The Wnt receptor Ryk reduces neuronal and cell survival capacity by repressing FOXO activity during the early phases of mutant huntingtin pathogenicity

Cendrine Tourette, Francesca Farina, Rafael P Vazquez-Manrique, Anne-Marie Orfila, Jessica Voisin, Sonia Hernandez, Nicolas Offner, J Alex Parker, Sophie Menet, Jinho Kim, Jungmok Lyu, Si Ho Choi, Kerry Cormier, Christina K Edgerly, Olivia L Bordiuk, Karen Smith, Anne Louise, Michael Halford, Steven Stacker, Jean-Philippe Vert, Robert J Ferrante, Wange Lu, Christian Neri, Cendrine Tourette, Francesca Farina, Rafael P Vazquez-Manrique, Anne-Marie Orfila, Jessica Voisin, Sonia Hernandez, Nicolas Offner, J Alex Parker, Sophie Menet, Jinho Kim, Jungmok Lyu, Si Ho Choi, Kerry Cormier, Christina K Edgerly, Olivia L Bordiuk, Karen Smith, Anne Louise, Michael Halford, Steven Stacker, Jean-Philippe Vert, Robert J Ferrante, Wange Lu, Christian Neri

Abstract

The Wnt receptor Ryk is an evolutionary-conserved protein important during neuronal differentiation through several mechanisms, including γ-secretase cleavage and nuclear translocation of its intracellular domain (Ryk-ICD). Although the Wnt pathway may be neuroprotective, the role of Ryk in neurodegenerative disease remains unknown. We found that Ryk is up-regulated in neurons expressing mutant huntingtin (HTT) in several models of Huntington's disease (HD). Further investigation in Caenorhabditis elegans and mouse striatal cell models of HD provided a model in which the early-stage increase of Ryk promotes neuronal dysfunction by repressing the neuroprotective activity of the longevity-promoting factor FOXO through a noncanonical mechanism that implicates the Ryk-ICD fragment and its binding to the FOXO co-factor β-catenin. The Ryk-ICD fragment suppressed neuroprotection by lin-18/Ryk loss-of-function in expanded-polyQ nematodes, repressed FOXO transcriptional activity, and abolished β-catenin protection of mutant htt striatal cells against cell death vulnerability. Additionally, Ryk-ICD was increased in the nucleus of mutant htt cells, and reducing γ-secretase PS1 levels compensated for the cytotoxicity of full-length Ryk in these cells. These findings reveal that the Ryk-ICD pathway may impair FOXO protective activity in mutant polyglutamine neurons, suggesting that neurons are unable to efficiently maintain function and resist disease from the earliest phases of the pathogenic process in HD.

Conflict of interest statement

The authors have declared that no competing interests exist. Dr. Robert Ferrante is listed as an author of our paper, but at the time of acceptance was not reachable or able to confirm details of his author contributions to the manuscript. The corresponding author, Christian Neri, has therefore supplied the information regarding his contribution to the manuscript and his competing interests and it is correct to the best of Christian Neri's knowledge.

Figures

Figure 1. Analysis of Ryk in polyQ…
Figure 1. Analysis of Ryk in polyQ nematodes and striatal cells derived from HdhQ111 mice.
(A) Modulation of touch response of polyQ nematodes by LOF of lin-18/RYK. Shown are the results in C. elegans transgenics expressing expanded (128Q) or normal (19Q) exon-1–like htt transgenes in touch receptor neurons. The 128Q-mediated loss of touch response is ameliorated by LOF of lin-18/RYK. No effects are detected in 19Q nematodes. Data are mean ± SEM with more than 200 animals tested per genotype. *p<0.001 versus 128Q controls. LOF of lin-18/RYK does not modify 128Q transgene expression levels as tested by qRT-PCR (right panel, data are mean ± SEM with n = 5). Significance was tested using one-way ANOVA, with correction for multiple testing by Tukey's Multiple Comparison Test. (B) Mutant htt (109Q/109Q) striatal cells have increased Ryk levels. Data for qRT-PCR are mean ± SEM (n = 7). **p<0.01 versus normal htt (7Q/7Q) cells. Data for Western blotting are mean ± SD (n = 3). **p<0.01 versus normal htt (7Q/7Q) cells. Significance was tested using paired t tests. (C) Reducing Ryk levels decreased the mortality of 109Q/109Q striatal cells induced by serum deprivation compared to scramble situation, with no effect detected in 7Q/7Q cells. Western blotting was used to test expression levels as 109Q/109Q cells do not display HTT aggregation. Mutant htt levels were unchanged by knockdown of Ryk. Data are mean ± SD (n = 4). *p<0.01 versus scramble. The RNA sequences shown are indicated in the Materials and Methods section. Significance was tested using paired t tests.
Figure 2. Neuroprotection by lin-18 /Ryk LOF…
Figure 2. Neuroprotection by lin-18/Ryk LOF is a cell-autonomous process that requires daf-16/FoxO activity.
(A) Expression of lin-18 cDNA in touch receptor neurons using the mec-3 promoter abolishes the neuroprotective activity of lin-18 LOF in 128Q nematodes with no effect detected in 19Q nematodes as tested in two independent extrachromosomal arrays (A1, A2) per polyQ genotype. The expression of lin-18 cDNA was confirmed by RT-PCR followed with enzymatic restriction for all of the arrays generated (unpublished data). Data are mean ± SEM (more than 200 animals tested). *p<0.001 compared to 128Q transgenics; **p<0.001 compared to 128Q;lin-18 nematodes. (B) Neuron dysfunction is aggravated by bar-1/β-catenin or daf-16/FoxO LOF in 128Q nematodes, and protection from 128Q toxicity by lin-18 LOF is reduced by LOF of bar-1 and suppressed by LOF of daf-16. Data are mean ± SEM (more than 200 animals tested). **p<0.001 compared to 128Q transgenics; ***p<0.001 compared to 128Q;lin-18 nematodes. (C) lin-18, bar-1 and daf-16 LOF alone or in combination do not change transgenic protein expression levels in 128Q nematodes. Data are mean ± SD (n = 3). Significance was tested using one-way ANOVA, with correction for multiple testing by Tukey's Multiple Comparison Test.
Figure 3. Ryk ICD binds to β-catenin.
Figure 3. Ryk ICD binds to β-catenin.
(A) β-catenin binds to Ryk. Constructs of Myc-tagged Ryk or uncleavable Ryk (Ryk:EGFR Rc) were transfected into 293T cells. Ryk proteins were immunoprecipitated with anti-Myc antibody, and beta-catenin associated with Ryk was determined by immunoblotting. Ryk ICD can be detected in the cells expressing wild-type Ryk. (B) The ICD of Ryk binds to β-catenin. Cells expressing Flag-tagged Ryk-ICD were used for anti-Flag immunoprecipitation. The ICD and associated β-catenin were determined by Western blot. NE, nuclear extract; FUIGW-FLAG, vector FUIGW plus a Flag sequence.
Figure 4. The Ryk ICD represses the…
Figure 4. The Ryk ICD represses the transcriptional activity of FOXO3a, a protein that protects from mutant HTT.
(A) Foxo3a siRNA treatment enhances the mortality of mutant htt striatal cells subjected to serum deprivation, whereas FOXO3a overexpression (O/E) has the opposite effect. Data are mean ± SD (n = 4). *p<0.001 compared to scramble; **p<0.001 compared to empty vector control. (B) Representative Western blots showing decreased (si-Foxo3a) or increased (FOXO3a O/E) FOXO3a levels and no change in HTT protein levels. (C) FOXO transcriptional activity was measured in normal htt mouse striatal cells. Cells were cultured in normal conditions and co-transfected with a construct encoding FOXO3a together with the reporter FHRE-luciferase, which contains three canonical FOXO binding sites, and an internal Renilla luciferase reporter construct. Luciferase and Renilla luciferase activities were measured and the ratio (luciferase/Renilla luciferase)10,000 calculated. Data are mean ± SD of four independent experiments performed in triplicate. Treatment with β-catenin siRNA, full-length Ryk cDNA, and Ryk-ICD cDNA reduces luciferase activity to similar levels, whereas treatment with uncleavable Ryk showed no effect. *p<0.001 compared to FHRE-luc, **p<0.001 compared to scramble RNA and ***p<0.001 compared to FOXO3a O/E. Significance was tested using one-way ANOVA, with correction for multiple testing by Tukey's Multiple Comparison Test. (D) Representative Western blots showing increased levels of FOXO3a and decreased levels of β-catenin, and expression of Myc-tagged Ryk, Myc-tagged Ryk-ICD, and Myc-tagged γ-secretase–uncleavable Ryk (all proteins with a Myc tag at the C-terminal end). The Myc-tagged Ryk and γ-secretase–uncleavable Ryk proteins were detected as two fragments, one corresponding to the full-length Ryk precursor (Ryk) and one corresponding to a Ryk CTF (Ryk CTF) resulting from proteolytic cleavage in the extracellular domain near the transmembrane domain. The full-length Ryk precursor is less abundant for wild-type Ryk expression compared to mutant Ryk expression (see Results for the discussion of Ryk expression profiles).
Figure 5. The Ryk ICD is cytotoxic…
Figure 5. The Ryk ICD is cytotoxic in C. elegans neurons and mouse striatal cells expressing expanded polyQs.
(A) Expression of LIN-18 ICD cDNA (4 ng/µl) in touch receptor neurons using the mec-3 promoter is sufficient to abolish the neuroprotective activity of lin-18 LOF in 128Q nematodes with no effect detected in 19Q nematodes as tested in two independent extrachromosomal arrays (128Q: A1 is ID1333, A2 is ID1334; 19Q: A1 is ID1331, A2 is ID1332; see also Table S8) per polyQ genotype. The expression of LIN-18 ICD cDNA was confirmed by RT-PCR for all of the arrays generated. Data are mean ± SEM (more than 200 animals tested). **p<0.001 compared to 128Q animals, **p<0.001 compared to 128Q;lin-18 animals. ns, not significant. Significance was tested using one-way ANOVA, with correction for multiple testing by Tukey's Multiple Comparison Test. (B) Expression of LIN-18 ICD cDNA (4 ng/µl) in touch receptor neurons using the mec-3 promoter is also sufficient to abolish the protective activity of lin-18 LOF on axonal swelling in the PLM neurons of 128Q nematodes as tested in two independent extrachromosomal arrays (Lin-18 ICD: A1 is ID1333, A2 is ID1334; Lin-18: A1 is ID1325, A2 is ID1326). The expression of LIN-18 ICD cDNA was confirmed by RT-PCR for all of the arrays generated. Expression of empty vector (4 ng/µl) showed no effect as tested in two independent extrachromosomal arrays (A1 is ID1339, A2 is ID1340). Data are mean ± SEM (more than 200 animals tested). *p<0.001 compared to 128Q animals, **p<0.001 compared to 128Q;lin-18 animals. ns, not significant. Significance was tested using one-way ANOVA, with correction for multiple testing by Tukey's Multiple Comparison Test. The lower panel shows a representative image of axonal swelling in the anterior process of posterior touch receptor neurons of 128Q nematodes co-expressing HTT1-57::CFP and YFP . Swelling (white arrows, YFP signals are pseudocolored in green) and HTT::CFP aggregation (yellow arrows, CFP signals are pseudocolored in red) are shown. Magnification is 100× and scale bar is 5 µM. (C) Overexpressing either V5-tagged β-catenin or Myc-tagged Ryk-ICD or both has no effect on the mortality induced by serum deprivation in normal htt striatal cells. Overexpressing β-catenin reduces the mortality induced by serum deprivation in mutant htt striatal cells, whereas overexpressing the Ryk-ICD aggravates cell mortality. Co-expressing Ryk-ICD and β-catenin resulted in cell mortality levels that are similar to those induced by empty vector overexpression. Data are mean ± SD (n = 4). *p<0.01 and **p<0.05 compared to empty vector. ns, not significant. Significance was tested using paired t tests. (D) Representative Western blot showing increased V5-tagged β-catenin and Myc-tagged Ryk-ICD levels after transfection of 7Q/7Q and 109Q/109Q striatal cells.
Figure 6. Reducing presenilin 1 expression counteracts…
Figure 6. Reducing presenilin 1 expression counteracts the cytotoxicity of full-length Ryk overexpression in mutant htt striatal cells.
Assays were performed using caspase 3/7 activity in cells subjected to serum deprivation. (A) PS1 and PS2 siRNA treatment enhances the viability of mutant htt striatal cells. Data are mean ± SD (n = 3), *p<0.01 compared to scramble RNA. Significance was tested using one-way ANOVA, with correction for multiple testing by Tukey's Multiple Comparison Test. (B) Representative Western blots showing decreased levels of PS1/PS2. (C) Knockdown of PS1 reduces the cytotoxic effects of overexpressing full-length Ryk in mutant htt striatal cells, with no effect detected on the cytotoxic effects of overexpressing Ryk-ICD. Data are mean ± SD (n = 4), *p<0.05 and **p<0.01 compared to scramble RNA. Significance was tested using one-way ANOVA, with correction for multiple testing by Tukey's Multiple Comparison Test. EV, empty vector; ns, not significant. (D) Representative Western blots showing decreased levels of PS1 and expression of Myc-tagged Ryk species and Myc-tagged Ryk-ICD. *Nonspecific signal.
Figure 7. The ICD of Ryk is…
Figure 7. The ICD of Ryk is increased in the nucleus of mutant htt striatal cells.
(A) Representative confocal microscopy images showing the pattern of Ryk-ICD immunoreactivity in normal htt (7Q/7Q) and mutant htt (109Q/109Q) striatal cells under normal culture conditions (no serum starvation) as detected using the rabbit polyclonal antibody anti-RykICD. (B) Quantification of Ryk-ICD immunoreactivity in mouse striatal cells. 7Q/7Q and 109Q/109Q cells were grown on the same slides. Comparisons were performed for cells with a nucleus size in the range of 150–250 pixels. Ryk-ICD immunoreactivity was increased in the nucleus of 109Q/109Q cells compared to 7Q/7Q cells. Data are mean ± SEM for the ratio Intensity/Area as detected in either nucleus or cytoplasm (n = 3 for a total of at least 100 cells analyzed), ***p<0.0001 compared to normal htt cells; n.s., not significant. Significance was tested using one-way ANOVA, with correction for multiple testing by Tukey's Multiple Comparison Test. Ryk siRNA treatments were observed to reduce nuclear Ryk-ICD immunoreactivity in 109Q/109Q cells (see Figure S6). (C) Representative confocal microscopy images showing the pattern of Ryk-ICD immunoreactivity in normal htt (7Q/7Q) and mutant htt (109Q/109Q) striatal cells under normal culture conditions (no serum starvation) as detected using the rabbit polyclonal antibody anti-RykICD and mouse Pol2 antibody 7C2. (D) Quantification of Ryk-ICD and Pol2 immunoreactivity in mouse striatal cells. 7Q/7Q and 109Q/109Q cells were grown on the same slides. Comparisons were performed for cells with a nucleus size in the range of 150–250 pixels. The ratio for Ryk-ICD/Pol2 immunoreactivity was increased in the nucleus of 109Q/109Q cells compared to 7Q/7Q cells. Data are mean ± SEM for the ratio Intensity Ryk-ICD/Intensity Pol2 as detected in the nucleus (n = 4 for a total of at least 100 cells analyzed), **p<0.002 compared to normal htt cells. Significance was tested using Welch's t test.

References

    1. Kikis EA, Gidalevitz T, Morimoto RI (2010) Protein homeostasis in models of aging and age-related conformational disease. Adv Exp Med Biol 694: 138–159.
    1. Eijkelenboom A, Burgering BM (2013) FOXOs: signalling integrators for homeostasis maintenance. Nat Rev Mol Cell Biol 14: 83–97.
    1. Salih DA, Brunet A (2008) FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr Opin Cell Biol 20: 126–136.
    1. Neri C (2012) Role and therapeutic potential of the pro-longevity factor FOXO and its regulators in neurodegenerative disease. Front Pharmacol 3: 15.
    1. Parmentier F, Lejeune FX, Neri C (2013) Pathways to decoding the clinical potential of stress response FOXO-interaction networks for Huntington's disease: of gene prioritization and context dependence. Front Aging Neurosci 5: 22.
    1. Gil JM, Rego AC (2008) Mechanisms of neurodegeneration in Huntington's disease. Eur J Neurosci 27: 2803–2820.
    1. Parker JA, Arango M, Abderrahmane S, Lambert E, Tourette C, et al. (2005) Resveratrol rescues mutant polyglutamine cytotoxicity in nematode and mammalian neurons. Nat Genet 37: 349–350.
    1. Cohen E, Bieschke J, Perciavalle RM, Kelly JW, Dillin A (2006) Opposing activities protect against age-onset proteotoxicity. Science 313: 1604–1610.
    1. Essers MA, de Vries-Smits LM, Barker N, Polderman PE, Burgering BM, et al. (2005) Functional interaction between beta-catenin and FOXO in oxidative stress signaling. Science 308: 1181–1184.
    1. Parker JA, Vazquez-Manrique RP, Tourette C, Farina F, Offner N, et al. (2012) Integration of beta-catenin, sirtuin, and FOXO signaling protects from mutant huntingtin toxicity. J Neurosci 32: 12630–12640.
    1. Inestrosa NC, Arenas E (2010) Emerging roles of Wnts in the adult nervous system. Nat Rev Neurosci 11: 77–86.
    1. Caricasole A, Bakker A, Copani A, Nicoletti F, Gaviraghi G, et al. (2005) Two sides of the same coin: Wnt signaling in neurodegeneration and neuro-oncology. Biosci Rep 25: 309–327.
    1. Carmichael J, Sugars KL, Bao Y, Rubinsztein DC (2002) GSK-3beta inhibitors prevent cellular polyglutamine toxicity caused by the Huntington's disease mutation. J Biol Chem 3: 3.
    1. Gines S, Ivanova E, Seong IS, Saura CA, MacDonald ME (2003) Enhanced Akt signaling is an early pro-survival response that reflects N-methyl-D-aspartate receptor activation in Huntington's disease knock-in striatal cells. J Biol Chem 278: 50514–50522.
    1. Parker JA, Connolly JB, Wellington C, Hayden M, Dausset J, et al. (2001) Expanded polyglutamines in Caenorhabditis elegans cause axonal abnormalities and severe dysfunction of PLM mechanosensory neurons without cell death. Proc Natl Acad Sci U S A 98: 13318–13323.
    1. Lyu J, Yamamoto V, Lu W (2008) Cleavage of the Wnt receptor Ryk regulates neuronal differentiation during cortical neurogenesis. Dev Cell 15: 773–780.
    1. Zhong J, Kim HT, Lyu J, Yoshikawa K, Nakafuku M, et al. (2011) The Wnt receptor Ryk controls specification of GABAergic neurons versus oligodendrocytes during telencephalon development. Development 138: 409–419.
    1. Keeble TR, Halford MM, Seaman C, Kee N, Macheda M, et al. (2006) The Wnt receptor Ryk is required for Wnt5a-mediated axon guidance on the contralateral side of the corpus callosum. J Neurosci 26: 5840–5848.
    1. Macheda ML, Sun WW, Kugathasan K, Hogan BM, Bower NI, et al. (2012) The Wnt receptor Ryk plays a role in mammalian planar cell polarity signaling. J Biol Chem 287: 29312–29323.
    1. Andre P, Wang Q, Wang N, Gao B, Schilit A, et al. (2012) The Wnt coreceptor Ryk regulates Wnt/planar cell polarity by modulating the degradation of the core planar cell polarity component Vangl2. J Biol Chem 287: 44518–44525.
    1. Hollis ER 2nd, Zou Y (2012) Reinduced Wnt signaling limits regenerative potential of sensory axons in the spinal cord following conditioning lesion. Proc Natl Acad Sci U S A 109: 14663–14668.
    1. Povinelli BJ, Nemeth MJ (2013) Wnt5a regulates hematopoietic stem cell proliferation and repopulation through the Ryk receptor. Stem Cells 32: 105–115.
    1. Trettel F, Rigamonti D, Hilditch-Maguire P, Wheeler VC, Sharp AH, et al. (2000) Dominant phenotypes produced by the HD mutation in STHdh(Q111) striatal cells. Hum Mol Genet 9: 2799–2809.
    1. Hickey MA, Kosmalska A, Enayati J, Cohen R, Zeitlin S, et al. (2008) Extensive early motor and non-motor behavioral deficits are followed by striatal neuronal loss in knock-in Huntington's disease mice. Neuroscience 157: 280–295.
    1. Lejeune FX, Mesrob L, Parmentier F, Bicep C, Vazquez-Manrique RP, et al. (2012) Large-scale functional RNAi screen in C. elegans identifies genes that regulate the dysfunction of mutant polyglutamine neurons. BMC Genomics 13: 91.
    1. Lu W, Yamamoto V, Ortega B, Baltimore D (2004) Mammalian Ryk is a Wnt coreceptor required for stimulation of neurite outgrowth. Cell 119: 97–108.
    1. Colavita A, Krishna S, Zheng H, Padgett RW, Culotti JG (1998) Pioneer axon guidance by UNC-129, a C. elegans TGF-beta. Science 281: 706–709.
    1. Russ AP, Lampel S (2005) The druggable genome: an update. Drug Discov Today 10: 1607–1610.
    1. Hodges A, Strand AD, Aragaki AK, Kuhn A, Sengstag T, et al. (2006) Regional and cellular gene expression changes in human Huntington's disease brain. Hum Mol Genet 15: 965–977.
    1. Arango M, Holbert S, Zala D, Brouillet E, Pearson J, et al. (2006) CA150 expression delays striatal cell death in overexpression and knock-in conditions for mutant huntingtin neurotoxicity. J Neurosci 26: 4649–4659.
    1. Berndt JD, Aoyagi A, Yang P, Anastas JN, Tang L, et al. (2011) Mindbomb 1, an E3 ubiquitin ligase, forms a complex with RYK to activate Wnt/beta-catenin signaling. J Cell Biol 194: 737–750.
    1. Mojsilovic-Petrovic J, Nedelsky N, Boccitto M, Mano I, Georgiades SN, et al. (2009) FOXO3a is broadly neuroprotective in vitro and in vivo against insults implicated in motor neuron diseases. J Neurosci 29: 8236–8247.
    1. Halford MM, Macheda ML, Parish CL, Takano EA, Fox S, et al. (2013) A fully human inhibitory monoclonal antibody to the Wnt receptor RYK. PLoS ONE 8: e75447 doi:
    1. Cupers P, Orlans I, Craessaerts K, Annaert W, De Strooper B (2001) The amyloid precursor protein (APP)-cytoplasmic fragment generated by gamma-secretase is rapidly degraded but distributes partially in a nuclear fraction of neurones in culture. J Neurochem 78: 1168–1178.
    1. Sastre M, Steiner H, Fuchs K, Capell A, Multhaup G, et al. (2001) Presenilin-dependent gamma-secretase processing of beta-amyloid precursor protein at a site corresponding to the S3 cleavage of Notch. EMBO Rep 2: 835–841.
    1. Kegel KB, Sapp E, Alexander J, Reeves P, Bleckmann D, et al. (2010) Huntingtin cleavage product A forms in neurons and is reduced by gamma-secretase inhibitors. Mol Neurodegener 5: 58.
    1. Hersch SM, Rosas HR, Ferrante RJ (2004) Neuropathology and pathophysiology of Huntington's disease. Watts, RL and Koller, WC (eds), Movement disorders: neurologic principles and practice. New York: McGraw-Hill: 503–523.
    1. Landis JN, Murphy CT (2010) Integration of diverse inputs in the regulation of Caenorhabditis elegans DAF-16/FOXO. Dev Dyn 239: 1405–1412.
    1. Renault VM, Rafalski VA, Morgan AA, Salih DA, Brett JO, et al. (2009) FoxO3 regulates neural stem cell homeostasis. Cell Stem Cell 5: 527–539.
    1. Liu Y, Shi J, Lu CC, Wang ZB, Lyuksyutova AI, et al. (2005) Ryk-mediated Wnt repulsion regulates posterior-directed growth of corticospinal tract. Nat Neurosci 8: 1151–1159.
    1. Bovolenta P, Rodriguez J, Esteve P (2006) Frizzled/RYK mediated signalling in axon guidance. Development 133: 4399–4408.
    1. Zhang Z, Hartmann H, Do VM, Abramowski D, Sturchler-Pierrat C, et al. (1998) Destabilization of beta-catenin by mutations in presenilin-1 potentiates neuronal apoptosis. Nature 395: 698–702.
    1. Francis R, McGrath G, Zhang J, Ruddy DA, Sym M, et al. (2002) aph-1 and pen-2 are required for Notch pathway signaling, gamma-secretase cleavage of betaAPP, and presenilin protein accumulation. Dev Cell 3: 85–97.
    1. Webb AE, Pollina EA, Vierbuchen T, Urban N, Ucar D, et al. (2013) FOXO3 shares common targets with ASCL1 genome-wide and inhibits ASCL1-dependent neurogenesis. Cell Rep 4: 477–491.
    1. Li X, Li YH, Yu S, Liu Y (2008) Upregulation of Ryk expression in rat dorsal root ganglia after peripheral nerve injury. Brain Res Bull 77: 178–184.
    1. Liu Y, Wang X, Lu CC, Kerman R, Steward O, et al. (2008) Repulsive Wnt signaling inhibits axon regeneration after CNS injury. J Neurosci 28: 8376–8382.
    1. Brenner S (1974) The genetics of Caenorhabditis elegans. Genetics 77: 71–94.
    1. Parker JA, Metzler M, Georgiou J, Mage M, Roder JC, et al. (2007) Huntingtin-interacting protein 1 influences worm and mouse presynaptic function and protects Caenorhabditis elegans neurons against mutant polyglutamine toxicity. J Neurosci 27: 11056–11064.
    1. Vazquez-Manrique RP, Nagy AI, Legg JC, Bales OA, Ly S, et al. (2008) Phospholipase C-epsilon regulates epidermal morphogenesis in Caenorhabditis elegans. PLoS Genet 4: e1000043.
    1. Duerr JS (2006) Immunohistochemistry. WormBook: 1–61.
    1. Zhang Y, Ma C, Delohery T, Nasipak B, Foat BC, et al. (2002) Identification of genes expressed in C. elegans touch receptor neurons. Nature 418: 331–335.
    1. Gauthier LR, Charrin BC, Borrell-Pages M, Dompierre JP, Rangone H, et al. (2004) Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell 118: 127–138.
    1. Miller JP, Holcomb J, Al-Ramahi I, de Haro M, Gafni J, et al. (2010) Matrix metalloproteinases are modifiers of huntingtin proteolysis and toxicity in Huntington's disease. Neuron 67: 199–212.
    1. Lyu J, Wesselschmidt RL, Lu W (2009) Cdc37 regulates Ryk signaling by stabilizing the cleaved Ryk intracellular domain. J Biol Chem 284(19): 12940–12948.
    1. Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, et al. (1999) Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96: 857–868.
    1. Menalled LB, Sison JD, Dragatsis I, Zeitlin S, Chesselet MF (2003) Time course of early and neuropathological anomalies in a knock-in mouse model of Huntington's disease with 140 CAG repeats. J Comp Neurol 465: 11–26.
    1. Vonsattel JP, Myers RH, Stevens TJ, Ferrante RJ, Bird ED, Richardson EP Jr (1985) Neuropathological classification of Huntington's disease. J Neuropathol Exp Neurol 44: 559–577.
    1. Remm M, Storm CE, Sonnhammer EL (2001) Automatic clustering of orthologs and in-paralogs from pairwise species comparisons. J Mol Biol 314: 1041–1052.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, et al. (2005) Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102: 15545–15550.
    1. Murphy CT, McCarroll SA, Bargmann CI, Fraser A, Kamath RS, et al. (2003) Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditis elegans. Nature 424: 277–283.
    1. McElwee J, Bubb K, Thomas JH (2003) Transcriptional outputs of the Caenorhabditis elegans forkhead protein DAF-16. Aging Cell 2: 111–121.
    1. Oh SW, Mukhopadhyay A, Dixit BL, Raha T, Green MR, et al. (2006) Identification of direct DAF-16 targets controlling longevity, metabolism and diapause by chromatin immunoprecipitation. Nat Genet 38: 251–257.
    1. Kuhn A, Goldstein DR, Hodges A, Strand AD, Sengstag T, et al. (2007) Mutant huntingtin's effects on striatal gene expression in mice recapitulate changes observed in human Huntington's disease brain and do not differ with mutant huntingtin length or wild-type huntingtin dosage. Hum Mol Genet 16: 1845–1861.
    1. Thomas EA, Coppola G, Tang B, Kuhn A, Kim S, et al. (2011) In vivo cell-autonomous transcriptional abnormalities revealed in mice expressing mutant huntingtin in striatal but not cortical neurons. Hum Mol Genet 20: 1049–1060.
    1. Borovecki F, Lovrecic L, Zhou J, Jeong H, Then F, et al. (2005) Genome-wide expression profiling of human blood reveals biomarkers for Huntington's disease. Proc Natl Acad Sci U S A 102: 11023–11028.
    1. The HD iPSC Consortium (2012) Induced pluripotent stem cells from patients with Huntington's disease show CAG-repeat-expansion-associated phenotypes. Cell Stem Cell 11: 264–278.

Source: PubMed

3
订阅