Skip to main content

In Vitro Screening Platforms for Identifying Autophagy Modulators in Mammalian Cells

  • Protocol
  • First Online:
Book cover Autophagy

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1880))

Abstract

Autophagy is a vital homeostatic pathway essential for cellular survival and human health. It primarily functions as an intracellular degradation process for the turnover of aggregation-prone proteins and unwanted organelles. Dysregulation of autophagy underlying diverse human diseases reduces cell viability, whereas stimulation of autophagy is cytoprotective in a number of transgenic disease models including neurodegenerative disorders. Thus, therapeutic exploitation of autophagy is considered a potential treatment strategy in certain human diseases, and therefore, chemical inducers of autophagy have tremendous biomedical relevance. In this review, we describe the in vitro screening platforms to identify autophagy modulators in mammalian cells using various methodologies including fluorescence and high-content imaging, flow cytometry, fluorescence and luminescence detection by microplate reader, immunoblotting, and immunofluorescence. The commonly used autophagy reporters in these screening platforms are either based on autophagy marker like LC3 or autophagy substrate such as aggregation-prone proteins or p62/SQSTM1. The reporters and assays for monitoring autophagy are evolving over time to become more sensitive in measuring autophagic flux with the capability of high-throughput applications for drug discovery. Here we highlight these developments and also describe the stringent secondary autophagy assays for characterizing the autophagy modulators arising from the primary screen. Since autophagy is implicated in myriad human physiological and pathological conditions, these technologies will enable identifying novel chemical modulators or genetic regulators of autophagy that will be of biomedical and fundamental importance to human health.

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

Access this chapter

Protocol
USD 49.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 189.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 249.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Mizushima N, Levine B, Cuervo AM, Klionsky DJ (2008) Autophagy fights disease through cellular self-digestion. Nature 451:1069–1075

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ravikumar B, Sarkar S, Davies JE, Futter M, Garcia-Arencibia M, Green-Thompson ZW, Jimenez-Sanchez M, Korolchuk VI, Lichtenberg M, Luo S, Massey DC, Menzies FM, Moreau K, Narayanan U, Renna M, Siddiqi FH, Underwood BR, Winslow AR, Rubinsztein DC (2010) Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev 90:1383–1435

    Article  CAS  PubMed  Google Scholar 

  3. Levine B, Kroemer G (2008) Autophagy in the pathogenesis of disease. Cell 132:27–42

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Boya P, Reggiori F, Codogno P (2013) Emerging regulation and functions of autophagy. Nat Cell Biol 15:713–720

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Jiang P, Mizushima N (2014) Autophagy and human diseases. Cell Res 24:69–79

    Article  CAS  PubMed  Google Scholar 

  6. Rubinsztein DC, Codogno P, Levine B (2012) Autophagy modulation as a potential therapeutic target for diverse diseases. Nat Rev Drug Discov 11:709–730

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Levine B, Packer M, Codogno P (2015) Development of autophagy inducers in clinical medicine. J Clin Investig 125:14–24

    Article  PubMed  PubMed Central  Google Scholar 

  8. Sarkar S (2013) Regulation of autophagy by mTOR-dependent and mTOR-independent pathways: autophagy dysfunction in neurodegenerative diseases and therapeutic application of autophagy enhancers. Biochem Soc Trans 41:1103–1130

    Article  CAS  PubMed  Google Scholar 

  9. Nixon RA (2013) The role of autophagy in neurodegenerative disease. Nat Med 19:983–997

    Article  CAS  PubMed  Google Scholar 

  10. Sarkar S, Ravikumar B, Floto RA, Rubinsztein DC (2009) Rapamycin and mTOR-independent autophagy inducers ameliorate toxicity of polyglutamine-expanded huntingtin and related proteinopathies. Cell Death Differ 16:46–56

    Article  CAS  PubMed  Google Scholar 

  11. Renna M, Jimenez-Sanchez M, Sarkar S, Rubinsztein DC (2010) Chemical inducers of autophagy that enhance the clearance of mutant proteins in neurodegenerative diseases. J Biol Chem 285:11061–11067

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yang Z, Klionsky DJ (2010) Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol 22:124–131

    Article  CAS  PubMed  Google Scholar 

  13. Mizushima N (2007) Autophagy: process and function. Genes Dev 21:2861–2873

    Article  CAS  PubMed  Google Scholar 

  14. Ktistakis NT, Tooze SA (2016) Digesting the expanding mechanisms of autophagy. Trends Cell Biol 26:624–635

    Article  CAS  PubMed  Google Scholar 

  15. Ganley IG (2013) Autophagosome maturation and lysosomal fusion. Essays Biochem 55:65–78

    Article  CAS  PubMed  Google Scholar 

  16. Reggiori F, Ungermann C (2017) Autophagosome maturation and fusion. J Mol Biol 429:486–496

    Article  CAS  PubMed  Google Scholar 

  17. Mizushima N, Yoshimori T, Ohsumi Y (2011) The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol 27:107–132

    Article  CAS  PubMed  Google Scholar 

  18. Mizushima N, Noda T, Yoshimori T, Tanaka Y, Ishii T, George MD, Klionsky DJ, Ohsumi M, Ohsumi Y (1998) A protein conjugation system essential for autophagy. Nature 395:395–398

    Article  CAS  PubMed  Google Scholar 

  19. Klionsky DJ, Schulman BA (2014) Dynamic regulation of macroautophagy by distinctive ubiquitin-like proteins. Nat Struct Mol Biol 21:336–345

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, Kominami E, Ohsumi Y, Yoshimori T (2000) LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J 19:5720–5728

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kimura S, Noda T, Yoshimori T (2007) Dissection of the autophagosome maturation process by a novel reporter protein, tandem fluorescent-tagged LC3. Autophagy 3:452–460

    Article  CAS  PubMed  Google Scholar 

  22. Kaizuka T, Morishita H, Hama Y, Tsukamoto S, Matsui T, Toyota Y, Kodama A, Ishihara T, Mizushima T, Mizushima N (2016) An autophagic flux probe that releases an internal control. Mol Cell 64:835–849

    Article  CAS  PubMed  Google Scholar 

  23. Mizushima N, Yoshimori T, Levine B (2010) Methods in mammalian autophagy research. Cell 140:313–326

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Mizushima N, Levine B (2010) Autophagy in mammalian development and differentiation. Nat Cell Biol 12:823–830

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kuma A, Hatano M, Matsui M, Yamamoto A, Nakaya H, Yoshimori T, Ohsumi Y, Tokuhisa T, Mizushima N (2004) The role of autophagy during the early neonatal starvation period. Nature 432:1032–1036

    Article  CAS  PubMed  Google Scholar 

  26. Komatsu M, Waguri S, Ueno T, Iwata J, Murata S, Tanida I, Ezaki J, Mizushima N, Ohsumi Y, Uchiyama Y, Kominami E, Tanaka K, Chiba T (2005) Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J Cell Biol 169:425–434

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, Yokoyama M, Mishima K, Saito I, Okano H, Mizushima N (2006) Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 441:885–889

    Article  CAS  PubMed  Google Scholar 

  28. Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, Ueno T, Koike M, Uchiyama Y, Kominami E, Tanaka K (2006) Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature 441:880–884

    Article  CAS  PubMed  Google Scholar 

  29. Sarkar S, Ravikumar B, Rubinsztein DC (2009) Autophagic clearance of aggregate-prone proteins associated with neurodegeneration. Methods Enzymol 453:83–110

    Article  CAS  PubMed  Google Scholar 

  30. Bjorkoy G, Lamark T, Pankiv S, Overvatn A, Brech A, Johansen T (2009) Monitoring autophagic degradation of p62/SQSTM1. Methods Enzymol 452:181–197

    Article  PubMed  CAS  Google Scholar 

  31. Zhu Y, Chen G, Chen L, Zhang W, Feng D, Liu L, Chen Q (2014) Monitoring mitophagy in mammalian cells. Methods Enzymol 547:39–55

    Article  CAS  PubMed  Google Scholar 

  32. Ezaki J, Komatsu M, Yokota S, Ueno T, Kominami E (2009) Method for monitoring pexophagy in mammalian cells. Methods Enzymol 452:215–226

    Article  CAS  PubMed  Google Scholar 

  33. Lystad AH, Simonsen A (2015) Assays to monitor aggrephagy. Methods 75:112–119

    Article  CAS  PubMed  Google Scholar 

  34. Kim YC, Guan KL (2015) mTOR: a pharmacologic target for autophagy regulation. J Clin Investig 125:25–32

    Article  PubMed  PubMed Central  Google Scholar 

  35. Blommaart EF, Luiken JJ, Blommaart PJ, van Woerkom GM, Meijer AJ (1995) Phosphorylation of ribosomal protein S6 is inhibitory for autophagy in isolated rat hepatocytes. J Biol Chem 270:2320–2326

    Article  CAS  PubMed  Google Scholar 

  36. Mizushima N (2010) The role of the Atg1/ULK1 complex in autophagy regulation. Curr Opin Cell Biol 22:132–139

    Article  CAS  PubMed  Google Scholar 

  37. Sarkar S, Floto RA, Berger Z, Imarisio S, Cordenier A, Pasco M, Cook LJ, Rubinsztein DC (2005) Lithium induces autophagy by inhibiting inositol monophosphatase. J Cell Biol 170:1101–1111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Williams A, Sarkar S, Cuddon P, Ttofi EK, Saiki S, Siddiqi FH, Jahreiss L, Fleming A, Pask D, Goldsmith P, O'Kane CJ, Floto RA, Rubinsztein DC (2008) Novel targets for Huntington’s disease in an mTOR-independent autophagy pathway. Nat Chem Biol 4:295–305

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Thoreen CC, Kang SA, Chang JW, Liu Q, Zhang J, Gao Y, Reichling LJ, Sim T, Sabatini DM, Gray NS (2009) An ATP-competitive mammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1. J Biol Chem 284:8023–8032

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Sarkar S, Davies JE, Huang Z, Tunnacliffe A, Rubinsztein DC (2007) Trehalose, a novel mTOR-independent autophagy enhancer, accelerates the clearance of mutant huntingtin and alpha-synuclein. J Biol Chem 282:5641–5652

    Article  CAS  PubMed  Google Scholar 

  41. Sarkar S, Perlstein EO, Imarisio S, Pineau S, Cordenier A, Maglathlin RL, Webster JA, Lewis TA, O'Kane CJ, Schreiber SL, Rubinsztein DC (2007) Small molecules enhance autophagy and reduce toxicity in Huntington’s disease models. Nat Chem Biol 3:331–338

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sarkar S (2013) Chemical screening platforms for autophagy drug discovery to identify therapeutic candidates for Huntington’s disease and other neurodegenerative disorders. Drug Discov Today Technol 10:e137–e144

    Article  PubMed  Google Scholar 

  43. Joachim J, Jiang M, McKnight NC, Howell M, Tooze SA (2015) High-throughput screening approaches to identify regulators of mammalian autophagy. Methods 75:96–104

    Article  CAS  PubMed  Google Scholar 

  44. Fleming A, Noda T, Yoshimori T, Rubinsztein DC (2011) Chemical modulators of autophagy as biological probes and potential therapeutics. Nat Chem Biol 7:9–17

    Article  CAS  PubMed  Google Scholar 

  45. Shu CW, Liu PF, Huang CM (2012) High throughput screening for drug discovery of autophagy modulators. Comb Chem High Throughput Screen 15:721–729

    Article  CAS  PubMed  Google Scholar 

  46. Allen GF, Toth R, James J, Ganley IG (2013) Loss of iron triggers PINK1/Parkin-independent mitophagy. EMBO Rep 14:1127–1135

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Mishra P, Dauphinee AN, Ward C, Sarkar S, Gunawardena A, Manjithaya R (2017) Discovery of pan autophagy inhibitors through a high-throughput screen highlights macroautophagy as an evolutionarily conserved process across 3 eukaryotic kingdoms. Autophagy 13:1556–1572

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. DeBosch BJ, Heitmeier MR, Mayer AL, Higgins CB, Crowley JR, Kraft TE, Chi M, Newberry EP, Chen Z, Finck BN, Davidson NO, Yarasheski KE, Hruz PW, Moley KH (2016) Trehalose inhibits solute carrier 2A (SLC2A) proteins to induce autophagy and prevent hepatic steatosis. Sci Signal 9:ra21

    Article  PubMed  PubMed Central  Google Scholar 

  49. Palmieri M, Pal R, Nelvagal HR, Lotfi P, Stinnett GR, Seymour ML, Chaudhury A, Bajaj L, Bondar VV, Bremner L, Saleem U, Tse DY, Sanagasetti D, Wu SM, Neilson JR, Pereira FA, Pautler RG, Rodney GG, Cooper JD, Sardiello M (2017) mTORC1-independent TFEB activation via Akt inhibition promotes cellular clearance in neurodegenerative storage diseases. Nat Commun 8:14338

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Maetzel D, Sarkar S, Wang H, Abi-Mosleh L, Xu P, Cheng AW, Gao Q, Mitalipova M, Jaenisch R (2014) Genetic and chemical correction of cholesterol accumulation and impaired autophagy in hepatic and neural cells derived from Niemann-Pick Type C patient-specific iPS cells. Stem Cell Reports 2:866–880

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Perucho J, Casarejos MJ, Gomez A, Solano RM, de Yebenes JG, Mena MA (2012) Trehalose protects from aggravation of amyloid pathology induced by isoflurane anesthesia in APP(swe) mutant mice. Curr Alzheimer Res 9:334–343

    Article  CAS  PubMed  Google Scholar 

  52. Schaeffer V, Lavenir I, Ozcelik S, Tolnay M, Winkler DT, Goedert M (2012) Stimulation of autophagy reduces neurodegeneration in a mouse model of human tauopathy. Brain 135:2169–2177

    Article  PubMed  PubMed Central  Google Scholar 

  53. Rodriguez-Navarro JA, Rodriguez L, Casarejos MJ, Solano RM, Gomez A, Perucho J, Cuervo AM, Garcia de Yebenes J, Mena MA (2010) Trehalose ameliorates dopaminergic and tau pathology in parkin deleted/tau overexpressing mice through autophagy activation. Neurobiol Dis 39:423–438

    Article  CAS  PubMed  Google Scholar 

  54. Castillo K, Nassif M, Valenzuela V, Rojas F, Matus S, Mercado G, Court FA, van Zundert B, Hetz C (2013) Trehalose delays the progression of amyotrophic lateral sclerosis by enhancing autophagy in motoneurons. Autophagy 9:1308–1320

    Article  CAS  PubMed  Google Scholar 

  55. Zhang X, Chen S, Song L, Tang Y, Shen Y, Jia L, Le W (2014) MTOR-independent, autophagic enhancer trehalose prolongs motor neuron survival and ameliorates the autophagic flux defect in a mouse model of amyotrophic lateral sclerosis. Autophagy 10:588–602

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Belzile JP, Sabalza M, Craig M, Clark E, Morello CS, Spector DH (2015) Trehalose, an mTOR-independent inducer of autophagy, inhibits human cytomegalovirus infection in multiple cell types. J Virol 90:1259–1277

    Article  PubMed  CAS  Google Scholar 

  57. Kruger U, Wang Y, Kumar S, Mandelkow EM (2012) Autophagic degradation of tau in primary neurons and its enhancement by trehalose. Neurobiol Aging 33:2291–2305

    Article  PubMed  CAS  Google Scholar 

  58. Aguib Y, Heiseke A, Gilch S, Riemer C, Baier M, Schatzl HM, Ertmer A (2009) Autophagy induction by trehalose counteracts cellular prion infection. Autophagy 5:361–369

    Article  CAS  PubMed  Google Scholar 

  59. Tien NT, Karaca I, Tamboli IY, Walter J (2016) Trehalose alters subcellular trafficking and the metabolism of the Alzheimer-associated amyloid precursor protein. J Biol Chem 291:10528–10540

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Yamamoto A, Tagawa Y, Yoshimori T, Moriyama Y, Masaki R, Tashiro Y (1998) Bafilomycin A1 prevents maturation of autophagic vacuoles by inhibiting fusion between autophagosomes and lysosomes in rat hepatoma cell line, H-4-II-E cells. Cell Struct Funct 23:33–42

    Article  CAS  PubMed  Google Scholar 

  61. Fass E, Shvets E, Degani I, Hirschberg K, Elazar Z (2006) Microtubules support production of starvation-induced autophagosomes but not their targeting and fusion with lysosomes. J Biol Chem 281:36303–36316

    Article  CAS  PubMed  Google Scholar 

  62. Klionsky DJ, Elazar Z, Seglen PO, Rubinsztein DC (2008) Does bafilomycin A1 block the fusion of autophagosomes with lysosomes? Autophagy 4:849–850

    Article  CAS  PubMed  Google Scholar 

  63. Klionsky DJ et al (2016) Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 12:1–222

    Article  PubMed  PubMed Central  Google Scholar 

  64. Larsen KB, Lamark T, Overvatn A, Harneshaug I, Johansen T, Bjorkoy G (2010) A reporter cell system to monitor autophagy based on p62/SQSTM1. Autophagy 6:784–793

    Article  CAS  PubMed  Google Scholar 

  65. Brown A, Patel S, Ward C, Lorenz A, Ortiz M, DuRoss A, Wieghardt F, Esch A, Otten EG, Heiser LM, Korolchuk VI, Sun C, Sarkar S, Sahay G (2016) PEG-lipid micelles enable cholesterol efflux in Niemann-Pick Type C1 disease-based lysosomal storage disorder. Sci Rep 6:31750

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Wyttenbach A, Swartz J, Kita H, Thykjaer T, Carmichael J, Bradley J, Brown R, Maxwell M, Schapira A, Orntoft TF, Kato K, Rubinsztein DC (2001) Polyglutamine expansions cause decreased CRE-mediated transcription and early gene expression changes prior to cell death in an inducible cell model of Huntington’s disease. Hum Mol Genet 10:1829–1845

    Article  CAS  PubMed  Google Scholar 

  67. Webb JL, Ravikumar B, Atkins J, Skepper JN, Rubinsztein DC (2003) Alpha-Synuclein is degraded by both autophagy and the proteasome. J Biol Chem 278:25009–25013

    Article  CAS  PubMed  Google Scholar 

  68. Yoshii SR, Mizushima N (2017) Monitoring and measuring autophagy. Int J Mol Sci 18:E1865

    Article  PubMed  CAS  Google Scholar 

  69. Kabeya Y, Mizushima N, Yamamoto A, Oshitani-Okamoto S, Ohsumi Y, Yoshimori T (2004) LC3, GABARAP and GATE16 localize to autophagosomal membrane depending on form-II formation. J Cell Sci 117:2805–2812

    Article  CAS  PubMed  Google Scholar 

  70. Zhang L, Yu J, Pan H, Hu P, Hao Y, Cai W, Zhu H, Yu AD, Xie X, Ma D, Yuan J (2007) Small molecule regulators of autophagy identified by an image-based high-throughput screen. Proc Natl Acad Sci U S A 104:19023–19028

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Balgi AD, Fonseca BD, Donohue E, Tsang TC, Lajoie P, Proud CG, Nabi IR, Roberge M (2009) Screen for chemical modulators of autophagy reveals novel therapeutic inhibitors of mTORC1 signaling. PLoS One 4:e7124

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Li Y, McGreal S, Zhao J, Huang R, Zhou Y, Zhong H, Xia M, Ding WX (2016) A cell-based quantitative high-throughput image screening identified novel autophagy modulators. Pharmacol Res 110:35–49

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. McKnight NC, Jefferies HB, Alemu EA, Saunders RE, Howell M, Johansen T, Tooze SA (2012) Genome-wide siRNA screen reveals amino acid starvation-induced autophagy requires SCOC and WAC. EMBO J 31:1931–1946

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Lipinski MM, Hoffman G, Ng A, Zhou W, Py BF, Hsu E, Liu X, Eisenberg J, Liu J, Blenis J, Xavier RJ, Yuan J (2010) A genome-wide siRNA screen reveals multiple mTORC1 independent signaling pathways regulating autophagy under normal nutritional conditions. Dev Cell 18:1041–1052

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Chan EY, Kir S, Tooze SA (2007) siRNA screening of the kinome identifies ULK1 as a multidomain modulator of autophagy. J Biol Chem 282:25464–25474

    Article  CAS  PubMed  Google Scholar 

  76. He P, Peng Z, Luo Y, Wang L, Yu P, Deng W, An Y, Shi T, Ma D (2009) High-throughput functional screening for autophagy-related genes and identification of TM9SF1 as an autophagosome-inducing gene. Autophagy 5:52–60

    Article  CAS  PubMed  Google Scholar 

  77. Orvedahl A, Sumpter R Jr, Xiao G, Ng A, Zou Z, Tang Y, Narimatsu M, Gilpin C, Sun Q, Roth M, Forst CV, Wrana JL, Zhang YE, Luby-Phelps K, Xavier RJ, Xie Y, Levine B (2011) Image-based genome-wide siRNA screen identifies selective autophagy factors. Nature 480:113–117

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Li M, Yang Z, Vollmer LL, Gao Y, Fu Y, Liu C, Chen X, Liu P, Vogt A, Yin XM (2015) AMDE-1 is a dual function chemical for autophagy activation and inhibition. PLoS One 10:e0122083

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Szyniarowski P, Corcelle-Termeau E, Farkas T, Hoyer-Hansen M, Nylandsted J, Kallunki T, Jaattela M (2011) A comprehensive siRNA screen for kinases that suppress macroautophagy in optimal growth conditions. Autophagy 7:892–903

    Article  CAS  PubMed  Google Scholar 

  80. Pietrocola F, Lachkar S, Enot DP, Niso-Santano M, Bravo-San Pedro JM, Sica V, Izzo V, Maiuri MC, Madeo F, Marino G, Kroemer G (2015) Spermidine induces autophagy by inhibiting the acetyltransferase EP300. Cell Death Differ 22:509–516

    Article  CAS  PubMed  Google Scholar 

  81. Lao Y, Wan G, Liu Z, Wang X, Ruan P, Xu W, Xu D, Xie W, Zhang Y, Xu H, Xu N (2014) The natural compound oblongifolin C inhibits autophagic flux and enhances antitumor efficacy of nutrient deprivation. Autophagy 10:736–749

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Peppard JV, Rugg C, Smicker M, Dureuil C, Ronan B, Flamand O, Durand L, Pasquier B (2014) Identifying small molecules which inhibit autophagy: a phenotypic screen using image-based high-content cell analysis. Curr Chem Genom Transl Med 8:3–15

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Kuo SY, Castoreno AB, Aldrich LN, Lassen KG, Goel G, Dancik V, Kuballa P, Latorre I, Conway KL, Sarkar S, Maetzel D, Jaenisch R, Clemons PA, Schreiber SL, Shamji AF, Xavier RJ (2015) Small-molecule enhancers of autophagy modulate cellular disease phenotypes suggested by human genetics. Proc Natl Acad Sci U S A 112:E4281–E4287

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Sarkar S, Korolchuk V, Renna M, Winslow A, Rubinsztein DC (2009) Methodological considerations for assessing autophagy modulators: a study with calcium phosphate precipitates. Autophagy 5:307–313

    Article  CAS  PubMed  Google Scholar 

  85. Chauhan S, Ahmed Z, Bradfute SB, Arko-Mensah J, Mandell MA, Won Choi S, Kimura T, Blanchet F, Waller A, Mudd MH, Jiang S, Sklar L, Timmins GS, Maphis N, Bhaskar K, Piguet V, Deretic V (2015) Pharmaceutical screen identifies novel target processes for activation of autophagy with a broad translational potential. Nat Commun 6:8620

    Article  CAS  PubMed  Google Scholar 

  86. Pampaloni F, Mayer B, Kabat Vel-Job K, Ansari N, Hotte K, Kogel D, Stelzer EHK (2017) A novel cellular spheroid-based autophagy screen applying live fluorescence microscopy identifies nonactin as a strong inducer of autophagosomal turnover. SLAS Discov 22:558–570

    CAS  PubMed  Google Scholar 

  87. Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Overvatn A, Bjorkoy G, Johansen T (2007) p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 282:24131–24145

    Article  CAS  PubMed  Google Scholar 

  88. N'Diaye EN, Kajihara KK, Hsieh I, Morisaki H, Debnath J, Brown EJ (2009) PLIC proteins or ubiquilins regulate autophagy-dependent cell survival during nutrient starvation. EMBO Rep 10:173–179

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Shaner NC, Steinbach PA, Tsien RY (2005) A guide to choosing fluorescent proteins. Nat Methods 2:905–909

    Article  CAS  PubMed  Google Scholar 

  90. Zhou C, Zhong W, Zhou J, Sheng F, Fang Z, Wei Y, Chen Y, Deng X, Xia B, Lin J (2012) Monitoring autophagic flux by an improved tandem fluorescent-tagged LC3 (mTagRFP-mWasabi-LC3) reveals that high-dose rapamycin impairs autophagic flux in cancer cells. Autophagy 8:1215–1226

    Article  CAS  PubMed  Google Scholar 

  91. Zhang JH, Chung TD, Oldenburg KR (1999) A simple statistical parameter for use in evaluation and validation of high throughput screening assays. J Biomol Screen 4:67–73

    Article  CAS  PubMed  Google Scholar 

  92. Malo N, Hanley JA, Cerquozzi S, Pelletier J, Nadon R (2006) Statistical practice in high-throughput screening data analysis. Nat Biotechnol 24:167–175

    Article  CAS  PubMed  Google Scholar 

  93. Ravikumar B, Sarkar S, Rubinsztein DC (2008) Clearance of mutant aggregate-prone proteins by autophagy. Methods Mol Biol 445:195–211

    Article  CAS  PubMed  Google Scholar 

  94. Rubinsztein DC (2006) The roles of intracellular protein-degradation pathways in neurodegeneration. Nature 443:780–786

    Article  CAS  PubMed  Google Scholar 

  95. Ravikumar B, Duden R, Rubinsztein DC (2002) Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy. Hum Mol Genet 11:1107–1117

    Article  CAS  PubMed  Google Scholar 

  96. Bjorkoy G, Lamark T, Brech A, Outzen H, Perander M, Overvatn A, Stenmark H, Johansen T (2005) p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol 171:603–614

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Stolz A, Ernst A, Dikic I (2014) Cargo recognition and trafficking in selective autophagy. Nat Cell Biol 16:495–501

    Article  CAS  PubMed  Google Scholar 

  98. Hale CM, Cheng Q, Ortuno D, Huang M, Nojima D, Kassner PD, Wang S, Ollmann MM, Carlisle HJ (2016) Identification of modulators of autophagic flux in an image-based high content siRNA screen. Autophagy 12:713–726

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. DeJesus R, Moretti F, McAllister G, Wang Z, Bergman P, Liu S, Frias E, Alford J, Reece-Hoyes JS, Lindeman A, Kelliher J, Russ C, Knehr J, Carbone W, Beibel M, Roma G, Ng A, Tallarico JA, Porter JA, Xavier RJ, Mickanin C, Murphy LO, Hoffman GR, Nyfeler B (2016) Functional CRISPR screening identifies the ufmylation pathway as a regulator of SQSTM1/p62. eLife 5:e17290

    Article  PubMed  PubMed Central  Google Scholar 

  100. Strohecker AM, Joshi S, Possemato R, Abraham RT, Sabatini DM, White E (2015) Identification of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase as a novel autophagy regulator by high content shRNA screening. Oncogene 34:5662–5676

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Tsuganezawa K, Shinohara Y, Ogawa N, Tsuboi S, Okada N, Mori M, Yokoyama S, Noda NN, Inagaki F, Ohsumi Y, Tanaka A (2013) Two-colored fluorescence correlation spectroscopy screening for LC3-P62 interaction inhibitors. J Biomol Screen 18:1103–1109

    Article  PubMed  CAS  Google Scholar 

  102. Farkas T, Hoyer-Hansen M, Jaattela M (2009) Identification of novel autophagy regulators by a luciferase-based assay for the kinetics of autophagic flux. Autophagy 5:1018–1025

    Article  CAS  PubMed  Google Scholar 

  103. Farkas T, Daugaard M, Jaattela M (2011) Identification of small molecule inhibitors of phosphatidylinositol 3-kinase and autophagy. J Biol Chem 286:38904–38912

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Rubinsztein DC, Cuervo AM, Ravikumar B, Sarkar S, Korolchuk V, Kaushik S, Klionsky DJ (2009) In search of an “autophagomometer”. Autophagy 5:585–589

    Article  CAS  PubMed  Google Scholar 

  105. Sarkar S, Korolchuk VI, Renna M, Imarisio S, Fleming A, Williams A, Garcia-Arencibia M, Rose C, Luo S, Underwood BR, Kroemer G, O'Kane CJ, Rubinsztein DC (2011) Complex inhibitory effects of nitric oxide on autophagy. Mol Cell 43:19–32

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Sun N, Malide D, Liu J, Rovira II, Combs CA, Finkel T (2017) A fluorescence-based imaging method to measure in vitro and in vivo mitophagy using mt-Keima. Nat Protoc 12:1576–1587

    Article  CAS  PubMed  Google Scholar 

  107. Shibutani ST, Yoshimori T (2014) A current perspective of autophagosome biogenesis. Cell Res 24:58–68

    Article  CAS  PubMed  Google Scholar 

  108. Mizushima N, Yamamoto A, Hatano M, Kobayashi Y, Kabeya Y, Suzuki K, Tokuhisa T, Ohsumi Y, Yoshimori T (2001) Dissection of autophagosome formation using Apg5-deficient mouse embryonic stem cells. J Cell Biol 152:657–668

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Fujita N, Itoh T, Omori H, Fukuda M, Noda T, Yoshimori T (2008) The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Mol Biol Cell 19:2092–2100

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Axe EL, Walker SA, Manifava M, Chandra P, Roderick HL, Habermann A, Griffiths G, Ktistakis NT (2008) Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum. J Cell Biol 182:685–701

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Proikas-Cezanne T, Pfisterer SG (2009) Assessing mammalian autophagy by WIPI-1/Atg18 puncta formation. Methods Enzymol 452:247–260

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We thank the funding agencies for supporting our research. SS is funded by Wellcome Trust Seed Award (109626/Z/15/Z), UKIERI (UK-India Education and Research Initiative) DST Thematic Partnership Award (2016-17-0087), and Birmingham Fellowship. TRR is funded by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP; 2015/02041-1), Fundação de Apoio à Pesquisa da Faculdade de Ciências Médicas da Santa Casa de São Paulo (FAP FCMSCSP), and Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq)/Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES). SS is also a Former Fellow for life at Hughes Hall, University of Cambridge, UK.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sovan Sarkar .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Science+Business Media, LLC, part of Springer Nature

About this protocol

Check for updates. Verify currency and authenticity via CrossMark

Cite this protocol

Seranova, E., Ward, C., Chipara, M., Rosenstock, T.R., Sarkar, S. (2019). In Vitro Screening Platforms for Identifying Autophagy Modulators in Mammalian Cells. In: Ktistakis, N., Florey, O. (eds) Autophagy. Methods in Molecular Biology, vol 1880. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-8873-0_26

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-8873-0_26

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-8872-3

  • Online ISBN: 978-1-4939-8873-0

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics