期刊论文详细信息
Chemistry Central Journal
Using C. elegans to discover therapeutic compounds for ageing-associated neurodegenerative diseases
Xi Chen1  Jeff W. Barclay2  Robert D. Burgoyne2  Alan Morgan2 
[1] Centre for Neurodegenerative Science, Van Andel Research Institute, 333 Bostwick Avenue NE, Grand Rapids, Michigan 49503, MI, USA
[2] Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown St, Liverpool L69 3BX, UK
关键词: Parkinson’s disease;    Neurodegeneration;    Huntington’s disease;    Frontotemporal dementia;    Compound screening;    Caenorhabditis elegans;    Amyotrophic lateral sclerosis;    Alzheimer’s disease;    Aging;    Adult onset neuronal ceroid lipofuscinosis;   
Others  :  1234570
DOI  :  10.1186/s13065-015-0143-y
 received in 2015-08-27, accepted in 2015-11-15,  发布年份 2015
PDF
【 摘 要 】

Age-associated neurodegenerative disorders such as Alzheimer’s disease are a major public health challenge, due to the demographic increase in the proportion of older individuals in society. However, the relatively few currently approved drugs for these conditions provide only symptomatic relief. A major goal of neurodegeneration research is therefore to identify potential new therapeutic compounds that can slow or even reverse disease progression, either by impacting directly on the neurodegenerative process or by activating endogenous physiological neuroprotective mechanisms that decline with ageing. This requires model systems that can recapitulate key features of human neurodegenerative diseases that are also amenable to compound screening approaches. Mammalian models are very powerful, but are prohibitively expensive for high-throughput drug screens. Given the highly conserved neurological pathways between mammals and invertebrates, Caenorhabditis elegans has emerged as a powerful tool for neuroprotective compound screening. Here we describe how C. elegans has been used to model various human ageing-associated neurodegenerative diseases and provide an extensive list of compounds that have therapeutic activity in these worm models and so may have translational potential.

【 授权许可】

   
2015 Chen et al.

【 预 览 】
附件列表
Files Size Format View
20151203010326894.pdf 1765KB PDF download
Fig.4. 67KB Image download
Fig.3. 41KB Image download
Fig.2. 83KB Image download
Fig.1. 73KB Image download
【 图 表 】

Fig.1.

Fig.2.

Fig.3.

Fig.4.

【 参考文献 】
  • [1]Muchowski PJ. Protein misfolding, amyloid formation, and neurodegeneration: a critical role for molecular chaperones? Neuron. 2002; 35(1):9-12.
  • [2]Taylor JP, Hardy J, Fischbeck KH. Toxic proteins in neurodegenerative disease. Science. 2002; 296(5575):1991-1995.
  • [3]Soto C, Estrada LD. Protein misfolding and neurodegeneration. Arch Neurol. 2008; 65(2):184-189.
  • [4]Ehrnhoefer DE, Wong BK, Hayden MR. Convergent pathogenic pathways in Alzheimer’s and Huntington’s diseases: shared targets for drug development. Nat Rev Drug Discovery. 2011; 10(11):853-867.
  • [5]Hardaway JA, Hardie SL, Whitaker SM, Baas SR, Zhang B, Bermingham DP, Lichtenstein AJ, Blakely RD. Forward genetic analysis to identify determinants of dopamine signaling in Caenorhabditis elegans using swimming-induced paralysis. G3. 2012; 2(8):961-975.
  • [6]Barclay JW, Morgan A, Burgoyne RD. Neurotransmitter release mechanisms studied in Caenorhabditis elegans. Cell Calcium. 2012; 52(3–4):289-295.
  • [7]Link CD. Expression of human beta-amyloid peptide in transgenic Caenorhabditis elegans. Proc Natl Acad Sci USA. 1995; 92(20):9368-9372.
  • [8]Nass R, Miller DM, Blakely RD. C. elegans: a novel pharmacogenetic model to study Parkinson’s disease. Parkinsonism Relat Disord. 2001; 7(3):185-191.
  • [9]Satyal SH, Schmidt E, Kitagawa K, Sondheimer N, Lindquist S, Kramer JM, Morimoto RI. Polyglutamine aggregates alter protein folding homeostasis in Caenorhabditis elegans. Proc Natl Acad Sci USA. 2000; 97(11):5750-5755.
  • [10]Amor S, Puentes F, Baker D, van der Valk P. Inflammation in neurodegenerative diseases. Immunology. 2010; 129(2):154-169.
  • [11]Burns AR, Wallace IM, Wildenhain J, Tyers M, Giaever G, Bader GD, Nislow C, Cutler SR, Roy PJ. A predictive model for drug bioaccumulation and bioactivity in Caenorhabditis elegans. Nat Chem Biol. 2010; 6(7):549-557.
  • [12]Cabreiro F, Au C, Leung KY, Vergara-Irigaray N, Cocheme HM, Noori T, Weinkove D, Schuster E, Greene ND, Gems D. Metformin retards aging in C. elegans by altering microbial folate and methionine metabolism. Cell. 2013; 153(1):228-239.
  • [13]van Ham T, Breitling R, Swertz M, Nollen E. Neurodegenerative diseases: lessons from genome-wide screens in small model organisms. EMBO Mol Med. 2009; 1(8–9):360-370.
