56
Views
24
CrossRef citations to date
0
Altmetric
Research Article

RNA Binding Protein HuR Promotes Autophagosome Formation by Regulating Expression of Autophagy-Related Proteins 5, 12, and 16 in Human Hepatocellular Carcinoma Cells

, , , , , , , , & show all
Article: e00508-18 | Received 22 Oct 2018, Accepted 18 Dec 2018, Published online: 03 Mar 2023

REFERENCES

  • Glick D, Barth S, Macleod KF. 2010. Autophagy: cellular and molecular mechanisms. J Pathol 221:3–12. https://doi.org/10.1002/path.2697.
  • Cadwell K, Stappenbeck TS, Virgin HW. 2009. Role of autophagy and autophagy genes in inflammatory bowel disease. Curr Top Microbiol Immunol 335:141–167. https://doi.org/10.1007/978-3-642-00302-8_7.
  • Murrow L, Debnath J. 2013. Autophagy as a stress-response and quality-control mechanism: implications for cell injury and human disease. Annu Rev Pathol 8:105–137. https://doi.org/10.1146/annurev-pathol-020712-163918.
  • Levine B, Kroemer G. 2008. Autophagy in the pathogenesis of disease. Cell 132:27–42. https://doi.org/10.1016/j.cell.2007.12.018.
  • Mizushima N, Noda T, Yoshimori T, Tanaka Y, Ishii T, George MD, Klionsky DJ, Ohsumi M, Ohsumi Y. 1998. A protein conjugation system essential for autophagy. Nature 395:395–398. https://doi.org/10.1038/26506.
  • Mizushima N, Noda T, Ohsumi Y. 1999. Apg16p is required for the function of the Apg12p-Apg5p conjugate in the yeast autophagy pathway. EMBO J 18:3888–3896. https://doi.org/10.1093/emboj/18.14.3888.
  • Fujita N, Itoh T, Omori H, Fukuda M, Noda T, Yoshimori T. 2008. The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Mol Biol Cell 19:2092–2100. https://doi.org/10.1091/mbc.e07-12-1257.
  • Hanada T, Noda NN, Satomi Y, Ichimura Y, Fujioka Y, Takao T, Inagaki F, Ohsumi Y. 2007. The Atg12-Atg5 conjugate has a novel E3-like activity for protein lipidation in autophagy. J Biol Chem 282:37298–37302. https://doi.org/10.1074/jbc.C700195200.
  • Kirisako T, Ichimura Y, Okada H, Kabeya Y, Mizushima N, Yoshimori T, Ohsumi M, Takao T, Noda T, Ohsumi Y. 2000. The reversible modification regulates the membrane-binding state of Apg8/Aut7 essential for autophagy and the cytoplasm to vacuole targeting pathway. J Cell Biol 151:263–276. https://doi.org/10.1083/jcb.151.2.263.
  • Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, Kominami E, Ohsumi Y, Yoshimori T. 2000. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J 19:5720–5728. https://doi.org/10.1093/emboj/19.21.5720.
  • Mizushima N, Yamamoto A, Hatano M, Kobayashi Y, Kabeya Y, Suzuki K, Tokuhisa T, Ohsumi Y, Yoshimori T. 2001. Dissection of autophagosome formation using Apg5-deficient mouse embryonic stem cells. J Cell Biol 152:657–668. https://doi.org/10.1083/jcb.152.4.657.
  • Malhotra R, Warne JP, Salas E, Xu AW, Debnath J. 2015. Loss of Atg12, but not Atg5, in pro-opiomelanocortin neurons exacerbates diet-induced obesity. Autophagy 11:145–154.
  • Saitoh T, Fujita N, Jang MH, Uematsu S, Yang BG, Satoh T, Omori H, Noda T, Yamamoto N, Komatsu M, Tanaka K, Kawai T, Tsujimura T, Takeuchi O, Yoshimori T, Akira S. 2008. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature 456:264–268. https://doi.org/10.1038/nature07383.
  • White E. 2012. Deconvoluting the context-dependent role for autophagy in cancer. Nat Rev Cancer 12:401–410. https://doi.org/10.1038/nrc3262.
