Heme Oxygenase-1 Inhibition Potentiates the Effects of Nab-Paclitaxel-Gemcitabine and Modulates the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma
Abstract
:Simple Summary
Abstract
1. Introduction
2. Materials and Methods
2.1. Cell Culture and Experimental Reagents
2.2. Preparation of Cytosolic and Nuclear Extracts
2.3. Western Blots
2.4. Apoptosis Assay
2.5. CellTiter 96 Non-Radioactive Cell Proliferation Assay (MTT)
2.6. Confocal Microscopy
2.7. Glutathione (GSH) Analysis
2.8. In Vivo Mice Model Studies
2.9. Immunohistochemistry (IHC) and Confocal Studies
2.10. RNA
2.11. Eukaryotic RNA Seq
2.12. TCGA Bioinformatics Analysis
2.13. Statistical Analysis
3. Results
3.1. HO-1 Expression in Human Pancreatic Tissues Correlates with Clinical Data
3.2. NPG Induces Ho-1 Expression in PDAC Cells through P38 Pathway and Increases Nuclear Translocation of HO-1
3.3. Inhibition of HO-1 Reduces Proliferation and Enhances the Cytotoxic Effects of NPG in PDAC and GEM-Resistant PDAC Cells but Not Ferroptosis
3.4. NPG and HO-1 Inhibition Generates Reactive Oxygen Species (ROS) and Increases Oxidized Glutathione
3.5. Inhibiting HO-1 Augments NPG-Induced Apoptosis in PDAC Cells
3.6. HO-1 Inhibition Augments NPG Cytotoxicity in the PDAC Orthotopic Xenograft Mouse Model
3.7. Transcriptome and Enrichment Analyses between NPG and NPG with HO-1 Inhibition Treatment Groups
3.8. HO-1 Inhibition with NPG Increases In Vivo Apoptosis, Decreases Macrophage and Treg Recruitment, and Increases CD8 T Cell Infiltration
4. Discussion
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
- Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [Green Version]
- Dumont, R.; Puleo, F.; Collignon, J.; Meurisse, N.; Chavez, M.; Seidel, L.; Gast, P.; Polus, M.; Loly, C.; Delvenne, P.; et al. A single center experience in resectable pancreatic ductal adenocarcinoma: The limitations of the surgery-first approach. Critical review of the literature and proposals for practice update. Acta Gastroenterol. Belg. 2017, 80, 451–461. [Google Scholar] [PubMed]
- Labori, K.J.; Katz, M.H.; Tzeng, C.W.; Bjornbeth, B.A.; Cvancarova, M.; Edwin, B.; Kure, E.H.; Eide, T.J.; Dueland, S.; Buanes, T.; et al. Impact of early disease progression and surgical complications on adjuvant chemotherapy completion rates and survival in patients undergoing the surgery first approach for resectable pancreatic ductal adenocarcinoma—A population-based cohort study. Acta Oncol. 2016, 55, 265–277. [Google Scholar] [CrossRef] [PubMed]
- Hammel, P.; Huguet, F.; van Laethem, J.L.; Goldstein, D.; Glimelius, B.; Artru, P.; Borbath, I.; Bouche, O.; Shannon, J.; Andre, T.; et al. Effect of Chemoradiotherapy vs. chemotherapy on survival in patients with locally advanced pancreatic cancer controlled after 4 months of gemcitabine with or without erlotinib: The LAP07 randomized clinical trial. JAMA 2016, 315, 1844–1853. [Google Scholar] [CrossRef] [PubMed]
- Reiser-Erkan, C.; Erkan, M.; Pan, Z.; Bekasi, S.; Giese, N.A.; Streit, S.; Michalski, C.W.; Friess, H.; Kleeff, J. Hypoxia-inducible proto-oncogene Pim-1 is a prognostic marker in pancreatic ductal adenocarcinoma. Cancer Biol. Ther. 2008, 7, 1352–1359. [Google Scholar] [CrossRef] [Green Version]
- Koong, A.C.; Mehta, V.K.; Le, Q.T.; Fisher, G.A.; Terris, D.J.; Brown, J.M.; Bastidas, A.J.; Vierra, M. Pancreatic tumors show high levels of hypoxia. Int. J. Radiat. Oncol. Biol. Phys. 2000, 48, 919–922. [Google Scholar] [CrossRef]
- Wykoff, C.C.; Pugh, C.W.; Harris, A.L.; Maxwell, P.H.; Ratcliffe, P.J. The HIF pathway: Implications for patterns of gene expression in cancer. Novartis Found. Symp. 2001, 240, 212–231. [Google Scholar] [CrossRef]
