Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Proteasome Inhibitors in Cancer Therapy and their Relation to Redox Regulation

Author(s): Gulce Sari, Zehra Okat, Ali Sahin and Betul Karademir*

Volume 24, Issue 44, 2018

Page: [5252 - 5267] Pages: 16

DOI: 10.2174/1381612825666190201120013

Price: $65

Abstract

Redox homeostasis is important for the maintenance of cell survival. Under physiological conditions, redox system works in a balance and involves activation of many signaling molecules. Regulation of redox balance via signaling molecules is achieved by different pathways and proteasomal system is a key pathway in this process. Importance of proteasomal system on signaling pathways has been investigated for many years. In this direction, many proteasome targeting molecules have been developed. Some of them are already in the clinic for cancer treatment and some are still under investigation to highlight underlying mechanisms. Although there are many studies done, molecular mechanisms of proteasome inhibitors and related signaling pathways need more detailed explanations. This review aims to discuss redox status and proteasomal system related signaling pathways. In addition, cancer therapies targeting proteasomal system and their effects on redox-related pathways have been summarized.

Keywords: Proteasome inhibitors, redox status, signaling molecules, cancer therapy, redox homeostasis, proteasomal system.

[1]
Dahlmann B. Mammalian proteasome subtypes: Their diversity in structure and function. Arch Biochem Biophys 2016; 591: 132-40.
[2]
Jung T, Höhn A, Grune T. The proteasome and the degradation of oxidized proteins: Part II - protein oxidation and proteasomal degradation. Redox Biol 2013; 2C: 99-104.
[3]
Tanaka K. The proteasome: overview of structure and functions. Proc Jpn Acad, Ser B, Phys Biol Sci 2009; 85: 12-36.
[4]
Jung T, Catalgol B, Grune T. The proteasomal system. Mol Aspects Med 2009; 30: 191-296.
[5]
Kuehn L, Dahlmann B. Proteasome activator PA28 and its interaction with 20 S proteasomes. Arch Biochem Biophys 1996; 329: 87-96.
[6]
Glickman MH, Ciechanover A. The ubiquitin-proteasome proteolytic pathway: destruction for the sake of construction. Physiol Rev 2002; 82(2): 373-428.
[7]
Catalgol B. Proteasome and cancer. Prog Mol Biol Transl Sci 2012; 109: 277-93.
[8]
Kamata H, Hirata H. Redox regulation of cellular signalling. Cell Signal 1999; 11(1): 1-14.
[9]
Dröge W. Free radicals in the physiological control of cell function. Physiol Rev 2002; 82(1): 47-95.
[10]
Le Bras M, Clément MV, Pervaiz S, Brenner C. Reactive oxygen species and the mitochondrial signaling pathway of cell death. Histol Histopathol 2005; 20(1): 205-19.
[11]
Kehrer JP. The Haber-Weiss reaction and mechanisms of toxicity. Toxicology 2000; 149(1): 43-50.
[12]
Szabó C, Ischiropoulos H, Radi R. Peroxynitrite: biochemistry, pathophysiology and development of therapeutics. Nat Rev Drug Discov 2007; 6(8): 662-80.
[13]
Faraci FM, Didion SP. Vascular protection: superoxide dismutase isoforms in the vessel wall. Arterioscler Thromb Vasc Biol 2004; 24(8): 1367-73.
[14]
Brigelius-Flohé R. Glutathione peroxidases and redox-regulated transcription factors. Biol Chem 2006; 387(10-11): 1329-35.
[15]
Nakamura H, Nakamura K, Yodoi J. Redox regulation of cellular activation. Annu Rev Immunol 1997; 15: 351-69.
[16]
Lillig CH, Holmgren A. Thioredoxin and related molecules--from biology to health and disease. Antioxid Redox Signal 2007; 9(1): 25-47.
[17]
Holmgren A, Aslund F. Glutaredoxin. Methods Enzymol 1995; 252: 283-92.
[18]
Wood ZA, Schröder E, Robin Harris J, Poole LB. Structure, mechanism and regulation of peroxiredoxins. Trends Biochem Sci 2003; 28(1): 32-40.
[19]
Valko M, Leibfritz D, Moncol J, Cronin MTD, Mazur M, Telser J. Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol 2007; 39: 44-84.
[20]
Kisselev AF, Goldberg AL. Proteasome inhibitors: from research tools to drug candidates. Chem Biol 2001; 8(8): 739-58.
[21]
Rock KL, Gramm C, Rothstein L, et al. Inhibitors of the proteasome block the degradation of most cell proteins and the generation of peptides presented on MHC class I molecules. Cell 1994; 78(5): 761-71.