  • [14]Chen X, Burgoyne RD. Identification of common genetic modifiers of neurodegenerative diseases from an integrative analysis of diverse genetic screens in model organisms. BMC Genom. 2012; 13:71.
  • [15]Wu Y, Wu Z, Butko P, Christen Y, Lambert MP, Klein WL, Link CD, Luo Y. Amyloid-beta-induced pathological behaviors are suppressed by Ginkgo biloba extract EGb 761 and ginkgolides in transgenic Caenorhabditis elegans. J Neurosci. 2006; 26(50):13102-13113.
  • [16]Gutierrez-Zepeda A, Santell R, Wu Z, Brown M, Wu Y, Khan I, Link CD, Zhao B, Luo Y. Soy isoflavone glycitein protects against beta amyloid-induced toxicity and oxidative stress in transgenic Caenorhabditis elegans. BMC neuroscience. 2005; 6:54.
  • [17]Abbas S, Wink M. Epigallocatechin gallate from green tea (Camellia sinensis) increases lifespan and stress resistance in Caenorhabditis elegans. Planta Med. 2009; 75(3):216-221.
  • [18]Abbas S, Wink M. Epigallocatechin gallate inhibits beta amyloid oligomerization in Caenorhabditis elegans and affects the daf-2/insulin-like signaling pathway. Phytomedicine. 2010; 17(11):902-909.
  • [19]Dostal V, Roberts CM, Link CD. Genetic mechanisms of coffee extract protection in a Caenorhabditis elegans model of beta-amyloid peptide toxicity. Genetics. 2010; 186(3):857-866.
  • [20]Lublin A, Isoda F, Patel H, Yen K, Nguyen L, Hajje D, Schwartz M, Mobbs C. FDA-approved drugs that protect mammalian neurons from glucose toxicity slow aging dependent on Cbp and protect against proteotoxicity. PLoS One. 2011; 6(11):e27762.
  • [21]Alavez S, Vantipalli MC, Zucker DJ, Klang IM, Lithgow GJ. Amyloid-binding compounds maintain protein homeostasis during ageing and extend lifespan. Nature. 2011; 472(7342):226-229.
  • [22]Arya U, Dwivedi H, Subramaniam JR. Reserpine ameliorates Abeta toxicity in the Alzheimer’s disease model in Caenorhabditis elegans. Exp Gerontol. 2009; 44(6–7):462-466.
  • [23]Jagota S, Rajadas J. Effect of phenolic compounds against Abeta aggregation and Abeta-induced toxicity in transgenic C. elegans. Neurochem Res. 2012; 37(1):40-48.
  • [24]Keowkase R, Aboukhatwa M, Luo Y. Fluoxetine protects against amyloid-beta toxicity, in part via daf-16 mediated cell signaling pathway, Caenorhabditis elegans. Neuropharmacology. 2010; 59(4–5):358-365.
  • [25]Smith JV, Luo Y. Elevation of oxidative free radicals in Alzheimer’s disease models can be attenuated by Ginkgo biloba extract EGb 761. J Alzheimers Dis. 2003; 5(4):287-300.
  • [26]Diomede L, Cassata G, Fiordaliso F, Salio M, Ami D, Natalello A, Doglia SM, De Luigi A, Salmona M. Tetracycline and its analogues protect Caenorhabditis elegans from beta amyloid-induced toxicity by targeting oligomers. Neurobiol Dis. 2010; 40(2):424-431.
  • [27]Sangha JS, Sun X, Wally OSD, Zhang K, Ji X, Wang Z, Wang Y, Zidichouski J, Prithiviraj B, Zhang J. Liuwei Dihuang (LWDH), a traditional Chinese medicinal formula, protects against β-Amyloid toxicity in transgenic Caenorhabditis elegans. PLoS One. 2012; 7(8):e43990.
  • [28]Diomede L, Rigacci S, Romeo M, Stefani M, Salmona M. Oleuropein aglycone protects transgenic C. elegans strains expressing Abeta42 by reducing plaque load and motor deficit. PLoS One. 2013; 8(3):e58893.
  • [29]Tardiff DF, Tucci ML, Caldwell KA, Caldwell GA, Lindquist S. Different 8-hydroxyquinolines protect models of TDP-43 protein, alpha-synuclein, and polyglutamine proteotoxicity through distinct mechanisms. J Biol Chem. 2012; 287(6):4107-4120.
  • [30]Matlack KE, Tardiff DF, Narayan P, Hamamichi S, Caldwell KA, Caldwell GA, Lindquist S. Clioquinol promotes the degradation of metal-dependent amyloid-beta (Abeta) oligomers to restore endocytosis and ameliorate Abeta toxicity. Proc Natl Acad Sci USA. 2014; 111(11):4013-4018.
  • [31]McColl G, Roberts BR, Pukala TL, Kenche VB, Roberts CM, Link CD, Ryan TM, Masters CL, Barnham KJ, Bush AI et al.. Utility of an improved model of amyloid-beta (Aβ 1-42 ) toxicity in Caenorhabditis elegans for drug screening for Alzheimer’s disease. Mol Neurodegener. 2012; 7:57.
  • [32]Mandelkow EM, Mandelkow E. Tau in Alzheimer’s disease. Trends Cell Biol. 1998; 8(11):425-427.
