13.07.2015 Views

Autophagy and cancer: Dynamism of the ... - ResearchGate

Autophagy and cancer: Dynamism of the ... - ResearchGate

Autophagy and cancer: Dynamism of the ... - ResearchGate

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

ARTICLE IN PRESS2 K. Tsuchihara et al. / Cancer Letters xxx (2008) xxx–xxxFinally, maturated autophagosomes fuse with <strong>the</strong> lysosome.The engulfed components, as well as <strong>the</strong> inner membrane<strong>of</strong> <strong>the</strong> autophagosome, are degraded by lysosomalhydrolases such as ca<strong>the</strong>psins. Autophagic processes havebeen well characterized in yeast, <strong>and</strong> more than 30autophagy-related genes (ATG) that encode <strong>the</strong> proteinsexecuting autophagy have been identified in <strong>the</strong> field <strong>of</strong>yeast genetics [6]. Similar autophagic machineries havebeen observed in mammalian cells, <strong>and</strong> mammalian orthologsfor ATGs <strong>and</strong> o<strong>the</strong>r autophagy regulating moleculeshave also been identified [7]. The detailed molecularevents involved in <strong>the</strong>se processes are described in ano<strong>the</strong>rcomprehensive review [8].Compared with <strong>the</strong> ubiquitin–proteasome system,which recognizes specific targets for degradation, autophagyhas been thought to engulf cytoplasmic constituentsnon-selectively. Recently, selective sequestration targetingspecific organelles or invasive microbes has also attractedattention [9].A very concise description <strong>of</strong> <strong>the</strong> function <strong>of</strong> autophagyis recycling; reducing waste <strong>and</strong> yielding resources. Damagedorganelles <strong>and</strong> misfolded proteins accumulate insenescent cells or cells under various stresses, such as oxidativestress <strong>and</strong> infection. The autophagic machinery degrades<strong>and</strong> reduces this cellular ‘‘garbage”. This functionis particularly effective in postmitotic cells, like neurons<strong>and</strong> cardiac myocytes, since <strong>the</strong>se cells cannot dilute suchsuperfluous components by undergoing cell division. Basalautophagy is constitutively observed in <strong>the</strong>se postmitoticcells <strong>and</strong> may have an important function for <strong>the</strong> qualitycontrol <strong>of</strong> cellular components [10]. On <strong>the</strong> o<strong>the</strong>r h<strong>and</strong>,self-digested components provide nutrients <strong>and</strong> materialsfor newly constructed cellular structures. The amino acids<strong>and</strong> fatty acids generated by autophagic degradation areused by <strong>the</strong> tricarboxylic acid (TCA) cycle to produce ATP,a main energy source for various cellular events. Methylpyruvate,a membrane-permeable derivative <strong>of</strong> pyruvatethat serves as a substrate for <strong>the</strong> TCA cycle, restored ATPproduction in autophagy-deficient cells. Fur<strong>the</strong>rmore, supplementationwith methylpyruvate rescued <strong>the</strong>se cellsfrom metabolic stress-induced cell death [11].Observations <strong>of</strong> clinical samples <strong>and</strong> animal <strong>and</strong> cellularmodels have suggested a variety <strong>of</strong> physiological <strong>and</strong> pathologicalroles <strong>of</strong> autophagy, such as development, aging,host defense system, neurodegenerative diseases, muscle<strong>and</strong> cardiac diseases <strong>and</strong> <strong>cancer</strong> [12,13]. However, <strong>the</strong> relevance<strong>of</strong> autophagy in tumor formation <strong>and</strong> progression isstill controversial. Although previous findings stronglysuggest that autophagy contributes to sustainable cell survival,anti-tumorigenic roles <strong>of</strong> autophagy have been alsomentioned. Here, we will summarize <strong>and</strong> discuss recentstudies <strong>of</strong> autophagy in <strong>cancer</strong> biology with <strong>the</strong> goal <strong>of</strong>clarifying this issue.2. <strong>Autophagy</strong> protects <strong>cancer</strong> cells from starvationCancer cells in solid tumors obtain <strong>the</strong>ir necessarynutrients from blood flow. Aberrantly proliferating <strong>cancer</strong>cells may have high bioenergetic dem<strong>and</strong>s <strong>and</strong> requiremore nutrients than non-<strong>cancer</strong>ous cells. Tumor angiogenesisis a reasonable way to increase blood flow. During <strong>the</strong>initial phase <strong>of</strong> tumorigenesis, tumor vessels have not yetbeen induced <strong>and</strong> <strong>the</strong> nutritional dem<strong>and</strong>s <strong>of</strong> tumor cellsare likely to surpass <strong>the</strong> supply from normal vasculature.Moreover, even after tumor vessels have been established,<strong>the</strong> oxygen tension <strong>and</strong> glucose concentration in locallyadvanced tumors remain at a low level. This suggests that<strong>the</strong> tumor microvasculature is functionally <strong>and</strong> structurallyimmature to support sufficient blood flow [14–16].Or, even once functionally <strong>and</strong> structurally adequate tumorvessels were established, soon or later, <strong>the</strong> balance <strong>of</strong> supply<strong>and</strong> dem<strong>and</strong> would be ruined by disorganized tumorcell proliferation resulting in <strong>the</strong> disastrous dysfunction<strong>of</strong> <strong>the</strong> tumor vessels. The reduction <strong>of</strong> functional bloodflow is significant in clinically hypovascular tumors suchas pancreatic <strong>cancer</strong>s [17]. Under <strong>the</strong>se conditions, <strong>cancer</strong>cells are likely to encounter chronic ischemia leading to ashortage <strong>of</strong> nutrients. Cancer cells might adapt <strong>the</strong>mselvesto such a harsh microenvironment. In experimental cellculture systems, several <strong>cancer</strong> cells were resistant tonutrient-deprived conditions. For example, several pancreatic<strong>cancer</strong>- <strong>and</strong> colorectal <strong>cancer</strong>-derived cell linesshowed a survival rate <strong>of</strong> more than 50% after 48 h <strong>of</strong> culturein a medium completely lacking carbon <strong>and</strong> nitrogensources. Contrastingly, non-transformed fibroblasts werecompletely abolished within a day under <strong>the</strong> same conditions[18]. These findings suggest that <strong>cancer</strong> cells usealternative metabolic processes for <strong>the</strong>ir survival understarved conditions.Metabolic stresses reportedly induce apoptosis [19]. Inmany tumor cells, apoptosis is suppressed by <strong>the</strong> overexpression<strong>of</strong> anti-apoptotic molecules or by <strong>the</strong> lack <strong>of</strong>pro-apoptotic molecules. Less apoptosis may explain <strong>the</strong>limited death <strong>of</strong> <strong>cancer</strong> cells under metabolic stresses,but <strong>the</strong> manner in which <strong>cancer</strong> cells obtain <strong>the</strong>ir necessarynutrients remains a mystery. One possible solutionis that <strong>the</strong> cells digest <strong>the</strong>ir own components <strong>and</strong> obtainamino acids as an alternative energy source. <strong>Autophagy</strong>seems ideal for <strong>cancer</strong> cells to maintain energy homeostasisin such an autonomous fashion. <strong>Autophagy</strong> is known tobe induced by different forms <strong>of</strong> metabolic stress, includingnutrient deprivation, growth factor deprivation, <strong>and</strong>hypoxia [13,20]. Since <strong>the</strong>se conditions are <strong>of</strong>ten observedin physiological tumor microenvironments, autophagy islikely activated in <strong>cancer</strong> cells.We should also remind <strong>the</strong> apparent discrepancy between<strong>the</strong> tumor ‘‘cell” doubling time <strong>and</strong> <strong>the</strong> tumor ‘‘volume”doubling time. The potential tumor cell doublingtime estimated by in vivo measurement <strong>of</strong> S-phase durationwas remarkably rapid in many solid tumors, a medianvalue <strong>of</strong> <strong>the</strong> order <strong>of</strong> 5 days. However, tumor volume doublingtime determined by radiological measurement <strong>of</strong> <strong>the</strong>size <strong>of</strong> <strong>the</strong>se tumors was much slower, months or years[21,22]. It suggests that about half <strong>of</strong> <strong>the</strong> proliferating <strong>cancer</strong>cells are subsequently lost under tumor microenvironmentthough many <strong>of</strong> <strong>the</strong>m have already acquired antiapoptoticfunction. Is <strong>the</strong>re any physiological relevance <strong>of</strong>such dynamism <strong>of</strong> cell kinetics? A potential scenario is that<strong>the</strong> vanishing cells serve nutritional sources for surroundingsurviving cells. In this sense, inducing cell death <strong>and</strong>supporting cell survival under metabolic stresses are notPlease cite this article in press as: K. Tsuchihara et al., <strong>Autophagy</strong> <strong>and</strong> <strong>cancer</strong>: <strong>Dynamism</strong> <strong>of</strong> <strong>the</strong> metabolism <strong>of</strong> tumor cells..., Cancer Lett. (2008), doi:10.1016/j.canlet.2008.09.040


ARTICLE IN PRESSK. Tsuchihara et al. / Cancer Letters xxx (2008) xxx–xxx 3contradicting. <strong>Autophagy</strong> is also known to induce celldeath <strong>and</strong> possibly sacrifice some part <strong>of</strong> individual tumorcell for supporting a prolonged survival <strong>and</strong> continuousgrowth <strong>of</strong> o<strong>the</strong>r cells in <strong>the</strong> tumor mass.Several experimental methods have been used to identify<strong>the</strong> activation <strong>of</strong> autophagy (Table 1) [23]. Measuring<strong>the</strong> turnover <strong>of</strong> long-lived proteins provides a biochemicalclue to autophagy. Transmission electron microscopy reveals<strong>the</strong> ultrastructure characteristics <strong>of</strong> <strong>the</strong> autophagosome.Autophagosome formation is also convenientlymonitored by following a phosphatidylethanolamine (PE)-conjugated form <strong>of</strong> yeast Atg8 or mammalian LC3, one <strong>of</strong><strong>the</strong> mammalian orthologs <strong>of</strong> Atg8. During autophagy, LC3shifts from a soluble form to a membrane-bound form(LC3-II) <strong>and</strong> is incorporated into <strong>the</strong> autophagosomal innermembrane. The existence <strong>of</strong> autophagosome-incorporatedLC3-II can be detected biochemically (immunoblottingagainst LC3) or microscopically (immunocytochemistryagainst LC3 or exogenously expressed, fluorescently taggedLC3).The ultrastructure <strong>of</strong> autophagosomes in tumor cellshas been observed in several experimental systems,including a rat pancreatic <strong>cancer</strong> model [24]. A high potentialfor autophagic protein degradation was observed in anundifferentiated colon <strong>cancer</strong> cell line, HT29, <strong>and</strong> o<strong>the</strong>rtransformed cells [25]. Nutrient deprivation-induced LC3-II turnover was observed in colorectal <strong>cancer</strong>-derived celllines that showed resistance to starvation [26]. In additionto <strong>the</strong>se findings in cultured <strong>cancer</strong> cells, <strong>the</strong> increasedexpression <strong>of</strong> autophagy-related proteins, including BNIP3<strong>and</strong> LC3, was observed specifically in colorectal <strong>and</strong> gastric<strong>cancer</strong> epi<strong>the</strong>lia in surgically-resected specimens [26,27].Increasing evidence implies that autophagy has a cytoprotectiverole in <strong>cancer</strong> cells under metabolic stress.Transplanted epi<strong>the</strong>lial tumors in which <strong>the</strong> Beclin 1 orAtg5 alleles were deleted showed a reduction in autophagy<strong>and</strong> an increase in cell death in regions exposed to metabolicstress [28–30]. Similarly, a cytoprotective function<strong>of</strong> autophagy was observed in cultured <strong>cancer</strong> cells. The geneticinactivation <strong>of</strong> autophagy by <strong>the</strong> suppression <strong>of</strong> ATGexpression using RNA interference or <strong>the</strong> constitutive activation<strong>of</strong> PI3K induced cell death in response to metabolicstresses [28–30]. RNA interference <strong>of</strong> ATG5, Beclin 1 <strong>and</strong>ATG7 enhanced tamoxifen-induced apoptosis in tamoxifen-resistantbreast <strong>cancer</strong> cell lines [31]. ATG5 knockdownalso enhanced <strong>the</strong> effect <strong>of</strong> alkylating drug-inducedcell death [32]. The pharmacological inhibition <strong>of</strong> autophagyalso induced nutrient deprivation-induced cell death[26]. Chloroquine, which interferes with lysosomal function,inhibited autophagy <strong>and</strong> suppressed Myc-inducedlymphomagenesis in a transgenic mouse model [32].Though <strong>the</strong> above findings suggest that autophagy maycontribute to tumor cell survival <strong>and</strong> tumor formation, <strong>the</strong>molecular mechanisms underlying <strong>the</strong> acquisition <strong>of</strong> vigorousautophagic activity in <strong>cancer</strong> cells have remained unclear.Recently, Kroemer <strong>and</strong> his colleagues reported apotential anti-autophagic function <strong>of</strong> cytoplasm-localizingp53 [33]. They observed <strong>the</strong> degradation <strong>of</strong> p53 under metabolicstresses followed by <strong>the</strong> induction <strong>of</strong> autophagy.Contrastingly, <strong>the</strong> loss <strong>of</strong> p53 resulted in <strong>the</strong> consistentactivation <strong>of</strong> autophagy in a series <strong>of</strong> cell lines. While overexpression<strong>of</strong> wild-type p53 reduced <strong>the</strong> aberrant autophagosomeformation, a mutant p53 protein whichharbors a codon 175 mutation (R175H), which is frequentlyidentified in clinical human <strong>cancer</strong>s, did not affect<strong>the</strong> higher basal autophagic activity <strong>of</strong> p53-null cells.Though most <strong>of</strong> <strong>the</strong>ir observation was limited in <strong>the</strong> culturecell system, fur<strong>the</strong>r studies which clarify its relevanceto tumor formation <strong>and</strong> progression are awaited.3. <strong>Autophagy</strong> <strong>and</strong> tumor suppressionAn opposite perspective was presented by a studyexamining a Bcl-2-binding protein, Beclin 1. Levine <strong>and</strong>Table 1Recomm<strong>and</strong>ed methods for monitoring autophagy in higher eukaryotes [23].CriteriaMethodsMonitoring phagophore <strong>and</strong> autophagosome formation by steady state methods1. Electron microscopy (increase in autophagosome quantity) Quantitative electron microscopy, immunoelectron microscopy2. Atg8/LC3 Western blotting <strong>and</strong> ubiquitin-like protein conjugation Western blotsystems(increase in <strong>the</strong> amount <strong>of</strong> LC3-II, <strong>and</strong> Atg12–Atg5 conjugation)3. Fluorescence microscopy (increase in punctate LC3 (or Atg18)) Fluorescence, immun<strong>of</strong>luorescence <strong>and</strong> immunoelectron microscopy4. TOR <strong>and</strong> Atg1 kinase activity Western blot, immunoprecipitation or kinase assays5. Transcriptional regulation Nor<strong>the</strong>rn blot or qRT-PCRMonitoring autophagy by flux measurements1. Autophagic protein degradation Turnover <strong>of</strong> long-lived proteins2. Turnover <strong>of</strong> LC3-II Western blot +/ lysosomal fusion or degradation inhibitors3. GFP-Atg8/LC3 lysosomal delivery, <strong>and</strong> proteolysisFluorescence microscopy, FACS Western blot +/ lysosomal fusion or degradation(to generate free GFP)inhibitors4. p62 Western blot Western blot with qRT-PCR or Nor<strong>the</strong>rn blot to assess transcription5. Autophagic sequestration assays Lysosomal accumulation by biochemical or multilabel fluorescence techniques6. Turnover <strong>of</strong> autophagic compartments Electron microscopy morphometry/stereology7. Autophagosome-lysosome colocalization <strong>and</strong> dequenching assay Fluorescence microscopy8. Sequestration <strong>and</strong> processing assays in plants Chimeric RFP fluorescence <strong>and</strong> processing, light <strong>and</strong> electron microscopy9. T<strong>and</strong>em mRFP-GFP fluorescence microscopy Fluorescence microscopy <strong>of</strong> t<strong>and</strong>em mRFP-GFP-LC310. Tissue fractionation Centrifugation, Western blot <strong>and</strong> electron microscopy11. Analyses in vivo Fluorescence microscopy <strong>and</strong> immunohistochemistryPlease cite this article in press as: K. Tsuchihara et al., <strong>Autophagy</strong> <strong>and</strong> <strong>cancer</strong>: <strong>Dynamism</strong> <strong>of</strong> <strong>the</strong> metabolism <strong>of</strong> tumor cells..., Cancer Lett. (2008), doi:10.1016/j.canlet.2008.09.040


ARTICLE IN PRESS4 K. Tsuchihara et al. / Cancer Letters xxx (2008) xxx–xxxher colleagues first identified Beclin 1, a mammalian ortholog<strong>of</strong> ATG6, as a c<strong>and</strong>idate tumor suppressor. Beclin 1interacts with class III PI3-kinase, Vps34, <strong>and</strong> this interactionwas crucial for <strong>the</strong> induction <strong>of</strong> autophagy <strong>and</strong> suppression<strong>of</strong> <strong>the</strong> growth <strong>of</strong> <strong>the</strong> xenografted breast <strong>cancer</strong>cell lines [34–36]. Vps34 is also required for normal proteintrafficking pathways such as <strong>the</strong> delivery <strong>of</strong> proteasesfrom <strong>the</strong> trans-Golgi network to <strong>the</strong> lysosomes. Atg6(Vsp30) was reportedly involved in <strong>the</strong> regulation <strong>of</strong> bothautophagy <strong>and</strong> endosomal membrane trafficking in yeast.Whe<strong>the</strong>r mammalian Beclin 1 takes part in <strong>the</strong> membranetrafficking is still controversial [34,35].The hemiallelic loss <strong>of</strong> <strong>the</strong> Beclin 1 coding gene was observedin 40–75% <strong>of</strong> sporadic human <strong>cancer</strong>s in <strong>the</strong> breast,ovary <strong>and</strong> prostate [37]. A gene-targeted mouse model <strong>of</strong>Beclin 1 provided ano<strong>the</strong>r clue [38,39]. The homozygousdeletion <strong>of</strong> Beclin 1 led to embryonic lethality. Meanwhile,Beclin 1 heterozygous mutant mice showed decreasedautophagy <strong>and</strong> increased spontaneous tumors, includinglung <strong>and</strong> liver <strong>cancer</strong>s <strong>and</strong> lymphomas. In <strong>the</strong>se mice, noloss <strong>of</strong> heterozygosity (LOH) was observed <strong>and</strong> <strong>the</strong> remainingwild-type allele <strong>of</strong> Beclin 1 was intact. Fur<strong>the</strong>rmore,Beclin 1 protein expression was reduced but not completelydiminished in mouse tumors as well as human clinicalsamples [36]. These findings suggest that Beclin 1 is ahaplo-insufficient tumor-suppressor gene. The tumor-suppressivefunction <strong>of</strong> Beclin 1 was reinforced by <strong>the</strong> relevance<strong>of</strong> Beclin 1-associated proteins to tumorsuppression. UVRAG was initially identified from <strong>the</strong> cDNAlibrary, which partially rescued <strong>the</strong> UV sensitivity <strong>of</strong> axeroderma pigmentosum (XP) cell line. The screening <strong>of</strong>Beclin 1-binding proteins revealed that UVRAG was recruitedto <strong>the</strong> Beclin 1–class III PI3K complex. UVRAG activatedBeclin 1 <strong>and</strong> induced autophagosome formation [40].In addition, UVRAG is involved in autophagosome maturation.UVRAG interacts with class C te<strong>the</strong>ring proteins(Vps11, Vps16, Vps18 <strong>and</strong> Vps33) resulting in activation<strong>of</strong> a small GTPase, Rab7 which enhances <strong>the</strong> fusion <strong>of</strong> autophagosomesto endosomes [41]. As seen with Beclin 1, UV-RAG is monoallelically mutated in human colorectal<strong>cancer</strong>s [40]. Bif-1 (also known as Endophilin B1) interactswith Beclin 1 through UVRAG. Bif-1 activated class III PI3K<strong>and</strong> induced autophagy. Spontaneous tumor formationwas increased in Bif-1 deficient mice [42].During investigations <strong>of</strong> autophagy in mammalian cells,several signaling pathways have been revealed to regulateautophagy. Interestingly, correlations between pro-autophagicmolecules <strong>and</strong> tumor suppressors <strong>and</strong> betweenanti-autophagic molecules <strong>and</strong> oncogene products can bepointed out (Fig. 1). The mammalian target <strong>of</strong> rapamycin(mTOR) is a key molecule for regulating <strong>cancer</strong> cell proliferation.Rapamycin inhibits mTOR function followed byautophagy induction [11,43]. Molecules known to suppressmTOR, including PTEN <strong>and</strong> TSC, both <strong>of</strong> which are regardedas tumor-suppressor gene products, induce autophagy[44,45]. Meanwhile, mTOR-activating molecules like classI PI3K <strong>and</strong> Akt, which are frequently activated in various<strong>cancer</strong> cells, inhibit autophagy [11,46]. The involvement <strong>of</strong>p53 in <strong>the</strong> activation <strong>of</strong> autophagy has also been suggested.In contrast to <strong>the</strong> inhibitory effect <strong>of</strong> cytoplasmic p53 men-Fig. 1. Cellular events during autophagy (macroautophagy). In <strong>the</strong> first step <strong>of</strong> autophagy, <strong>the</strong> isolation membrane (phagophore) is elongated <strong>and</strong>sequestrates <strong>the</strong> cytoplasmic constituents. Fusion <strong>of</strong> <strong>the</strong> edges <strong>of</strong> <strong>the</strong> isolation membrane forms an autophagosome, which contains <strong>the</strong> sequestratedcomponents. A lysosome <strong>the</strong>n fuses with <strong>the</strong> autophagosome, <strong>and</strong> lysosomal hydrolases degrade <strong>the</strong> engulfed components along with <strong>the</strong> inner membrane<strong>of</strong> <strong>the</strong> autophagosome. Mammalian homologues <strong>of</strong> ATG (autophagy-related genes) have been identified. Class I PI3K <strong>and</strong> mTOR signaling inhibits autophagyactivation, whereas tumor-suppressor proteins like p53 <strong>and</strong> PTEN induce autophagy. Rapamycin inhibits mTOR <strong>and</strong> induces autophagy. 3-Methyladenine,chloroquine, bafilomycin A1, E64d <strong>and</strong> pepstatin A inhibit autophagy at different points. These small molecules are used to inhibit autophagy inexperimental model systems.Please cite this article in press as: K. Tsuchihara et al., <strong>Autophagy</strong> <strong>and</strong> <strong>cancer</strong>: <strong>Dynamism</strong> <strong>of</strong> <strong>the</strong> metabolism <strong>of</strong> tumor cells..., Cancer Lett. (2008), doi:10.1016/j.canlet.2008.09.040


ARTICLE IN PRESSK. Tsuchihara et al. / Cancer Letters xxx (2008) xxx–xxx 5tioned above, <strong>the</strong> transactivating function <strong>of</strong> nuclear p53takes a role in <strong>the</strong> autophagy induction. DRAM is a directtranscriptional target <strong>of</strong> p53, <strong>and</strong> its product localizes in<strong>the</strong> lysosomal membrane. The activation <strong>of</strong> p53 inducedan increase in autophagy in a DRAM-dependent manner[47]. p53 was also reported to affect autophagy via <strong>the</strong> suppression<strong>of</strong> mTOR [45]. DAPk, death-associated protein kinase,induces autophagy as well as apoptosis. The tumorsuppressivefunction <strong>of</strong> DAPk has also been described, <strong>and</strong>promoter hypermethylation <strong>of</strong> <strong>the</strong> DAPk locus has been reportedin several human <strong>cancer</strong>s [48].These findings suggest <strong>the</strong> likely relevance <strong>of</strong> autophagyto tumor suppression. However, <strong>the</strong> above-mentionedpro- <strong>and</strong> anti-autophagic molecules, like p53 <strong>and</strong> mTOR,are pluripotent proteins that regulate cell proliferation<strong>and</strong> death via various molecular events. The net contribution<strong>of</strong> autophagy to cell fate decisions during tumorigenesisshould be carefully estimated, but this objective hasnot yet been successfully accomplished.4. <strong>Autophagy</strong> <strong>and</strong> cell death<strong>Autophagy</strong> has attracted much attention in connectionwith cell death. ‘‘Type II” or ‘‘autophagic” non-apoptoticprogrammed cell death is morphologically defined by <strong>the</strong>existence <strong>of</strong> autophagosomes [49]. Autophagic death hasbeen reported in <strong>cancer</strong> cells, especially those that havebeen treated with chemo<strong>the</strong>rapeutic or radio<strong>the</strong>rapeuticagents [50]. This cytotoxic effect has been supposed to be<strong>the</strong> main reason for <strong>the</strong> tumor-suppressive function <strong>of</strong>autophagy. Self-cannibalism, that is, <strong>the</strong> situation in whichexcess autophagic catabolism exceeds <strong>the</strong> capacity for cellularanabolism, is frequently argued as a potential mechanismfor autophagic death. Though autophagy has beenregarded to degrade cytoplasmic constituents non-specifically,<strong>the</strong> specific autophagic degradation <strong>of</strong> target proteins<strong>and</strong> organelles has also been reported. The specific degradation<strong>of</strong> cytoprotective factors is ano<strong>the</strong>r undeniablemechanism <strong>of</strong> autophagic death [51]. However, experimentalevidence supporting <strong>the</strong>se ideas has not yet beenobtained. Moreover, cytotoxic stimuli, such as oxidativestress, induces both autophagy <strong>and</strong> cell death, but whe<strong>the</strong>rautophagy is an active death-inducing mechanism (celldeath by autophagy) or a result <strong>of</strong> an unsuccessful effortto prolong <strong>the</strong> survival <strong>of</strong> damaged cells (cell death withautophagy) is difficult to distinguish.Recent findings have raised a question about <strong>the</strong> relevance<strong>of</strong> autophagic death to tumor suppression. The allelicloss <strong>of</strong> Beclin 1 inhibited both basal <strong>and</strong> stress-inducedautophagy in immortalized baby mouse kidney epi<strong>the</strong>lialcells. Metabolic stress-induced cell death was apparentlyincreased, suggesting loss <strong>of</strong> autophagy led impaired cytoprotectivefunction. Despite impairment in autophagymediatedcell survival, Beclin 1+/ cells were more tumorigenicthan <strong>the</strong> wild-type control cells [28]. To make aplausible explanation for <strong>the</strong>se contradictory findings, <strong>the</strong>loss <strong>of</strong> cytoprotection induces tumorigenesis, is stillchallenging.5. Potential mechanisms for <strong>the</strong> tumor-suppressivefunction <strong>of</strong> autophagyRecently, White <strong>and</strong> colleagues have proposed interestinghypo<strong>the</strong>ses about <strong>the</strong> above issues (Fig. 2). Under meta-Fig. 2. Both <strong>the</strong> gain <strong>and</strong> loss <strong>of</strong> autophagy are possibly involved in tumor formation <strong>and</strong> growth. The activation <strong>of</strong> autophagy supplies nutrients to tumorcells under metabolic stress. On <strong>the</strong> o<strong>the</strong>r h<strong>and</strong>, <strong>the</strong> eradication <strong>of</strong> tumor cells by autophagy-related cell death is assumed to occur, <strong>and</strong> <strong>the</strong> loss <strong>of</strong>autophagy may lead to tumor cell survival. A recent mouse model using transplanted autophagy-deficient tumor cells revealed that <strong>the</strong> loss <strong>of</strong> autophagyinduced DNA damage <strong>and</strong> inflammation, enhancing tumor formation <strong>and</strong> growth.Please cite this article in press as: K. Tsuchihara et al., <strong>Autophagy</strong> <strong>and</strong> <strong>cancer</strong>: <strong>Dynamism</strong> <strong>of</strong> <strong>the</strong> metabolism <strong>of</strong> tumor cells..., Cancer Lett. (2008), doi:10.1016/j.canlet.2008.09.040


ARTICLE IN PRESS6 K. Tsuchihara et al. / Cancer Letters xxx (2008) xxx–xxxbolic stress, Beclin 1+/ epi<strong>the</strong>lial cells overexpressing antiapoptoticBcl-2 showed an increase in DNA double str<strong>and</strong>breaks, gene amplification, <strong>and</strong> chromosomal number disorder.They assumed that <strong>the</strong> loss <strong>of</strong> autophagy inducesDNA damage <strong>and</strong> chromosomal instability, followed by increasedtumor susceptibility [30]. The exact mechanismsfor how autophagy maintains genome stability remain unclear.One possibility is that cells lacking autophagy are unableto reduce damaged mitochondria <strong>and</strong> peroxisomes.These damaged organelles are potential sources <strong>of</strong> reactiveoxygen species that induce genotoxic stress.When <strong>the</strong> same autophagy-deficient epi<strong>the</strong>lial cellswere exposed to starvation, apparent necrotic cell deathwas observed when apoptosis was inhibited. Fur<strong>the</strong>rmore,significant inflammation was induced in <strong>the</strong> tumor tissuesin which autophagy-deficient cells were transplanted [28].Necrosis is <strong>of</strong>ten associated with macrophage infiltrationin vivo, <strong>and</strong> tumor-associated macrophages enhance tumorprogression [52]. It is an interesting hypo<strong>the</strong>sis that <strong>the</strong>inflammation induced by a lack <strong>of</strong> autophagy may be correlatedwith tumor progression in vivo.The above scenarios are not incompatible with <strong>the</strong> ideathat autophagy protects <strong>cancer</strong> cells from metabolic stressinduceddeath, <strong>and</strong> <strong>the</strong>se potential mechanisms are worthy<strong>of</strong> fur<strong>the</strong>r discussion to elucidate <strong>the</strong> physiological roles <strong>of</strong>autophagy in tumorigenesis.6. Perspective – remaining tasksThe relevance <strong>of</strong> autophagy to <strong>cancer</strong> remains a frustratingtopic to discuss. It is necessary to underst<strong>and</strong> <strong>the</strong>consequences <strong>of</strong> <strong>the</strong> loss or gain <strong>of</strong> autophagy in <strong>the</strong> context<strong>of</strong> <strong>the</strong> tumor microenvironment. Previously proposedhypo<strong>the</strong>ses for how autophagy contributes to tumor biology,ei<strong>the</strong>r pro- or anti-tumorigenic, are possible <strong>and</strong> interesting,but most <strong>of</strong> <strong>the</strong>m have arisen from artificialexperimental systems <strong>and</strong> no direct clinico-pathologicalevidence supports <strong>the</strong>se ideas. Evaluating autophagy inclinical tumor samples has been difficult, mainly because<strong>of</strong> <strong>the</strong> lack <strong>of</strong> appropriate markers for detecting activeautophagy. Autophagosome formation in tumor tissueshas been morphologically confirmed using electronmicroscopy, but this method is not suitable for h<strong>and</strong>linga large number <strong>of</strong> specimens. Recently, anti-LC3 antibodiesfor immunostaining have become available [26,53,54].LC3-II proteins incorporated into autophagosome membranesexhibit a punctuate cytoplasmic staining patternduring active autophagy. LC3 immunostaining is an easiermethod <strong>of</strong> evaluating autophagosome formation in conventionalformaldehyde-fixed surgically-resected specimens.We applied polyclonal anti-LC3 antibody for <strong>the</strong>immunostaining <strong>of</strong> colorectal <strong>cancer</strong> specimens. In over90% <strong>of</strong> <strong>the</strong> cases, LC3 accumulated specifically in <strong>cancer</strong>ousepi<strong>the</strong>lia but not in adjacent non-<strong>cancer</strong>ous mucosa [26].Similar <strong>cancer</strong>-specific accumulation <strong>of</strong> LC3 was observedin pancreatic <strong>cancer</strong>. Interestingly, strong LC3 expressionin <strong>the</strong> peripheral area <strong>of</strong> <strong>cancer</strong> tissue was correlated witha poor prognosis (Fig. 3) [55]. As we mentioned earlier,both pancreatic <strong>and</strong> colorectal <strong>cancer</strong>-derived cell linesare <strong>of</strong>ten resistant to <strong>the</strong> nutrient-starvation. It seemsmore than coincidence that specific LC3 accumulationwas observed in <strong>the</strong> clinical samples <strong>of</strong> <strong>the</strong>se tumors. Fur<strong>the</strong>rinvestigation using various <strong>cancer</strong> tissue samples mayreveal <strong>the</strong> contribution <strong>of</strong> <strong>the</strong> gain or loss <strong>of</strong> autophagy intumor formation <strong>and</strong> progression more clearly.<strong>Autophagy</strong> is executed by dynamic <strong>and</strong> multiple cellularprocesses, including <strong>the</strong> formation <strong>of</strong> <strong>the</strong> autophagosome,<strong>the</strong> delivery <strong>of</strong> cytoplasmic constituents to <strong>the</strong>lysosome, <strong>and</strong> <strong>the</strong> digestion <strong>and</strong> recycling <strong>of</strong> <strong>the</strong>se targetmolecules <strong>and</strong> organelles. For <strong>the</strong> precise evaluation <strong>of</strong>autophagic activity, <strong>the</strong> ‘‘autophagic flux” must be estimated.For example, <strong>the</strong> accumulation <strong>of</strong> autophagosomesreflects ei<strong>the</strong>r an increase in autophagosome formation(activation <strong>of</strong> autophagy) or a reduction in <strong>the</strong> degradation<strong>of</strong> autophagosomes (inhibition <strong>of</strong> autophagy). To determine<strong>the</strong> flux, <strong>the</strong> rate <strong>of</strong> long-lived protein degradationmust be measured or <strong>the</strong> changes in appropriate autophagymarkers, such as <strong>the</strong> LC3-II protein level, must be assessedwith or without <strong>the</strong> arrest <strong>of</strong> autophagic flux at agiven point <strong>of</strong> blockage. Recently, Klionsky <strong>and</strong> 231 o<strong>the</strong>rscientists published guidelines for <strong>the</strong> use <strong>and</strong> interpretation<strong>of</strong> assays for monitoring autophagy in higher eukaryotes(Table 1) [23]. In <strong>the</strong>se guidelines, <strong>the</strong> authors stronglyrecommended <strong>the</strong> measurement <strong>of</strong> autophagic flux formonitoring autophagy. Detecting autophagosome formationusing steady state methods, including electronmicroscopy or LC3 immunostaining, should be combinedwith flux measurements. However, applying such dynamicassays to clinical samples is <strong>of</strong>ten technically difficult.The above-mentioned guidelines do not provide a fullresolution <strong>of</strong> <strong>the</strong> above difficulties. Fur<strong>the</strong>r discussionmight be needed to establish st<strong>and</strong>ard methods for evaluatingautophagy in clinical samples. To assess autophagicactivity using steady state methods, not only one parameter,but multiple autophagy-related molecules should beapplied. For example, while <strong>the</strong> loss <strong>of</strong> Beclin 1 implies areduction in autophagosome formation, this outcomeshould be confirmed by a decrease in autophagosome-specificmarkers, such as LC3-II. Mimicking in vivo events onex vivo systems may support <strong>the</strong> findings obtained in clinicalsamples using steady state methods. In such cases, itshould be remembered that autophagy is induced undercomplex microenvironments in <strong>cancer</strong> tissues. As mentionedpreviously, <strong>cancer</strong> cells are <strong>of</strong>ten exposed to chronicischemia. Under this situation, not only nutrients but alsooxygen, growth factors <strong>and</strong> o<strong>the</strong>r components providedby blood flow might be decreased. Meanwhile, metabolites<strong>and</strong> o<strong>the</strong>r components excreted from <strong>cancer</strong> cells may notbe properly removed from <strong>the</strong> <strong>cancer</strong> tissues. Cancer cellsare also directly <strong>and</strong> indirectly influenced by surroundingstromal cells <strong>and</strong> extracellular matrices. Reconstitutingall <strong>the</strong>se factors in an experimental cell culture system ispractically unfeasible, but <strong>the</strong> above restrictions should alwaysbe taken into account when evaluating <strong>the</strong> resultsobtained from any model system.To analyze autophagy function under more physiologicalconditions, appropriate animal models are eagerlyneeded. Xenografted <strong>and</strong>/or isografted animals in which<strong>the</strong> transplanted transformed cells lacking autophagy-relatedgenes are useful, but not perfect. Transplanted tumorsdo not always reproduce <strong>the</strong> microenvironmentalPlease cite this article in press as: K. Tsuchihara et al., <strong>Autophagy</strong> <strong>and</strong> <strong>cancer</strong>: <strong>Dynamism</strong> <strong>of</strong> <strong>the</strong> metabolism <strong>of</strong> tumor cells..., Cancer Lett. (2008), doi:10.1016/j.canlet.2008.09.040


ARTICLE IN PRESSK. Tsuchihara et al. / Cancer Letters xxx (2008) xxx–xxx 7Fig. 3. Autophagosome formation in <strong>the</strong> surgically-resected specimens. (A) Autophagosome-incorporated LC3 protein was detected using GFP-LC3 fusionprotein in a cultured colorectal <strong>cancer</strong> cell line, SW480, during amino acid-deprivation. (B) Tumor cell-specific LC3 accumulation in colorectal <strong>cancer</strong> tissuewas detected using immunohistochemistry with anti-LC3 antibody <strong>and</strong> hematoxylin <strong>and</strong> eosin staining. (C) LC3 accumulation at <strong>the</strong> peripheral area <strong>of</strong> <strong>the</strong>pancreatic <strong>cancer</strong> tissue. Nv: nerve cells in which LC3 is constitutively expressed. (inlet) Higher magnification <strong>of</strong> (B) <strong>and</strong> (C). LC3 proteins were localized in<strong>the</strong> cytoplasm with irregular condensation. (D) The group with strongly positive expression <strong>of</strong> LC3 protein in <strong>the</strong> peripheral area <strong>of</strong> <strong>the</strong> pancreatic <strong>cancer</strong>tissue had a significantly shorter survival time. (A) <strong>and</strong> (B) are quoted from Ref. [26], (C) <strong>and</strong> (D) are quoted from Ref. [55].conditions <strong>of</strong> clinical tumors. Fur<strong>the</strong>rmore, such models donot provide information regarding <strong>the</strong> initial steps <strong>of</strong>tumorigenesis, in which autophagy might have criticalroles for pampering aberrantly proliferating <strong>cancer</strong> cellswithout extra blood supply. Examining <strong>the</strong> effect <strong>of</strong> variousautophagy inhibitors in tumorigenic animals is ano<strong>the</strong>rway to evaluate <strong>the</strong> roles <strong>of</strong> autophagy in vivo. But it has<strong>the</strong> drawback that <strong>the</strong>se reagents are not completelyautophagy-specific [32]. Recently, several genetically engineeredmouse models targeting autophagy-related geneshave been reported [12]. However, <strong>the</strong>se mice <strong>of</strong>ten showembryonic or neonatal lethality, hampering <strong>the</strong> long-termobservation <strong>of</strong> tumorigenesis in adult animals. Tissue-specificor inducible conditional knockout systems may beuseful for this purpose.Whe<strong>the</strong>r <strong>the</strong> control <strong>of</strong> autophagy is useful for <strong>cancer</strong><strong>the</strong>rapy <strong>and</strong> prevention is ano<strong>the</strong>r important issue. Severalsmall molecules are known to activate or inhibit autophagy(Fig. 1) [56]. Inhibiting <strong>the</strong> class I PI3K-mTOR axis byrapamycin induces autophagy. The inhibition <strong>of</strong> autophagycan be achieved by targeting several points. A class III PI3Kinhibitor (3-methyladenine), lysosomotropic alkalines(chloroquine <strong>and</strong> 3-hydroxychloroquine), a lysosomal protonpump inhibitor (bafilomycin A1) <strong>and</strong> lysosomal enzymeinhibitors (E64d <strong>and</strong> pepstatin A) are available forexperimental use. Since <strong>the</strong> contribution <strong>of</strong> autophagy totumorigenesis is still controversial, we should carefullyconsider <strong>the</strong> application <strong>of</strong> autophagy inhibitors or activatorsfor <strong>the</strong>rapeutic use. Increasing evidence emphasizes<strong>the</strong> importance <strong>of</strong> basal autophagy for cellular quality control[57]. The disturbance <strong>of</strong> autophagy might result inunexpected adverse effects, especially in postmitotic cellsin <strong>the</strong> central nervous system <strong>and</strong> cardiovascular system.The specificity <strong>of</strong> such activators <strong>and</strong> inhibitors is ano<strong>the</strong>rissue to consider. The above-mentioned chemicals are notautophagy-specific. Also, <strong>the</strong> targeted kinases (PI3K,mTOR) or organelles (lysosome) have broad functionso<strong>the</strong>r than autophagy regulation.In <strong>the</strong> last decade, autophagy has appeared in <strong>the</strong> centerstage <strong>of</strong> <strong>cancer</strong> biology <strong>and</strong> is now attracting much attentionin <strong>the</strong> development <strong>of</strong> new <strong>cancer</strong> <strong>the</strong>rapeutics. Asmentioned above, a considerable number <strong>of</strong> issues remainto be clarified. However, similar to its molecular process,<strong>the</strong> research field <strong>of</strong> autophagy is very dynamic. Novelstrategies for monitoring autophagy <strong>and</strong> experimentalmodels are appearing, <strong>and</strong> various small molecules regulatingautophagic process will be available. Fur<strong>the</strong>r investigationwill reveal <strong>the</strong> dynamism <strong>of</strong> metabolism in <strong>cancer</strong>cells <strong>and</strong> tissues that is still behind <strong>the</strong> scene now. And itwill likely open up a new field <strong>of</strong> <strong>cancer</strong> biology.Conflicts <strong>of</strong> interest statementNone declared.AcknowledgementsThis work was supported by Grants for <strong>the</strong> Third-TermComprehensive 10-Year Strategy for Cancer Control, <strong>and</strong> aGrant-in-Aid for Cancer Research from <strong>the</strong> Ministry <strong>of</strong>Health, Labour <strong>and</strong> Welfare <strong>and</strong> a Grant-in-Aid for ScientificResearch on Priority Areas from <strong>the</strong> Ministry <strong>of</strong> Education,Culture, Sports, Science <strong>and</strong> Technology – Japan.References[1] D.J. Klionsky, The molecular machinery <strong>of</strong> autophagy: unansweredquestions, J. Cell Sci. 118 (2005) 7–18.Please cite this article in press as: K. Tsuchihara et al., <strong>Autophagy</strong> <strong>and</strong> <strong>cancer</strong>: <strong>Dynamism</strong> <strong>of</strong> <strong>the</strong> metabolism <strong>of</strong> tumor cells..., Cancer Lett. (2008), doi:10.1016/j.canlet.2008.09.040


ARTICLE IN PRESS8 K. Tsuchihara et al. / Cancer Letters xxx (2008) xxx–xxx[2] D.J. Klionsky, <strong>Autophagy</strong>: from phenomenology to molecularunderst<strong>and</strong>ing in less than a decade, Nat. Rev. Mol. Cell Biol. 8(2007) 931–937.[3] B. Levine, D.J. Klionsky, Development by self-digestion: molecularmechanisms <strong>and</strong> biological functions <strong>of</strong> autophagy, Dev. Cell 6(2004) 463–477.[4] N. Mizushima, <strong>Autophagy</strong>: process <strong>and</strong> function, Genes Dev. 21(2007) 2861–2873.[5] N. Mizushima, D.J. Klionsky, Protein turnover via autophagy:implications for metabolism, Annu. Rev. Nutr. 27 (2007) 19–40.[6] D.J. Klionsky, J.M. Cregg, W.A. Dunn Jr., S.D. Emr, Y. Sakai, I.V.S<strong>and</strong>oval, A. Sibirny, S. Subramani, M. Thumm, M. Veenhuis, Y.Ohsumi, A unified nomenclature for yeast autophagy-related genes,Dev. Cell 5 (2003) 539–545.[7] P. Codogno, A.J. Meijer, <strong>Autophagy</strong> <strong>and</strong> signaling: <strong>the</strong>ir role in cellsurvival <strong>and</strong> cell death, Cell Death Differ. 12 (Suppl. 2) (2005) 1509–1518.[8] Z. Xie, D.J. Klionsky, Autophagosome formation: core machinery <strong>and</strong>adaptations, Nat. Cell Biol. 9 (2007) 1102–1109.[9] L. Yu, L. Str<strong>and</strong>berg, M.J. Lenardo, The selectivity <strong>of</strong> autophagy <strong>and</strong> itsrole in cell death <strong>and</strong> survival, <strong>Autophagy</strong> 4 (2008).[10] M. Komatsu, T. Ueno, S. Waguri, Y. Uchiyama, E. Kominami, K.Tanaka, Constitutive autophagy: vital role in clearance <strong>of</strong>unfavorable proteins in neurons, Cell Death Differ. 14 (2007) 887–894.[11] J.J. Lum, D.E. Bauer, M. Kong, M.H. Harris, C. Li, T. Lindsten, C.B.Thompson, Growth factor regulation <strong>of</strong> autophagy <strong>and</strong> cell survivalin <strong>the</strong> absence <strong>of</strong> apoptosis, Cell 120 (2005) 237–248.[12] B. Levine, G. Kroemer, <strong>Autophagy</strong> in <strong>the</strong> pathogenesis <strong>of</strong> disease, Cell132 (2008) 27–42.[13] N. Mizushima, B. Levine, A.M. Cuervo, D.J. Klionsky, <strong>Autophagy</strong> fightsdisease through cellular self-digestion, Nature 451 (2008) 1069–1075.[14] A.L. Harris, Hypoxia – a key regulatory factor in tumour growth, Nat.Rev. Cancer 2 (2002) 38–47.[15] R.K. Jain, Molecular regulation <strong>of</strong> vessel maturation, Nat. Med. 9(2003) 685–693.[16] P. Vaupel, O. Thews, M. Hoeckel, Treatment resistance <strong>of</strong> solidtumors: role <strong>of</strong> hypoxia <strong>and</strong> anemia, Med. Oncol. 18 (2001) 243–259.[17] M. Kitano, M. Kudo, K. Maekawa, Y. Suetomi, H. Sakamoto, N. Fukuta,R. Nakaoka, T. Kawasaki, Dynamic imaging <strong>of</strong> pancreatic diseases bycontrast enhanced coded phase inversion harmonicultrasonography, Gut 53 (2004) 854–859.[18] K. Izuishi, K. Kato, T. Ogura, T. Kinoshita, H. Esumi, Remarkabletolerance <strong>of</strong> tumor cells to nutrient deprivation: possible newbiochemical target for <strong>cancer</strong> <strong>the</strong>rapy, Cancer Res. 60 (2000) 6201–6207.[19] D.A. Nelson, T.T. Tan, A.B. Rabson, D. Anderson, K. Degenhardt, E.White, Hypoxia <strong>and</strong> defective apoptosis drive genomic instability<strong>and</strong> tumorigenesis, Genes Dev. 18 (2004) 2095–2107.[20] G.L. Semenza, Mitochondrial autophagy: life <strong>and</strong> breath <strong>of</strong> <strong>the</strong> cell,<strong>Autophagy</strong> 4 (2008).[21] S. Friberg, S. Mattson, On <strong>the</strong> growth rates <strong>of</strong> human malignanttumors: implications for medical decision making, J. Surg. Oncol. 65(1997) 284–297.[22] D.A. Rew, G.D. Wilson, Cell production rates in human tissues <strong>and</strong>tumours <strong>and</strong> <strong>the</strong>ir significance. Part II: clinical data, Eur. J. Surg.Oncol. 26 (2000) 405–417.[23] K. Heidenreich, J.A. Hill, M. Hoyer-Hansen, X. Hu, W.P. Huang, A.Iwasaki, M. Jaattela, W.T. Jackson, X. Jiang, S. Jin, T. Johansen, J.U.Jung, M. Kadowaki, C. Kang, A. Kelekar, D.H. Kessel, J.A. Kiel, H.P. Kim,A. Kimchi, T.J. Kinsella, K. Kiselyov, K. Kitamoto, E. Knecht, et al,Guidelines for <strong>the</strong> use <strong>and</strong> interpretation <strong>of</strong> assays for monitoringautophagy in higher eukaryotes, <strong>Autophagy</strong> 4 (2008) 151–175.[24] G. Rez, S. Toth, Z. Palfia, Cellular autophagic capacity is highlyincreased in azaserine-induced premalignant atypical acinar nodulecells, Carcinogenesis 20 (1999) 1893–1898.[25] E. Ogier-Denis, J.J. Houri, C. Bauvy, P. Codogno, Guanine nucleotideexchange on heterotrimeric Gi3 protein controls autophagicsequestration in HT-29 cells, J. Biol. Chem. 271 (1996) 28593–28600.[26] K. Sato, K. Tsuchihara, S. Fujii, M. Sugiyama, T. Goya, Y. Atomi, T.Ueno, A. Ochiai, H. Esumi, <strong>Autophagy</strong> is activated in colorectal<strong>cancer</strong> cells <strong>and</strong> contributes to <strong>the</strong> tolerance to nutrient deprivation,Cancer Res. 67 (2007) 9677–9684.[27] S.H. Lee, E.G. Jeong, N.J. Yoo, Mutational <strong>and</strong> expressional analysis <strong>of</strong>BNIP3, a pro-apoptotic Bcl-2 member, in gastric carcinomas, APMIS115 (2007) 1274–1280.[28] K. Degenhardt, R. Ma<strong>the</strong>w, B. Beaudoin, K. Bray, D. Anderson, G.Chen, C. Mukherjee, Y. Shi, C. Gelinas, Y. Fan, D.A. Nelson, S. Jin, E.White, <strong>Autophagy</strong> promotes tumor cell survival <strong>and</strong> restrictsnecrosis, inflammation, <strong>and</strong> tumorigenesis, Cancer Cell 10 (2006)51–64.[29] V. Karantza-Wadsworth, S. Patel, O. Kravchuk, G. Chen, R. Ma<strong>the</strong>w, S.Jin, E. White, <strong>Autophagy</strong> mitigates metabolic stress <strong>and</strong> genomedamage in mammary tumorigenesis, Genes Dev. 21 (2007) 1621–1635.[30] R. Ma<strong>the</strong>w, S. Kongara, B. Beaudoin, C.M. Karp, K. Bray, K.Degenhardt, G. Chen, S. Jin, E. White, <strong>Autophagy</strong> suppresses tumorprogression by limiting chromosomal instability, Genes Dev. 21(2007) 1367–1381.[31] M.A. Qadir, B. Kwok, W.H. Dragowska, K.H. To, D. Le, M.B. Bally, S.M.Gorski, Macroautophagy inhibition sensitizes tamoxifen-resistantbreast <strong>cancer</strong> cells <strong>and</strong> enhances mitochondrial depolarization,Breast Cancer Res. Treat. (2008).[32] R.K. Amaravadi, D. Yu, J.J. Lum, T. Bui, M.A. Christophorou, G.I. Evan,A. Thomas-Tikhonenko, C.B. Thompson, <strong>Autophagy</strong> inhibitionenhances <strong>the</strong>rapy-induced apoptosis in a Myc-induced model <strong>of</strong>lymphoma, J. Clin. Invest. 117 (2007) 326–336.[33] E. Tasdemir, M.C. Maiuri, L. Galluzzi, I. Vitale, M. Djavaheri-Mergny,M. D’Amelio, A. Criollo, E. Morselli, C. Zhu, F. Harper, U. Nannmark, C.Samara, P. Pinton, J.M. Vicencio, R. Carnuccio, U.M. Moll, F. Madeo, P.Paterlini-Brechot, R. Rizzuto, G. Szabadkai, G. Pierron, K. Blomgren,N. Tavernarakis, P. Codogno, F. Cecconi, G. Kroemer, Regulation <strong>of</strong>autophagy by cytoplasmic p53, Nat. Cell Biol. 10 (2008) 676–687.[34] N. Furuya, J. Yu, M. Byfield, S. Pattingre, B. Levine, The evolutionarilyconserved domain <strong>of</strong> Beclin 1 is required for Vps34 binding,autophagy <strong>and</strong> tumor suppressor function, <strong>Autophagy</strong> 1 (2005)46–52.[35] X. Zeng, J.H. Overmeyer, W.A. Maltese, Functional specificity <strong>of</strong> <strong>the</strong>mammalian Beclin-Vps34 PI 3-kinase complex in macroautophagyversus endocytosis <strong>and</strong> lysosomal enzyme trafficking, J. Cell Sci. 119(2006) 259–270.[36] X.H. Liang, S. Jackson, M. Seaman, K. Brown, B. Kempkes, H.Hibshoosh, B. Levine, Induction <strong>of</strong> autophagy <strong>and</strong> inhibition <strong>of</strong>tumorigenesis by beclin 1, Nature 402 (1999) 672–676.[37] V.M. Aita, X.H. Liang, V.V. Murty, D.L. Pincus, W. Yu, E. Cayanis, S.Kalachikov, T.C. Gilliam, B. Levine, Cloning <strong>and</strong> genomic organization<strong>of</strong> beclin 1, a c<strong>and</strong>idate tumor suppressor gene on chromosome17q21, Genomics 59 (1999) 59–65.[38] X. Qu, J. Yu, G. Bhagat, N. Furuya, H. Hibshoosh, A. Troxel, J. Rosen,E.L. Eskelinen, N. Mizushima, Y. Ohsumi, G. Cattoretti, B. Levine,Promotion <strong>of</strong> tumorigenesis by heterozygous disruption <strong>of</strong> <strong>the</strong>beclin 1 autophagy gene, J. Clin. Invest. 112 (2003) 1809–18020.[39] Z. Yue, S. Jin, C. Yang, A.J. Levine, N. Heintz, Beclin 1, an autophagygene essential for early embryonic development, is ahaploinsufficient tumor suppressor, Proc. Natl. Acad. Sci. USA 100(2003) 15077–15082.[40] C. Liang, P. Feng, B. Ku, I. Dotan, D. Canaani, B.H. Oh, J.U. Jung,Autophagic <strong>and</strong> tumour suppressor activity <strong>of</strong> a novel Beclin1-binding protein UVRAG, Nat. Cell Biol. 8 (2006) 688–699.[41] C. Liang, J.S. Lee, K.S. Inn, M.U. Gack, Q. Li, E.A. Roberts, I. Vergne, V.Deretic, P. Feng, C. Akazawa, J.U. Jung, Beclin1-binding UVRAG targets<strong>the</strong> class C Vps complex to coordinate autophagosome maturation<strong>and</strong> endocytic trafficking, Nat. Cell Biol. 10 (2008) 776–787.[42] Y. Takahashi, D. Coppola, N. Matsushita, H.D. Cualing, M. Sun, Y. Sato,C. Liang, J.U. Jung, J.Q. Cheng, J.J. Mul, W.J. Pledger, H.G. Wang, Bif-1interacts with Beclin 1 through UVRAG <strong>and</strong> regulates autophagy <strong>and</strong>tumorigenesis, Nat. Cell Biol. 9 (2007) 1142–1151.[43] B. Ravikumar, C. Vacher, Z. Berger, J.E. Davies, S. Luo, L.G. Oroz, F.Scaravilli, D.F. Easton, R. Duden, C.J. O’Kane, D.C. Rubinsztein,Inhibition <strong>of</strong> mTOR induces autophagy <strong>and</strong> reduces toxicity <strong>of</strong>polyglutamine expansions in fly <strong>and</strong> mouse models <strong>of</strong> Huntingtondisease, Nat. Genet. 36 (2004) 585–595.[44] S. Arico, A. Petiot, C. Bauvy, P.F. Dubbelhuis, A.J. Meijer, P. Codogno,E. Ogier-Denis, The tumor suppressor PTEN positively regulatesmacroautophagy by inhibiting <strong>the</strong> phosphatidylinositol 3-kinase/protein kinase B pathway, J. Biol. Chem. 276 (2001) 35243–35246.[45] Z. Feng, H. Zhang, A.J. Levine, S. Jin, The coordinate regulation <strong>of</strong> <strong>the</strong>p53 <strong>and</strong> mTOR pathways in cells, Proc. Natl. Acad. Sci. USA 102(2005) 8204–8209.[46] H. Takeuchi, Y. Kondo, K. Fujiwara, T. Kanzawa, H. Aoki, G.B. Mills, S.Kondo, Synergistic augmentation <strong>of</strong> rapamycin-induced autophagyin malignant glioma cells by phosphatidylinositol 3-kinase/proteinkinase B inhibitors, Cancer Res. 65 (2005) 3336–3346.[47] D. Crighton, S. Wilkinson, J. O’Prey, N. Syed, P. Smith, P.R. Harrison,M. Gasco, O. Garrone, T. Crook, K.M. Ryan, DRAM, a p53-inducedmodulator <strong>of</strong> autophagy, is critical for apoptosis, Cell 126 (2006)121–134.Please cite this article in press as: K. Tsuchihara et al., <strong>Autophagy</strong> <strong>and</strong> <strong>cancer</strong>: <strong>Dynamism</strong> <strong>of</strong> <strong>the</strong> metabolism <strong>of</strong> tumor cells..., Cancer Lett. (2008), doi:10.1016/j.canlet.2008.09.040


ARTICLE IN PRESSK. Tsuchihara et al. / Cancer Letters xxx (2008) xxx–xxx 9[48] B. Inbal, S. Bialik, I. Sabanay, G. Shani, A. Kimchi, DAP kinase <strong>and</strong>DRP-1 mediate membrane blebbing <strong>and</strong> <strong>the</strong> formation <strong>of</strong> autophagicvesicles during programmed cell death, J. Cell Biol. 157 (2002) 455–468.[49] P.G. Clarke, Developmental cell death: morphological diversity <strong>and</strong>multiple mechanisms, Anat. Embryol. Berl. 181 (1990) 195–213.[50] Y. Kondo, T. Kanzawa, R. Sawaya, S. Kondo, The role <strong>of</strong> autophagy in<strong>cancer</strong> development <strong>and</strong> response to <strong>the</strong>rapy, Nat. Rev. Cancer 5(2005) 726–734.[51] M.C. Maiuri, E. Zalckvar, A. Kimchi, G. Kroemer, Self-eating <strong>and</strong> selfkilling:crosstalk between autophagy <strong>and</strong> apoptosis, Nat. Rev. Mol.Cell Biol. 8 (2007) 741–752.[52] A. Mantovani, T. Schioppa, C. Porta, P. Allavena, A. Sica, Role <strong>of</strong>tumor-associated macrophages in tumor progression <strong>and</strong> invasion,Cancer Metastasis Rev. 25 (2006) 315–322.[53] H. Aoki, Y. Kondo, K. Aldape, A. Yamamoto, E. Iwado, T. Yokoyama,E.F. Hollingsworth, R. Kobayashi, K. Hess, N. Shinojima, T. Shingu, Y.Tamada, L. Zhang, C. Conrad, O. Bogler, G. Mills, R. Sawaya, S. Kondo,Monitoring autophagy in glioblastoma with antibody againstis<strong>of</strong>orm b <strong>of</strong> human microtubule-associated protein 1 light chain3, <strong>Autophagy</strong> 4 (2008).[54] S. Jager, C. Bucci, I. Tanida, T. Ueno, E. Kominami, P. Saftig, E.L.Eskelinen, Role for Rab7 in maturation <strong>of</strong> late autophagic vacuoles, J.Cell Sci. 117 (2004) 4837–4848.[55] S. Fujii, S. Mitsunaga, M. Yamazaki, T. Hasebe, G. Ishii, M. Kojima, T.Kinoshita, T. Ueno, H. Esumi, A. Ochiai, <strong>Autophagy</strong> is activated inpancreatic <strong>cancer</strong> cells <strong>and</strong> correlates with poor patient outcome,Cancer Sci. 99 (2008) 1813–1819.[56] D.C. Rubinsztein, J.E. Gestwicki, L.O. Murphy, D.J. Klionsky, Potential<strong>the</strong>rapeutic applications <strong>of</strong> autophagy, Nat. Rev. Drug Discov. 6(2007) 304–312.[57] T. Yorimitsu, D.J. Klionsky, Eating <strong>the</strong> endoplasmic reticulum:quality control by autophagy, Trends Cell Biol. 17 (2007) 279–285.Please cite this article in press as: K. Tsuchihara et al., <strong>Autophagy</strong> <strong>and</strong> <strong>cancer</strong>: <strong>Dynamism</strong> <strong>of</strong> <strong>the</strong> metabolism <strong>of</strong> tumor cells..., Cancer Lett. (2008), doi:10.1016/j.canlet.2008.09.040

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!