[22]
Heinemeyer W, Fischer M, Krimmer T, Stachon U, Wolf DH. The active sites of the eukaryotic 20 S proteasome and their involvement in subunit precursor processing. J Biol Chem 1997; 272: 25200-9.
[23]
Adams J, Palombella VJ, Sausville EA, et al. Proteasome inhibitors: a novel class of potent and effective antitumor agents. Cancer Res 1999; 59: 2615-22.
[24]
Lee DH, Goldberg AL. Selective inhibitors of the proteasome-dependent and vacuolar pathways of protein degradation in Saccharomyces cerevisiae. J Biol Chem 1996; 271(44): 27280-4.
[25]
Tsubuki S, Saito Y, Tomioka M, Ito H, Kawashima S. Differential inhibition of calpain and proteasome activities by peptidyl aldehydes of di-leucine and tri-leucine. J Biochem 1996; 119(3): 572-6.
[26]
Adams J, Behnke M, Chen S, et al. Potent and selective inhibitors of the proteasome: dipeptidyl boronic acids. Bioorg Med Chem Lett 1998; 8: 333-8.
[27]
Richardson PG, Barlogie B, Berenson J, et al. A phase 2 study of bortezomib in relapsed, refractory myeloma. N Engl J Med 2003; 348(26): 2609-17.
[28]
Fisher RI, Bernstein SH, Kahl BS, et al. Multicenter Phase II Study of Bortezomib in Patients With Relapsed or Refractory Mantle Cell Lymphoma. J Clin Oncol 2004; 24(30): 4867-74.
[29]
Nazif T, Bogyo M. Global analysis of proteasomal substrate specificity using positional-scanning libraries of covalent inhibitors. Proc Natl Acad Sci USA 2001; 98(6): 2967-72.
[30]
Lynas JF, Harriott P, Healy A, McKervey MA, Walker B. Inhibitors of the chymotrypsin-like activity of proteasome based on di- and tri-peptidyl alpha-keto aldehydes (glyoxals). Bioorg Med Chem Lett 1998; 8(4): 373-8.
[31]
Chatterjee S, Dunn D, Mallya S, Ator MAP. ’-extended a-ketoamide inhibitors of proteasome. Bioorg Med Chem Lett 1999; 9: 2603-6.
[32]
Myung J, Kim KB, Crews CM. The ubiquitin-proteasome pathway and proteasome inhibitors. Med Res Rev 2001; 21(4): 245-73.
[33]
Fenteany G, Standaert RF, Lane WS, Choi S, Corey EJ, Schreiber SL. Inhibition of proteasome activities and subunit-specific amino-terminal threonine modification by lactacystin. Science 1995; 268(5211): 726-31.
[34]
Adams J. Development of the proteasome inhibitor PS-341. Oncologist 2002; 7: 9-16.
[35]
Goy A, Gilles F. Update on the proteasome inhibitor bortezomib in hematologic malignancies. Clin Lymphoma 2004; 4: 230-7.
[36]
O’Connor OA, Wright J, Moskowitz C, et al. Phase II clinical experience with the novel proteasome inhibitor bortezomib in patients with indolent non-Hodgkin’s lymphoma and mantle cell lymphoma. J Clin Oncol 2005; 23: 676-84.
[37]
Kaplan GS, Torcun CC, Grune T, Ozer NK, Karademir B. Proteasome inhibitors in cancer therapy: Treatment regimen and peripheral neuropathy as a side effect. Free Radic Biol Med 2017; 103: 1-13.
[38]
Karademir B, Sari G, Jannuzzi AT, et al. , Musunuri S, Wicher G, Grune T, Mi J, Hacioglu-Bay H, Forsberg-Nilsson K, Bergquist J, Jung TProteomic approach for understanding milder neurotoxicity of Carfilzomib against Bortezomib. Sci Rep 2018; 8(1): 16318.
[39]
Kuhn DJ, Chen Q, Voorhees PM, et al. Potent activity of carfilzomib, a novel, irreversible inhibitor of the ubiquitin-proteasome pathway, against preclinical models of multiple myeloma. Blood 2007; 110(9): 3281-90.
[40]
Demo SD, Kirk CJ, Aujay MA, et al. Antitumor Activity of PR-171, a Novel Irreversible Inhibitor of the Proteasome. Cancer Res 2007; 67(13): 6383-91.
[41]
Teicher BA, Tomaszewski JE. Proteasome Inhibitors. Biochem Pharmacol 2015; 96: 1-9.
[42]
Kupperman E, Lee EC, Cao Y, et al. Evaluation of the Proteasome Inhibitor MLN9708 in Preclinical Models of Human Cancer. Cancer Res 2010; 70(5): 1970-80.
[43]
Chauhan D, Hideshima T, Anderson KC. A novel proteasome inhibitor NPI-0052 as an anticancer therapy. Br J Cancer 2006; 95: 961-5.
