Cancer may be a pathway to cell survival under persistent hypoxia and elevated ROS: A model for solid-cancer initiation and early development
Chi Zhang
Computational Systems Biology Laboratory, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA
Search for more papers by this authorSha Cao
Computational Systems Biology Laboratory, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA
Search for more papers by this authorBryan P. Toole
Department of Regenerative Medicine and Cell Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
Search for more papers by this authorCorresponding Author
Ying Xu
Computational Systems Biology Laboratory, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA
College of Computer Science and Technology, Jilin University, Changchun, Jilin, China
Correspondence to: Ying Xu, Computational Systems Biology Laboratory, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA, USA, Tel.: 706-542-9779, E-mail: [email protected]Search for more papers by this authorChi Zhang
Computational Systems Biology Laboratory, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA
Search for more papers by this authorSha Cao
Computational Systems Biology Laboratory, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA
Search for more papers by this authorBryan P. Toole
Department of Regenerative Medicine and Cell Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC
Search for more papers by this authorCorresponding Author
Ying Xu
Computational Systems Biology Laboratory, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA
College of Computer Science and Technology, Jilin University, Changchun, Jilin, China
Correspondence to: Ying Xu, Computational Systems Biology Laboratory, Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA, USA, Tel.: 706-542-9779, E-mail: [email protected]Search for more papers by this authorAbstract
A number of proposals have been made in the past century regarding what may drive sporadic cancers to initiate and develop. Yet the problem remains largely unsolved as none of the proposals have been widely accepted as cancer-initiation drivers. We propose here a driver model for the initiation and early development of solid cancers associated with inflammation-induced chronic hypoxia and reactive oxygen species (ROS) accumulation. The model consists of five key elements: (i)human cells tend to have a substantial gap between ATP demand and supply during chronic hypoxia, which would inevitably lead to increased uptake of glucose and accumulation of its metabolites; (ii) the accumulation of these metabolites will cast mounting pressure on the cells and ultimately result in the production and export of hyaluronic acid; (iii) the exported hyaluronic acid will be degraded into fragments of various sizes, serving as tissue-repair signals, including signals for cell proliferation, cell survival and angiogenesis, which lead to the initial proliferation of the underlying cells; (iv) cell division provides an exit for the accumulated glucose metabolites using them towards macromolecular synthesis for the new cell, and hence alleviate the pressure from the metabolite accumulation; and (v) this process continues as long as the hypoxic condition persists. In tandem, genetic mutations may be selected to make cell divisions and hence survival more sustainable and efficient, also increasingly more uncontrollable. This model also applies to some hereditary cancers as their key mutations, such as BRCA for breast cancer, generally lead to increased ROS and ultimately to repression of mitochondrial activities and up-regulation of glycolysis, as well as hypoxia; hence the energy gap, glucose-metabolite accumulation, hyaluronic acid production and continuous cell division for survival.
Supporting Information
Additional Supporting Information may be found in the online version of this article.
Filename | Description |
---|---|
ijc28975-sup-0001-suppinfo.doc2.3 MB |
Supplementary Information |
Please note: The publisher is not responsible for the content or functionality of any supporting information supplied by the authors. Any queries (other than missing content) should be directed to the corresponding author for the article.
References
- 1Warburg O. On the origin of cancer cells. Science 1956; 123: 309–14.
- 2Munoz N, Bosch FX, de Sanjose S, et al. Epidemiologic classification of human papillomavirus types associated with cervical cancer. N Engl J Med 2003; 348: 518–27.
- 3Perz JF, Armstrong GL, Farrington LA, et al. The contributions of hepatitis B virus and hepatitis C virus infections to cirrhosis and primary liver cancer worldwide. J Hepatol 2006; 45: 529–38.
- 4DeBerardinis RJ, Lum JJ, Hatzivassiliou G, et al. The biology of cancer: metabolic reprogramming fuels cell growth and proliferation. Cell Metab 2008; 7: 11–20.
- 5Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 2009; 324: 1029–33.
- 6Stehelin D, Varmus HE, Bishop JM, et al. DNA related to the transforming gene(s) of avian sarcoma viruses is present in normal avian DNA. Nature 1976; 260: 170–73.
- 7Knudson AG. Mutation and cancer: statistical study of retinoblastoma. Proc Natl Acad Sci USA 1971; 68: 820–23.
- 8Nishisho I, Nakamura Y, Miyoshi Y, et al. Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science 1991; 2253: 665–69.
- 9Nowell PC, Hungerford DA. A minute chromosome in human chronic granulocytic leukemia. Science 1960; 142.
- 10Greenman C, Stephens P, Smith R, et al. Patterns of somatic mutation in human cancer genomes. Nature 2007; 446: 153–8.
- 11Zhu X, Assoian RK. Integrin-dependent activation of MAP kinase: a link to shape-dependent cell proliferation. Mol Biol Cell 1995; 6: 273–82.
- 12Senoo H, Hata R. Extracellular matrix regulates cell morphology, proliferation, and tissue formation. Kaibogaku zasshi. J Anat 1994; 69: 719–33.
- 13Wells RG. The role of matrix stiffness in regulating cell behavior. Hepatology 2008; 47: 1394–400.
- 14Seluanov A, Hine C, Azpurua J, et al. Hypersensitivity to contact inhibition provides a clue to cancer resistance of naked mole-rat. Proc Natl Acad Sci USA 2009; 106: 19352–7.
- 15Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144: 646–74.
- 16Huang S, Ingber DE. Shape-dependent control of cell growth, differentiation, and apoptosis: switching between attractors in cell regulatory networks. Exp Cell Res 2000; 261: 91–103.
- 17Beachy PA, Karhadkar SS, Berman DM. Tissue repair and stem cell renewal in carcinogenesis. Nature 2004; 432: 324–31.
- 18Akagi K, Li J, Broutian TR, et al. Genome-wide analysis of HPV integration in human cancers reveals recurrent, focal genomic instability. Genome Res 2014; 24: 185–99.
- 19Chandel NS, Maltepe E, Goldwasser E, et al. Mitochondrial reactive oxygen species trigger hypoxia-induced transcription. Proc Natl Acad Sci USA 1998; 95: 11715–20.
- 20Bartrons R, Caro J. Hypoxia, glucose metabolism and the Warburg's effect. J Bioenerg Biomembr 2007; 39: 223–9.
- 21Elinav E, Nowarski R, Thaiss CA, et al. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer 2013; 13: 759–71.
- 22Shacter E, Weitzman SA. Chronic inflammation and cancer. Oncology 2002; 16: 217–26, 29; discussion 30–2.
- 23Vaupel P, Mayer A. Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev 2007; 26: 225–39.
- 24Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N Engl J Med 2011; 364: 656–65.
- 25Bartels K, Grenz A, Eltzschig HK. Hypoxia and inflammation are two sides of the same coin. Proc Natl Acad Sci USA 2013; 110: 18351–2.
- 26Gupta SC, Hevia D, Patchva S, et al. Upsides and downsides of reactive oxygen species for cancer: the roles of reactive oxygen species in tumorigenesis, prevention, and therapy. Antioxid Redox Signal 2012; 16: 1295–322.
- 27Frezza C, Zheng L, Tennant DA, et al. Metabolic profiling of hypoxic cells revealed a catabolic signature required for cell survival. PLoS One 2011; 6: e24411.
- 28Yevdokimova NY. Elevated level of ambient glucose stimulates the synthesis of high-molecular-weight hyaluronic acid by human mesangial cells. The involvement of transforming growth factor beta1 and its activation by thrombospondin-1. Acta Biochim Pol 2006; 53: 383–93.
- 29Stern R, Asari AA, Sugahara KN. Hyaluronan fragments: an information-rich system. Eur J Cell Biol 2006; 85: 699–715.
- 30Ruben LN, Clothier RH, Balls M. Cancer resistance in amphibians. Altern Lab Anim 2007; 35: 463–70.
- 31Manov I, Hirsh M, Iancu TC, et al. Pronounced cancer resistance in a subterranean rodent, the blind mole-rat, Spalax: in vivo and in vitro evidence. BMC Biol 2013; 11: 91.
- 32Tian X, Azpurua J, Hine C, et al. High-molecular-mass hyaluronan mediates the cancer resistance of the naked mole rat. Nature 2013; 499: 346–9.
- 33Krivoruchko A, Storey KB. Forever young: mechanisms of natural anoxia tolerance and potential links to longevity. Oxid Med Cell Longev 2010; 3: 186–98.
- 34Caulin AF, Maley CC. Peto's Paradox: evolution's prescription for cancer prevention. Trends Ecol Evol 2011; 26: 175–82.
- 35Rolfe D, Brown G. Cellular energy utilization and molecular origin of standard metabolic rate in mammals. Physiol Rev 1997; 77: 731–58.
- 36Buttgereit F, Brand MD. A hierarchy of ATP-consuming processes in mammalian cells. Biochem J 1995; 312 (Pt 1): 163–7.
- 37Nathaniel TI, Otukonyong E, Abdellatif A, et al. Effect of hypoxia on metabolic rate, core body temperature, and c-fos expression in the naked mole rat. Int J Dev Neurosci 2012; 30: 539–44.
- 38Hochachka PW, Buck LT, Doll CJ, et al. Unifying theory of hypoxia tolerance: molecular/metabolic defense and rescue mechanisms for surviving oxygen lack. Proc Natl Acad Sci USA 1996; 93: 9493–8.
- 39St-Pierre J, Brand MD, Boutilier RG. The effect of metabolic depression on proton leak rate in mitochondria from hibernating frogs. J Exp Biol 2000; 203: 1469–76.
- 40Kim EB, Fang X, Fushan AA, et al. Genome sequencing reveals insights into physiology and longevity of the naked mole rat. Nature 2011; 479: 223–7.
- 41Azpurua J, Seluanov A. Long-lived cancer-resistant rodents as new model species for cancer research. Front Genet 2012; 3: 319.
- 42Shams I, Avivi A, Nevo E. Hypoxic stress tolerance of the blind subterranean mole rat: expression of erythropoietin and hypoxia-inducible factor 1 alpha. Proc Natl Acad Sci USA 2004; 101: 9698–703.
- 43Kano M, Tsutsumi S, Kawahara N, et al. A meta-clustering analysis indicates distinct pattern alteration between two series of gene expression profiles for induced ischemic tolerance in rats. Physiol Genomics 2005; 21: 274–83.
- 44Gesser H, Johansen K, Maloiy GM. Tissue metabolism and enzyme activities in the rodent Heterocephalus glaber, a poor temperature regulator. Comp Biochem Physiol 1977; 57: 293–6.
- 45Schwanhausser B, Busse D, Li N, et al. Global quantification of mammalian gene expression control. Nature 2011; 473: 337–42.
- 46Widmer HR, Hoppeler H, Nevo E, et al. Working underground: respiratory adaptations in the blind mole rat. Proc Natl Acad Sci USA 1997; 94: 2062–7.
- 47Edrey YH, Park TJ, Kang H, et al. Endocrine function and neurobiology of the longest-living rodent, the naked mole-rat. Exp Gerontol 2011; 46: 116–23.
- 48Bickler PE, Buck LT. Hypoxia tolerance in reptiles, amphibians, and fishes: life with variable oxygen availability. Annu Rev Physiol 2007; 69: 145–70.
- 49Ramirez J, Folkow L, Blix A. Hypoxia tolerance in mammals and birds: from the wilderness to the clinic. Annu Rev Physiol 2007; 69: 113–43.
- 50Kubasiak LA, Hernandez OM, Bishopric NH, et al. Hypoxia and acidosis activate cardiac myocyte death through the Bcl-2 family protein BNIP3. Proc Natl Acad Sci USA 2002; 99: 12825–30.
- 51Schaffer J. Lipotoxicity: when tissues overeat. Curr Opin Lipidol 2003; 14: 281–7.
- 52Stern R. Hyaluronan in cancer biology, 1st edn. San Diego, CA: Academic Press, 2009.
10.1016/B978-012374178-3.10001-8 Google Scholar
- 53Toole B. Hyaluronan-CD44 interactions in cancer: paradoxes and possibilities. Clin Cancer Res 2009; 15: 7462–68.
- 54Sironen R, Tammi M, Tammi R, et al. Hyaluronan in human malignancies. Exp Cell Res 2011; 317: 383–91.
- 55Jiang D, Liang J, Noble PW. Hyaluronan in tissue injury and repair. Annu Rev Cell Dev Biol 2007; 23: 435–61.
- 56Tammi RH, Passi AG, Rilla K, et al. Transcriptional and post-translational regulation of hyaluronan synthesis. FEBS J 2011; 278: 1419–28.
- 57Fantus IG, Goldberg H, Whiteside C, et al. The hexosamine biosynthesis pathway. Contemporary diabetes. The diabetic kidney. 2006. Chapter 7: 117–33.
- 58Guillaumond F, Leca J, Olivares O, et al. Strengthened glycolysis under hypoxia supports tumor symbiosis and hexosamine biosynthesis in pancreatic adenocarcinoma. Proc Natl Acad Sci USA 2013; 110: 3919–24.
- 59Pelletier J, Bellot G, Gounon P, et al. Glycogen synthesis is induced in hypoxia by the hypoxia-inducible factor and promotes cancer cell survival. Front Oncol 2012; 2: 18.
- 60Pescador N, Villar D, Cifuentes D, et al. Hypoxia promotes glycogen accumulation through hypoxia inducible factor (HIF)-mediated induction of glycogen synthase 1. PLoS One 2010; 5: e9644.
- 61Guillaumond F, Leca J, Olivares O, et al. Strengthened glycolysis under hypoxia supports tumor symbiosis and hexosamine biosynthesis in pancreatic adenocarcinoma. Proc Natl Acad Sci USA 2013; 110: 3919–24.
- 62Bontemps Y, Vuillermoz B, Antonicelli F, et al. Specific protein-1 is a universal regulator of UDP-glucose dehydrogenase expression: its positive involvement in transforming growth factor-beta signaling and inhibition in hypoxia. J Biol Chem 2003; 278: 21566–75.
- 63Melillo G. Hypoxia: jump-starting inflammation. Blood 2011; 117: 2561–2.
- 64Wahl SM. Transforming growth factor beta (TGF-beta) in inflammation: a cause and a cure. J Clin Immunol 1992; 12: 61–74.
- 65Medina AP, Lin J, Weigel PH. Hyaluronan synthase mediates dye translocation across liposomal membranes. BMC Biochem 2012; 13: 2.
- 66Schulz T, Schumacher U, Prehm P. Hyaluronan export by the ABC transporter MRP5 and its modulation by intracellular cGMP. J Biol Chem 2007; 282: 20999–1004.
- 67Agren UM, Tammi RH, Tammi MI. Reactive oxygen species contribute to epidermal hyaluronan catabolism in human skin organ culture. Free Radic Biol Med 1997; 23: 996–1001.
- 68Gao F, Okunieff P, Han Z, et al. Hypoxia-induced alterations in hyaluronan and hyaluronidase. Adv Exp Med Biol 2005; 566: 249–56.
- 69Tusher VG, Tibshirani R, Chu G. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA 2001; 98: 5116–21.
- 70Schulz T, Schumacher U, Prante C, et al. Cystic fibrosis transmembrane conductance regulator can export hyaluronan. Pathobiology 2010; 77: 200–9.
- 71Xu H, Ito T, Tawada A, et al. Effect of hyaluronan oligosaccharides on the expression of heat shock protein 72. J Biol Chem 2002; 277: 17308–14.
- 72Tsatas D, Kanagasundaram V, Kaye A, et al. EGF receptor modifies cellular responses to hyaluronan in glioblastoma cell lines. J Clin Neurosci 2002; 9: 282–8.
- 73Savani RC, Zhao H, Cui Z, et al. Increased inflammation, hyaluronan & respiratory distress in mice overexpressing the hyaluronan receptor RHAMM in macrophages. FASEB J 2007; 21: 374.8.
- 74Kosaki R, Watanabe K, Yamaguchi Y. Overproduction of hyaluronan by expression of the hyaluronan synthase Has2 enhances anchorage-independent growth and tumorigenicity. Cancer Res 1999; 59: 1141–5.
- 75Toole BP. Hyaluronan promotes the malignant phenotype. Glycobiology 2002; 12: 37R–42R.
- 76Itano N, Atsumi F, Sawai T, et al. Abnormal accumulation of hyaluronan matrix diminishes contact inhibition of cell growth and promotes cell migration. Proc Natl Acad Sci USA 2002; 99: 3609–14.
- 77Hashimoto K, Fukuda K, Yamazaki K, et al. Hypoxia-induced hyaluronan synthesis by articular chondrocytes: the role of nitric oxide. Inflamm Res 2006; 55: 72–7.
- 78Stenfeldt A-L, Wennerås C. Danger signals derived from stressed and necrotic epithelial cells activate human eosinophils. Immunology 2004; 112: 605–14.
- 79Zong W-X, Thompson CB. Necrotic death as a cell fate. Genes Dev 2006; 20: 1–15.
- 80Kayyali US, Pennella CM, Trujillo C, et al. Cytoskeletal changes in hypoxic pulmonary endothelial cells are dependent on MAPK-activated protein kinase MK2. J Biol Chem 2002; 277: 42596–602.
- 81Assoian RK, Zhu X. Cell anchorage and the cytoskeleton as partners in growth factor dependent cell cycle progression. Curr Opin Cell Biol 1997; 9: 93–98.
- 82Théry M, Bornens M. Cell shape and cell division. Curr Opin Cell Biol 2006; 18: 648–57.
- 83Bedogni B, Welford SM, Cassarino DS, et al. The hypoxic microenvironment of the skin contributes to Akt-mediated melanocyte transformation. Cancer Cell 2005; 8: 443–54.
- 84Nishi H, Nakada T, Kyo S, et al. Hypoxia-inducible factor 1 mediates upregulation of telomerase (hTERT). Mol Cell Biol 2004; 24: 6076–83.
- 85Bristow R, Hill R. Hypoxia and metabolism. Hypoxia, DNA repair and genetic instability. Nat Rev Cancer 2008; 8: 180–92.
- 86Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med 2003; 9: 677–84.
- 87Fujiwara S, Nakagawa K, Harada H, et al. Silencing hypoxia-inducible factor-1alpha inhibits cell migration and invasion under hypoxic environment in malignant gliomas. Int J Oncol 2007; 30: 793–802.
- 88Yabu M, Shime H, Hara H, et al. IL-23-dependent and -independent enhancement pathways of IL-17A production by lactic acid. Int Immunol 2011; 23: 29–41.
- 89Beckert S, Farrahi F, Aslam RS, et al. Lactate stimulates endothelial cell migration. Wound Repair Regen 2006; 14: 321–24.
- 90Hirschhaeuser F, Sattler UGA, Mueller-Klieser W. Lactate: a metabolic key player in cancer. Cancer Res 2011; 71: 6921–25.
- 91Satgé D. Analysis of somatic mutations in cancer tissues challenges the somatic mutation theory of cancer. eLS. 2013. DOI: doi/10.1002/9780470015902.a0024465.
10.1002/9780470015902.a0024465 Google Scholar
- 92Soto AM, Sonnenschein C. The somatic mutation theory of cancer: growing problems with the paradigm? BioEssays 2004; 26: 1097–107.
- 93Mazurek S, Boschek C, Hugo F, et al. Pyruvate kinase type M2 and its role in tumor growth and spreading. Semin Cancer Biol 2005; 15: 300–8.
- 94Anastasiou D, Poulogiannis G, Asara JM, et al. Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses. Science 2011; 334: 1278–83.
- 95Sgambato A, Cittadini A, Faraglia B, et al. Multiple functions of p27Kip1 and its alterations in tumor cells: a review. J Cell Physiol 2000; 183: 18–27.
10.1002/(SICI)1097-4652(200004)183:1<18::AID-JCP3>3.0.CO;2-S CAS PubMed Web of Science® Google Scholar
- 96Kim S, Chin K, Gray JW, et al. A screen for genes that suppress loss of contact inhibition: identification of ING4 as a candidate tumor suppressor gene in human cancer. Proc Natl Acad Sci USA 2004; 101: 16251–6.
- 97Kim S, Welm AL, Bishop JM. A dominant mutant allele of the ING4 tumor suppressor found in human cancer cells exacerbates MYC-initiated mouse mammary tumorigenesis. Cancer Res 2010; 70: 5155–62.
- 98McFarland CD, Korolev KS, Kryukov GV, et al. Impact of deleterious passenger mutations on cancer progression. Proc Natl Acad Sci USA 2013; 110: 2910–5.
- 99Easton DF, Ford D, Bishop DT. Breast and ovarian cancer incidence in BRCA1-mutation carriers. Breast Cancer Linkage Consortium. Am J Hum Genet 1995; 56: 265–71.
- 100Toro JR, Nickerson ML, Wei MH, et al. Mutations in the fumarate hydratase gene cause hereditary leiomyomatosis and renal cell cancer in families in North America. Am J Hum Genet 2003; 73: 95–106.
- 101Morin PJ, Sparks AB, Korinek V, et al. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 1997; 275: 1787–90.
- 102Hogg A, Onadim Z, Baird PN, et al. Detection of heterozygous mutations in the RB1 gene in retinoblastoma patients using single-strand conformation polymorphism analysis and polymerase chain reaction sequencing. Oncogene 1992; 7: 1445–51.
- 103Malkin D, Li FP, Strong LC, et al. Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science 1990; 250: 1233–8.
- 104Liaw D, Marsh DJ, Li J, et al. Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat Genet 1997; 16: 64–7.
- 105Iliopoulos O, Kibel A, Gray S, et al. Tumour suppression by the human von Hippel-Lindau gene product. Nat Med 1995; 1: 822–6.
- 106Martinez-Outschoorn U, Balliet R, Lin Z, et al. BRCA1 mutations drive oxidative stress and glycolysis in the tumor microenvironment: implications for breast cancer prevention with antioxidant therapies. Cell Cycle 2012; 11: 4402–13.
- 107Sudarshan S, Sourbier C, Kong H-S, et al. Fumarate hydratase deficiency in renal cancer induces glycolytic addiction and hypoxia-inducible transcription factor 1α stabilization by glucose-dependent generation of reactive oxygen species. Mol Cell Biol 2009; 29: 4080–90.
- 108Sunaga N, Kohno T, Kolligs FT, et al. Constitutive activation of the Wnt signaling pathway by CTNNB1 (beta-catenin) mutations in a subset of human lung adenocarcinoma. Genes Chromosomes Cancer 2001; 30: 316–21.
- 109Myant KB, Cammareri P, McGhee EJ, et al. ROS production and NF-κB activation triggered by RAC1 facilitate WNT-driven intestinal stem cell proliferation and colorectal cancer initiation. Cell Stem Cell 2013; 12: 761–73.
- 110Bremner R, Zacksenhaus E. Cyclins, Cdks, E2f, Skp2, and more at the first international RB tumor suppressor meeting. Cancer Res 2010; 70: 6114–8.
- 111Jain AK, Bloom DA, Jaiswal AK. Nuclear import and export signals in control of Nrf2. J Biol Chem 2005; 280: 29158–68.
- 112He X, Ni Y, Wang Y, et al. Naturally occurring germline and tumor-associated mutations within the ATP-binding motifs of PTEN lead to oxidative damage of DNA associated with decreased nuclear p53. Hum Mol Genet 2011; 20: 80–9.
- 113Block K, Gorin Y, New DD, et al. The NADPH oxidase subunit p22phox inhibits the function of the tumor suppressor protein tuberin. Am J Pathol 2010; 176: 2447–55.
- 114Mauro C, Leow SC, Anso E, et al. NF-kappaB controls energy homeostasis and metabolic adaptation by upregulating mitochondrial respiration. Nat Cell Biol 2011; 13: 1272–9.
- 115Campisi J, Andersen J, Kapahi P, et al. Cellular senescence: a link between cancer and age-related degenerative disease? Semin Cancer Biol 2011; 21: 354–9.