  • [33]Brandt R, Gergou A, Wacker I, Fath T, Hutter H. A Caenorhabditis elegans model of tau hyperphosphorylation: induction of developmental defects by transgenic overexpression of Alzheimer’s disease-like modified tau. Neurobiol Aging. 2009; 30(1):22-33.
  • [34]Kraemer BC, Zhang B, Leverenz JB, Thomas JH, Trojanowski JQ, Schellenberg GD. Neurodegeneration and defective neurotransmission in a Caenorhabditis elegans model of tauopathy. Proc Natl Acad Sci USA. 2003; 100(17):9980-9985.
  • [35]Miyasaka T, Ding Z, Gengyo-Ando K, Oue M, Yamaguchi H, Mitani S, Ihara Y. Progressive neurodegeneration in C. elegans model of tauopathy. Neurobiol Dis. 2005; 20(2):372-383.
  • [36]Fatouros C, Pir GJ, Biernat J, Koushika SP, Mandelkow E, Mandelkow E-M, Schmidt E, Baumeister R. Inhibition of tau aggregation in a novel Caenorhabditis elegans model of tauopathy mitigates proteotoxicity. Hum Mol Genet. 2012; 21(16):3587-3603.
  • [37]McCormick AV, Wheeler JM, Guthrie CR, Liachko NF, Kraemer BC. Dopamine D2 receptor antagonism suppresses tau aggregation and neurotoxicity. Biol Psychiatry. 2013; 73(5):464-471.
  • [38]Chen X, McCue FH, Wong SQ, Kashyap SS, Kraemer BC, Barclay JW, Burgoyne RD, Morgan A. Ethosuximide ameliorates neurodegenerative disease phenotypes by modulating DAF-16/FOXO target gene expression. Mol Neurodegener. 2015; 10(1):51.
  • [39]Teixeira-Castro A, Ailion M, Jalles A, Brignull HR, Vilaca JL, Dias N, Rodrigues P, Oliveira JF, Neves-Carvalho A, Morimoto RI et al.. Neuron-specific proteotoxicity of mutant ataxin-3 in C. elegans: rescue by the DAF-16 and HSF-1 pathways. Hum Mol Genet. 2011; 20(15):2996-3009.
  • [40]Brignull HR, Morley JF, Garcia SM, Morimoto RI. Modeling polyglutamine pathogenesis in C. elegans. Methods Enzymol. 2006; 412:256-282.
  • [41]Faber PW, Voisine C, King DC, Bates EA, Hart AC. Glutamine/proline-rich PQE-1 proteins protect Caenorhabditis elegans neurons from huntingtin polyglutamine neurotoxicity. Proc Natl Acad Sci USA. 2002; 99(26):17131-17136.
  • [42]Voisine C, Varma H, Walker N, Bates EA, Stockwell BR, Hart AC. Identification of potential therapeutic drugs for huntington’s disease using Caenorhabditis elegans. PLoS One. 2007; 2(6):e504.
  • [43]Varma H, Cheng R, Voisine C, Hart AC, Stockwell BR. Inhibitors of metabolism rescue cell death in Huntington’s disease models. Proc Natl Acad Sci. 2007; 104(36):14525-14530.
  • [44]Parker J, Arango M, Abderrahmane S, Lambert E, Tourette C, Catoire H, Neri C. Resveratrol rescues mutant polyglutamine cytotoxicity in nematode and mammalian neurons. Nat Genet. 2005; 37(4):349-350.
  • [45]Shin BH, Lim Y, Oh HJ, Park SM, Lee S-K, Ahnn J, Kim DH, Song WK, Kwak TH, Park WJ. Pharmacological activation of Sirt1 ameliorates polyglutamine-induced toxicity through the regulation of autophagy. PLoS One. 2013; 8(6):e64953.
  • [46]Yang X, Zhang P, Wu J, Xiong S, Jin N, Huang Z. The neuroprotective and lifespan-extension activities of Damnacanthus officinarum extracts in Caenorhabditis elegans. J Ethnopharmacol. 2012; 141(1):41-47.
  • [47]Xiao L, Li H, Zhang J, Yang F, Huang A, Deng J, Liang M, Ma F, Hu M, Huang Z. Salidroside protects Caenorhabditis elegans neurons from polyglutamine-mediated toxicity by reducing oxidative stress. Molecules. 2014; 19(6):7757-7769.
  • [48]Haldimann P, Muriset M, Vigh L, Goloubinoff P. The novel hydroxylamine derivative NG-094 suppresses polyglutamine protein toxicity in Caenorhabditis elegans. J Biol Chem. 2011; 286(21):18784-18794.
  • [49]Cai WJ, Huang JH, Zhang SQ, Wu B, Kapahi P, Zhang XM, Shen ZY (2011) Icariin and its derivative Icariside II extend healthspan via Insulin/IGF-1 pathway in C. elegans. Plos One 6:e28835
  • [50]Ching TT, Chiang WC, Chen CS, Hsu AL. Celecoxib extends C. elegans lifespan via inhibition of insulin-like signaling but not cyclooxygenase-2 activity. Aging Cell. 2011; 10(3):506-519.