[44]
Fenical W, Jensen PR, Palladino MA, Lam KS, Lloyd GK, Potts BC. Discovery and development of the anticancer agent salinosporamide A (NPI-0052). Bioorg Med Chem 2009; 17(6): 2175-80.
[45]
Potts BC, Albitar MX, Anderson KC, et al. Marizomib, a proteasome inhibitor for all seasons: preclinical profile and a framework for clinical trials. Curr Cancer Drug Targets 2011; (3): 254-84.
[46]
Chauhan D, Singh AV, Aujay M, et al. A novel orally active proteasome inhibitor ONX 0912 triggers in vitro and in vivo cytotoxicity in multiple myeloma. Blood 2010; 116(23): 4906-15.
[47]
Zang Y, Thomas SM, Chan ET, et al. The next generation proteasome inhibitors carfilzomib and oprozomib activate prosurvival autophagy via induction of the unfolded protein response and ATF4. Autophagy 2012; 8(12): 1873-4.
[48]
Piva R, Ruggeri B, Williams M, et al. CEP-18770: A novel, orally active proteasome inhibitor with a tumor-selective pharmacologic profile competitive with bortezomib. Blood 2008; 111(5): 2765-75.
[49]
Dick LR, Cruikshank AA, Grenier L, Melandri FD, Nunes SL, Stein RL. Mechanistic Studies on the Inactivation of the Proteasome by Lactacystin: A central role for clasto-lactacystin β-lactone. J Biol Chem 1996; 271: 7273-6.
[50]
Landis-Piwowar KR, Huo C, Chen D, et al. A novel prodrug ofthe green tea polyphenol (-)-epigallocatechin-3-gallate as a potential anticancer agent. Cancer Res 2007; 67: 4303-10.
[51]
Kuhn DJ, Burns AC, Kazi A, Dou QP. Direct inhibition of the ubiquitin-proteasome pathway by ester bond-containing green tea polyphenolsis associated with increased expression ofsterol regulatory element-binding protein 2 and LDL receptor. Biochim Biophys Acta 2004; 1682: 1-10.
[52]
Dou QP. Molecular mechanisms of green tea polyphenols. Nutr Cancer 2009; 61: 827-35.
[53]
Salvioli S, Sikora E, Cooper EL, Franceschi C. Curcumin in Cell Death Processes: A Challenge for CAM of Age-Related Pathologies. Evid Based Complement Alternat Med 2007; 4(2): 181-90.
[54]
O’Sullivan-Coyne G, O’Sullivan GC, O’Donovan TR, Piwocka K, McKenna SL. Curcumin induces apoptosis-independent death in oesophageal cancer cells. Br J Cancer 2009; 101: 1585-95.
[55]
Aoki H, Takada Y, Kondo S, Sawaya R, Aggarwal BB, Kondo Y. Evidence that curcumin suppresses the growth of malignant gliomas in vitro and in vivo through induction of autophagy: role of Akt and extracellular signal-regulated kinase signaling pathways. Mol Pharmacol 2007; 72: 29-39.
[56]
Colland F. The therapeutic potential of deubiquitinating enzyme inhibitors. Biochem Soc Trans 2010; 38(1): 137-43.
[57]
Reverdy C, Conrath S, Lopez R, et al. Discovery of Specific Inhibitors of Human USP7/HAUSP Deubiquitinating Enzyme. Chem Biol 2012; 19(4): 467-77.
[58]
Wada T, Yamashita Y, Saga Y, et al. Screening for genetic abnormalities involved in ovarian carcinogenesis using retroviral expression libraries. Int J Oncol 2009; 35(5): 973-6.
[59]
Wang Y, Rishi AK, Puliyappadamba VT, et al. Targeted proteasome inhibition by Velcade induces apoptosis in human mesothelioma and breast cancer cell lines. Cancer Chemother Pharmacol 2011; 66: 455-66.
[60]
Blackburn C, Gigstad KM, Hales P, et al. Characterization of a new series of noncovalent proteasome inhibitors with exquisite potency and selectivity for the 20S beta5-subunit. Biochem J 2010; 430: 461-76.
[61]
Parlati F, Lee SJ, Aujay M, et al. Carfilzomib can induce tumor cell death through selective inhibition of the chymotrypsin-like activity of the proteasome. Blood 2009; 114: 3439-47.
[62]
Ho YK, Bargagna-Mohan P, Wehenkel M, Mohan R, Kim KB. LPM2-Specific Inhibitors: Chemical Genetic Tools for Proteasome Biology. Chem Biol 2007; 14: 419-30.
[63]
Jang ER, Lee NR, Han S, et al. Revisiting the role of the immunoproteasome in the activation of the canonical NF-kappaB pathway. Mol Biosyst 2012; 8: 2295-02.