- Harris, A.L. Hypoxia—A key regulatory factor in tumour growth. Nat. Rev. Cancer 2002, 2, 38–47. [Google Scholar] [CrossRef]
- Shibaji, T.; Nagao, M.; Ikeda, N.; Kanehiro, H.; Hisanaga, M.; Ko, S.; Fukumoto, A.; Nakajima, Y. Prognostic significance of HIF-1 alpha overexpression in human pancreatic cancer. Anticancer Res. 2003, 23, 4721–4727. [Google Scholar]
- Ye, L.Y.; Zhang, Q.; Bai, X.L.; Pankaj, P.; Hu, Q.D.; Liang, T.B. Hypoxia-inducible factor 1alpha expression and its clinical significance in pancreatic cancer: A meta-analysis. Pancreatology 2014, 14, 391–397. [Google Scholar] [CrossRef] [PubMed]
- Abdalla, M.Y.; Ahmad, I.M.; Rachagani, S.; Banerjee, K.; Thompson, C.M.; Maurer, H.C.; Olive, K.P.; Bailey, K.L.; Britigan, B.E.; Kumar, S. Enhancing responsiveness of pancreatic cancer cells to gemcitabine treatment under hypoxia by heme oxygenase-1 inhibition. Transl. Res. 2019, 207, 56–69. [Google Scholar] [CrossRef] [PubMed]
- Morita, T.; Mitsialis, S.A.; Koike, H.; Liu, Y.; Kourembanas, S. Carbon monoxide controls the proliferation of hypoxic vascular smooth muscle cells. J. Biol. Chem. 1997, 272, 32804–32809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Panchenko, M.V.; Farber, H.W.; Korn, J.H. Induction of heme oxygenase-1 by hypoxia and free radicals in human dermal fibroblasts. Am. J. Physiol. Cell Physiol. 2000, 278, C92–C101. [Google Scholar] [CrossRef] [PubMed]
- Frese, K.K.; Neesse, A.; Cook, N.; Bapiro, T.E.; Lolkema, M.P.; Jodrell, D.I.; Tuveson, D.A. nab-Paclitaxel potentiates gemcitabine activity by reducing cytidine deaminase levels in a mouse model of pancreatic cancer. Cancer Discov. 2012, 2, 260–269. [Google Scholar] [CrossRef] [Green Version]
- Maines, M.D.; Gibbs, P.E. 30 some years of heme oxygenase: From a “molecular wrecking ball” to a “mesmerizing” trigger of cellular events. Biochem. Biophys. Res. Commun. 2005, 338, 568–577. [Google Scholar] [CrossRef]
- Ryter, S.W.; Alam, J.; Choi, A.M.K. Heme oxygenase-1/carbon monoxide: From basic science to therapeutic applications. Physiol. Rev. 2006, 86, 583–650. [Google Scholar] [CrossRef]
- Otterbein, L.E.; Kolls, J.K.; Mantell, L.L.; Cook, J.L.; Alam, J.; Choi, A.M.K. Exogenous administration of heme oxygenase-1 by gene transfer provides protection against hyperoxia-induced lung injury. J. Clin. Investig. 1999, 103, 1047–1054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, F.; Kaide, J.I.; Yang, L.; Jiang, H.; Quan, S.; Kemp, R.; Gong, W.; Balazy, M.; Abraham, N.G.; Nasjletti, A. CO modulates pulmonary vascular response to acute hypoxia: Relation to endothelin. Am. J. Physiol. Heart Circ. Physiol. 2004, 286, H137–H144. [Google Scholar] [CrossRef]
- Ryter, S.W.; Kim, H.P.; Nakahira, K.; Zuckerbraun, B.S.; Morse, D.; Choi, A.M. Protective functions of heme oxygenase-1 and carbon monoxide in the respiratory system. Antioxid. Redox Signal. 2007, 9, 2157–2173. [Google Scholar] [CrossRef]
- Abdalla, M.Y.; Ahmad, I.M.; Switzer, B.; Britigan, B.E. Induction of heme oxygenase-1 contributes to survival of Mycobacterium abscessus in human macrophages-like THP-1 cells. Redox Biol. 2015, 4, 328–339. [Google Scholar] [CrossRef] [Green Version]
- Shibahara, S.; Han, F.; Li, B.; Takeda, K. Hypoxia and heme oxygenases: Oxygen sensing and regulation of expression. Antioxid. Redox Signal. 2007, 9, 2209–2225. [Google Scholar] [CrossRef]
- Alam, J.; Igarashi, K.; Immenschuh, S.; Shibahara, S.; Tyrrell, R.M. Regulation of heme oxygenase-1 gene transcription: Recent advances and highlights from the International Conference (Uppsala, 2003) on Heme Oxygenase. Antioxid. Redox Signal. 2004, 6, 924–933. [Google Scholar] [CrossRef] [PubMed]
- Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rajeshkumar, N.V.; Yabuuchi, S.; Pai, S.G.; Tong, Z.; Hou, S.; Bateman, S.; Pierce, D.W.; Heise, C.; von Hoff, D.D.; Maitra, A.; et al. Superior therapeutic efficacy of nab-paclitaxel over cremophor-based paclitaxel in locally advanced and metastatic models of human pancreatic cancer. Br. J. Cancer 2016, 115, 442–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thota, R.; Pauff, J.M.; Berlin, J.D. Treatment of metastatic pancreatic adenocarcinoma: A review. Oncology 2014, 28, 70–74. [Google Scholar]
- Shukla, S.K.; Purohit, V.; Mehla, K.; Gunda, V.; Chaika, N.V.; Vernucci, E.; King, R.J.; Abrego, J.; Goode, G.D.; Dasgupta, A.; et al. MUC1 and HIF-1alpha signaling crosstalk induces anabolic glucose metabolism to impart gemcitabine resistance to pancreatic cancer. Cancer Cell 2017, 32, 392. [Google Scholar] [CrossRef]
- Goldman, M.J.; Craft, B.; Hastie, M.; Repecka, K.; McDade, F.; Kamath, A.; Banerjee, A.; Luo, Y.; Rogers, D.; Brooks, A.N.; et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat. Biotechnol. 2020, 38, 675–678. [Google Scholar] [CrossRef]
- Nakashima, K.; Sato, T.; Shigemori, S.; Shimosato, T.; Shinkai, M.; Kaneko, T. Regulatory role of heme oxygenase-1 in silica-induced lung injury. Respir. Res. 2018, 19, 144. [Google Scholar] [CrossRef]
- Schwer, C.I.; Guerrero, A.M.; Humar, M.; Roesslein, M.; Goebel, U.; Stoll, P.; Geiger, K.K.; Pannen, B.H.; Hoetzel, A.; Schmidt, R. Heme oxygenase-1 inhibits the proliferation of pancreatic stellate cells by repression of the extracellular signal-regulated kinase1/2 pathway. J. Pharmacol. Exp. Ther. 2008, 327, 863–871. [Google Scholar] [CrossRef]
- Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, W.S.; Ramaratnam, R.S.; Welsch, M.E.; Shimada, K.; Skouta, R.; Viswanathan, V.S.; Cheah, J.H.; Clemons, P.A.; Shamji, A.F.; Clish, C.B.; et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 2014, 156, 317–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kasukabe, T.; Honma, Y.; Okabe-Kado, J.; Higuchi, Y.; Kato, N.; Kumakura, S. Combined treatment with cotylenin A and phenethyl isothiocyanate induces strong antitumor activity mainly through the induction of ferroptotic cell death in human pancreatic cancer cells. Oncol. Rep. 2016, 36, 968–976. [Google Scholar] [CrossRef] [Green Version]
- Ju, H.Q.; Gocho, T.; Aguilar, M.; Wu, M.; Zhuang, Z.N.; Fu, J.; Yanaga, K.; Huang, P.; Chiao, P.J. Mechanisms of overcoming intrinsic resistance to gemcitabine in pancreatic ductal adenocarcinoma through the redox modulation. Mol. Cancer Ther. 2015, 14, 788–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ngo, E.O.; Nutter, L.M. Status of glutathione and glutathione-metabolizing enzymes in menadione-resistant human cancer cells. Biochem. Pharmacol. 1994, 47, 421–424. [Google Scholar] [CrossRef]
- Tew, K.D. Glutathione-associated enzymes in anticancer drug resistance. Cancer Res. 1994, 54, 4313–4320. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.F.; Hua, R.; Liu, D.J.; Liu, W.; Huo, Y.M.; Sun, Y.W. Effect of CD74 on the prognosis of patients with resectable pancreatic cancer. Hepatobiliary Pancreat. Dis. Int. 2014, 13, 81–86. [Google Scholar] [CrossRef]
- Koide, N.; Yamada, T.; Shibata, R.; Mori, T.; Fukuma, M.; Yamazaki, K.; Aiura, K.; Shimazu, M.; Hirohashi, S.; Nimura, Y.; et al. Establishment of perineural invasion models and analysis of gene expression revealed an invariant chain (CD74) as a possible molecule involved in perineural invasion in pancreatic cancer. Clin. Cancer Res. 2006, 12, 2419–2426. [Google Scholar] [CrossRef] [Green Version]
- Anz, D.; Mueller, W.; Golic, M.; Kunz, W.G.; Rapp, M.; Koelzer, V.H.; Ellermeier, J.; Ellwart, J.W.; Schnurr, M.; Bourquin, C.; et al. CD103 is a hallmark of tumor-infiltrating regulatory T cells. Int. J. Cancer 2011, 129, 2417–2426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Siret, C.; Collignon, A.; Silvy, F.; Robert, S.; Cheyrol, T.; Andre, P.; Rigot, V.; Iovanna, J.; van de Pavert, S.; Lombardo, D.; et al. Deciphering the crosstalk between myeloid-derived suppressor cells and regulatory t cells in pancreatic ductal adenocarcinoma. Front. Immunol. 2019, 10, 3070. [Google Scholar] [CrossRef] [PubMed]
- Daley, D.; Mani, V.R.; Mohan, N.; Akkad, N.; Ochi, A.; Heindel, D.W.; Lee, K.B.; Zambirinis, C.P.; Pandian, G.S.B.; Savadkar, S.; et al. Dectin 1 activation on macrophages by galectin 9 promotes pancreatic carcinoma and peritumoral immune tolerance. Nat. Med. 2017, 23, 556–567. [Google Scholar] [CrossRef]
- Watanabe, N.; Gavrieli, M.; Sedy, J.R.; Yang, J.; Fallarino, F.; Loftin, S.K.; Hurchla, M.A.; Zimmerman, N.; Sim, J.; Zang, X.; et al. BTLA is a lymphocyte inhibitory receptor with similarities to CTLA-4 and PD-1. Nat. Immunol. 2003, 4, 670–679. [Google Scholar] [CrossRef]
- Derre, L.; Rivals, J.P.; Jandus, C.; Pastor, S.; Rimoldi, D.; Romero, P.; Michielin, O.; Olive, D.; Speiser, D.E. BTLA mediates inhibition of human tumor-specific CD8+ T cells that can be partially reversed by vaccination. J. Clin. Investig. 2010, 120, 157–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oguro, S.; Ino, Y.; Shimada, K.; Hatanaka, Y.; Matsuno, Y.; Esaki, M.; Nara, S.; Kishi, Y.; Kosuge, T.; Hiraoka, N. Clinical significance of tumor-infiltrating immune cells focusing on BTLA and Cbl-b in patients with gallbladder cancer. Cancer Sci. 2015, 106, 1750–1760. [Google Scholar] [CrossRef]
- Bian, B.; Fanale, D.; Dusetti, N.; Roque, J.; Pastor, S.; Chretien, A.S.; Incorvaia, L.; Russo, A.; Olive, D.; Iovanna, J. Prognostic significance of circulating PD-1, PD-L1, pan-BTN3As, BTN3A1 and BTLA in patients with pancreatic adenocarcinoma. Oncoimmunology 2019, 8, e1561120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andersen, M.H.; Sorensen, R.B.; Brimnes, M.K.; Svane, I.M.; Becker, J.C.; thor Straten, P. Identification of heme oxygenase-1-specific regulatory CD8+ T cells in cancer patients. J. Clin. Investig. 2009, 119, 2245–2256. [Google Scholar] [CrossRef]
- Alaluf, E.; Vokaer, B.; Detavernier, A.; Azouz, A.; Splittgerber, M.; Carrette, A.; Boon, L.; Libert, F.; Soares, M.; le Moine, A.; et al. Heme oxygenase-1 orchestrates the immunosuppressive program of tumor-associated macrophages. JCI Insight 2020, 5. [Google Scholar] [CrossRef] [PubMed]
- Shi, C.; Washington, M.K.; Chaturvedi, R.; Drosos, Y.; Revetta, F.L.; Weaver, C.J.; Buzhardt, E.; Yull, F.E.; Blackwell, T.S.; Sosa-Pineda, B.; et al. Fibrogenesis in pancreatic cancer is a dynamic process regulated by macrophage-stellate cell interaction. Lab. Investig. 2014, 94, 409–421. [Google Scholar] [CrossRef]
- Yang, M.; McKay, D.; Pollard, J.W.; Lewis, C.E. Diverse functions of macrophages in different tumor microenvironments. Cancer Res. 2018, 78, 5492–5503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- DeNardo, D.G.; Ruffell, B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382. [Google Scholar] [CrossRef]
- Fukunaga, A.; Miyamoto, M.; Cho, Y.; Murakami, S.; Kawarada, Y.; Oshikiri, T.; Kato, K.; Kurokawa, T.; Suzuoki, M.; Nakakubo, Y.; et al. CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas 2004, 28, e26–e31. [Google Scholar] [CrossRef] [PubMed]
- Norian, L.A.; Rodriguez, P.C.; O’Mara, L.A.; Zabaleta, J.; Ochoa, A.C.; Cella, M.; Allen, P.M. Tumor-infiltrating regulatory dendritic cells inhibit CD8+ T cell function via L-arginine metabolism. Cancer Res. 2009, 69, 3086–3094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bellone, G.; Turletti, A.; Artusio, E.; Mareschi, K.; Carbone, A.; Tibaudi, D.; Robecchi, A.; Emanuelli, G.; Rodeck, U. Tumor-associated transforming growth factor-beta and interleukin-10 contribute to a systemic Th2 immune phenotype in pancreatic carcinoma patients. Am. J. Pathol. 1999, 155, 537–547. [Google Scholar] [CrossRef]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef]
- Erkan, M.; Hausmann, S.; Michalski, C.W.; Fingerle, A.A.; Dobritz, M.; Kleeff, J.; Friess, H. The role of stroma in pancreatic cancer: Diagnostic and therapeutic implications. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 454–467. [Google Scholar] [CrossRef]
- Kang, J.; Hwang, I.; Yoo, C.; Kim, K.P.; Jeong, J.H.; Chang, H.M.; Lee, S.S.; Park, D.H.; Song, T.J.; Seo, D.W.; et al. Nab-paclitaxel plus gemcitabine versus FOLFIRINOX as the first-line chemotherapy for patients with metastatic pancreatic cancer: Retrospective analysis. Investig. New Drugs 2018, 36, 732–741. [Google Scholar] [CrossRef] [PubMed]
- Von Hoff, D.D.; Ramanathan, R.K.; Borad, M.J.; Laheru, D.A.; Smith, L.S.; Wood, T.E.; Korn, R.L.; Desai, N.; Trieu, V.; Iglesias, J.L.; et al. Gemcitabine plus nab-paclitaxel is an active regimen in patients with advanced pancreatic cancer: A phase I/II trial. J. Clin. Oncol. 2011, 29, 4548–4554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ireland, L.; Santos, A.; Ahmed, M.S.; Rainer, C.; Nielsen, S.R.; Quaranta, V.; Weyer-Czernilofsky, U.; Engle, D.D.; Perez-Mancera, P.A.; Coupland, S.E.; et al. Chemoresistance in pancreatic cancer is driven by stroma-derived insulin-like growth factors. Cancer Res. 2016, 76, 6851–6863. [Google Scholar] [CrossRef] [Green Version]
- Hockel, M.; Vaupel, P. Tumor hypoxia: Definitions and current clinical, biologic, and molecular aspects. J. Natl. Cancer Inst. 2001, 93, 266–276. [Google Scholar] [CrossRef] [Green Version]
- Wang, G.L.; Jiang, B.H.; Rue, E.A.; Semenza, G.L. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc. Natl. Acad. Sci. USA 1995, 92, 5510–5514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seimiya, H.; Tanji, M.; Oh-hara, T.; Tomida, A.; Naasani, I.; Tsuruo, T. Hypoxia up-regulates telomerase activity via mitogen-activated protein kinase signaling in human solid tumor cells. Biochem. Biophys. Res. Commun. 1999, 260, 365–370. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.Z.; Zou, A.P. Transcriptional regulation of heme oxygenases by HIF-1à in renal medullary interstitial cells. Am. J. Physiol. Ren. Physiol. 2001, 281, F900–F908. [Google Scholar] [CrossRef] [Green Version]
- Lee, P.J.; Jiang, B.-H.; Chin, B.Y.; Iyer, N.V.; Alam, J.; Semenza, G.L.; Choi, A.M.K. Hypoxia-inducible factor-1 mediates transcriptional activation of the heme oxygenase-1 gene in resonse to hypoxia. J. Biol. Chem. 1997, 272, 5375–5381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ryter, S.W.; Otterbein, L.E.; Morse, D.; Choi, A.M.K. Heme oxygenase/carbon monoxide signaling pathways: Regulation and functional significance. Mol. Cell. Biochem. 2002, 234, 249–263. [Google Scholar] [CrossRef] [PubMed]
- Stocker, R.; Yamamoto, Y.; McDonagh, A.F.; Glazer, A.N.; Ames, B.N. Bilirubin is an antioxidant of possible physiological importance. Science 1987, 235, 1043–1046. [Google Scholar] [CrossRef] [PubMed]
- Van der Zee, J.; Krootjes, B.B.H.; Chignell, C.F.; Dubbelman, T.M.A.R.; van Steveninck, J. Hydroxyl radical generation by a light-dependent Fenton reaction. Free Radic. Biol. Med. 1993, 14, 105–113. [Google Scholar]
- Abraham, N.G.; Kappas, A. Pharmacological and clinical aspects of heme oxygenase. Pharmacol. Rev. 2008, 60, 79–127. [Google Scholar] [CrossRef] [Green Version]
- Tibullo, D.; Barbagallo, I.; Giallongo, C.; la Cava, P.; Parrinello, N.; Vanella, L.; Stagno, F.; Palumbo, G.A.; li Volti, G.; di Raimondo, F. Nuclear translocation of heme oxygenase-1 confers resistance to imatinib in chronic myeloid leukemia cells. Curr. Pharm. Des. 2013, 19, 2765–2770. [Google Scholar] [CrossRef]
- Biswas, C.; Shah, N.; Muthu, M.; La, P.; Fernando, A.P.; Sengupta, S.; Yang, G.; Dennery, P.A. Nuclear heme oxygenase-1 (HO-1) modulates subcellular distribution and activation of Nrf2, impacting metabolic and anti-oxidant defenses. J. Biol. Chem. 2014, 289, 26882–26894. [Google Scholar] [CrossRef] [Green Version]
- Rueff, J.; Rodrigues, A.S. Cancer drug resistance: A brief overview from a genetic viewpoint. Methods Mol. Biol. 2016, 1395, 1–18. [Google Scholar] [CrossRef]
- Nuhn, P.; Kunzli, B.M.; Hennig, R.; Mitkus, T.; Ramanauskas, T.; Nobiling, R.; Meuer, S.C.; Friess, H.; Berberat, P.O. Heme oxygenase-1 and its metabolites affect pancreatic tumor growth in vivo. Mol. Cancer 2009, 8, 37. [Google Scholar] [CrossRef] [Green Version]
- Nitti, M.; Piras, S.; Marinari, U.M.; Moretta, L.; Pronzato, M.A.; Furfaro, A.L. HO-1 induction in cancer progression: A matter of cell adaptation. Antioxidants 2017, 6, 29. [Google Scholar] [CrossRef]
- Loboda, A.; Jozkowicz, A.; Dulak, J. HO-1/CO system in tumor growth, angiogenesis and metabolism—Targeting HO-1 as an anti-tumor therapy. Vascul. Pharmacol. 2015, 74, 11–22. [Google Scholar] [CrossRef]
- Cheng, C.C.; Guan, S.S.; Yang, H.J.; Chang, C.C.; Luo, T.Y.; Chang, J.; Ho, A.S. Blocking heme oxygenase-1 by zinc protoporphyrin reduces tumor hypoxia-mediated VEGF release and inhibits tumor angiogenesis as a potential therapeutic agent against colorectal cancer. J. Biomed. Sci. 2016, 23, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Estrela, J.M.; Ortega, A.; Obrador, E. Glutathione in cancer biology and therapy. Crit. Rev. Clin. Lab. Sci. 2006, 43, 143–181. [Google Scholar] [CrossRef] [PubMed]
- Calvert, P.; Yao, K.S.; Hamilton, T.C.; O’Dwyer, P.J. Clinical studies of reversal of drug resistance based on glutathione. Chem. Biol. Interact. 1998, 111–112, 213–224. [Google Scholar] [CrossRef]
- Dodd, S.; Dean, O.; Copolov, D.L.; Malhi, G.S.; Berk, M. N-acetylcysteine for antioxidant therapy: Pharmacology and clinical utility. Expert Opin. Biol. Ther. 2008, 8, 1955–1962. [Google Scholar] [CrossRef]
- Morse, D.; Lin, L.; Choi, A.M.; Ryter, S.W. Heme oxygenase-1, a critical arbitrator of cell death pathways in lung injury and disease. Free Radic. Biol. Med. 2009, 47, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chau, L.Y. Heme oxygenase-1: Emerging target of cancer therapy. J. Biomed. Sci. 2015, 22, 22. [Google Scholar] [CrossRef] [Green Version]
- Arnold, J.N.; Magiera, L.; Kraman, M.; Fearon, D.T. Tumoral immune suppression by macrophages expressing fibroblast activation protein-alpha and heme oxygenase-1. Cancer Immunol. Res. 2014, 2, 121–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sunamura, M.; Duda, D.G.; Ghattas, M.H.; Lozonschi, L.; Motoi, F.; Yamauchi, J.; Matsuno, S.; Shibahara, S.; Abraham, N.G. Heme oxygenase-1 accelerates tumor angiogenesis of human pancreatic cancer. Angiogenesis 2003, 6, 15–24. [Google Scholar] [CrossRef]
- Mielgo, A.; Schmid, M.C. Impact of tumour associated macrophages in pancreatic cancer. BMB Rep. 2013, 46, 131–138. [Google Scholar] [CrossRef] [Green Version]
- Protti, M.P.; de Monte, L. Immune infiltrates as predictive markers of survival in pancreatic cancer patients. Front. Physiol. 2013, 4, 210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kurahara, H.; Takao, S.; Maemura, K.; Mataki, Y.; Kuwahata, T.; Maeda, K.; Sakoda, M.; Iino, S.; Ishigami, S.; Ueno, S.; et al. M2-polarized tumor-associated macrophage infiltration of regional lymph nodes is associated with nodal lymphangiogenesis and occult nodal involvement in pN0 pancreatic cancer. Pancreas 2013, 42, 155–159. [Google Scholar] [CrossRef]
- Gordon, S.; Martinez, F.O. Alternative activation of macrophages: Mechanism and functions. Immunity 2010, 32, 593–604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, C.Y.; Xu, J.Y.; Shi, X.Y.; Huang, W.; Ruan, T.Y.; Xie, P.; Ding, J.L. M2-polarized tumor-associated macrophages promoted epithelial-mesenchymal transition in pancreatic cancer cells, partially through TLR4/IL-10 signaling pathway. Lab. Investig. 2013, 93, 844–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mitchem, J.B.; Brennan, D.J.; Knolhoff, B.L.; Belt, B.A.; Zhu, Y.; Sanford, D.E.; Belaygorod, L.; Carpenter, D.; Collins, L.; Piwnica-Worms, D.; et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013, 73, 1128–1141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thyagarajan, A.; Alshehri, M.S.A.; Miller, K.L.R.; Sherwin, C.M.; Travers, J.B.; Sahu, R.P. Myeloid-derived suppressor cells and pancreatic cancer: Implications in novel therapeutic approaches. Cancers 2019, 11, 1627. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.Q.; Liu, L.; Xu, H.X.; Wu, C.T.; Xiang, J.F.; Xu, J.; Liu, C.; Long, J.; Ni, Q.X.; Yu, X.J. Infiltrating immune cells and gene mutations in pancreatic ductal adenocarcinoma. Br. J. Surg. 2016, 103, 1189–1199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, Y.F.; Lu, Y.; Cheng, H.; Shi, S.; Xu, J.; Long, J.; Liu, L.; Liu, C.; Yu, X. Abnormal distribution of peripheral lymphocyte subsets induced by PDAC modulates overall survival. Pancreatology 2014, 14, 295–301. [Google Scholar] [CrossRef]
- Liu, C.; Cheng, H.; Luo, G.; Lu, Y.; Jin, K.; Guo, M.; Ni, Q.; Yu, X. Circulating regulatory T cell subsets predict overall survival of patients with unresectable pancreatic cancer. Int. J. Oncol. 2017, 51, 686–694. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schulz, S.; Chisholm, K.M.; Zhao, H.; Kalish, F.; Yang, Y.; Wong, R.J.; Stevenson, D.K. Heme oxygenase-1 confers protection and alters T-cell populations in a mouse model of neonatal intestinal inflammation. Pediatr. Res. 2015, 77, 640–648. [Google Scholar] [CrossRef] [Green Version]
- Wong, T.H.; Chen, H.A.; Gau, R.J.; Yen, J.H.; Suen, J.L. Heme oxygenase-1-expressing dendritic cells promote Foxp3+ regulatory T cell differentiation and Induce less severe airway inflammation in murine models. PLoS ONE 2016, 11, e0168919. [Google Scholar] [CrossRef]
- Smyth, M.J.; Dunn, G.P.; Schreiber, R.D. Cancer immunosurveillance and immunoediting: The roles of immunity in suppressing tumor development and shaping tumor immunogenicity. Adv. Immunol. 2006, 90, 1–50. [Google Scholar] [CrossRef] [PubMed]
- Ademmer, K.; Ebert, M.; Muller-Ostermeyer, F.; Friess, H.; Buchler, M.W.; Schubert, W.; Malfertheiner, P. Effector T lymphocyte subsets in human pancreatic cancer: Detection of CD8+CD18+ cells and CD8+CD103+ cells by multi-epitope imaging. Clin. Exp. Immunol. 1998, 112, 21–26. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.S.; Park, Y.S.; Kim, J.Y.; Kim, Y.G.; Kim, Y.J.; Lee, H.K.; Kim, H.S.; Hong, J.T.; Kim, Y.; Han, S.B. Inhibition of human pancreatic tumor growth by cytokine-induced killer cells in nude mouse xenograft model. Immune Netw. 2012, 12, 247–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ino, Y.; Yamazaki-Itoh, R.; Shimada, K.; Iwasaki, M.; Kosuge, T.; Kanai, Y.; Hiraoka, N. Immune cell infiltration as an indicator of the immune microenvironment of pancreatic cancer. Br. J. Cancer 2013, 108, 914–923. [Google Scholar] [CrossRef]
- Smirne, C.; Camandona, M.; Alabiso, O.; Bellone, G.; Emanuelli, G. High serum levels of transforming growth factor-beta1, interleukin-10 and vascular endothelial growth factor in pancreatic adenocarcinoma patients. Minerva Gastroenterol. Dietol. 1999, 45, 21–27. [Google Scholar]
- Feng, L.; Qi, Q.; Wang, P.; Chen, H.; Chen, Z.; Meng, Z.; Liu, L. Serum levels of IL-6, IL-8, and IL-10 are indicators of prognosis in pancreatic cancer. J. Int. Med. Res. 2018, 46, 5228–5236. [Google Scholar] [CrossRef] [Green Version]
- Von Bernstorff, W.; Voss, M.; Freichel, S.; Schmid, A.; Vogel, I.; Johnk, C.; Henne-Bruns, D.; Kremer, B.; Kalthoff, H. Systemic and local immunosuppression in pancreatic cancer patients. Clin. Cancer Res. 2001, 7, 925s–932s. [Google Scholar] [PubMed]
- Tan, M.C.; Goedegebuure, P.S.; Belt, B.A.; Flaherty, B.; Sankpal, N.; Gillanders, W.E.; Eberlein, T.J.; Hsieh, C.S.; Linehan, D.C. Disruption of CCR5-dependent homing of regulatory T cells inhibits tumor growth in a murine model of pancreatic cancer. J. Immunol. 2009, 182, 1746–1755. [Google Scholar] [CrossRef] [PubMed]
- Saraiva, M.; O’Garra, A. The regulation of IL-10 production by immune cells. Nat. Rev. Immunol. 2010, 10, 170–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sierra-Filardi, E.; Vega, M.A.; Sanchez-Mateos, P.; Corbi, A.L.; Puig-Kroger, A. Heme Oxygenase-1 expression in M-CSF-polarized M2 macrophages contributes to LPS-induced IL-10 release. Immunobiology 2010, 215, 788–795. [Google Scholar] [CrossRef] [PubMed]
- Lee, T.S.; Chau, L.Y. Heme oxygenase-1 mediates the anti-inflammatory effect of interleukin-10 in mice. Nat. Med. 2002, 8, 240–246. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ahmad, I.M.; Dafferner, A.J.; O’Connell, K.A.; Mehla, K.; Britigan, B.E.; Hollingsworth, M.A.; Abdalla, M.Y. Heme Oxygenase-1 Inhibition Potentiates the Effects of Nab-Paclitaxel-Gemcitabine and Modulates the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma. Cancers 2021, 13, 2264. https://doi.org/10.3390/cancers13092264
Ahmad IM, Dafferner AJ, O’Connell KA, Mehla K, Britigan BE, Hollingsworth MA, Abdalla MY. Heme Oxygenase-1 Inhibition Potentiates the Effects of Nab-Paclitaxel-Gemcitabine and Modulates the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma. Cancers. 2021; 13(9):2264. https://doi.org/10.3390/cancers13092264
Chicago/Turabian StyleAhmad, Iman M., Alicia J. Dafferner, Kelly A. O’Connell, Kamiya Mehla, Bradley E. Britigan, Michael A. Hollingsworth, and Maher Y. Abdalla. 2021. "Heme Oxygenase-1 Inhibition Potentiates the Effects of Nab-Paclitaxel-Gemcitabine and Modulates the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma" Cancers 13, no. 9: 2264. https://doi.org/10.3390/cancers13092264
APA StyleAhmad, I. M., Dafferner, A. J., O’Connell, K. A., Mehla, K., Britigan, B. E., Hollingsworth, M. A., & Abdalla, M. Y. (2021). Heme Oxygenase-1 Inhibition Potentiates the Effects of Nab-Paclitaxel-Gemcitabine and Modulates the Tumor Microenvironment in Pancreatic Ductal Adenocarcinoma. Cancers, 13(9), 2264. https://doi.org/10.3390/cancers13092264