  • [51]Ayyadevara S, Bharill P, Dandapat A, Hu C, Khaidakov M, Mitra S, Shmookler Reis RJ, Mehta JL. Aspirin inhibits oxidant stress, reduces age-associated functional declines, and extends lifespan of Caenorhabditis elegans. Antioxid Redox Signal. 2013; 18(5):481-490.
  • [52]Wolozin B, Gabel C, Ferree A, Guillily M, Ebata A. Watching worms whither: modeling neurodegeneration in C. elegans. Prog Mol Biol Transl Sci. 2011; 100:499-514.
  • [53]Ved R, Saha S, Westlund B, Perier C, Burnam L, Sluder A, Hoener M, Rodrigues CM, Alfonso A, Steer C et al.. Similar patterns of mitochondrial vulnerability and rescue induced by genetic modification of alpha-synuclein, parkin, and DJ-1 in Caenorhabditis elegans. J Biol Chem. 2005; 280(52):42655-42668.
  • [54]Braungart E, Gerlach M, Riederer P, Baumeister R, Hoener MC. Caenorhabditis elegans MPP + model of Parkinson’s disease for high-throughput drug screenings. Neurodegener Dis. 2004; 1(4–5):175-183.
  • [55]Marvanova M, Nichols CD. Identification of neuroprotective compounds of Caenorhabditis elegans dopaminergic neurons against 6-OHDA. J Mol Neurosci. 2007; 31(2):127-137.
  • [56]Locke CJ, Fox SA, Caldwell GA, Caldwell KA. Acetaminophen attenuates dopamine neuron degeneration in animal models of Parkinson’s disease. Neurosci Lett. 2008; 439(2):129-133.
  • [57]Kautu BB, Carrasquilla A, Hicks ML, Caldwell KA, Caldwell GA. Valproic acid ameliorates C. elegans dopaminergic neurodegeneration with implications for ERK-MAPK signaling. Neurosci Lett. 2013; 541:116-119.
  • [58]Fu R-H, Harn H-J, Liu S-P, Chen C-S, Chang W-L, Chen Y-M, Huang J-E, Li R-J, Tsai S-Y, Hung H-S et al.. n-Butylidenephthalide protects against dopaminergic neuron degeneration and α-synuclein accumulation in Caenorhabditis elegans models of Parkinson’s Disease. PLoS One. 2014; 9(1):e85305.
  • [59]Fu R-H, Wang Y-C, Chen C-S, Tsai R-T, Liu S-P, Chang W-L, Lin H-L, Lu C-H, Wei J-R, Wang Z-W et al.. Acetylcorynoline attenuates dopaminergic neuron degeneration and α-synuclein aggregation in animal models of Parkinson’s disease. Neuropharmacology. 2014; 82:108-120.
  • [60]Liu Z, Hamamichi S, Lee BD, Yang D, Ray A, Caldwell GA, Caldwell KA, Dawson TM, Smith WW, Dawson VL. Inhibitors of LRRK2 kinase attenuate neurodegeneration and Parkinson-like phenotypes in Caenorhabditis elegans and Drosophila Parkinson’s disease models. Hum Mol Genet. 2011; 20(20):3933-3942.
  • [61]Yao C, Johnson WM, Gao Y, Wang W, Zhang J, Deak M, Alessi DR, Zhu X, Mieyal JJ, Roder H et al (2012) Kinase inhibitors arrest neurodegeneration in cell and C. elegans models of LRRK2 toxicity. Hum Mol Genet 22:328–344
  • [62]Vaccaro A, Patten SA, Ciura S, Maios C, Therrien M, Drapeau P, Kabashi E, Parker JA. Methylene Blue protects against TDP-43 and FUS neuronal toxicity in C. elegans and D. rerio. PLoS One. 2012; 7(7):e42117.
  • [63]Tauffenberger A, Julien C, Parker JA. Evaluation of longevity enhancing compounds against transactive response DNA-binding protein-43 neuronal toxicity. Neurobiol Aging. 2013; 34(9):2175-2182.
  • [64]Liachko NF, McMillan PJ, Guthrie CR, Bird TD, Leverenz JB, Kraemer BC. CDC7 inhibition blocks pathological TDP-43 phosphorylation and neurodegeneration. Ann Neurol. 2013; 74(1):39-52.
  • [65]Haltia M. The neuronal ceroid-lipofuscinoses. J Neuropathol Exp Neurol. 2003; 62(1):1-13.
  • [66]Haltia M, Goebel HH. The neuronal ceroid-lipofuscinoses: a historical introduction. Biochim Biophys Acta. 2013; 1832(11):1795-1800.
  • [67]Noskova L, Stranecky V, Hartmannova H, Pristoupilova A, Baresova V, Ivanek R, Hulkova H, Jahnova H, van der Zee J, Staropoli JF et al.. Mutations in DNAJC5, encoding cysteine-string protein alpha, cause autosomal-dominant adult-onset neuronal ceroid lipofuscinosis. Am J Hum Genet. 2011; 89(2):241-252.
  • [68]Benitez BA, Alvarado D, Cai Y, Mayo K, Chakraverty S, Norton J, Morris JC, Sands MS, Goate A, Cruchaga C. Exome-sequencing confirms DNAJC5 mutations as cause of adult neuronal ceroid-lipofuscinosis. PLoS One. 2011; 6(11):e26741.
  • [69]Velinov M, Dolzhanskaya N, Gonzalez M, Powell E, Konidari I, Hulme W, Staropoli JF, Xin W, Wen GY, Barone R et al.. Mutations in the gene DNAJC5 cause autosomal dominant Kufs disease in a proportion of cases: study of the Parry family and 8 other families. PLoS One. 2012; 7(1):e29729.
  • [70]Cadieux-Dion M, Andermann E, Lachance-Touchette P, Ansorge O, Meloche C, Barnabe A, Kuzniecky RI, Andermann F, Faught E, Leonberg S et al.. Recurrent mutations in DNAJC5 cause autosomal dominant Kufs disease. Clin Genet. 2013; 83(6):571-575.
  • [71]Kashyap SS, Johnson JR, McCue HV, Chen X, Edmonds MJ, Ayala M, Graham ME, Jenn RC, Barclay JW, Burgoyne RD et al.. Caenorhabditis elegans dnj-14, the orthologue of the DNAJC5 gene mutated in adult onset neuronal ceroid lipofuscinosis, provides a new platform for neuroprotective drug screening and identifies a SIR-2.1-independent action of resveratrol. Hum Mol Genet. 2014; 23(22):5916-5927.
  • [72]Burgoyne RD, Morgan A. Cysteine string protein (CSP) and its role in preventing neurodegeneration. Semin Cell Dev Biol. 2015; 40:153-159.
  • [73]Bizat N, Peyrin JM, Haik S, Cochois V, Beaudry P, Laplanche JL, Neri C. Neuron dysfunction is induced by prion protein with an insertional mutation via a Fyn kinase and reversed by sirtuin activation in Caenorhabditis elegans. J Neurosci. 2010; 30(15):5394-5403.
  • [74]Karuppagounder SS, Pinto JT, Xu H, Chen HL, Beal MF, Gibson GE. Dietary supplementation with resveratrol reduces plaque pathology in a transgenic model of Alzheimer’s disease. Neurochem Int. 2009; 54(2):111-118.
  • [75]Tiwari SK, Seth B, Agarwal S, Yadav A, Karmakar M, Gupta SK, Choubey V, Sharma A, Chaturvedi RK (2015) Ethosuximide induces hippocampal neurogenesis and reverses cognitive deficits in amyloid-beta toxin induced Alzheimer’s rat model via PI3 K/Akt/Wnt/beta-catenin pathway. J Biol Chem 290:28540–28558
  • [76]McCue HV, Chen X, Barclay JW, Morgan A, Burgoyne RD. Expression profile of a Caenorhabditis elegans model of adult neuronal ceroid lipofuscinosis reveals down regulation of ubiquitin E3 ligase components. Sci Rep. 2015; 5:14392.
  • [77]Sleigh JN, Buckingham SD, Esmaeili B, Viswanathan M, Cuppen E, Westlund BM, Sattelle DB. A novel Caenorhabditis elegans allele, smn-1(cb131), mimicking a mild form of spinal muscular atrophy, provides a convenient drug screening platform highlighting new and pre-approved compounds. Hum Mol Genet. 2011; 20(2):245-260.
  • [78]Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA. 2005; 294(15):1934-1943.
  • [79]Crunelli V, Leresche N. Block of thalamic T-type Ca(2+) channels by ethosuximide is not the whole story. Epilepsy Curr Am Epilepsy Soc. 2002; 2(2):53-56.
  • [80]Park SJ, Ahmad F, Philp A, Baar K, Williams T, Luo H, Ke H, Rehmann H, Taussig R, Brown AL et al.. Resveratrol ameliorates aging-related metabolic phenotypes by inhibiting cAMP phosphodiesterases. Cell. 2012; 148(3):421-433.
  • [81]Hubbard BP, Gomes AP, Dai H, Li J, Case AW, Considine T, Riera TV, Lee JE, SY S, Lamming DW et al.. Evidence for a common mechanism of SIRT1 regulation by allosteric activators. Science. 2013; 339(6124):1216-1219.
  • [82]Evason K, Huang C, Yamben I, Covey DF, Kornfeld K. Anticonvulsant medications extend worm life-span. Science. 2005; 307(5707):258-262.
  • [83]Baur JA. Resveratrol, sirtuins, and the promise of a DR mimetic. Mech Ageing Dev. 2010; 131(4):261-269.
  • [84]Pani G. Neuroprotective effects of dietary restriction: evidence and mechanisms. Semin Cell Dev Biol. 2015; 40:106-114.
  • [85]Swinney DC, Anthony J. How were new medicines discovered? Nat Rev Drug Discovery. 2011; 10(7):507-519.
  • [86]Swinney DC. The contribution of mechanistic understanding to phenotypic screening for first-in-class medicines. J Biomol Screen. 2013; 18(10):1186-1192.
  • [87]Weiss RA. Special anniversary review: 25 years of human immunodeficiency virus research: successes and challenges. Clin Exp Immunol. 2008; 152(2):201-210.
  • [88]Frokjaer-Jensen C. Exciting prospects for precise engineering of Caenorhabditis elegans genomes with CRISPR/Cas9. Genetics. 2013; 195(3):635-642.
  • [89]Fay DS, Fluet A, Johnson CJ, Link CD. In vivo aggregation of beta-amyloid peptide variants. J Neurochem. 1998; 71(4):1616-1625.
  • [90]Link CD, Johnson CJ, Fonte V, Paupard M-C, Hall DH, Styren S, Mathis CA, Klunk WE. Visualization of fibrillar amyloid deposits in living, transgenic Caenorhabditis elegans animals using the sensitive amyloid dye, X-34. Neurobiol Aging. 2001; 22(2):217-226.
  • [91]Yatin SM, Yatin M, Aulick T, Ain KB, Butterfield DA. Alzheimer’s amyloid beta-peptide associated free radicals increase rat embryonic neuronal polyamine uptake and ornithine decarboxylase activity: protective effect of vitamin E. Neurosci Lett. 1999; 263(1):17-20.
  • [92]Drake J, Link CD, Butterfield DA. Oxidative stress precedes fibrillar deposition of Alzheimer’s disease amyloid beta-peptide (1–42) in a transgenic Caenorhabditis elegans model. Neurobiol Aging. 2003; 24(3):415-420.
  • [93]Florez-McClure ML, Hohsfield LA, Fonte G, Bealor MT, Link CD. Decreased insulin-receptor signaling promotes the autophagic degradation of beta-amyloid peptide in C. elegans. Autophagy. 2007; 3(6):569-580.
  • [94]Link CD, Taft A, Kapulkin V, Duke K, Kim S, Fei Q, Wood DE, Sahagan BG. Gene expression analysis in a transgenic Caenorhabditis elegans Alzheimer’s disease model. Neurobiol Aging. 2003; 24(3):397-413.
  • [95]Aparecida Paiva F, de Freitas Bonomo L, Ferreira Boasquivis P, Borges Raposo de Paula IT, Guerra JF, Mendes Leal W, Silva ME, Pedrosa ML, Oliveira Rde P. Carqueja (Baccharis trimera) Protects against oxidative stress and beta-amyloid-induced toxicity in Caenorhabditis elegans. Oxid Med Cell Longev. 2015; 2015:740162.
  • [96]Link CD. C. elegans models of age-associated neurodegenerative diseases: lessons from transgenic worm models of Alzheimer’s disease. Exp Gerontol. 2006; 41(10):1007-1013.
  • [97]Dosanjh LE, Brown MK, Rao G, Link CD, Luo Y. Behavioral phenotyping of a transgenic Caenorhabditis elegans expressing neuronal amyloid-β. J Alzheimers Dis. 2010; 19(2):681-690.
  • [98]Treusch S, Hamamichi S, Goodman JL, Matlack KES, Chung CY, Baru V, Shulman JM, Parrado A, Bevis BJ, Valastyan JS et al.. Functional links between Aβ toxicity, endocytic trafficking, and Alzheimer’s disease risk factors in yeast. Science. 2011; 334(6060):1241-1245.
  • [99]Hornsten A, Lieberthal J, Fadia S, Malins R, Ha L, Xu X, Daigle I, Markowitz M, O’Connor G, Plasterk R et al.. APL-1, a Caenorhabditis elegans protein related to the human beta-amyloid precursor protein, is essential for viability. Proc Natl Acad Sci USA. 2007; 104(6):1971-1976.
  • [100]Ewald CY, Cheng R, Tolen L, Shah V, Gillani A, Nasrin A, Li C. Pan-neuronal expression of APL-1, an APP-related protein, disrupts olfactory, gustatory, and touch plasticity in Caenorhabditis elegans. J Neurosci. 2012; 32(30):10156-10169.
  • [101]Oeda T, Shimohama S, Kitagawa N, Kohno R, Imura T, Shibasaki H, Ishii N. Oxidative stress causes abnormal accumulation of familial amyotrophic lateral sclerosis-related mutant SOD1 in transgenic Caenorhabditis elegans. Hum Mol Genet. 2001; 10(19):2013-2023.
  • [102]Wang J, Farr GW, Hall DH, Li F, Furtak K, Dreier L, Horwich AL. An ALS-linked mutant SOD1 produces a locomotor defect associated with aggregation and synaptic dysfunction when expressed in neurons of Caenorhabditis elegans. PLoS Genet. 2009; 5(1):e1000350.
  • [103]Witan H, Kern A, Koziollek-Drechsler I, Wade R, Behl C, Clement AM. Heterodimer formation of wild-type and amyotrophic lateral sclerosis-causing mutant Cu/Zn-superoxide dismutase induces toxicity independent of protein aggregation. Hum Mol Genet. 2008; 17(10):1373-1385.
  • [104]Murakami A, Kojima K, Ohya K, Imamura K, Takasaki Y. A new conformational epitope generated by the binding of recombinant 70-kd protein and U1 RNA to anti-U1 RNP autoantibodies in sera from patients with mixed connective tissue disease. Arthritis Rheum. 2002; 46(12):3273-3282.
  • [105]Gidalevitz T, Krupinski T, Garcia S, Morimoto RI. Destabilizing protein polymorphisms in the genetic background direct phenotypic expression of mutant SOD1 toxicity. PLoS Genet. 2009; 5(3):e1000399.
  • [106]Silva DF, Esteves AR, Oliveira CR, Cardoso SM. Mitochondria: the common upstream driver of amyloid-beta and tau pathology in Alzheimer’s disease. Curr Alzheimer Res. 2011; 8(5):563-572.
