The effect of environmental chemicals on the tumor microenvironment
Jazyk angličtina Země Velká Británie, Anglie Médium print
Typ dokumentu časopisecké články, Research Support, N.I.H., Extramural, práce podpořená grantem, přehledy
Grantová podpora
R01 CA113447
NCI NIH HHS - United States
P30 ES000210
NIEHS NIH HHS - United States
P50CA114747
NCI NIH HHS - United States
T32CA09151
NCI NIH HHS - United States
R01CA552679
NCI NIH HHS - United States
P01CA034233
NCI NIH HHS - United States
U56CA112973
NCI NIH HHS - United States
R01 CA170378
NCI NIH HHS - United States
R01CA17037801
NCI NIH HHS - United States
T32 CA009151
NCI NIH HHS - United States
R01CA105102
NCI NIH HHS - United States
R01 CA92306-S1
NCI NIH HHS - United States
U01 CA188383
NCI NIH HHS - United States
T32ES007015
NIEHS NIH HHS - United States
T32 CA009686
NCI NIH HHS - United States
R01 CA92306
NCI NIH HHS - United States
F32CA177139
NCI NIH HHS - United States
R01CA89305
NCI NIH HHS - United States
R01 CA092306
NCI NIH HHS - United States
T32 ES007015
NIEHS NIH HHS - United States
R13 ES023276
NIEHS NIH HHS - United States
R01 CA118374
NCI NIH HHS - United States
PubMed
26106136
PubMed Central
PMC4565612
DOI
10.1093/carcin/bgv035
PII: bgv035
Knihovny.cz E-zdroje
- MeSH
- karcinogeneze chemicky indukované MeSH
- lidé MeSH
- nádorové mikroprostředí účinky léků MeSH
- nádory chemicky indukované MeSH
- nebezpečné látky škodlivé účinky MeSH
- vystavení vlivu životního prostředí škodlivé účinky MeSH
- zvířata MeSH
- Check Tag
- lidé MeSH
- zvířata MeSH
- Publikační typ
- časopisecké články MeSH
- práce podpořená grantem MeSH
- přehledy MeSH
- Research Support, N.I.H., Extramural MeSH
- Názvy látek
- nebezpečné látky MeSH
Potentially carcinogenic compounds may cause cancer through direct DNA damage or through indirect cellular or physiological effects. To study possible carcinogens, the fields of endocrinology, genetics, epigenetics, medicine, environmental health, toxicology, pharmacology and oncology must be considered. Disruptive chemicals may also contribute to multiple stages of tumor development through effects on the tumor microenvironment. In turn, the tumor microenvironment consists of a complex interaction among blood vessels that feed the tumor, the extracellular matrix that provides structural and biochemical support, signaling molecules that send messages and soluble factors such as cytokines. The tumor microenvironment also consists of many host cellular effectors including multipotent stromal cells/mesenchymal stem cells, fibroblasts, endothelial cell precursors, antigen-presenting cells, lymphocytes and innate immune cells. Carcinogens can influence the tumor microenvironment through effects on epithelial cells, the most common origin of cancer, as well as on stromal cells, extracellular matrix components and immune cells. Here, we review how environmental exposures can perturb the tumor microenvironment. We suggest a role for disrupting chemicals such as nickel chloride, Bisphenol A, butyltins, methylmercury and paraquat as well as more traditional carcinogens, such as radiation, and pharmaceuticals, such as diabetes medications, in the disruption of the tumor microenvironment. Further studies interrogating the role of chemicals and their mixtures in dose-dependent effects on the tumor microenvironment could have important general mechanistic implications for the etiology and prevention of tumorigenesis.
Cancer Research UK Cambridge Institute University of Cambridge Robinson Way CB2 0RE Cambridge UK
Center for Experimental Molecular Medicine Meibergdreef 9 1105 AZ Amsterdam The Netherlands
Centre De Recherche En Cancerologie De Lyon U1052 UMR5286 Université de Lyon 69007 Lyon France
Charles University Prague 3rd Faculty of Medicine 100 00 Prague 10 Czech Republic
Department of Experimental and Clinical Medicine University of Firenze 50134 Florence Italy
Department of Medicine University of Louisville Louisville KY 40202 USA
Department of Pathology Kuwait University 13110 Safat Kuwait
Department of Radiation Oncology NYU School of Medicine New York NY 10016 USA
Department of Science and Biomedical Technology University of Cagliari 09124 Cagliari Italy
Division of Oncology Departments of Medicine and Pathology Stanford University Stanford CA 94305 USA
Faculty of Medicine and Health Sciences Universiti Putra Malaysia Serdang 43400 Selangor Malaysia
Institute of Molecular Genetics National Research Council 27100 Pavia Italy
Mediterranean Institute of Oncology 95029 Viagrande Italy
Pathology Unit Mediterranean Institute of Oncology 95029 Viagrande Italy
School of Medicine and Public Health University of Wisconsin Madison Madison WI 53705 USA
University of Pennsylvania School of Medicine Philadelphia PA 19104 USA
Urology Department Kasr Al Ainy School of Medicine Cairo University El Manial Cairo 11562 Egypt
Zobrazit více v PubMed
Birnbaum L.S. (2013) State of the science of endocrine disruptors. Environ. Health Perspect., 121, A107. PubMed PMC
Bergman Å. et al.(eds) (2013) The State-of-the-Science of Endocrine Disrupting Chemicals – 2012, UNEP/WHO, Geneva, Switzerland.
Olumi A.F., et al. (1999) Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res., 59, 5002–5011. PubMed PMC
Le Bitoux M.A., et al. (2008) Tumor-host interactions: the role of inflammation. Histochem. Cell Biol., 130, 1079–1090. PubMed
Maffini M.V., et al. (2005) Stromal regulation of neoplastic development: age-dependent normalization of neoplastic mammary cells by mammary stroma. Am. J. Pathol., 167, 1405–1410. PubMed PMC
Weaver V.M., et al. (2004) Watch thy neighbor: cancer is a communal affair. J. Cell Sci., 117, 1287–1290. PubMed
Haddow A. (1938) Cellular inhibition and the origin of cancer. Acta Unio Int. Contra Cancrum, 3, 342–353.
Solt DaF E. (1976) New principle for the analysis of chemical carcinogenesis. Nature, 263, 701–703.
Laconi S., et al. (2001) A growth-constrained environment drives tumor progression invivo. Proc. Natl Acad. Sci. USA, 98, 7806–7811. PubMed PMC
Barcellos-Hoff M.H., et al. (2000) Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Res., 60, 1254–1260. PubMed
Laconi E., et al. (2000) The resistance phenotype in the development and treatment of cancer. Lancet Oncol., 1, 235–241. PubMed
Fleenor C.J., et al. (2010) Ionizing radiation and hematopoietic malignancies: altering the adaptive landscape. Cell Cycle, 9, 3005–3011. PubMed PMC
Coppé J.P., et al. (2008) Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol., 6, 2853–2868. PubMed PMC
Laconi E. (2007) The evolving concept of tumor microenvironments. Bioessays, 29, 738–744. PubMed
Gillies R.J., et al. (2007) Hypoxia and adaptive landscapes in the evolution of carcinogenesis. Cancer Metastasis Rev., 26, 311–317. PubMed
Potter J.D. (2007) Morphogens, morphostats, microarchitecture and malignancy. Nat. Rev. Cancer, 7, 464–474. PubMed
Marongiu F., et al. (2012) Cancer as a disease of tissue pattern formation. Prog. Histochem. Cytochem., 47, 175–207. PubMed
Tharp A.P., et al. (2012) Bisphenol A alters the development of the rhesus monkey mammary gland. Proc. Natl Acad. Sci. USA, 109, 8190–8195. PubMed PMC
Li L., et al. (2007) Non-monotonic dose-response relationship in steroid hormone receptor-mediated gene expression. J. Mol. Endocrinol., 38, 569–585. PubMed
Chai H., et al. (2009) Field effect in cancer-an update. Ann. Clin. Lab. Sci., 39, 331–337. PubMed
Lippman S.M., et al. (2009) Cancer prevention: from 1727 to milestones of the past 100 years. Cancer Res., 69, 5269–5284. PubMed
Landgren O. (2013) Monoclonal gammopathy of undetermined significance and smoldering multiple myeloma: biological insights and early treatment strategies. Hematology Am. Soc. Hematol. Educ. Program, 2013, 478–487. PubMed
Sandberg A.A., et al. (2010) Cytogenetics and genetics of human cancer: methods and accomplishments. Cancer Genet. Cytogenet., 203, 102–126. PubMed
Botti C., et al. (2000) Incidence of chromosomes 1 and 17 aneusomy in breast cancer and adjacent tissue: an interphase cytogenetic study. J. Am. Coll. Surg., 190, 530–539. PubMed
Ai H., et al. (1999) Identification of individuals at high risk for head and neck carcinogenesis using chromosome aneuploidy detected by fluorescence in situ hybridization. Mutat. Res., 439, 223–232. PubMed
Hsieh J.C., et al. (2013) Large chromosome deletions, duplications, and gene conversion events accumulate with age in normal human colon crypts. Aging Cell, 12, 269–279. PubMed PMC
Chandran U.R., et al. (2005) Differences in gene expression in prostate cancer, normal appearing prostate tissue adjacent to cancer and prostate tissue from cancer free organ donors. BMC Cancer, 5, 45. PubMed PMC
Hu B., et al. (2012) Multifocal epithelial tumors and field cancerization from loss of mesenchymal CSL signaling. Cell, 149, 1207–1220. PubMed PMC
Crowder S.W., et al. (2013) Passage-dependent cancerous transformation of human mesenchymal stem cells under carcinogenic hypoxia. FASEB J., 27, 2788–2798. PubMed PMC
Cameron K.S., et al. (2011) Exploring the molecular mechanisms of nickel-induced genotoxicity and carcinogenicity: a literature review. Rev. Environ. Health, 26, 81–92. PubMed PMC
Esteller M. (2007) Epigenetic gene silencing in cancer: the DNA hypermethylome. Hum. Mol. Genet., 16(Spec No 1), R50–R59. PubMed
Pogribny I.P., et al. (2013) DNA methylome alterations in chemical carcinogenesis. Cancer Lett., 334, 39–45. PubMed
Bollati V., et al. (2007) Changes in DNA methylation patterns in subjects exposed to low-dose benzene. Cancer Res., 67, 876–880. PubMed
Balkwill F., et al. (2001) Inflammation and cancer: back to Virchow? Lancet, 357, 539–545. PubMed
Lutgens M.W., et al. (2013) Declining risk of colorectal cancer in inflammatory bowel disease: an updated meta-analysis of population-based cohort studies. Inflamm. Bowel Dis., 19, 789–799. PubMed
Hubbard R., et al. (2000) Lung cancer and cryptogenic fibrosing alveolitis. A population-based cohort study. Am. J. Respir. Crit. Care Med., 161, 5–8. PubMed
Grivennikov S.I., et al. (2010) Immunity, inflammation, and cancer. Cell, 140, 883–899. PubMed PMC
Meira L.B., et al. (2008) DNA damage induced by chronic inflammation contributes to colon carcinogenesis in mice. J. Clin. Invest., 118, 2516–2525. PubMed PMC
Hasselbalch H.C. (2013) Chronic inflammation as a promotor of mutagenesis in essential thrombocythemia, polycythemia vera and myelofibrosis. A human inflammation model for cancer development? Leuk. Res., 37, 214–220. PubMed
Schwitalla S., et al. (2013) Intestinal tumorigenesis initiated by dedifferentiation and acquisition of stem-cell-like properties. Cell, 152, 25–38. PubMed
Barker N., et al. (2009) Crypt stem cells as the cells-of-origin of intestinal cancer. Nature, 457, 608–611. PubMed
Dostert C., et al. (2008) Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science, 320, 674–677. PubMed PMC
Liu G., et al. (2013) Molecular basis of asbestos-induced lung disease. Annu. Rev. Pathol., 8, 161–187. PubMed PMC
Cheng C.Y., et al. (2009) IL-1 beta induces urokinase-plasminogen activator expression and cell migration through PKC alpha, JNK1/2, and NF-kappaB in A549 cells. J. Cell. Physiol., 219, 183–193. PubMed
Hong Y.S., et al. (2012) Methylmercury exposure and health effects. J. Prev. Med. Public Health, 45, 353–363. PubMed PMC
He X., et al. (2012) Resveratrol inhibits paraquat-induced oxidative stress and fibrogenic response by activating the nuclear factor erythroid 2-related factor 2 pathway. J. Pharmacol. Exp. Ther., 342, 81–90. PubMed PMC
Murphy G., et al. (2008) Progress in matrix metalloproteinase research. Mol. Aspects Med., 29, 290–308. PubMed PMC
Shuman Moss L.A., et al. (2012) Matrix metalloproteinases: changing roles in tumor progression and metastasis. Am. J. Pathol., 181, 1895–1899. PubMed PMC
Birkedal-Hansen H., et al. (1993) Matrix metalloproteinases: a review. Crit. Rev. Oral Biol. Med., 4, 197–250. PubMed
Knights A.J., et al. (2012) Holding tight: cell junctions and cancer spread. Trends Cancer Res., 8, 61–69. PubMed PMC
Orlichenko L.S., et al. (2008) Matrix metalloproteinases stimulate epithelial-mesenchymal transition during tumor development. Clin. Exp. Metastasis, 25, 593–600. PubMed
Rico-Leo E.M., et al. (2013) Dioxin receptor expression inhibits basal and transforming growth factor β-induced epithelial-to-mesenchymal transition. J. Biol. Chem., 288, 7841–7856. PubMed PMC
Chen Y., et al. (2012) Nuclear receptors in the multidrug resistance through the regulation of drug-metabolizing enzymes and drug transporters. Biochem. Pharmacol., 83, 1112–1126. PubMed PMC
Pontillo C.A., et al. (2013) Action of hexachlorobenzene on tumor growth and metastasis in different experimental models. Toxicol. Appl. Pharmacol., 268, 331–342. PubMed
Haque M., et al. (2005) Aryl hydrocarbon exposure induces expression of MMP-9 in human prostate cancer cell lines. Cancer Lett., 225, 159–166. PubMed
Villano C.M., et al. (2006) 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induces matrix metalloproteinase (MMP) expression and invasion in A2058 melanoma cells. Toxicol. Appl. Pharmacol., 210, 212–224. PubMed
Peng T.L., et al. (2009) Aryl hydrocarbon receptor pathway activation enhances gastric cancer cell invasiveness likely through a c-Jun-dependent induction of matrix metalloproteinase-9. BMC Cell Biol., 10, 27. PubMed PMC
Ishida M., et al. (2010) Activation of the aryl hydrocarbon receptor pathway enhances cancer cell invasion by upregulating the MMP expression and is associated with poor prognosis in upper urinary tract urothelial cancer. Carcinogenesis, 31, 287–295. PubMed
Pande K., et al. (2005) Aspects of dioxin toxicity are mediated by interleukin 1-like cytokines. Mol. Pharmacol., 67, 1393–1398. PubMed
Suganuma M., et al. (1999) Essential role of tumor necrosis factor alpha (TNF-alpha) in tumor promotion as revealed by TNF-alpha-deficient mice. Cancer Res., 59, 4516–4518. PubMed
Popivanova B.K., et al. (2008) Blocking TNF-alpha in mice reduces colorectal carcinogenesis associated with chronic colitis. J. Clin. Invest., 118, 560–570. PubMed PMC
Moore R.J., et al. (1999) Mice deficient in tumor necrosis factor-alpha are resistant to skin carcinogenesis. Nat. Med., 5, 828–831. PubMed
Krelin Y., et al. (2007) Interleukin-1beta-driven inflammation promotes the development and invasiveness of chemical carcinogen-induced tumors. Cancer Res., 67, 1062–1071. PubMed
Tran C., et al. (2013) Inhibition of constitutive aryl hydrocarbon receptor (AhR) signaling attenuates androgen independent signaling and growth in (C4-2) prostate cancer cells. Biochem. Pharmacol., 85, 753–762. PubMed PMC
Wormke M., et al. (2003) The aryl hydrocarbon receptor mediates degradation of estrogen receptor alpha through activation of proteasomes. Mol. Cell. Biol., 23, 1843–1855. PubMed PMC
Rüegg J., et al. (2008) The transcription factor aryl hydrocarbon receptor nuclear translocator functions as an estrogen receptor beta-selective coactivator, and its recruitment to alternative pathways mediates antiestrogenic effects of dioxin. Mol. Endocrinol., 22, 304–316. PubMed PMC
Sorrell J.M., et al. (2009) Fibroblasts-a diverse population at the center of it all. Int. Rev. Cell Mol. Biol., 276, 161–214. PubMed
Cirri P., et al. (2011) Cancer associated fibroblasts: the dark side of the coin. Am. J. Cancer Res., 1, 482–497. PubMed PMC
Shimoda M., et al. (2010) Carcinoma-associated fibroblasts are a rate-limiting determinant for tumour progression. Semin. Cell Dev. Biol., 21, 19–25. PubMed PMC
Mulero-Navarro S., et al. (2005) Immortalized mouse mammary fibroblasts lacking dioxin receptor have impaired tumorigenicity in a subcutaneous mouse xenograft model. J. Biol. Chem., 280, 28731–28741. PubMed
Santiago-Josefat B., et al. (2004) Overexpression of latent transforming growth factor-beta binding protein 1 (LTBP-1) in dioxin receptor-null mouse embryo fibroblasts. J. Cell Sci., 117, 849–859. PubMed
Shimizu T., et al. (2005) Suppression of matrix metalloproteinase production in nasal fibroblasts by tranilast, an antiallergic agent, in vitro . Mediators Inflamm., 2005, 150–159. PubMed PMC
Yashiro M., et al. (2003) Tranilast (N-3,4-dimethoxycinamoyl anthranilic acid): a novel inhibitor of invasion-stimulating interaction between gastric cancer cells and orthotopic fibroblasts. Anticancer Res., 23, 3899–3904. PubMed
Montesano R., et al. (2001) Environmental causes of human cancers. Eur. J. Cancer, 37(suppl. 8), S67–S87. PubMed
Fontham E.T., et al. ; ACS Cancer and the Environment Subcommittee. (2009) American Cancer Society perspectives on environmental factors and cancer. CA Cancer J. Clin., 59, 343–351. PubMed
Hanahan D., et al. (2011) Hallmarks of cancer: the next generation. Cell, 144, 646–674. PubMed
Patan S. (2004) Vasculogenesis and angiogenesis. Cancer Treat. Res., 117, 3–32. PubMed
Seaman S., et al. (2007) Genes that distinguish physiological and pathological angiogenesis. Cancer Cell, 11, 539–554. PubMed PMC
Chinoy M.R., et al. (2002) Angiopoietin-1 and VEGF in vascular development and angiogenesis in hypoplastic lungs. Am. J. Physiol. Lung Cell. Mol. Physiol., 283, L60–L66. PubMed
Noden D.M. (1989) Embryonic origins and assembly of blood vessels. Am. Rev. Respir. Dis., 140, 1097–1103. PubMed
Jarzynka M.J., et al. (2006) Estradiol and nicotine exposure enhances A549 bronchioloalveolar carcinoma xenograft growth in mice through the stimulation of angiogenesis. Int. J. Oncol., 28, 337–344. PubMed PMC
Takahama M., et al. (1999) Expression of vascular endothelial growth factor and its receptors during lung carcinogenesis by N-nitrosobis(2-hydroxypropyl)amine in rats. Mol. Carcinog., 24, 287–293. PubMed
Duffield J.S., et al. (2013) Host responses in tissue repair and fibrosis. Annu. Rev. Pathol., 8, 241–276. PubMed PMC
Hernandez-Gea V., et al. (2013) Role of the microenvironment in the pathogenesis and treatment of hepatocellular carcinoma. Gastroenterology, 144, 512–527. PubMed PMC
Houghton A.M. (2013) Mechanistic links between COPD and lung cancer. Nat. Rev. Cancer, 13, 233–245. PubMed
Sueblinvong V., et al. (2012) Predisposition for disrepair in the aged lung. Am. J. Med. Sci., 344, 41–51. PubMed PMC
Atkinson M.J. (2013) Radiation treatment effects on the proteome of the tumour microenvironment. Adv. Exp. Med. Biol., 990, 49–60. PubMed
Jensen K., et al. (2012) General mechanisms of nicotine-induced fibrogenesis. FASEB J., 26, 4778–4787. PubMed PMC
Koval M., et al. (2010) Extracellular matrix influences alveolar epithelial claudin expression and barrier function. Am. J. Respir. Cell Mol. Biol., 42, 172–180. PubMed PMC
Bollyky P.L., et al. (2012) The role of hyaluronan and the extracellular matrix in islet inflammation and immune regulation. Curr. Diab. Rep., 12, 471–480. PubMed PMC
Rosin F.C., et al. (2011) Identification of myeloid-derived suppressor cells and T regulatory cells in lung microenvironment after Urethane-induced lung tumor. Int. Immunopharmacol., 11, 873–878. PubMed
Guo H., et al. (2010) Bisphenol A in combination with TNF-alpha selectively induces Th2 cell-promoting dendritic cells in vitro with an estrogen-like activity. Cell. Mol. Immunol., 7, 227–234. PubMed PMC
Yamashita U., et al. (2005) Effect of endocrine disrupters on macrophage functions in vitro . J. UOEH, 27, 1–10. PubMed
Spinardi-Barbisan A.L., et al. (2004) Infiltrating CD8+ T lymphocytes, natural killer cells, and expression of IL-10 and TGF-beta1 in chemically induced neoplasms in male Wistar rats. Toxicol. Pathol., 32, 548–557. PubMed
Hurt K., et al. (2013) Tributyltin and dibutyltin alter secretion of tumor necrosis factor alpha from human natural killer cells and a mixture of T cells and natural killer cells. J. Appl. Toxicol., 33, 503–510. PubMed PMC
Dudimah F.D., et al. (2007) Effect of tributyltin (TBT) on ATP levels in human natural killer (NK) cells: relationship to TBT-induced decreases in NK function. J. Appl. Toxicol., 27, 86–94. PubMed
Goud S.N., et al. (1999) Inhibition of natural killer cell activity in mice treated with tobacco specific carcinogen NNK. J. Toxicol. Environ. Health A, 56, 131–144. PubMed
Hindley J.P., et al. (2011) Analysis of the T-cell receptor repertoires of tumor-infiltrating conventional and regulatory T cells reveals no evidence for conversion in carcinogen-induced tumors. Cancer Res., 71, 736–746. PubMed PMC
Granville C.A., et al. (2009) A central role for Foxp3+ regulatory T cells in K-Ras-driven lung tumorigenesis. PLoS One, 4, e5061. PubMed PMC
Hoser G., et al. (2003) Lymphocyte subsets differences in smokers and nonsmokers with primary lung cancer: a flow cytometry analysis of bronchoalveolar lavage fluid cells. Med. Sci. Monit., 9, BR310–BR315. PubMed
Mann K.K., et al. (2001) The role of NF-kappaB as a survival factor in environmental chemical-induced pre-B cell apoptosis. Mol. Pharmacol., 59, 302–309. PubMed
Benigni R. (2012) Alternatives to the carcinogenicity bioassay for toxicity prediction: are we there yet? Expert Opin. Drug Metab. Toxicol., 8, 407–417. PubMed
Alavanja M.C., et al. (2013) Increased cancer burden among pesticide applicators and others due to pesticide exposure. CA Cancer J. Clin., 63, 120–142. PubMed
Gallo F., et al. (1993) The immune system response during development and progression of carcinogen-induced rat mammary tumors: prevention of tumor growth and restoration of immune system responsiveness by thymopentin. Breast Cancer Res. Treat., 27, 221–237. PubMed
Sengupta M., et al. (2002) Effect of lead and arsenic on murine macrophage response. Drug Chem. Toxicol., 25, 459–472. PubMed
Jerrett M., et al. (2009) Long-term ozone exposure and mortality. N. Engl. J. Med., 360, 1085–1095. PubMed PMC
Zelikoff J.T., et al. (1991) Immunomodulating effects of ozone on macrophage functions important for tumor surveillance and host defense. J. Toxicol. Environ. Health, 34, 449–467. PubMed
Hecker E. (1981) Cocarcinogenesis and tumor promoters of the diterpene ester type as possible carcinogenic risk factors. J. Cancer Res. Clin. Oncol., 99, 103–124. PubMed
Keller R., et al. (1982) Tumor-promoting diterpene esters prevent macrophage activation and suppress macrophage tumoricidal capacity. Exp. Cell Biol., 50, 121–134. PubMed
Rioux N., et al. (2000) The induction of cyclooxygenase-1 by a tobacco carcinogen in U937 human macrophages is correlated to the activation of NF-kappaB. Carcinogenesis, 21, 1745–1751. PubMed
Therriault M.J., et al. (2003) Immunomodulatory effects of the tobacco-specific carcinogen, NNK, on alveolar macrophages. Clin. Exp. Immunol., 132, 232–238. PubMed PMC
Massa T., et al. (1990) A host-mediated in vivo/in vitro assay with peritoneal murine macrophages for the detection of carcinogenic chemicals. J. Cancer Res. Clin. Oncol., 116, 357–364. PubMed
Lorimore S.A., et al. (2001) Inflammatory-type responses after exposure to ionizing radiation in vivo: a mechanism for radiation-induced bystander effects? Oncogene, 20, 7085–7095. PubMed
Kamp D.W., et al. (1999) The molecular basis of asbestos induced lung injury. Thorax, 54, 638–652. PubMed PMC
Broaddus V.C. (2001) Apoptosis and asbestos-induced disease: is there a connection? J. Lab. Clin. Med., 137, 314–315. PubMed
Kamp D.W., et al. (2002) Asbestos-induced alveolar epithelial cell apoptosis: role of mitochondrial dysfunction caused by iron-derived free radicals. Mol. Cell. Biochem., 234–235, 153–160. PubMed
Knaapen A.M., et al. (2005) Nitrite enhances neutrophil-induced DNA strand breakage in pulmonary epithelial cells by inhibition of myeloperoxidase. Carcinogenesis, 26, 1642–1648. PubMed
Malejka-Giganti D., et al. (1994) Peroxidative metabolism of carcinogenic N-arylhydroxamic acids: implications for tumorigenesis. Environ. Health Perspect., 102(suppl. 6), 75–81. PubMed PMC
Malejka-Giganti D., et al. (1993) Metabolism of the carcinogen N-hydroxy-N-2-fluorenylacetamide by rat peritoneal neutrophils. Carcinogenesis, 14, 341–346. PubMed
Vikis H.G., et al. (2012) Neutrophils are required for 3-methylcholanthrene-initiated, butylated hydroxytoluene-promoted lung carcinogenesis. Mol. Carcinog., 51, 993–1002. PubMed PMC
Hernández L.G., et al. (2009) Mechanisms of non-genotoxic carcinogens and importance of a weight of evidence approach. Mutat. Res., 682, 94–109. PubMed
Ben-Jonathan N., et al. (2009) Effects of bisphenol A on adipokine release from human adipose tissue: implications for the metabolic syndrome. Mol. Cell. Endocrinol., 304, 49–54. PubMed PMC
Coccini T., et al. (2013) Pulmonary toxicity of instilled cadmium-doped silica nanoparticles during acute and subacute stages in rats. Histol. Histopathol., 28, 195–209. PubMed
Geens T., et al. (2012) Distribution of bisphenol-A, triclosan and n-nonylphenol in human adipose tissue, liver and brain. Chemosphere, 87, 796–802. PubMed
Khan J.A., et al. (2011) Magnetite (Fe3O4) nanocrystals affect the expression of genes involved in the TGF-beta signalling pathway. Mol. Biosyst., 7, 1481–1486. PubMed
Ma J.Y., et al. (2012) Induction of pulmonary fibrosis by cerium oxide nanoparticles. Toxicol. Appl. Pharmacol., 262, 255–264. PubMed PMC
Chapellier M., et al. (2015) Disequilibrium of BMP2 levels in the breast stem cell niche launches epithelial transformation by overamplifying BMPR1B cell response. Stem Cell Reports, 4, 239–254. PubMed PMC
McHale C.M., et al. (2011) Current understanding of the mechanism of benzene-induced leukemia in humans: implications for risk assessment. Carcinogenesis, 33, 240–252. PubMed PMC
Maguer-Satta V. (2011) The stem cell niche: the black master of cancer. In Shostak, S (ed.) InTech. Cancer Stem Cells Theories and Practices.
Zolghadr F., et al. (2012) How benzene and its metabolites affect human marrow derived mesenchymal stem cells. Toxicol. Lett., 214, 145–153. PubMed
Chamorro-García R., et al. (2012) Bisphenol A diglycidyl ether induces adipogenic differentiation of multipotent stromal stem cells through a peroxisome proliferator-activated receptor gamma-independent mechanism. Environ. Health Perspect., 120, 984–989. PubMed PMC
Vigneri P., et al. (2009) Diabetes and cancer. Endocr. Relat. Cancer, 16, 1103–1123. PubMed
Kasper J.S., et al. (2006) A meta-analysis of diabetes mellitus and the risk of prostate cancer. Cancer Epidemiol. Biomarkers Prev., 15, 2056–2062. PubMed
Pollak M. (2008) Insulin and insulin-like growth factor signalling in neoplasia. Nat. Rev. Cancer, 8, 915–928. PubMed
Pollak M. (2012) The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat. Rev. Cancer, 12, 159–169. PubMed
Samani A.A., et al. (2007) The role of the IGF system in cancer growth and metastasis: overview and recent insights. Endocr. Rev., 28, 20–47. PubMed
Nathan D.M., et al. ; American Diabetes Association; European Association for Study of Diabetes (2009) Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care, 32, 193–203. PubMed PMC
Viollet B., et al. (2012) Cellular and molecular mechanisms of metformin: an overview. Clin. Sci. (Lond)., 122, 253–270. PubMed PMC
Evans J.M., et al. (2005) Metformin and reduced risk of cancer in diabetic patients. BMJ, 330, 1304–1305. PubMed PMC
Anisimov V.N., et al. (2005) Effect of metformin on life span and on the development of spontaneous mammary tumors in HER-2/neu transgenic mice. Exp. Gerontol., 40, 685–693. PubMed
Huang X., et al. (2008) Important role of the LKB1-AMPK pathway in suppressing tumorigenesis in PTEN-deficient mice. Biochem. J., 412, 211–221. PubMed
Tomimoto A., et al. (2008) Metformin suppresses intestinal polyp growth in ApcMin/+ mice. Cancer Sci., 99, 2136–2141. PubMed PMC
He G., et al. (2006) Thiazolidinediones inhibit insulin-like growth factor-i-induced activation of p70S6 kinase and suppress insulin-like growth factor-I tumor-promoting activity. Cancer Res., 66, 1873–1878. PubMed
Xiang X., et al. (2004) AMP-activated protein kinase activators can inhibit the growth of prostate cancer cells by multiple mechanisms. Biochem. Biophys. Res. Commun., 321, 161–167. PubMed
Grisouard J., et al. (2011) Targeting AMP-activated protein kinase in adipocytes to modulate obesity-related adipokine production associated with insulin resistance and breast cancer cell proliferation. Diabetol. Metab. Syndr., 3, 16. PubMed PMC
Salminen A., et al. (2011) AMP-activated protein kinase inhibits NF-κB signaling and inflammation: impact on healthspan and lifespan. J. Mol. Med. (Berl)., 89, 667–676. PubMed PMC
Xavier D.O., et al. (2010) Metformin inhibits inflammatory angiogenesis in a murine sponge model. Biomed. Pharmacother., 64, 220–225. PubMed
Ersoy C., et al. (2008) The effect of metformin treatment on VEGF and PAI-1 levels in obese type 2 diabetic patients. Diabetes Res. Clin. Pract., 81, 56–60. PubMed
Vazquez-Martin A., et al. (2009) The antidiabetic drug metformin suppresses HER2 (erbB-2) oncoprotein overexpression via inhibition of the mTOR effector p70S6K1 in human breast carcinoma cells. Cell Cycle, 8, 88–96. PubMed
Zhuang Y., et al. (2008) Cell cycle arrest in Metformin treated breast cancer cells involves activation of AMPK, downregulation of cyclin D1, and requires p27Kip1 or p21Cip1. J. Mol. Signal., 3, 18. PubMed PMC
Isakovic A., et al. (2007) Dual antiglioma action of metformin: cell cycle arrest and mitochondria-dependent apoptosis. Cell. Mol. Life Sci., 64, 1290–1302. PubMed PMC
Liu B., et al. (2009) Metformin induces unique biological and molecular responses in triple negative breast cancer cells. Cell Cycle, 8, 2031–2040. PubMed
Ben Sahra I., et al. (2010) Targeting cancer cell metabolism: the combination of metformin and 2-deoxyglucose induces p53-dependent apoptosis in prostate cancer cells. Cancer Res., 70, 2465–2475. PubMed
Jones R.G., et al. (2005) AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. Mol. Cell, 18, 283–293. PubMed
Buzzai M., et al. (2007) Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Cancer Res., 67, 6745–6752. PubMed
Algire C., et al. (2011) Diet and tumor LKB1 expression interact to determine sensitivity to anti-neoplastic effects of metformin in vivo . Oncogene, 30, 1174–1182. PubMed
Ashinuma H., et al. (2012) Antiproliferative action of metformin in human lung cancer cell lines. Oncol. Rep., 28, 8–14. PubMed
Rizos C.V., et al. (2013) Metformin and cancer. Eur. J. Pharmacol., 705, 96–108. PubMed
Lea M.A., et al. (2011) Addition of 2-deoxyglucose enhances growth inhibition but reverses acidification in colon cancer cells treated with phenformin. Anticancer Res., 31, 421–426. PubMed
Appleyard M.V., et al. (2012) Phenformin as prophylaxis and therapy in breast cancer xenografts. Br. J. Cancer, 106, 1117–1122. PubMed PMC
Anisimov V.N., et al. (2013) The key role of growth hormone-insulin-IGF-1 signaling in aging and cancer. Crit. Rev. Oncol. Hematol., 87, 201–223. PubMed PMC
Hattori Y., et al. (2006) Metformin inhibits cytokine-induced nuclear factor kappaB activation via AMP-activated protein kinase activation in vascular endothelial cells. Hypertension, 47, 1183–1188. PubMed
Arai M., et al. (2010) Metformin, an antidiabetic agent, suppresses the production of tumor necrosis factor and tissue factor by inhibiting early growth response factor-1 expression in human monocytes in vitro . J. Pharmacol. Exp. Ther., 334, 206–213. PubMed
Pearce E.L., et al. (2009) Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature, 460, 103–107. PubMed PMC
Williams T., et al. (2011) AMP-activated protein kinase (AMPK) activity is not required for neuronal development but regulates axogenesis during metabolic stress. Proc. Natl Acad. Sci. USA, 108, 5849–5854. PubMed PMC
Hwang Y.P., et al. (2010) Metformin blocks migration and invasion of tumour cells by inhibition of matrix metalloproteinase-9 activation through a calcium and protein kinase Calpha-dependent pathway: phorbol-12-myristate-13-acetate-induced/extracellular signal-regulated kinase/activator protein-1. Br. J. Pharmacol., 160, 1195–1211. PubMed PMC
Tan B.K., et al. (2011) Metformin treatment exerts antiinvasive and antimetastatic effects in human endometrial carcinoma cells. J. Clin. Endocrinol. Metab., 96, 808–816. PubMed
Hirsch H.A., et al. (2013) Metformin inhibits the inflammatory response associated with cellular transformation and cancer stem cell growth. Proc. Natl Acad. Sci. USA, 110, 972–977. PubMed PMC
Jorgensen L.N., et al. (1992) Carcinogenic effect of the human insulin analogue B10Asp in female rats. Diabetologia, 35, A3.
Berger M. (2000) Safety of insulin glargine. Lancet, 356, 2013–2014. PubMed
Kurtzhals P., et al. (2000) Correlations of receptor binding and metabolic and mitogenic potencies of insulin analogs designed for clinical use. Diabetes, 49, 999–1005. PubMed
Currie C.J., et al. (2009) The influence of glucose-lowering therapies on cancer risk in type 2 diabetes. Diabetologia, 52, 1766–1777. PubMed
Hemkens L.G., et al. (2009) Risk of malignancies in patients with diabetes treated with human insulin or insulin analogues: a cohort study. Diabetologia, 52, 1732–1744. PubMed PMC
Jonasson J.M., et al. (2009) Insulin glargine use and short-term incidence of malignancies-a population-based follow-up study in Sweden. Diabetologia, 52, 1745–1754. PubMed
Colhoun H.M; SDRN Epidemiology Group (2009) Use of insulin glargine and cancer incidence in Scotland: a study from the Scottish Diabetes Research Network Epidemiology Group. Diabetologia, 52, 1755–1765. PubMed PMC
Taubes G. (2012) Cancer research. Cancer prevention with a diabetes pill? Science, 335, 29. PubMed
Ronckers C.M., et al. (2005) Radiation and breast cancer: a review of current evidence. Breast Cancer Res., 7, 21–32. PubMed PMC
Shore R.E., et al. (1993) Thyroid cancer among persons given X-ray treatment in infancy for an enlarged thymus gland. Am. J. Epidemiol., 137, 1068–1080. PubMed
Shore R.E., et al. (1985) Thyroid tumors following thymus irradiation. J. Natl Cancer Inst., 74, 1177–1184. PubMed
Preston D.L., et al. (2007) Solid cancer incidence in atomic bomb survivors: 1958–1998. Radiat. Res., 168, 1–64. PubMed
Terzaghi M., et al. (1976) X-radiation-induced transformation in a C3H mouse embryo-derived cell line. Cancer Res., 36, 1367–1374. PubMed
Terzaghi M., et al. (1979) Dynamics of neoplastic development in carcinogen-exposed tracheal mucosa. Cancer Res., 39, 4003–4010. PubMed
Kennedy A.R., et al. (1980) Relationship between x-ray exposure and malignant transformation in C3H 10T1/2 cells. Proc. Natl Acad. Sci. USA, 77, 7262–7266. PubMed PMC
Engelmann I., et al. (2000) Ex vivo tumor cell lines are resistant to intercellular induction of apoptosis and independent of exogenous survival factors. Anticancer Res., 20, 2361–2370. PubMed
Terzaghi-Howe M. (1987) Inhibition of carcinogen-altered rat tracheal epithelial cell proliferation by normal epithelial cells in vivo . Carcinogenesis, 8, 145–150. PubMed
Häufel T., et al. (1999) Three distinct roles for TGF-beta during intercellular induction of apoptosis: a review. Anticancer Res., 19, 105–111. PubMed
Portess D.I., et al. (2007) Low-dose irradiation of nontransformed cells stimulates the selective removal of precancerous cells via intercellular induction of apoptosis. Cancer Res., 67, 1246–1253. PubMed
KAPLAN H.S., et al. (1956) Indirect induction of lymphomas in irradiated mice. I. Tumor incidence and morphology in mice bearing nonirradiated thymic grafts. Cancer Res., 16, 422–425. PubMed
Morgan J.E., et al. (2002) Myogenic cell proliferation and generation of a reversible tumorigenic phenotype are triggered by preirradiation of the recipient site. J. Cell Biol., 157, 693–702. PubMed PMC
Ohuchida K., et al. (2004) Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res., 64, 3215–3222. PubMed
Nguyen D.H., et al. (2011) Radiation acts on the microenvironment to affect breast carcinogenesis by distinct mechanisms that decrease cancer latency and affect tumor type. Cancer Cell, 19, 640–651. PubMed PMC
Gonda T.A., et al. (2009) Chronic inflammation, the tumor microenvironment and carcinogenesis. Cell Cycle, 8, 2005–2013. PubMed
Claisse D. (1989) Chemical contamination of French coasts: the results of a ten years mussel watch. Marine Pollution Bull., 20, 523–528.
O’Connor T.P., et al. (2006) Trends in chemical concentrations in mussels and oysters collected along the US coast: update to 2003. Mar. Environ. Res., 62, 261–285. PubMed
Krishnakumar P.K., et al. (1994) Effects of environmental contaminants on the health of Mytilus edulis from Puget sound, Washington, USA. I. Cytochemical measures of lysosomal responses in the digestive cells using automatic image analysis. Mar. Ecol. Prog. Ser., 106, 249–261.
Ramu K., et al. (2007) Asian Mussel Watch Program: contamination status of polybrominated diphenyl ethers and organochlorines in coastal waters of Asian countries. Environ. Sci. Technol., 41, 4580–4586. PubMed
Melwani A.R., et al. (2013) Mussel watch update: long-term trends in selected contaminants from coastal California, 1977–2010. Mar. Pollut. Bull, 81, 291–302. PubMed
Bushkin-Bedient S., et al. (2010) Benefits versus risks associated with consumption of fish and other seafood. Rev. Environ. Health, 25, 161–191. PubMed
Smith A.G., et al. (2002) Organochlorine chemicals in seafood: occurrence and health concerns. Food Chem. Toxicol., 40, 767–779. PubMed
Landers D.H., et al. (2010) The Western Airborne Contaminant Assessment Project (WACAP): an interdisciplinary evaluation of the impacts of airborne contaminants in western U.S. National Parks. Environ. Sci. Technol., 44, 855–859. PubMed
Thomas G.O., et al. (2006) Organohalogen chemicals in human blood from the United Kingdom. Environ. Pollut., 141, 30–41. PubMed
Qin Y.Y., et al. (2011) Halogenated POPs and PAHs in blood plasma of Hong Kong residents. Environ. Sci. Technol., 45, 1630–1637. PubMed
Huss H.H., et al. (2003) Assessment and Management of Seafood Safety and Quality. FAO Fisheries Technical Paper. No. 444. FAO, Rome, 230 p.
EC (2008) Regulation (EC) No 629/2008 of the European parliament and the council of 2 July 2008 amending Regulation (EC) No 1881/2006 setting maximum levels for certain contaminants in foodstuffs. OJEC, L173/176–179.
FDA US (2011) Fish and Fishery Products Hazards and Controls Guidance. Food and Drug Administration, Center for Food Safety and Applied Nutrition, U.S. Department of Health and Human Services; http://www.fda.gov/FoodGuidances (3 December 2015, date last accessed).
Lafferty K.D., et al. (2004) Are diseases increasing in the ocean?*. Annu. Rev. Ecol. Evol. Syst., 35, 31–54.
Morley N.J. (2010) Interactive effects of infectious diseases and pollution in aquatic molluscs. Aquat. Toxicol., 96, 27–36. PubMed
Bossart G.D. (2011) Marine mammals as sentinel species for oceans and human health. Vet. Pathol., 48, 676–690. PubMed
Stentiford G., et al. (2009) Site-specific disease profiles in fish and their use in environmental monitoring. Mar. Ecol. Progr. Ser., 381, 1–15.
Myers M.S., et al. (1998) Toxicopathic hepatic lesions in subadult English sole (pleuronectes vetuls) from Puget Sound, Washington, USA: relationships with other biomarkers of contaminant exposure. Mar. Environ. Res., 45, 47–67.
Reichert W.L., et al. (1998) Molecular epizootiology of genotoxic events in marine fish: linking contaminant exposure, DNA damage, and tissue-level alterations. Mutat. Res., 411, 215–225. PubMed
Elston R.A., et al. (1992) Disseminated neoplasia of bivalve molluscs. Rev. Aquat. Sci., 6, 405–466.
Krishnakumar P.K., et al. (1999) Environmental contaminants and the prevalence of hemic neoplasia (leukemia) in the common mussel (Mytilus edulis complex) from Puget Sound, Washington, U.S.A. J. Invertebr. Pathol., 73, 135–146. PubMed
Ruiz P., et al. (2013) Biomarkers and transcription levels of cancer-related genes in cockles Cerastoderma edule from Galicia (NW Spain) with disseminated neoplasia. Aquat. Toxicol., 136–137, 101–111. PubMed
Di Y., et al. (2011) Tissue-specific expression of p53 and ras genes in response to the environmental genotoxicant benzo(α)pyrene in marine mussels. Environ. Sci. Technol., 45, 8974–8981. PubMed
Walker C.W., et al. (2011) p53 Superfamily proteins in marine bivalve cancer and stress biology. Adv. Mar. Biol., 59, 1–36. PubMed
Mössner S., et al. (1997) Marine mammals as global pollution indicators for organochlorines. Chemosphere, 34, 1285–1296. PubMed
Pierce G.J., et al. (2008) Bioaccumulation of persistent organic pollutants in female common dolphins (Delphinus delphis) and harbour porpoises (Phocoena phocoena) from western European seas: geographical trends, causal factors and effects on reproduction and mortality. Environ. Pollut., 153, 401–415. PubMed
Newman S.J., et al. (2006) Marine mammal neoplasia: a review. Vet. Pathol., 43, 865–880. PubMed
Guido C., et al. (2012) Mitochondrial fission induces glycolytic reprogramming in cancer-associated myofibroblasts, driving stromal lactate production, and early tumor growth. Oncotarget, 3, 798–810. PubMed PMC
Ralph S.J., et al. (2010) The causes of cancer revisited: “mitochondrial malignancy” and ROS-induced oncogenic transformation - why mitochondria are targets for cancer therapy. Mol. Aspects Med., 31, 145–170. PubMed
Ushio-Fukai M., et al. (2008) Reactive oxygen species and angiogenesis: NADPH oxidase as target for cancer therapy. Cancer Lett., 266, 37–52. PubMed PMC
Simon H.U., et al. (2000) Role of reactive oxygen species (ROS) in apoptosis induction. Apoptosis, 5, 415–418. PubMed
Zhou F., et al. (2013) Novel roles of reactive oxygen species in the pathogenesis of acute myeloid leukemia. J. Leukoc. Biol., 94, 423–429. PubMed PMC
Chang X., et al. (2013) Paraquat inhibits cell viability via enhanced oxidative stress and apoptosis in human neural progenitor cells. Chem. Biol. Interact., 206, 248–255. PubMed
Radisky D., et al. (2005) Rac1b and reactive oxygen species mediate MMP-3-induced EMT and genomic instability. Nature, 436, 123–127. PubMed PMC
Durando M., et al. (2011) Prenatal exposure to bisphenol A promotes angiogenesis and alters steroid-mediated responses in the mammary glands of cycling rats. J. Steroid Biochem. Mol. Biol., 127, 35–43. PubMed
Zhu M., et al. (1997) Expression of exogenous NF-IL6 induces apoptosis in Sp2/0-Ag14 myeloma cells. DNA Cell Biol., 16, 127–135. PubMed
Ye F., et al. (2010) Alterations of dendritic cell subsets in the peripheral circulation of patients with cervical carcinoma. J. Exp. Clin. Cancer Res., 29, 78. PubMed PMC
Yamagiwa Y., et al. (2006) Interleukin-6 decreases senescence and increases telomerase activity in malignant human cholangiocytes. Life Sci., 78, 2494–2502. PubMed PMC
Kelso A., et al. (1992) Survival of the myeloid progenitor cell line FDC-P1 is prolonged by interferon-gamma or interleukin-4. Growth Factors, 6, 233–242. PubMed
Touboul C., et al. (2013) Mesenchymal stem cells enhance ovarian cancer cell infiltration through IL6 secretion in an amniochorionic membrane based 3D model. J. Transl. Med., 11, 28. PubMed PMC
Na Y.R., et al. (2013) Interleukin-6-induced Twist and N-cadherin enhance melanoma cell metastasis. Melanoma Res., 23, 434–443. PubMed
Grivennikov S.I., et al. (2011) Inflammatory cytokines in cancer: tumour necrosis factor and interleukin 6 take the stage. Ann. Rheum. Dis., 70, 104–108. PubMed
Textor S., et al. (2011) Human NK cells are alerted to induction of p53 in cancer cells by upregulation of the NKG2D ligands ULBP1 and ULBP2. Cancer Res., 71, 5998–6009. PubMed
Yao L., et al. (1999) Contribution of natural killer cells to inhibition of angiogenesis by interleukin-12. Blood, 93, 1612–1621. PubMed
Langers I., et al. (2012) Natural killer cells: role in local tumor growth and metastasis. Biologics, 6, 73–82. PubMed PMC
Reiners K.S., et al. (2013) Soluble ligands for NK cell receptors promote evasion of chronic lymphocytic leukemia cells from NK cell anti-tumor activity. Blood, 121, 3658–3665. PubMed PMC
Abarca-Rojano E., et al. (2009) Re-organization of mitochondria at the NK cell immune synapse. Immunol. Lett., 122, 18–25. PubMed
Sceneay J., et al. (2012) Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res., 72, 3906–3911. PubMed
Wu J., et al. (2003) Natural killer cells and cancer. Adv. Cancer Res., 90, 127–156. PubMed
Fiaschi T., et al. (2012) Oxidative stress, tumor microenvironment, and metabolic reprogramming: a diabolic liaison. Int. J. Cell Biol., 2012, 762825. PubMed PMC
Taguchi K., et al. (2011) Molecular mechanisms of the Keap1–Nrf2 pathway in stress response and cancer evolution. Genes Cells, 16, 123–140. PubMed
Reuter S., et al. (2010) Oxidative stress, inflammation, and cancer: how are they linked? Free Radic. Biol. Med., 49, 1603–1616. PubMed PMC
Aitken R.J., et al. (2001) Oxidative stress, DNA damage and the Y chromosome. Reproduction, 122, 497–506. PubMed
Carvour M., et al. (2008) Chronic low-dose oxidative stress induces caspase-3-dependent PKCdelta proteolytic activation and apoptosis in a cell culture model of dopaminergic neurodegeneration. Ann. N. Y. Acad. Sci., 1139, 197–205. PubMed PMC
Conti A., et al. (2010) Role of inflammation and oxidative stress mediators in gliomas. Cancers (Basel), 2, 693–712. PubMed PMC
Lorenz O., et al. (2009) Proteomics reveals acute pro-inflammatory and protective responses in rat Kupffer cells and hepatocytes after chemical initiation of liver cancer and after LPS and IL-6. Proteomics. Clin. Appl., 3, 947–967. PubMed
Tertil M., et al. (2010) Oxidative stress in tumor angiogenesis- therapeutic targets. Curr. Pharm. Des., 16, 3877–3894. PubMed
Butler L.M., et al. (2011) Kaposi’s sarcoma-associated herpesvirus infection of endothelial cells inhibits neutrophil recruitment through an interleukin-6-dependent mechanism: a new paradigm for viral immune evasion. J. Virol., 85, 7321–7332. PubMed PMC
Rodier F., et al. (2009) Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nat. Cell Biol., 11, 973–979. PubMed PMC
Bhattacharjee R.N., et al. (2010) Telomere-mediated chromosomal instability triggers TLR4 induced inflammation and death in mice. PLoS One, 5, e11873. PubMed PMC
Li J., et al. (2004) Nickel compounds act through phosphatidylinositol-3-kinase/Akt-dependent, p70(S6k)-independent pathway to induce hypoxia inducible factor transactivation and Cap43 expression in mouse epidermal Cl41 cells. Cancer Res., 64, 94–101. PubMed
Lu H., et al. (2005) Carcinogenic effect of nickel compounds. Mol. Cell. Biochem., 279, 45–67. PubMed
Ding J., et al. (2006) Nickel compounds render anti-apoptotic effect to human bronchial epithelial Beas-2B cells by induction of cyclooxygenase-2 through an IKKbeta/p65-dependent and IKKalpha- and p50-independent pathway. J. Biol. Chem., 281, 39022–39032. PubMed
Freitas M., et al. (2013) Nickel induces apoptosis in human neutrophils. Biometals, 26, 13–21. PubMed
Reed J.E., et al. (2006) Stabilization of G-quadruplex DNA and inhibition of telomerase activity by square-planar nickel(II) complexes. J. Am. Chem. Soc., 128, 5992–5993. PubMed
Campbell N.H., et al. (2012) Molecular basis of structure-activity relationships between salphen metal complexes and human telomeric DNA quadruplexes. J. Med. Chem., 55, 209–222. PubMed
Zhang J., et al. (2013) The alteration of miR-222 and its target genes in nickel-induced tumor. Biol. Trace Elem. Res., 152, 267–274. PubMed
Wu C.H., et al. (2012) Nickel-induced epithelial-mesenchymal transition by reactive oxygen species generation and E-cadherin promoter hypermethylation. J. Biol. Chem., 287, 25292–25302. PubMed PMC
Ding J., et al. (2009) TNF-alpha induction by nickel compounds is specific through ERKs/AP-1-dependent pathway in human bronchial epithelial cells. Curr. Cancer Drug Targets, 9, 81–90. PubMed
van Esterik J.C., et al. (2014) Programming of metabolic effects in C57BL/6JxFVB mice by exposure to bisphenol A during gestation and lactation. Toxicology, 321, 40–52. PubMed
Allard P., et al. (2010) Bisphenol A impairs the double-strand break repair machinery in the germline and causes chromosome abnormalities. Proc. Natl Acad. Sci. USA, 107, 20405–20410. PubMed PMC
Terasaka H., et al. (2005) Cytotoxicity and apoptosis-inducing activity of bisphenol A and hydroquinone in HL-60 cells. Anticancer Res., 25, 2241–2247. PubMed
Xu J., et al. (2002) Bisphenol A induces apoptosis and G2-to-M arrest of ovarian granulosa cells. Biochem. Biophys. Res. Commun., 292, 456–462. PubMed
Takahashi A., et al. (2004) Bisphenol A from dental polycarbonate crown upregulates the expression of hTERT. J. Biomed. Mater. Res. B. Appl. Biomater., 71, 214–221. PubMed
Pupo M., et al. (2012) Bisphenol A induces gene expression changes and proliferative effects through GPER in breast cancer cells and cancer-associated fibroblasts. Environ. Health Perspect., 120, 1177–1182. PubMed PMC
Hassan Z.K., et al. (2012) Bisphenol A induces hepatotoxicity through oxidative stress in rat model. Oxid. Med. Cell. Longev., 2012, 194829. PubMed PMC
Whalen M.M., et al. (2001) Butyltin exposure causes a rapid decrease in cyclic AMP levels in human lymphocytes. Toxicol. Appl. Pharmacol., 171, 141–148. PubMed
Person R.J., et al. (2010) Effects of butyltin exposures on MAP kinase-dependent transcription regulators in human natural killer cells. Toxicol. Mech. Methods, 20, 227–233. PubMed PMC
Sasaki Y.F., et al. (1993) Increasing effect of tri-n-butyltins and triphenyltins on the frequency of chemically induced chromosome aberrations in cultured Chinese hamster cells. Mutat. Res., 300, 5–14. PubMed
Nakano K., et al. (2004) Tributyltin (TBT) increases TNFα mRNA expression and induces apoptosis in the murine macrophage cell line in vitro . Environ. Health Prev. Med., 9, 266–271. PubMed PMC
Nakatsu Y., et al. (2007) Concentration dependence of the mechanisms of tributyltin-induced apoptosis. Toxicol. Sci., 97, 438–447. PubMed
Osman A.M., et al. (2012) Phosphoproteomic analysis of mouse thymoma cells treated with tributyltin oxide: TBTO affects proliferation and energy sensing pathways. Toxicol. Sci., 126, 84–100. PubMed
Glaser V., et al. (2010) Oxidative stress-mediated inhibition of brain creatine kinase activity by methylmercury. Neurotoxicology, 31, 454–460. PubMed
Ni M., et al. (2010) Methylmercury induces acute oxidative stress, altering Nrf2 protein level in primary microglial cells. Toxicol. Sci., 116, 590–603. PubMed PMC
Toyama T., et al. (2007) Cytoprotective role of Nrf2/Keap1 system in methylmercury toxicity. Biochem. Biophys. Res. Commun., 363, 645–650. PubMed
Sherwani S.I., et al. (2013) Eicosanoid signaling and vascular dysfunction: methylmercury-induced phospholipase D activation in vascular endothelial cells. Cell Biochem. Biophys., 67, 317–329. PubMed
Betti C., et al. (1992) Genotoxic activity of methyl mercury chloride and dimethyl mercury in human lymphocytes. Mutat. Res., 281, 255–260. PubMed
Ceccatelli S., et al. (2010) Methylmercury-induced neurotoxicity and apoptosis. Chem. Biol. Interact., 188, 301–308. PubMed
Sokolowski K., et al. (2011) Methylmercury (MeHg) elicits mitochondrial-dependent apoptosis in developing hippocampus and acts at low exposures. Neurotoxicology, 32, 535–544. PubMed PMC
Usuki F., et al. (2008) Methylmercury activates ASK1/JNK signaling pathways, leading to apoptosis due to both mitochondria- and endoplasmic reticulum (ER)-generated processes in myogenic cell lines. Neurotoxicology, 29, 22–30. PubMed
Kunimoto M. (1994) Methylmercury induces apoptosis of rat cerebellar neurons in primary culture. Biochem. Biophys. Res. Commun., 204, 310–317. PubMed
Sager P.R. (1988) Selectivity of methyl mercury effects on cytoskeleton and mitotic progression in cultured cells. Toxicol. Appl. Pharmacol., 94, 473–486. PubMed
Patel E., et al. (2013) Methylmercury impairs motor function in early development and induces oxidative stress in cerebellar granule cells. Toxicol. Lett., 222, 265–272. PubMed
Ilbäck N.G., et al. (1996) Effects of methyl mercury on cytokines, inflammation and virus clearance in a common infection (coxsackie B3 myocarditis). Toxicol. Lett., 89, 19–28. PubMed
Black A.T., et al. (2008) Increased oxidative stress and antioxidant expression in mouse keratinocytes following exposure to paraquat. Toxicol. Appl. Pharmacol., 231, 384–392. PubMed PMC
Ribas G., et al. (1997) Genotoxic evaluation of the herbicide paraquat in cultured human lymphocytes. Teratog. Carcinog. Mutagen., 17, 339–347. PubMed
Yang W., et al. (2008) Paraquat-induced apoptosis in human neuroblastoma SH-SY5Y cells: involvement of p53 and mitochondria. J. Toxicol. Environ. Health A, 71, 289–299. PubMed
Rio M.J., et al. (2008) Paraquat induces apoptosis in human lymphocytes: protective and rescue effects of glucose, cannabinoids and insulin-like growth factor-1. Growth Factors, 26, 49–60. PubMed
Paolillo N., et al. (2011) Effects of paraquat and capsaicin on the expression of genes related to inflammatory, immune responses and cell death in immortalized human HaCat keratinocytes. Int. J. Immunopathol. Pharmacol., 24, 861–868. PubMed
Sallmyr A., et al. (2008) Genomic instability in myeloid malignancies: increased reactive oxygen species (ROS), DNA double strand breaks (DSBs) and error-prone repair. Cancer Lett., 270, 1–9. PubMed
Passos J.F., et al. (2007) Mitochondrial dysfunction accounts for the stochastic heterogeneity in telomere-dependent senescence. PLoS Biol., 5, e110. PubMed PMC
Jin S., et al. (2013) Non-canonical Notch signaling activates IL-6/JAK/STAT signaling in breast tumor cells and is controlled by p53 and IKKα/IKKβ. Oncogene, 32, 4892–4902. PubMed PMC
Kurz D.J., et al. (2004) Chronic oxidative stress compromises telomere integrity and accelerates the onset of senescence in human endothelial cells. J. Cell Sci., 117, 2417–2426. PubMed
Take M., et al. (2014) Estimation of chloroform inhalation dose by other routes based on the relationship of area under the blood concentration-time curve (AUC)-inhalation dose to chloroform distribution in the blood of rats. J. Environ. Sci. Health A Tox. Hazard. Subst. Environ. Eng., 49, 253–261. PubMed
Matsumoto M., et al. (2013) Subchronic toxicity and carcinogenicity studies of 1,2-dichloropropane inhalation to mice. Inhal. Toxicol., 25, 435–443. PubMed
Kano H., et al. (2009) Carcinogenicity studies of 1,4-dioxane administered in drinking-water to rats and mice for 2 years. Food Chem. Toxicol., 47, 2776–2784. PubMed
Vandenberg L.N. (2014) Low-dose effects of hormones and endocrine disruptors. Vitam. Horm., 94, 129–165. PubMed
Vandenberg L.N. (2014) Non-monotonic dose responses in studies of endocrine disrupting chemicals: bisphenol a as a case study. Dose. Response., 12, 259–276. PubMed PMC
Conolly R.B., et al. (2004) Nonmonotonic dose-response relationships: mechanistic basis, kinetic modeling, and implications for risk assessment. Toxicol. Sci., 77, 151–157. PubMed
Vandenberg L.N., et al. (2012) Hormones and endocrine-disrupting chemicals: low-dose effects and nonmonotonic dose responses. Endocr. Rev., 33, 378–455. PubMed PMC
Welshons W.V., et al. (2003) Large effects from small exposures. I. Mechanisms for endocrine-disrupting chemicals with estrogenic activity. Environ. Health Perspect., 111, 994–1006. PubMed PMC
Kortenkamp A. (2008) Low dose mixture effects of endocrine disrupters: implications for risk assessment and epidemiology. Int. J. Androl., 31, 233–240. PubMed
Schug T.T., et al. (2013) Designing Endocrine Disruption Out of the Next Generation of Chemicals. Green Chem., 15, 181–198. PubMed PMC
Bisson W. (2012) Editorial: computational chemogenomics in drug design and discovery. Curr. Top. Med. Chem., 12, 1867–1868. PubMed
Gerlach C., et al. (2014) Mono-substituted isopropylated triaryl phosphate, a major component of Firemaster 550, is an AHR agonist that exhibits AHR-independent cardiotoxicity in zebrafish. Aquat. Toxicol., 154, 71–79. PubMed PMC
Benninghoff A.D., et al. (2011) Estrogen-like activity of perfluoroalkyl acids in vivo and interaction with human and rainbow trout estrogen receptors in vitro . Toxicol. Sci., 120, 42–58. PubMed PMC