AMPK activation protects from neuronal dysfunction and vulnerability across nematode, cellular and mouse models of Huntington's disease

Rafael P Vázquez-Manrique, Francesca Farina, Karine Cambon, María Dolores Sequedo, Alex J Parker, José María Millán, Andreas Weiss, Nicole Déglon, Christian Neri, Rafael P Vázquez-Manrique, Francesca Farina, Karine Cambon, María Dolores Sequedo, Alex J Parker, José María Millán, Andreas Weiss, Nicole Déglon, Christian Neri

Abstract

The adenosine monophosphate activated kinase protein (AMPK) is an evolutionary-conserved protein important for cell survival and organismal longevity through the modulation of energy homeostasis. Several studies suggested that AMPK activation may improve energy metabolism and protein clearance in the brains of patients with vascular injury or neurodegenerative disease. However, in Huntington's disease (HD), AMPK may be activated in the striatum of HD mice at a late, post-symptomatic phase of the disease, and high-dose regiments of the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide may worsen neuropathological and behavioural phenotypes. Here, we revisited the role of AMPK in HD using models that recapitulate the early features of the disease, including Caenorhabditis elegans neuron dysfunction before cell death and mouse striatal cell vulnerability. Genetic and pharmacological manipulation of aak-2/AMPKα shows that AMPK activation protects C. elegans neurons from the dysfunction induced by human exon-1 huntingtin (Htt) expression, in a daf-16/forkhead box O-dependent manner. Similarly, AMPK activation using genetic manipulation and low-dose metformin treatment protects mouse striatal cells expressing full-length mutant Htt (mHtt), counteracting their vulnerability to stress, with reduction of soluble mHtt levels by metformin and compensation of cytotoxicity by AMPKα1. Furthermore, AMPK protection is active in the mouse brain as delivery of gain-of-function AMPK-γ1 to mouse striata slows down the neurodegenerative effects of mHtt. Collectively, these data highlight the importance of considering the dynamic of HD for assessing the therapeutic potential of stress-response targets in the disease. We postulate that AMPK activation is a compensatory response and valid approach for protecting dysfunctional and vulnerable neurons in HD.

© The Author 2015. Published by Oxford University Press.

Figures

Figure 1.
Figure 1.
aak-2/AMPKα is neuroprotective in 128Q worms. (A) Ablation of the aak-2 gene results in enhancement of the touch phenotype in 128Q worms. (B) Metformin alleviates the touch phenotype of 128Q animals, without affecting the behaviour of 19Q worms. (C) Metformin rescue of the worms depends mostly on the presence of the aak-2 gene. In all panels, values are mean ± SEM (N = 3 with a total of at least 100 animals tested per condition). ANOVA tests, with Tukey post hoc analysis. Ns: not significant. ***P < 0.001.
Figure 2.
Figure 2.
aak-2 requires daf-16 for neuroprotection in a tissue-specific-dependent manner. The deletion of both daf-16 and aak-2 induce a similar enhancement of the touch phenotype. Combining both mutations do not show a synergistic effect, neither an additive effect, suggesting that both genes induce neuroprotecttion though the same pathway. Rescuing aak-2 and daf-16 in mechanosensory neurons rescues the touch phenotype at different levels. Rescue of the touch phenotype by aak-2 overexpression requires daf-16, further suggesting that both work in the same signalling pathway. Values are mean ± SEM (N = 3 with a total of at least 100 animals tested per condition). ANOVA tests, with Tukey post hoc analysis were used. ns: not significant. **P < 0.01 and ***P < 0.001.
Figure 3.
Figure 3.
Nucleo-cytoplasmic expression of AMPKα1 and AMPKα2 in 7Q/7Q and 109Q/109Q mouse striatal cells. (A) RT-PCR analysis of the expression of AMPKα subunits shows that AMPKα1 mRNA levels are higher in 109Q/109Q cells versus 7Q/7Q cells and that AMPKα2 mRNA levels are lower in 109Q/109Q cells versus 7Q/7Q cells. Values are mean ± SD (N = 3 in triplicate). ***P < 0.001. (B) Representative images of antibody staining of both subunits showing that AMPKα1 and AMPKα2 have variable nucleo-cytoplasmic distributions across 7Q/7Q and 109Q/109Q cells. Scale bar is 20 µm. (C) Quantification of nuclear signal intensities for AMPKα1 and AMPKα2 showing that there is more AMPKα1 in 109Q/109Q cells compared with 7Q/7Q cells, which is also observed for AMPKα2. Values are mean ± SEM (N = 3 for a total of at least 90 cells tested per condition). ***P < 0.001. (D) Quantification of nucleo-cytoplasmic distribution for AMPKα1 showing that there is more AMPKα1 in the nucleus of 7Q/7Q cells, which is more pronounced in 109Q/109Q cells. Values are mean ± SEM (N = 3 for a total of at least 90 cells). ***P < 0.001. Statistics were performed using t tests in all panels.
Figure 4.
Figure 4.
Nucleo-cytoplasmic expression of AMPKα-phosphorylated in 7Q/7Q and 109Q/109Q mouse striatal cells. (A) Representative confocal images illustrating that AMPKα-phosphorylated is evenly distributed in the nucleus and cytoplasm in 7Q/7Q and 109Q/109Q cells. Scale bar is 20 µm. (B) The upper two panels show an example of the AMPKα-phosphorylated signal along a line (yellow) that crosses the cytoplasm and nucleus. The blue curve shows DAPI staining, pointing out the region covered by the nuclei, and the green line shows fluorescent due to AMPKα-phosphorylated staining. Colour boxes indicate the area used for the quantification of AMPKα-phosphorylated signals on either side of the nuclear membrane. The lower panel shows the levels of AMPKα-phosphorylated signals, with increase of the signal in both the cytoplasmic and nuclear compartment induced by metformin treatment of 7Q/7Q and 109Q/109Q cells. Values are mean ± SD (N = 3 for a total of at least 50 cells/condition). ANOVA tests, with Tukey post hoc analysis for each genotype, were used. *P < 0.05, **P < 0.01 and ***P < 0.001.
Figure 5.
Figure 5.
AMPK has a protective function in and in vitro model of HD. (A) Growing striatal cells in metformin 2 mm reduces mortality induced by serum deprivation in 109Q/109Q mouse striatal cells (***P < 0.001, N = 3). The right panel shows representative western blots to show that AMPKγ1GOF induces phosphorylation of AMPKα in striatal cells. The levels of Htt do not change. (B) Silencing AMPKα1 by RNAi in 109Q/109Q mouse striatal cells enhances cell mortality in response to serum deprivation with no effect detected in 7Q/7Q cells (***P < 0.001; ns: not significant; N = 3). The right panel shows representative western blots in which AMPKα1 siRNAs reduce AMPKα1 protein levels (N = 3, mean ± SD = 0.4 ± 0.1, P = 0.018) with no change detected in the level of Htt (N = 3, mean ± SD = 1.1 ± 0.5, P = 0.71). (C) Overexpression of an over-activated form of AMPKγ1, AMPKγ1GOF, reduces mortality of 109Q/109Q cells in response to serum deprivation (**P < 0.005, N = 3). The right panel shows representative western blots in which AMPKγ1GOF expression increases AMPKγ1 protein levels (as detected using a c-Myc tag), phosphorylated AMPKα1 levels (N = 3, mean ± SD = 1.6 ± 0.1, P = 0.027). The effect on Htt levels is addressed in (D). (D) Overexpression of the over-activated form of AMPKγ1 induces clearance of misfolded mHtt (***P < 0.001, N = 3). The right panel shows representative western blots in which AMPKγ1GOF increases AMPKγ1 levels (as detected using a c-Myc tag), and phosphorylated AMPKα levels (N = 4, mean ± SD = 1.7 ± 0.4, P = 0.021). In all panels: ns: not significant.
Figure 6.
Figure 6.
In vivo activation of AMPK rescues striatal cell degeneration in a mouse model of HD. (A) Diagram showing the treatment of both sides of the brains of mice with lentiviruses expressing the N-terminal fragment of mHtt [Htt171-82Q (Htt in the diagram)], or the AMPKγ1GOF. Below is shown a representative picture of DARPP32 staining of the sections of the brains of these mice, which highlights the area of the lesion caused by mutant Htt. The right panel shows the analysis of the volume of the lesion caused by mutant Htt, with overexpression of AMPKγ1GOF reducing cell death. Values are mean ± SEM. *P < 0.05 (P = 0.03, N = 13). (B) Representative pictures showing UBI staining and pointing out to inclusion bodies. The right panel shows the quantification of the number of UBI-positive aggregates. Values are mean ± SEM. The overexpression of AMPKγ1GOF does not change the number of inclusion bodies (P = 0.87, N = 13). Statistics use the Wilcoxon test for paired samples. ns: not significant.

References

    1. Carling D. (2004) The AMP-activated protein kinase cascade—a unifying system for energy control. Trends. Biochem. Sci., 29, 18–24.
    1. Carling D., Thornton C., Woods A., Sanders M.J. (2012) AMP-activated protein kinase: new regulation, new roles? Biochem. J., 445, 11–27.
    1. Viana R., Aguado C., Esteban I., Moreno D., Viollet B., Knecht E., Sanz P. (2008) Role of AMP-activated protein kinase in autophagy and proteasome function. Biochem. Biophys. Res. Commun., 369, 964–968.
    1. Alers S., Loffler A.S., Wesselborg S., Stork B. (2012) Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks. Mol. Cell Biol., 32, 2–11.
    1. Chiacchiera F., Simone C. (2010) The AMPK-FoxO3A axis as a target for cancer treatment. Cell Cycle, 9, 1091–1096.
    1. Mihaylova M.M., Shaw R.J. (2011) The AMPK signalling pathway coordinates cell growth, autophagy and metabolism. Nat. Cell. Biol., 13, 1016–1023.
    1. Shaw R.J., Lamia K.A., Vasquez D., Koo S.H., Bardeesy N., Depinho R.A., Montminy M., Cantley L.C. (2005) The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science, 310, 1642–1646.
    1. Canto C., Auwerx J. (2009) PGC-1alpha, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr. Opin. Lipidol., 20, 98–105.
    1. Egan D.F., Shackelford D.B., Mihaylova M.M., Gelino S., Kohnz R.A., Mair W., Vasquez D.S., Joshi A., Gwinn D.M., Taylor R. et al. (2011) Phosphorylation of ULK1 (hATG1) by AMP-activated protein kinase connects energy sensing to mitophagy. Science, 331, 456–461.
    1. Zhou G., Myers R., Li Y., Chen Y., Shen X., Fenyk-Melody J., Wu M., Ventre J., Doebber T., Fujii N. et al. (2001) Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Invest., 108, 1167–1174.
    1. He H., Ke R., Lin H., Ying Y., Liu D., Luo Z. (2015) Metformin, an old drug, brings a new era to cancer therapy. Cancer J., 21, 70–74.
    1. Nasri H., Rafieian-Kopaei M. (2014) Metformin and diabetic kidney disease: a mini-review on recent findings. Iran J. Pediatr., 24, 565–568.
    1. Apfeld J., O'Connor G., McDonagh T., DiStefano P.S., Curtis R. (2004) The AMP-activated protein kinase AAK-2 links energy levels and insulin-like signals to lifespan in C. elegans. Genes Dev., 18, 3004–3009.
    1. Mair W., Morantte I., Rodrigues A.P., Manning G., Montminy M., Shaw R.J., Dillin A. (2011) Lifespan extension induced by AMPK and calcineurin is mediated by CRTC-1 and CREB. Nature, 470, 404–408.
    1. Salminen A., Kaarniranta K., Haapasalo A., Soininen H., Hiltunen M. (2011) AMP-activated protein kinase: a potential player in Alzheimer's disease. J. Neurochem., 118, 460–474.
    1. Brown K.A., Samarajeewa N.U., Simpson E.R. (2013) Endocrine-related cancers and the role of AMPK. Mol. Cell. Endocrinol., 366, 170–179.
    1. Steinberg G.R., Kemp B.E. (2009) AMPK in health and disease. Physiol. Rev., 89, 1025–1078.
    1. Zaha V.G., Young L.H. (2012) AMP-activated protein kinase regulation and biological actions in the heart. Circ. Res., 111, 800–814.
    1. Sarkaki A., Farbood Y., Badavi M., Khalaj L., Khodagholi F., Ashabi G. (2015) Metformin improves anxiety-like behaviors through AMPK-dependent regulation of autophagy following transient forebrain ischemia. Metab. Brain Dis., 30, 1139–1150.
    1. Ashabi G., Khalaj L., Khodagholi F., Goudarzvand M., Sarkaki A. (2015) Pre-treatment with metformin activates Nrf2 antioxidant pathways and inhibits inflammatory responses through induction of AMPK after transient global cerebral ischemia. Metab. Brain Dis., 30, 747–754.
    1. Jiang T., Yu J.T., Zhu X.C., Zhang Q.Q., Tan M.S., Cao L., Wang H.F., Shi J.Q., Gao L., Qin H. et al. (2015) Ischemic preconditioning provides neuroprotection by induction of AMP-activated protein kinase-dependent autophagy in a rat model of ischemic stroke. Mol. Neurobiol., 51, 220–229.
    1. Du L.L., Chai D.M., Zhao L.N., Li X.H., Zhang F.C., Zhang H.B., Liu L.B., Wu K., Liu R., Wang J.Z. et al. (2015) AMPK activation ameliorates Alzheimer's disease-like pathology and spatial memory impairment in a streptozotocin-induced Alzheimer's disease model in rats. J. Alzheimers Dis., 43, 775–784.
    1. Lu J., Wu D.M., Zheng Y.L., Hu B., Zhang Z.F., Shan Q., Zheng Z.H., Liu C.M., Wang Y.J. (2010) Quercetin activates AMP-activated protein kinase by reducing PP2C expression protecting old mouse brain against high cholesterol-induced neurotoxicity. J. Pathol., 222, 199–212.
    1. Ma T., Chen Y., Vingtdeux V., Zhao H., Viollet B., Marambaud P., Klann E. (2014) Inhibition of AMP-activated protein kinase signaling alleviates impairments in hippocampal synaptic plasticity induced by amyloid beta. J. Neurosci., 34, 12230–12238.
    1. DiTacchio K.A., Heinemann S.F., Dziewczapolski G. (2015) Metformin treatment alters memory function in a mouse model of Alzheimer's disease. J. Alzheimers Dis., 44, 43–48.
    1. Ma T.C., Buescher J.L., Oatis B., Funk J.A., Nash A.J., Carrier R.L., Hoyt K.R. (2007) Metformin therapy in a transgenic mouse model of Huntington's disease. Neurosci. Lett., 411, 98–103.
    1. Ju T.C., Chen H.M., Lin J.T., Chang C.P., Chang W.C., Kang J.J., Sun C.P., Tao M.H., Tu P.H., Chang C. et al. (2011) Nuclear translocation of AMPK-alpha1 potentiates striatal neurodegeneration in Huntington's disease. J. Cell Biol., 194, 209–227.
    1. Ju T.C., Chen H.M., Chen Y.C., Chang C.P., Chang C., Chern Y. (2014) AMPK-alpha1 functions downstream of oxidative stress to mediate neuronal atrophy in Huntington's disease. Biochim. Biophys. Acta., 1842, 1668–1680.
    1. Tourette C., Farina F., Vazquez-Manrique R.P., Orfila A.M., Voisin J., Hernandez S., Offner N., Parker J.A., Menet S., Kim J. et al. (2014) The Wnt receptor Ryk reduces neuronal and cell survival capacity by repressing FOXO activity during the early phases of mutant huntingtin pathogenicity. PLoS Biol., 12, e1001895.
    1. Trettel F., Rigamonti D., Hilditch-Maguire P., Wheeler V.C., Sharp A.H., Persichetti F., Cattaneo E., MacDonald M.E. (2000) Dominant phenotypes produced by the HD mutation in STHdh(Q111) striatal cells. Hum. Mol. Genet., 9, 2799–2809.
    1. Curtis R., O'Connor G., DiStefano P.S. (2006) Aging networks in Caenorhabditis elegans: AMP-activated protein kinase (aak-2) links multiple aging and metabolism pathways. Aging Cell, 5, 119–126.
    1. Lee H., Cho J.S., Lambacher N., Lee J., Lee S.J., Lee T.H., Gartner A., Koo H.S. (2008) The Caenorhabditis elegans AMP-activated protein kinase AAK-2 is phosphorylated by LKB1 and is required for resistance to oxidative stress and for normal motility and foraging behavior. J. Biol. Chem., 283, 14988–14993.
    1. Weimer S., Priebs J., Kuhlow D., Groth M., Priebe S., Mansfeld J., Merry T.L., Dubuis S., Laube B., Pfeiffer A.F. et al. (2014) D-Glucosamine supplementation extends life span of nematodes and of ageing mice. Nat. Commun., 5, 3563.
    1. Lejeune F.X., Mesrob L., Parmentier F., Bicep C., Vazquez-Manrique R.P., Parker J.A., Vert J.P., Tourette C., Neri C. (2012) Large-scale functional RNAi screen in C. elegans identifies genes that regulate the dysfunction of mutant polyglutamine neurons. BMC Genomics, 13, 91.
    1. Owen M.R., Doran E., Halestrap A.P. (2000) Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem. J., 348(Pt 3), 607–614.
    1. Parker J.A., Arango M., Abderrahmane S., Lambert E., Tourette C., Catoire H., Neri C. (2005) Resveratrol rescues mutant polyglutamine cytotoxicity in nematode and mammalian neurons. Nat. Genet., 37, 349–350.
    1. Parker J.A., Vazquez-Manrique R.P., Tourette C., Farina F., Offner N., Mukhopadhyay A., Orfila A.M., Darbois A., Menet S., Tissenbaum H.A. et al. (2012) Integration of beta-catenin, sirtuin, and FOXO signaling protects from mutant huntingtin toxicity. J. Neurosci., 32, 12630–12640.
    1. Greer E.L., Oskoui P.R., Banko M.R., Maniar J.M., Gygi M.P., Gygi S.P., Brunet A. (2007) The energy sensor AMP-activated protein kinase directly regulates the mammalian FOXO3 transcription factor. J. Biol. Chem., 282, 30107–30119.
    1. Jeong H., Cohen D.E., Cui L., Supinski A., Savas J.N., Mazzulli J.R., Yates J.R. III, Bordone L., Guarente L., Krainc D. (2012) Sirt1 mediates neuroprotection from mutant huntingtin by activation of the TORC1 and CREB transcriptional pathway. Nat. Med., 18, 159–165.
    1. Jiang M., Wang J., Fu J., Du L., Jeong H., West T., Xiang L., Peng Q., Hou Z., Cai H. et al. (2012) Neuroprotective role of Sirt1 in mammalian models of Huntington's disease through activation of multiple Sirt1 targets. Nat. Med., 18, 153–158.
    1. Park S.J., Ahmad F., Philp A., Baar K., Williams T., Luo H., Ke H., Rehmann H., Taussig R., Brown A.L. et al. (2012) Resveratrol ameliorates aging-related metabolic phenotypes by inhibiting cAMP phosphodiesterases. Cell, 148, 421–433.
    1. Burnett C., Valentini S., Cabreiro F., Goss M., Somogyvari M., Piper M.D., Hoddinott M., Sutphin G.L., Leko V., McElwee J.J. et al. (2011) Absence of effects of Sir2 overexpression on lifespan in C. elegans and Drosophila. Nature, 477, 482–485.
    1. Arango M., Holbert S., Zala D., Brouillet E., Pearson J., Regulier E., Thakur A.K., Aebischer P., Wetzel R., Deglon N. 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. Salt I., Celler J.W., Hawley S.A., Prescott A., Woods A., Carling D., Hardie D.G. (1998) AMP-activated protein kinase: greater AMP dependence, and preferential nuclear localization, of complexes containing the alpha2 isoform. Biochem. J., 334(Pt 1), 177–187.
    1. Inoki K., Li Y., Xu T., Guan K.L. (2003) Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev., 17, 1829–1834.
    1. Sanchez A.M., Csibi A., Raibon A., Cornille K., Gay S., Bernardi H., Candau R. (2012) AMPK promotes skeletal muscle autophagy through activation of forkhead FoxO3a and interaction with Ulk1. J. Cell Biochem., 113, 695–710.
    1. Baldo B., Paganetti P., Grueninger S., Marcellin D., Kaltenbach L.S., Lo D.C., Semmelroth M., Zivanovic A., Abramowski D., Smith D. et al. (2012) TR-FRET-based duplex immunoassay reveals an inverse correlation of soluble and aggregated mutant huntingtin in Huntington's disease. Chem. Biol., 19, 264–275.
    1. Tabrizi S.J., Scahill R.I., Durr A., Roos R.A., Leavitt B.R., Jones R., Landwehrmeyer G.B., Fox N.C., Johnson H., Hicks S.L. et al. (2011) Biological and clinical changes in premanifest and early stage Huntington's disease in the TRACK-HD study: the 12-month longitudinal analysis. Lancet Neurol., 10, 31–42.
    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. Kim M., Lee H.S., LaForet G., McIntyre C., Martin E.J., Chang P., Kim T.W., Williams M., Reddy P.H., Tagle D. et al. (1999) Mutant huntingtin expression in clonal striatal cells: dissociation of inclusion formation and neuronal survival by caspase inhibition. J. Neurosci., 19, 964–973.
    1. Saudou F., Finkbeiner S., Devys D., Greenberg M.E. (1998) Huntingtin acts in the nucleus to induce apoptosis but death does not correlate with the formation of intranuclear inclusions. Cell, 95, 55–66.
    1. Zala D., Benchoua A., Brouillet E., Perrin V., Gaillard M.C., Zurn A.D., Aebischer P., Deglon N. (2005) Progressive and selective striatal degeneration in primary neuronal cultures using lentiviral vector coding for a mutant huntingtin fragment. Neurobiol. Dis., 20, 785–798.
    1. Arrasate M., Mitra S., Schweitzer E.S., Segal M.R., Finkbeiner S. (2004) Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature, 431, 805–810.
    1. Mitra S., Tsvetkov A.S., Finkbeiner S. (2009) Single neuron ubiquitin-proteasome dynamics accompanying inclusion body formation in Huntington disease. J. Biol. Chem., 284, 4398–4403.
    1. Ben Sahra I., Regazzetti C., Robert G., Laurent K., Le Marchand-Brustel Y., Auberger P., Tanti J.F., Giorgetti-Peraldi S., Bost F. (2011) Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res., 71, 4366–4372.
    1. Foretz M., Hebrard S., Leclerc J., Zarrinpashneh E., Soty M., Mithieux G., Sakamoto K., Andreelli F., Viollet B. (2010) Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease in hepatic energy state. J. Clin. Invest., 120, 2355–2369.
    1. Onken B., Driscoll M. (2010) Metformin induces a dietary restriction-like state and the oxidative stress response to extend C. elegans Healthspan via AMPK, LKB1, and SKN-1. PLoS One, 5, e8758.
    1. Anisimov V.N., Berstein L.M., Popovich I.G., Zabezhinski M.A., Egormin P.A., Piskunova T.S., Semenchenko A.V., Tyndyk M.L., Yurova M.N., Kovalenko I.G. et al. (2011) If started early in life, metformin treatment increases life span and postpones tumors in female SHR mice. Aging (Albany NY), 3, 148–157.
    1. Cabreiro F., Au C., Leung K.Y., Vergara-Irigaray N., Cocheme H.M., Noori T., Weinkove D., Schuster E., Greene N.D., Gems D. (2013) Metformin retards aging in C. elegans by altering microbial folate and methionine metabolism. Cell, 153, 228–239.
    1. Laberge R.M., Sun Y., Orjalo A.V., Patil C.K., Freund A., Zhou L., Curran S.C., Davalos A.R., Wilson-Edell K.A., Liu S. et al. (2015) MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat. Cell. Biol., 17, 1049–1061.
    1. Coughlan K.S., Mitchem M.R., Hogg M.C., Prehn J.H. (2015) “Preconditioning” with latrepirdine, an adenosine 5′-monophosphate-activated protein kinase activator, delays amyotrophic lateral sclerosis progression in SOD1(G93A) mice. Neurobiol. Aging, 36, 1140–1150.
    1. Wong V.K., Wu A.G., Wang J.R., Liu L., Law B.Y. (2015) Neferine attenuates the protein level and toxicity of mutant huntingtin in PC-12 cells via induction of autophagy. Molecules, 20, 3496–3514.
    1. Wu A.G., Wong V.K., Xu S.W., Chan W.K., Ng C.I., Liu L., Law B.Y. (2013) Onjisaponin B derived from Radix Polygalae enhances autophagy and accelerates the degradation of mutant alpha-synuclein and huntingtin in PC-12 cells. Int. J. Mol. Sci., 14, 22618–22641.
    1. Vingtdeux V., Chandakkar P., Zhao H., d'Abramo C., Davies P., Marambaud P. (2011) Novel synthetic small-molecule activators of AMPK as enhancers of autophagy and amyloid-beta peptide degradation. FASEB J., 25, 219–231.
    1. Brenner S. (1974) The genetics of Caenorhabditis elegans. Genetics, 77, 71–94.
    1. Parker J.A., Connolly J.B., Wellington C., Hayden M., Dausset J., Neri C. (2001) Expanded polyglutamines in Caenorhabditis elegans cause axonal abnormalities and severe dysfunction of PLM mechanosensory neurons without cell death. Proc. Natl Acad. Sci. USA, 98, 13318–13323.
    1. Lin K., Dorman J.B., Rodan A., Kenyon C. (1997) daf-16: An HNF-3/forkhead family member that can function to double the life-span of Caenorhabditis elegans. Science (New York), 278, 1319–1322.
    1. Tissenbaum H.A., Guarente L. (2001) Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans. Nature, 410, 227–230.
    1. Walker D.S., Vazquez-Manrique R.P., Gower N.J., Gregory E., Schafer W.R., Baylis H.A. (2009) Inositol 1,4,5-trisphosphate signalling regulates the avoidance response to nose touch in Caenorhabditis elegans. PLoS Genet., 5, e1000636.
    1. Mello C., Fire A. (1995) DNA transformation. Methods Cell Biol., 48, 451–482.
    1. Gauthier L.R., Charrin B.C., Borrell-Pages M., Dompierre J.P., Rangone H., Cordelieres F.P., De Mey J., MacDonald M.E., Lessmann V., Humbert S. et al. (2004) Huntingtin controls neurotrophic support and survival of neurons by enhancing BDNF vesicular transport along microtubules. Cell, 118, 127–138.
    1. Weiss A., Abramowski D., Bibel M., Bodner R., Chopra V., DiFiglia M., Fox J., Kegel K., Klein C., Grueninger S. et al. (2009) Single-step detection of mutant huntingtin in animal and human tissues: a bioassay for Huntington's disease. Anal. Biochem., 395, 8–15.
    1. Wild E.J., Boggio R., Langbehn D., Robertson N., Haider S., Miller J.R., Zetterberg H., Leavitt B.R., Kuhn R., Tabrizi S.J. et al. (2015) Quantification of mutant huntingtin protein in cerebrospinal fluid from Huntington's disease patients. J. Clin. Invest., 125, 1979–1986.
    1. de Almeida L.P., Ross C.A., Zala D., Aebischer P., Deglon N. (2002) Lentiviral-mediated delivery of mutant huntingtin in the striatum of rats induces a selective neuropathology modulated by polyglutamine repeat size, huntingtin expression levels, and protein length. J. Neurosci., 22, 3473–3483.
    1. Hottinger A.F., Azzouz M., Deglon N., Aebischer P., Zurn A.D. (2000) Complete and long-term rescue of lesioned adult motoneurons by lentiviral-mediated expression of glial cell line-derived neurotrophic factor in the facial nucleus. J. Neurosci., 20, 5587–5593.
    1. Drouet V., Perrin V., Hassig R., Dufour N., Auregan G., Alves S., Bonvento G., Brouillet E., Luthi-Carter R., Hantraye P. et al. (2009) Sustained effects of nonallele-specific Huntingtin silencing. Ann. Neurol., 65, 276–285.

Source: PubMed

3
订阅