  • [107]Zhang T, Mullane PC, Periz G, Wang J. TDP-43 neurotoxicity and protein aggregation modulated by heat shock factor and insulin/IGF-1 signaling. Hum Mol Genet. 2011; 20(10):1952-1965.
  • [108]Li J, Huang KX, Wd L. WD: Establishing a novel C. elegans model to investigate the role of autophagy in amyotrophic lateral sclerosis. Acta Pharmacol Sin. 2013; 34(5):644-650.
  • [109]Liachko NF, Guthrie CR, Kraemer BC. Phosphorylation promotes neurotoxicity in a Caenorhabditis elegans model of TDP-43 proteinopathy. J Neurosci. 2010; 30(48):16208-16219.
  • [110]Ash PE, Zhang YJ, Roberts CM, Saldi T, Hutter H, Buratti E, Petrucelli L, Link CD. Neurotoxic effects of TDP-43 overexpression in C. elegans. Hum Mol Genet. 2010; 19(16):3206-3218.
  • [111]Morley JF, Brignull HR, Weyers JJ, Morimoto RI. The threshold for polyglutamine-expansion protein aggregation and cellular toxicity is dynamic and influenced by aging in Caenorhabditis elegans. Proc Natl Acad Sci. 2002; 99(16):10417-10422.
  • [112]Wang H, Lim PJ, Yin C, Rieckher M, Vogel BE, Monteiro MJ. Suppression of polyglutamine-induced toxicity in cell and animal models of Huntington’s disease by ubiquilin. Hum Mol Genet. 2006; 15(6):1025-1041.
  • [113]Yamanaka K, Okubo Y, Suzaki T, Ogura T. Analysis of the two p97/VCP/Cdc48p proteins of Caenorhabditis elegans and their suppression of polyglutamine-induced protein aggregation. J Struct Biol. 2004; 146(1–2):242-250.
  • [114]Parker J, Connolly J, Wellington C, Hayden M, Dausset J, Neri C. Expanded polyglutamines in Caenorhabditis elegans cause axonal abnormalities and severe dysfunction of PLM mechanosensory neurons without cell death. Proc Natl Acad Sci USA. 2001; 98(23):13318-13323.
  • [115]Lejeune F-X, Mesrob L, Parmentier F, Bicep C, Vazquez-Manrique R, Parker JA, Vert J-P, Tourette C, Neri C. Large-scale functional RNAi screen in C. elegans identifies genes that regulate the dysfunction of mutant polyglutamine neurons. BMC Genom. 2012; 13(1):91.
  • [116]Teixeira-Castro A, Ailion M, Jalles A, Brignull HR, Vilaça JL, Dias N, Rodrigues P, Oliveira JF, Neves-Carvalho A, Morimoto RI, Maciel P. Neuron-specific proteotoxicity of mutant ataxin-3 in C. elegans: rescue by the DAF-16 and HSF-1 pathways. Hum Mol Genet. 2011; 20:2996-3009.
  • [117]Christie NT, Lee AL, Fay HG, Gray AA, Kikis EA. Novel polyglutamine model uncouples proteotoxicity from aging. PLoS One. 2014; 9(5):e96835.
  • [118]Hamamichi S, Rivas RN, Knight AL, Cao S, Caldwell KA, Caldwell GA. Hypothesis-based RNAi screening identifies neuroprotective genes in a Parkinson’s disease model. Proc Natl Acad Sci USA. 2008; 105(2):728-733.
  • [119]van Ham TJ, Thijssen KL, Breitling R, Hofstra RMW, Plasterk RHA, Nollen EAA. C. elegans model identifies genetic modifiers of α-Synuclein inclusion formation during aging. PLoS Genet. 2008; 4(3):e1000027.
  • [120]Lakso M, Vartiainen S, Moilanen AM, Sirvio J, Thomas JH, Nass R, Blakely RD, Wong G. Dopaminergic neuronal loss and motor deficits in Caenorhabditis elegans overexpressing human alpha-synuclein. J Neurochem. 2003; 86(1):165-172.
  • [121]Settivari R, LeVora J, Nass R (2009) The divalent metal transporter homologues SMF-1/2 mediates dopamine neuron sensitivity in Caenorhabditis elegans models of manganism and Parkinson’s disease. J Biol Chem M109.051409
  • [122]Cao S, Gelwix CC, Caldwell KA, Caldwell GA. Torsin-mediated protection from cellular stress in the dopaminergic neurons of Caenorhabditis elegans. J Neurosci. 2005; 25(15):3801-3812.
  • [123]Kuwahara T, Koyama A, Gengyo-Ando K, Masuda M, Kowa H, Tsunoda M, Mitani S, Iwatsubo T. Familial Parkinson mutant alpha-synuclein causes dopamine neuron dysfunction in transgenic Caenorhabditis elegans. J Biol Chem. 2006; 281(1):334-340.
  • [124]Buttner S, Broeskamp F, Sommer C, Markaki M, Habernig L, Alavian-Ghavanini A, Carmona-Gutierrez D, Eisenberg T, Michael E, Kroemer G et al.. Spermidine protects against alpha-synuclein neurotoxicity. Cell Cycle. 2014; 13(24):3903-3908.
