Skip to main content

Advertisement

Log in

Emerging tale of UPR and cancer: an essentiality for malignancy

  • Review
  • Published:
Tumor Biology

Abstract

A set of cellular response to counter any alteration in homeostasis of a cell originating at endoplasmic reticulum is collectively termed as unfolded protein response (UPR). It initially is adaptive in nature as to restore cellular normalcy failing in course often activates pro-apoptotic signaling pathway resulting in cell death. UPR has emerged as an essential adaptation mechanism that cross talk with various cellular processes for cancer pathogenesis. Interestingly, it plays diverse role in plethora of signaling pathways instrumental in transformation, cell invasion, cell migration, metastasis, neovascularization, proliferation, and maintenance of energy metabolism of cancerous cells. In cancerous cells, it is triggered by change in microenvironment of a cell usually driven by hypoxia, acidosis, and nutrient deprivation, which often leads to positive selection pressure involving the reprogramming of energy metabolism which promotes channelization of limited metabolites into the hexosamine biosynthetic pathway (HBP). Substantial evidences suggest the role of UPR in oncogene (Myc, mTOR, RAS, HER2) driven cancer transformation and progression. In this review, we have comprehensively underlined the role played by UPR in adaptation, transformation, proliferation, invasion, and metastasis of cancerous cells.

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

Access this article

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

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Abbreviations

HIF-1:

Hypoxia-inducible factor 1

GCN2:

General control nonderepresible 2

HBP:

Hexosamine biosynthetic pathway

FGF2:

Fibroblast growth factor 2

GFAT1:

Glutamine:fructose-6-phosphate amidotransferase 1

VEGF:

Vascular endothelial growth factor

LAMP3:

Lysosomal-associated membrane protein 3

UPR:

Unfolded protein response

CHOP:

C/EBP (CCAAT/enhancer binding protein) homologous protein

ATF4:

Activating transcription factor 4

ATF6:

Activating transcription factor 6

GADD34:

Growth arrest and DNA damage 34

XBP1:

X-box binding protein 1

References

  1. Brewer JW. Regulatory crosstalk within the mammalian unfolded protein response. Cell Mol Life Sci. 2014;71(6):1067–79.

    Article  CAS  PubMed  Google Scholar 

  2. Helenius A, Aebi M. Roles of N-linked glycans in the endoplasmic reticulum. Annu Rev Biochem. 2004;73:1019–49.

    Article  CAS  PubMed  Google Scholar 

  3. Schroder M. Endoplasmic reticulum stress responses. Cell Mol Life Sci. 2008;65(6):862–94.

    Article  CAS  PubMed  Google Scholar 

  4. Berridge MJ. The endoplasmic reticulum: a multifunctional signaling organelle. Cell Calcium. 2002;32(5–6):235–49.

    Article  CAS  PubMed  Google Scholar 

  5. Niforou K, Cheimonidou C, Trougakos IP. Molecular chaperones and proteostasis regulation during redox imbalance. Redox Biol. 2014;2:323–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Arif Bashir, Naveed Nazir Shah, Younis Mohammad Hazari, Mudasir Habib, Samirul Bashir, Nazia Hilal, Mariam Banday, Syed Asrafuzzaman, Khalid Majid Fazili, (2016) Novel variants of SERPIN1A gene: Interplay between alpha1-antitrypsin deficiency and chronic obstructive pulmonary disease. Respir Med 117:139–149

  7. Kim H, Bhattacharya A, Qi L. Endoplasmic reticulum quality control in cancer: friend or foe. Semin Cancer Biol. 2015;33:25–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Dejeans N, Barroso K, Fernandez-Zapico ME, Samali A, Chevet E. Novel roles of the unfolded protein response in the control of tumor development and aggressiveness. Semin Cancer Biol. 2015;33:67–73.

    Article  CAS  PubMed  Google Scholar 

  9. Hollien J, Weissman JS. Decay of endoplasmic reticulum-localized mRNAs during the unfolded protein response. Science (New York, NY). 2006;313(5783):104–7.

    Article  CAS  Google Scholar 

  10. Tirasophon W, Lee K, Callaghan B, Welihinda A, Kaufman RJ. The endoribonuclease activity of mammalian IRE1 autoregulates its mRNA and is required for the unfolded protein response. Genes Dev. 2000;14(21):2725–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yang Q, Kim Y-S, Lin Y, Lewis J, Neckers L, Liu Z-G. Tumour necrosis factor receptor 1 mediates endoplasmic reticulum stress-induced activation of the MAP kinase JNK. EMBO Reports. 2006 05/0510/06/received03/15/revised03/27/accepted;7(6):622–7.

