Heat shock proteins and Drosophila aging

https://doi.org/10.1016/j.exger.2010.09.002Get rights and content

Abstract

Since their discovery in Drosophila, the heat shock proteins (Hsps) have been shown to regulate both stress resistance and life-span. Aging is characterized by increased oxidative stress and the accumulation of abnormal (malfolded) proteins, and these stresses induce Hsp gene expression through the transcription factor HSF. In addition, a subset of Hsps is induced by oxidative stress through the JNK signaling pathway and the transcription factor Foxo. The Hsps counteract the toxicity of abnormal proteins by facilitating protein refolding and turnover, and through other mechanisms including inhibition of apoptosis. The Hsps are up-regulated in tissue-specific patterns during aging, and their expression correlates with, and sometimes predicts, life span, making them ideal biomarkers of aging. The tools available for experimentally manipulating gene function and assaying healthspan in Drosophila provides an unparalleled opportunity to further study the role of Hsps in aging.

Research Highlights

►Review of the expression and function of hsps in response to stress and aging. ►The role of the HSF pathway and the JNK/Foxo pathway. ►Antagonistic pleiotropy, sexual antagonistic pleiotropy, p53 and foxo. ►Protein turnover, proteotoxicity, and the role of hsps in aging. ►Hsps as predictive biomarkers of aging and life span.

Introduction

The laboratory fruit-fly, Drosophila melanogaster, has been a leading model for the study of aging for almost a century because of its ease of culture, short life cycle, and highly developed genetics and molecular biology (Partridge and Tower, 2008). Moreover, the heat shock response and the Hsp genes were first discovered in Drosophila, as revealed by the “puffing” pattern of polytene chromosomes in response to heat and oxidative stress (Ritossa, 1962, Ritossa, 1996). Since then the Drosophila model has been key to understanding the regulation of the heat shock response, the function of the Hsps, and the intimate connection between the Hsps and the aging process.

At a genetic level, aging in Drosophila and other organisms is thought to result from antagonistic pleiotropy of gene function between developmental stages and the sexes (Hughes and Reynolds, 2005, Magwire et al., 2010, Tower, 2006, Tower and Arbeitman, 2009). The force of natural selection declines with age, allowing for the accumulation of inherited alleles with late-acting deleterious effects (“mutation accumulation”). These same aging-promoting alleles may be maintained by positive selection if they provide a benefit earlier in life (“antagonistic plieotropy”). Because the sexes are subject to different and sometimes opposing selective pressures, inherited alleles may accumulate that are relatively beneficial to one sex but relatively deleterious to the other sex, or detrimental to both sexes (“sexual antagonistic plieotropy”). Antagonistic pleiotropy is one possible explanation for why pathways that promote growth and differentiation, such as the insulin/IGF1-like signaling (IIS) pathway, the target-of-rapamycin (TOR) pathway, and the sex-determination pathway (X/Y) are such important modulators of aging, as differentiation should be epistatic to the antagonist–pleiotropic activities of many genes (Fig. 1). For example, these growth and differentiation-promoting pathways may act by up-regulating expression of life-span shortening genes, and down-regulating expression of longevity-promoting genes (such as Hsps), as was observed for IIS in C. elegans (Ayyadevara et al., 2008, Curran et al., 2009, Kenyon, 2010, Murphy et al., 2003). One of the first genes suspected of exhibiting antagonistic pleiotropy between developmental stages was mammalian p53, based upon its beneficial function in young animals to prevent cancer, but a possible detrimental effect in old animals by promoting counter-productive cell senescence or apoptosis (Campisi, 2003). Recently Drosophila p53 has been shown to exhibit sexual antagonistic pleiotropy, as indicated by opposing effects of p53 mutations and transgenes on life span in males versus females (Shen and Tower, 2010, Waskar et al., 2009). Interestingly the sexual-dimorphism of p53 transgene effects on life span were found to be regulated by the IIS target transcription factor Foxo, consistent with a role for IIS and Foxo in promoting sexual antagonistic pleiotropy (Shen and Tower, 2010). The activity of p53 is regulated by Hsps, making p53 one likely component of the mechanism(s) by which Hsps can affect aging and life span.

Mechanistically, aging appears to involve an imbalance between damage and repair of macromolecules, including DNA damage and mutations, loss of epigenetic regulation, and telomere erosion (Vijg, 2008), as well as the response of the animal and its cells to such damage (Campisi and Vijg, 2009). Proteins are particularly subject to aging-related damages, including cleavage, covalent modifications, oxidative lesions, glycation, crosslinking, and denaturation (Semba et al., 2010, Stadtman, 2006). The correct synthesis, folding, and turnover of proteins is one of the most energetically costly functions of the cell, and proteotoxicity is a key component of aging and aging-related diseases (Morimoto and Cuervo, 2009). Mitochondrial malfunction is increasingly implicated in the aging process, and abnormal mitochondria have been found to accumulate with age in several Drosophila tissues (Calleja et al., 1993, Fleming et al., 1985, Schwarze et al., 1998, Sohal, 1975, Walker and Benzer, 2004). The resultant oxidative stress and reduction in ATP synthesis may play a central role in the creation and accumulation of abnormal proteins during aging (Bueler, 2009, Linford and Pletcher, 2009, Marzetti et al., 2009, Morrow and Tanguay, 2008).

The Hsps are defined by their ability to bind to denatured proteins, their ability to alter the folded structure of other proteins, and by the induction of their expression in response to stresses that cause protein denaturation, such as heat and oxidative stress (Morimoto, 2008, Morrow et al., 2006). By mediating either protein refolding or degradation, the Hsps counteract proteotoxicity and favor stress resistance. These functions of the Hsps may underlie their ability to favor life span and counteract aging-related malfunction in several systems (Lithgow and Walker, 2002, Morrow and Tanguay, 2003). In addition to their role in response to global protein damage, the Hsps also have more specific targets, and are involved in regulating pathways that are central to aging phenotypes such as protein turnover, cellular senescence, cancer, and apoptosis/programmed cell death (Morimoto and Cuervo, 2009, Tower, 2009), and the specific modulation of such pathways may also underlie the effects of Hsps on aging and life span. Finally, the expression of Hsps correlates with, and sometimes predicts, life span, making Hsps among the best-known biomarkers of aging in Drosophila and other animals (Johnson, 2006, Tower, 2009).

Section snippets

Hsp classes and functions

The heat shock proteins are molecular chaperones — a class of proteins characterized by their ability to modulate the structure and folding of other proteins (client proteins)(Kim et al., 2007). The expression of the Hsps is induced through the HSF (heat shock transcription factor) pathway, by stresses that cause protein denaturation (unfolding), such as heat stress, oxidative stress and other stresses (Fig. 2). Activated HSF binds to heat shock response elements (HSEs) in the promoters of the

Hsps and protein turnover

The Hsps can facilitate the refolding of denatured client proteins, and/or facilitate their entry into several degradation pathways, including the lysosome, autophagic vesicles, and the ubiquitin/proteosome pathway. By facilitating the clearance of damaged proteins, the Hsps are key components of the cells response to proteotoxicity, and can confer upon cells increased resistance to heat and other stresses.

Chaperone-mediated autophagy is a process where specific cytosolic proteins are targeted

Hsps as modulators of stress resistance and proteotoxicity

Hsps have been demonstrated to be key players in conferring resistance to heat and other stresses in Drosophila (Morrow and Tanguay, 2003, Vermeulen and Loeschcke, 2007). For example, the mutation of hsp22 decreases survival upon heat stress (Morrow et al., 2004a), as does mutation of all six copies of the hsp70 gene (Gong and Golic, 2006). Mutation of hsp83 sensitizes flies to the toxic effects of sleep deprivation (Shaw et al., 2002). Over-expression of certain Hsps can increase resistance to

Regulation of Hsp expression in response to proteotoxic stress and aging

Denaturation of proteins and the resultant exposure of hydrophobic residues is the main mechanism for the induction of Hsp genes by the transcription factor HSF (Fig. 2a). HSF is consitutively expressed, and normally resides in the cytoplasm in a complex with Hsps that hold HSF in an inactive state (Morimoto, 2002). When heat, ROS or other stress causes protein denaturation, this exposes hydrophobic residues that bind Hsps and titrate them away from HSF. This allows HSF to assume an active

Regulation of Hsp expression in response to ROS and aging, a role for JNK and Foxo

A subset of Hsp genes are regulated by the transcription factor Foxo in both Drosophila and C. elegans. Foxo is particularly relevant to aging because in C. elegans the Foxo homolog Daf16, along with HSF, has been shown to be required for the life span extension caused by reductions in IIS (Kenyon, 2010). Notably, RNAi inhibition of the expression of sHsp genes has been shown to block part of this life span extension, suggesting that induction of sHsps by Foxo and HSF is a part of the mechanism

Hsps as modulators of life span

Laboratory selection of Drosophila strains for increased life span produced flies with increased expression of sHsp genes as young adults, suggesting that increased sHsp expression might favor longevity (Kurapati et al., 2000). Consistent with this idea, mutation of hsp70 or hsp22 can reduce adult fly survival, and chemicals that increased life span, such as HDAC inhibitors, produced correlated increases in hsp70 and sHsp gene expression (Kang et al., 2002, Zhao et al., 2005). Direct testing of

Hsps are predictive biomarkers of life span

Drosophila undergo a number of functional declines that correlate with increased age and increased mortality rate, including decreased spontaneous movement, decreased climbing speed, decreased memory, decreased heart function, and decreased reproductive capacity (Grotewiel et al., 2005, Iliadi and Boulianne, 2010, Piazza et al., 2009, Reenan and Rogina, 2008). Correlated molecular changes include decreases in protein turnover system activities (ubiquitin/proteosome and autophagy/lysosomal),

Summary

Altered expression of Hsps was among the first molecular changes characterized during aging in Drosophila. Since then the Hsps have been shown to directly regulate aging phenotypes, including life span, stress resistance and function. The tractability of the Drosophila system promises to allow continued discovery of the role of Hsps in aging, and facilitate our understanding of human aging and disease.

Acknowledgements

The author was supported by a grant from the Department of Health and Human Services (AG011833) and by Scientific Opportunity Funds from the Genetics of Longevity Consortium (U19 AG032122).

References (160)

  • C.A. Dickey et al.

    Brain CHIP: removing the culprits in neurodegenerative disease

    Trends Mol. Med.

    (2007)
  • D.A. Drummond et al.

    Mistranslation-induced protein misfolding as a dominant constraint on coding-sequence evolution

    Cell

    (2008)
  • M.S. Grotewiel et al.

    Functional senescence in Drosophila melanogaster

    Ageing Res. Rev.

    (2005)
  • T.E. Johnson

    Recent results: biomarkers of aging

    Exp. Gerontol.

    (2006)
  • H.J. Kim et al.

    Heat shock responses for understanding diseases of protein denaturation

    Mol. Cells

    (2007)
  • H.J. Kim et al.

    Gene expression profiling implicates OXPHOS complexes in lifespan extension of flies over-expressing a small mitochondrial chaperone, Hsp22

    Exp. Gerontol.

    (2010)
  • V. King et al.

    Aging-specific expression of Drosophila hsp22

    Dev. Biol.

    (1999)
  • U. Kurzik-Dumke et al.

    Sequence of the new Drosophila melanogaster small heat-shock-related gene, lethal(2) essential for life [l(2)efl], at locus 59F4, 5

    Gene

    (1995)
  • P.C. Liao et al.

    The effect of neuronal expression of heat shock proteins 26 and 27 on lifespan, neurodegeneration, and apoptosis in Drosophila

    Biochem. Biophys. Res. Commun.

    (2008)
  • N.J. Linford et al.

    Aging: fruit flies break the chain to a longer life

    Curr. Biol.

    (2009)
  • G.J. Lithgow et al.

    Stress resistance as a determinate of C. elegans lifespan

    Mech. Ageing Dev.

    (2002)
  • H.R. Massie et al.

    Iron accumulation during development and ageing of Drosophila

    Mech. Ageing Dev.

    (1985)
  • P. Mehlen et al.

    Small stress proteins as novel regulators of apoptosis. Heat shock protein 27 blocks Fas/APO-1- and staurosporine-induced cell death

    J. Biol. Chem.

    (1996)
  • N. Minois et al.

    Locomotor activity as a function of age and life span in Drosophila melanogaster overexpressing hsp70

    Exp. Gerontol.

    (2001)
  • R.I. Morimoto

    Dynamic remodeling of transcription complexes by molecular chaperones

    Cell

    (2002)
  • G. Morrow et al.

    Heat shock proteins and aging in Drosophila melanogaster

    Semin. Cell Dev. Biol.

    (2003)
  • G. Morrow et al.

    The small heat shock protein Hsp22 of Drosophila melanogaster is a mitochondrial protein displaying oligomeric organization

    J. Biol. Chem.

    (2000)
  • G. Morrow et al.

    Decreased lifespan in the absence of expression of the mitochondrial small heat shock protein Hsp22 in Drosophila

    J. Biol. Chem.

    (2004)
  • L.D. Mueller et al.

    Predicting death in female Drosophila

    Exp. Gerontol.

    (2009)
  • S.N. Agoff et al.

    Regulation of the human hsp70 promoter by p53

    Science

    (1993)
  • S. Ayyadevara et al.

    Remarkable longevity and stress resistance of nematode PI3K-null mutants

    Aging Cell

    (2008)
  • J.F. Beaulieu et al.

    Interaction of Drosophila 27,000 Mr heat-shock protein with the nucleus of heat-shocked and ecdysone-stimulated culture cells

    J. Cell Sci.

    (1989)
  • J. Bilen et al.

    Genome-wide screen for modifiers of ataxin-3 neurodegeneration in Drosophila

    PLoS Genet.

    (2007)
  • M.V. Blagosklonny et al.

    Mutant conformation of p53 translated in vitro or in vivo requires functional HSP90

    Proc. Natl. Acad. Sci. USA

    (1996)
  • J.M. Burger et al.

    Sex-specific effects of interventions that extend fly life span

    Sci. Aging Knowledge Environ.

    (2004)
  • J. Campisi

    Cancer and ageing: rival demons?

    Nat. Rev. Cancer

    (2003)
  • J. Campisi et al.

    Does damage to DNA and other macromolecules play a role in aging? If so, how?

    J. Gerontol. A Biol. Sci. Med. Sci.

    (2009)
  • H.C. Chang et al.

    SnapShot: molecular chaperones, Part I

    Cell

    (2007)
  • C.G. Concannon et al.

    On the role of Hsp27 in regulating apoptosis

    Apoptosis

    (2003)
  • S.P. Curran et al.

    A soma-to-germline transformation in long-lived Caenorhabditis elegans mutants

    Nature

    (2009)
  • C. Curtis et al.

    Transcriptional profiling of MnSOD-mediated lifespan extension in Drosophila reveals a species-general network of aging and metabolic genes

    Genome Biol.

    (2007)
  • D.A. Drummond et al.

    The evolutionary consequences of erroneous protein synthesis

    Nat. Rev. Genet.

    (2009)
  • J.H. Feder et al.

    The consequences of expressing hsp70 in Drosophila cells at normal temperatures

    Genes Dev.

    (1992)
  • J.E. Fleming et al.

    Age dependent changes in mitochondria

    Basic Life Sci.

    (1985)
  • J.E. Fleming et al.

    Aging results in an unusual expression of Drosophila heat shock proteins

    Proc. Natl. Acad. Sci. USA

    (1988)
  • N.J. Fuda et al.

    Defining mechanisms that regulate RNA polymerase II transcription in vivo

    Nature

    (2009)
  • R. Garesse et al.

    A Drosophila model of mitochondrial DNA replication: proteins, genes and regulation

    IUBMB Life

    (2005)
  • C. Garrido et al.

    Heat shock proteins 27 and 70: anti-apoptotic proteins with tumorigenic properties

    Cell Cycle

    (2006)
  • H. Gershon et al.

    Detection of inactive enzyme molecules in aging organisms

    Nature

    (1970)
  • S. Ghosh et al.

    Comparison of pathways controlling toxicity in the eye and brain in Drosophila models of human neurodegenerative diseases

    Hum. Mol. Genet.

    (2004)
  • Cited by (0)

    View full text