  • [125]Karpinar DP, Balija MB, Kugler S, Opazo F, Rezaei-Ghaleh N, Wender N, Kim HY, Taschenberger G, Falkenburger BH, Heise H et al.. Pre-fibrillar alpha-synuclein variants with impaired beta-structure increase neurotoxicity in Parkinson’s disease models. EMBO J. 2009; 28(20):3256-3268.
  • [126]Kuwahara T, Koyama A, Koyama S, Yoshina S, Ren C-H, Kato T, Mitani S, Iwatsubo T. A systematic RNAi screen reveals involvement of endocytic pathway in neuronal dysfunction in α-synuclein transgenic C. elegans. Hum Mol Genet. 2008; 17(19):2997-3009.
  • [127]Kuwahara T, Tonegawa R, Ito G, Mitani S, Iwatsubo T. Phosphorylation of α-synuclein protein at Ser-129 reduces neuronal dysfunction by lowering its membrane binding property in Caenorhabditis elegans. J Biol Chem. 2012; 287(10):7098-7109.
  • [128]Saha S, Guillily MD, Ferree A, Lanceta J, Chan D, Ghosh J, Hsu CH, Segal L, Raghavan K, Matsumoto K et al.. LRRK2 modulates vulnerability to mitochondrial dysfunction in Caenorhabditis elegans. J Neurosci. 2009; 29(29):9210-9218.
  • [129]Yao C, El Khoury R, Wang W, Byrd TA, Pehek EA, Thacker C, Zhu X, Smith MA, Wilson-Delfosse AL, Chen SG. LRRK2-mediated neurodegeneration and dysfunction of dopaminergic neurons in a Caenorhabditis elegans model of Parkinson’s disease. Neurobiol Dis. 2010; 40(1):73-81.
  • [130]Bizat N, Peyrin J-M, Haïk S, Cochois V, Beaudry P, Laplanche J-L, Néri C. Neuron dysfunction is induced by prion protein with an insertional mutation via a Fyn kinase and reversed by Sirtuin activation in Caenorhabditis elegans. J Neurosci. 2010; 30(15):5394-5403.
  • [131]Park K-W, Li L. Prion protein in Caenorhabditis elegans: distinct models of anti-BAX and neuropathology. Prion. 2011; 5(1):28-38.
  • [132]Nussbaum-Krammer CI, Park K-W, Li L, Melki R, Morimoto RI. Spreading of a prion domain from cell-to-cell by vesicular transport in Caenorhabditis elegans. PLoS Genet. 2013; 9(3):e1003351.
  • [133]Lakowski B, Hekimi S. The genetics of caloric restriction in Caenorhabditis elegans. Proc Natl Acad Sci USA. 1998; 95(22):13091-13096.
  • [134]Levitan D, Greenwald I. Effects of SEL-12 presenilin on LIN-12 localization and function in Caenorhabditis elegans. Development. 1998; 125(18):3599-3606.
  • [135]Wittenburg N, Eimer S, Lakowski B, Rohrig S, Rudolph C, Baumeister R. Presenilin is required for proper morphology and function of neurons in C. elegans. Nature. 2000; 406(6793):306-309.
  • [136]Springer W, Hoppe T, Schmidt E, Baumeister R. A Caenorhabditis elegans Parkin mutant with altered solubility couples alpha-synuclein aggregation to proteotoxic stress. Hum Mol Genet. 2005; 14(22):3407-3423.
  • [137]Sämann J, Hegermann J, von Gromoff E, Eimer S, Baumeister R, Schmidt E. Caenorhabditits elegans LRK-1 and PINK-1 act antagonistically in stress response and neurite outgrowth. J Biol Chem. 2009; 284(24):16482-16491.
  • [138]Briese M, Esmaeili B, Fraboulet S, Burt EC, Christodoulou S, Towers PR, Davies KE, Sattelle DB. Deletion of smn-1, the Caenorhabditis elegans ortholog of the spinal muscular atrophy gene, results in locomotor dysfunction and reduced lifespan. Hum Mol Genet. 2009; 18(1):97-104.
  • [139]Nass R, Miller DM, Blakely RD. C. elegans: a novel pharmacogenetic model to study Parkinson’s disease. Parkinsonism Relat Disord. 2001; 7(3):185-191.
  • [140]Ruan Q, Harrington AJ, Caldwell KA, Caldwell GA, Standaert DG. VPS41, a protein involved in lysosomal trafficking, is protective in Caenorhabditis elegans and mammalian cellular models of Parkinson’s disease. Neurobiol Dis. 2010; 37(2):330-338.
  • [141]Liu J, Banskota AH, Critchley AT, Hafting J, Prithiviraj B. Neuroprotective effects of the cultivated Chondrus crispus in a C. elegans model of Parkinson’s disease. Mar Drugs. 2015; 13(4):2250-2266.
  • [142]Caldwell KA, Tucci ML, Armagost J, Hodges TW, Chen J, Memon SB, Blalock JE, DeLeon SM, Findlay RH, Ruan Q et al.. Investigating bacterial sources of toxicity as an environmental contributor to dopaminergic neurodegeneration. PLoS One. 2009; 4(10):e7227.
  文献评价指标  
  下载次数:47次 浏览次数:34次