3025_ftp

19
INVITED REVIEW Journal of Pathology J Pathol 2012; 226: 255–273 Published online in Wiley Online Library (wileyonlinelibrary.com) DOI: 10.1002/path.3025 Autophagy and disease: always two sides to a problem Sunandini Sridhar, 1 Yair Botbol, 2 Fernando Macian 2,3 and Ana Maria Cuervo 1,3 * 1 Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, USA 2 Department of Pathology, Albert Einstein College of Medicine, New York, USA 3 Institute for Aging Studies, Albert Einstein College of Medicine, New York, USA *Correspondence to: Ana Maria Cuervo, Department of Developmental and Molecular Biology, Institute for Aging Research, Albert Einstein College of Medicine, Chanin Building, Room 504, 1300 Morris Park Avenue, Bronx, NY 10461, USA. e-mail: [email protected] Abstract Autophagy is a process traditionally known to contribute to cellular cleaning through the removal of intracellular components in lysosomes. In recent years, intensive scrutiny at the molecular level to which autophagy has been subjected has also contributed to expanding our understanding of the physiological role of this pathway. Added to the well-characterized role in quality control, autophagy has proved to be important in the maintenance of cellular homeostasis and of the energetic balance, in cellular and tissue remodelling, and cellular defence against extracellular insults and pathogens. It is not a surprise that, in light of this growing number of physiological functions, connections between autophagic malfunction and human pathologies have also been strengthened. In this review, we focus on several pathological conditions associated with primary or secondary defects in autophagy and comment on a recurring theme for many of them, ie the fact that autophagy can often exert both beneficial and aggravating effects on the progression of disease. Elucidating the factors that determine the switch between these dual functions of autophagy in disease has become a priority when considering the potential therapeutic implications of the pharmacological modulation of autophagy in many of these pathological conditions. Copyright 2011 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd. Keywords: cancer; cardiomyopathy; immunity; lysosomes; neurodegeneration; protein aggregation; proteolysis; proteotoxicity; T cell function Received 6 August 2011; Revised 25 September 2011; Accepted 3 October 2011 No conflicts of interest were declared. Introduction to autophagy The Greek term ‘autophagy’ (‘self-eating’) was coined almost half a century ago by Christian DeDuve [1]. Today, this intensely investigated pathway has come to be recognized as an evolutionarily conserved intra- cellular process through which cytosolic components, ranging from proteins, lipids, sugars and nucleotides to whole organelles and invading pathogens, are targeted for lysosomal degradation [2,3]. Autophagy contributes to both the removal of damaged long-lived proteins and organelles and the normal turnover of these intracellu- lar components as part of its role in cellular quality control. In addition, autophagy also serves as a cellu- lar adaptive response to compromised conditions, such as metabolic stress, when degradation of intracellular materials through this pathway becomes an alternative source of energy [4]. In recent years, a growing number of functions have been added to these two basic cellular functions of the autophagic process—quality control and maintenance of the cellular energetic balance—helping to shape the physiological relevance of this pathway. Furthermore, a better understanding of the molecular mechanisms that mediate the autophagic process has helped to establish connections between autophagy and the pathogenesis of different disorders and of ageing [3,5,6]. Findings in recent years also support the existence of more than one type of autophagy and the fact that different autophagic pathways co-exist in most cells. In this review, we briefly describe the main characteristics and molecular components of the different autophagic variants and then provide a general view of the multiple physiological functions of this cellular process. In the second part, we comment on specific autophagy-related pathologies, which either result from a primary defect in the autophagic process, or for which changes in autophagy have been described to exert a modulatory effect on the course of the disease. The molecular dissection of the different autophagic pathways To date, three basic forms of autophagy have been described, namely, macroautophagy, microautophagy and chaperone-mediated autophagy (CMA), which pri- marily differ in the way in which cytosolic components (cargo) are delivered to lysosomes [2,7,8] (Figure 1). Macroautophagy and microautophagy were both first Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273 Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

Transcript of 3025_ftp

INVITED REVIEWJournal of PathologyJ Pathol 2012; 226: 255–273Published online in Wiley Online Library(wileyonlinelibrary.com) DOI: 10.1002/path.3025

Autophagy and disease: always two sides to a problemSunandini Sridhar,1 Yair Botbol,2 Fernando Macian2,3 and Ana Maria Cuervo1,3*

1 Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, USA2 Department of Pathology, Albert Einstein College of Medicine, New York, USA3 Institute for Aging Studies, Albert Einstein College of Medicine, New York, USA

*Correspondence to: Ana Maria Cuervo, Department of Developmental and Molecular Biology, Institute for Aging Research, Albert EinsteinCollege of Medicine, Chanin Building, Room 504, 1300 Morris Park Avenue, Bronx, NY 10461, USA. e-mail: [email protected]

AbstractAutophagy is a process traditionally known to contribute to cellular cleaning through the removal of intracellularcomponents in lysosomes. In recent years, intensive scrutiny at the molecular level to which autophagy has beensubjected has also contributed to expanding our understanding of the physiological role of this pathway. Addedto the well-characterized role in quality control, autophagy has proved to be important in the maintenance ofcellular homeostasis and of the energetic balance, in cellular and tissue remodelling, and cellular defence againstextracellular insults and pathogens. It is not a surprise that, in light of this growing number of physiologicalfunctions, connections between autophagic malfunction and human pathologies have also been strengthened. Inthis review, we focus on several pathological conditions associated with primary or secondary defects in autophagyand comment on a recurring theme for many of them, ie the fact that autophagy can often exert both beneficialand aggravating effects on the progression of disease. Elucidating the factors that determine the switch betweenthese dual functions of autophagy in disease has become a priority when considering the potential therapeuticimplications of the pharmacological modulation of autophagy in many of these pathological conditions.Copyright 2011 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.

Keywords: cancer; cardiomyopathy; immunity; lysosomes; neurodegeneration; protein aggregation; proteolysis; proteotoxicity; T cellfunction

Received 6 August 2011; Revised 25 September 2011; Accepted 3 October 2011

No conflicts of interest were declared.

Introduction to autophagy

The Greek term ‘autophagy’ (‘self-eating’) was coinedalmost half a century ago by Christian DeDuve [1].Today, this intensely investigated pathway has cometo be recognized as an evolutionarily conserved intra-cellular process through which cytosolic components,ranging from proteins, lipids, sugars and nucleotides towhole organelles and invading pathogens, are targetedfor lysosomal degradation [2,3]. Autophagy contributesto both the removal of damaged long-lived proteins andorganelles and the normal turnover of these intracellu-lar components as part of its role in cellular qualitycontrol. In addition, autophagy also serves as a cellu-lar adaptive response to compromised conditions, suchas metabolic stress, when degradation of intracellularmaterials through this pathway becomes an alternativesource of energy [4].

In recent years, a growing number of functions havebeen added to these two basic cellular functions of theautophagic process—quality control and maintenanceof the cellular energetic balance—helping to shape thephysiological relevance of this pathway. Furthermore, abetter understanding of the molecular mechanisms thatmediate the autophagic process has helped to establish

connections between autophagy and the pathogenesisof different disorders and of ageing [3,5,6]. Findingsin recent years also support the existence of morethan one type of autophagy and the fact that differentautophagic pathways co-exist in most cells. In thisreview, we briefly describe the main characteristicsand molecular components of the different autophagicvariants and then provide a general view of the multiplephysiological functions of this cellular process. In thesecond part, we comment on specific autophagy-relatedpathologies, which either result from a primary defectin the autophagic process, or for which changes inautophagy have been described to exert a modulatoryeffect on the course of the disease.

The molecular dissection of the differentautophagic pathways

To date, three basic forms of autophagy have beendescribed, namely, macroautophagy, microautophagyand chaperone-mediated autophagy (CMA), which pri-marily differ in the way in which cytosolic components(cargo) are delivered to lysosomes [2,7,8] (Figure 1).Macroautophagy and microautophagy were both first

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

256 S Sridhar et al

Figure 1. Autophagic pathways. Three different general mechanisms of lysosomal delivery of cargo set the basis for the different typesof autophagy described in mammalian cells. (A) Macroautophagy : different extra- and intracellular signals activate the recruitment ofthe macroautophagy initiation complex to the sites of autophagosome formation. Shuttling of proteins and lipids to these regions andposttranslational modifications of the lipids initiate the formation of a limiting membrane, which grows through the assembly of proteinsconjugated to proteins or lipids while it sequesters components of the cytosol. Once the membrane seals to form the autophagosome,this double membrane vesicle is delivered to lysosomes where, upon membrane fusion, lysosomal hydrolases gain access to cargo.(B) Microautophagy : through stimuli that remain poorly identified, cytosolic soluble proteins and organelles are directly sequestered byinvaginations in the boundary membrane of lysosomes and late endosomes. Cargo internalized in the small luminal vesicles is degradedafter the vesicles pinch off from the limiting membrane. Although most microautophagy probably occurs in bulk, selective targeting byhsc70 of cytosolic proteins to microvesicles as they form in late endosomes has also been described. (C) Chaperone-mediated autophagy(CMA) is induced by stimuli such as prolonged starvation, oxidative stress and other conditions resulting in protein damage, but thesignalling mechanism activated by these stimuli remains unknown. When CMA is activated, selective cytosolic proteins bearing a targetingmotif are recruited by hsc70 and co-chaperones to the surface of lysosomes. Upon binding to the receptor protein LAMP-2A, substratescross the membrane through the LAMP-2A-dependent translocation complex and are then rapidly degraded in the lumen.

described as mechanisms for ‘in-bulk’ sequestra-tion of cytoplasmic components (including entireorganelles) into vesicular compartments. In macroau-tophagy, these vesicles form de novo from a limit-ing membrane of non-lysosomal origin that seals uponitself to form free-standing double membrane carriersknown as autophagosomes [9,10]. Fusion of lysosomeswith the limiting membrane of the autophagosomegrants lysosomal hydrolases access to the sequesteredcargo, assuring its complete degradation. In the caseof microautophagy, the engulfment of cargo occursthrough deformation of the lysosomal membrane itself,to form vesicles that invaginate towards the lysoso-mal lumen [11,12]. Pinch-off of these vesicles fromthe lysosomal membrane into the lumen, and the sub-sequent degradation of their limiting membrane by thelysosomal hydrolases, precede cargo degradation.

Both macroautophagy and microautophagy were ini-tially described in mammalian cells, but their molec-ular characterization has been mostly carried out inyeast, taking advantage of the genetic analysis that canbe performed in this model organism. These geneticscreenings have identified that more than 30 differ-ent genes (known as ATG or autophagy-related genes)

participate in the execution and regulation of macroau-tophagy [2,13] (Figure 1). The protein products ofthese genes (Atg or autophagy-related proteins) orga-nize into functional complexes that mediate the fol-lowing steps in the autophagic process: nucleationof the limiting membrane, elongation of the mem-brane, sealing to form a vesicle and fusion of thedouble membrane vesicle with lysosomes. Nucleationis attained through post-translational modifications ofpre-existing lipids in the membranes of different intra-cellular organelles [endoplasmic reticulum (ER), mito-chondria, Golgi] as well as in the plasma membrane[14–16]. Coordinated recruitment of lipid-modifyingenzymes and proteins at the place of autophagosomeformation gives rise to the limiting membrane [17].Growth of this membrane occurs through two enzyme-regulated processes that mediate conjugation of (1) aprotein—light chain protein 3 or LC3—to one of theconstituent membrane lipids and (2) of two cytoso-lic proteins (Atg5 and Atg12) between them [9,18].SNARE-like proteins, molecular motors and additionallipid-modifying enzymes (phosphatases and kinases)participate in autophagosome-lysosome fusion. Mostof the regulatory components exert their action onthe nucleation complexes. For example, the Target of

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

The pathophysiology of autophagy 257

Rapamycin, a well-characterized negative regulator ofthis pathway, prevents the recruitment and interactionof specific Atgs of the nucleation complex to the siteof autophagosome formation [19–21]. Although lessis known about the regulation of later steps of theautophagic process, recent studies have identified thetranscription factor EB as a global regulator of Atgsthat can up-regulate initiation, elongation and fusion,as well as lysosomal biogenesis in response to nutri-tional stress [22]. Homologues for most yeast Atgshave been identified in many other species (Drosophila[23], Caenorhabditis elegans [24], Dictyostelium [25],Arabidopsis [26], Trypanosoma [27]) and in mammals,where these genes often have several variants withdiverse functions [28].

The number of yeast genes shown to participatein microautophagy does not exceed a dozen, butthis process also partially depends on Atgs sharedwith macroautophagy [29]. Microautophagy-specificproteins contribute to the formation and sealing ofthe vacuolar membrane (equivalent of the lysosome inyeast) [30]. Failure to identify mammalian homologuesof the yeast microautophagy genes initially led to theproposal that microautophagy was not evolutionarilyconserved. However, recent studies show otherwise,as a microautophagy-like process has been describedto occur in mammalian late endosomes. This process,known as endosomal microautophagy, has adopted themachinery involved in biogenesis of multivesicularbodies to form the invaginating vesicles that sequestercytosolic material for degradation [31].

In recent years, in addition to this ‘in bulk’ non-selective degradation of cytosolic components, specificsequestration of cargo has also been described forboth macro- and microautophagy. In the case of selec-tive macroautophagy, the distinctive characteristic isthe presence of a single type of cytosolic material inautophagosomes, in clear contrast to the ‘in-bulk’ pro-cess, where heterogeneous cargo occupies the lumenof the autophagosome. Selective forms of macroau-tophagy are named depending on the sequestered mate-rial, giving rise to terms such as mitophagy [32,33],pexophagy [34,35], reticulophagy [36,37], lipophagy[38], ribophagy [39] and aggrephagy [40]. Similar cri-teria apply to selective forms of microautophagy, suchas micropexophagy [34,35] micronucleophagy [41,42]and microglycophagy [43]. All these selective formsof autophagy utilize the same essential componentsof the autophagic machinery described for their ‘in-bulk’ variants, but they add an extra step related tocargo recognition. A specific subset of proteins, knownas cargo-recognition proteins, have been involved inthis step and include, among others, p62, neighbourof BRCA1 (NBR1), Nix and the PINK1/Parkin pair[44,45]. These adaptor proteins connect the degra-dation machinery with particular components—oftenubiquitin moieties—on the surface of the cytosolicorganelle to be sequestered [44,46,47]. Cytosolic chap-erones, in particular the heat shock cognate proteinof 70 kDa, or hsc70, and BAG-3, have also been

recently described to contribute to selective recogni-tion of aggregates by macroautophagy in chaperone-assisted selective autophagy (CASA) [48].

Selectivity is the distinctive feature of CMA, thethird type of mammalian autophagy, wherein particularsoluble proteins bearing in their sequence a pentapep-tide targeting motif biochemically related to KFERQ,are recognized by hsc70, which mediates their deliv-ery to the lysosomes for direct translocation acrossthe lysosomal membrane [8,49] (Figure 1). Althoughyeast genetic approaches could not be applied to themolecular dissection of CMA, since this pathway hasso far been described only in mammals, about eightdifferent proteins have been identified to participatein this pathway using biochemical procedures. Mostof the identified CMA components have promiscuousfunctions and are shared with other intracellular pro-cesses. Such is the case for the cytosolic chaperoneshsc70 and hsp90 [50,51] and the novel pair of reg-ulators, glial fibrillary acidic protein and elongationfactor 1α [52]. Interestingly, lysosome-specific vari-ants have been identified for each of these proteins,suggesting that post-translational modifications of thecytosolic proteins determine their association with thisdegradative compartment and their commitment toCMA. So far, the only CMA-exclusive component isthe lysosome-associated membrane protein type 2A(LAMP-2A), which acts as a receptor for substrates ofthis pathway [53]. Once the cytosolic proteins destinedfor CMA degradation bind to this single transmem-brane protein, they promote its assembly into a higher-order molecular complex required for translocation ofthe CMA substrates into the lysosomal lumen [51].

Interested readers are referred to more focusedreviews [2,3,8,29,54] for a comprehensive descriptionof the molecular apparatus executing these differentautophagic pathways. For the purposes of this review,the term ‘autophagy’ will represent macroautophagy,unless otherwise mentioned.

Physiological relevance of autophagy

As described in the Introduction, a growing number ofcellular functions have been attributed to autophagy.We highlight here some of the best-characterized func-tions that have contributed to expanding the physio-logical relevance of this catabolic process. The roleof autophagy in cellular defence, by participating ininnate and adaptive immunity, is discussed in detailin the section dedicated to autophagy and the immunefunction.

Autophagy in the cellular energetic balanceBoth macroautophagy and CMA are induced as anacute adaptive response to a variety of metabolicstressors, including, among others, nutrient starvation,growth factor withdrawal, high lipid content chal-lenges or hypoxia [4,55–57]. Under these conditions,

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

258 S Sridhar et al

free amino acids (especially branched-chain aminoacids), released by autophagic proteolysis of intracel-lular proteins and organelles, are recycled to main-tain protein synthesis even when nutrients are scarce.Autophagy in the liver converts this organ into amain source of amino acids that are then deliveredto other organs through the blood stream during star-vation [5,56,58,59]. In fact, the low plasma concen-trations of essential amino acids and decreased ATPlevels in most tissues observed in autophagy defectiveAtg5−/− and Atg7−/− neonates confirm the impor-tance of this pathway during nutritional deprivation,in this case as a result of the cessation in transpla-cental nutrient supply at birth [60,61]. Amino acidsresulting from autophagic breakdown could also beutilized for the production of cellular ATP throughdirect oxidation, or by fuelling the TCA cycle and glu-coneogenesis with intermediates such as oxaloacetate.This contribution of amino acids to energy homeostasishas been confirmed in specific conditions, such as inthe case of interleukin-3-deprived haematopoietic cells,where the addition of TCA cycle substrates, such asmethylpyruvate, is enough to preserve cellular viability[62]. However, amino acids are not an efficient sourceof energy. Instead, the array of energy stores mobi-lized by autophagy is now recognized to include moreenergetically efficient molecules, such as lipids, glyco-gen and nucleic acids. The hydrolytic products of thesemolecules, free fatty acids, glucose and nucleotides,can be funnelled into the TCA cycle, gluconeogenesisor glycolysis to produce ATP [4,43].

This capability of autophagy to maintain ATP pro-duction and support macromolecular synthesis makesit a pro-survival pathway of particular importance inorgans with high energetic requirements, such as theheart or skeletal muscles. Alterations of this specificautophagic function also constitute the basis of somecommon metabolic disorders.

Autophagy and cellular quality control

The proteome and cellular organelles are susceptible todifferent toxic insults that can lead to the generation ofmisfolded or modified soluble proteins, protein cross-linking and oligomerization into high-order irreversiblestructures or aggregates, and accumulation of defec-tive, no longer functional, organelles that could becomeharmful for the cells [63–65]. Basal autophagy, oftenreferred to as ‘quality control autophagy’, is integralto the cellular surveillance machinery responsible forrecognition and removal of these aberrant structures[66,67]. Basal autophagy is particularly important innon-dividing post-mitotic cells that cannot dilute thecellular damage through division. For example, condi-tional knock-out of the essential autophagy genes atg7or atg5 in hepatocytes, neurons and cardiomyocytesleads to marked accumulation of ubiquitin-positive pro-tein aggregates and damaged organelles, even in theabsence of an added toxic challenge [60,68,69].

Exposure to stressors, such as oxidative stress, ERstress or other conditions resulting in massive amountsof unfolded proteins and organelle damage, elicits acti-vation of inducible forms of autophagy [70–73]. In thiscontext, activation of CMA contributes to the selec-tive removal of soluble (aggregate-prone) proteins,whereas macroautophagy facilitates the clearance ofprotein aggregates [40,63,74,75] and whole organelles[33,36,37]. However, it is possible that macroau-tophagy ameliorates proteotoxicity not only throughaggregate removal but also by the in-bulk sequestra-tion of ‘still-soluble’ forms of the pathogenic proteinsbefore they aggregate [68,76,77]. Autophagy is alsoimportant to restore organelle homeostasis [36,72,78],to eliminate damaged organelles after stress [37,79]and to assist cells to adapt their organelle content tothe changing environmental conditions (eg it controlsperoxisome number during nutrient auxotrophy or totalmitochondria mass for energetic balance) [80].

Autophagy-mediated control of protein and organellequality, as well as organelle number, is essential for themaintenance of cellular homeostasis and to guaranteecellular survival during stress.

Cellular remodelling and autophagyCellular differentiation often requires the eliminationof large subsets of proteins, nucleic acids or organellesin order to switch into the next developmental stage orfinal differentiated state. Furthermore, cellular remod-elling is an energy-demanding process often associ-ated with nutrient-deprived developmental phases. Itis no surprise, therefore, that autophagy has emergedthrough evolution as one of the favoured cellulartools to accomplish these developmental remodellingtasks. The capability to eliminate whole regions of thecytosol, unique to some of the autophagy variants, andits recycling properties underlie the important role thatthis process plays during development. Autophagy hasbeen shown to be necessary in yeast during sporula-tion [81], in C. elegans during dauer formation [24],in Drosophila during the transition from the larval tothe pupal stage [82] and in Dictyostelium for switch-ing from amoeba to fruiting body [25]. Examplesof autophagy-mediated differentiation events in mam-malian cells include, among others, the removal ofmaternal macromolecules during early embryogenesis[83] and the clearance of mitochondria during ery-throcyte [84–86], lymphocyte [87,88] and adipocyte[89,90] differentiation.

Autophagy in cellular death and cell survivalAs indicated in the previous sections, the ability ofautophagy to assist cells to adapt to the changing envi-ronment, to protect them against the damage caused bytoxic insults and to defend them from pathogens has ledto the classification of this pathway as a cell-survivalmechanism. However, interplay between autophagyand mechanisms related to cell death have also beendescribed. The best-accepted example of autophagy as

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

The pathophysiology of autophagy 259

a cell death effector is the degradation of the salivaryglands during pupal transition in Drosophila [82,91]. Incontrast, direct evidence of a similar role for autophagyin mammals is sparse [92]. In fact, many of the orig-inal claims of ‘cell death by autophagy’ have cur-rently been revised and reformulated as ‘cell deathwith autophagy’, since the main support for autophagicinvolvement in many of these cases was the obser-vation of autophagic vacuoles in the dying cell [93].Although an increase in the number of autophagic vac-uoles could result from their increased formation, thelarge capability of the lysosomal system often preventstheir accumulation by facilitating their rapid clearance.It is more frequent that the increase in autophagosomecontent results from a compromise in their clearanceby lysosomes and, consequently, from impaired ratherthan enhanced macroautophagy. Systematic analysis ofautophagic flux, rather than autophagic markers, shouldhelp to clarify these issues. However, independent ofthis initial controversy, it has become clear that, asdescribed in the following sections, once in a diseasecontext interactions between autophagy and apoptosis(or type I cell death) are complex and need to be anal-ysed in the specific disorder or cell type [94–96]. Infact, autophagy and apoptosis share some molecularcomponents and appear to be in a delicate balance witheach other in most systems examined [97,98].

Pathology of autophagy

The broad array of physiological functions attributedto autophagy justifies why alterations in this catabolicprocess lead to cellular malfunctioning and often celldeath, and has set the basis for its contribution to thepathogenesis of severe human disorders. Rather thanproviding an exhaustive list of disease conditions inwhich autophagy has been shown to be altered, inthe following sections we have selected four commonpathological conditions, cancer, neurodegeneration,heart dysfunction and immune and infectious diseases,to comment on their interplay with the autophagic sys-tem. The reason behind our selections is the fact thatin all of these pathologies growing evidence supportsa dual role of autophagy in their pathogenesis and dis-ease progression, which leads to the question, ‘Shouldwe inhibit or activate autophagy in these pathologies?’.

Autophagy and cancerCancer was the first human pathology connectedto autophagy through the discovery that Beclin1(Atg6/VPS30 homologue), a core component of theautophagosome nucleation complex, was monoalleli-cally deleted in 40–75% of sporadic human breast,ovarian and prostate cancers [99]. Independent stud-ies verified that heterozygous Beclin1+/− mice developspontaneous tumours, including lymphomas, lung car-cinomas, hepatocellular carcinomas and mammary pre-cancerous lesions [100,101]. Analogous generation

of spontaneous hyperproliferating tumours was alsoreported later upon deletion of other autophagy-relatedgenes, such as the ultraviolet resistance-associatedgene (UVRAG), the Bax interacting factor-1 (Bif-1 )and the LC3 (Atg8 homologue) processing proteaseAtg4c [102–104]. Furthermore, the products of com-mon oncogenes, such as class I PI3K, PKB, TOR andBcl-2, have been shown to act as autophagy repres-sors, whereas tumour suppressor gene products suchas p53, PTEN, DAPk and TSC1/TSC2 exert a stim-ulatory effect on autophagy (reviewed in [3,98,105]).Although, in light of this opposite effect of oncogenesand tumour suppressors on autophagy, this process wasinitially classified as an anti-oncogenic mechanism,this conclusion has been challenged by experimen-tal evidence supporting that under certain conditionsautophagy can also be pro-oncogenic (Figure 2).

From the perspective of cellular energetics, auto-phagy is likely a pro-survival mechanism in both earlyand late stages of cancer development, as it suppliescells with the energy and essential macromolecules tosustain normal cellular functions. However, it has beenproposed that in the early stages of cancer develop-ment, quality control by autophagy, particularly overgenome maintenance, inhibits tumourigenesis, confer-ring anti-oncogenic functions upon this pathway. Inaddition, autophagy could also play a role in the main-tenance or entry of cells into the G0 phase and con-sequently, prevent spontaneous hyperproliferation ofcells [106,107]. In contrast, in the late stages of onco-genesis, autophagy is necessary for cancer survival,as it contributes both energy for the rapidly dividingcancer cells and quality control functions to eliminateintracellular damage caused by the aggressive tumourmicroenvironment and by anti-oncogenic interventions(Figure 2). In fact, consistent with the pro-survivalfunction of autophagy, compromise of this pathway(ie Beclin1 interference) in apoptosis-deficient tumourcells impairs their survival in metabolic stress condi-tions in vitro and in vivo [108]. In general, blockageof autophagy sensitizes cells to metabolic stress, oftenleading to necrotic cell death accompanied by inflam-mation, a condition shown to promote tumourigene-sis [109–111]. Metabolic stress is intrinsic to rapidlygrowing tumours, in which poor vascularization resultsin lack of nutrients and oxygen for long periods of time[112–116]. Activation of macroautophagy under theseconditions has the potential to accommodate the acuteenergy demands by providing cancer cells with aminoacids, free fatty acids and glucose, required to gener-ate energy through the TCA cycle and via β-oxidation(Figure 2). Macroautophagy has also been recentlyimplicated in facilitating the unique utilization of glu-cose by cancer cells, known as the ‘Warburg effect’[117,118], by which cells favour glycolysis to accel-erate ATP production and generate glycolysis inter-mediates required for the transformed cells [119,120].Blockage of autophagy has been shown to prevent Ras-mediated oncogenic transformation by reducing glu-cose uptake and its utilization toward glycolysis [121].

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

260 S Sridhar et al

Figure 2. Autophagy and cancer. Autophagy may play opposite roles in the oncogenic process. Anti-tumoural effect (left): activemaintenance of cellular quality control for cytosolic pro-oncogenic proteins such as p62 prevents malignant transformation of non-tumoural cells. In addition, the supply of energy provided through macroautophagy activation reduces the dependence on glycolysis,while assuring the energy required for maintenance of a stable genome, further preventing oncogenesis. Pro-oncogenic effect (right): thereduction in macroautophagic activity in early stages of the oncogenic process favours malignant transformation, as the accumulationof molecules such as p62 activates signalling mechanisms that promote necrosis and inflammation. Poor quality control as a result ofdiminished macroautophagy can also result in accumulation of defective mitochondria, with the subsequent release of harmful molecules(cytochrome c and reactive oxygen species) that contribute to further altering genome maintenance. However, as the tumour progresses,activation of macroautophagy is observed in many oncogenic processes, in part to compensate for the poor nutritional supply associatedwith rapidly growing tumours and to defend cancer cells against damage induced by anti-oncogenic therapies. In addition, enhancedmitochondrial degradation in this stage may contribute to the up-regulation of glycolysis to maintain the energetic balance (Warburgeffect) characteristic of malignant cells.

Although it is clear that interrupting glycolysis elimi-nates the energetic advantage of the cancer cells andcompromises proliferation, the mechanism by whichautophagy up-regulates glycolysis is not known. It hasbeen proposed that this effect could in part be medi-ated by activation of the removal of mitochondria viaautophagy (mitophagy) to force energy dependence onglycolysis [119,120,122] (Figure 2). Autophagy couldimpact oncogenic transformation in multiple ways; inaddition to the proposed effect on glycolysis, a bettermaintenance of highly efficient mitochondria throughmitophagy has been recently proposed to also con-tribute to meeting the energetic requirements of thetransforming cells [123]. Furthermore, an additionalfactor in this already complex scenario is the possi-ble impact that metabolic changes in the surroundingstroma cells could have on the cancer cells. In fact,an autophagy-dependent metabolic switch in the can-cer stroma fibroblasts has been recently proposed. Thisswitch in the stroma could provide cancer cells withglycolytic intermediates necessary for efficient ATPproduction by oxidative phosphorylation [122].

Reduction in cellular energy upon autophagic com-promise also decreases the fidelity of cellular pro-cesses such as DNA replication and mitosis, result-ing in genetic aberrations [124]. The occurrence of

these aberrations in tumour cells with defective qual-ity control of organelles and toxic proteins due to theautophagic compromise can precipitate death of thecancer cells. In this respect, accumulation of the pro-tein p62 (normally degraded by macroautophagy) whenthis pathway is compromised leads to the formationof p62 aggregates capable of stimulating inflammationby inhibition of nuclear factor-κB (NF-κB) and activa-tion of NF-E2 related factor 2 (Nrf-2) [109,125,126](Figure 2).

Little is known to date about the contributions ofother forms of autophagy (CMA and microautophagy)to cancer biology. Recent studies have revealed thatCMA contributes to the turnover of inactive forms ofthe M2 isoform of pyruvate kinase, a key enzymein the maintenance of glycolysis in cancer cells[127]. A number of additional glycolytic enzymeshave been recognized as CMA substrates, includ-ing glyderaldehyde-3-phosphate dehydrogenase, phos-phoglycerate kinase, aspartate aminotransferase andaldolase B, strengthening the possible link betweenCMA and the energetic balance of cancer cells[49,128].

Collectively, the relationship between autophagy andcancer has proved to be a very complex one. This dualcapability of autophagy to, on the one hand facilitate

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

The pathophysiology of autophagy 261

tumour progression and, on the other hand diminishmalignant transformation, has reinforced the need tocritically evaluate autophagy in a disease-context andstage-specific manner, especially during the designof targeted therapy, as autophagy function is tightlyrelated to these factors [78,94].

Effectors and modulators of macroautophagy havebecome attractive targets in the treatment of can-cer. Autophagy up-regulation has been described inmany different types of cancers in response to anti-oncogenic drugs, when it often exerts a protectiveeffect [129,130]. Based on these findings, down-regulation of autophagy should be an effective way ofenhancing the cytotoxic effect of different chemother-apy protocols. In fact, inhibition of macroautophagyhas proved to be effective in enhancing the toxicity ofdrugs commonly used in cancer therapy, such as oxalo-platin [129] and the kinase inhibitors sorafenib [131],epirubicin [132] or 5-fluorouracil [133].

Different reports have also documented that the cyto-toxic effect of some common anti-cancer agents isin part due to their ability to up-regulate autophagy[134–138]. These findings now raise a note of cautionin the universal applicability of autophagy inhibitorsas anti-cancer drugs. However, additional studies arerequired to clarify the contribution of the observedincrease in autophagy in cancer cell death under theseconditions. Often, the proposed increase in autophagyis based on the fact that levels of autophagy-relatedproteins are higher during the treatment. Up-regulationof autophagy markers does not necessarily meanincreased autophagic flux. In fact, if autophagy is func-tioning properly, accumulation of autophagic compart-ments rarely occurs. It is thus possible that the observedincrease in autophagic markers in some of these studiesis actually a reflection of a compromise in the clear-ance of autophagic compartments, rather than in theirformation. Accumulation of autophagolysosomes cancontribute to cell toxicity and be detrimental for theaffected cells. Interestingly, this toxic effect of the‘congestion’ of the autophagic pathway is currentlybeing explored for therapeutic purposes. Althoughsome success has been obtained by directly targetingthe final steps of autophagy, the lysosomal degrada-tion, the characteristic plasticity of the lysosomal com-partment makes a very prolonged inhibition necessarybefore this intervention becomes detrimental. One wayaround this problem, which is currently utilized in dif-ferent clinical trials, is to combine the lysosomal block-age with enhanced formation of autophagosomes (egby inhibition of the mTOR complex with rapamycin)[139–141]. The higher content of autophagic vacuoles,as a result of both increased formation and reducedclearance, can compromise cell viability by differentmechanisms. Accumulation of these compartments inthe cytoplasm interferes with intracellular traffickingand prevents recycling of essential cellular compo-nents (amino acids, free fatty acids, etc.). In addition,as autophagolysosomes persist inside cells longer than

usual, their membranes often become leaky, facilitat-ing the release of potent lysosomal enzymes in thecytosol, further enhancing cellular damage and toxi-city.

Autophagy and neurodegenerationMaintenance of cellular homeostasis is essential inneurons, a typical example of non-dividing differen-tiated cells. The importance of autophagy in neuronalhomeostasis has now been well supported, despite orig-inal disagreement on whether autophagy was activein neurons. The reason for this contention was thefact that autophagosomes, the morphological featureof macroautophagy, were rarely observed in neurons.However, recent studies support that this pathway isconstitutively active and that it can be further up-regulated in neurons in response to very different stres-sors [142–144]. Perhaps the most conclusive supportfor the existence of basal autophagy in neurons and forits contribution to the maintenance of neuronal survivalcame from studies in transgenic mouse models con-ditionally knocked out for essential autophagy genesin the central nervous system [68,69]. These animals,even in the absence of any added stressor, displayedmarked neurodegeneration associated with the accumu-lation of intracellular protein inclusions and neuronalloss. The efficient capability of the lysosomal systemto remove newly formed autophagosomes could bebehind the low occurrence of detectable autophago-somes in neurons in a given moment.

As with many other cells, neurons up-regulateautophagy in response to common stressors but alsoin defence against neuron-specific injuries, such asaxotomy, neuronal ischaemia or excitatory toxicity[145–147] (Figure 3). Often, failure to up-regulateautophagy under these conditions or primary com-promise of autophagic activity during the stress pre-cipitates neuronal death post-injury [148]. However,activation of autophagy in neurons may have a rela-tively narrow window of opportunity, because recentreports have revealed that genetic or chemical inhibi-tion of autophagy becomes beneficial at some specificstates post-injury [105]. These findings suggest thateither excessive up-regulation of autophagy, or up-regulation of this pathway under conditions in whichautophagosome formation or clearance cannot be guar-anteed, may be harmful to neurons [149]. In furthersupport of the possible detrimental effect of massiveup-regulation of autophagy, recent studies have shownthat, under specific conditions, autophagosomes canbecome a source of reactive oxygen species and thusaggravate neurotoxicity [150].

Changes in autophagic activity have been describedin many protein conformational disorders, and amongthem neurodegenerative diseases have received particu-lar attention. The feature shared by all these conditionsis the presence of pathogenic proteins that accumulateinside neurons, organized in the form of oligomericor multimeric structures. The disease originates from

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

262 S Sridhar et al

Figure 3. Autophagy and Neurodegeneration. (A) Protective autophagy : macroautophagy and CMA both contribute to the maintenance ofneuronal homeostasis and are necessary in the defence of neurons against injury and stressors. (B) Defective macroautophagy : defects inmacroautophagy have been described to occur at very different levels in neurodegenerative diseases. Some of the possible steps affectedin this process are highlighted in the model, and the conditions in which they have been observed are described in the text. (C) DefectiveCMA: primary defects in CMA have been described both in Parkinson’s disease and in certain tauopathies. While in the former condition,pathogenic proteins such as α-synuclein can block access of other cytosolic proteins to lysosomes via CMA by abnormally binding to thetranslocation machinery, in tauopathies the accumulation at the surface of lysosomes of oligomeric forms of pathogenic tau targetedvia CMA destabilizes the lysosomal membrane and results in leakage of lysosomal enzymes into the cytosol, which often triggers cellulardeath.

both the loss of function of the pathogenic protein andthe toxicity associated with the presence of the abnor-mal protein structures, which often leads to neuronaldeath. The first connection between neurodegenera-tion and autophagy originated from the observationthat protein aggregates can be eliminated by autophagy[40,74] (Figure 3). In fact, pharmacological activationof this catabolic process in experimental animal mod-els of Huntington’s disease reduced cellular toxicityand slowed down progression of the disease [151].After these initial reports, the degradation of manyother pathogenic proteins by autophagy and the ben-eficial effect of up-regulation of this pathway haveproved to be true in experimental models of manyother neurodegenerative conditions [152]. In fact, notonly the acute pharmacological activation of autophagybut even chronic conditions that lead to a progressivemaintained up-regulation of this process, such as underconditions when ER stress is maintained, have beenshown to have a positive effect [153]. These findingshave now opened the possibility of utilizing modulatorsof autophagy as the basis for therapeutic approaches for

these types of pathology and, in fact, ongoing chemi-cal screenings aim at identifying chemical modulatorsof this pathway with higher selectivity and potency[154].

However, there are certain limitations to the uni-versal use of up-regulation of macroautophagy foranti-neurodegenerative purposes. In fact, it has beenreported that not all protein aggregates are recog-nized by the macroautophagic machinery. Despitethe current dissection of molecules that participatein autophagic recognition of aggregates (p62, NBR1and several cytosolic chaperones, including hsc70 andBAG-3), the presence of these proteins in an aggre-gate, although necessary, does not seem to be suffi-cient for autophagic recognition. Thus, some proteininclusions positive for these cargo-recognition proteinsremain unnoticed by the autophagic machinery, mak-ing superfluous any attempt to enhance autophago-some formation as a way to eliminate these toxicprotein products [155]. Further studies are needed toaddress whether expression of other cellular compo-nents or specific post-translational modifications in the

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

The pathophysiology of autophagy 263

aggregated proteins could enhance their recognition bythe autophagic systems.

A second scenario, in which chemical up-regulationof macroautophagy may not be effective againstneurodegeneration, is in those conditions in whichmacroautophagy itself is compromised. Understandingthe macroautophagy step or steps defective in specificneurodegenerative disorders is a priority if autophagyenhancers are to be moved forward in the treatment ofthese conditions. Examples of defects in almost all ofthe steps of macroautophagy have been described indifferent neurodegenerative disorders [66] (Figure 3).For example, initiation of autophagy may be com-promised because of altered signalling through theinsulin or mTOR pathways, which is tightly bound toactivation of autophagy. Also, conditions resulting inlower content of available Atgs will reduce neuronalautophagic capability, such as the recently describeddepletion of LC3-II through abnormal interaction andaggregation with p62 in the presence of dopaminergicneurotoxin [156]. Along the same lines, abnormal inter-action between mutant α-synuclein (the protein thataccumulates in protein inclusions in Parkinson’s dis-ease) and Rab1 has been shown to misplace Atg9, aprotein required for the formation of autophagosomes,out of the ER membrane [157]. Under other conditions,problems arise at the level of cargo recognition, eitherbecause of alterations in the organelle-specific mark-ers for degradation or in the autophagic machinery. Aprimary defect at the level of the limiting membranethat seems to prevent selective recognition of cargo hasbeen recently described in cellular and animal modelsof Huntington’s disease [158]. In these cases, mutanthuntingtin binds tightly to the inner surface of the form-ing autophagosome, resulting in an abnormally highinteraction with p62 in this compartment that affects itsability to bind cargo. In other conditions, autophago-somes form and sequester the pertinent cargo but theyfail to be cleared from the cytosol. Problems withclearance could result from alterations at very differ-ent levels. For example, problems with vesicular traf-ficking could indirectly interfere with the mobilizationof autophagosomes toward the lysosomal compartment[159]. Pathogenic proteins can also interfere with thefusion step, which, although still not completely elu-cidated at the molecular level, is known to dependon different SNARE proteins, the actin cytoskeletonand the histone deacetylase 6 (HDAC6) [160]. Con-sequently, changes in any of these components couldlead to intracellular accumulation of autophagosomes.Lastly, primary defects in the lysosomal compartmentalso have a negative impact on clearance of autophago-somes in different neurological disorders. The rea-sons for lysosomal failure could be multiple. In prin-ciple, most lysosomal storage disorders could com-promise autophagosome clearance because often theaccumulation of the undegraded product inside lyso-somes limits their degradative capacity [161]. In addi-tion, other conditions that alter lysosomal membrane

stability, decrease lysosomal biogenesis or change lyso-somal pH could also alter autophagosome clearance.In fact, defective acidification of lysosomes, becauseof compromised targeting of a component of the pro-ton pump from the ER to the lysosomal membrane,has been already identified as causative of the reducedrates of autophagy in some models of Alzheimer’sdisease [162]. As functional analysis of autophagybecomes more broadly utilized in the study of neurode-generative diseases, it is likely that new connectionsbetween these pathologies and autophagy will becomeevident.

Alterations of CMA have also been described insome neurodegenerative disorders (Figure 3). Althoughin many of them, this pathway becomes up-regulatedas a consequence of the failure in macroautophagy, dif-ferent pathogenic proteins have been shown to directlyinterfere with CMA activity [163]. Mutant forms of α-synuclein can be targeted to lysosomes via CMA but,in contrast to the wild-type protein that binds to thelysosomal receptor and rapidly reaches the lumen fordegradation, the mutant variants fail to translocate. Per-sistence of mutant α-synuclein tightly bound to theCMA receptor at the lysosomal membrane inhibitslysosomal degradation of other cytosolic proteinsvia this pathway [164]. A similar effect was laterdescribed for wild-type α-synuclein upon modificationby dopamine, which makes CMA blockage relevant forsporadic forms of Parkinson’s disease, more commonthan the familial forms [165]. Later studies have alsoproposed interference of CMA by other Parkinson’sdisease-related proteins, such as mutant forms of UCH-L1 [166]. A higher degree of lysosomal compromise asa result of lysosomal targeting of pathogenic proteinsvia CMA has been described in certain tauopathies,where the mutant forms of the protein not only interactabnormally with CMA components blocking this path-way, but also disrupt the lysosomal membrane as theyorganize into toxic oligomeric species on their surface[167].

To date, systematic studies analysing the contribu-tion of alterations in microautophagy to neurodegen-eration have not been performed. However, the factthat protein unfolding is not a prerequisite for microau-tophagy makes attractive the possibility that micro-aggregates can be delivered to this pathway for degra-dation.

Growing evidence supports that neurons utilizediverse ways to eliminate pathogenic proteins and thatmultiple steps of the autophagic process are oftenaffected in a given neurodegenerative disorder. Thus,for example, in the case of Alzheimer’s disease (AD),macroautophagy contributes to the elimination of thetoxic product of the amyloid precursor protein (APP)A-β. However, clearance of A-β is compromised as thedisease progresses, due to the failure in this autophagicpathway [168]. In most forms of AD, the autophagicdefect occurs in the late steps of macroautophagy.Thus, autophagosomes form properly sequestering theusual cargo but they are not eliminated through the

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

264 S Sridhar et al

lysosomal system [168]. As described above, deficientacidification of the lysosomal lumen, due to the inabil-ity to target to lysosomes one of the subunits of theproton pump that usually acidifies this compartment,has been recently described in some types of AD [169].Persistence of autophagosomes in AD neurons is par-ticularly toxic because the enzymes responsible forprocessing of APP into A-β as well as APP accumu-late in these compartments. Autophagosomes becomein this case a new site of intracellular production oftoxic A-β [168]. Failure of the autophagic system com-promises the elimination of aggregate forms of tau, aprotein that also accumulates in AD neurons. In fact,for certain types of tau mutations, this pathogenic pro-tein could contribute to the failure of macroautophagy,due to the toxic effect that the still soluble forms ofthe protein exert in the membrane of lysosomes whenthey are delivered to this compartment by CMA [167].

Similarly, in the case of Parkinson’s disease (PD)the most common pathogenic protein, α-synuclein, isusually degraded by different autophagic and non-autophagic pathways. Soluble forms of the proteinare substrates of both the ubiquitin proteasome sys-tem and of CMA [164,170]. However, macroautophagyis the only plausible way for the elimination of thepathogenic variants of this protein once they aggregate[171]. As in the other disorders, pathogenic forms of α-synuclein interfere with the normal functioning of theubiquitin proteasome pathway [170], CMA [164,165]and even macroautophagy at the level of autophago-some formation [157]. The recently identified physio-logical role for Parkin, a protein also related to PD,in mitophagy, suggests that alterations of this selec-tive form of autophagy could also contribute to thepathogenesis of this disease [172].

Macroautophagy has proved to be an effective mech-anism for the degradation of aggregates of huntingtin,the protein mutated in Huntington’s disease (HD)and main component of the inclusions observed inthe affected neurons [151,173]. Huntingtin is a verydynamic protein that undergoes physiological cleavageinto different functional fragments that likely are turnedover by different proteolytic systems. Mutant hunt-ingtin, even before aggregation, seems to be primarily atarget for macroautophagic degradation [174], althoughspecific posttranslational modifications can also favourits elimination by CMA [175]. As in the other neu-rodegenerative disorders, growing evidence supportsa direct toxic effect of the mutant protein on theautophagic system. For example, mutant huntingtin hasbeen described to accumulate on the luminal side of thelimiting membrane as the autophagosome forms andthus interfere with the recognition of cytosolic cargo,including huntingtin protein aggregates [158]. Alter-ations at the level of autophagosome trafficking andclearance have also been described in some experimen-tal models of HD [176]. Although huntingtin bears sev-eral CMA-targeting motifs in its amino acid sequence,the contribution of this pathway to the removal of thepathogenic protein is, overall, modest. However, recent

studies support a beneficial effect of the artificial tar-geting of mutant huntingtin for CMA degradation thatcould be explored for therapeutic purposes [177].

It is anticipated that a similar dual interaction ofdifferent pathogenic proteins with the autophagic sys-tems—by which they can be eliminated through thesepathways but also interfere with their normal function-ing—could also contribute to pathogenesis in otherneurodegenerative disorders.

Autophagy and the failing heartThe heart is comprised of long-lived, post-mitotic cellswith little regenerative capacity, which are continu-ally subjected to stress, such as ischaemia, pressureoverload and ischaemia–reperfusion injury. Adapta-tion to these conditions is attained through remodelling(myocytes elongate and undergo hypertrophy) and fail-ure to do so frequently constitutes the basis of coronaryartery disease, hypertension and congestive heart fail-ure [178,179]. Evidence for the role of basal autophagyin the quality control and housekeeping of cardiomy-ocytes was first obtained through genetic models ofLAMP-2 knock-out mice (defective for autophago-some–lysosome fusion) that demonstrated cardiomy-opathy and abnormal accumulation of autophagicvacuoles, similar to that observed in Danon’s diseasepatients [180,181]. In fact, mutations in the LAMP-2 gene were subsequently described in these patients[182]. Studies in cardiomyocyte-specific Atg5 and Atg7knock-out models have reiterated the need for func-tional autophagy to preserve normal cardiac function,both under basal conditions and in response to stress[183]. In fact, pronounced loss of autophagy in car-diomyocytes with age seems to be behind differentforms of age-related cardiomyopathy [184].

Added to the key role of basal macroautophagyin cardiomyocyte homeostasis, growing evidence alsosupports an important contribution of this pathwayin the heart in response to stressors. As describedin the brain, macroautophagy has also proved tobe essential for the prevention of cardiomyocyteproteinopathies, such as those arising from desminand α-β-crystallin accumulation [185,186]. Autophagyinduced in response to cardiac stress (especially duringischaemia–reperfusion injury) also plays a cytopro-tective role in the heart. In fact, in support of thisprosurvival function, different studies have demon-strated that pharmacological inhibition of autophagyduring mild ischaemic stress enhances cardiomyocytedeath [187,188], whereas macroautophagy activationis cardioprotective [189,190]. The beneficial effect ofmacroautophagy up-regulation under these conditionscould result, in part, from an efficient removal ofthe mitochondrial population compromised under theseconditions. Thus, mitochondrial membrane depolar-ization and increased production of reactive oxygenspecies by mitochondria are common features asso-ciated to ischaemia–reperfusion. Degradation of thealtered mitochondria by macroautophagy under these

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

The pathophysiology of autophagy 265

conditions has been shown to efficiently amelioratecellular damage by reducing the activation of pro-apoptotic cascades [191–193].

However, induction of macroautophagy in thestressed heart can also be detrimental and contributeto heart failure, judging by the fact that pharmacolog-ical or genetic blockage of macroautophagy enhancescell survival post-ischaemia–reperfusion injury in spe-cific settings [194]. The timing and the nature of thecondition that induced cardiac stress may be essentialfor the switch in the effect of macroautophagy. Thus,some studies suggest that macroautophagy is cardio-protective during ischaemic recovery but maladaptiveduring reperfusion recovery [195]. It is possible that,during reperfusion, the need for removal of dysfunc-tional mitochondria and oxidatively damaged cellularstructures that accumulated during the ischaemic periodcould drive the autophagic response to be more aggres-sive than desired, resulting in cell death. In fact, geneticup-regulation of autophagy in response to the haemo-dynamic stress-induced hypertrophic growth responsehas also proved to be maladaptive and to result inpathological cardiac hypertrophy [196].

In summary, as in the case of tumour biologyand neurodegeneration, the role of autophagy in heartpathology is also context-dependent and excessive orinsufficient macroautophagy is often associated withdisease. However, when maintained under strict con-trol, up-regulation of macroautophagy by compoundssuch as resveratrol has proved to be a useful cardiopro-tective strategy during ischaemia–reperfusion injury[197–199].

Although the heart is one of the organs in whichCMA is up-regulated most rapidly in response to star-vation [200], the contribution of CMA to cardiomy-ocyte homeostasis and to heart pathology has not beenexplored in depth. As mentioned above, mutations inLAMP-2 occur in patients with Danon’s cardiomyopa-thy, but the phenotype seems, for the most part, to berelated to the presence of large autophagic vacuolesin muscle, reflective of compromised macroautophagy.This could be explained by the fact that the lamp2 geneundergoes splicing, giving rise to three protein variants,A, B and C [201] and that mutations in only the B vari-ant, tightly related to macroautophagy, are enough toreproduce the full vacuolar phenotype [182], whereaschanges in LAMP-2A, the variant required for CMA,do not affect macroautophagy [202].

Autophagy, infectious disorders and autoimmunityThe autophagy machinery can act as a cell-autonomousdefence against invading pathogens through a processknown as xenophagy [203]. Activation of this processhas been extensively reported, for example, in responseto Group A Streptococcus infection in epithelial cells(Figure 4). The fact that Atg5 deficiency allows bacte-ria to survive and multiply robustly supports the normalcontribution of autophagy in the elimination of thispathogen [204]. Autophagic surveillance is not limited

to bacteria as, in fact, a protective role for autophagyhas been also demonstrated for the vesicular stomatitisvirus, herpes simplex virus 1 or HIV-1, among others[205–207].

Cytosolic autophagy often becomes a second surveil-lance point for pathogens such as Listeria monocy-togenes that can escape phagosomes by puncturingtheir membrane to replicate in the cytoplasm. How-ever, once in the cytoplasm, the autophagic surveil-lance mechanism is activated to entrap the escapedbacteria and deliver them to endocytic and lysoso-mal compartments for degradation [208]. Pathogensthat survive inside vesicular compartments can also becontrolled by macroautophagy. For example, Mycobac-terium tuberculosis resides in phagosomes by avoidingtheir fusion to lysosomes. Interestingly, this blockagecan be overcome by induction of autophagy, leading tothe degradation of this pathogen [209].

Autophagy proteins may also help to acceleratephagosome maturation through a process that has beentermed ‘LC3-associated phagocytosis’. Engagement oftoll-like receptors has been shown to induce recruit-ment of LC3 to the phagosomal membrane and favourfusion with the lysosome [210].

Despite this active involvement of autophagy inpathogen elimination, some pathogens have evolvedto take advantage of the different compartments ofthe autophagy pathway to establish replicative niches[211,212] (Figure 4). Evidence that the autophagosomecompartment is required for the replication of cer-tain pathogens, such as Porphyromonas gingivalis orCoxiella burnetti was provided by demonstrating thattreatment with 3-methyladenine, a well-characterizedinhibitor of autophagosome formation, was efficient indecreasing the survival of these pathogens [213,214].In fact, some of these microorganisms that utilize com-partments of the autophagic system have developedmechanisms to expand the size and number of thesecompartments. For example, Staphylococcus aureus iscapable of secreting a factor that activates autophagy[215]. Seclusion inside the autophagosomes may bebeneficial for pathogens as a way to escape the cytoso-lic surveillance mechanisms, but these compartmentsmay also contain cytosolic materials that can be uti-lized as an energy source by the microorganisms.

This bivalent role of autophagy in pathogen defencesuggests that, although it should be possible to phar-macologically manipulate the autophagic system toeliminate different pathogens, it is essential to firstunderstand the characteristics of the pathogen–autophagy interaction in each of the individualinstances [216,217].

Apart from its role in cell-autonomous immunity,autophagy plays a role in activation of adaptiveimmunity through its recently described involvementin antigen processing and presentation to lympho-cytes [218–220] (Figure 4). In professional APCs,phagosome-processed extracellular antigens are pre-sented through MHC class II molecules to CD4 T

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

266 S Sridhar et al

Figure 4. Autophagy and the immune system. (A) Macroautophagy against pathogens: macroautophagy contributes to the elimination ofdifferent types of pathogens—bacteria and viruses—when they escape to the cytosol after internalization in the phagosome. Under certainconditions, fusion of autophagosomes with phagosomes is required before degradation can occur. (B) Pathogens using macroautophagy :growing evidence supports that certain pathogens have evolved to utilize autophagosomes as a site of replication and can activelyprevent the fusion of this compartment, with lysosomes to guarantee their survival. (C) Antigen presentation: all three forms of autophagy,macroautophagy, CMA and microautophagy have been shown to contribute to antigen loading of MHC class II molecules for presentationof antigens to activate T cells.

cells. However, several cytosolic and nuclear pro-teins have been found associated with MHC IImolecules [221,222]. In fact, pharmacological inhibi-tion of macroautophagy has been shown to be efficientin reducing MHC class II intracellular antigen pre-sentation [223–225], whereas activation of autophagypromotes this type of presentation [226]. Later stud-ies have confirmed the presence of intracellular anti-gens in autophagosomes, and marked reduction ofMHC II presentation in lymphoblastoid cell lines uponknock-down of essential Atgs, as well as in vivo inAtg5-deficient dendritic cells [227,228]. Interestingly,presentation of self-antigens by class II molecules isnot limited to macroautophagy but may also involveother types of autophagy. In fact, over-expression ofLAMP-2A, the receptor for CMA, has been shown toenhance MHC II presentation of intracellular antigens[229], and a new microautophagy-dependent processof antigen delivery into late endosomes in dendriticcells has also been recently characterized [31]. Fur-thermore, participation of autophagy in antigen pre-sentation may not be limited to presentation throughMHC II. Although it is traditionally accepted that MHCclass I presents intracellular antigens processed throughthe proteasomal pathway, growing evidence supportsthat macroautophagy could also assist in MHC I

presentation by collaborating with the proteasome inthe processing of intracellular proteins [230,231].

Also of interest is the recently proposed contributionof autophagy in the establishment of tolerance to selfantigens. Self-reactive T cells are usually actively elim-inated through MHC class II presentation in epithelialcells of the thymus to allow T cell tolerance to self-antigens [232]. Proof that autophagy regulates centralT cell tolerance has been provided by a recent studyshowing that transplantation into athymic nude hostsof Atg5-deficient thymus results in defects in posi-tive and negative selection and the development ofautoimmunity [233]. Interestingly, the antigen-loadingcompartments for MHC class II in immature dendriticcells, which are involved in peripheral tolerance, con-tinuously receive input from autophagosomes [234].Furthermore, genetic polymorphisms in two differentautophagy genes (ATG16L1 and IRGM ) have beenlinked to Crohn’s disease [235,236]. Compromisedautophagic function may result in insufficient induc-tion of tolerance against commensals or self-antigensin the gut, [235,236], although it may also affect thesecretion of antimicrobial proteins by Paneth cells [7].

This novel role of autophagy in the establishmentand maintenance of tolerance also suggests a possi-ble role for autophagy in autoimmunity [237]. In this

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

The pathophysiology of autophagy 267

respect, autophagy is known to contribute to apoptotic-cell clearance [238], and defective clearance of apop-totic cells has been proposed to lead to autoimmunediseases, such as systemic lupus erythematosus [239].

As any other cell type, cells of the immune systemalso depend on autophagy for the maintenance of theirhomeostasis [240], but changes in autophagy in T cellsin particular have also been described to contribute tomodulation of specific cell functions. Thus, the activa-tion of macroautophagy observed upon T cell activa-tion is required to promote T cell differentiation andsurvival and allows cells to accommodate the bioener-getic requirements of these conditions [241–243].

Overall, recent studies support that autophagydefends the cell in a bimodal fashion; first, it directlyeliminates invading pathogens [244]; and second, itsimultaneously assists the immune system of the hostorganism to mount a specialized immune responseagainst the invader by processing pathogenic anti-gens for presentation to T cells [6,203,245]. Thereforeautophagy can be envisaged as an immediate as wellas a persistent mechanism of cellular defence.

Conclusions and future perspectives

The better molecular characterizations of the differ-ent autophagic pathways, as well as the possibilityof genetically manipulating these cellular processes,have helped to establish tight connections betweenautophagic malfunctioning and disease. The initialexcitement about the possibility of modulating auto-phagy with therapeutic purposes in different disordershas been followed by some degree of confusion asto whether autophagy should be up- or down-regulatein these conditions. As described in this review, bothup- and down-regulation of macroautophagy have beenshown to be protective against cancer, neurodegener-ation, infectious diseases and ischaemic insult in theheart. How to decide what to do? Further analysis inmost of these conditions is required to fully under-stand the contribution of autophagic malfunctioning tothe disease. Questions, such as when in the course ofthe disease autophagic function becomes compromised,what changes in each autophagic pathway and whetherthese changes are primary or compensatory to failure inother systems, need to be answered before autophagymodulators can be systematically used in the treatmentof these conditions.

A considerable advance in this respect has beenthe introduction of functional read-outs of autophagyto complement the initial morphological analysis.For example, the presence of a higher number ofautophagosomes in many disease conditions, initiallyinterpreted as an increase in macroautophagy, is nowmore cautiously analysed, because blockage in the latesteps of this pathway can also have a similar mor-phological signature. Thus, conditions initially labelledas having ‘too much autophagy’ are being currently

revised as having ‘a blockage in autophagic clearance’.Blocking autophagosome formation may only be ben-eficial then, when excessive autophagosome contentand autophagic clogging contribute to the pathology,but not when autophagy is up-regulated in the diseaseto compensate for failure in other systems. Along thesame lines, the ultimate goal may not be to repressautophagosome formation in some of these cases, butinstead to facilitate their clearance by directly repair-ing possible defects in the late steps of the autophagicprocess.

In addition to determining the right timing for mod-ulating autophagy in many of these diseases, andwhether this process should be up- or down-regulated,further expansion of the chemical options to manip-ulate autophagy is needed for it to become a routinetherapeutic target. To date, most of the macroautophagyinhibitors used in clinical trials act during the late stepsof this process, on the lysosomal compartment, whichis also shared by other autophagic pathways. In con-trast, for activators, most of the available drugs havean effect on initiation and autophagosome formation,but few compounds have been shown to be effective atincreasing the overall autophagic flux. Ongoing chem-ical screenings are currently addressing the need forthese types of modulators and should render usablemolecules in a timely fashion [154].

AcknowledgmentWe thank Dr Susmita Kaushik for critically review-ing the manuscript. Work in our laboratories is sup-ported by the National Institutes of Health/NationalInstitute on Aging (Grant Nos. AG021904 to AMCand AG031782 to AMC and FM) and the NationalInstitute of Allergy and Infectious Diseases (GrantNo. AI059738 to FM). AMC is also supported by aHirschl/Weill–Caulier Career Scientist Award.

Author’s contributions

All authors contributed separate sections to this reviewand all read and edited the final submitted version.

References

1. De Duve C, Wattiaux R. Functions of lysosomes [review]. AnnRev Physiol 1966; 28: 435–492.

2. Yang Z, Klionsky DJ. Eaten alive: a history of macroautophagy.Nat Cell Biol 2010; 12: 814–822.

3. Mizushima N, Levine B, Cuervo AM, et al . Autophagy fightsdisease through cellular self-digestion. Nature 2008; 451:1069–1075.

4. Singh R, Cuervo AM. Autophagy in the cellular energetic bal-ance. Cell Metabolism 2011; 13: 495–504.

5. Mizushima N. The pleiotropic role of autophagy: from proteinmetabolism to bactericide. Cell Death Differ 2005; 12(suppl 2):1535–1541.

6. Levine B, Kroemer G. Autophagy in the pathogenesis of disease.Cell 2008; 132: 27–42.

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

268 S Sridhar et al

7. Cadwell K, Liu JY, Brown SL, et al . A key role for autophagyand the autophagy gene Atg16l1 in mouse and human intestinalPaneth cells. Nature 2008; 456: 259–263.

8. Cuervo AM. Chaperone-mediated autophagy: selectivity pays off.Trends Endocrinol Metab 2010; 21: 142–150.

9. Kirisako T, Baba M, Ishihara N, et al . Formation process ofautophagosome is traced with Apg8/Aut7p in yeast. J Cell Biol

1999; 147: 435–446.10. Tanida I. Autophagosome formation and molecular mechanism of

autophagy. Antioxid Redox Signal 2011; 14: 2201–2214.11. Mortimore GE, Lardeux BR, Adams CE. Regulation of microau-

tophagy and basal protein turnover in rat liver. Effects of short-term starvation. J Biol Chem 1988; 263: 2506–2512.

12. Mayer A. Cell-free reconstitution of microautophagy in yeast.Methods Enzymol 2008; 451: 151–162.

13. Klionsky DJ, Cregg JM, Dunn WA, Jr., et al . A unified nomen-clature for yeast autophagy-related genes. Dev Cell 2003; 5:539–545.

14. Axe EL, Walker SA, Manifava M, et al . Autophagosome forma-tion from membrane compartments enriched in phosphatidylinos-itol 3-phosphate and dynamically connected to the endoplasmicreticulum. J Cell Biol 2008; 182: 685–701.

15. Hailey DW, Rambold AS, Satpute-Krishnan P, et al . Mitochon-dria supply membranes for autophagosome biogenesis during star-vation. Cell 2010; 141: 656–667.

16. Ravikumar B, Moreau K, Jahreiss L, et al . Plasma membranecontributes to the formation of pre-autophagosomal structures. Nat

Cell Biol 2010; 12: 747–757.17. Kang R, Zeh HJ, Lotze MT, et al . The Beclin 1 network regulates

autophagy and apoptosis. Cell Death Differ 2011; 18: 571–580.18. Mizushima N, Yamamoto A, Hatano M, et al . Dissection of

autophagosome formation using Apg5-deficient mouse embryonicstem cells. J Cell Biol 2001; 152: 657–668.

19. Noda T, Ohsumi Y. Tor, a phosphatidylinositol kinase homo-logue, controls autophagy in yeast. J Biol Chem 1998; 273:3963–3966.

20. Hosokawa N, Hara T, Kaizuka T, et al . Nutrient-dependentmTORC1 association with the ULK1–Atg13–FIP200 complexrequired for autophagy. Mol Biol Cell 2009; 20: 1981–1991.

21. Ganley IG, Lam du H, Wang J, et al . ULK1.ATG13.FIP200 com-plex mediates mTOR signaling and is essential for autophagy.J Biol Chem 2009; 284: 12297–12305.

22. Settembre C, Di Malta C, Polito VA, et al . TFEB links autophagyto lysosomal biogenesis. Science 2011; 332: 1429–1433.

23. Scott RC, Schuldiner O, Neufeld TP. Role and regulation ofstarvation-induced autophagy in the Drosophila fat body. Dev

Cell 2004; 7: 167–178.24. Melendez A, Talloczy Z, Seaman M, et al . Autophagy genes

are essential for dauer development and life-span extension inC. elegans. Science 2003; 301: 1387–1391.

25. Otto GP, Wu MY, Kazgan N, et al . Macroautophagy is requiredfor multicellular development of the social amoeba Dictyostelium

discoideum . J Biol Chem 2003; 278: 17636–17645.26. Yoshimoto K, Hanaoka H, Sato S, et al . Processing of ATG8s,

ubiquitin-like proteins, and their deconjugation by ATG4s areessential for plant autophagy. Plant Cell 2004; 16: 2967–2983.

27. Alvarez VE, Kosec G, Sant’Anna C, et al . Autophagy is involvedin nutritional stress response and differentiation in Trypanosoma

cruzi . J Biol Chem 2008; 283: 3454–3464.28. Itakura E, Mizushima N. Characterization of autophagosome for-

mation site by a hierarchical analysis of mammalian Atg proteins.Autophagy 2010; 6: 764–776.

29. Mijaljica D, Prescott M, Devenish RJ. Microautophagy in mam-malian cells: revisiting a 40 year-old conundrum. Autophagy

2011; 7: 673–682.

30. Tuttle DL, Lewin AS, Dunn WA, Jr. Selective autophagy ofperoxisomes in methylotrophic yeasts. Eur J Cell Biol 1993; 60:283–290.

31. Sahu R, Kaushik S, Cannizzo E, et al . Microautohagy of cytoso-lic proteins by late endosomes. Dev Cell 2011; 20:131–139.

32. Goldman SJ, Taylor R, Zhang Y, et al . Autophagy and the degra-dation of mitochondria. Mitochondrion 2010; 10: 309–315.

33. Tolkovsky AM. Mitophagy. Biochim Biophys Acta 2009; 1793:1508–1515.

34. Farre JC, Krick R, Subramani S, et al . Turnover of organelles byautophagy in yeast. Curr Opin Cell Biol 2009; 21: 522–530.

35. Veenhuis M, Salomons FA, Van Der Klei IJ. Peroxisome biogene-sis and degradation in yeast: a structure/function analysis. Microsc

Res Tech 2000; 51: 584–600.36. Bernales S, McDonald KL, Walter P. Autophagy counterbalances

endoplasmic reticulum expansion during the unfolded proteinresponse. PLoS Biol 2006; 4: e423.

37. Hamasaki M, Noda T, Baba M, et al . Starvation triggers thedelivery of the endoplasmic reticulum to the vacuole viaautophagy in yeast. Traffic 2005; 6: 56–65.

38. Singh R, Kaushik S, Wang Y, et al . Autophagy regulates lipidmetabolism. Nature 2009; 458: 1131–1135.

39. Kraft C, Deplazes A, Sohrmann M, et al . Mature ribosomes areselectively degraded upon starvation by an autophagy pathwayrequiring the Ubp3p/Bre5p ubiquitin protease. Nat Cell Biol 2008;10: 602–610.

40. Yamamoto A, Simonsen A. The elimination of accumulated andaggregated proteins: a role for aggrephagy in neurodegeneration.Neurobiol Dis 2011; 43: 17–28.

41. Krick R, Muhe Y, Prick T, et al . Piecemeal microautophagy ofthe nucleus: genetic and morphological traits. Autophagy 2009;5: 270–272.

42. Roberts P, Moshitch-Moshkovitz S, Kvam E, et al . Piecemealmicroautophagy of nucleus in Saccharomyces cerevisiae. Mol Biol

Cell 2003; 14: 129–141.43. Kotoulas OB, Kalamidas SA, Kondomerkos DJ. Glycogen

autophagy in glucose homeostasis. Pathol Res Pract 2006; 202:631–638.

44. Lamark T, Kirkin V, Dikic I, et al . NBR1 and p62 as cargoreceptors for selective autophagy of ubiquitinated targets. Cell

Cycle 2009; 8: 1986–1990.45. Schweers RL, Zhang J, Randall MS, et al . NIX is required for

programmed mitochondrial clearance during reticulocyte matura-tion. Proc Natl Acad Sci USA 2007; 104: 19500–19505.

46. Kirkin V, Lamark T, Johansen T, et al . NBR1 cooperates withp62 in selective autophagy of ubiquitinated targets. Autophagy

2009; 5: 732–733.47. Pankiv S, Clausen TH, Lamark T, et al . p62/SQSTM1 binds

directly to Atg8/LC3 to facilitate degradation of ubiquitinatedprotein aggregates by autophagy. J Biol Chem 2007; 282:24131–24145.

48. Arndt V, Dick N, Tawo R, et al . Chaperone-assisted selectiveautophagy is essential for muscle maintenance. Curr Biol 2010;20: 143–148.

49. Kaushik S, Bandyopadhyay U, Sridhar S, et al . Chaperone-mediated autophagy at a glance. J Cell Sci 2011; 124:495–499.

50. Agarraberes F, Terlecky S, Dice J. An intralysosomal hsp70 isrequired for a selective pathway of lysosomal protein degradation.J Cell Biol 1997; 137: 825–834.

51. Bandyopadhyay U, Kaushik S, Varticovski L, et al . Thechaperone-mediated autophagy receptor organizes in dynamicprotein complexes at the lysosomal membrane. Mol Cell Biol

2008; 28: 5747–5763.

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

The pathophysiology of autophagy 269

52. Bandyopadhyay U, Sridhar S, Kaushik S, et al . Identification ofregulators of chaperone-mediated autophagy. Mol Cell 2010; 39:535–547.

53. Cuervo AM, Dice JF. A receptor for the selective uptake anddegradation of proteins by lysosomes. Science 1996; 273:501–503.

54. Orenstein SJ, Cuervo AM. Chaperone-mediated autophagy:molecular mechanisms and physiological relevance. Semin Cell

Dev Biol 2010; 21: 719–726.55. Mizushima N, Yamamoto A, Matsui M, et al . In vivo analysis of

autophagy in response to nutrient starvation using transgenic miceexpressing a fluorescent autophagosome marker. Mol Biol Cell

2004; 15: 1101–1111.56. Harris RA, Goodwin GW, Paxton R, et al . Nutritional and hor-

monal regulation of the activity state of hepatic branched-chainα-keto acid dehydrogenase complex. Ann N Y Acad Sci 1989; 573:306–313.

57. Pursiheimo JP, Rantanen K, Heikkinen PT, et al . Hypoxia-activated autophagy accelerates degradation of SQSTM1/p62.Oncogene 2009; 28: 334–344.

58. Mizushima N, Klionsky DJ. Protein turnover via autophagy:implications for metabolism. Annu Rev Nutr 2007; 27:19–40.

59. Moriwaki H, Miwa Y, Tajika M, et al . Branched-chain aminoacids as a protein and energy source in liver cirrhosis. Biochem

Biophys Res Commun 2004; 313: 405–409.60. Komatsu M, Waguri S, Ueno T, et al . Impairment of starvation-

induced and constitutive autophagy in Atg7-deficient mice. J Cell

Biol 2005; 169: 425–434.61. Kuma A, Hatano M, Matsui M, et al . The role of autophagy

during the early neonatal starvation period. Nature 2004; 432:1032–1036.

62. Lum JJ, Bauer DE, Kong M, et al . Growth factor regulation ofautophagy and cell survival in the absence of apoptosis. Cell

2005; 120: 237–248.63. Tyedmers J, Mogk A, Bukau B. Cellular strategies for control-

ling protein aggregation. Nat Rev Mol Cell Biol 2010; 11:777–788.

64. Douglas PM, Summers DW, Cyr DM. Molecular chaperonesantagonize proteotoxicity by differentially modulating proteinaggregation pathways. Prion 2009; 3: 51–58.

65. Morimoto RI. Proteotoxic stress and inducible chaperone net-works in neurodegenerative disease and aging. Genes Dev 2008;22: 1427–1438.

66. Wong E, Cuervo AM. Autophagy gone awry in neurodegenera-tive diseases. Nat Neurosci 2010; 13: 805–811.

67. Menzies FM, Moreau K, Rubinsztein DC. Protein misfoldingdisorders and macroautophagy. Curr Opin Cell Biol 2011; 23:190–197.

68. Hara T, Nakamura K, Matsui M, et al . Suppression of basalautophagy in neural cells causes neurodegenerative disease inmice. Nature 2006; 441: 885–889.

69. Komatsu M, Waguri S, Chiba T, et al . Loss of autophagy in thecentral nervous system causes neurodegeneration in mice. Nature

2006; 441: 880–884.70. Schutt F, Bergmann M, Holz FG, et al . Proteins modified by

malondialdehyde, 4-hydroxynonenal, or advanced glycation endproducts in lipofuscin of human retinal pigment epithelium. Invest

Ophthalmol Vis Sci 2003; 44: 3663–3668.71. Dohm GL, Tapscott EB, Kasperek GJ. Protein degradation during

endurance exercise and recovery. Med Sci Sports Exerc 1987; 19:S166–171.

72. Dewaele M, Maes H, Agostinis P. ROS-mediated mechanismsof autophagy stimulation and their relevance in cancer therapy.Autophagy 2010; 6: 838–854.

73. Kiffin R, Christian C, Knecht E, et al . Activation of chaperone-

mediated autophagy during oxidative stress. Mol Biol Cell 2004;

15: 4829–4840.

74. Ravikumar B, Duden R, Rubinsztein DC. Aggregate-prone pro-

teins with polyglutamine and polyalanine expansions are degraded

by autophagy. Human Mol Genet 2002; 11: 1107–1117.

75. Sarkar S, Ravikumar B, Rubinsztein DC. Autophagic clearance

of aggregate-prone proteins associated with neurodegeneration.

Methods Enzymol 2009; 453: 83–110.

76. Moreau K, Luo S, Rubinsztein DC. Cytoprotective roles for

autophagy. Curr Opin Cell Biol 2010; 22: 206–211.

77. Arrasate M, Mitra S, Schweitzer ES, et al . Inclusion body forma-

tion reduces levels of mutant huntingtin and the risk of neuronal

death. Nature 2004; 431: 805–810.

78. Carew JS, Nawrocki ST, Kahue CN, et al . Targeting autophagy

augments the anticancer activity of the histone deacetylase

inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance.

Blood 2007; 110: 313–322.

79. Tolkovsky AM, Xue L, Fletcher GC, et al . Mitochondrial dis-

appearance from cells: a clue to the role of autophagy in pro-

grammed cell death and disease? Biochimie 2002; 84: 233–240.

80. Iwata J, Ezaki J, Komatsu M, et al . Excess peroxisomes are

degraded by autophagic machinery in mammals. J Biol Chem

2006; 281: 4035–4041.

81. Mukaiyama H, Kajiwara S, Hosomi A, et al . Autophagy-

deficient Schizosaccharomyces pombe mutants undergo partial

sporulation during nitrogen starvation. Microbiology 2009; 155:

3816–3826.

82. Berry DL, Baehrecke EH. Growth arrest and autophagy are

required for salivary gland cell degradation in Drosophila . Cell

2007; 131: 1137–1148.

83. Tsukamoto S, Kuma A, Murakami M, et al . Autophagy is essen-

tial for preimplantation development of mouse embryos. Science

2008; 321: 117–120.

84. Mortensen M, Ferguson DJ, Edelmann M, et al . Loss of

autophagy in erythroid cells leads to defective removal of mito-

chondria and severe anemia in vivo. Proc Natl Acad Sci USA

2010; 107: 832–837.

85. Sandoval H, Thiagarajan P, Dasgupta SK, et al . Essential role for

Nix in autophagic maturation of erythroid cells. Nature 2008; 454:

232–235.

86. Takano-Ohmuro H, Mukaida M, Kominami E, et al . Autophagy

in embryonic erythroid cells: its role in maturation. Eur J Cell

Biol 2000; 79: 759–764.

87. Pua HH, Guo J, Komatsu M, et al . Autophagy is essential for

mitochondrial clearance in mature T lymphocytes. J Immunol

2009; 182: 4046–4055.

88. Pua HH, He YW. Mitophagy in the little lymphocytes: an essen-

tial role for autophagy in mitochondrial clearance in T lympho-

cytes. Autophagy 2009; 5: 745–746.

89. Singh R, Xiang Y, Wang Y, et al . Autophagy regulates adipose

mass and differentiation in mice. J Clin Invest 2009; 119:

3329–3339.

90. Zhang Y, Goldman S, Baerga R, et al . Adipose-specific deletion

of autophagy-related gene 7 (atg7 ) in mice reveals a role in

adipogenesis. Proc Natl Acad Sci USA 2009; 106: 19860–19865.

91. Denton D, Shravage B, Simin R, et al . Autophagy, not apoptosis,

is essential for midgut cell death in Drosophila . Curr Biol 2009;

19: 1741–1746.

92. Grishchuk Y, Ginet V, Truttmann AC, et al . Beclin 1-independent

autophagy contributes to apoptosis in cortical neurons. Autophagy

2011; 7: 1115–1131.

93. Nishiyama J, Matsuda K, Kakegawa W, et al . Reevaluation of

neurodegeneration in lurcher mice: constitutive ion fluxes cause

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

270 S Sridhar et al

cell death with, not by, autophagy. J Neurosci 2010; 30:

2177–2187.

94. Amaravadi RK, Yu D, Lum JJ, et al . Autophagy inhibition

enhances therapy-induced apoptosis in a Myc-induced model of

lymphoma. J Clin Invest 2007; 117: 326–336.

95. Debnath J, Baehrecke EH, Kroemer G. Does autophagy con-

tribute to cell death? Autophagy 2005; 1: 66–74.

96. Uchiyama Y. Autophagic cell death and its execution by lysoso-

mal cathepsins. Arch Histol Cytol 2001; 64: 233–246.

97. Gorski SM, Chittaranjan S, Pleasance ED, et al . A SAGE

approach to discovery of genes involved in autophagic cell death.

Curr Biol 2003; 13: 358–363.

98. Levine B, Sinha S, Kroemer G. Bcl-2 family members: dual

regulators of apoptosis and autophagy. Autophagy 2008; 4:

600–606.

99. Liang XH, Jackson S, Seaman M, et al . Induction of autophagy

and inhibition of tumorigenesis by beclin 1. Nature 1999; 402:

672–676.

100. Qu X, Yu J, Bhagat G, et al . Promotion of tumorigenesis by

heterozygous disruption of the beclin 1 autophagy gene. J Clin

Invest 2003; 112: 1809–1820.

101. Yue Z, Jin S, Yang C, et al . Beclin 1, an autophagy gene essential

for early embryonic development, is a haploinsufficient tumor

suppressor. Proc Natl Acad Sci USA 2003; 100: 15077–15082.

102. Takahashi Y, Coppola D, Matsushita N, et al . Bif-1 interacts

with Beclin 1 through UVRAG and regulates autophagy and

tumorigenesis. Nat Cell Biol 2007; 9: 1142–1151.

103. Liang C, Feng P, Ku B, et al . Autophagic and tumour suppressor

activity of a novel Beclin1-binding protein UVRAG. Nat Cell Biol

2006; 8: 688–699.

104. Marino G, Salvador-Montoliu N, Fueyo A, et al . Tissue-specific

autophagy alterations and increased tumorigenesis in mice

deficient in Atg4C/autophagin-3. J Biol Chem 2007; 282:

18573–18583.

105. Cheng HC, Kim SR, Oo TF, et al . Akt suppresses retrograde

degeneration of dopaminergic axons by inhibition of macroau-

tophagy. J Neurosci 2011; 31: 2125–2135.

106. Scott RC, Juhasz G, Neufeld TP. Direct induction of autophagy

by Atg1 inhibits cell growth and induces apoptotic cell death.

Curr Biol 2007; 17: 1–11.

107. Koneri K, Goi T, Hirono Y, et al . Beclin 1 gene inhibits tumor

growth in colon cancer cell lines. Anticancer Res 2007; 27:

1453–1457.

108. Degenhardt K, Mathew R, Beaudoin B, et al . Autophagy pro-

motes tumor cell survival and restricts necrosis, inflammation,

and tumorigenesis. Cancer Cell 2006; 10: 51–64.

109. Mathew R, Karp CM, Beaudoin B, et al . Autophagy suppresses

tumorigenesis through elimination of p62. Cell 2009; 137:

1062–1075.

110. Mathew R, Kongara S, Beaudoin B, et al . Autophagy suppresses

tumor progression by limiting chromosomal instability. Genes

Dev 2007; 21: 1367–1381.

111. White E, Karp C, Strohecker AM, et al . Role of autophagy in

suppression of inflammation and cancer. Curr Opin Cell Biol

2010; 22: 212–217.

112. Balkwill F, Charles KA, Mantovani A. Smoldering and polarized

inflammation in the initiation and promotion of malignant disease.

Cancer Cell 2005; 7: 211–217.

113. Mantovani A, Allavena P, Sica A, et al . Cancer-related inflam-

mation. Nature 2008; 454: 436–444.

114. Folkman J. Angiogenesis and apoptosis. Semin Cancer Biol 2003;

13: 159–167.

115. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;

100: 57–70.

116. Jin S, White E. Role of autophagy in cancer: management ofmetabolic stress. Autophagy 2007; 3: 28–31.

117. Koppenol WH, Bounds PL, Dang CV. Otto Warburg’s contribu-tions to current concepts of cancer metabolism. Nat Rev Cancer

2011; 11: 325–337.118. Warburg O. On respiratory impairment in cancer cells. Science

1956; 124: 269–270.119. Thompson CB. Metabolic enzymes as oncogenes or tumor sup-

pressors. N Engl J Med 2009; 360: 813–815.120. Selak MA, Armour SM, MacKenzie ED, et al . Succinate links

TCA cycle dysfunction to oncogenesis by inhibiting HIF-α prolylhydroxylase. Cancer Cell 2005; 7: 77–85.

121. Lock R, Roy S, Kenific CM, et al . Autophagy facilitates glycoly-sis during Ras-mediated oncogenic transformation. Mol Biol Cell

2011; 22: 165–178.122. Martinez-Outschoorn UE, Pavlides S, Whitaker-Menezes D,

et al . Tumor cells induce the cancer associated fibroblast pheno-type via caveolin-1 degradation: implications for breast cancerand DCIS therapy with autophagy inhibitors. Cell Cycle 2010; 9:2423–2433.

123. Guo JY, Chen HY, Mathew R, et al . Activated Ras requiresautophagy to maintain oxidative metabolism and tumorigenesis.Genes Dev 2011; 25: 460–470.

124. Nelson DA, Tan TT, Rabson AB, et al . Hypoxia and defectiveapoptosis drive genomic instability and tumorigenesis. Genes Dev

2004; 18: 2095–2107.125. Kim JH, Kim HY, Lee YK, et al . Involvement of mitophagy in

oncogenic K-Ras-induced transformation: overcoming a cellularenergy deficit from glucose deficiency. Autophagy 2011; 7:1187–1198.

126. Komatsu M, Kurokawa H, Waguri S, et al . The selectiveautophagy substrate p62 activates the stress responsive transcrip-tion factor Nrf2 through inactivation of Keap1. Nat Cell Biol

2010; 12: 213–223.127. Lv L, Li D, Zhao D, et al . Acetylation targets the M2 Isoform

of pyruvate kinase for degradation through chaperone-mediatedautophagy and promotes tumor growth. Mol Cell 2011; 42:719–730.

128. Cuervo AM, Dice JF. Lysosomes, a meeting point of proteins,chaperones, and proteases. J Mol Med 1998; 76: 6–12.

129. Ding ZB, Hui B, Shi YH, et al . Autophagy activation in hepato-cellular carcinoma contributes to the tolerance of oxaliplatin viareactive oxygen species modulation. Clin Cancer Res 2011; 17:6229–6238.

130. Yang L, Yu Y, Kang R, et al . Up-regulated autophagy by endoge-nous HMGB1 promotes chemoresistance in leukemia cells.Leukemia Lymphoma 2011; (in press).

131. Shi YH, Ding ZB, Zhou J, et al . Targeting autophagy enhancessorafenib lethality for hepatocellular carcinoma via ER stress-related apoptosis. Autophagy 2011; 7: 1159–1172.

132. Sun WL, Chen J, Wang YP, et al . Autophagy protects breastcancer cells from epirubicin-induced apoptosis and facili-tates epirubicin-resistance development. Autophagy 2011; 7:1035–1044.

133. Li J, Hou N, Faried A, et al . Inhibition of autophagy augments5-fluorouracil chemotherapy in human colon cancer in vitro andin vivo model. Eur J Cancer 2010; 46: 1900–1909.

134. Cerquetti L, Sampaoli C, Amendola D, et al . Rosiglitazoneinduces autophagy in H295R and cell cycle deregulation in SW13adrenocortical cancer cells. Exp Cell Res 2011; 317: 1397–1410.

135. Griffin C, McNulty J, Pandey S. Pancratistatin induces apoptosisand autophagy in metastatic prostate cancer cells. Int J Oncol

2011; 38: 1549–1556.136. Lin CI, Whang EE, Donner DB, et al . Autophagy induction with

RAD001 enhances chemosensitivity and radiosensitivity through

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

The pathophysiology of autophagy 271

Met inhibition in papillary thyroid cancer. Mol Cancer Res MCR

2010; 8: 1217–1226.

137. Toepfer N, Childress C, Parikh A, et al . Atorvastatin induces

autophagy in prostate cancer PC3 cells through activation of LC3

transcription. Cancer Biol Ther 2011; 12: 691–699.

138. Zou CF, Jia L, Jin H, et al . Re-expression of ARHI (DIRAS3)

induces autophagy in breast cancer cells and enhances the

inhibitory effect of paclitaxel. BMC Cancer 2011; 11: 22.

139. Pimkina J, Murphy ME. ARF, autophagy and tumor suppression.

Autophagy 2009; 5: 397–399.

140. Amaravadi RK, Lippincott-Schwartz J, Yin X-M, et al . Princi-

ples and current strategies for targeting autophagy for cancer

treatment. Clin Cancer Res 2011; 17: 654–666.

141. Maclean KH, Dorsey FC, Cleveland JL, et al . Targeting lysoso-

mal degradation induces p53-dependent cell death and prevents

cancer in mouse models of lymphomagenesis. J Clin Invest 2008;

118: 79–88.

142. Alirezaei M, Kemball CC, Flynn CT, et al . Short-term fast-

ing induces profound neuronal autophagy. Autophagy 2010; 6:

702–710.

143. Boland B, Kumar A, Lee S, et al . Autophagy induction and

autophagosome clearance in neurons: relationship to autophagic

pathology in Alzheimer’s disease. J Neurosci 2008; 28:

6926–6937.

144. Young JE, Martinez RA, La Spada AR. Nutrient deprivation

induces neuronal autophagy and implicates reduced insulin signal-

ing in neuroprotective autophagy activation. J Biol Chem 2009;

284: 2363–2373.

145. Rodriguez-Muela N, Germain F, Marino G, et al . Autophagy

promotes survival of retinal ganglion cells after optic nerve

axotomy in mice. Cell Death Differ 2011; (in press).

146. Midorikawa R, Yamamoto-Hino M, Awano W, et al . Autophagy-

dependent rhodopsin degradation prevents retinal degeneration in

Drosophila . J Neurosci 2010; 30: 10703–10719.

147. Piras A, Gianetto D, Conte D, et al . Activation of autophagy in a

rat model of retinal ischemia following high intraocular pressure.

PLoS One 2011; 6: e22514.

148. Liu C, Gao Y, Barrett J, et al . Autophagy and protein aggregation

after brain ischemia. J Neurochem 2010; 115: 68–78.

149. Russo R, Berliocchi L, Adornetto A, et al . Calpain-mediated

cleavage of Beclin-1 and autophagy deregulation following retinal

ischemic injury in vivo. Cell Death Dis 2011; 2: e144.

150. Kubota C, Torii S, Hou N, et al . Constitutive reactive oxygen

species generation from autophagosome/lysosome in neuronal

oxidative toxicity. J Biol Chem 2010; 285: 667–674.

151. Ravikumar B, Vacher C, Berger Z, et al . Inhibition of mTOR

induces autophagy and reduces toxicity of polyglutamine expan-

sions in fly and mouse models of Huntington disease. Nat Genet

2004; 36: 585–595.

152. Nascimento-Ferreira I, Santos-Ferreira T, Sousa-Ferreira L, et al .

Overexpression of the autophagic beclin-1 protein clears mutant

ataxin-3 and alleviates Machado–Joseph disease. Brain 2011;

134: 1400–1415.

153. Nassif M, Hetz C. Targeting autophagy in ALS: a complex

mission. Autophagy 2011; 7: 450–453.

154. Fleming A, Noda T, Yoshimori T, et al . Chemical modulators of

autophagy as biological probes and potential therapeutics. Nat

Chem Biol 2011; 7: 9–17.

155. Wong ES, Tan JM, Soong WE, et al . Autophagy-mediated clear-

ance of aggresomes is not a universal phenomenon. Hum Mol

Genet 2008; 17: 2570–2582.

156. Lim J, Kim HW, Youdim MB, et al . Binding preference of

p62 towards LC3-ll during dopaminergic neurotoxin-induced

impairment of autophagic flux. Autophagy 2011; 7: 51–60.

157. Winslow AR, Chen CW, Corrochano S, et al . α-Synucleinimpairs macroautophagy: implications for Parkinson’s disease.J Cell Biol 2010; 190: 1023–1037.

158. Martinez-Vicente M, Talloczy Z, Wong E, et al . Cargo recogni-tion failure is responsible for inefficient autophagy in Hunting-ton’s disease. Nat Neursci 2010; 13: 567–576.

159. Ravikumar B, Acevedo-Arozena A, Imarisio S, et al . Dyneinmutations impair autophagic clearance of aggregate-prone pro-teins. Nat Genet 2005; 37: 771–776.

160. Lee JY, Koga H, Kawaguchi Y, et al . HDAC6 controlsautophagosome maturation essential for ubiquitin-selectivequality-control autophagy. EMBO J 2010; 29: 969–980.

161. Settembre C, Fraldi A, Jahreiss L, et al . A block of autophagy inlysosomal storage disorders. Hum Mol Genet 2008; 17: 119–129.

162. Lee J-H, Yu W, Kumar A, et al . PS1 mutations in Alzheimer’sdisease disrupt lysosomal proteolysis and autophagy. Cell 2010;7: 1146–1158.

163. Koga H, Cuervo AM. Chaperone-mediated autophagy dysfunc-tion in the pathogenesis of neurodegeneration. Neurobiol Dis

2011; 43: 29–37.164. Cuervo AM, Stefanis L, Fredenburg R, et al . Impaired degrada-

tion of mutant α-synuclein by chaperone-mediated autophagy.Science 2004; 305: 1292–1295.

165. Martinez-Vicente M, Talloczy Z, Kaushik S, et al . Dopamine-modified α-synuclein blocks chaperone-mediated autophagy.J Clin Invest 2008; 118: 777–788.

166. Kabuta T, Furuta A, Aoki S, et al . Aberrant interaction betweenParkinson disease-associated mutant UCH-L1 and the lysosomalreceptor for chaperone-mediated autophagy. J Biol Chem 2008;283: 23731–23738.

167. Wang Y, Martinez-Vicente M, Kruger U, et al . Tau fragmenta-tion, aggregation and clearance: the dual role of lysosomal pro-cessing. Hum Mol Genet 2009; 18: 4153–4170.

168. Yu W, Cuervo A, Kumar A, et al . Macroautophagy—a novelβ-amyloid peptide-generating pathway activated in Alzheimer’sdisease. J Cell Biol 2005; 171: 87–98.

169. Lee JH, Yu WH, Kumar A, et al . Lysosomal proteolysis andautophagy require Presenilin 1 and are disrupted by Alzheimer-related PS1 mutations. Cell 2010; 141: 1146–1191.

170. Stefanis L, Larsen K, Rideout H, et al . Expression of A53Tmutant but not wild-type α-synuclein in PC12 cells inducesalterations of the ubiquitin-dependent degradation system, lossof dopamine release, and autophagic cell death. J Neurosci 2001;21: 9549–9560.

171. Webb J, Ravikumar B, Atkins J, et al . α-Synuclein is degradedby both autophagy and the proteasome. J Biol Chem 2003; 278:25009–25013.

172. Geisler S, Holmstrom KM, Skujat D, et al . PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1.Nat Cell Biol 2010; 12: 119–131.

173. Iwata A, Christianson JC, Bucci M, et al . Increased suscepti-bility of cytoplasmic over nuclear polyglutamine aggregates toautophagic degradation. Proc Natl Acad Sci USA 2005; 102:13135–13140.

174. Jeong H, Then F, Melia TJ Jr, et al . Acetylation targets mutanthuntingtin to autophagosomes for degradation. Cell 2009; 137:60–72.

175. Thompson LM, Aiken CT, Kaltenbach LS, et al . IKK phospho-rylates huntingtin and targets it for degradation by the proteasomeand lysosome. J Cell Biol 2009; 187: 1083–1099.

176. Iwata A, Riley BE, Johnston JA, et al . HDAC6 and microtubulesare required for autophagic degradation of aggregated huntingtin.J Biol Chem 2005; 280: 40282–40292.

177. Bauer PO, Goswami A, Wong HK, et al . Harnessingchaperone-mediated autophagy for the selective degradation

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

272 S Sridhar et al

of mutant huntingtin protein. Nat Biotechnol 2010; 28:

256–263.

178. Cao DJ, Gillette TG, Hill JA. Cardiomyocyte autophagy: remod-

eling, repairing, and reconstructing the heart. Curr Hypert Rep

2009; 11: 406–411.

179. Hill JA, Olson EN. Cardiac plasticity. N Engl J Med 2008; 358:

1370–1380.

180. Saftig P, Tanaka Y, Lullmann-Rauch R, et al . Disease model:

LAMP-2 enlightens Danon disease. Trends Mol Med 2001; 7:

37–39.

181. Tanaka Y, Guhde G, Suter A, et al . Accumulation of autophagic

vacuoles and cardiomyopathy in LAMP-2-deficient mice. Nature

2000; 406: 902–906.

182. Lacoste-Collin L, Garcia V, Uro-Coste E, et al . Danon’s disease

(X-linked vacuolar cardiomyopathy and myopathy): a case with

a novel Lamp-2 gene mutation. Neuromuscul Disord 2002; 12:

882–885.

183. Nakai A, Yamaguchi O, Takeda T, et al . The role of autophagy in

cardiomyocytes in the basal state and in response to hemodynamic

stress. Nat Med 2007; 13: 619–624.

184. Taneike M, Yamaguchi O, Nakai A, et al . Inhibition of

autophagy in the heart induces age-related cardiomyopathy.

Autophagy 2010; 6: 600–606.

185. Wang X, Osinska H, Klevitsky R, et al . Expression of R120G-

αB-crystallin causes aberrant desmin and αB-crystallin aggrega-

tion and cardiomyopathy in mice. Circ Res 2001; 89: 84–91.

186. Tannous P, Zhu H, Johnstone JL, et al . Autophagy is an adaptive

response in desmin-related cardiomyopathy. Proc Natl Acad Sci

USA 2008; 105: 9745–9750.

187. Gurusamy N, Lekli I, Gorbunov NV, et al . Cardioprotection by

adaptation to ischaemia augments autophagy in association with

BAG-1 protein. J Cell Mol Med 2009; 13: 373–387.

188. Yan L, Vatner DE, Kim SJ, et al . Autophagy in chronically

ischemic myocardium. Proc Natl Acad Sci USA 2005; 102:

13807–13812.

189. Hamacher-Brady A, Brady NR, Gottlieb RA. Enhancing

macroautophagy protects against ischemia/reperfusion injury in

cardiac myocytes. J Biol Chem 2006; 281: 29776–29787.

190. Huang C, Yitzhaki S, Perry CN, et al . Autophagy induced

by ischemic preconditioning is essential for cardioprotection.

J Cardiovasc Transl Res 2010; 3: 365–373.

191. Yitzhaki S, Huang C, Liu W, et al . Autophagy is required for

preconditioning by the adenosine A1 receptor-selective agonist

CCPA. Basic Res Cardiol 2009; 104: 157–167.

192. Huang C, Andres AM, Ratliff EP, et al . Preconditioning involves

selective mitophagy mediated by Parkin and p62/SQSTM1. PLoS

One 2011; 6: e20975.

193. Dorn GW, 2nd. Mitochondrial pruning by Nix and BNip3: an

essential function for cardiac-expressed death factors. J Cardio-

vasc Transl Res 2010; 3: 374–383.

194. Valentim L, Laurence KM, Townsend PA, et al . Urocortin

inhibits Beclin1-mediated autophagic cell death in cardiac

myocytes exposed to ischaemia/reperfusion injury. J Mol Cell

Cardiol 2006; 40: 846–852.

195. Matsui Y, Takagi H, Qu X, et al . Distinct roles of autophagy in

the heart during ischemia and reperfusion: roles of AMP-activated

protein kinase and Beclin 1 in mediating autophagy. Circ Res

2007; 100: 914–922.

196. Zhu H, Tannous P, Johnstone JL, et al . Cardiac autophagy is a

maladaptive response to hemodynamic stress. J Clin Invest 2007;

117: 1782–1793.

197. Morselli E, Galluzzi L, Kepp O, et al . Autophagy mediates phar-

macological lifespan extension by spermidine and resveratrol.

Aging 2009; 1: 961–970.

198. Das M, Das DK. Resveratrol and cardiovascular health. Mol

Aspects Med 2010; 31: 503–512.

199. Lekli I, Ray D, Mukherjee S, et al . Co-ordinated autophagy with

resveratrol and gamma-tocotrienol confers synergetic cardiopro-

tection. J Cell Mol Med 2010; 14: 2506–2518.

200. Wing SS, Chiang HL, Goldberg AL, et al . Proteins containing

peptide sequences related to Lys–Phe–Glu–Arg–Gln are selec-

tively depleted in liver and heart, but not skeletal muscle, of fasted

rats. Biochem J 1991; 275: 165–169.

201. Eskelinen EL, Cuervo AM, Taylor MR, et al . Unifying nomen-

clature for the isoforms of the lysosomal membrane protein

LAMP-2. Traffic 2005; 6: 1058–1061.

202. Massey AC, Kaushik S, Sovak G, et al . Consequences of the

selective blockage of chaperone-mediated autophagy. Proc Natl

Acad Sci USA 2006; 103: 5805–5810.

203. Deretic V. Autophagy in immunity and cell-autonomous defense

against intracellular microbes. Immunol Rev 2011; 240: 92–104.

204. Nakagawa I, Amano A, Mizushima N, et al . Autophagy defends

cells against invading group A Streptococcus . Science 2004; 306:

1037–1040.

205. Blanchet FP, Moris A, Nikolic DS, et al . Human immunodefi-

ciency virus-1 inhibition of immunoamphisomes in dendritic cells

impairs early innate and adaptive immune responses. Immunity

2010; 32: 654–669.

206. Shelly S, Lukinova N, Bambina S, et al . Autophagy is an essen-

tial component of Drosophila immunity against vesicular stom-

atitis virus. Immunity 2009; 30: 588–598.

207. Orvedahl A, Alexander D, Talloczy Z, et al . HSV-1 ICP34.5

confers neurovirulence by targeting the Beclin 1 autophagy

protein. Cell Host Microbe 2007; 1: 23–35.

208. Rich KA, Burkett C, Webster P. Cytoplasmic bacteria can be

targets for autophagy. Cell Microbiol 2003; 5: 455–468.

209. Gutierrez MG, Master SS, Singh SB, et al . Autophagy is a

defense mechanism inhibiting BCG and Mycobacterium tubercu-

losis survival in infected macrophages. Cell 2004; 119: 753–766.

210. Sanjuan MA, Dillon CP, Tait SW, et al . Toll-like receptor sig-

nalling in macrophages links the autophagy pathway to phagocy-

tosis. Nature 2007; 450: 1253–1257.

211. Dorn BR, Dunn WA, Jr., Progulske-Fox A. Bacterial interactions

with the autophagic pathway. Cell Microbiol 2002; 4: 1–10.

212. Colombo MI. Autophagy: a pathogen driven process. IUBMB Life

2007; 59: 238–242.

213. Gutierrez MG, Vazquez CL, Munafo DB, et al . Autophagy

induction favours the generation and maturation of the Coxiella-

replicative vacuoles. Cell Microbiol 2005; 7: 981–993.

214. Dorn BR, Dunn WA, Jr., Progulske-Fox A. Porphyromonas gin-

givalis traffics to autophagosomes in human coronary artery

endothelial cells. Infect Immun 2001; 69: 5698–5708.

215. Mestre MB, Fader CM, Sola C, et al . α-Hemolysin is required

for the activation of the autophagic pathway in Staphylococcus

aureus-infected cells. Autophagy 2010; 6: 110–125.

216. Ogawa M, Yoshimori T, Suzuki T, et al . Escape of intracellular

Shigella from autophagy. Science 2005; 307: 727–731.

217. Kirkegaard K, Taylor MP, Jackson WT. Cellular autophagy: sur-

render, avoidance and subversion by microorganisms. Nat Rev

Microbiol 2004; 2: 301–314.

218. Munz C. Autophagy and antigen presentation. Cell Microbiol

2006; 8: 891–898.

219. Levine B, Deretic V. Unveiling the roles of autophagy in innate

and adaptive immunity. Nat Rev Immunol 2007; 7: 767–777.

220. Crotzer VL, Blum JS. Autophagy and its role in MHC-mediated

antigen presentation. J Immunol 2009; 182: 3335–3341.

221. Friede T, Gnau V, Jung G, et al . Natural ligand motifs of closely

related HLA-DR4 molecules predict features of rheumatoid

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com

The pathophysiology of autophagy 273

arthritis associated peptides. Biochim Biophys Acta 1996; 1316:85–101.

222. Dongre AR, Kovats S, de Roos P, et al . In vivo MHC class IIpresentation of cytosolic proteins revealed by rapid automatedtandem mass spectrometry and functional analyses. Eur J Immunol2001; 31: 1485–1494.

223. Nimmerjahn F, Milosevic S, Behrends U, et al . Major histocom-patibility complex class II-restricted presentation of a cytosolicantigen by autophagy. Eur J Immunol 2003; 33: 1250–1259.

224. Dorfel D, Appel S, Grunebach F, et al . Processing and presen-tation of HLA class I and II epitopes by dendritic cells aftertransfection with in vitro-transcribed MUC1 RNA. Blood 2005;105: 3199–3205.

225. Brazil MI, Weiss S, Stockinger B. Excessive degradation of intra-cellular protein in macrophages prevents presentation in the con-text of major histocompatibility complex class II molecules. EurJ Immunol 1997; 27: 1506–1514.

226. Dengjel J, Schoor O, Fischer R, et al . Autophagy promotes MHCclass II presentation of peptides from intracellular source proteins.Proc Natl Acad Sci USA 2005; 102: 7922–7927.

227. Paludan C, Schmid D, Landthaler M, et al . Endogenous MHCclass II processing of a viral nuclear antigen after autophagy.Science 2005; 307: 593–596.

228. Lee HK, Mattei LM, Steinberg BE, et al . In vivo requirement forAtg5 in antigen presentation by dendritic cells. Immunity 2010;32: 227–239.

229. Zhou D, Li P, Lin Y, et al . Lamp-2a facilitates MHC classII presentation of cytoplasmic antigens. Immunity 2005; 22:571–581.

230. Munz C. Antigen processing via autophagy—not only for MHCclass II presentation anymore? Curr Opin Immunol 2010; 22:89–93.

231. English L, Chemali M, Duron J, et al . Autophagy enhances thepresentation of endogenous viral antigens on MHC class Imolecules during HSV-1 infection. Nat Immunol 2009; 10:480–487.

232. Starr TK, Jameson SC, Hogquist KA. Positive and negative selec-tion of T cells. Annu Rev Immunol 2003; 21: 139–176.

233. Nedjic J, Aichinger M, Emmerich J, et al . Autophagy in thymicepithelium shapes the T-cell repertoire and is essential fortolerance. Nature 2008; 455: 396–400.

234. Schmid D, Pypaert M, Munz C. Antigen-loading compartmentsfor major histocompatibility complex class II molecules contin-uously receive input from autophagosomes. Immunity 2007; 26:79–92.

235. Rioux JD, Xavier RJ, Taylor KD, et al . Genome-wide associationstudy identifies new susceptibility loci for Crohn disease andimplicates autophagy in disease pathogenesis. Nat Genet 2007;39: 596–604.

236. Prescott NJ, Fisher SA, Franke A, et al . A nonsynonymous SNPin ATG16L1 predisposes to ileal Crohn’s disease and is inde-pendent of CARD15 and IBD5. Gastroenterology 2007; 132:1665–1671.

237. Lunemann JD, Munz C. Autophagy in CD4+ T cell immunityand tolerance. Cell Death Differ 2009; 16: 79–86.

238. Qu X, Zou Z, Sun Q, et al . Autophagy gene-dependent clearanceof apoptotic cells during embryonic development. Cell 2007; 128:931–946.

239. Bratton DL, Henson PM. Autoimmunity and apoptosis: refusingto go quietly. Nat Med 2005; 11: 26–27.

240. Jia W, Pua HH, Li QJ, et al . Autophagy regulates endoplasmicreticulum homeostasis and calcium mobilization in T lympho-cytes. J Immunol 2011; 186: 1564–1574.

241. Pua HH, Dzhagalov I, Chuck M, et al . A critical role for theautophagy gene Atg5 in T cell survival and proliferation. J ExpMed 2007; 204: 25–31.

242. Li C, Capan E, Zhao Y, et al . Autophagy is induced in CD4+ Tcells and important for the growth factor-withdrawal cell death.J Immunol 2006; 177: 5163–5168.

243. Hubbard VM, Valdor R, Patel B, et al . Macroautophagy regulatesenergy metabolism during effector T cell activation. J Immunol2010; 185: 7349–7357.

244. Levine B. Eating oneself and uninvited guests: autophagy-relatedpathways in cellular defense. Cell 2005; 120: 159–162.

245. Deretic V, Levine B. Autophagy, immunity, and microbial adap-tations. Cell Host Microbe 2009; 5: 527–549.

Copyright 2011 Pathological Society of Great Britain and Ireland. J Pathol 2012; 226: 255–273Published by John Wiley & Sons, Ltd. www.pathsoc.org.uk www.thejournalofpathology.com