginal discs of various genotypes as quantified by calculating total fluorescence intensity (a). Altered expression of dMyc did not make any negative impact on cell cycle as almost equal BrdU incorporation efficiency was evident at the eye morphogenetic furrows (arrow in b-e) of eye discs in controls (b), tauWT expressing (c), tauWT expressing with downregulated dMyc (d) and upregulated dMyc larvae (e). Assessment of the size of adult eye (f) and ommatidium (f’) of GMR-Gal4/+ and GMR-Gal4/+;UAS-dmycRNAi flies (g, g’) did not show any significant difference. (Scale bars: b-e = 100 μm; f’-g’ = 50 μm). (GIF 150 kb)

High Resolution Image (TIF 9289 kb)

Fig. S5

Downregulation of dMyc reduces the abnormal phosphorylation of tau during the early phase of disease pathogenesis. Larval eye imaginal disc stained with anti-human PHF-tauSer202/Thr205 (AT8) antibody showed relatively high level of tau phosphorylation in differentiated neurons of tauWT expressing larval eye disc (a, a’, c) which was reduced following downregulation of dMyc (d, d’, f) and enhanced significantly after dMyc overexpression (g, g’, i). DAPI counterstained discs are shown in (b, e, h). (Scale bars: a-i = 100 μm; a',d',g' = 10 μm). (GIF 181 kb)

High Resolution Image (TIF 5720 kb)

Three dimensional view of a normally developed mushroom body (MB) in adult brain of control Elav-Gal4/+ flies. (MP4 2518 kb)

Three dimensional view of a severely degenerated mushroom body (MB) and neuropil structures in tauWT expressing adult brain. (MP4 2658 kb)

Three dimensional view of a completely restored and normally developed mushroom body (MB) in tauWT expressing adult brain with reduced expression of dMyc. (MP4 2858 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chanu, S.I., Sarkar, S. Targeted Downregulation of dMyc Suppresses Pathogenesis of Human Neuronal Tauopathies in Drosophila by Limiting Heterochromatin Relaxation and Tau Hyperphosphorylation. Mol Neurobiol 54, 2706–2719 (2017). https://doi.org/10.1007/s12035-016-9858-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-016-9858-6

Keywords

Navigation