Skip to main content
Top
Published in: Tumor Biology 9/2015

01-09-2015 | Research Article

K-Ras promotes the non-small lung cancer cells survival by cooperating with sirtuin 1 and p27 under ROS stimulation

Authors: Dezhi Cheng, Liang Zhao, Yunsheng Xu, Rongying Ou, Gang Li, Han Yang, Wenfeng Li

Published in: Tumor Biology | Issue 9/2015

Login to get access

Abstract

Cigarette smoking might lead to lung cancer. However, the related signaling pathways at molecular level remained unknown until now. In this study, we studied the signaling processes associated between tobacco exposure and lung cancer. First, we detected and validated pathway-specific gene expression at bronchial epithelium. These proteins reflected the activation of signaling pathways relevant to tobacco exposure, including ATM, BCL2, GPX1, K-Ras, IKBKB, and SIRT1. Tobacco smoking was simulated via reactive oxygen species (ROS) pathway. ROS not only arrested cell cycle at G1/S stage but also increased expressions of Sirt1 and p27. Further studies showed that the expression of p27 was dependent on ERK1/2 activation, and p27 itself could halt cell cycle by inhibiting the activation of CDKs. Moreover, activation of K-Ras, the key regulator of Ras/ERK pathway, was tightly regulated by enzyme activity of Sirt1. Deacetylation of K-Ras by Sirt1 increased the transformation of Ras-GTP to Ras-GDP, promoting the activation of downstream of ERK1/2. In reverse, Ras/ERK pathway could also regulate Sirt1 transcription. In conclusion, inhibition of Sirt1 may be an effective strategy for the prevention of tumor progression in high-risk patients or as a therapeutic strategy in established tumors.
Literature
1.
go back to reference Shields PG. Molecular epidemiology of lung cancer. Ann Oncol Suppl. 1999;5:7–11.CrossRef Shields PG. Molecular epidemiology of lung cancer. Ann Oncol Suppl. 1999;5:7–11.CrossRef
2.
go back to reference Sundaresan M, Yu ZX, Ferrans VJ, Irani K, Finkel T. Requirement for generation of H2O2 for platelet-derived growth factor signal transduction. Science. 1995;270:296–9.CrossRefPubMed Sundaresan M, Yu ZX, Ferrans VJ, Irani K, Finkel T. Requirement for generation of H2O2 for platelet-derived growth factor signal transduction. Science. 1995;270:296–9.CrossRefPubMed
3.
go back to reference Bae YS, Kang SW, Seo MS, et al. Epidermal growth factor (EGF)-induced generation of hydrogen peroxide in EGF receptor-mediated tyrosine phosphorylation. J Biol Chem. 1997;272:217–21.CrossRefPubMed Bae YS, Kang SW, Seo MS, et al. Epidermal growth factor (EGF)-induced generation of hydrogen peroxide in EGF receptor-mediated tyrosine phosphorylation. J Biol Chem. 1997;272:217–21.CrossRefPubMed
4.
go back to reference Leto TL, Geiszt M. Role of NOX family NADPH oxidases in host defense. Antioxid Redox Signal. 2006;8:1549–61.CrossRefPubMed Leto TL, Geiszt M. Role of NOX family NADPH oxidases in host defense. Antioxid Redox Signal. 2006;8:1549–61.CrossRefPubMed
5.
go back to reference Thannickal VJ, Fanburg BL. Reactive oxygen species in cell signaling. Am J Physiol Lung Cell Mol Physiol. 2000;279:L1005–28.PubMed Thannickal VJ, Fanburg BL. Reactive oxygen species in cell signaling. Am J Physiol Lung Cell Mol Physiol. 2000;279:L1005–28.PubMed
6.
go back to reference Vafa O, Wade M, Kern S, et al. c-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: a mechanism for oncogene-induced genetic instability. Mol Cell. 2002;9:1031–44.CrossRefPubMed Vafa O, Wade M, Kern S, et al. c-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: a mechanism for oncogene-induced genetic instability. Mol Cell. 2002;9:1031–44.CrossRefPubMed
7.
go back to reference Gianni D, Taulet N, DerMardirossian C, Bokoch GM. c-Src-mediated phosphorylation of NoxA1 and Tks4 induces the reactive oxygen species (ROS)-dependent formation of functional in vadopodia in human colon cancer cells. Mol Biol Cell. 2010;21:4287–98.CrossRefPubMedPubMedCentral Gianni D, Taulet N, DerMardirossian C, Bokoch GM. c-Src-mediated phosphorylation of NoxA1 and Tks4 induces the reactive oxygen species (ROS)-dependent formation of functional in vadopodia in human colon cancer cells. Mol Biol Cell. 2010;21:4287–98.CrossRefPubMedPubMedCentral
8.
go back to reference Ferro E, Goitre L, Retta SF, Trabalzini L. The interplay between ROS and Ras GTPases: physiological and pathological implications. J Signal Transduct. 2012;2012:365769.PubMed Ferro E, Goitre L, Retta SF, Trabalzini L. The interplay between ROS and Ras GTPases: physiological and pathological implications. J Signal Transduct. 2012;2012:365769.PubMed
11.
go back to reference Laupent-Puig P, Lievre A, Blons H. Mutations and response to epidermal growth factor receptor inhibitors. Clin Cancer Res. 2009;15:1133–9.CrossRef Laupent-Puig P, Lievre A, Blons H. Mutations and response to epidermal growth factor receptor inhibitors. Clin Cancer Res. 2009;15:1133–9.CrossRef
12.
13.
go back to reference Frye RA. Phylogenetic classification of prokaryotic and eukaryotic Sir2-like proteins. Biochem Biophys Res Commun. 2000;273:793–8.CrossRefPubMed Frye RA. Phylogenetic classification of prokaryotic and eukaryotic Sir2-like proteins. Biochem Biophys Res Commun. 2000;273:793–8.CrossRefPubMed
15.
go back to reference Kaeberlein TL, Smith ED, Tsuchiya M, et al. Lifespan extension in Caenorhabditis elegans by complete removal of food. Aging Cell. 2006;5:487–94.CrossRefPubMed Kaeberlein TL, Smith ED, Tsuchiya M, et al. Lifespan extension in Caenorhabditis elegans by complete removal of food. Aging Cell. 2006;5:487–94.CrossRefPubMed
16.
go back to reference Imai S, Armstrong CM, Kaeberlein M, et al. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature. 2000;403:795–800.CrossRefPubMed Imai S, Armstrong CM, Kaeberlein M, et al. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature. 2000;403:795–800.CrossRefPubMed
17.
go back to reference Bordone L, Motta M, Picard F, et al. Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic beta cells. PLoS Biol. 2006;4:e31.CrossRefPubMed Bordone L, Motta M, Picard F, et al. Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic beta cells. PLoS Biol. 2006;4:e31.CrossRefPubMed
19.
20.
go back to reference Yeung F, Hoberg JE, Ramsey CS, et al. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004;23:2369–80.CrossRefPubMedPubMedCentral Yeung F, Hoberg JE, Ramsey CS, et al. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004;23:2369–80.CrossRefPubMedPubMedCentral
21.
go back to reference Roninson IB. Oncogenic functions of tumour suppressor p21(Waf1/Cip1/Sdi1): association with cell senescence and tumour-promoting activities of stromal fibroblasts. Cancer Lett. 2002;179:1–14.CrossRefPubMed Roninson IB. Oncogenic functions of tumour suppressor p21(Waf1/Cip1/Sdi1): association with cell senescence and tumour-promoting activities of stromal fibroblasts. Cancer Lett. 2002;179:1–14.CrossRefPubMed
22.
go back to reference Cohen HY, Miller C, Bitterman KJ, et al. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science. 2004;305:390–2.CrossRefPubMed Cohen HY, Miller C, Bitterman KJ, et al. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science. 2004;305:390–2.CrossRefPubMed
25.
go back to reference Shimizu T, Awai N, Takeda K. Complex regulation of CDKs and G1 arrest during the granulocytic differentiation of human myeloblastic leukemia ML-1 cells. Oncogene. 2000;19:4640–6.CrossRefPubMed Shimizu T, Awai N, Takeda K. Complex regulation of CDKs and G1 arrest during the granulocytic differentiation of human myeloblastic leukemia ML-1 cells. Oncogene. 2000;19:4640–6.CrossRefPubMed
26.
go back to reference Rajamohan SB. SIRT1 promotes cell survival under stress by deacetylation-dependent deactivation of poly(ADP-ribose) polymerase 1. Mol Cell Biol. 2009;29:4116–29.CrossRefPubMedPubMedCentral Rajamohan SB. SIRT1 promotes cell survival under stress by deacetylation-dependent deactivation of poly(ADP-ribose) polymerase 1. Mol Cell Biol. 2009;29:4116–29.CrossRefPubMedPubMedCentral
27.
go back to reference Ford J, Jiang M, Milner J. Cancer-specific functions of SIRT1 enable human epithelial cancer cell growth and survival. Cancer Res. 2005;65:10457–63.CrossRefPubMed Ford J, Jiang M, Milner J. Cancer-specific functions of SIRT1 enable human epithelial cancer cell growth and survival. Cancer Res. 2005;65:10457–63.CrossRefPubMed
28.
go back to reference Huang J, Gan Q, Han L, et al. SIRT1 overexpression antagonizes cellular senescence with activated ERK/S6k1 signaling in human diploid fibroblasts. PLoS One. 2008;3:e1710.CrossRefPubMedPubMedCentral Huang J, Gan Q, Han L, et al. SIRT1 overexpression antagonizes cellular senescence with activated ERK/S6k1 signaling in human diploid fibroblasts. PLoS One. 2008;3:e1710.CrossRefPubMedPubMedCentral
30.
go back to reference Post GR, Brown JH. G protein-coupled receptors and signaling pathways regulating growth responses. FASEB J. 1996;10:741–9.PubMed Post GR, Brown JH. G protein-coupled receptors and signaling pathways regulating growth responses. FASEB J. 1996;10:741–9.PubMed
31.
go back to reference Fanton CP, Mcmahon M, Pieper RO. Dual growth arrest pathways in astrocytes and astrocytic tumors in response to Raf-1 activation. J Biol Chem. 2001;276:18871–7.CrossRefPubMed Fanton CP, Mcmahon M, Pieper RO. Dual growth arrest pathways in astrocytes and astrocytic tumors in response to Raf-1 activation. J Biol Chem. 2001;276:18871–7.CrossRefPubMed
32.
go back to reference Malumbres M, Perez DE, Hernandes MI, et al. Cellular response to oncogenic Ras involves induction of the Cdk4 and Cdk6 inhibitor p15(INK4b). Mol Cell Biol. 2000;20:2915–25.CrossRefPubMedPubMedCentral Malumbres M, Perez DE, Hernandes MI, et al. Cellular response to oncogenic Ras involves induction of the Cdk4 and Cdk6 inhibitor p15(INK4b). Mol Cell Biol. 2000;20:2915–25.CrossRefPubMedPubMedCentral
33.
go back to reference Zhang JG, Zhao G, Qin Q, et al. Nicotinamide prohibits proliferation and enhances chemosensitivity of pancreatic cancer cells through deregulating SIRT1 and Ras/Akt pathways. Pancreatology. 2013;13:140–6.CrossRefPubMed Zhang JG, Zhao G, Qin Q, et al. Nicotinamide prohibits proliferation and enhances chemosensitivity of pancreatic cancer cells through deregulating SIRT1 and Ras/Akt pathways. Pancreatology. 2013;13:140–6.CrossRefPubMed
34.
go back to reference Heltweg B, Gatbonton T, Schuler AD, et al. Antitumor activity of a small-molecule inhibitor of human silent information regulator 2 enzymes. Cancer Res. 2006;66:4368–77.CrossRefPubMed Heltweg B, Gatbonton T, Schuler AD, et al. Antitumor activity of a small-molecule inhibitor of human silent information regulator 2 enzymes. Cancer Res. 2006;66:4368–77.CrossRefPubMed
35.
go back to reference Castro RE, Ferreira DM, Afonso MB, et al. miR-34a/SIRT1/p53 is suppressed by ursodeoxycholic acid in the rat liver and activated by disease severity in human non-alcoholic fatty liver disease. J Hepatol. 2013;58:119–25.CrossRefPubMed Castro RE, Ferreira DM, Afonso MB, et al. miR-34a/SIRT1/p53 is suppressed by ursodeoxycholic acid in the rat liver and activated by disease severity in human non-alcoholic fatty liver disease. J Hepatol. 2013;58:119–25.CrossRefPubMed
36.
go back to reference Mohannad HP, Zhang W, Prevas HS, et al. Loss of a single Hic1 allele accelerates polyp formation in Apc(Delta716) mice. Oncogene. 2011;30:2659–69.CrossRef Mohannad HP, Zhang W, Prevas HS, et al. Loss of a single Hic1 allele accelerates polyp formation in Apc(Delta716) mice. Oncogene. 2011;30:2659–69.CrossRef
37.
go back to reference Liu T, Liu PY, Marshall GM. The critical role of the class III histone deacetylase SIRT1 in cancer. Cancer Res. 2009;69:1702–5.CrossRefPubMed Liu T, Liu PY, Marshall GM. The critical role of the class III histone deacetylase SIRT1 in cancer. Cancer Res. 2009;69:1702–5.CrossRefPubMed
39.
go back to reference Burnstt C, Valentini S, Cabreiro F, et al. Absence of effects of Sir2 overexpression on lifespan in C. elegans and Drosophila. Nature. 2011;477:482–5.CrossRef Burnstt C, Valentini S, Cabreiro F, et al. Absence of effects of Sir2 overexpression on lifespan in C. elegans and Drosophila. Nature. 2011;477:482–5.CrossRef
40.
go back to reference Palacios JA, Herranz D, De Boins M, et al. SIRT1 contributes to telomere maintenance and augments global homologous recombination. J Cell Biol. 2010;191:1299–313.CrossRefPubMedPubMedCentral Palacios JA, Herranz D, De Boins M, et al. SIRT1 contributes to telomere maintenance and augments global homologous recombination. J Cell Biol. 2010;191:1299–313.CrossRefPubMedPubMedCentral
41.
go back to reference Ota H, Tokunaga E, Chang K, et al. Sirt1 inhibitor, Sirtinol, induces senescence-like growth arrest with attenuated Ras-MAPK signaling in human cancer cells. Oncogene. 2006;25:176–85.PubMed Ota H, Tokunaga E, Chang K, et al. Sirt1 inhibitor, Sirtinol, induces senescence-like growth arrest with attenuated Ras-MAPK signaling in human cancer cells. Oncogene. 2006;25:176–85.PubMed
42.
go back to reference Sun Y, Sun D, Li F, et al. Downregulation of Sirt1 by antisense oligonucleotides induces apoptosis and enhances radiation sensitization in A549 lung cancer cells. Lung Cancer. 2007;58:21–9.CrossRefPubMed Sun Y, Sun D, Li F, et al. Downregulation of Sirt1 by antisense oligonucleotides induces apoptosis and enhances radiation sensitization in A549 lung cancer cells. Lung Cancer. 2007;58:21–9.CrossRefPubMed
Metadata
Title
K-Ras promotes the non-small lung cancer cells survival by cooperating with sirtuin 1 and p27 under ROS stimulation
Authors
Dezhi Cheng
Liang Zhao
Yunsheng Xu
Rongying Ou
Gang Li
Han Yang
Wenfeng Li
Publication date
01-09-2015
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 9/2015
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-015-3429-8

Other articles of this Issue 9/2015

Tumor Biology 9/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine