The Dark Side of the Force: When the Immune System Is the Fuel of Tumor Onset
Abstract
1. Introduction
2. Environmental Factors
2.1. Tobacco Smoke
2.2. Ultraviolet Light
2.3. Asbestos Fibers
3. Lifestyle
Dietary Habits and Alcohol Consumption
4. Infection-Related
4.1. Helicobacter pylori and Gastric Cancer
4.2. Viral Hepatitis and Hepatocellular Carcinoma
4.3. HIV- and AIDS-Related Cancer
4.4. Other Pathogens
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Serhan, C.N. Novel lipid mediators and resolution mechanisms in acute inflammation: To resolve or not? Am. J. Pathol. 2010, 177, 1576–1591. [Google Scholar] [CrossRef]
- Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
- Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to Virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
- Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef] [PubMed]
- Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef] [PubMed]
- Bonavita, E.; Galdiero, M.R.; Jaillon, S.; Mantovani, A. Phagocytes as Corrupted Policemen in Cancer-Related Inflammation. Adv. Cancer Res. 2015, 128, 141–171. [Google Scholar] [CrossRef] [PubMed]
- Granja, S.; Tavares-Valente, D.; Queirós, O.; Baltazar, F. Value of pH regulators in the diagnosis, prognosis and treatment of cancer. Semin. Cancer Biol. 2017, 43, 17–34. [Google Scholar] [CrossRef]
- Maeda, H.; Akaike, T. Nitric oxide and oxygen radicals in infection, inflammation, and cancer. Biochemistry 1998, 63, 854–865. [Google Scholar]
- Shacter, E.; Weitzman, S.A. Chronic inflammation and cancer. Oncology (Williston Park) 2002, 16, 217–226. [Google Scholar]
- Bohr, V.A.; Stevnsner, T.; de Souza-Pinto, N.C. Mitochondrial DNA repair of oxidative damage in mammalian cells. Gene 2002, 286, 127–134. [Google Scholar] [CrossRef]
- Van Houten, B.; Woshner, V.; Santos, J.H. Role of mitochondrial DNA in toxic responses to oxidative stress. DNA Repair 2006, 5, 145–152. [Google Scholar] [CrossRef] [PubMed]
- Shukla, A.; Jung, M.; Stern, M.; Fukagawa, N.K.; Taatjes, D.J.; Sawyer, D.; Van Houten, B.; Mossman, B.T. Asbestos induces mitochondrial DNA damage and dysfunction linked to the development of apoptosis. Am. J. Physiol. Lung Cell Mol. Physiol. 2003, 285, L1018–L1025. [Google Scholar] [CrossRef] [PubMed]
- Lebedeva, M.A.; Eaton, J.S.; Shadel, G.S. Loss of p53 causes mitochondrial DNA depletion and altered mitochondrial reactive oxygen species homeostasis. Biochim. Biophys. Acta 2009, 1787, 328–334. [Google Scholar] [CrossRef]
- Ralph, S.J.; Rodríguez-Enríquez, S.; Neuzil, J.; Saavedra, E.; Moreno-Sánchez, R. The causes of cancer revisited: “mitochondrial malignancy” and ROS-induced oncogenic transformation—Why mitochondria are targets for cancer therapy. Mol. Aspects Med. 2010, 31, 145–170. [Google Scholar] [CrossRef]
- Tubbs, A.; Nussenzweig, A. Endogenous DNA Damage as a Source of Genomic Instability in Cancer. Cell 2017, 168, 644–656. [Google Scholar] [CrossRef]
- Cadet, J.; Douki, T.; Ravanat, J.L. Oxidatively generated base damage to cellular DNA. Free Radic. Biol. Med. 2010, 49, 9–21. [Google Scholar] [CrossRef]
- Cadet, J.; Wagner, J.R. DNA base damage by reactive oxygen species, oxidizing agents, and UV radiation. Cold Spring Harb. Perspect. Biol. 2013, 5. [Google Scholar] [CrossRef]
- David, S.S.; O’Shea, V.L.; Kundu, S. Base-excision repair of oxidative DNA damage. Nature 2007, 447, 941–950. [Google Scholar] [CrossRef]
- Wall, S.B.; Oh, J.Y.; Diers, A.R.; Landar, A. Oxidative modification of proteins: An emerging mechanism of cell signaling. Front. Physiol. 2012, 3, 369. [Google Scholar] [CrossRef]
- Yang, J.; Gupta, V.; Carroll, K.S.; Liebler, D.C. Site-specific mapping and quantification of protein S-sulphenylation in cells. Nat. Commun. 2014, 5, 4776. [Google Scholar] [CrossRef] [PubMed]
- Poole, L.B. The basics of thiols and cysteines in redox biology and chemistry. Free Radic. Biol. Med. 2015, 80, 148–157. [Google Scholar] [CrossRef] [PubMed]
- Weerapana, E.; Wang, C.; Simon, G.M.; Richter, F.; Khare, S.; Dillon, M.B.; Bachovchin, D.A.; Mowen, K.; Baker, D.; Cravatt, B.F. Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 2010, 468, 790–795. [Google Scholar] [CrossRef] [PubMed]
- Ying, J.; Clavreul, N.; Sethuraman, M.; Adachi, T.; Cohen, R.A. Thiol oxidation in signaling and response to stress: Detection and quantification of physiological and pathophysiological thiol modifications. Free Radic. Biol. Med. 2007, 43, 1099–1108. [Google Scholar] [CrossRef]
- Bravard, A.; Vacher, M.; Gouget, B.; Coutant, A.; de Boisferon, F.H.; Marsin, S.; Chevillard, S.; Radicella, J.P. Redox regulation of human OGG1 activity in response to cellular oxidative stress. Mol. Cell Biol. 2006, 26, 7430–7436. [Google Scholar] [CrossRef]
- Tashiro, H.; Brenner, M.K. Immunotherapy against cancer-related viruses. Cell Res. 2017, 27, 59–73. [Google Scholar] [CrossRef]
- Karin, M. Nuclear factor-kappaB in cancer development and progression. Nature 2006, 441, 431–436. [Google Scholar] [CrossRef]
- Yu, H.; Kortylewski, M.; Pardoll, D. Crosstalk between cancer and immune cells: Role of STAT3 in the tumour microenvironment. Nat. Rev. Immunol. 2007, 7, 41–51. [Google Scholar] [CrossRef]
- Voronov, E.; Shouval, D.S.; Krelin, Y.; Cagnano, E.; Benharroch, D.; Iwakura, Y.; Dinarello, C.A.; Apte, R.N. IL-1 is required for tumor invasiveness and angiogenesis. Proc. Natl. Acad. Sci. USA 2003, 100, 2645–2650. [Google Scholar] [CrossRef]
- Grivennikov, S.; Karin, M. Autocrine IL-6 signaling: A key event in tumorigenesis? Cancer Cell 2008, 13, 7–9. [Google Scholar] [CrossRef]
- Szlosarek, P.W.; Balkwill, F.R. Tumour necrosis factor alpha: A potential target for the therapy of solid tumours. Lancet Oncol. 2003, 4, 565–573. [Google Scholar] [CrossRef]
- Langowski, J.L.; Zhang, X.; Wu, L.; Mattson, J.D.; Chen, T.; Smith, K.; Basham, B.; McClanahan, T.; Kastelein, R.A.; Oft, M. IL-23 promotes tumour incidence and growth. Nature 2006, 442, 461–465. [Google Scholar] [CrossRef] [PubMed]
- Ridker, P.M.; MacFadyen, J.G.; Thuren, T.; Everett, B.M.; Libby, P.; Glynn, R.J.; Group, C.T. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: Exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet 2017, 390, 1833–1842. [Google Scholar] [CrossRef]
- Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef] [PubMed]
- Qian, B.Z.; Pollard, J.W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010, 141, 39–51. [Google Scholar] [CrossRef] [PubMed]
- Jinushi, M.; Chiba, S.; Yoshiyama, H.; Masutomi, K.; Kinoshita, I.; Dosaka-Akita, H.; Yagita, H.; Takaoka, A.; Tahara, H. Tumor-associated macrophages regulate tumorigenicity and anticancer drug responses of cancer stem/initiating cells. Proc. Natl. Acad. Sci. USA 2011, 108, 12425–12430. [Google Scholar] [CrossRef]
- Fan, Q.M.; Jing, Y.Y.; Yu, G.F.; Kou, X.R.; Ye, F.; Gao, L.; Li, R.; Zhao, Q.D.; Yang, Y.; Lu, Z.H.; et al. Tumor-associated macrophages promote cancer stem cell-like properties via transforming growth factor-beta1-induced epithelial-mesenchymal transition in hepatocellular carcinoma. Cancer Lett. 2014, 352, 160–168. [Google Scholar] [CrossRef]
- Kogure, A.; Kosaka, N.; Ochiya, T. Cross-talk between cancer cells and their neighbors via miRNA in extracellular vesicles: An emerging player in cancer metastasis. J. Biomed. Sci. 2019, 26, 7. [Google Scholar] [CrossRef]
- Takeuchi, Y.; Nishikawa, H. Roles of regulatory T cells in cancer immunity. Int. Immunol. 2016, 28, 401–409. [Google Scholar] [CrossRef]
- Shitara, K.; Nishikawa, H. Regulatory T cells: A potential target in cancer immunotherapy. Ann. N. Y. Acad. Sci. 2018, 1417, 104–115. [Google Scholar] [CrossRef]
- Thornton, A.M.; Lu, J.; Korty, P.E.; Kim, Y.C.; Martens, C.; Sun, P.D.; Shevach, E.M. Helios(+) and Helios(-) Treg subpopulations are phenotypically and functionally distinct and express dissimilar TCR repertoires. Eur. J. Immunol. 2019, 49, 398–412. [Google Scholar] [CrossRef] [PubMed]
- Chinen, T.; Kannan, A.K.; Levine, A.G.; Fan, X.; Klein, U.; Zheng, Y.; Gasteiger, G.; Feng, Y.; Fontenot, J.D.; Rudensky, A.Y. An essential role for the IL-2 receptor in Treg cell function. Nat. Immunol. 2016, 17, 1322–1333. [Google Scholar] [CrossRef] [PubMed]
- Taylor, A.; Verhagen, J.; Blaser, K.; Akdis, M.; Akdis, C.A. Mechanisms of immune suppression by interleukin-10 and transforming growth factor-beta: The role of T regulatory cells. Immunology 2006, 117, 433–442. [Google Scholar] [CrossRef] [PubMed]
- Collison, L.W.; Workman, C.J.; Kuo, T.T.; Boyd, K.; Wang, Y.; Vignali, K.M.; Cross, R.; Sehy, D.; Blumberg, R.S.; Vignali, D.A. The inhibitory cytokine IL-35 contributes to regulatory T-cell function. Nature 2007, 450, 566–569. [Google Scholar] [CrossRef]
- Cao, X.; Cai, S.F.; Fehniger, T.A.; Song, J.; Collins, L.I.; Piwnica-Worms, D.R.; Ley, T.J. Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity 2007, 27, 635–646. [Google Scholar] [CrossRef]
- Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors in cancer therapy: A focus on T-regulatory cells. Immunol. Cell Biol. 2018, 96, 21–33. [Google Scholar] [CrossRef]
- Hecht, S.S. Lung carcinogenesis by tobacco smoke. Int. J. Cancer 2012, 131, 2724–2732. [Google Scholar] [CrossRef]
- Zhou, G. Tobacco, air pollution, environmental carcinogenesis, and thoughts on conquering strategies of lung cancer. Cancer Biol. Med. 2019, 16, 700–713. [Google Scholar] [CrossRef]
- Shaykhiev, R.; Sackrowitz, R.; Fukui, T.; Zuo, W.L.; Chao, I.W.; Strulovici-Barel, Y.; Downey, R.J.; Crystal, R.G. Smoking-induced CXCL14 expression in the human airway epithelium links chronic obstructive pulmonary disease to lung cancer. Am. J. Respir. Cell Mol. Biol. 2013, 49, 418–425. [Google Scholar] [CrossRef]
- Hussain, S.P.; Hofseth, L.J.; Harris, C.C. Radical causes of cancer. Nat. Rev. Cancer 2003, 3, 276–285. [Google Scholar] [CrossRef]
- Karin, M.; Greten, F.R. NF-kappaB: Linking inflammation and immunity to cancer development and progression. Nat. Rev. Immunol. 2005, 5, 749–759. [Google Scholar] [CrossRef] [PubMed]
- De Boer, W.I.; Sont, J.K.; van Schadewijk, A.; Stolk, J.; van Krieken, J.H.; Hiemstra, P.S. Monocyte chemoattractant protein 1, interleukin 8, and chronic airways inflammation in COPD. J. Pathol. 2000, 190, 619–626. [Google Scholar] [CrossRef]
- Doz, E.; Noulin, N.; Boichot, E.; Guénon, I.; Fick, L.; Le Bert, M.; Lagente, V.; Ryffel, B.; Schnyder, B.; Quesniaux, V.F.; et al. Cigarette smoke-induced pulmonary inflammation is TLR4/MyD88 and IL-1R1/MyD88 signaling dependent. J. Immunol. 2008, 180, 1169–1178. [Google Scholar] [CrossRef] [PubMed]
- Robays, L.J.; Maes, T.; Joos, G.F.; Vermaelen, K.Y. Between a cough and a wheeze: Dendritic cells at the nexus of tobacco smoke-induced allergic airway sensitization. Mucosal Immunol. 2009, 2, 206–219. [Google Scholar] [CrossRef] [PubMed]
- Macnee, W. Pathogenesis of chronic obstructive pulmonary disease. Clin. Chest Med. 2007, 28, 479–513. [Google Scholar] [CrossRef] [PubMed]
- Cosio, M.G.; Saetta, M.; Agusti, A. Immunologic aspects of chronic obstructive pulmonary disease. N. Engl. J. Med. 2009, 360, 2445–2454. [Google Scholar] [CrossRef]
- Tockman, M.S.; Anthonisen, N.R.; Wright, E.C.; Donithan, M.G. Airways obstruction and the risk for lung cancer. Ann. Intern. Med. 1987, 106, 512–518. [Google Scholar] [CrossRef]
- Barnes, P.J. Immunology of asthma and chronic obstructive pulmonary disease. Nat. Rev. Immunol. 2008, 8, 183–192. [Google Scholar] [CrossRef]
- Wang, G.Z.; Cheng, X.; Li, X.C.; Liu, Y.Q.; Wang, X.Q.; Shi, X.; Wang, Z.Y.; Guo, Y.Q.; Wen, Z.S.; Huang, Y.C.; et al. Tobacco smoke induces production of chemokine CCL20 to promote lung cancer. Cancer Lett. 2015, 363, 60–70. [Google Scholar] [CrossRef]
- Shiels, M.S.; Pfeiffer, R.M.; Hildesheim, A.; Engels, E.A.; Kemp, T.J.; Park, J.H.; Katki, H.A.; Koshiol, J.; Shelton, G.; Caporaso, N.E.; et al. Circulating inflammation markers and prospective risk for lung cancer. J. Natl. Cancer Inst. 2013, 105, 1871–1880. [Google Scholar] [CrossRef]
- Albrengues, J.; Shields, M.A.; Ng, D.; Park, C.G.; Ambrico, A.; Poindexter, M.E.; Upadhyay, P.; Uyeminami, D.L.; Pommier, A.; Küttner, V.; et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 2018, 361. [Google Scholar] [CrossRef]
- Freedman, N.D.; Silverman, D.T.; Hollenbeck, A.R.; Schatzkin, A.; Abnet, C.C. Association between smoking and risk of bladder cancer among men and women. JAMA 2011, 306, 737–745. [Google Scholar] [CrossRef] [PubMed]
- Cumberbatch, M.G.; Rota, M.; Catto, J.W.; La Vecchia, C. The Role of Tobacco Smoke in Bladder and Kidney Carcinogenesis: A Comparison of Exposures and Meta-analysis of Incidence and Mortality Risks. Eur. Urol. 2016, 70, 458–466. [Google Scholar] [CrossRef] [PubMed]
- Pietzak, E.J.; Mucksavage, P.; Guzzo, T.J.; Malkowicz, S.B. Heavy Cigarette Smoking and Aggressive Bladder Cancer at Initial Presentation. Urology 2015, 86, 968–972. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.H.; Shun, C.T.; Huang, K.H.; Huang, C.Y.; Tsai, Y.C.; Yu, H.J.; Pu, Y.S. Stopping smoking might reduce tumour recurrence in nonmuscle-invasive bladder cancer. BJU Int. 2007, 100, 281–286. [Google Scholar] [CrossRef]
- Wilcox, A.N.; Silverman, D.T.; Friesen, M.C.; Locke, S.J.; Russ, D.E.; Hyun, N.; Colt, J.S.; Figueroa, J.D.; Rothman, N.; Moore, L.E.; et al. Smoking status, usual adult occupation, and risk of recurrent urothelial bladder carcinoma: Data from The Cancer Genome Atlas (TCGA) Project. Cancer Causes Control 2016, 27, 1429–1435. [Google Scholar] [CrossRef]
- Lerner, C.A.; Sundar, I.K.; Yao, H.; Gerloff, J.; Ossip, D.J.; McIntosh, S.; Robinson, R.; Rahman, I. Vapors produced by electronic cigarettes and e-juices with flavorings induce toxicity, oxidative stress, and inflammatory response in lung epithelial cells and in mouse lung. PLoS ONE 2015, 10, e0116732. [Google Scholar] [CrossRef]
- Higham, A.; Bostock, D.; Booth, G.; Dungwa, J.V.; Singh, D. The effect of electronic cigarette and tobacco smoke exposure on COPD bronchial epithelial cell inflammatory responses. Int. J. Chron Obstruct. Pulmon. Dis. 2018, 13, 989–1000. [Google Scholar] [CrossRef]
- Reidel, B.; Radicioni, G.; Clapp, P.W.; Ford, A.A.; Abdelwahab, S.; Rebuli, M.E.; Haridass, P.; Alexis, N.E.; Jaspers, I.; Kesimer, M. E-Cigarette Use Causes a Unique Innate Immune Response in the Lung, Involving Increased Neutrophilic Activation and Altered Mucin Secretion. Am. J. Respir. Crit. Care Med. 2018, 197, 492–501. [Google Scholar] [CrossRef]
- Thirión-Romero, I.; Pérez-Padilla, R.; Zabert, G.; Barrientos-Gutiérrez, I. Respiratory impact of electronic cigarettes and "low-RISK" tobacco. Rev. Invest. Clin. 2019, 71, 17–27. [Google Scholar] [CrossRef]
- Mueller, M.M. Inflammation in epithelial skin tumours: Old stories and new ideas. Eur. J. Cancer 2006, 42, 735–744. [Google Scholar] [CrossRef] [PubMed]
- Lim, J.L.; Stern, R.S. High levels of ultraviolet B exposure increase the risk of non-melanoma skin cancer in psoralen and ultraviolet A-treated patients. J. Invest. Dermatol. 2005, 124, 505–513. [Google Scholar] [CrossRef] [PubMed]
- Bhattacharya, U. Uv induced skin inflammation and cancer, where is the link? Int. J. Curr. Adv. Res. 2017, 6, 8454–8463. [Google Scholar]
- Kupper, T.S.; Chua, A.O.; Flood, P.; McGuire, J.; Gubler, U. Interleukin 1 gene expression in cultured human keratinocytes is augmented by ultraviolet irradiation. J. Clin. Invest. 1987, 80, 430–436. [Google Scholar] [CrossRef] [PubMed]
- Kondo, S.; Kono, T.; Sauder, D.N.; McKenzie, R.C. IL-8 gene expression and production in human keratinocytes and their modulation by UVB. J. Invest. Dermatol. 1993, 101, 690–694. [Google Scholar] [CrossRef]
- Strickland, I.; Rhodes, L.E.; Flanagan, B.F.; Friedmann, P.S. TNF-alpha and IL-8 are upregulated in the epidermis of normal human skin after UVB exposure: Correlation with neutrophil accumulation and E-selectin expression. J. Invest. Dermatol. 1997, 108, 763–768. [Google Scholar] [CrossRef]
- Scharffetter-Kochanek, K.; Wlaschek, M.; Brenneisen, P.; Schauen, M.; Blaudschun, R.; Wenk, J. UV-induced reactive oxygen species in photocarcinogenesis and photoaging. Biol. Chem. 1997, 378, 1247–1257. [Google Scholar]
- Takashima, A.; Bergstresser, P.R. Impact of UVB radiation on the epidermal cytokine network. Photochem. Photobiol. 1996, 63, 397–400. [Google Scholar] [CrossRef]
- Clydesdale, G.J.; Dandie, G.W.; Muller, H.K. Ultraviolet light induced injury: Immunological and inflammatory effects. Immunol. Cell Biol. 2001, 79, 547–568. [Google Scholar] [CrossRef]
- Magcwebeba, T.; Riedel, S.; Swanevelder, S.; Bouic, P.; Swart, P.; Gelderblom, W. Interleukin-1α Induction in Human Keratinocytes (HaCaT): An In Vitro Model for Chemoprevention in Skin. J. Skin Cancer 2012, 2012, 393681. [Google Scholar] [CrossRef]
- Byrne, S.N.; Beaugie, C.; O’Sullivan, C.; Leighton, S.; Halliday, G.M. The immune-modulating cytokine and endogenous Alarmin interleukin-33 is upregulated in skin exposed to inflammatory UVB radiation. Am. J. Pathol. 2011, 179, 211–222. [Google Scholar] [CrossRef] [PubMed]
- Choi, Y.S.; Choi, H.J.; Min, J.K.; Pyun, B.J.; Maeng, Y.S.; Park, H.; Kim, J.; Kim, Y.M.; Kwon, Y.G. Interleukin-33 induces angiogenesis and vascular permeability through ST2/TRAF6-mediated endothelial nitric oxide production. Blood 2009, 114, 3117–3126. [Google Scholar] [CrossRef] [PubMed]
- Kai, H.; Kadono, T.; Kakinuma, T.; Tomita, M.; Ohmatsu, H.; Asano, Y.; Tada, Y.; Sugaya, M.; Sato, S. CCR10 and CCL27 are overexpressed in cutaneous squamous cell carcinoma. Pathol. Res. Pract. 2011, 207, 43–48. [Google Scholar] [CrossRef] [PubMed]
- Sung, Y.M.; He, G.; Fischer, S.M. Lack of expression of the EP2 but not EP3 receptor for prostaglandin E2 results in suppression of skin tumor development. Cancer Res. 2005, 65, 9304–9311. [Google Scholar] [CrossRef] [PubMed]
- Hata, A.N.; Breyer, R.M. Pharmacology and signaling of prostaglandin receptors: Multiple roles in inflammation and immune modulation. Pharmacol. Ther. 2004, 103, 147–166. [Google Scholar] [CrossRef]
- López-Camarillo, C.; Ocampo, E.A.; Casamichana, M.L.; Pérez-Plasencia, C.; Alvarez-Sánchez, E.; Marchat, L.A. Protein kinases and transcription factors activation in response to UV-radiation of skin: Implications for carcinogenesis. Int. J. Mol. Sci. 2012, 13, 142–172. [Google Scholar] [CrossRef] [PubMed]
- Bald, T.; Quast, T.; Landsberg, J.; Rogava, M.; Glodde, N.; Lopez-Ramos, D.; Kohlmeyer, J.; Riesenberg, S.; van den Boorn-Konijnenberg, D.; Hömig-Hölzel, C.; et al. Ultraviolet-radiation-induced inflammation promotes angiotropism and metastasis in melanoma. Nature 2014, 507, 109–113. [Google Scholar] [CrossRef]
- Robinson, B.W.; Musk, A.W.; Lake, R.A. Malignant mesothelioma. Lancet 2005, 366, 397–408. [Google Scholar] [CrossRef]
- IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Arsenic, metals, fibres, and dusts. IARC Monogr. Eval. Carcinog. Risks Hum. 2012, 100, 11–465. [Google Scholar]
- Artvinli, M.; Bariş, Y.I. Malignant mesotheliomas in a small village in the Anatolian region of Turkey: An epidemiologic study. J. Natl. Cancer Inst. 1979, 63, 17–22. [Google Scholar]
- Comba, P.; Gianfagna, A.; Paoletti, L. Pleural mesothelioma cases in Biancavilla are related to a new fluoro-edenite fibrous amphibole. Arch. Environ. Health 2003, 58, 229–232. [Google Scholar] [CrossRef] [PubMed]
- Liu, G.; Cheresh, P.; Kamp, D.W. Molecular basis of asbestos-induced lung disease. Annu. Rev. Pathol. 2013, 8, 161–187. [Google Scholar] [CrossRef] [PubMed]
- Mossman, B.T.; Lippmann, M.; Hesterberg, T.W.; Kelsey, K.T.; Barchowsky, A.; Bonner, J.C. Pulmonary endpoints (lung carcinomas and asbestosis) following inhalation exposure to asbestos. J. Toxicol. Environ. Health B Crit. Rev. 2011, 14, 76–121. [Google Scholar] [CrossRef] [PubMed]
- Gaudino, G.; Xue, J.; Yang, H. How asbestos and other fibers cause mesothelioma. Transl. Lung Cancer Res. 2020, 9, S39–S46. [Google Scholar] [CrossRef]
- Guo, G.; Chmielecki, J.; Goparaju, C.; Heguy, A.; Dolgalev, I.; Carbone, M.; Seepo, S.; Meyerson, M.; Pass, H.I. Whole-exome sequencing reveals frequent genetic alterations in BAP1, NF2, CDKN2A, and CUL1 in malignant pleural mesothelioma. Cancer Res. 2015, 75, 264–269. [Google Scholar] [CrossRef]
- Benedetti, S.; Nuvoli, B.; Catalani, S.; Galati, R. Reactive oxygen species a double-edged sword for mesothelioma. Oncotarget 2015, 6, 16848–16865. [Google Scholar] [CrossRef]
- Shukla, A.; Gulumian, M.; Hei, T.K.; Kamp, D.; Rahman, Q.; Mossman, B.T. Multiple roles of oxidants in the pathogenesis of asbestos-induced diseases. Free Radic. Biol. Med. 2003, 34, 1117–1129. [Google Scholar] [CrossRef]
- Liu, W.; Ernst, J.D.; Broaddus, V.C. Phagocytosis of crocidolite asbestos induces oxidative stress, DNA damage, and apoptosis in mesothelial cells. Am. J. Respir. Cell Mol. Biol. 2000, 23, 371–378. [Google Scholar] [CrossRef]
- Kamp, D.W.; Graceffa, P.; Pryor, W.A.; Weitzman, S.A. The role of free radicals in asbestos-induced diseases. Free Radic. Biol. Med. 1992, 12, 293–315. [Google Scholar] [CrossRef]
- Colotta, F.; Allavena, P.; Sica, A.; Garlanda, C.; Mantovani, A. Cancer-related inflammation, the seventh hallmark of cancer: Links to genetic instability. Carcinogenesis 2009, 30, 1073–1081. [Google Scholar] [CrossRef]
- Marczynski, B.; Czuppon, A.B.; Marek, W.; Reichel, G.; Baur, X. Increased incidence of DNA double-strand breaks and anti-ds DNA antibodies in blood of workers occupationally exposed to asbestos. Hum. Exp. Toxicol. 1994, 13, 3–9. [Google Scholar] [CrossRef] [PubMed]
- Yang, H.; Bocchetta, M.; Kroczynska, B.; Elmishad, A.G.; Chen, Y.; Liu, Z.; Bubici, C.; Mossman, B.T.; Pass, H.I.; Testa, J.R.; et al. TNF-alpha inhibits asbestos-induced cytotoxicity via a NF-kappaB-dependent pathway, a possible mechanism for asbestos-induced oncogenesis. Proc. Natl. Acad. Sci. USA 2006, 103, 10397–10402. [Google Scholar] [CrossRef]
- Yang, H.; Rivera, Z.; Jube, S.; Nasu, M.; Bertino, P.; Goparaju, C.; Franzoso, G.; Lotze, M.T.; Krausz, T.; Pass, H.I.; et al. Programmed necrosis induced by asbestos in human mesothelial cells causes high-mobility group box 1 protein release and resultant inflammation. Proc. Natl. Acad. Sci. USA 2010, 107, 12611–12616. [Google Scholar] [CrossRef] [PubMed]
- Hillegass, J.M.; Miller, J.M.; MacPherson, M.B.; Westbom, C.M.; Sayan, M.; Thompson, J.K.; Macura, S.L.; Perkins, T.N.; Beuschel, S.L.; Alexeeva, V.; et al. Asbestos and erionite prime and activate the NLRP3 inflammasome that stimulates autocrine cytokine release in human mesothelial cells. Part. Fibre Toxicol. 2013, 10, 39. [Google Scholar] [CrossRef] [PubMed]
- Dostert, C.; Pétrilli, V.; Van Bruggen, R.; Steele, C.; Mossman, B.T.; Tschopp, J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 2008, 320, 674–677. [Google Scholar] [CrossRef]
- Kadariya, Y.; Menges, C.W.; Talarchek, J.; Cai, K.Q.; Klein-Szanto, A.J.; Pietrofesa, R.A.; Christofidou-Solomidou, M.; Cheung, M.; Mossman, B.T.; Shukla, A.; et al. Inflammation-Related IL1β/IL1R Signaling Promotes the Development of Asbestos-Induced Malignant Mesothelioma. Cancer Prev. Res. 2016, 9, 406–414. [Google Scholar] [CrossRef]
- Christ, A.; Lauterbach, M.; Latz, E. Western Diet and the Immune System: An Inflammatory Connection. Immunity 2019, 51, 794–811. [Google Scholar] [CrossRef]
- Uranga, J.A.; López-Miranda, V.; Lombó, F.; Abalo, R. Food, nutrients and nutraceuticals affecting the course of inflammatory bowel disease. Pharmacol. Rep. 2016, 68, 816–826. [Google Scholar] [CrossRef]
- Shoelson, S.E.; Herrero, L.; Naaz, A. Obesity, inflammation, and insulin resistance. Gastroenterology 2007, 132, 2169–2180. [Google Scholar] [CrossRef]
- Mozaffarian, D.; Hao, T.; Rimm, E.B.; Willett, W.C.; Hu, F.B. Changes in diet and lifestyle and long-term weight gain in women and men. N. Engl. J. Med. 2011, 364, 2392–2404. [Google Scholar] [CrossRef]
- Tanoue, T.; Honda, K. Induction of Treg cells in the mouse colonic mucosa: A central mechanism to maintain host-microbiota homeostasis. Semin. Immunol. 2012, 24, 50–57. [Google Scholar] [CrossRef] [PubMed]
- Buc, M. Role of regulatory T cells in pathogenesis and biological therapy of multiple sclerosis. Mediat. Inflamm. 2013, 2013, 963748. [Google Scholar] [CrossRef]
- Caprara, G.; Allavena, P.; Erreni, M. Intestinal Macrophages at the Crossroad between Diet, Inflammation, and Cancer. Int. J. Mol. Sci. 2020, 21, 4825. [Google Scholar] [CrossRef]
- Darfeuille-Michaud, A.; Boudeau, J.; Bulois, P.; Neut, C.; Glasser, A.L.; Barnich, N.; Bringer, M.A.; Swidsinski, A.; Beaugerie, L.; Colombel, J.F. High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology 2004, 127, 412–421. [Google Scholar] [CrossRef] [PubMed]
- Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermúdez-Humarán, L.G.; Gratadoux, J.J.; Blugeon, S.; Bridonneau, C.; Furet, J.P.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef] [PubMed]
- Sun, P.; Wang, H.; He, Z.; Chen, X.; Wu, Q.; Chen, W.; Sun, Z.; Weng, M.; Zhu, M.; Ma, D.; et al. Fasting inhibits colorectal cancer growth by reducing M2 polarization of tumor-associated macrophages. Oncotarget 2017, 8, 74649–74660. [Google Scholar] [CrossRef]
- Lee, J.Y.; Plakidas, A.; Lee, W.H.; Heikkinen, A.; Chanmugam, P.; Bray, G.; Hwang, D.H. Differential modulation of Toll-like receptors by fatty acids: Preferential inhibition by n-3 polyunsaturated fatty acids. J. Lipid Res. 2003, 44, 479–486. [Google Scholar] [CrossRef]
- Moreira, A.P.; Texeira, T.F.; Ferreira, A.B.; Peluzio, M.o.C.; Alfenas, R.e.C. Influence of a high-fat diet on gut microbiota, intestinal permeability and metabolic endotoxaemia. Br. J. Nutr. 2012, 108, 801–809. [Google Scholar] [CrossRef]
- Zitvogel, L.; Pietrocola, F.; Kroemer, G. Nutrition, inflammation and cancer. Nat. Immunol. 2017, 18, 843–850. [Google Scholar] [CrossRef]
- Ferrante, A.W. The immune cells in adipose tissue. Diabetes Obes. Metab. 2013, 15 (Suppl. 3), 34–38. [Google Scholar] [CrossRef]
- Weisberg, S.P.; McCann, D.; Desai, M.; Rosenbaum, M.; Leibel, R.L.; Ferrante, A.W. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Invest. 2003, 112, 1796–1808. [Google Scholar] [CrossRef] [PubMed]
- McNelis, J.C.; Olefsky, J.M. Macrophages, immunity, and metabolic disease. Immunity 2014, 41, 36–48. [Google Scholar] [CrossRef] [PubMed]
- Day, S.D.; Enos, R.T.; McClellan, J.L.; Steiner, J.L.; Velázquez, K.T.; Murphy, E.A. Linking inflammation to tumorigenesis in a mouse model of high-fat-diet-enhanced colon cancer. Cytokine 2013, 64, 454–462. [Google Scholar] [CrossRef]
- Wunderlich, C.M.; Ackermann, P.J.; Ostermann, A.L.; Adams-Quack, P.; Vogt, M.C.; Tran, M.L.; Nikolajev, A.; Waisman, A.; Garbers, C.; Theurich, S.; et al. Obesity exacerbates colitis-associated cancer via IL-6-regulated macrophage polarisation and CCL-20/CCR-6-mediated lymphocyte recruitment. Nat. Commun. 2018, 9, 1646. [Google Scholar] [CrossRef] [PubMed]
- Sabharwal, S.S.; Schumacker, P.T. Mitochondrial ROS in cancer: Initiators, amplifiers or an Achilles’ heel? Nat. Rev. Cancer 2014, 14, 709–721. [Google Scholar] [CrossRef]
- Dikalov, S.I.; Nazarewicz, R.R. Angiotensin II-induced production of mitochondrial reactive oxygen species: Potential mechanisms and relevance for cardiovascular disease. Antioxid. Redox Signal. 2013, 19, 1085–1094. [Google Scholar] [CrossRef]
- Bowers, L.W.; Maximo, I.X.; Brenner, A.J.; Beeram, M.; Hursting, S.D.; Price, R.S.; Tekmal, R.R.; Jolly, C.A.; deGraffenried, L.A. NSAID use reduces breast cancer recurrence in overweight and obese women: Role of prostaglandin-aromatase interactions. Cancer Res. 2014, 74, 4446–4457. [Google Scholar] [CrossRef]
- Cui, Y.; Deming-Halverson, S.L.; Shrubsole, M.J.; Beeghly-Fadiel, A.; Cai, H.; Fair, A.M.; Shu, X.O.; Zheng, W. Use of nonsteroidal anti-inflammatory drugs and reduced breast cancer risk among overweight women. Breast Cancer Res. Treat. 2014, 146, 439–446. [Google Scholar] [CrossRef]
- Shebl, F.M.; Hsing, A.W.; Park, Y.; Hollenbeck, A.R.; Chu, L.W.; Meyer, T.E.; Koshiol, J. Non-steroidal anti-inflammatory drugs use is associated with reduced risk of inflammation-associated cancers: NIH-AARP study. PLoS ONE 2014, 9, e114633. [Google Scholar] [CrossRef]
- Mittal, S.; El-Serag, H.B. Epidemiology of hepatocellular carcinoma: Consider the population. J. Clin. Gastroenterol. 2013; 47, S2–S6. [Google Scholar] [CrossRef]
- Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef]
- Yang, Y.M.; Kim, S.Y.; Seki, E. Inflammation and Liver Cancer: Molecular Mechanisms and Therapeutic Targets. Semin. Liver Dis. 2019, 39, 26–42. [Google Scholar] [CrossRef]
- Louvet, A.; Mathurin, P. Alcoholic liver disease: Mechanisms of injury and targeted treatment. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 231–242. [Google Scholar] [CrossRef]
- Forner, A.; Reig, M.; Bruix, J. Hepatocellular carcinoma. Lancet 2018, 391, 1301–1314. [Google Scholar] [CrossRef]
- Bishayee, A. The role of inflammation and liver cancer. Adv. Exp. Med. Biol. 2014, 816, 401–435. [Google Scholar] [CrossRef] [PubMed]
- Testino, G.; Leone, S.; Borro, P. Alcohol and hepatocellular carcinoma: A review and a point of view. World J. Gastroenterol. 2014, 20, 15943–15954. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Yang, J.L.; Yu, K.K.; Xu, M.; Xu, Y.Z.; Chen, L.; Lu, Y.M.; Fang, H.S.; Wang, X.Y.; Hu, Z.Q.; et al. Activation of the NF-κB pathway as a mechanism of alcohol enhanced progression and metastasis of human hepatocellular carcinoma. Mol. Cancer 2015, 14, 10. [Google Scholar] [CrossRef]
- Buzzetti, E.; Pinzani, M.; Tsochatzis, E.A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 2016, 65, 1038–1048. [Google Scholar] [CrossRef] [PubMed]
- Akazawa, Y.; Nakao, K. To die or not to die: Death signaling in nonalcoholic fatty liver disease. J. Gastroenterol. 2018, 53, 893–906. [Google Scholar] [CrossRef] [PubMed]
- Anstee, Q.M.; Reeves, H.L.; Kotsiliti, E.; Govaere, O.; Heikenwalder, M. From NASH to HCC: Current concepts and future challenges. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 411–428. [Google Scholar] [CrossRef]
- Kuper, H.; Adami, H.O.; Trichopoulos, D. Infections as a major preventable cause of human cancer. J. Intern. Med. 2000, 248, 171–183. [Google Scholar] [CrossRef]
- Oh, J.K.; Weiderpass, E. Infection and cancer: Global distribution and burden of diseases. Ann. Glob. Health 2014, 80, 384–392. [Google Scholar] [CrossRef] [PubMed]
- Martins-Green, M.; Boudreau, N.; Bissell, M.J. Inflammation is responsible for the development of wound-induced tumors in chickens infected with Rous sarcoma virus. Cancer Res. 1994, 54, 4334–4341. [Google Scholar] [PubMed]
- Paver, E.C.; Currie, A.M.; Gupta, R.; Dahlstrom, J.E. Human papilloma virus related squamous cell carcinomas of the head and neck: Diagnosis, clinical implications and detection of HPV. Pathology 2020, 52, 179–191. [Google Scholar] [CrossRef] [PubMed]
- de Martel, C.; Franceschi, S. Infections and cancer: Established associations and new hypotheses. Crit Rev. Oncol. Hematol. 2009, 70, 183–194. [Google Scholar] [CrossRef] [PubMed]
- Matsunaga, S.; Nishiumi, S.; Tagawa, R.; Yoshida, M. Alterations in metabolic pathways in gastric epithelial cells infected with Helicobacter pylori. Microb. Pathog. 2018, 124, 122–129. [Google Scholar] [CrossRef] [PubMed]
- Osman, A.G.A. Molecular Detection of Helicobacter pylori GLmM Gene among Gastritis and Duodenitis Patients in Albogaa Specialized Hospital—Omdurman; Sudan University: Khartoum, Sudan, 2019. [Google Scholar]
- Bravo, D.; Hoare, A.; Soto, C.; Valenzuela, M.A.; Quest, A.F. Helicobacter pylori in human health and disease: Mechanisms for local gastric and systemic effects. World J. Gastroenterol 2018, 24, 3071–3089. [Google Scholar] [CrossRef]
- Khan, M.A.; Howden, C.W. Helicobacter pylori and related diseases. In Essential Medical Disorders of the Stomach and Small Intestine, A Clinical Casebook; Springer: Berlin/Heidelberg, Germany, 2019; pp. 141–154. [Google Scholar]
- Heneghan, M.A.; McCarthy, C.F.; Moran, A.P. Relationship of blood group determinants on Helicobacter pylori lipopolysaccharide with host lewis phenotype and inflammatory response. Infect. Immun. 2000, 68, 937–941. [Google Scholar] [CrossRef]
- Miyahara, R.; Niwa, Y.; Matsuura, T.; Maeda, O.; Ando, T.; Ohmiya, N.; Itoh, A.; Hirooka, Y.; Goto, H. Prevalence and prognosis of gastric cancer detected by screening in a large Japanese population: Data from a single institute over 30 years. J. Gastroenterol. Hepatol. 2007, 22, 1435–1442. [Google Scholar] [CrossRef]
- Correa, P. Gastric cancer: Overview. Gastroenterol. Clin. N. Am. 2013, 42, 211–217. [Google Scholar] [CrossRef]
- Waldum, H.L.; Kleveland, P.M.; Sørdal, Ø. Helicobacter pylori and gastric acid: An intimate and reciprocal relationship. Ther. Adv. Gastroenterol. 2016, 9, 836–844. [Google Scholar] [CrossRef]
- Machado, A.M.; Figueiredo, C.; Seruca, R.; Rasmussen, L.J. Helicobacter pylori infection generates genetic instability in gastric cells. Biochim. Biophys. Acta 2010, 1806, 58–65. [Google Scholar] [CrossRef] [PubMed]
- Park, D.I.; Park, S.H.; Kim, S.H.; Kim, J.W.; Cho, Y.K.; Kim, H.J.; Sohn, C.I.; Jeon, W.K.; Kim, B.I.; Cho, E.Y.; et al. Effect of Helicobacter pylori infection on the expression of DNA mismatch repair protein. Helicobacter 2005, 10, 179–184. [Google Scholar] [CrossRef] [PubMed]
- Kim, J.J.; Tao, H.; Carloni, E.; Leung, W.K.; Graham, D.Y.; Sepulveda, A.R. Helicobacter pylori impairs DNA mismatch repair in gastric epithelial cells. Gastroenterology 2002, 123, 542–553. [Google Scholar] [CrossRef] [PubMed]
- Kidane, D.; Murphy, D.L.; Sweasy, J.B. Accumulation of abasic sites induces genomic instability in normal human gastric epithelial cells during Helicobacter pylori infection. Oncogenesis 2014, 3, e128. [Google Scholar] [CrossRef] [PubMed]
- Chmiela, M.; Karwowska, Z.; Gonciarz, W.; Allushi, B.; Stączek, P. Host pathogen interactions in Helicobacter pylori related gastric cancer. World J. Gastroenterol. 2017, 23, 1521–1540. [Google Scholar] [CrossRef]
- Hudson, J.D.; Shoaibi, M.A.; Maestro, R.; Carnero, A.; Hannon, G.J.; Beach, D.H. A proinflammatory cytokine inhibits p53 tumor suppressor activity. J. Exp. Med. 1999, 190, 1375–1382. [Google Scholar] [CrossRef]
- Yamanishi, Y.; Boyle, D.L.; Rosengren, S.; Green, D.R.; Zvaifler, N.J.; Firestein, G.S. Regional analysis of p53 mutations in rheumatoid arthritis synovium. Proc. Natl. Acad. Sci. USA 2002, 99, 10025–10030. [Google Scholar] [CrossRef]
- Yang, J.D.; Hainaut, P.; Gores, G.J.; Amadou, A.; Plymoth, A.; Roberts, L.R. A global view of hepatocellular carcinoma: Trends, risk, prevention and management. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 589–604. [Google Scholar] [CrossRef]
- Ding, X.X.; Zhu, Q.G.; Zhang, S.M.; Guan, L.; Li, T.; Zhang, L.; Wang, S.Y.; Ren, W.L.; Chen, X.M.; Zhao, J.; et al. Precision medicine for hepatocellular carcinoma: Driver mutations and targeted therapy. Oncotarget 2017, 8, 55715–55730. [Google Scholar] [CrossRef]
- Lee, D.H.; Lee, J.M. Primary malignant tumours in the non-cirrhotic liver. Eur. J. Radiol. 2017, 95, 349–361. [Google Scholar] [CrossRef]
- El-Serag, H.B. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology 2012, 142, 1264–1273. [Google Scholar] [CrossRef] [PubMed]
- Puigvehí, M.; Moctezuma-Velázquez, C.; Villanueva, A.; Llovet, J.M. The oncogenic role of hepatitis delta virus in hepatocellular carcinoma. JHEP Rep. 2019, 1, 120–130. [Google Scholar] [CrossRef] [PubMed]
- D’souza, S.; Lau, K.C.; Coffin, C.S.; Patel, T.R. Molecular mechanisms of viral hepatitis induced hepatocellular carcinoma. World J. Gastroenterol. 2020, 26, 5759–5783. [Google Scholar] [CrossRef] [PubMed]
- Bandiera, S.; Billie Bian, C.; Hoshida, Y.; Baumert, T.F.; Zeisel, M.B. Chronic hepatitis C virus infection and pathogenesis of hepatocellular carcinoma. Curr. Opin. Virol. 2016, 20, 99–105. [Google Scholar] [CrossRef]
- Luedde, T.; Schwabe, R.F. NF-κB in the liver--linking injury, fibrosis and hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2011, 8, 108–118. [Google Scholar] [CrossRef]
- Yang, D.; Liu, N.; Zuo, C.; Lei, S.; Wu, X.; Zhou, F.; Liu, C.; Zhu, H. Innate host response in primary human hepatocytes with hepatitis C virus infection. PLoS ONE 2011, 6, e27552. [Google Scholar] [CrossRef]
- Thomas, E.; Gonzalez, V.D.; Li, Q.; Modi, A.A.; Chen, W.; Noureddin, M.; Rotman, Y.; Liang, T.J. HCV infection induces a unique hepatic innate immune response associated with robust production of type III interferons. Gastroenterology 2012, 142, 978–988. [Google Scholar] [CrossRef]
- Finkin, S.; Yuan, D.; Stein, I.; Taniguchi, K.; Weber, A.; Unger, K.; Browning, J.L.; Goossens, N.; Nakagawa, S.; Gunasekaran, G.; et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat. Immunol. 2015, 16, 1235–1244. [Google Scholar] [CrossRef]
- Yoshida, T.; Hanada, T.; Tokuhisa, T.; Kosai, K.; Sata, M.; Kohara, M.; Yoshimura, A. Activation of STAT3 by the hepatitis C virus core protein leads to cellular transformation. J. Exp. Med. 2002, 196, 641–653. [Google Scholar] [CrossRef]
- Bromberg, J.; Darnell, J.E. The role of STATs in transcriptional control and their impact on cellular function. Oncogene 2000, 19, 2468–2473. [Google Scholar] [CrossRef]
- Presser, L.D.; Haskett, A.; Waris, G. Hepatitis C virus-induced furin and thrombospondin-1 activate TGF-β1: Role of TGF-β1 in HCV replication. Virology 2011, 412, 284–296. [Google Scholar] [CrossRef] [PubMed]
- Lin, W.; Tsai, W.L.; Shao, R.X.; Wu, G.; Peng, L.F.; Barlow, L.L.; Chung, W.J.; Zhang, L.; Zhao, H.; Jang, J.Y.; et al. Hepatitis C virus regulates transforming growth factor beta1 production through the generation of reactive oxygen species in a nuclear factor kappaB-dependent manner. Gastroenterology 2010, 138, 2509–2518. [Google Scholar] [CrossRef] [PubMed]
- Bataller, R.; Lemon, S.M. Fueling fibrosis in chronic hepatitis C. Proc. Natl. Acad. Sci. USA 2012, 109, 14293–14294. [Google Scholar] [CrossRef] [PubMed]
- Ioannou, G.N.; Feld, J.J. What Are the Benefits of a Sustained Virologic Response to Direct-Acting Antiviral Therapy for Hepatitis C Virus Infection? Gastroenterology 2019, 156, 446–460. [Google Scholar] [CrossRef]
- Morgan, R.L.; Baack, B.; Smith, B.D.; Yartel, A.; Pitasi, M.; Falck-Ytter, Y. Eradication of hepatitis C virus infection and the development of hepatocellular carcinoma: A meta-analysis of observational studies. Ann. Intern. Med. 2013, 158, 329–337. [Google Scholar] [CrossRef]
- Meringer, H.; Shibolet, O.; Deutsch, L. Hepatocellular carcinoma in the post-hepatitis C virus era: Should we change the paradigm? World J. Gastroenterol. 2019, 25, 3929–3940. [Google Scholar] [CrossRef]
- Block, T.M.; Mehta, A.S.; Fimmel, C.J.; Jordan, R. Molecular viral oncology of hepatocellular carcinoma. Oncogene 2003, 22, 5093–5107. [Google Scholar] [CrossRef]
- Vandeven, N.; Nghiem, P. Pathogen-driven cancers and emerging immune therapeutic strategies. Cancer Immunol. Res. 2014, 2, 9–14. [Google Scholar] [CrossRef]
- Li, B.; Yan, C.; Zhu, J.; Chen, X.; Fu, Q.; Zhang, H.; Tong, Z.; Liu, L.; Zheng, Y.; Zhao, P.; et al. Anti-PD-1/PD-L1 Blockade Immunotherapy Employed in Treating Hepatitis B Virus Infection-Related Advanced Hepatocellular Carcinoma: A Literature Review. Front. Immunol. 2020, 11, 1037. [Google Scholar] [CrossRef]
- Weiss, R.A. How does HIV cause AIDS? Science 1993, 260, 1273–1279. [Google Scholar] [CrossRef]
- Powell, M.K.; Benková, K.; Selinger, P.; Dogoši, M.; Kinkorová Luňáčková, I.; Koutníková, H.; Laštíková, J.; Roubíčková, A.; Špůrková, Z.; Laclová, L.; et al. Opportunistic Infections in HIV-Infected Patients Differ Strongly in Frequencies and Spectra between Patients with Low CD4+ Cell Counts Examined Postmortem and Compensated Patients Examined Antemortem Irrespective of the HAART Era. PLoS ONE 2016, 11, e0162704. [Google Scholar] [CrossRef] [PubMed]
- Hernández-Ramírez, R.U.; Shiels, M.S.; Dubrow, R.; Engels, E.A. Cancer risk in HIV-infected people in the USA from 1996 to 2012: A population-based, registry-linkage study. Lancet HIV 2017, 4, e495–e504. [Google Scholar] [CrossRef]
- Engels, E.A.; Yanik, E.L.; Wheeler, W.; Gill, M.J.; Shiels, M.S.; Dubrow, R.; Althoff, K.N.; Silverberg, M.J.; Brooks, J.T.; Kitahata, M.M.; et al. Cancer-Attributable Mortality Among People with Treated Human Immunodeficiency Virus Infection in North America. Clin. Infect. Dis. 2017, 65, 636–643. [Google Scholar] [CrossRef] [PubMed]
- Shmakova, A.; Germini, D.; Vassetzky, Y. HIV-1, HAART and cancer: A complex relationship. Int. J. Cancer 2020, 146, 2666–2679. [Google Scholar] [CrossRef] [PubMed]
- Khan, S.A.; Tavolari, S.; Brandi, G. Cholangiocarcinoma: Epidemiology and risk factors. Liver Int. 2019, 39 (Suppl. 1), 19–31. [Google Scholar] [CrossRef]
- Inobaya, M.T.; Olveda, R.M.; Chau, T.N.; Olveda, D.U.; Ross, A.G. Prevention and control of schistosomiasis: A current perspective. Res. Rep. Trop. Med. 2014, 2014, 65–75. [Google Scholar] [CrossRef]
- IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Biological agents. Volume 100 B. A review of human carcinogens. IARC Monogr. Eval. Carcinog. Risks Hum. 2012, 100, 1–441. [Google Scholar]
- Sithithaworn, P.; Yongvanit, P.; Duenngai, K.; Kiatsopit, N.; Pairojkul, C. Roles of liver fluke infection as risk factor for cholangiocarcinoma. J. Hepatobiliary Pancreat. Sci. 2014, 21, 301–308. [Google Scholar] [CrossRef]
- Pak, J.H.; Lee, J.Y.; Jeon, B.Y.; Dai, F.; Yoo, W.G.; Hong, S.J. Cytokine Production in Cholangiocarcinoma Cells in Response to Clonorchis sinensis Excretory-Secretory Products and Their Putative Protein Components. Korean J. Parasitol 2019, 57, 379–387. [Google Scholar] [CrossRef]
- Zaghloul, M.S. Bladder cancer and schistosomiasis. J. Egypt. Natl. Cancer Inst. 2012, 24, 151–159. [Google Scholar] [CrossRef]
- Jones, J.L.; Hanby, A.M.; Wells, C.; Calaminici, M.; Johnson, L.; Turton, P.; Deb, R.; Provenzano, E.; Shaaban, A.; Ellis, I.O.; et al. Breast implant-associated anaplastic large cell lymphoma (BIA-ALCL): An overview of presentation and pathogenesis and guidelines for pathological diagnosis and management. Histopathology 2019, 75, 787–796. [Google Scholar] [CrossRef] [PubMed]
- Bizjak, M.; Selmi, C.; Praprotnik, S.; Bruck, O.; Perricone, C.; Ehrenfeld, M.; Shoenfeld, Y. Silicone implants and lymphoma: The role of inflammation. J. Autoimmun 2015, 65, 64–73. [Google Scholar] [CrossRef] [PubMed]
- Hu, H.; Johani, K.; Almatroudi, A.; Vickery, K.; Van Natta, B.; Kadin, M.E.; Brody, G.; Clemens, M.; Cheah, C.Y.; Lade, S.; et al. Bacterial Biofilm Infection Detected in Breast Implant-Associated Anaplastic Large-Cell Lymphoma. Plast. Reconstr. Surg. 2016, 137, 1659–1669. [Google Scholar] [CrossRef]
- Loch-Wilkinson, A.; Beath, K.J.; Knight, R.J.W.; Wessels, W.L.F.; Magnusson, M.; Papadopoulos, T.; Connell, T.; Lofts, J.; Locke, M.; Hopper, I.; et al. Breast Implant-Associated Anaplastic Large Cell Lymphoma in Australia and New Zealand: High-Surface-Area Textured Implants Are Associated with Increased Risk. Plast. Reconstr. Surg. 2017, 140, 645–654. [Google Scholar] [CrossRef] [PubMed]
- Cordeiro, P.G.; Ghione, P.; Ni, A.; Hu, Q.; Ganesan, N.; Galasso, N.; Dogan, A.; Horwitz, S.M. Risk of breast implant associated anaplastic large cell lymphoma (BIA-ALCL) in a cohort of 3546 women prospectively followed long term after reconstruction with textured breast implants. J. Plast. Reconstr. Aesthet. Surg. 2020, 73, 841–846. [Google Scholar] [CrossRef]
- Jones, P.; Mempin, M.; Hu, H.; Chowdhury, D.; Foley, M.; Cooter, R.; Adams, W.P.; Vickery, K.; Deva, A.K. The Functional Influence of Breast Implant Outer Shell Morphology on Bacterial Attachment and Growth. Plast. Reconstr. Surg. 2018, 142, 837–849. [Google Scholar] [CrossRef]
- Rothman, N.; Skibola, C.F.; Wang, S.S.; Morgan, G.; Lan, Q.; Smith, M.T.; Spinelli, J.J.; Willett, E.; De Sanjose, S.; Cocco, P.; et al. Genetic variation in TNF and IL10 and risk of non-Hodgkin lymphoma: A report from the InterLymph Consortium. Lancet Oncol. 2006, 7, 27–38. [Google Scholar] [CrossRef]
- Blombery, P.; Thompson, E.R.; Jones, K.; Arnau, G.M.; Lade, S.; Markham, J.F.; Li, J.; Deva, A.; Johnstone, R.W.; Khot, A.; et al. Whole exome sequencing reveals activating JAK1 and STAT3 mutations in breast implant-associated anaplastic large cell lymphoma anaplastic large cell lymphoma. Haematologica 2016, 101, e387–e390. [Google Scholar] [CrossRef]
- Di Napoli, A.; Jain, P.; Duranti, E.; Margolskee, E.; Arancio, W.; Facchetti, F.; Alobeid, B.; Santanelli di Pompeo, F.; Mansukhani, M.; Bhagat, G. Targeted next generation sequencing of breast implant-associated anaplastic large cell lymphoma reveals mutations in JAK/STAT signalling pathway genes, TP53 and DNMT3A. Br. J. Haematol. 2018, 180, 741–744. [Google Scholar] [CrossRef]
Infectious or Inflammatory Agents | Cancer Type | Mechanisms | References |
---|---|---|---|
Environmental Factors | |||
Tobacco smoke | Lung cancer | ROS, chronic airway inflammation | [49,51,52,53] |
Bladder cancer | [59,62,63,65] | ||
E-cigarettes | Lung cancer (potential) | Bronchial inflammation, structural damage | [69,70] |
Ultraviolet light (UV-B) | Squamous cell carcinoma | ROS, prostaglandins | [84] |
Basal cell carcinoma | Skin inflammation | [74,75,76] | |
Primary cutaneous melanoma | IL-33 | [81,82] | |
Asbestos fibers | Mesothelioma | Chronic inflammation | [92,93,94] |
ROS/RNS | [99,100] | ||
HMGB1 | [102,103] | ||
NLRP3 inflammasome | [104,105] | ||
Lifestyle | |||
Dietary habits and alcohol consumption | Colorectal cancer | Dysbiosis and Chronic low-grade systemic inflammation (IL-6-CCL20/CCR6 axis) | [123,124] |
Hepatocellular carcinoma | ROS | [128,129] | |
Infection-Related | |||
Helicobacter pylori | Gastric cancer | Downregulation of repair mechanisms, oxidative stress, MIF | [154,157,158,159] |
CagA, VacA → inflammation | [9] | ||
HCV | Hepatocellular carcinoma | Chronic inflammation (cirrhosis). NF-κB, STAT3, oxidative stress | [168,169,170,171,172,173,178] |
HBV | Chronic infection and ROS | [166,181] | |
HIV | Kaposi’s sarcoma | Immunodepression→ Reactivation of HHV-8 | [185,186] |
Non-Hodgkin lymphoma | Immunodepression→ Re-activation of EBV | ||
Cervical cancer | Immunodepression→ Reactivation of HPV | ||
Other pathogens | Cholangiocarcinoma | O. viverrini and C. sinensis→chronic inflammation | [188,191,192] |
Bladder cancer | S. haematobium→ N-nitrosamines and free radicals | [193] | |
BIA-ALCL | Capsule contents/surface material→inflammation | [194,196,201,202] |
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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Digifico, E.; Balinzo, S.; Belgiovine, C. The Dark Side of the Force: When the Immune System Is the Fuel of Tumor Onset. Int. J. Mol. Sci. 2021, 22, 1224. https://doi.org/10.3390/ijms22031224
Digifico E, Balinzo S, Belgiovine C. The Dark Side of the Force: When the Immune System Is the Fuel of Tumor Onset. International Journal of Molecular Sciences. 2021; 22(3):1224. https://doi.org/10.3390/ijms22031224
Chicago/Turabian StyleDigifico, Elisabeth, Silvia Balinzo, and Cristina Belgiovine. 2021. "The Dark Side of the Force: When the Immune System Is the Fuel of Tumor Onset" International Journal of Molecular Sciences 22, no. 3: 1224. https://doi.org/10.3390/ijms22031224
APA StyleDigifico, E., Balinzo, S., & Belgiovine, C. (2021). The Dark Side of the Force: When the Immune System Is the Fuel of Tumor Onset. International Journal of Molecular Sciences, 22(3), 1224. https://doi.org/10.3390/ijms22031224