[64]
Wehenkel M, Ban JO, Ho YK, Carmony KC, Hong JT, Kim KB. A selective inhibitor of the immunoproteasome subunit LMP2 induces apoptosis in PC-3 cells and suppresses tumour growth in nude mice. Br J Cancer 2012; 107(1): 53-62.
[65]
Muchamuel T, Basler M, Aujay MA, et al. A selective inhibitor of the immunoproteasome subunit LMP7 blocks cytokine production and attenuates progression of experimental arthritis. Nat Med 2009; 15: 781-7.
[66]
Ichikawa HT, Conley T, Muchamuel T, et al. Novel Proteasome Inhibitors Have a Beneficial Effect in Murine Lupus via the dual inhibition of Type I Interferon and autoantibody secreting cells. Arthritis Rheum 2012; 64(2): 493-503.
[67]
Kuhn DJ, Hunsucker SA, Chen Q, Voorhees PM, Orlowski M, Orlowski RZ. Targeted inhibition of the immunoproteasome is a potent strategy against models of multiple myeloma that overcomes resistance to conventional drugs and nonspecific proteasome inhibitors. Blood 2009; 113: 4667-76.
[68]
Singh AV, Bandi M, Aujay MA, et al. PR-924, a selective inhibitor of the immunoproteasome subunit LMP-7, blocks multiple myeloma cell growth both in vitro and in vivo. Br J Haematol 2011; 152: 155-63.
[69]
McConkey DJ, Zhu K. Mechanisms of proteasome inhibitor action and resistance in cancer. Drug Resist Updat 2008; 11(4-5): 164-79.
[70]
Rastogi N, Mishra DP. Therapeutic targeting of cancer cell cycle using proteasome inhibitors. Cell Div 2012; 7: 26.
[71]
Verma IM, Stevenson JK, Schwarz EM, Van Antwerp D, Miyamoto S. Rel/NF-kB/IkB family: intimate tales of association and dissociation. Genes Dev 1995; 9: 2723-35.
[72]
Urban MB, Schreck R, Baeuerle PA. NF-κB contacts DNA by a heterodimer of the p50 and p65 subunit. EMBO J 1991; 10(7): 1817-25.
[73]
Traenckner EMB, Pahl HL, Schmidt KN, Wilk S, Baeuerle PA. Phosphorylation of human IκB on serine 32 and 26 controls IκB-α proteolysis and NF-κB activation in response to diverse stimuli. EMBO J 1995; 14: 2876-83.
[74]
Chen Z, Hagler J, Palombella VJ, et al. Signal-induced site-specific phosphorylation targets Iκβα to the ubiquitin-proteasome pathway. Genes Dev 1995; 9: 1586-97.
[75]
Hoffmann A, Natoli G, Ghosh G. Transcriptional regulation via the NF-κB signaling module. Oncogene 2006; 25: 6706-16.
[76]
Henkel T, Machleidt T, Alkalay I, Krönke M, Ben-Neriah Y, Baeuerle PA. Rapid proteolysis of I kappa B-alpha is necessary for activation of transcription factor NF-kappa B. Nature 1993; 365(6442): 182-5.
[77]
Traenckner EMB, Wilk S, Baeuerle PA. A proteasome inhibitor prevents activation of NF-kappa B and stabilizes a newly phosphorylated form of I kappa B-alpha that is still bound to NF-kappa B. EMBO J 1994; 13(22): 5433-41.
[78]
Pineda-Molina E, Klatt P, Vázquez J, et al. Glutathionylation of the p50 subunit of NF-kappa B: a mechanism for redox-induced inhibition of DNA binding. Biochemistry 2001; 40(47): 14134-42.
[79]
Guttridge DC, Albanese C, Reuther JY, Pestell RG, Baldwin AS Jr. NF-kB controls cell growth and differentiation through transcriptional regulation of cyclin D1. Mol Cell Biol 1999; 19: 5785-99.
[80]
Tomonari A, Nishio K, Kurokawa H, et al. Identification of cis-Acting DNA Elements of the Human g-Glutamylcysteine Synthetase Heavy Subunit Gene. Biochem Biophys Res Commun 1997; 232: 522-7.
[81]
Rojo AI, Salinas M, Martin D, Perona R, Cuadrado A. Regulation of Cu/Zn-superoxide dismutase expression via the phosphatidylinositol 3 kinase/Akt pathway and nuclear factor-kappaB. J Neurosci 2004; 24: 7324-34.
[82]
Delic J, Mashedors P, Omura S, et al. proteasome inhibitor lactacystin induces apoptosis and sensitizes chemo- and radioresistant human chronic lymphocytic leukaemia lymphocytes to TNF-alpha-initiated apoptosis. Br J Cancer 1998; 77(7): 1103-7.
[83]
Nawrocki ST, Bruns CJ, Harbison MT, et al. Effects of the proteasome inhibitor PS-341 on apoptosis and angiogenesis in orthotopic human pancreatic tumor xenografts. Mol Cancer Ther 2002; 1(14): 1243-53.
[84]
Ma MH, Yang HH, Parker K, et al. The proteasome inhibitor PS-341 markedly enhances sensitivity of multiple myeloma tumor cells to chemotherapeutic agents. Clin Cancer Res 2003; 9: 1136-44.
[85]
Angel P, Karin M. The role of Jun, Fos, and the AP-1 complex in cell-proliferation and transformation. Biochim Biophys Acta 1991; 1072: 129-57.
[86]
Sen CK, Packer L. Antioxidant and redox regulation of gene transcription. FASEB J 1996; 10: 709-20.
[87]
Gius D, Botero A, Shah S, Curry HA. Intracellular oxidation/reduction status in the regulation of transcription factors NF-kappaB and AP-1. Toxicol Lett 1999; 106(2-3): 93-106.
[88]
Meyer M, Pahl HL, Bauerle PA. Regulation of the transcription factors NF-kB and AP-1 by redox changes. Chem Biol Interact 1994; 91: 91-100.
[89]
Shaulian E, Karin M. AP-1 as a regulator of cell life and death. Nat Cell Biol 2002; 4: 131-6.
[90]
Jariel-Encontre I, Salvat C, Steff AM, et al. Complex mechanisms for c-fos and c-jun degradation. Mol Biol Rep 1997; 24(1): 51-6.
[91]
Hirota K, Matsui M, Iwata S, Nishiyama A, Mori K, Yodoi J. AP-1 transcriptional activity is regulated by a direct association between thioredoxin and Ref-1. Proc Natl Acad Sci USA 1997; 94(8): 3633-8.
[92]
Meriin AB, Gabai VL, Yaglom J, Shifrin VI, Sherman MY. Proteasome inhibitors activate stress kinases and induce Hsp72. Diverse effects on apoptosis. J Biol Chem 1998; 273(11): 6373-9.
[93]
Nakayama K, Furusu A, Xu Q, Konta T, Kitamura M. Unexpected transcriptional induction of monocyte chemoattractant protein 1 by proteasome inhibition: involvement of the c-Jun N-terminal kinase-activator protein 1 pathway. J Immunol 2001; 167(3): 1145-50.
[94]
Yang Y, Ikezoe T, Saito T, Kobayashi M, Koeffler HP, Taguchi H. Proteasome inhibitor PS-341 induces growth arrest and apoptosis of non-small cell lung cancer cells via the JNK/c-Jun/AP-1 signaling. Cancer Sci 2004; 95(2): 176-80.
[95]
Blattner C, Sparks A, Lane D. Transcription factor E2F-1 is upregulated in response to DNA damage in a manner analogous to that of p53. Mol Cell Biol 1999; 19: 3704-13.
[96]
Lin WC, Lin FT, Nevins JR. Selective induction of E2F1 in response to DNA damage, mediated by ATM-dependent phosphorylation. Genes Dev 2001; 15: 1833-44.
[97]
Pediconi N, Ianari A, Costanzo A, et al. Differential regulation of E2F1 apoptotic target genes in response to DNA damage. Nat Cell Biol 2003; 5: 552-8.
[98]
Marti A, Wirbelauer C, Scheffner M, Krek W. Interaction between ubiquitin-protein ligase SCFSKP2 and E2F-1 underlies the regulation of E2F-1 degradation. Nat Cell Biol 1999; 1: 14-9.
[99]
Lohmann DR, Gallie BL. Retinoblastoma: revisiting the model prototype of inherited cancer. Am J Med Genet C Semin Med Genet 2004; 129C: 23-8.
[100]
Berezutskaya E, Bagchi S. The human papillomavirus E7 oncoprotein functionally interacts with the S4 subunit of the 26 S proteasome. J Biol Chem 1997; 272: 30135-40.
[101]
Boyer SN, Wazer DE, Band V. E7 protein of human papilloma virus-16 induces degradation of retinoblastoma protein through the ubiquitin-proteasome pathway. Cancer Res 1996; 56: 4620-4.
[102]
Higashitsuji H, Itoh K, Nagao T, et al. Reduced stability of retinoblastoma protein by gankyrin, an oncogenic ankyrin-repeat protein overexpressed in hepatomas. Nat Med 2000; 6: 96-9.
[103]
Knight JS, Sharma N, Robertson ES. Epstein-Barr virus latent antigen 3C can mediate the degradation of the retinoblastoma protein through an SCF cellular ubiquitin ligase. Proc Natl Acad Sci USA 2005; 102: 18562-6.
[104]
Wang J, Sampath A, Raychaudhuri P, Bagchi S. Both Rb and E7 are regulated by the ubiquitin proteasome pathway in HPV-containing cervical tumor cells. Oncogene 2001; 20: 4740-9.
[105]
Albero MP, Vaquer JM, Andreu EJ, et al. Bortezomib decreases Rb phosphorylation and induces caspase-dependent apoptosis in Imatinib-sensitive and -resistant Bcr-Abl1-expressing cells. Oncogene 2010; 29: 3276-86.
[106]
Baiz D, Pozzato G, Dapas B, et al. Bortezomib arrests the proliferation of hepatocellular carcinoma cells HepG2 and JHH6 by differentially affecting E2F1, p21 and p27 levels. Biochimie 2009; 91(3): 373-82.
[107]
Lim JH, Chang YC, Park YB, Park JW, Kwon TK. Transcriptional repression of E2F gene by proteasome inhibitors in human osteosarcoma cells. Biochem Biophys Res Commun 2004; 318(4): 868-72.
[108]
Ceresa C, Giovannetti E, Voortman J, et al. Bortezomib induces schedule-dependent modulation of gemcitabine pharmacokinetics and pharmacodynamics in non-small cell lung cancer and blood mononuclear cells. Mol Cancer Ther 2009; 8(5): 1026-36.
[109]
Azuma-Hara M, Taniura H, Uetsuki T, Niinobe M, Yoshikawa K. Regulation and deregulation of E2F1 in postmitotic neurons differentiated from embryonal carcinoma P19 cells. Exp Cell Res 1999; 251(2): 442-51.
[110]
Farra R, Dapas B, Baiz D, et al. Impairment of the Pin1/E2F1 axis in the anti-proliferative effect of bortezomib in hepatocellular carcinoma cells. Biochimie 2015; 112: 85-95.
[111]
Liu R, Fu C, Sun J, et al. A New Perspective for Osteosarcoma Therapy: Proteasome Inhibition by MLN9708/2238 Successfully Induces Apoptosis and Cell Cycle Arrest and Attenuates the Invasion Ability of Osteosarcoma Cells in Vitro. Cell Physiol Biochem 2017; 41(2): 451-65.
[112]
Carelli S, Ceriotti A, Cabibbo A, Fassina G, Ruvo M, Sitia R. Cysteine and glutathione secretion in response toprotein disulfide bond formation in the ER. Science 1997; 277: 1681-4.
[113]
Ginn-Pease ME, Whisler RL. Optimal NF-kB mediated transcriptional responses in Jurkat T cells exposed to oxidative stress are dependent on intracellular glutathione and costimulatory signals. Biochem Biophys Res Commun 1996; 226: 695-702.
[114]
Esposito F, Agosti V, Morrone G, et al. Inhibition of the differentiationof human myeloid cell lines by redox changes induced through glutathione depletion. Biochem J 1994; 301: 649-53.
[115]
Westerheide SD, Morimoto RI. Heat shock response modulators as therapeutic tools for diseases of protein conformation. J Biol Chem 2005; 280: 33097-100.
[116]
Liu H, Lightfoot R, Stevens JL. Activation of heat shock factor by alkylating agents is triggered by glutathione depletion and oxidation of protein thiols. J Biol Chem 1996; 271: 4805-12.
[117]
Ling YH, Liebes L, Zou Y, Perez-Soler R. Reactive oxygen species generation and mitochondrial dysfunction in the apoptotic response to Bortezomib, a novel proteasome inhibitor, in human H460 non-small cell lung cancer cells. J Biol Chem 2003; 278(36): 33714-23.
[118]
Qiu JH, Asai A, Chi S, Saito N, Hamada H, Kirino T. Proteasome inhibitors induce cytochrome c-caspase-3-likeprotease-mediated apoptosis in cultured cortical neurons. J Neurosci 2000; 20(1): 259-65.
[119]
Yang CF, Shen HM, Ong CN. Ebselen induces apoptosis in HepG2 cells through rapid depletion of intracellular thiols. Arch Biochem Biophys 2000; 374(2): 142-52.
[120]
Zou W, Yue P, Lin N, et al. Vitamin C inactivates the proteasome inhibitor PS-341 in human cancer cells. Clin Cancer Res 2006; 12(1): 273-80.
[121]
Du ZX, Zhang HY, Meng X, Guan Y, Wang HQ. Role of oxidative stress and intracellular glutathione in the sensitivity to apoptosis induced by proteasome inhibitor in thyroid cancer cells. BMC Cancer 2009; 9: 56.
[122]
Demasi M, Shringarpure R, Davies KJA. Glutathiolation of the Proteasome Is Enhanced by Proteolytic Inhibitors. Arch Biochem Biophys 2001; 389(2): 254-63.
[123]
Vousden KH, Lane DP. p53 in health and disease. Nat Rev Mol Cell Biol 2007; 8: 275-83.
[124]
Petitjean A, Achatz MI, Borresen-Dale AL, Hainaut P, Olivier M. TP53 mutations in human cancers: functional selection and impact on cancer prognosis and outcomes. Oncogene 2007; 26: 2157-65.
[125]
Yee KS, Vousden KH. Complicating the complexity of p53. Carcinogenesis 2005; 26: 1317-22.
[126]
Brooks CL, Gu W. Ubiquitination, phosphorylation and acetylation: the molecular basis for p53 regulation. Curr Opin Cell Biol 2003; 15: 164-71.
[127]
Buzek J, Latonen L, Kurki S, Peltonen K, Laiho M. Redox state of tumor suppressor p53 regulates its sequence-specific DNA binding in DNA-damaged cells by cysteine 277. Nucleic Acids Res 2002; 30: 2340-8.
[128]
Cho Y, Gorina S, Jeffrey PD, Pavletich NP. Crystal structure of a p53 tumor suppressor-DNA complex: understanding tumorigenic mutations. Science 1994; 265: 346-54.
[129]
Cano CE, Gommeaux J, Pietri S, et al. Tumor Protein 53-Induced Nuclear Protein 1 Is a Major Mediator of p53Antioxidant Function. Cancer Res 2009; 69: 219-26.
[130]
Haupt Y, Maya R, Kazaz A, Oren M. Mdm2 promotes the rapid degradation of p53. Nature 1997; 387: 296-9.
[131]
Honda R, Tanaka H, Yasuda H. Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett 1997; 420: 25-7.
[132]
Marine JC, Lozano G. Mdm2-mediated ubiquitylation: p53 and beyond. Cell Death Differ 2010; 17: 93-102.
[133]
Russo A, Bronte G, Fulfaro F, et al. Bortezomib: a new pro-apoptotic agent in cancer treatment. Curr Cancer Drug Targets 2010; 10: 55-67.
[134]
Halasi M, Pandit B, Gartel AL. Proteasome inhibitors suppress the protein expression of mutant p53. Cell Cycle 2014; 13(20): 3202-6.
[135]
Behrens J, von Kries JP, Kühl M, et al. Functional interaction of β-catenin with the transcription factor LEF-1. Nature 1996; 382: 638-42.
[136]
Hoogeboom D, Burgering BMT. Should I stay or should I go: β-catenin decides under stress. Rev Can 2009; 1796(2): 63-74.
[137]
McManus EJ, Sakamoto K, Armit LJ, et al. Role that phosphorylation of GSK3 plays in insulin and Wnt signalling defined by knockin analysis. EMBO J 2005; 24: 1571-83.
[138]
Qiang YW, Hu B, Chen Y, et al. Bortezomib induces osteoblast differentiation via Wnt-independent activation of β-catenin/TCF signaling. Blood 2009; 113(18): 4319-30.
[139]
Chong KY, Hsu CJ, Hung TH, et al. Wnt pathway activation and ABCB1 expression account for attenuation of proteasome inhibitor-mediated apoptosis in multidrug-resistant cancer cells. Cancer Biol Ther 2015; 16(1): 149-59.
[140]
Morrison DK. MAP Kinase Pathways. Cold Spring Harb Perspect Biol 2012; 4: a011254.
[141]
Hehner SP, Breitkreutz R, Shubinsky G, et al. Enhancement of T cell receptor signaling by a mild oxidative shift in the intracellular thiol pool. J Immunol 2000; 165: 4319-28.
[142]
Lo YYC, Wong JMS, Cruz TF. Reactive oxygen species mediate cytokine activation of c-Jun NH2-terminal kinases. J Biol Chem 1996; 271: 15703-7.
[143]
Davis RJ. The Mitogen-activated Protein Kinase Signal Transduction Pathway. The Journal of Biological Chemistry 1993; 268: 14553-6.
[144]
Orlowski RZ, Small GW, Shi YY. Evidence that inhibition of p44/42 mitogen-activated protein kinase signaling is a factor in proteasome inhibitor-mediated apoptosis. J Biol Chem 2002; 277: 27864-71.
[145]
Befani CD, Vlachostergios PJ, Hatzidaki E, et al. Bortezomib represses HIF-1alpha protein expression and nuclear accumulation by inhibiting both PI3K/Akt/TOR and MAPK pathways in prostate cancer cells. J Mol Med (Berl) 2012; 90(1): 45-54.
[146]
Yin D, Zhou H, Kumagai T, et al. Liu G, Ong JM, Black KL, Koeffler HP. Proteasome inhibitor PS-341 causes cell growth arrest and apoptosis in human glioblastoma multiforme (GBM). Oncogene 24: 344-54.2005;
[147]
Li C, Johnson DE. Bortezomib induces autophagy in head and neck squamous cell carcinoma cells via JNK activation. Cancer Lett 2012; 314: 102-7.
[148]
Nishizuka Y. The molecular heterogeneity of protein kinase C and its implications for cellular regulation. Nature 1988; 334: 661-5.
[149]
Avdonin PR. Receptor-dependent regulation of (Ca2+)i and phospholipase C in vascular endothelial cells. J Recept Signal Transduct Res 2000; 20(4): 235-54.
[150]
Newton AC. Regulation of the ABC kinases by phosphorylation: protein kinase C as a paradigm. Biochem J 2003; 370(2): 361-71.
[151]
Sonnenburg ED, Gao T, Newton AC. The phosphoinositi-dedependent kinase, PDK-1, phosphorylates conventional protein kinase C isozymes by a mechanism that is independent of phosphoinositide 3-kinase. J Biol Chem 2001; 276(48): 45289-97.
[152]
Newton AC, Koshland DE Jr. Protein kinase C autophosphorylates by an intrapeptide reaction. J Biol Chem 1987; 262(21): 10185-8.
[153]
Junoy B, Maccario H, Mas JL, Enjalbert A, Drouva SV. Proteasome Implication in Phorbol Ester- and GnRH-Induced Selective Down-Regulation of PKC (α, ε, ζ) in αT3-1 and LβT2 Gonadotrope Cell Lines. Endocrinology 2002; 143: 1386-403.
[154]
Lee HW, Smith L, Pettit GR, Smith JB. Bryostatin 1and Phorbol Ester Down-Modulate Protein Kinase C-alpha and -epsilon viathe Ubiquitin/Proteasome Pathway in Human Fibroblasts. Mol Pharmacol 1997; 51: 439-47.
[155]
Takagi K, Saito Y, Sawada J. Proteasome inhibitor enhances growth hormone-binding protein release. Mol Cell Endocrinol 2001; 182: 157-63.
[156]
Ghosh S, Baltimore D. Activation in vitro of NF-kappa B by phosphorylation of its inhibitor I kappa B. Nature 1990; 344(6267): 678-82.
[157]
Diaz-Meco MT, Dominguez I, Sanz L, et al. zeta PKC induces phosphorylation and inactivation of I kappa B-alpha in vitro. EMBO J 1994; 13(12): 2842-8.
[158]
Kahana S, Finniss S, Cazacu S, et al. Proteasome inhibitors sensitize glioma cells and glioma stem cells to TRAIL-induced apoptosis by PKCε-dependent downregulation of AKT and XIAP expressions. Cell Signal 2011; 23(8): 1348-57.
[159]
Xu L, Su L, Liu X. PKCδ regulates death receptor 5 expression induced by PS-341 through ATF4-ATF3/CHOP axis in human lung cancer cells. Mol Cancer Ther 2012; 11(10): 2174-82.
[160]
Xiao X, Zuo X, Davis AA, et al. HSF1 is required for extra-embryonic development, postnatal growth and protection during inflammatory responses in mice. EMBO J 1999; 18: 5943-52.
[161]
Garigan D, Hsu AL, Fraser AG, Kamath RS, Ahringer J, Kenyon C. Genetic analysis of tissue aging in Caenorhabditis elegans: A role for heat-shock factor and bacterial proliferation. Genetics 2002; 161(3): 1101-12.
[162]
Hsu AL, Murphy CT, Kenyon C. Regulation of Aging and Age-Related Disease by DAF-16 and Heat-Shock Factor. Science 2003; 300(5622): 1142-5.
[163]
Zuo J, Baler R, Dahl G, Voellmy R. Activation of the DNA binding ability of human heat shock transcription factor 1 may involve the transition from an intramolecular to an intermolecular triple-stranded coiled-coil structure. Mol Cell Biol 1994; 14: 7557-68.
[164]
Ahn SG, Thiele DJ. Redox regulation of mammalian heat shock factor 1 is essential for Hsp gene activation and protection from stress. Genes and Development 2003; 17: 516-28.
[165]
Mathew A, Mathur SK, Morimoto RI. The heat shock response and protein degradation: regulation of HSF2 by the ubiquitin proteasome pathway. Mol Cell Biol 1998; 18: 5091-8.
[166]
Kim D, Kim SH, Li GC. Proteasome inhibitors MG132 and lactacystin hyperphosphorylate HSF1 and induce hsp70 and hsp27 expression. Biochem Biophys Res Commun 1999; 254: 264-8.
[167]
Shah SP, Nooka AK, Jaye DL, Bahlis NJ, Lonial S, Boise LH. Bortezomib-induced heat shock response protects multiple myeloma cells and is activated by heat shock factor 1 serine 326 phosphorylation. Oncotarget 2016; 7(37): 59727-41.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy