Skip to main content
Log in

A functional unfolded protein response is required for chronological aging in Saccharomyces cerevisiae

  • Original Article
  • Published:
Current Genetics Aims and scope Submit manuscript

Abstract

Progressive impairment of proteostasis and accumulation of toxic misfolded proteins are associated with the cellular aging process. Here, we employed chronologically aged yeast cells to investigate how activation of the unfolded protein response (UPR) upon accumulation of misfolded proteins in the endoplasmic reticulum (ER) affects lifespan. We found that cells lacking a functional UPR display a significantly reduced chronological lifespan, which contrasts previous findings in models of replicative aging. We find exacerbated UPR activation in aged cells, indicating an increase in misfolded protein burden in the ER during the course of aging. We also observed that caloric restriction, which promotes longevity in various model organisms, extends lifespan of UPR-deficient strains. Similarly, aging in pH-buffered media extends lifespan, albeit independently of the UPR. Thus, our data support a role for caloric restriction and reduced acid stress in improving ER homeostasis during aging. Finally, we show that UPR-mediated upregulation of the ER chaperone Kar2 and functional ER-associated degradation (ERAD) are essential for proper aging. Our work documents the central role of secretory protein homeostasis in chronological aging in yeast and highlights that the requirement for a functional UPR can differ between post-mitotic and actively dividing eukaryotic cells.

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

Access this article

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

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  • Aerts AM, Zabrocki P, Govaert G, Mathys J, Carmona-Gutierrez D, Madeo F et al (2009) Mitochondrial dysfunction leads to reduced chronological lifespan and increased apoptosis in yeast. FEBS Lett 583:113–117

    CAS  PubMed  Google Scholar 

  • Aragón T, van Anken E, Pincus D, Serafimova IM, Korennykh AV, Rubio CA, Walter P (2009) Messenger RNA targeting to endoplasmic reticulum stress signalling sites. Nature 457:736–740

    PubMed  Google Scholar 

  • Baird NA, Douglas PM, Simic MS, Grant AR, Moresco JJ, Wolff SC et al (2014) HSF-1-mediated cytoskeletal integrity determines thermotolerance and life span. Science 346:360–363

    CAS  PubMed  PubMed Central  Google Scholar 

  • Beaupere C, Labunskyy VM (2019) (Un)folding mechanisms of adaptation to ER stress: lessons from aneuploidy. Curr Genet 65:467–471

    CAS  PubMed  Google Scholar 

  • Beaupere C, Dinatto L, Wasko BM, Chen RB, VanValkenburg L, Kiflezghi MG et al (2018) Genetic screen identifies adaptive aneuploidy as a key mediator of ER stress resistance in yeast. Proc Natl Acad Sci USA 115:9586–9591

    CAS  PubMed  PubMed Central  Google Scholar 

  • Ben-Zvi A, Miller EA, Morimoto RI (2009) Collapse of proteostasis represents an early molecular event in Caenorhabditis elegans aging. Proc Natl Acad Sci USA 106:14914–14919

    CAS  PubMed  PubMed Central  Google Scholar 

  • Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D (2000) Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat Cell Biol 2:326–332

    CAS  PubMed  Google Scholar 

  • Biederer T, Volkwein C, Sommer T (1997) Role of Cue1p in ubiquitination and degradation at the ER surface. Science 278:1806–1809

    CAS  PubMed  Google Scholar 

  • Brachmann CB, Davies A, Cost GJ, Caputo E, Li J, Hieter P, Boeke JD (1998) Designer deletion strains derived from Saccharomyces cerevisiae S288C: a useful set of strains and plasmids for PCR-mediated gene disruption and other applications. Yeast 14:115–132

    CAS  PubMed  Google Scholar 

  • Brehme M, Voisine C (2016) Model systems of protein-misfolding diseases reveal chaperone modifiers of proteotoxicity. Dis Model Mech 9:823–838

    CAS  PubMed  PubMed Central  Google Scholar 

  • Breitenbach M, Laun P, Dickinson JR, Klocker A, Rinnerthaler M, Dawes IW et al (2012) The role of mitochondria in the aging processes of yeast. Subcell Biochem 57:55–78

    CAS  PubMed  Google Scholar 

  • Breslow DK, Cameron DM, Collins SR, Schuldiner M, Stewart-Ornstein J, Newman HW et al (2008) A comprehensive strategy enabling high-resolution functional analysis of the yeast genome. Nat Methods 5:711–718

    CAS  PubMed  PubMed Central  Google Scholar 

  • Brown MK, Naidoo N (2012) The endoplasmic reticulum stress response in aging and age-related diseases. Front Physiol 3:263

    PubMed  PubMed Central  Google Scholar 

  • Burtner CR, Murakami CJ, Kaeberlein M (2009a) A genomic approach to yeast chronological aging. Methods Mol Biol 548:101–114

    CAS  PubMed  Google Scholar 

  • Burtner CR, Murakami CJ, Kennedy BK, Kaeberlein M (2009b) A molecular mechanism of chronological aging in yeast. Cell Cycle 8:1256–1270

    CAS  PubMed  Google Scholar 

  • Carvalho P, Goder V, Rapoport TA (2006) Distinct ubiquitin-ligase complexes define convergent pathways for the degradation of ER proteins. Cell 126:361–373

    CAS  PubMed  Google Scholar 

  • Chadwick SR, Lajoie P (2019) Endoplasmic reticulum stress coping mechanisms and lifespan regulation in health and diseases. Front Cell Dev Biol 7:84. https://doi.org/10.3389/fcell.2019.00084

    Article  PubMed  PubMed Central  Google Scholar 

  • Chadwick SR, Pananos AD, Di Gregorio SE, Park AE, Etedali-Zadeh P, Duennwald ML, Lajoie P (2016) A toolbox for rapid quantitative assessment of chronological lifespan and survival in Saccharomyces cerevisiae. Traffic 17:689–703

    CAS  PubMed  Google Scholar 

  • Choi K-M, Kwon Y-Y, Lee C-K (2013) Characterization of global gene expression during assurance of lifespan extension by caloric restriction in budding yeast. Exp Gerontol 48:1455–1468

    CAS  PubMed  Google Scholar 

  • Cohen A, Weindling E, Rabinovich E, Nachman I, Fuchs S, Chuartzman S et al (2016) Water-transfer slows aging in Saccharomyces cerevisiae. PLoS One 11:e0148650

    PubMed  PubMed Central  Google Scholar 

  • Colman RJ, Anderson RM, Johnson SC, Kastman EK, Kosmatka KJ, Beasley TM et al (2009) Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 325:201–204

    CAS  PubMed  PubMed Central  Google Scholar 

  • Cox JS, Walter P (1996) A novel mechanism for regulating activity of a transcription factor that controls the unfolded protein response. Cell 87:391–404

    CAS  PubMed  Google Scholar 

  • Cox JS, Shamu CE, Walter P (1993) Transcriptional induction of genes encoding endoplasmic reticulum resident proteins requires a transmembrane protein kinase. Cell 73:1197–1206

    CAS  PubMed  Google Scholar 

  • Demir AB, Koc A (2010) Assessment of chronological lifespan dependent molecular damages in yeast lacking mitochondrial antioxidant genes. Biochem Biophys Res Commun 400:106–110

    CAS  PubMed  Google Scholar 

  • Denoth Lippuner A, Julou T, Barral Y (2014) Budding yeast as a model organism to study the effects of age. FEMS Microbiol Rev 38:300–325

    CAS  PubMed  Google Scholar 

  • Denzel MS, Storm NJ, Gutschmidt A, Baddi R, Hinze Y, Jarosch E et al (2014) Hexosamine pathway metabolites enhance protein quality control and prolong life. Cell 156:1167–1178

    CAS  PubMed  Google Scholar 

  • Di Gregorio SE, Duennwald ML (2018) Yeast as a model to study protein misfolding in aged cells. FEMS Yeast Res. https://doi.org/10.1093/femsyr/foy054

    Article  PubMed  Google Scholar 

  • Douglas PM, Baird NA, Simic MS, Uhlein S, McCormick MA, Wolff SC et al (2015) Heterotypic signals from neural HSF-1 separate thermotolerance from longevity. Cell Rep 12:1196–1204

    CAS  PubMed  PubMed Central  Google Scholar 

  • Duennwald ML, Lindquist S (2008) Impaired ERAD and ER stress are early and specific events in polyglutamine toxicity. Genes Dev 22:3308–3319

    CAS  PubMed  PubMed Central  Google Scholar 

  • Fabrizio P, Longo VD (2007) The chronological life span of Saccharomyces cerevisiae. Methods Mol Biol 371:89–95

    CAS  PubMed  Google Scholar 

  • Fabrizio P, Pozza F, Pletcher SD, Gendron CM, Longo VD (2001) Regulation of longevity and stress resistance by Sch9 in yeast. Science 292:288–290

    CAS  PubMed  Google Scholar 

  • Fabrizio P, Gattazzo C, Battistella L, Wei M, Cheng C, McGrew K, Longo VD (2005) Sir2 blocks extreme life-span extension. Cell 123:655–667

    CAS  PubMed  Google Scholar 

  • Friedlander R, Jarosch E, Urban J, Volkwein C, Sommer T (2000) A regulatory link between ER-associated protein degradation and the unfolded-protein response. Nat Cell Biol 2:379–384

    CAS  PubMed  Google Scholar 

  • Garay E, Campos SE, González de la Cruz J, Gaspar AP, Jinich A, Deluna A (2014) High-resolution profiling of stationary-phase survival reveals yeast longevity factors and their genetic interactions. PLoS Genet 10:e1004168

    PubMed  PubMed Central  Google Scholar 

  • Gardner BM, Walter P (2011) Unfolded proteins are Ire1-activating ligands that directly induce the unfolded protein response. Science 333:1891–1894

    CAS  PubMed  PubMed Central  Google Scholar 

  • Gavilán MP, Pintado C, Gavilán E, Jiménez S, Ríos RM, Vitorica J et al (2009) Dysfunction of the unfolded protein response increases neurodegeneration in aged rat hippocampus following proteasome inhibition. Aging Cell 8:654–665

    PubMed  Google Scholar 

  • Ghavidel A, Baxi K, Ignatchenko V, Prusinkiewicz M, Arnason TG, Kislinger T et al (2015) A genome scale screen for mutants with delayed exit from mitosis: Ire1-independent induction of autophagy integrates ER homeostasis into mitotic lifespan. PLoS Genet 11:e1005429

    PubMed  PubMed Central  Google Scholar 

  • Griac P, Henry SA (1999) The yeast inositol-sensitive upstream activating sequence, UASINO, responds to nitrogen availability. Nucleic Acids Res 27:2043–2050

    CAS  PubMed  PubMed Central  Google Scholar 

  • Guarente L, Kenyon C (2000) Genetic pathways that regulate ageing in model organisms. Nature 408:255–262

    CAS  PubMed  Google Scholar 

  • Henis-Korenblit S, Zhang P, Hansen M, McCormick M, Lee S-J, Cary M, Kenyon C (2010) Insulin/IGF-1 signaling mutants reprogram ER stress response regulators to promote longevity. Proc Natl Acad Sci USA 107:9730–9735

    CAS  PubMed  PubMed Central  Google Scholar 

  • Herker E, Jungwirth H, Lehmann KA, Maldener C, Fröhlich K-U, Wissing S et al (2004) Chronological aging leads to apoptosis in yeast. J Cell Biol 164:501–507

    CAS  PubMed  PubMed Central  Google Scholar 

  • Höhn A, Weber D, Jung T, Ott C, Hugo M, Kochlik B et al (2017) Happily (n) ever after: aging in the context of oxidative stress, proteostasis loss and cellular senescence. Redox Biol 11:482–501

    PubMed  Google Scholar 

  • Hsu C-L, Prasad R, Blackman C, Ng DTW (2012) Endoplasmic reticulum stress regulation of the Kar2p/BiP chaperone alleviates proteotoxicity via dual degradation pathways. Mol Biol Cell 23:630–641

    CAS  PubMed  PubMed Central  Google Scholar 

  • Hu J, Wei M, Mirisola MG, Longo VD (2013) Assessing chronological aging in Saccharomyces cerevisiae. Methods Mol Biol 965:463–472

    CAS  PubMed  PubMed Central  Google Scholar 

  • Hussain SG, Ramaiah KVA (2007) Reduced eIF2alpha phosphorylation and increased proapoptotic proteins in aging. Biochem Biophys Res Commun 355:365–370

    CAS  PubMed  Google Scholar 

  • Jiang JC, Jaruga E, Repnevskaya MV, Jazwinski SM (2000) An intervention resembling caloric restriction prolongs life span and retards aging in yeast. FASEB J 14:2135–2137

    CAS  PubMed  Google Scholar 

  • Jiang Y, Chadwick SR, Lajoie P (2016) Endoplasmic reticulum stress: the cause and solution to Huntington’s disease? Brain Res 1648:650–657

    CAS  PubMed  Google Scholar 

  • Jin H, Mimura N, Kashio M, Koseki H, Aoe T (2014) Late-onset of spinal neurodegeneration in knock-in mice expressing a mutant BiP. PLoS One 9:e112837

    PubMed  PubMed Central  Google Scholar 

  • Jo MC, Liu W, Gu L, Dang W, Qin L (2015) High-throughput analysis of yeast replicative aging using a microfluidic system. Proc Natl Acad Sci USA 112:9364–9369

    CAS  PubMed  PubMed Central  Google Scholar 

  • Johnson SC, Rabinovitch PS, Kaeberlein M (2013) mTOR is a key modulator of ageing and age-related disease. Nature 493:338–345

    CAS  PubMed  PubMed Central  Google Scholar 

  • Jonikas MC, Collins SR, Denic V, Oh E, Quan EM, Schmid V et al (2009) Comprehensive characterization of genes required for protein folding in the endoplasmic reticulum. Science 323:1693–1697

    CAS  PubMed  PubMed Central  Google Scholar 

  • Kabani M, Kelley SS, Morrow MW, Montgomery DL, Sivendran R, Rose MD et al (2003) Dependence of endoplasmic reticulum-associated degradation on the peptide binding domain and concentration of BiP. Mol Biol Cell 14:3437–3448

    CAS  PubMed  PubMed Central  Google Scholar 

  • Kaeberlein M, Burtner CR, Kennedy BK (2007) Recent developments in yeast aging. PLoS Genet 3:e84

    PubMed  PubMed Central  Google Scholar 

  • Kapahi P, Zid BM, Harper T, Koslover D, Sapin V, Benzer S (2004) Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway. Curr Biol 14:885–890

    CAS  PubMed  PubMed Central  Google Scholar 

  • Kawazoe N, Kimata Y, Izawa S (2017) Acetic acid causes endoplasmic reticulum stress and induces the unfolded protein response in Saccharomyces cerevisiae. Front Microbiol 8:1192

    PubMed  PubMed Central  Google Scholar 

  • Kennedy BK, Smith ED, Kaeberlein M (2005) The enigmatic role of Sir2 in aging. Cell 123:548–550

    CAS  PubMed  Google Scholar 

  • Kimata Y, Kimata YI, Shimizu Y, Abe H, Farcasanu IC, Takeuchi M et al (2003) Genetic evidence for a role of BiP/Kar2 that regulates Ire1 in response to accumulation of unfolded proteins. Mol Biol Cell 14:2559–2569

    CAS  PubMed  PubMed Central  Google Scholar 

  • Kimata Y, Ishiwata-Kimata Y, Ito T, Hirata A, Suzuki T, Oikawa D et al (2007) Two regulatory steps of ER-stress sensor Ire1 involving its cluster formation and interaction with unfolded proteins. J Cell Biol 179:75–86

    CAS  PubMed  PubMed Central  Google Scholar 

  • Kirstein J, Morito D, Kakihana T, Sugihara M, Minnen A, Hipp MS et al (2015) Proteotoxic stress and ageing triggers the loss of redox homeostasis across cellular compartments. EMBO J 34:2334–2349

    CAS  PubMed  PubMed Central  Google Scholar 

  • Klaips CL, Jayaraj GG, Hartl FU (2018) Pathways of cellular proteostasis in aging and disease. J Cell Biol 217:51–63

    CAS  PubMed  PubMed Central  Google Scholar 

  • Kohno K, Normington K, Sambrook J, Gething MJ, Mori K (1993) The promoter region of the yeast KAR2 (BiP) gene contains a regulatory domain that responds to the presence of unfolded proteins in the endoplasmic reticulum. Mol Cell Biol 13:877–890

    CAS  PubMed  PubMed Central  Google Scholar 

  • Koike N, Hatano Y, Ushimaru T (2018) Heat shock transcriptional factor mediates mitochondrial unfolded protein response. Curr Genet 64:907–917

    CAS  PubMed  Google Scholar 

  • Kono N, Amin-Wetzel N, Ron D (2017) Generic membrane-spanning features endow IRE1α with responsiveness to membrane aberrancy. Mol Biol Cell 28:2318–2332

    CAS  PubMed  PubMed Central  Google Scholar 

  • Kozutsumi Y, Segal M, Normington K, Gething MJ, Sambrook J (1988) The presence of malfolded proteins in the endoplasmic reticulum signals the induction of glucose-regulated proteins. Nature 332:462–464

    CAS  PubMed  Google Scholar 

  • Labunskyy VM, Gerashchenko MV, Delaney JR, Kaya A, Kennedy BK, Kaeberlein M, Gladyshev VN (2014) Lifespan extension conferred by endoplasmic reticulum secretory pathway deficiency requires induction of the unfolded protein response. PLoS Genet 10:e1004019

    PubMed  PubMed Central  Google Scholar 

  • Lajoie P, Moir RD, Willis IM, Snapp EL (2012) Kar2p availability defines distinct forms of endoplasmic reticulum stress in living cells. Mol Biol Cell 23:955–964

    CAS  PubMed  PubMed Central  Google Scholar 

  • Laun P, Rinnerthaler M, Bogengruber E, Heeren G, Breitenbach M (2006) Yeast as a model for chronological and reproductive aging—a comparison. Exp Gerontol 41:1208–1212

    CAS  PubMed  Google Scholar 

  • Lin SJ, Defossez PA, Guarente L (2000) Requirement of NAD and SIR2 for life-span extension by calorie restriction in Saccharomyces cerevisiae. Science 289:2126–2128

    CAS  PubMed  Google Scholar 

  • Lin SS, Manchester JK, Gordon JI (2001) Enhanced gluconeogenesis and increased energy storage as hallmarks of aging in Saccharomyces cerevisiae. J Biol Chem 276:36000–36007

    CAS  PubMed  Google Scholar 

  • Liu Y, Chang A (2008) Heat shock response relieves ER stress. EMBO J 27:1049–1059

    CAS  PubMed  PubMed Central  Google Scholar 

  • Luis NM, Wang L, Ortega M, Deng H, Katewa SD, Li PW-L et al (2016) Intestinal IRE1 is required for increased triglyceride metabolism and longer lifespan under dietary restriction. Cell Rep 17:1207–1216

    CAS  PubMed  PubMed Central  Google Scholar 

  • Ma Y, Hendershot LM (2004) ER chaperone functions during normal and stress conditions. J Chem Neuroanat 28:51–65

    CAS  PubMed  Google Scholar 

  • Martínez G, Duran-Aniotz C, Cabral-Miranda F, Vivar JP, Hetz C (2017) Endoplasmic reticulum proteostasis impairment in aging. Aging Cell 16:615–623

    PubMed  PubMed Central  Google Scholar 

  • Matecic M, Smith DL, Pan X, Maqani N, Bekiranov S, Boeke JD, Smith JS (2010) A microarray-based genetic screen for yeast chronological aging factors. PLoS Genet 6:e1000921

    PubMed  PubMed Central  Google Scholar 

  • Mayer TU, Braun T, Jentsch S (1998) Role of the proteasome in membrane extraction of a short-lived ER-transmembrane protein. EMBO J 17:3251–3257

    CAS  PubMed  PubMed Central  Google Scholar 

  • Mercado G, Castillo V, Soto P, López N, Axten JM, Sardi SP et al (2018) Targeting PERK signaling with the small molecule GSK2606414 prevents neurodegeneration in a model of Parkinson’s disease. Neurobiol Dis 112:136–148

    CAS  PubMed  Google Scholar 

  • Merksamer PI, Trusina A, Papa FR (2008) Real-time redox measurements during endoplasmic reticulum stress reveal interlinked protein folding functions. Cell 135:933–947

    CAS  PubMed  PubMed Central  Google Scholar 

  • Mirisola MG, Longo VD (2012) Acetic acid and acidification accelerate chronological and replicative aging in yeast. Cell Cycle 11:3532–3533

    CAS  PubMed  PubMed Central  Google Scholar 

  • Moir RD, Gross DA, Silver DL, Willis IM (2012) SCS3 and YFT2 link transcription of phospholipid biosynthetic genes to ER stress and the UPR. PLoS Genet 8:e1002890

    CAS  PubMed  PubMed Central  Google Scholar 

  • Mori K, Sant A, Kohno K, Normington K, Gething MJ, Sambrook JF (1992) A 22 bp cis-acting element is necessary and sufficient for the induction of the yeast KAR2 (BiP) gene by unfolded proteins. EMBO J 11:2583–2593

    CAS  PubMed  PubMed Central  Google Scholar 

  • Mori K, Ma W, Gething MJ, Sambrook J (1993) A transmembrane protein with a cdc2 +/CDC28-related kinase activity is required for signaling from the ER to the nucleus. Cell 74:743–756

    CAS  PubMed  Google Scholar 

  • Murakami C, Kaeberlein M (2009) Quantifying yeast chronological life span by outgrowth of aged cells. J Vis Exp. https://doi.org/10.3791/1156

    Article  PubMed  PubMed Central  Google Scholar 

  • Naidoo N (2009) ER and aging-protein folding and the ER stress response. Ageing Res Rev 8:150–159

    CAS  PubMed  Google Scholar 

  • Naidoo N, Ferber M, Master M, Zhu Y, Pack AI (2008) Aging impairs the unfolded protein response to sleep deprivation and leads to proapoptotic signaling. J Neurosci 28:6539–6548

    CAS  PubMed  PubMed Central  Google Scholar 

  • Nikawa J, Yamashita S (1992) IRE1 encodes a putative protein kinase containing a membrane-spanning domain and is required for inositol phototrophy in Saccharomyces cerevisiae. Mol Microbiol 6:1441–1446

    CAS  PubMed  Google Scholar 

  • Nuss JE, Choksi KB, DeFord JH, Papaconstantinou J (2008) Decreased enzyme activities of chaperones PDI and BiP in aged mouse livers. Biochem Biophys Res Commun 365:355–361

    CAS  PubMed  Google Scholar 

  • Paz Gavilán M, Vela J, Castaño A, Ramos B, del Río JC, Vitorica J, Ruano D (2006) Cellular environment facilitates protein accumulation in aged rat hippocampus. Neurobiol Aging 27:973–982

    PubMed  Google Scholar 

  • Pincus D, Chevalier MW, Aragón T, van Anken E, Vidal SE, El-Samad H, Walter P (2010) BiP binding to the ER-stress sensor Ire1 tunes the homeostatic behavior of the unfolded protein response. PLoS Biol 8:e1000415

    PubMed  PubMed Central  Google Scholar 

  • Pineau L, Colas J, Dupont S, Beney L, Fleurat-Lessard P, Berjeaud J-M et al (2009) Lipid-induced ER stress: synergistic effects of sterols and saturated fatty acids. Traffic 10:673–690

    CAS  PubMed  Google Scholar 

  • Postnikoff SDL, Johnson JE, Tyler JK (2017) The integrated stress response in budding yeast lifespan extension. Microb Cell 4:368–375

    CAS  PubMed  PubMed Central  Google Scholar 

  • Promlek T, Ishiwata-Kimata Y, Shido M, Sakuramoto M, Kohno K, Kimata Y (2011) Membrane aberrancy and unfolded proteins activate the endoplasmic reticulum stress sensor Ire1 in different ways. Mol Biol Cell 22:3520–3532

    CAS  PubMed  PubMed Central  Google Scholar 

  • Ron D, Walter P (2007) Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol 8:519–529

    CAS  PubMed  Google Scholar 

  • Rubenstein EM, Kreft SG, Greenblatt W, Swanson R, Hochstrasser M (2012) Aberrant substrate engagement of the ER translocon triggers degradation by the Hrd1 ubiquitin ligase. J Cell Biol 197:761–773

    CAS  PubMed  PubMed Central  Google Scholar 

  • Ruegsegger C, Saxena S (2016) Proteostasis impairment in ALS. Brain Res 1648:571–579

    CAS  PubMed  Google Scholar 

  • Ruetenik AL, Ocampo A, Ruan K, Zhu Y, Li C, Zhai RG, Barrientos A (2016) Attenuation of polyglutamine-induced toxicity by enhancement of mitochondrial OXPHOS in yeast and fly models of aging. Microb Cell 3:338–351

    CAS  PubMed  PubMed Central  Google Scholar 

  • Salminen A, Kaarniranta K (2010) ER stress and hormetic regulation of the aging process. Ageing Res Rev 9:211–217

    CAS  PubMed  Google Scholar 

  • Scheper W, Hoozemans JJM (2015) The unfolded protein response in neurodegenerative diseases: a neuropathological perspective. Acta Neuropathol 130:315–331

    CAS  PubMed  PubMed Central  Google Scholar 

  • Schieke SM, Finkel T (2007) TOR and aging: less is more. Cell Metab 5:233–235

    CAS  PubMed  Google Scholar 

  • Schindelin J, Rueden CT, Hiner MC, Eliceiri KW (2015) The ImageJ ecosystem: an open platform for biomedical image analysis. Mol Reprod Dev 82:518–529

    CAS  PubMed  PubMed Central  Google Scholar 

  • Schuck S, Prinz WA, Thorn KS, Voss C, Walter P (2009) Membrane expansion alleviates endoplasmic reticulum stress independently of the unfolded protein response. J Cell Biol 187:525–536

    CAS  PubMed  PubMed Central  Google Scholar 

  • Sekiya M, Maruko-Otake A, Hearn S, Sakakibara Y, Fujisaki N, Suzuki E et al (2017) EDEM function in ERAD protects against chronic ER proteinopathy and age-related physiological decline in Drosophila. Dev Cell 41:652–664.e5

    CAS  PubMed  PubMed Central  Google Scholar 

  • Shen Z-J, Postnikoff S, Tyler JK (2019) Is Gcn4-induced autophagy the ultimate downstream mechanism by which hormesis extends yeast replicative lifespan? Curr Genet 65:717–720

    CAS  PubMed  PubMed Central  Google Scholar 

  • Shiu RP, Pouyssegur J, Pastan I (1977) Glucose depletion accounts for the induction of two transformation-sensitive membrane proteins in Rous sarcoma virus-transformed chick embryo fibroblasts. Proc Natl Acad Sci USA 74:3840–3844

    CAS  PubMed  PubMed Central  Google Scholar 

  • Singh P, Li R (2018) Emerging roles for sphingolipids in cellular aging. Curr Genet 64:761–767

    CAS  PubMed  Google Scholar 

  • Smith DL, McClure JM, Matecic M, Smith JS (2007) Calorie restriction extends the chronological lifespan of Saccharomyces cerevisiae independently of the sirtuins. Aging Cell 6:649–662

    CAS  PubMed  Google Scholar 

  • Steffen KK, Kennedy BK, Kaeberlein M (2009) Measuring replicative life span in the budding yeast. J Vis Exp. https://doi.org/10.3791/1209

    Article  PubMed  PubMed Central  Google Scholar 

  • Taylor RC, Dillin A (2013) XBP-1 is a cell-nonautonomous regulator of stress resistance and longevity. Cell 153:1435–1447

    CAS  PubMed  PubMed Central  Google Scholar 

  • Travers KJ, Patil CK, Wodicka L, Lockhart DJ, Weissman JS, Walter P (2000) Functional and genomic analyses reveal an essential coordination between the unfolded protein response and ER-associated degradation. Cell 101:249–258

    CAS  PubMed  Google Scholar 

  • Tsuchiya M, Dang N, Kerr EO, Hu D, Steffen KK, Oakes JA et al (2006) Sirtuin-independent effects of nicotinamide on lifespan extension from calorie restriction in yeast. Aging Cell 5:505–514

    CAS  PubMed  Google Scholar 

  • van der Vlies D, Woudenberg J, Post JA (2003) Protein oxidation in aging: endoplasmic reticulum as a target. Amino Acids 25:397–407

    PubMed  Google Scholar 

  • Vidal RL, Hetz C (2012) Crosstalk between the UPR and autophagy pathway contributes to handling cellular stress in neurodegenerative disease. Autophagy 8:970–972

    CAS  PubMed  PubMed Central  Google Scholar 

  • Vidal RL, Figueroa A, Court FA, Thielen P, Molina C, Wirth C et al (2012) Targeting the UPR transcription factor XBP1 protects against Huntington’s disease through the regulation of FoxO1 and autophagy. Hum Mol Genet 21:2245–2262

    CAS  PubMed  PubMed Central  Google Scholar 

  • Viswanathan M, Kim SK, Berdichevsky A, Guarente L (2005) A role for SIR-2.1 regulation of ER stress response genes in determining C. elegans life span. Dev Cell 9:605–615

    CAS  PubMed  Google Scholar 

  • Volmer R, van der Ploeg K, Ron D (2013) Membrane lipid saturation activates endoplasmic reticulum unfolded protein response transducers through their transmembrane domains. Proc Natl Acad Sci USA 110:4628–4633

    CAS  PubMed  PubMed Central  Google Scholar 

  • Walter P, Ron D (2011) The unfolded protein response: from stress pathway to homeostatic regulation. Science 334:1081–1086

    CAS  PubMed  Google Scholar 

  • Wang L, Zeng X, Ryoo HD, Jasper H (2014) Integration of UPRER and oxidative stress signaling in the control of intestinal stem cell proliferation. PLoS Genet 10:e1004568

    PubMed  PubMed Central  Google Scholar 

  • Wang L, Ryoo HD, Qi Y, Jasper H (2015) PERK limits drosophila lifespan by promoting intestinal stem cell proliferation in response to ER stress. PLoS Genet 11:e1005220

    PubMed  PubMed Central  Google Scholar 

  • Wasko BM, Carr DT, Tung H, Doan H, Schurman N, Neault JR et al (2013) Buffering the pH of the culture medium does not extend yeast replicative lifespan. [version 1; peer review: 2 approved]. F1000Res 2:216

    PubMed  PubMed Central  Google Scholar 

  • Weindling E, Bar-Nun S (2015) Sir2 links the unfolded protein response and the heat shock response in a stress response network. Biochem Biophys Res Commun 457:473–478

    CAS  PubMed  Google Scholar 

  • Williams AJ, Paulson HL (2008) Polyglutamine neurodegeneration: protein misfolding revisited. Trends Neurosci 31:521–528

    CAS  PubMed  PubMed Central  Google Scholar 

  • Yang S, Huang S, Gaertig MA, Li X-J, Li S (2014) Age-dependent decrease in chaperone activity impairs MANF expression, leading to Purkinje cell degeneration in inducible SCA17 mice. Neuron 81:349–365

    CAS  PubMed  PubMed Central  Google Scholar 

  • Yoshida H, Matsui T, Hosokawa N, Kaufman RJ, Nagata K, Mori K (2003) A time-dependent phase shift in the mammalian unfolded protein response. Dev Cell 4:265–271

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Peter Walter (UCSF) and Davis Ng (National University of Singapore) for yeast strains and plasmids. SRC, PEZ, ENF, and JG generated the data and analyzed the results with PL. SRC, MLD, and PL wrote the manuscript. All authors read and approved the final version of the manuscript.

Funding

This study is supported by an operating grant from the Canadian Institutes for Health Research (CIHR) to MLD and PL (MOP 137041). PL is the recipient of a John R. Evans Leaders Fund award (#35183) with matching fund from the Ontario Research Fund. SRC was supported by an Ontario Graduate Scholarship and now holds an Alexander Graham Bell Canada Graduate Scholarship from the Natural Sciences and Engineering Research Council of Canada (NSERC).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Patrick Lajoie.

Additional information

Communicated by M. Kupiec.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

294_2019_1019_MOESM1_ESM.pdf

A) Wild-type and ire1Δ cells were grown in SC media for 8h (day 0), 1 day, and 2 days. Cells were stained with propidium iodide and imaged with a UV trans-illuminator to view fluorescence. Boiled cells were used as a positive control, unstained cells were used as a negative control (n=3). B) Wild-type and ire1Δ cells were aged in 2% SC glucose media for indicated time periods. Cells were stained with propidium iodide and fluorescence data was analyzed to generate survival curves and survival integrals with day 3 set to 100% survival (as opposed to day 1) and statistical analysis was performed using t-tests (n=5, P = 0.0004). (PDF 29 kb)

294_2019_1019_MOESM2_ESM.pdf

A) Wild-type and ire1Δ cells were grown for either 8 hours (Day 0) or overnight (Day 1) in SC media then spotted in serial fivefold dilutions on SC glucose plates. B) Cells expressing the fluorescent protein UPR-mCherry were grown overnight in the indicated media, and then analyzed by fluorescent microscopy. Relative fluorescence from individual cells was quantified and plotted, and statistically analyzed using t-tests (n=122-145, P < 0.0001). ire1Δ+UPR-mCherry is used as a negative control. C) Wild-type and ire1Δ cells were aged for 1-3 days, then RNA was isolated, reverse transcribed to cDNA, then INO1 expression was quantified using Real-Time qPCR. (PDF 168 kb)

294_2019_1019_MOESM3_ESM.pdf

C) Aged wild-type and ire1Δ cells were spotted on YPD or YPG (2% glycerol) plates to assess the effect of switching the cells from fermentation to mitochondrial respiration. (PDF 43 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chadwick, S.R., Fazio, E.N., Etedali-Zadeh, P. et al. A functional unfolded protein response is required for chronological aging in Saccharomyces cerevisiae. Curr Genet 66, 263–277 (2020). https://doi.org/10.1007/s00294-019-01019-0

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00294-019-01019-0

Keywords

Navigation