  • Maes H, Rubio N, Garg AD, Agostinis P. 2013. Autophagy: shaping the tumor microenvironment and therapeutic response. Trends Mol Med 19:428–446. https://doi.org/10.1016/j.molmed.2013.04.005.
  • Wang C, Hu Q, Shen HM. 2016. Pharmacological inhibitors of autophagy as novel cancer therapeutic agents. Pharmacol Res 105:164–175. https://doi.org/10.1016/j.phrs.2016.01.028.
  • Lei Y, Zhang D, Yu J, Dong H, Zhang J, Yang S. 2017. Targeting autophagy in cancer stem cells as an anticancer therapy. Cancer Lett 393:33–39. https://doi.org/10.1016/j.canlet.2017.02.012.
  • Bento CF, Renna M, Ghislat G, Puri C, Ashkenazi A, Vicinanza M, Menzies FM, Rubinsztein DC. 2016. Mammalian autophagy: how does it work?. Annu Rev Biochem 85:685–713. https://doi.org/10.1146/annurev-biochem-060815-014556.
  • Frankel LB, Lund AH. 2012. MicroRNA regulation of autophagy. Carcinogenesis 33:2018–2025. https://doi.org/10.1093/carcin/bgs266.
  • Zhai H, Fesler A, Ju J. 2013. MicroRNA: a third dimension in autophagy. Cell Cycle 12:246–250. https://doi.org/10.4161/cc.23273.
  • Gozuacik D, Akkoc Y, Ozturk DG, Kocak M. 2017. Autophagy-regulating microRNAs and cancer. Front Oncol 7:65. https://doi.org/10.3389/fonc.2017.00065.
  • Hinman MN, Lou H. 2008. Diverse molecular functions of Hu proteins. Cell Mol Life Sci 65:3168–3181. https://doi.org/10.1007/s00018-008-8252-6.
  • Abdelmohsen K, Gorospe M. 2010. Posttranscriptional regulation of cancer traits by HuR. WIREs RNA 1:214–229. https://doi.org/10.1002/wrna.4.
  • Srikantan S, Gorospe M. 2012. HuR function in disease. Front Biosci 17:189–205. https://doi.org/10.2741/3921.
  • Lopez de Silanes I, Fan J, Yang X, Zonderman AB, Potapova O, Pizer ES, Gorospe M. 2003. Role of the RNA-binding protein HuR in colon carcinogenesis. Oncogene 22:7146–7154. https://doi.org/10.1038/sj.onc.1206862.
  • Wang J, Wang B, Bi J, Zhang C. 2011. Cytoplasmic HuR expression correlates with angiogenesis, lymphangiogenesis, and poor outcome in lung cancer. Med Oncol 28:577. https://doi.org/10.1007/s12032-010-9734-6.
  • Miyata Y, Watanabe S, Sagara Y, Mitsunari K, Matsuo T, Ohba K, Sakai H. 2013. High expression of HuR in cytoplasm, but not nuclei, is associated with malignant aggressiveness and prognosis in bladder cancer. PLoS One 8:e59095. https://doi.org/10.1371/journal.pone.0059095.
  • Kotta-Loizou I, Vasilopoulos SN, Coutts RH, Theocharis S. 2016. Current evidence and future perspectives on HuR and breast cancer development, prognosis, and treatment. Neoplasia 18:674–688. https://doi.org/10.1016/j.neo.2016.09.002.
  • Jimbo M, Blanco FF, Huang YH, Telonis AG, Screnci BA, Cosma GL, Alexeev V, Gonye GE, Yeo CJ, Sawicki JA, Winter JM, Brody JR. 2015. Targeting the mRNA-binding protein HuR impairs malignant characteristics of pancreatic ductal adenocarcinoma cells. Oncotarget 6:27312–27331. https://doi.org/10.18632/oncotarget.4743.
  • Blanco FF, Preet R, Aguado A, Vishwakarma V, Stevens LE, Vyas A, Padhye S, Xu L, Weir SJ, Anant S, Meisner-Kober N, Brody JR, Dixon DA. 2016. Impact of HuR inhibition by the small molecule MS-444 on colorectal cancer cell tumorigenesis. Oncotarget 7:74043–74058. https://doi.org/10.18632/oncotarget.12189.
  • Viiri J, Amadio M, Marchesi N, Hyttinen JM, Kivinen N, Sironen R, Rilla K, Akhtar S, Provenzani A, D’Agostino VG, Govoni S, Pascale A, Agostini H, Petrovski G, Salminen A, Kaarniranta K. 2013. Autophagy activation clears ELAVL1/HuR-mediated accumulation of SQSTM1/p62 during proteasomal inhibition in human retinal pigment epithelial cells. PLoS One 8:e69563. https://doi.org/10.1371/journal.pone.0069563.
  • Marchesi N, Thongon N, Pascale A, Provenzani A, Koskela A, Korhonen E, Smedowski A, Govoni S, Kauppinen A, Kaarniranta K, Amadio M. 2018. Autophagy stimulus promotes early HuR protein activation and p62/SQSTM1 protein synthesis in ARPE-19 cells by triggering Erk1/2, p38(MAPK), and JNK kinase pathways. Oxid Med Cell Longev 2018:4956080. https://doi.org/10.1155/2018/4956080.
  • Kimura S, Noda T, Yoshimori T. 2007. Dissection of the autophagosome maturation process by a novel reporter protein, tandem fluorescent-tagged LC3. Autophagy 3:452–460. https://doi.org/10.4161/auto.4451.
  • Lebedeva S, Jens M, Theil K, Schwanhausser B, Selbach M, Landthaler M, Rajewsky N. 2011. Transcriptome-wide analysis of regulatory interactions of the RNA-binding protein HuR. Mol Cell 43:340–352. https://doi.org/10.1016/j.molcel.2011.06.008.
  • Kim C, Kim W, Lee H, Ji E, Choe YJ, Martindale JL, Akamatsu W, Okano H, Kim HS, Nam SW, Gorospe M, Lee EK. 2014. The RNA-binding protein HuD regulates autophagosome formation in pancreatic beta cells by promoting autophagy-related gene 5 expression. J Biol Chem 289:112–121. https://doi.org/10.1074/jbc.M113.474700.
  • Kim KM, Cho H, Choi K, Kim J, Kim BW, Ko YG, Jang SK, Kim YK. 2009. A new MIF4G domain-containing protein, CTIF, directs nuclear cap-binding protein CBP80/20-dependent translation. Genes Dev 23:2033–2045. https://doi.org/10.1101/gad.1823409.
  • Mukaka MM. 2012. Statistics corner: a guide to appropriate use of correlation coefficient in medical research. Malawi Med J 24:69–71.
  • Palanisamy K, Tsai TH, Yu TM, Sun KT, Yu SH, Lin FY, Wang IK, Li CY. 2018. RNA-binding protein, human antigen R regulates hypoxia-induced autophagy by targeting ATG7/ATG16L1 expressions and autophagosome formation. J Cell Physiol https://doi.org/10.1002/jcp.27502.
  • Keene JD. 2007. RNA regulons: coordination of post-transcriptional events. Nat Rev Genet 8:533–543. https://doi.org/10.1038/nrg2111.
  • Kang MJ, Ryu BK, Lee MG, Han J, Lee JH, Ha TK, Byun DS, Chae KS, Lee BH, Chun HS, Lee KY, Kim HJ, Chi SG. 2008. NF-kappaB activates transcription of the RNA-binding factor HuR, via PI3K-AKT signaling, to promote gastric tumorigenesis. Gastroenterology 135:2030–2042. https://doi.org/10.1053/j.gastro.2008.08.009.
  • Abdelmohsen K, Srikantan S, Yang X, Lal A, Kim HH, Kuwano Y, Galban S, Becker KG, Kamara D, de Cabo R, Gorospe M. 2009. Ubiquitin-mediated proteolysis of HuR by heat shock. EMBO J 28:1271–1282. https://doi.org/10.1038/emboj.2009.67.
  • Lucchesi C, Sheikh MS, Huang Y. 2016. Negative regulation of RNA-binding protein HuR by tumor-suppressor ECRG2. Oncogene 35:2565–2573. https://doi.org/10.1038/onc.2015.339.
  • Cui Y, Bi M, Su T, Liu H, Lu SH. 2010. Molecular cloning and characterization of a novel esophageal cancer related gene. Int J Oncol 37:1521–1528.
  • Abdelmohsen K, Srikantan S, Kuwano Y, Gorospe M. 2008. miR-519 reduces cell proliferation by lowering RNA-binding protein HuR levels. Proc Natl Acad Sci U S A 105:20297–20302. https://doi.org/10.1073/pnas.0809376106.
  • Xu F, Zhang X, Lei Y, Liu X, Liu Z, Tong T, Wang W. 2010. Loss of repression of HuR translation by miR-16 may be responsible for the elevation of HuR in human breast carcinoma. J Cell Biochem 111:727–734. https://doi.org/10.1002/jcb.22762.
  • Roff AN, Craig TJ, August A, Stellato C, Ishmael FT. 2014. MicroRNA-570-3p regulates HuR and cytokine expression in airway epithelial cells. Am J Clin Exp Immunol 3:68–83.
  • Guo J, Li M, Meng X, Sui J, Dou L, Tang W, Huang X, Man Y, Wang S, Li J. 2014. MiR-291b-3p induces apoptosis in liver cell line NCTC1469 by reducing the level of RNA-binding protein HuR. Cell Physiol Biochem 33:810–822. https://doi.org/10.1159/000358654.
  • Abdelmohsen K, Kim MM, Srikantan S, Mercken EM, Brennan SE, Wilson GM, Cabo R, Gorospe M. 2010. miR-519 suppresses tumor growth by reducing HuR levels. Cell Cycle 9:1354–1359. https://doi.org/10.4161/cc.9.7.11164.
  • Linsley PS, Schelter J, Burchard J, Kibukawa M, Martin MM, Bartz SR, Johnson JM, Cummins JM, Raymond CK, Dai H, Chau N, Cleary M, Jackson AL, Carleton M, Lim L. 2007. Transcripts targeted by the microRNA-16 family cooperatively regulate cell cycle progression. Mol Cell Biol 27:2240–2252. https://doi.org/10.1128/MCB.02005-06.
  • D'Angelo D, Palmieri D, Mussnich P, Roche M, Wierinckx A, Raverot G, Fedele M, Croce CM, Trouillas J, Fusco A. 2012. Altered microRNA expression profile in human pituitary GH adenomas: down-regulation of miRNA targeting HMGA1, HMGA2, and E2F1. J Clin Endocrinol Metab 97:E1128–E1138. https://doi.org/10.1210/jc.2011-3482.
  • Komatsu M, Waguri S, Ueno T, Iwata J, Murata S, Tanida I, Ezaki J, Mizushima N, Ohsumi Y, Uchiyama Y, Kominami E, Tanaka K, Chiba T. 2005. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J Cell Biol 169:425–434. https://doi.org/10.1083/jcb.200412022.
  • Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, Yokoyama M, Mishima K, Saito I, Okano H, Mizushima N. 2006. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 441:885–889. https://doi.org/10.1038/nature04724.
  • Yang L, Li P, Fu S, Calay ES, Hotamisligil GS. 2010. Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance. Cell Metab 11:467–478. https://doi.org/10.1016/j.cmet.2010.04.005.
  • Chang Y, Lin J, Tsung A. 2012. Manipulation of autophagy by MIR375 generates antitumor effects in liver cancer. Autophagy 8:1833–1834. https://doi.org/10.4161/auto.21796.
  • Liu L, Liao JZ, He XX, Li PY. 2017. The role of autophagy in hepatocellular carcinoma: friend or foe. Oncotarget 8:57707–57722. https://doi.org/10.18632/oncotarget.17202.
  • Sun K, Guo XL, Zhao QD, Jing YY, Kou XR, Xie XQ, Zhou Y, Cai N, Gao L, Zhao X, Zhang SS, Song JR, Li D, Deng WJ, Li R, Wu MC, Wei LX. 2013. Paradoxical role of autophagy in the dysplastic and tumor-forming stages of hepatocarcinoma development in rats. Cell Death Dis 4:e501. https://doi.org/10.1038/cddis.2013.35.
  • Tian Y, Kuo CF, Sir D, Wang L, Govindarajan S, Petrovic LM, Ou JH. 2015. Autophagy inhibits oxidative stress and tumor suppressors to exert its dual effect on hepatocarcinogenesis. Cell Death Differ 22:1025–1034. https://doi.org/10.1038/cdd.2014.201.
  • Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A, Schwartz M, Porta C, Zeuzem S, Bolondi L, Greten TF, Galle PR, Seitz JF, Borbath I, Haussinger D, Giannaris T, Shan M, Moscovici M, Voliotis D, Bruix J, SHARP Investigators Study Group. 2008. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med 359:378–390. https://doi.org/10.1056/NEJMoa0708857.
  • Yuan H, Li AJ, Ma SL, Cui LJ, Wu B, Yin L, Wu MC. 2014. Inhibition of autophagy significantly enhances combination therapy with sorafenib and HDAC inhibitors for human hepatoma cells. World J Gastroenterol 20:4953–4962. https://doi.org/10.3748/wjg.v20.i17.4953.
  • Dash S, Chava S, Chandra PK, Aydin Y, Balart LA, Wu T. 2016. Autophagy in hepatocellular carcinomas: from pathophysiology to therapeutic response. Hepat Med 8:9–20.
  • Takamura A, Komatsu M, Hara T, Sakamoto A, Kishi C, Waguri S, Eishi Y, Hino O, Tanaka K, Mizushima N. 2011. Autophagy-deficient mice develop multiple liver tumors. Genes Dev 25:795–800. https://doi.org/10.1101/gad.2016211.
  • Kessler SM, Laggai S, Barghash A, Schultheiss CS, Lederer E, Artl M, Helms V, Haybaeck J, Kiemer AK. 2015. IMP2/p62 induces genomic instability and an aggressive hepatocellular carcinoma phenotype. Cell Death Dis 6:e1894. https://doi.org/10.1038/cddis.2015.241.
  • Xie Y, Kang R, Sun X, Zhong M, Huang J, Klionsky DJ, Tang D. 2015. Posttranslational modification of autophagy-related proteins in macroautophagy. Autophagy 11:28–45. https://doi.org/10.4161/15548627.2014.984267.
  • Wani WY, Boyer-Guittaut M, Dodson M, Chatham J, Darley-Usmar V, Zhang J. 2015. Regulation of autophagy by protein post-translational modification. Lab Invest 95:14–25. https://doi.org/10.1038/labinvest.2014.131.
  • Botti-Millet J, Nascimbeni AC, Dupont N, Morel E, Codogno P. 2016. Fine-tuning autophagy: from transcriptional to posttranslational regulation. Am J Physiol Cell Physiol 311:C351. https://doi.org/10.1152/ajpcell.00129.2016.
  • Pietrocola F, Izzo V, Niso-Santano M, Vacchelli E, Galluzzi L, Maiuri MC, Kroemer G. 2013. Regulation of autophagy by stress-responsive transcription factors. Semin Cancer Biol 23:310–322. https://doi.org/10.1016/j.semcancer.2013.05.008.
  • Fullgrabe J, Klionsky DJ, Joseph B. 2014. The return of the nucleus: transcriptional and epigenetic control of autophagy. Nat Rev Mol Cell Biol 15:65–74. https://doi.org/10.1038/nrm3716.
  • Lapierre LR, Kumsta C, Sandri M, Ballabio A, Hansen M. 2015. Transcriptional and epigenetic regulation of autophagy in aging. Autophagy 11:867–880. https://doi.org/10.1080/15548627.2015.1034410.
  • Yang L, Wang H, Shen Q, Feng L, Jin H. 2017. Long non-coding RNAs involved in autophagy regulation. Cell Death Dis 8:e3073. https://doi.org/10.1038/cddis.2017.464.
  • Hu G, McQuiston T, Bernard A, Park YD, Qiu J, Vural A, Zhang N, Waterman SR, Blewett NH, Myers TG, Maraia RJ, Kehrl JH, Uzel G, Klionsky DJ, Williamson PR. 2015. A conserved mechanism of TOR-dependent RCK-mediated mRNA degradation regulates autophagy. Nat Cell Biol 17:930–942. https://doi.org/10.1038/ncb3189.
  • Bose JK, Huang CC, Shen CK. 2011. Regulation of autophagy by neuropathological protein TDP-43. J Biol Chem 286:44441–44448. https://doi.org/10.1074/jbc.M111.237115.
  • Sánchez-Jiménez C, Izquierdo JM. 2013. T-cell intracellular antigen (TIA)-proteins deficiency in murine embryonic fibroblasts alters cell cycle progression and induces autophagy. PLoS One 8:e75127. https://doi.org/10.1371/journal.pone.0075127.
  • Yan C, Chen J, Chen N. 2016. Long noncoding RNA MALAT1 promotes hepatic steatosis and insulin resistance by increasing nuclear SREBP-1c protein stability. Sci Rep 6:22640. https://doi.org/10.1038/srep22640.
  • Chen W, Zhang L, Zhou ZQ, Ren YQ, Sun LN, Man YL, Ma ZW, Wang ZK. 2018. Effects of long non-coding RNA LINC00963 on renal interstitial fibrosis and oxidative stress of rats with chronic renal failure via the Foxo signaling pathway. Cell Physiol Biochem 46:815–828. https://doi.org/10.1159/000488739.
  • Carrascoso I, Alcalde J, Sanchez-Jimenez C, Gonzalez-Sanchez P, Izquierdo JM. 2017. T-cell intracellular antigens and Hu antigen R antagonistically modulate mitochondrial activity and dynamics by regulating optic atrophy 1 gene expression. Mol Cell Biol 37:e00174-17. https://doi.org/10.1128/MCB.00174-17.
  • Huang Y, Guerrero-Preston R, Ratovitski EA. 2012. Phospho-DeltaNp63alpha-dependent regulation of autophagic signaling through transcription and micro-RNA modulation. Cell Cycle 11:1247–1259. https://doi.org/10.4161/cc.11.6.19670.
  • Li Y, Zhou D, Ren Y, Zhang Z, Guo X, Ma M, Xue Z, Lv J, Liu H, Xi Q, Jia L, Zhang L, Liu Y, Zhang Q, Yan J, Da Y, Gao F, Yue J, Yao Z, Zhang R. 2018. Mir223 restrains autophagy and promotes CNS inflammation by targeting ATG16L1. Autophagy 14:1–15. https://doi.org/10.1080/15548627.2018.1522467.
  • Huang H, Tang J, Zhang L, Bu Y, Zhang X. 2018. miR-874 regulates multiple-drug resistance in gastric cancer by targeting ATG16L1. Int J Oncol 53:2769–2779. https://doi.org/10.3892/ijo.2018.4593.
  • Choe YJ, Ha TJ, Ko KW, Lee SY, Shin SJ, Kim HS. 2012. Anthocyanins in the black soybean (Glycine max L.) protect U2OS cells from apoptosis by inducing autophagy via the activation of adenosyl monophosphate-dependent protein kinase. Oncol Rep 28:2049–2056. https://doi.org/10.3892/or.2012.2034.
  • Schneider CA, Rasband WS, Eliceiri KW. 2012. NIH Image to ImageJ: 25 years of image analysis. Nat Methods 9:671–675. https://doi.org/10.1038/nmeth.2089.
  • Lee EK, Kim W, Tominaga K, Martindale JL, Yang X, Subaran SS, Carlson OD, Mercken EM, Kulkarni RN, Akamatsu W, Okano H, Perrone-Bizzozero NI, de Cabo R, Egan JM, Gorospe M. 2012. RNA-binding protein HuD controls insulin translation. Mol Cell 45:826–835. https://doi.org/10.1016/j.molcel.2012.01.016.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.