  12. Han D, Lerner AG, Vande Walle L, Upton JP, Xu W, Hagen A, et al. IRE1alpha kinase activation modes control alternate endoribonuclease outputs to determine divergent cell fates. Cell. 2009;138(3):562–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Bertolotti A, Zhang Y, Hendershot LM, Harding HP, Ron D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat Cell Biol. 2000;2(6):326–32.

    Article  CAS  PubMed  Google Scholar 

  14. Koromilas AE. Roles of the translation initiation factor eIF2alpha serine 51 phosphorylation in cancer formation and treatment. Biochim Biophys Acta. 2015;7:871–80.

    Article  CAS  Google Scholar 

  15. Holcik M, Sonenberg N. Translational control in stress and apoptosis. Nat Rev Mol Cell Biol. 2005;6(4):318–27.

    Article  CAS  PubMed  Google Scholar 

  16. Sano R, Reed JCER. Stress-induced cell death mechanisms. Biochim Biophys Acta. 2013;12(70).

  17. Brush MH, Weiser DC, Shenolikar S. Growth arrest and DNA damage-inducible protein GADD34 targets protein phosphatase 1α to the endoplasmic reticulum and promotes dephosphorylation of the α subunit of eukaryotic translation initiation factor 2. Molecular and cellular biology. 2003 09/09/received10/08/revised11/22/accepted;23(4):1292–303.

  18. Yerlikaya A, Kimball SR, Stanley BA. Phosphorylation of eIF2alpha in response to 26S proteasome inhibition is mediated by the haem-regulated inhibitor (HRI) kinase. The Biochemical journal. 2008;412(3):579–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Guo FJ, Xiong Z, Lu X, Ye M, Han X, Jiang R. ATF6 upregulates XBP1S and inhibits ER stress-mediated apoptosis in osteoarthritis cartilage. Cell Signal. 2014;26(2):332–42.

    Article  CAS  PubMed  Google Scholar 

  20. Adachi Y, Yamamoto K, Okada T, Yoshida H, Harada A, Mori K. ATF6 is a transcription factor specializing in the regulation of quality control proteins in the endoplasmic reticulum. Cell Struct Funct. 2008;33(1):75–89.

    Article  CAS  PubMed  Google Scholar 

  21. Okada T, Yoshida H, Akazawa R, Negishi M, Mori K. Distinct roles of activating transcription factor 6 (ATF6) and double-stranded RNA-activated protein kinase-like endoplasmic reticulum kinase (PERK) in transcription during the mammalian unfolded protein response. The Biochemical journal. 2002;366(Pt 2):585–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sutherland RM, Ausserer WA, Murphy BJ, Laderoute KR. Tumor hypoxia and heterogeneity: challenges and opportunities for the future. Semin Radiat Oncol. 1996;6(1):59–70.

    Article  PubMed  Google Scholar 

  23. Dufey E, Urra H, Hetz CER. Proteostasis addiction in cancer biology: novel concepts. Semin Cancer Biol. 2015;33:40–7.

    Article  CAS  PubMed  Google Scholar 

  24. Graeber TG, Osmanian C, Jacks T, Housman DE, Koch CJ, Lowe SW, et al. Hypoxia-mediated selection of cells with diminished apoptotic potential in solid tumours. Nature. 1996;379(6560):88–91.

    Article  CAS  PubMed  Google Scholar 

  25. Blais JD, Addison CL, Edge R, Falls T, Zhao H, Wary K, et al. Perk-dependent translational regulation promotes tumor cell adaptation and angiogenesis in response to hypoxic stress. Mol Cell Biol. 2006;26(24):9517–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Gutierrez T, Simmen T. Endoplasmic reticulum chaperones and oxidoreductases: critical regulators of tumor cell survival and immunorecognition. Front Oncol. 2014;4:291.

    PubMed  PubMed Central  Google Scholar 

  27. Koritzinsky M, Levitin F, van den Beucken T, Rumantir RA, Harding NJ, Chu KC, et al. Two phases of disulfide bond formation have differing requirements for oxygen. J Cell Biol. 2013;203(4):615–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tu BP, Weissman JS. The FAD- and O(2)-dependent reaction cycle of Ero1-mediated oxidative protein folding in the endoplasmic reticulum. Mol Cell. 2002;10(5):983–94.

    Article  CAS  PubMed  Google Scholar 

  29. Semenza GL. Hypoxia-inducible factors in physiology and medicine. Cell. 2012;148(3):399–408.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Maxwell PH, Dachs GU, Gleadle JM, Nicholls LG, Harris AL, Stratford IJ, et al. Hypoxia-inducible factor-1 modulates gene expression in solid tumors and influences both angiogenesis and tumor growth. Proc Natl Acad Sci U S A. 1997;94(15):8104–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ryan HE, Poloni M, McNulty W, Elson D, Gassmann M, Arbeit JM, et al. Hypoxia-inducible factor-1alpha is a positive factor in solid tumor growth. Cancer Res. 2000;60(15):4010–5.

    CAS  PubMed  Google Scholar 

  32. Hochachka PW, Buck LT, Doll CJ, Land SC. Unifying theory of hypoxia tolerance: molecular/metabolic defense and rescue mechanisms for surviving oxygen lack. Proc Natl Acad Sci U S A. 1996;93(18):9493–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Wouters BG, Koritzinsky M. Hypoxia signalling through mTOR and the unfolded protein response in cancer. Nature reviews. Cancer. 2008;8(11):851–64.

    CAS  PubMed  Google Scholar 

  34. Koumenis C, Naczki C, Koritzinsky M, Rastani S, Diehl A, Sonenberg N, et al. Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2alpha. Mol Cell Biol. 2002;22(21):7405–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Blais JD, Filipenko V, Bi M, Harding HP, Ron D, Koumenis C, et al. Activating transcription factor 4 is translationally regulated by hypoxic stress. Mol Cell Biol. 2004;24(17):7469–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Fels DR, Koumenis C. The PERK/eIF2alpha/ATF4 module of the UPR in hypoxia resistance and tumor growth. Cancer biology & therapy. 2006;5(7):723–8.

    Article  CAS  Google Scholar 

  37. Bi M, Naczki C, Koritzinsky M, Fels D, Blais J, Hu N, et al. ER stress-regulated translation increases tolerance to extreme hypoxia and promotes tumor growth. EMBO J. 2005;24(19):3470–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ameri K, Lewis CE, Raida M, Sowter H, Hai T, Harris AL. Anoxic induction of ATF-4 through HIF-1-independent pathways of protein stabilization in human cancer cells. Blood. 2004;103(5):1876–82.

    Article  CAS  PubMed  Google Scholar 

  39. Rouschop KM, van den Beucken T, Dubois L, Niessen H, Bussink J, Savelkouls K, et al. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5. J Clin Invest. 2010;120(1):127–41.

    Article  CAS  PubMed  Google Scholar 

  40. Pike LR, Singleton DC, Buffa F, Abramczyk O, Phadwal K, Li JL, et al. Transcriptional up-regulation of ULK1 by ATF4 contributes to cancer cell survival. The Biochemical journal. 2013;449(2):389–400.

    Article  CAS  PubMed  Google Scholar 

  41. Rouschop KM, Dubois LJ, Keulers TG, van den Beucken T, Lambin P, Bussink J, et al. PERK/eIF2alpha signaling protects therapy resistant hypoxic cells through induction of glutathione synthesis and protection against ROS. Proc Natl Acad Sci U S A. 2013;110(12):4622–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Dewhirst MW, Cao Y, Moeller B. Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response. Nature reviews. Cancer. 2008;8(6):425–37.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Romero-Ramirez L, Cao H, Nelson D, Hammond E, Lee AH, Yoshida H, et al. XBP1 is essential for survival under hypoxic conditions and is required for tumor growth. Cancer Res. 2004;64(17):5943–7.

    Article  CAS  PubMed  Google Scholar 

  44. Chen X, Iliopoulos D, Zhang Q, Tang Q, Greenblatt MB, Hatziapostolou M, et al. XBP1 promotes triple-negative breast cancer by controlling the HIF1alpha pathway. Nature. 2014;508(7494):103–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pereira ER, Frudd K, Awad W, Hendershot LM. Endoplasmic reticulum (ER) stress and hypoxia response pathways interact to potentiate hypoxia-inducible factor 1 (HIF-1) transcriptional activity on targets like vascular endothelial growth factor (VEGF. J Biol Chem. 2014;289(6):3352–64.

    Article  CAS  PubMed  Google Scholar 

  46. Tay KH, Luan Q, Croft A, Jiang CC, Jin L, Zhang XD, et al. Sustained IRE1 and ATF6 signaling is important for survival of melanoma cells undergoing ER stress. Cell Signal. 2014;26(2):287–94.

    Article  CAS  PubMed  Google Scholar 

  47. Tang CH, Ranatunga S, Kriss CL, Cubitt CL, Tao J, Pinilla-Ibarz JA, et al. Inhibition of ER stress-associated IRE-1/XBP-1 pathway reduces leukemic cell survival. J Clin Invest. 2014;124(6):2585–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Arai M, Kondoh N, Imazeki N, Hada A, Hatsuse K, Kimura F, et al. Transformation-associated gene regulation by ATF6alpha during hepatocarcinogenesis. FEBS Lett. 2006;580(1):184–90.

    Article  CAS  PubMed  Google Scholar 

  49. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.

    Article  CAS  PubMed  Google Scholar 

  50. Wek RC, Cavener DR. Translational control and the unfolded protein response. Antioxid Redox Signal. 2007;9(12):2357–71.

    Article  CAS  PubMed  Google Scholar 

  51. Zhang P, McGrath BC, Reinert J, Olsen DS, Lei L, Gill S, et al. The GCN2 eIF2alpha kinase is required for adaptation to amino acid deprivation in mice. Mol Cell Biol. 2002;22(19):6681–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Shan J, Ord D, Ord T, Kilberg MS. Elevated ATF4 expression, in the absence of other signals, is sufficient for transcriptional induction via CCAAT enhancer-binding protein-activating transcription factor response elements. J Biol Chem. 2009;284(32):21241–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wek RC, Ramirez M, Jackson BM, Hinnebusch AG. Identification of positive-acting domains in GCN2 protein kinase required for translational activation of GCN4 expression. Mol Cell Biol. 1990;10(6):2820–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Wek SA, Zhu S, Wek RC. The histidyl-tRNA synthetase-related sequence in the eIF-2 alpha protein kinase GCN2 interacts with tRNA and is required for activation in response to starvation for different amino acids. Mol Cell Biol. 1995;15(8):4497–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Wang Y, Alam GN, Ning Y, Visioli F, Dong Z, Nor JE, et al. The unfolded protein response induces the angiogenic switch in human tumor cells through the PERK/ATF4 pathway. Cancer Res. 2012;72(20):5396–406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Wang Y, Ning Y, Alam GN, Jankowski BM, Dong Z. Nor JE, et al. amino acid deprivation promotes tumor angiogenesis through the GCN2/ATF4 pathway. Neoplasia. 2013;15(8):989–97.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Ward PS, Thompson CB. Signaling in control of cell growth and metabolism. Cold Spring Harb Perspect Biol. 2012;4(7):a006783.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. DeBerardinis RJI. Cancer a disease of abnormal cellular metabolism? New angles on an old idea. Genet Med. 2008;10(11):767–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Osthus RC, Shim H, Kim S, Li Q, Reddy R, Mukherjee M, et al. Deregulation of glucose transporter 1 and glycolytic gene expression by c-Myc. J Biol Chem. 2000;275(29):21797–800.

    Article  CAS  PubMed  Google Scholar 

  60. Ying H, Kimmelman AC, Lyssiotis CA, Hua S, Chu GC, Fletcher-Sananikone E, et al. Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism. Cell. 2012;149(3):656–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zachara NE, O’Donnell N, Cheung WD, Mercer JJ, Marth JD, Hart GW. Dynamic O-GlcNAc modification of nucleocytoplasmic proteins in response to stress. A survival response of mammalian cells. J Biol Chem. 2004;279(29):30133–42.

    Article  CAS  PubMed  Google Scholar 

  62. Vasseur S, Manie SNER. Stress and hexosamine pathway during tumourigenesis: a pas de deux? Semin Cancer Biol. 2015;33:34–9.

    Article  CAS  PubMed  Google Scholar 

  63. Caldwell SA, Jackson SR, Shahriari KS, Lynch TP, Sethi G, Walker S, et al. Nutrient sensor O-GlcNAc transferase regulates breast cancer tumorigenesis through targeting of the oncogenic transcription factor FoxM1. Oncogene. 2010;29(19):2831–42.

    Article  CAS  PubMed  Google Scholar 

  64. Gu Y, Mi W, Ge Y, Liu H, Fan Q, Han C, et al. GlcNAcylation plays an essential role in breast cancer metastasis. Cancer Res. 2010;70(15):6344–51.

    Article  CAS  PubMed  Google Scholar 

  65. Lynch TP, Ferrer CM, Jackson SR, Shahriari KS, Vosseller K, Reginato MJ. Critical role of O-linked beta-N-acetylglucosamine transferase in prostate cancer invasion, angiogenesis, and metastasis. J Biol Chem. 2012;287(14):11070–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Mi W, Gu Y, Han C, Liu H, Fan Q, Zhang X, et al. O-GlcNAcylation is a novel regulator of lung and colon cancer malignancy. Biochim Biophys Acta. 2011;1812(4):514–9.

    Article  CAS  PubMed  Google Scholar 

  67. Yehezkel G, Cohen L, Kliger A, Manor E, Khalaila IO. Linked beta-N-acetylglucosaminylation (O-GlcNAcylation) in primary and metastatic colorectal cancer clones and effect of N-acetyl-beta-D-glucosaminidase silencing on cell phenotype and transcriptome. J Biol Chem. 2012;287(34):28755–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Zhu Q, Zhou L, Yang Z, Lai M, Xie H, Wu L, et al. O-GlcNAcylation plays a role in tumor recurrence of hepatocellular carcinoma following liver transplantation. Med Oncol. 2012;29(2):985–93.

    Article  CAS  PubMed  Google Scholar 

  69. Shi Y, Tomic J, Wen F, Shaha S, Bahlo A, Harrison R, et al. Aberrant O-GlcNAcylation characterizes chronic lymphocytic leukemia. Leukemia. 2010;24(9):1588–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Hauselmann I, Borsig L. Altered tumor-cell glycosylation promotes metastasis. Front Oncol. 2014;4:28.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Radhakrishnan P, Dabelsteen S, Madsen FB, Francavilla C, Kopp KL, Steentoft C, et al. Immature truncated O-glycophenotype of cancer directly induces oncogenic features. Proc Natl Acad Sci U S A. 2014;111(39):E4066–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Yuneva M, Zamboni N, Oefner P, Sachidanandam R, Lazebnik Y. Deficiency in glutamine but not glucose induces Myc-dependent apoptosis in human cells. J Cell Biol. 2007;178(1):93–105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Wise DR, DeBerardinis RJ, Mancuso A, Sayed N, Zhang XY, Pfeiffer HK, et al. Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction. Proc Natl Acad Sci U S A. 2008;105(48):18782–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Gao P, Tchernyshyov I, Chang TC, Lee YS, Kita K, Ochi T, et al. C-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature. 2009;458(7239):762–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Guillaumond F, Leca J, Olivares O, Lavaut MN, Vidal N, Berthezene P, et al. Strengthened glycolysis under hypoxia supports tumor symbiosis and hexosamine biosynthesis in pancreatic adenocarcinoma. Proc Natl Acad Sci U S A. 2013;110(10):3919–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Myatt SS, Lam EW. The emerging roles of forkhead box (fox) proteins in cancer. Nat Rev Cancer. 2007;7(11):847–59.

    Article  CAS  PubMed  Google Scholar 

  77. Ma Z, Vocadlo DJ, Vosseller K. Hyper-O-GlcNAcylation is anti-apoptotic and maintains constitutive NF-kappaB activity in pancreatic cancer cells. J Biol Chem. 2013;288(21):15121–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Park SY, Kim HS, Kim NH, Ji S, Cha SY, Kang JG, et al. Snail1 is stabilized by O-GlcNAc modification in hyperglycaemic condition. EMBO J. 2010;29(22):3787–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Wang ZV, Deng Y, Gao N, Pedrozo Z, Li DL, Morales CR, et al. Spliced X-box binding protein 1 couples the unfolded protein response to hexosamine biosynthetic pathway. Cell. 2014;156(6):1179–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Denzel MS, Storm NJ, Gutschmidt A, Baddi R, Hinze Y, Jarosch E, et al. Hexosamine pathway metabolites enhance protein quality control and prolong life. Cell. 2014;156(6):1167–78.

    Article  CAS  PubMed  Google Scholar 

  81. Romero-Ramirez L, Cao H, Regalado MP, Kambham N, Siemann D, Kim JJ, et al. X box-binding protein 1 regulates angiogenesis in human pancreatic adenocarcinomas. Transl Oncol. 2009;2(1):31–8.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Ghosh R, Lipson KL, Sargent KE, Mercurio AM, Hunt JS, Ron D, et al. Transcriptional regulation of VEGF-A by the unfolded protein response pathway. PLoS One. 2010;5(3):e9575.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Auf G, Jabouille A, Guerit S, Pineau R, Delugin M, Bouchecareilh M, et al. Inositol-requiring enzyme 1alpha is a key regulator of angiogenesis and invasion in malignant glioma. Proc Natl Acad Sci U S A. 2010;107(35):15553–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Karali E, Bellou S, Stellas D, Klinakis A, Murphy C, Fotsis TVEGF. Signals through ATF6 and PERK to promote endothelial cell survival and angiogenesis in the absence of ER stress. Mol Cell. 2014;54(4):559–72.

    Article  CAS  PubMed  Google Scholar 

  85. Urra H, Hetz CA. Novel ER stress-independent function of the UPR in angiogenesis. Mol Cell. 2014;54(4):542–4.

    Article  CAS  PubMed  Google Scholar 

  86. Friedberg ECDNA. Damage and repair. Nature. 2003;421(6921):436–40.

    Article  PubMed  CAS  Google Scholar 

  87. Yamamori T, Meike S, Nagane M, Yasui H, Inanami OER. Stress suppresses DNA double-strand break repair and sensitizes tumor cells to ionizing radiation by stimulating proteasomal degradation of Rad51. FEBS Lett. 2013;587(20):3348–53.

    Article  CAS  PubMed  Google Scholar 

  88. Mujcic H, Nagelkerke A, Rouschop KM, Chung S, Chaudary N, Span PN, et al. Hypoxic activation of the PERK/eIF2alpha arm of the unfolded protein response promotes metastasis through induction of LAMP3. Clin Cancer Res. 2013;19(22):6126–37.

    Article  CAS  PubMed  Google Scholar 

  89. Mujcic H, Rzymski T, Rouschop KM, Koritzinsky M, Milani M, Harris AL, et al. Hypoxic activation of the unfolded protein response (UPR) induces expression of the metastasis-associated gene LAMP3. Radiother Oncol. 2009;92(3):450–9.

    Article  CAS  PubMed  Google Scholar 

  90. Nagelkerke A, Bussink J, Mujcic H, Wouters BG, Lehmann S, Sweep FC, et al. Hypoxia stimulates migration of breast cancer cells via the PERK/ATF4/LAMP3-arm of the unfolded protein response. Breast Cancer Res. 2013a;15(1):R2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Nagelkerke A, Bussink J, van der Kogel AJ, Sweep FC, Span PN. The PERK/ATF4/LAMP3-arm of the unfolded protein response affects radioresistance by interfering with the DNA damage response. Radiother Oncol. 2013b;108(3):415–21.

    Article  CAS  PubMed  Google Scholar 

  92. Nagelkerke A, Sweep FC, Stegeman H, Grenman R, Kaanders JH, Bussink J, et al. Hypoxic regulation of the PERK/ATF4/LAMP3-arm of the unfolded protein response in head and neck squamous cell carcinoma. Head Neck. 2015;37(6):896–905.

    Article  PubMed  Google Scholar 

  93. Dejeans N, Pluquet O, Lhomond S, Grise F, Bouchecareilh M, Juin A, et al. Autocrine control of glioma cells adhesion and migration through IRE1alpha-mediated cleavage of SPARC mRNA. J Cell Sci. 2012;125(Pt 18):4278–87.

    Article  CAS  PubMed  Google Scholar 

  94. Kunigal S, Gondi C, Gujrati M, Lakka SS, Dinh DH, Olivero WC, et al. SPARC-induced migration of glioblastoma cell lines via uPA-uPAR signaling and activation of small GTPase RhoA. Int J Oncol. 2006;29(6):1349–57.

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Mahadevan NR, Rodvold J, Sepulveda H, Rossi S, Drew AF, Zanetti M. Transmission of endoplasmic reticulum stress and pro-inflammation from tumor cells to myeloid cells. Proc Natl Acad Sci U S A. 2011;108(16):6561–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Mumm JB, Oft M. Cytokine-based transformation of immune surveillance into tumor-promoting inflammation. Oncogene. 2008;27(45):5913–9.

    Article  CAS  PubMed  Google Scholar 

  97. Dang CVMYC. On the path to cancer. Cell. 2012;149(1):22–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Hart LS, Cunningham JT, Datta T, Dey S, Tameire F, Lehman SL, et al. ER stress-mediated autophagy promotes Myc-dependent transformation and tumor growth. J Clin Invest. 2012;122(12):4621–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Qiu B, Simon MC. Oncogenes strike a balance between cellular growth and homeostasis. Semin Cell Dev Biol. 2015;43:3–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Carroll PA, Diolaiti D, McFerrin L, Gu H, Djukovic D, Du J, et al. Deregulated Myc requires MondoA/mlx for metabolic reprogramming and tumorigenesis. Cancer Cell. 2015;27(2):271–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462(7274):739–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Qing G, Li B, Vu A, Skuli N, Walton ZE, Liu X, et al. ATF4 regulates MYC-mediated neuroblastoma cell death upon glutamine deprivation. Cancer Cell. 2012;22(5):631–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Denoyelle C, Abou-Rjaily G, Bezrookove V, Verhaegen M, Johnson TM, Fullen DR, et al. Anti-oncogenic role of the endoplasmic reticulum differentially activated by mutations in the MAPK pathway. Nat Cell Biol. 2006;8(10):1053–63.

    Article  CAS  PubMed  Google Scholar 

  104. Platz A, Egyhazi S, Ringborg U, Hansson J. Human cutaneous melanoma; a review of NRAS and BRAF mutation frequencies in relation to histogenetic subclass and body site. Mol Oncol. 2008;1(4):395–405.

    Article  PubMed  Google Scholar 

  105. Corazzari M, Rapino F, Ciccosanti F, Giglio P, Antonioli M, Conti B, et al. Oncogenic BRAF induces chronic ER stress condition resulting in increased basal autophagy and apoptotic resistance of cutaneous melanoma. Cell Death Differ. 2015;22(6):946–58.

    Article  CAS  PubMed  Google Scholar 

  106. Ma XH, Piao SF, Dey S, McAfee Q, Karakousis G, Villanueva J, et al. Targeting ER stress-induced autophagy overcomes BRAF inhibitor resistance in melanoma. J Clin Invest. 2014;124(3):1406–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Croft A, Tay KH, Boyd SC, Guo ST, Jiang CC, Lai F, et al. Oncogenic activation of MEK/ERK primes melanoma cells for adaptation to endoplasmic reticulum stress. J Invest Dermatol. 2014;134(2):488–97.

    Article  CAS  PubMed  Google Scholar 

  108. Ursini-Siegel J, Schade B, Cardiff RD, Muller WJ. Insights from transgenic mouse models of ERBB2-induced breast cancer. Nat Rev Cancer. 2007;7(5):389–97.

    Article  CAS  PubMed  Google Scholar 

  109. Arteaga CL, Sliwkowski MX, Osborne CK, Perez EA, Puglisi F, Gianni L. Treatment of HER2-positive breast cancer: current status and future perspectives. Nat Rev Clin Oncol. 2012;9(1):16–32.

    Article  CAS  Google Scholar 

  110. Nam S, Chang HR, Jung HR, Gim Y, Kim NY, Grailhe R, et al. A pathway-based approach for identifying biomarkers of tumor progression to trastuzumab-resistant breast cancer. Cancer Lett. 2015;356(2 Pt B):880–90.

    Article  CAS  PubMed  Google Scholar 

  111. Hazari YM, Habib M, Bashir S, Bashir A, Hilal N, Irfan S, et al. Natural osmolytes alleviate GRP78 and ATF-4 levels: corroboration for potential modulators of unfolded protein response. Life Sci. 2016;146:148–53.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

The financial support provided by grants from the DBT (No. BT/PR7240/MED/30/915/2012), DST (No. SB/SO/AS-126/2012), FIST (No. SR/FST/LSI-384/2008), SAP, and UGC (No. F.3-26/2011 (SAP-II)) facilities extended by University of Kashmir are gratefully acknowledged. Department of Science and Technology (DST) fellowship to Younis Mohammad Hazari and Department of Biotechnology (DBT) fellowship to Arif Bashir is acknowledged.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Khalid Majid Fazili.

Ethics declarations

Conflicts of interest

None.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hazari, Y.M., Bashir, A., Haq, E.u. et al. Emerging tale of UPR and cancer: an essentiality for malignancy. Tumor Biol. 37, 14381–14390 (2016). https://doi.org/10.1007/s13277-016-5343-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13277-016-5343-0

Keywords

Navigation