Research article

Forskolin sensitizes pancreatic cancer cells to gemcitabine via Stat3 and Erk1/2 inhibition

  • Received: 05 May 2017 Accepted: 15 June 2017 Published: 19 June 2017
  • Cytotoxic chemotherapy, including gemcitabine-based regimens, represents the mainstay of treatment for locally advanced and metastatic pancreatic cancer. However, the response to chemotherapy is absolutely unsatisfactory and the prognosis of pancreatic cancer remains very poor. Therefore, it would be greatly beneficial to develop therapeutic approaches that cause pancreatic cancer cells to increase their sensitivity to chemotherapeutic drug gemcitabine. Forskolin is a natural cAMP elevating agent used for centuries in traditional medicine and its safety has been also documented in modern medicine. Notably, forskolin is emerging as a very interesting molecule to possibly use in cancer therapy. In the present study, we investigated the effects of forskolin on the proliferation, migration and sensitivity to gemcitabine of pancreatic cancer cells, carrying out flow cytometry-based assays of cell-cycle progression and cell death, wound-healing and MTT assays, direct cell number counting and immunoblotting experiments. Here, we show that forskolin exerts significant growth and migratory inhibitory effects on Panc-1 and AsPC-1 pancreatic cancer cells. In addition, we report that forskolin strongly enhances gemcitabine-induced antiproliferative effects by both cell cycle inhibition and cell death induction. Importantly, the forskolin-induced potentiation of antiproliferative effect by gemcitabine is preceded and accompanied by a strong inhibition of phosphorylation levels of Stat3 and Erk1/2 proteins. Altogether, our data enforce the evidence of forskolin acting as a compound with anticancer activity and provide a rationale for the design of in vivo/clinical studies exploring forskolin as a gemcitabine sensitizer/adjuvant to possibly use in pancreatic cancer patients.

    Citation: Michela Illiano, Luigi Sapio, Ilaria Caiafa, Emilio Chiosi, Annamaria Spina, Silvio Naviglio. Forskolin sensitizes pancreatic cancer cells to gemcitabine via Stat3 and Erk1/2 inhibition[J]. AIMS Molecular Science, 2017, 4(2): 224-240. doi: 10.3934/molsci.2017.2.224

    Related Papers:

  • Cytotoxic chemotherapy, including gemcitabine-based regimens, represents the mainstay of treatment for locally advanced and metastatic pancreatic cancer. However, the response to chemotherapy is absolutely unsatisfactory and the prognosis of pancreatic cancer remains very poor. Therefore, it would be greatly beneficial to develop therapeutic approaches that cause pancreatic cancer cells to increase their sensitivity to chemotherapeutic drug gemcitabine. Forskolin is a natural cAMP elevating agent used for centuries in traditional medicine and its safety has been also documented in modern medicine. Notably, forskolin is emerging as a very interesting molecule to possibly use in cancer therapy. In the present study, we investigated the effects of forskolin on the proliferation, migration and sensitivity to gemcitabine of pancreatic cancer cells, carrying out flow cytometry-based assays of cell-cycle progression and cell death, wound-healing and MTT assays, direct cell number counting and immunoblotting experiments. Here, we show that forskolin exerts significant growth and migratory inhibitory effects on Panc-1 and AsPC-1 pancreatic cancer cells. In addition, we report that forskolin strongly enhances gemcitabine-induced antiproliferative effects by both cell cycle inhibition and cell death induction. Importantly, the forskolin-induced potentiation of antiproliferative effect by gemcitabine is preceded and accompanied by a strong inhibition of phosphorylation levels of Stat3 and Erk1/2 proteins. Altogether, our data enforce the evidence of forskolin acting as a compound with anticancer activity and provide a rationale for the design of in vivo/clinical studies exploring forskolin as a gemcitabine sensitizer/adjuvant to possibly use in pancreatic cancer patients.


    加载中
    [1] Siegel RL, Miller KD, Jemal A (2016) Cancer statistics, 2016. CA Cancer J Clin 66: 7-30. doi: 10.3322/caac.21332
    [2] Ying H, Dey P, Yao W, et al. (2016) Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev 30: 355-385. doi: 10.1101/gad.275776.115
    [3] Rebelo A, Molpeceres J, Rijo P, et al. (2017) Pancreatic Cancer Therapy Review: from classic therapeutic agents to modern nanotechnologies. Curr Drug Metab 18: 346-359. doi: 10.2174/1389200218666170201151135
    [4] Fogel EL, Shahda S, Sandrasegaran K, et al. (2017) A Multidisciplinary Approach to Pancreas Cancer in 2016: A Review. Am J Gastroenterol 112: 537-554. doi: 10.1038/ajg.2016.610
    [5] Turek M, Krzyczmonik M, Bałczewski P (2016) New hopes in cancer battle - a review of new molecules and treatment strategies. Med Chem 12: 700-719. doi: 10.2174/1573406412666160502153700
    [6] Millimouno FM, Dong J, Yang L, et al. (2014) Targeting apoptosis pathways in cancer and perspectives with natural compounds from mother nature. Cancer Prev Res (Phila) 7: 1081-1107. doi: 10.1158/1940-6207.CAPR-14-0136
    [7] Naviglio S, Della Ragione F (2013) Naturally occurring molecules and anticancer combination therapies in the era of personalized medicine and economic crisis. Curr Pharm Des 19: 5325-5326. doi: 10.2174/1381612811319300001
    [8] Shanmugam MK, Lee JH, Chai EZ, et al. (2016) Cancer prevention and therapy through the modulation of transcription factors by bioactive natural compounds. Semin Cancer Biol 40-41: 35-47. doi: 10.1016/j.semcancer.2016.03.005
    [9] Sapio L, Gallo M, Illiano M, et al. (2017) The natural cAMP elevating compound forskolin in cancer therapy: Is it time? J Cell Physiol 232: 922-927. doi: 10.1002/jcp.25650
    [10] Kanne H, Burte NP, Prasanna V, et al. (2015) Extraction and elemental analysis of Coleus forskohlii extract. Pharmacognosy Res 7: 237-241. doi: 10.4103/0974-8490.157966
    [11] Godard MP, Johnson BA, Richmond SR (2005) Body composition and hormonal adaptations associated with forskolin consumption in overweight and obese men. Obes Res 13: 1335-1343. doi: 10.1038/oby.2005.162
    [12] Henderson S, Magu B, Rasmussen C, et al. (2005) Effects of Coleus forskohlii supplementation on body composition and hematological profiles in mildly overweight women. J Int Soc Sports Nutr 2: 54-62. doi: 10.1186/1550-2783-2-2-54
    [13] Loftus HL, Astell KJ, Mathai M, et al. (2015) Coleus forskohlii Extract Supplementation in Conjunction with a Hypocaloric Diet Reduces the Risk Factors of Metabolic Syndrome in Overweight and Obese Subjects: A Randomized Controlled Trial. Nutrients 7: 9508-9522. doi: 10.3390/nu7115483
    [14] Beavo JA, Brunton LL (2002) Cyclic nucleotide research -- still expanding after half a century. Nat Rev Mol Cell Biol 3: 710-718. doi: 10.1038/nrm911
    [15] Gancedo JM (2013) Biological roles of cAMP: variations on a theme in the different kingdoms of life. Biol Rev Camb Philos Soc 88: 645-668. doi: 10.1111/brv.12020
    [16] Pattabiraman DR, Bierie B, Kober KI, et al. (2016) Activation of PKA leads to mesenchymal-to-epithelial transition and loss of tumor-initiating ability. Science 351: aad3680. doi: 10.1126/science.aad3680
    [17] Follin-Arbelet V, Misund K, Hallan Naderi E, et al. (2015) The natural compound forskolin synergizes with dexamethasone to induce cell death in myeloma cells via BIM. Sci Rep 5: 13001. doi: 10.1038/srep13001
    [18] Naviglio S, Di Gesto D, Illiano F, et al. (2010) Leptin potentiates antiproliferative action of cAMP elevation via protein kinase A down-regulation in breast cancer cells. J Cell Physiol 225: 801-809. doi: 10.1002/jcp.22288
    [19] Dong H, Claffey KP, Brocke S, et al. (2015) Inhibition of breast cancer cell migration by activation of cAMP signaling. Breast Cancer Res Treat 152: 17-28. doi: 10.1007/s10549-015-3445-9
    [20] Park JY, Juhnn YS (2016) cAMP signaling increases histone deacetylase 8 expression by inhibiting JNK-dependent degradation via autophagy and the proteasome system in H1299 lung cancer cells. Biochem Biophys Res Commun 470: 336-342. doi: 10.1016/j.bbrc.2016.01.049
    [21] Cristóbal I, Rincón R, Manso R, et al. (2014) Hyperphosphorylation of PP2A in colorectal cancer and the potential therapeutic value showed by its forskolin-induced dephosphorylation and activation. Biochim Biophys Acta 1842: 1823-1829. doi: 10.1016/j.bbadis.2014.06.032
    [22] Burdyga A, Conant A, Haynes L, et al. (2013) cAMP inhibits migration, ruffling and paxillin accumulation in focal adhesions of pancreatic ductal adenocarcinoma cells: effects of PKA and EPAC. Biochim Biophys Acta 1833: 2664-2672. doi: 10.1016/j.bbamcr.2013.06.011
    [23] Quinn SN, Graves SH, Dains-McGahee C, et al. (2017) Adenylyl cyclase 3/adenylyl cyclase-associated protein 1 (CAP1) complex mediates the anti-migratory effect of forskolin in pancreatic cancer cells. Mol Carcinog 56: 1344-1360. doi: 10.1002/mc.22598
    [24] Spina A, Di Maiolo F, Esposito A, et al. (2012) cAMP Elevation Down-Regulates β3 Integrin and Focal Adhesion Kinase and Inhibits Leptin-Induced Migration of MDA-MB-231 Breast Cancer Cells. Biores Open Access 1: 324-332. doi: 10.1089/biores.2012.0270
    [25] Sapio L, Sorvillo L, Illiano M, et al. (2015) Inorganic Phosphate Prevents Erk1/2 and Stat3 Activation and Improves Sensitivity to Doxorubicin of MDA-MB-231 Breast Cancer Cells. Molecules 20: 15910-15928. doi: 10.3390/molecules200915910
    [26] Crowley LC, Scott AP, Marfell BJ, et al. (2016) Measuring Cell Death by Propidium Iodide Uptake and Flow Cytometry. Cold Spring Harb Protoc 2016: pdb.prot087163.
    [27] Thoennissen NH, Iwanski GB, Doan NB, et al. (2009) Cucurbitacin B induces apoptosis by inhibition of the JAK/STAT pathway and potentiates antiproliferative effects of gemcitabine on pancreatic cancer cells. Cancer Res 69: 5876-5884. doi: 10.1158/0008-5472.CAN-09-0536
    [28] Zhang Q, Wang H, Ran L, et al. (2016) The preclinical evaluation of TIC10/ONC201 as an anti-pancreatic cancer agent. Biochem Biophys Res Commun 476: 260-266. doi: 10.1016/j.bbrc.2016.05.106
    [29] Nagaraju GP, Mezina A, Shaib WL, et al. (2016) Targeting the Janus-activated kinase-2-STAT3 signalling pathway in pancreatic cancer using the HSP90 inhibitor ganetespib. Eur J Cancer 52: 109-119. doi: 10.1016/j.ejca.2015.10.057
    [30] Jung KH, Yan HH, Fang Z, et al. (2014) HS-104, a PI3K inhibitor, enhances the anticancer efficacy of gemcitabine in pancreatic cancer. Int J Oncol 45: 311-321.
    [31] Zimmerman NP, Roy I, Hauser AD, et al. (2015) Cyclic AMP regulates the migration and invasion potential of human pancreatic cancer cells. Mol Carcinog 54: 203-215. doi: 10.1002/mc.22091
    [32] Lee BY, Timpson P, Horvath LG, et al. (2015) FAK signaling in human cancer as a target for therapeutics. Pharmacol Ther 146: 132-149. doi: 10.1016/j.pharmthera.2014.10.001
    [33] Canel M, Serrels A, Frame MC, et al. (2013) E-cadherin-integrin crosstalk in cancer invasion and metastasis. J Cell Sci 126: 393-401. doi: 10.1242/jcs.100115
    [34] Lieberman MD, Paty P, Li XK, et al. (1996) Elevation of intracellular cyclic adenosine monophosphate inhibits the epidermal growth factor signal transduction pathway and cellular growth in pancreatic adenocarcinoma cell lines. Surgery 120: 354-359. doi: 10.1016/S0039-6060(96)80309-6
    [35] Burris HA 3rd, Moore MJ, Andersen J, et al. (1997) Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol 15: 2403-2413. doi: 10.1200/JCO.1997.15.6.2403
    [36] Ellenrieder V, König A, Seufferlein T (2016) Current Standard and Future Perspectives in First- and Second-Line Treatment of Metastatic Pancreatic Adenocarcinoma. Digestion 94: 44-49. doi: 10.1159/000447739
    [37] Moon SU, Kim JW, Sung JH, et al. (2015) p21-Activated Kinase 4 (PAK4) as a Predictive Marker of Gemcitabine Sensitivity in Pancreatic Cancer Cell Lines. Cancer Res Treat 47: 501-508.
    [38] Sumiyoshi H, Matsushita A, Nakamura Y, et al. (2016) Suppression of STAT5b in pancreatic cancer cells leads to attenuated gemcitabine chemoresistance, adhesion and invasion. Oncol Rep 35: 3216-3226.
    [39] Miao X, Koch G, Ait-Oudhia S, et al. (2016) Pharmacodynamic Modeling of Cell Cycle Effects for Gemcitabine and Trabectedin Combinations in Pancreatic Cancer Cells. Front Pharmacol 7: 421.
    [40] Morgan MA, Parsels LA, Parsels JD, et al. (2005) Role of checkpoint kinase 1 in preventing premature mitosis in response to gemcitabine. Cancer Res 65: 6835-6842. doi: 10.1158/0008-5472.CAN-04-2246
    [41] Zhang JG, Hong DF, Zhang CW, et al. (2014) Sirtuin 1 facilitates chemoresistance of pancreatic cancer cells by regulating adaptive response to chemotherapy-induced stress. Cancer Sci 105: 445-454. doi: 10.1111/cas.12364
    [42] Pan Y, Zheng M, Zhong L, et al. (2015) A preclinical evaluation of SKLB261, a multikinase inhibitor of EGFR/Src/VEGFR2, as a therapeutic agent against pancreatic cancer. Mol Cancer Ther 14: 407-418. doi: 10.1158/1535-7163.MCT-14-0485
    [43] Ozaki T, Nakamura M, Ogata T, et al. (2016) Depletion of pro-oncogenic RUNX2 enhances gemcitabine (GEM) sensitivity of p53-mutated pancreatic cancer Panc-1 cells through the induction of pro-apoptotic TAp63. Oncotarget 7: 71937-71950.
    [44] Mann KM, Ying H, Juan J, et al. (2016) KRAS-related proteins in pancreatic cancer. Pharmacol Ther 168: 29-42. doi: 10.1016/j.pharmthera.2016.09.003
    [45] Wu P, Wu D, Zhao L, et al. (2016) Prognostic role of STAT3 in solid tumors: a systematic review and meta-analysis. Oncotarget 7: 19863-19883.
    [46] Singh NS, Bernier M, Wainer IW (2016) Selective GPR55 antagonism reduces chemoresistance in cancer cells. Pharmacol Res 111: 757-766. doi: 10.1016/j.phrs.2016.07.013
    [47] He X, Wang J, Wei W, et al. (2016) Hypoxia regulates ABCG2 activity through the activation of ERK1/2/HIF-1α and contributes to chemoresistance in pancreatic cancer cells. Cancer Biol Ther 17: 188-198. doi: 10.1080/15384047.2016.1139228
    [48] Chai X, Chu H, Yang X, et al. (2015) Metformin Increases Sensitivity of Pancreatic Cancer Cells to Gemcitabine by Reducing CD133+ Cell Populations and Suppressing ERK/P70S6K Signaling. Sci Rep 5: 14404. doi: 10.1038/srep14404
    [49] Vena F, Li Causi E, Rodriguez-Justo M, et al. (2015) The MEK1/2 Inhibitor Pimasertib Enhances Gemcitabine Efficacy in Pancreatic Cancer Models by Altering Ribonucleotide Reductase Subunit-1 (RRM1). Clin Cancer Res 21: 5563-5577. doi: 10.1158/1078-0432.CCR-15-0485
    [50] Lee J, Han SI, Yun JH, et al. (2015) Quercetin 3-O-glucoside suppresses epidermal growth factor-induced migration by inhibiting EGFR signaling in pancreatic cancer cells. Tumour Biol 36: 9385-9393. doi: 10.1007/s13277-015-3682-x
    [51] Wang M, Lu X, Dong X, et al. (2015) pERK1/2 silencing sensitizes pancreatic cancer BXPC-3 cell to gemcitabine-induced apoptosis via regulating Bax and Bcl-2 expression. World J Surg Oncol 13: 66. doi: 10.1186/s12957-015-0451-7
    [52] Zheng C, Jiao X, Jiang Y, et al. (2013) ERK1/2 activity contributes to gemcitabine resistance in pancreatic cancer cells. J Int Med Res 41: 300-306. doi: 10.1177/0300060512474128
    [53] Tang Y, Liu F, Zheng C, et al. (2012) Knockdown of clusterin sensitizes pancreatic cancer cells to gemcitabine chemotherapy by ERK1/2 inactivation. J Exp Clin Cancer Res 31: 73. doi: 10.1186/1756-9966-31-73
    [54] Venkatasubbarao K, Peterson L, Zhao S, et al. (2013) Inhibiting signal transducer and activator of transcription-3 increases response to gemcitabine and delays progression of pancreatic cancer. Mol Cancer 12: 104.
    [55] Li L, Leung PS (2014) Use of herbal medicines and natural products: an alternative approach to overcoming the apoptotic resistance of pancreatic cancer. Int J Biochem Cell Biol 53: 224-236. doi: 10.1016/j.biocel.2014.05.021
    [56] Vendrely V, Peuchant E, Buscail E, et al. (2017) Resveratrol and capsaicin used together as food complements reduce tumor growth and rescue full efficiency of low dose gemcitabine in a pancreatic cancer model. Cancer Lett 390: 91-102. doi: 10.1016/j.canlet.2017.01.002
    [57] Ren X, Zhao W, Du Y, et al. (2016) Activator protein 1 promotes gemcitabine-induced apoptosis in pancreatic cancer by upregulating its downstream target Bim. Oncol Lett 12: 4732-4738.
    [58] Finbloom DS, Larner AC (1995) Regulation of the Jak/STAT signalling pathway. Cell Signal 7: 739-745. doi: 10.1016/0898-6568(95)02004-7
    [59] Yuan J, Zhang F, Niu R (2015) Multiple regulation pathways and pivotal biological functions of STAT3 in cancer. Sci Rep 5: 17663.
    [60] Li MX, Bi XY, Huang Z, et al. (2015) Prognostic Role of Phospho-STAT3 in Patients with Cancers of the Digestive System: A Systematic Review and Meta-Analysis. PLoS One 10: e0127356. doi: 10.1371/journal.pone.0127356
    [61] Zulkifli AA, Tan FH, Putoczki TL, et al. (2017) STAT3 signaling mediates tumour resistance to EGFR targeted therapeutics. Mol Cell Endocrinol 451: 15-23. doi: 10.1016/j.mce.2017.01.010
    [62] Zhao C, Li H, Lin HJ, et al. (2015) Feedback Activation of STAT3 as a Cancer Drug-Resistance Mechanism. Trends Pharmacol Sci 37: 47-61.
    [63] Johnston PA, Grandis JR (2011) STAT3 signaling: anticancer strategies and challenges. Mol Interv 11: 18-26. doi: 10.1124/mi.11.1.4
    [64] Furtek SL, Backos DS, Matheson CJ, et al. (2016) Strategies and Approaches of Targeting STAT3 for Cancer Treatment. ACS Chem Biol 11: 308-318. doi: 10.1021/acschembio.5b00945
    [65] Furukawa T (2015) Impacts of activation of the mitogen-activated protein kinase pathway in pancreatic cancer. Front Oncol 5: 23.
    [66] Neuzillet C, Hammel P, Tijeras-Raballand A, et al. (2013) Targeting the Ras-ERK pathway in pancreatic adenocarcinoma. Cancer Metastasis Rev 32: 147-162. doi: 10.1007/s10555-012-9396-2
    [67] Spina A, Di Maiolo F, Esposito A, et al. (2013) Integrating leptin and cAMP signalling pathways in triple-negative breast cancer cells. Front Biosci (Landmark Ed) 18: 133-144. doi: 10.2741/4092
    [68] Follin-Arbelet V, Torgersen ML, Naderi EH, et al. (2013) Death of multiple myeloma cells induced by cAMP-signaling involves downregulation of Mcl-1 via the JAK/STAT pathway. Cancer Lett 335: 323-331. doi: 10.1016/j.canlet.2013.02.042
  • Reader Comments
  • © 2017 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(5090) PDF downloads(1163) Cited by(5)

Article outline

Figures and Tables

Figures(5)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog