|
1.Ferlay, J., Soerjomataram, I., Dikshit, R., Eser, S., Mathers, C., Rebelo, M., Parkin, D. M., Forman, D., and Bray, F. (2015) Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. International Journal of Cancer 136, E359-E386 2.El-Serag, H. B., and Rudolph, K. L. (2007) Hepatocellular carcinoma: epidemiology and molecular carcinogenesis. Gastroenterology 132, 2557-2576 3.Torre, L. A., Bray, F., Siegel, R. L., Ferlay, J., Lortet-Tieulent, J., and Jemal, A. (2015) Global cancer statistics, 2012. CA: A Cancer Journal for Clinicians 65, 87-108 4.Llovet, J. M., Burroughs, A., and Bruix, J. (2003) Hepatocellular carcinoma. The Lancet 362, 1907-1917 5.Park, J. W., Chen, M., Colombo, M., Roberts, L. R., Schwartz, M., Chen, P. J., Kudo, M., Johnson, P., Wagner, S., and Orsini, L. S. (2015) Global patterns of hepatocellular carcinoma management from diagnosis to death: the BRIDGE Study. Liver International 35, 2155-2166 6.Berasain, C., Castillo, J., Perugorria, M. J., Latasa, M. U., Prieto, J., and Avila, M. A. (2009) Inflammation and Liver Cancer. Annals of the New York Academy of Sciences 1155, 206-221 7.Marotta, F., Vangieri, B., Cecere, A., and Gattoni, A. (2004) The pathogenesis of hepatocellular carcinoma is multifactorial event. Novel immunological treatment in prospect. La Clinica terapeutica 155, 187-199 8.Mapara, M. Y., and Sykes, M. (2004) Tolerance and Cancer: Mechanisms of Tumor Evasion and Strategies for Breaking Tolerance. Journal of Clinical Oncology 22, 1136-1151 9.Zou, W. (2005) Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer 5, 263-274 10.Boon, T., and van der Bruggen, P. (1996) Human tumor antigens recognized by T lymphocytes. The Journal of Experimental Medicine 183, 725-729 11.Nagano, J., Shimizu, M., Hara, T., Shirakami, Y., Kochi, T., Nakamura, N., Ohtaki, H., Ito, H., Tanaka, T., Tsurumi, H., Saito, K., Seishima, M., and Moriwaki, H. (2013) Effects of Indoleamine 2,3-Dioxygenase Deficiency on High-Fat Diet-Induced Hepatic Inflammation. PLOS ONE 8, e73404 12.Ito, H., Hoshi, M., Ohtaki, H., Taguchi, A., Ando, K., Ishikawa, T., Osawa, Y., Hara, A., Moriwaki, H., Saito, K., and Seishima, M. (2010) Ability of IDO To Attenuate Liver Injury in α-Galactosylceramide–Induced Hepatitis Model. The Journal of Immunology 185, 4554-4560 13.Uyttenhove, C., Pilotte, L., Theate, I., Stroobant, V., Colau, D., Parmentier, N., Boon, T., and Van den Eynde, B. J. (2003) Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med 9, 1269-1274 14.Mellor, A. L., and Munn, D. H. (2004) IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol 4, 762-774 15.Takikawa, O. (2005) Biochemical and medical aspects of the indoleamine 2,3-dioxygenase-initiated l-tryptophan metabolism. Biochemical and Biophysical Research Communications 338, 12-19 16.Taylor, M. W., and Feng, G. (1991) Relationship between interferon-gamma, indoleamine 2, 3-dioxygenase, and tryptophan catabolism. The FASEB Journal 5, 2516-2522 17.Basu, G. D., Tinder, T. L., Bradley, J. M., Tu, T., Hattrup, C. L., Pockaj, B. A., and Mukherjee, P. (2006) Cyclooxygenase-2 inhibitor enhances the efficacy of a breast cancer vaccine: role of IDO. The Journal of Immunology 177, 2391-2402 18.Babcock, T. A., and Carlin, J. M. (2000) TRANSCRIPTIONAL ACTIVATION OF INDOLEAMINE DIOXYGENASE BY INTERLEUKIN 1 AND TUMOR NECROSIS FACTOR α IN INTERFERON-TREATED EPITHELIAL CELLS. Cytokine 12, 588-594 19.Robinson, C. M., Shirey, K. A., and Carlin, J. M. (2003) Synergistic transcriptional activation of indoleamine dioxygenase by IFN-γ and tumor necrosis factor-α. Journal of interferon & cytokine research 23, 413-421 20.Munn, D. H., Shafizadeh, E., Attwood, J. T., Bondarev, I., Pashine, A., and Mellor, A. L. (1999) Inhibition of T Cell Proliferation by Macrophage Tryptophan Catabolism. The Journal of Experimental Medicine 189, 1363-1372 21.Frumento, G., Rotondo, R., Tonetti, M., Damonte, G., Benatti, U., and Ferrara, G. B. (2002) Tryptophan-derived Catabolites Are Responsible for Inhibition of T and Natural Killer Cell Proliferation Induced by Indoleamine 2,3-Dioxygenase. The Journal of Experimental Medicine 196, 459-468 22.Nishikawa, H., Kato, T., Tawara, I., Takemitsu, T., Saito, K., Wang, L., Ikarashi, Y., Wakasugi, H., Nakayama, T., Taniguchi, M., Kuribayashi, K., Old, L. J., and Shiku, H. (2005) Accelerated chemically induced tumor development mediated by CD4+CD25+ regulatory T cells in wild-type hosts. Proceedings of the National Academy of Sciences of the United States of America 102, 9253-9257 23.Okamoto, A., Nikaido, T., Ochiai, K., Takakura, S., Saito, M., Aoki, Y., Ishii, N., Yanaihara, N., Yamada, K., Takikawa, O., Kawaguchi, R., Isonishi, S., Tanaka, T., and Urashima, M. (2005) Indoleamine 2,3-Dioxygenase Serves as a Marker of Poor Prognosis in Gene Expression Profiles of Serous Ovarian Cancer Cells. Clinical Cancer Research 11, 6030-6039 24.Brandacher, G., Perathoner, A., Ladurner, R., Schneeberger, S., Obrist, P., Winkler, C., Werner, E. R., Werner-Felmayer, G., Weiss, H. G., and Georg, G. (2006) Prognostic value of indoleamine 2, 3-dioxygenase expression in colorectal cancer: effect on tumor-infiltrating T cells. Clinical Cancer Research 12, 1144-1151 25.Fukuno, K., Hara, T., Tsurumi, H., Shibata, Y., Mabuchi, R., Nakamura, N., Kitagawa, J., Shimizu, M., Ito, H., Saito, K., and Moriwaki, H. (2015) Expression of indoleamine 2,3-dioxygenase in leukemic cells indicates an unfavorable prognosis in acute myeloid leukemia patients with intermediate-risk cytogenetics. Leukemia & Lymphoma 56, 1398-1405 26.Mabuchi, R., Hara, T., Matsumoto, T., Shibata, Y., Nakamura, N., Nakamura, H., Kitagawa, J., Kanemura, N., Goto, N., Shimizu, M., Ito, H., Yamamoto, Y., Saito, K., Moriwaki, H., and Tsurumi, H. (2016) High serum concentration of L-kynurenine predicts unfavorable outcomes in patients with acute myeloid leukemia. Leukemia & Lymphoma 57, 92-98 27.Yoshikawa, T., Hara, T., Tsurumi, H., Goto, N., Hoshi, M., Kitagawa, J., Kanemura, N., Kasahara, S., Ito, H., Takemura, M., Saito, K., Seishima, M., Takami, T., and Moriwaki, H. (2010) Serum concentration of L-kynurenine predicts the clinical outcome of patients with diffuse large B-cell lymphoma treated with R-CHOP. European Journal of Haematology 84, 304-309 28.Ninomiya, S., Hara, T., Tsurumi, H., Hoshi, M., Kanemura, N., Goto, N., Kasahara, S., Shimizu, M., Ito, H., Saito, K., Hirose, Y., Yamada, T., Takahashi, T., Seishima, M., Takami, T., and Moriwaki, H. (2011) Indoleamine 2,3-dioxygenase in tumor tissue indicates prognosis in patients with diffuse large B-cell lymphoma treated with R-CHOP. Annals of Hematology 90, 409-416 29.Ninomiya, S., Hara, T., Tsurumi, H., Goto, N., Saito, K., Seishima, M., Takami, T., and Moriwaki, H. (2012) Indoleamine 2, 3-dioxygenase expression and serum kynurenine concentrations in patients with diffuse large B-cell lymphoma. Leukemia & lymphoma 53, 1143-1145 30.Muller, A. J., DuHadaway, J. B., Donover, P. S., Sutanto-Ward, E., and Prendergast, G. C. (2005) Inhibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat Med 11, 312-319 31.Hou, D.-Y., Muller, A. J., Sharma, M. D., DuHadaway, J., Banerjee, T., Johnson, M., Mellor, A. L., Prendergast, G. C., and Munn, D. H. (2007) Inhibition of Indoleamine 2,3-Dioxygenase in Dendritic Cells by Stereoisomers of 1-Methyl-Tryptophan Correlates with Antitumor Responses. Cancer Research 67, 792-801 32.Ogawa, K., Hara, T., Shimizu, M., Ninomiya, S., Nagano, J., Sakai, H., Hoshi, M., Ito, H., Tsurumi, H., Saito, K., Seishima, M., Tanaka, T., and Moriwaki, H. (2012) Suppression of azoxymethane-induced colonic preneoplastic lesions in rats by 1-methyltryptophan, an inhibitor of indoleamine 2,3-dioxygenase. Cancer Science 103, 951-958 33.Nakamura, N., Hara, T., Shimizu, M., Mabuchi, R., Nagano, J., Ohno, T., Kochi, T., Kubota, M., Shirakami, Y., and Goto, N. (2015) Effects of indoleamine 2, 3-dioxygenase inhibitor in non-Hodgkin lymphoma model mice. International journal of hematology 102, 327-334 34.Opitz, C. A., Litzenburger, U. M., Sahm, F., Ott, M., Tritschler, I., Trump, S., Schumacher, T., Jestaedt, L., Schrenk, D., and Weller, M. (2011) An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 478, 197-203 35.Bessede, A., Gargaro, M., Pallotta, M. T., Matino, D., Servillo, G., Brunacci, C., Bicciato, S., Mazza, E. M. C., Macchiarulo, A., Vacca, C., Iannitti, R., Tissi, L., Volpi, C., Belladonna, M. L., Orabona, C., Bianchi, R., Lanz, T. V., Platten, M., Della Fazia, M. A., Piobbico, D., Zelante, T., Funakoshi, H., Nakamura, T., Gilot, D., Denison, M. S., Guillemin, G. J., DuHadaway, J. B., Prendergast, G. C., Metz, R., Geffard, M., Boon, L., Pirro, M., Iorio, A., Veyret, B., Romani, L., Grohmann, U., Fallarino, F., and Puccetti, P. (2014) Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature 511, 184-190 36.Bersten, D. C., Sullivan, A. E., Peet, D. J., and Whitelaw, M. L. (2013) bHLH-PAS proteins in cancer. Nat Rev Cancer 13, 827-841 37.Whitlock Jr, J. P. (1993) Mechanistic aspects of dioxin action. Chemical research in toxicology 6, 754-763 38.Hankinson, O. (1995) The aryl hydrocarbon receptor complex. Annual review of pharmacology and toxicology 35, 307-340 39.Rowlands, J. C., and Gustafsson, J.-Å. (1997) Aryl hydrocarbon receptor-mediated signal transduction. Critical reviews in toxicology 27, 109-134 40.Swanson, H. I., and Bradfield, C. A. (1993) The AH-receptor: genetics, structure and function. Pharmacogenetics and Genomics 3, 213-230 41.Denison, M. S., Pandini, A., Nagy, S. R., Baldwin, E. P., and Bonati, L. (2002) Ligand binding and activation of the Ah receptor. Chemico-biological interactions 141, 3-24 42.Bock, K. W., and Köhle, C. (2005) Ah receptor-and TCDD-mediated liver tumor promotion: clonal selection and expansion of cells evading growth arrest and apoptosis. Biochemical pharmacology 69, 1403-1408 43.Knerr, S., and Schrenk, D. (2006) Carcinogenicity of 2, 3, 7, 8‐tetrachlorodibenzo‐p‐dioxin in experimental models. Molecular nutrition & food research 50, 897-907 44.Beischlag, T. V., Morales, J. L., Hollingshead, B. D., and Perdew, G. H. (2008) The aryl hydrocarbon receptor complex and the control of gene expression. Critical Reviews™ in Eukaryotic Gene Expression 18 45.ujisawa-Sehara, A., ogawa, K., Nishi, C., and Fujii-Kuriyama, Y. (1986) Regulatory DNA elements localized remotely upstream from the drug-metabolizing cytochrome P-450c gene. Nucleic acids research 14, 1465-1477 46.Jin, U.-H., Lee, S.-O., Sridharan, G., Lee, K., Davidson, L. A., Jayaraman, A., Chapkin, R. S., Alaniz, R., and Safe, S. (2014) Microbiome-derived tryptophan metabolites and their aryl hydrocarbon receptor-dependent agonist and antagonist activities. Molecular pharmacology 85, 777-788 47.Zelante, T., Iannitti, R. G., Cunha, C., De Luca, A., Giovannini, G., Pieraccini, G., Zecchi, R., D’Angelo, C., Massi-Benedetti, C., and Fallarino, F. (2013) Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 39, 372-385 48.Fukumoto, S., Toshimitsu, T., Matsuoka, S., Maruyama, A., Oh-oka, K., Takamura, T., Nakamura, Y., Ishimaru, K., Fujii-Kuriyama, Y., and Ikegami, S. (2014) Identification of a probiotic bacteria-derived activator of the aryl hydrocarbon receptor that inhibits colitis. Immunology and cell biology 92, 460 49.Magiatis, P., Pappas, P., Gaitanis, G., Mexia, N., Melliou, E., Galanou, M., Vlachos, C., Stathopoulou, K., Skaltsounis, A. L., and Marselos, M. (2013) Malassezia yeasts produce a collection of exceptionally potent activators of the Ah (dioxin) receptor detected in diseased human skin. Journal of Investigative Dermatology 133, 2023-2030 50.van den Bogaard, E. H., Bergboer, J. G., Vonk-Bergers, M., van Vlijmen-Willems, I. M., Hato, S. V., van der Valk, P. G., Schröder, J. M., Joosten, I., Zeeuwen, P. L., and Schalkwijk, J. (2013) Coal tar induces AhR-dependent skin barrier repair in atopic dermatitis. The Journal of clinical investigation 123, 917 51.Murray, I. A., Patterson, A. D., and Perdew, G. H. (2014) Aryl hydrocarbon receptor ligands in cancer: friend and foe. Nature Reviews Cancer 14, 801-814 52.Puga, A., Xia, Y., and Elferink, C. (2002) Role of the aryl hydrocarbon receptor in cell cycle regulation. Chemico-biological interactions 141, 117-130 53.Oesch-Bartlomowicz, B., Huelster, A., Wiss, O., Antoniou-Lipfert, P., Dietrich, C., Arand, M., Weiss, C., Bockamp, E., and Oesch, F. (2005) Aryl hydrocarbon receptor activation by cAMP vs. dioxin: divergent signaling pathways. Proceedings of the National Academy of Sciences of the United States of America 102, 9218-9223 54.Fernandez-Salguero, P. M. (2010) A remarkable new target gene for the dioxin receptor: the Vav3 proto-oncogene links AhR to adhesion and migration. Cell adhesion & migration 4, 172-175 55.DiNatale, B. C., Smith, K., John, K., Krishnegowda, G., Amin, S. G., and Perdew, G. H. (2012) Ah receptor antagonism represses head and neck tumor cell aggressive phenotype. Molecular cancer research 10, 1369-1379 56.Ikuta, T., and Kawajiri, K. (2006) Zinc finger transcription factor Slug is a novel target gene of aryl hydrocarbon receptor. Experimental cell research 312, 3585-3594 57.Gumbiner, B. M. (1996) Cell adhesion: the molecular basis of tissue architecture and morphogenesis. Cell 84, 345-357 58.Funayama, N., Fagotto, F., McCrea, P., and Gumbiner, B. M. (1995) Embryonic axis induction by the armadillo repeat domain of beta-catenin: evidence for intracellular signaling. The Journal of cell biology 128, 959-968 59.Gumbiner, B. M. (1995) Signal transduction by β-catenin. Current opinion in cell biology 7, 634-640 60.Peifer, M., McCrea, P. D., Green, K. J., Wieschaus, E., and Gumbiner, B. M. (1992) The vertebrate adhesive junction proteins beta-catenin and plakoglobin and the Drosophila segment polarity gene armadillo form a multigene family with similar properties. The Journal of cell biology 118, 681-691 61.Provost, E., and Rimm, D. L. (1999) Controversies at the cytoplasmic face of the cadherin-based adhesion complex. Current opinion in cell biology 11, 567-572 62.Jiang, W. (1996) E‐cadherin and its associated protein catenins, cancer invasion and metastasis. British journal of surgery 83, 437-446 63.Osada, T., Sakamoto, M., Ino, Y., Iwamatsu, A., Matsuno, Y., Muto, T., and Hirohashi, S. (1996) E‐cadherin is involved in the intrahepatic metastasis of hepatocellular carcinoma. Hepatology 24, 1460-1467 64.Miller, J. R., Hocking, A. M., Brown, J. D., and Moon, R. T. (1999) Mechanism and function of signal transduction by the Wnt/beta-catenin and Wnt/Ca2+ pathways. Oncogene 18, 7860-7872 65.Barker, N., Morin, P. J., and Clevers, H. (1999) The Yin-Yang of TCF/β-catenin signaling. Advances in cancer research 77, 1-24 66.Polakis, P. (1999) The oncogenic activation of β-catenin. Current opinion in genetics & development 9, 15-21 67.Munemitsu, S., Albert, I., Souza, B., Rubinfeld, B., and Polakis, P. (1995) Regulation of intracellular beta-catenin levels by the adenomatous polyposis coli (APC) tumor-suppressor protein. Proceedings of the National Academy of Sciences 92, 3046-3050 68.Yost, C., Torres, M., Miller, J. R., Huang, E., Kimelman, D., and Moon, R. T. (1996) The axis-inducing activity, stability, and subcellular distribution of beta-catenin is regulated in Xenopus embryos by glycogen synthase kinase 3. Genes & development 10, 1443-1454 69.Hart, M. J., de los Santos, R., Albert, I. N., Rubinfeld, B., and Polakis, P. (1998) Downregulation of β-catenin by human Axin and its association with the APC tumor suppressor, β-catenin and GSK3β. Current Biology 8, 573-581 70.Kitagawa, M., Hatakeyama, S., Shirane, M., Matsumoto, M., Ishida, N., Hattori, K., Nakamichi, I., Kikuchi, A., Nakayama, K. i., and Nakayama, K. (1999) An F‐box protein, FWD1, mediates ubiquitin‐dependent proteolysis of β‐catenin. The EMBO journal 18, 2401-2410 71.Aberle, H., Bauer, A., Stappert, J., Kispert, A., and Kemler, R. (1997) β‐catenin is a target for the ubiquitin–proteasome pathway. The EMBO journal 16, 3797-3804 72.Kikuchi, A. (2000) Regulation of β-catenin signaling in the Wnt pathway. Biochemical and biophysical research communications 268, 243-248 73.Omer, C. A., Miller, P. J., Diehl, R. E., and Kral, A. M. (1999) Identification of Tcf4 residues involved in high-affinity β-catenin binding. Biochemical and biophysical research communications 256, 584-590 74.Behrens, J., von Kries, J. P., Kühl, M., Bruhn, L., Wedlich, D., Grosschedl, R., and Birchmeier, W. (1996) Functional interaction of β-catenin with the transcription factor LEF-1. Nature 382, 638-642 75.Stahl, S., Ittrich, C., Marx‐Stoelting, P., Köhle, C., Altug‐Teber, Ö., Riess, O., Bonin, M., Jobst, J., Kaiser, S., and Buchmann, A. (2005) Genotype–phenotype relationships in hepatocellular tumors from mice and man. Hepatology 42, 353-361 76.Loeppen, S., Koehle, C., Buchmann, A., and Schwarz, M. (2005) A β-catenin-dependent pathway regulates expression of cytochrome P450 isoforms in mouse liver tumors. Carcinogenesis 26, 239-248 77.Tan, X., Behari, J., Cieply, B., Michalopoulos, G. K., and Monga, S. P. (2006) Conditional deletion of β-catenin reveals its role in liver growth and regeneration. Gastroenterology 131, 1561-1572 78.Sekine, S., Lan, B. Y. A., Bedolli, M., Feng, S., and Hebrok, M. (2006) Liver‐specific loss of β‐catenin blocks glutamine synthesis pathway activity and cytochrome p450 expression in mice. Hepatology 43, 817-825 79.Godoy, P., Hewitt, N. J., Albrecht, U., Andersen, M. E., Ansari, N., Bhattacharya, S., Bode, J. G., Bolleyn, J., Borner, C., and Böttger, J. (2013) Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Archives of toxicology 87, 1315-1530 80.Braeuning, A., Köhle, C., Buchmann, A., and Schwarz, M. (2011) Coordinate regulation of cytochrome P450 1A1 expression in mouse liver by the aryl hydrocarbon receptor and the β-catenin pathway. Toxicological sciences 122, 16-25 81.Okumura, N., Yoshida, H., Kitagishi, Y., Murakami, M., Nishimura, Y., and Matsuda, S. (2012) PI3K/AKT/PTEN signaling as a molecular target in leukemia angiogenesis. Advances in hematology 2012 82.Gao, C., Hölscher, C., Liu, Y., and Li, L. (2012) GSK3: a key target for the development of novel treatments for type 2 diabetes mellitus and Alzheimer disease. Reviews in the Neurosciences 23, 1-11 83.Welcker, M., Singer, J., Loeb, K. R., Grim, J., Bloecher, A., Gurien-West, M., Clurman, B. E., and Roberts, J. M. (2003) Multisite phosphorylation by Cdk2 and GSK3 controls cyclin E degradation. Molecular cell 12, 381-392 84.Sherr, C. J., and Roberts, J. M. (1999) CDK inhibitors: positive and negative regulators of G1-phase progression. Genes & development 13, 1501-1512 85.Diehl, J. A., Cheng, M., Roussel, M. F., and Sherr, C. J. (1998) Glycogen synthase kinase-3β regulates cyclin D1 proteolysis and subcellular localization. Genes & development 12, 3499-3511 86.Leis, H., Segrelles, C., Ruiz, S., Santos, M., and Paramio, J. M. (2002) Expression, localization, and activity of glycogen synthase kinase 3β during mouse skin tumorigenesis. Molecular carcinogenesis 35, 180-185 87.Doble, B. W., and Woodgett, J. R. (2003) GSK-3: tricks of the trade for a multi-tasking kinase. Journal of cell science 116, 1175-1186 88.Song, M. S., Salmena, L., and Pandolfi, P. P. (2012) The functions and regulation of the PTEN tumour suppressor. Nature reviews. Molecular cell biology 13, 283 89.Pan, K., Wang, H., Chen, M. S., Zhang, H. K., Weng, D. S., Zhou, J., Huang, W., Li, J. J., Song, H. F., and Xia, J. C. (2008) Expression and prognosis role of indoleamine 2,3-dioxygenase in hepatocellular carcinoma. J Cancer Res Clin Oncol 134, 1247-1253 90.Higashitani, K., Kanto, T., Kuroda, S., Yoshio, S., Matsubara, T., Kakita, N., Oze, T., Miyazaki, M., Sakakibara, M., Hiramatsu, N., Mita, E., Imai, Y., Kasahara, A., Okuno, A., Takikawa, O., Hayashi, N., and Takehara, T. (2013) Association of enhanced activity of indoleamine 2,3-dioxygenase in dendritic cells with the induction of regulatory T cells in chronic hepatitis C infection. J Gastroenterol 48, 660-670 91.Moennikes, O., Loeppen, S., Buchmann, A., Andersson, P., Ittrich, C., Poellinger, L., and Schwarz, M. (2004) A constitutively active dioxin/aryl hydrocarbon receptor promotes hepatocarcinogenesis in mice. Cancer research 64, 4707-4710 92.Andersson, P., McGuire, J., Rubio, C., Gradin, K., Whitelaw, M. L., Pettersson, S., Hanberg, A., and Poellinger, L. (2002) A constitutively active dioxin/aryl hydrocarbon receptor induces stomach tumors. Proceedings of the National Academy of Sciences 99, 9990-9995 93.Fernandez-Salguero, P., Pineau, T., Hilbert, D. M., and McPhail, T. (1995) Immune system impairment and hepatic fibrosis in mice lacking the dioxin-binding Ah receptor. Science 268, 722 94.Gramatzki, D., Pantazis, G., Schittenhelm, J., Tabatabai, G., Köhle, C., Wick, W., Schwarz, M., Weller, M., and Tritschler, I. (2009) Aryl hydrocarbon receptor inhibition downregulates the TGF-[beta]/Smad pathway in human glioblastoma cells. Oncogene 28, 2593 95.Denison, M. S., and Nagy, S. R. (2003) Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annual review of pharmacology and toxicology 43, 309-334 96.Liu, Y., Peterson, D. A., Kimura, H., and Schubert, D. (1997) Mechanism of cellular 3‐(4, 5‐dimethylthiazol‐2‐yl)‐2, 5‐diphenyltetrazolium bromide (MTT) reduction. Journal of neurochemistry 69, 581-593 97.Berridge, M. V., and Tan, A. S. (1993) Characterization of the cellular reduction of 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT): subcellular localization, substrate dependence, and involvement of mitochondrial electron transport in MTT reduction. Archives of biochemistry and biophysics 303, 474-482 98.Nelson, J. D., Denisenko, O., and Bomsztyk, K. (2006) Protocol for the fast chromatin immunoprecipitation (ChIP) method. Nature protocols 1, 179 99.Lai, D.-W., Liu, S.-H., Karlsson, A. I., Lee, W.-J., Wang, K.-B., Chen, Y.-C., Shen, C.-C., Wu, S.-M., Liu, C.-Y., and Tien, H.-R. (2014) The novel Aryl hydrocarbon receptor inhibitor biseugenol inhibits gastric tumor growth and peritoneal dissemination. Oncotarget 5, 7788 100.Takikawa, O., Kuroiwa, T., Yamazaki, F., and Kido, R. (1988) Mechanism of interferon-gamma action. Characterization of indoleamine 2, 3-dioxygenase in cultured human cells induced by interferon-gamma and evaluation of the enzyme-mediated tryptophan degradation in its anticellular activity. Journal of Biological Chemistry 263, 2041-2048 101.Chung, T. W., Tan, K. T., Chan, H. L., Lai, M. D., Yen, M. C., Li, Y. R., Lin, S. H., and Lin, C. C. (2014) Induction of indoleamine 2,3-dioxygenase (IDO) enzymatic activity contributes to interferon-gamma induced apoptosis and death receptor 5 expression in human non-small cell lung cancer cells. Asian Pac J Cancer Prev 15, 7995-8001 102.Mahanonda, R., Sa-Ard-Iam, N., Montreekachon, P., Pimkhaokham, A., Yongvanichit, K., Fukuda, M. M., and Pichyangkul, S. (2007) IL-8 and IDO expression by human gingival fibroblasts via TLRs. The Journal of Immunology 178, 1151-1157 103.Peng, T.-L., Chen, J., Mao, W., Liu, X., Tao, Y., Chen, L.-Z., and Chen, M.-H. (2009) Potential therapeutic significance of increased expression of aryl hydrocarbon receptor in human gastric cancer. World journal of gastroenterology: WJG 15, 1719 104.Xu, H., Rajesan, R., Harper, P., Kim, R. B., Lonnerdal, B., Yang, M., Uematsu, S., Hutson, J., Watson‐MacDonell, J., and Ito, S. (2005) Induction of cytochrome P450 1A by cow milk‐based formula: a comparative study between human milk and formula. British journal of pharmacology 146, 296-305 105.Sagan, D., Kocki, T., Kocki, J., and Szumilo, J. (2012) Serum kynurenic acid: possible association with invasiveness of non-small cell lung cancer. Asian Pacific Journal of Cancer Prevention 13, 4241-4244 106.Fujigaki, H., Seishima, M., and Saito, K. (2012) Posttranslational modification of indoleamine 2, 3-dioxygenase. Analytical and bioanalytical chemistry 403, 1777-1782 107.Ozaki, Y., Edelstein, M. P., and Duch, D. S. (1988) Induction of indoleamine 2, 3-dioxygenase: a mechanism of the antitumor activity of interferon gamma. Proceedings of the National Academy of Sciences 85, 1242-1246 108.Luis, M., and Peterson, E. M. (1988) Dependence of the in vitro antiproliferative activity of recombinant human γ-interferon on the concentration of tryptophan in culture media. Cancer research 48, 346-350 109.Thaker, A. I., Rao, M. S., Bishnupuri, K. S., Kerr, T. A., Foster, L., Marinshaw, J. M., Newberry, R. D., Stenson, W. F., and Ciorba, M. A. (2013) IDO1 metabolites activate β-catenin signaling to promote cancer cell proliferation and colon tumorigenesis in mice. Gastroenterology 145, 416-425. e414 110.Nelson, W. J., and Nusse, R. (2004) Convergence of Wnt, ß-Catenin, and Cadherin Pathways. Science 303, 1483-1487 111.Thiery, J. P. (2002) Epithelial-mesenchymal transitions in tumour progression. Nature reviews. Cancer 2, 442 112.Nieto, M. A. (2002) The snail superfamily of zinc-finger transcription factors. Nature reviews. Molecular cell biology 3, 155 113.Batlle, E., Sancho, E., Francí, C., Domínguez, D., Monfar, M., Baulida, J., and de Herreros, A. G. (2000) The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nature cell biology 2, 84 114.Sato, N., Saga, Y., Mizukami, H., Wang, D., Takahashi, S., Nonaka, H., Fujiwara, H., Takei, Y., Machida, S., and Takikawa, O. (2012) Downregulation of indoleamine-2, 3-dioxygenase in cervical cancer cells suppresses tumor growth by promoting natural killer cell accumulation. Oncology reports 28, 1574-1578 115.Wang, D., Saga, Y., Mizukami, H., Sato, N., Nonaka, H., Fujiwara, H., Takei, Y., Machida, S., Takikawa, O., and Ozawa, K. (2012) Indoleamine-2, 3-dioxygenase, an immunosuppressive enzyme that inhibits natural killer cell function, as a useful target for ovarian cancer therapy. International journal of oncology 40, 929-934 116.Bui, L.-C., Tomkiewicz, C., Chevallier, A., Pierre, S., Bats, A.-S., Mota, S., Raingeaud, J., Pierre, J., Diry, M., and Transy, C. (2009) Nedd9/Hef1/Cas-L mediates the effects of environmental pollutants on cell migration and plasticity. Oncogene 28, 3642 117.Zudaire, E., Cuesta, N., Murty, V., Woodson, K., Adams, L., Gonzalez, N., Martínez, A., Narayan, G., Kirsch, I., and Franklin, W. (2008) The aryl hydrocarbon receptor repressor is a putative tumor suppressor gene in multiple human cancers. The Journal of clinical investigation 118, 640 118.Nguyen, L. P., and Bradfield, C. A. (2007) The search for endogenous activators of the aryl hydrocarbon receptor. Chemical research in toxicology 21, 102-116 119.Zhang, H., Yao, Y., Chen, Y., Yue, C., Chen, J., Tong, J., Jiang, Y., and Chen, T. (2016) Crosstalk between AhR and wnt/β-catenin signal pathways in the cardiac developmental toxicity of PM2.5 in zebrafish embryos. Toxicology 355, 31-38 120.Wincent, E., Stegeman, J. J., and Jönsson, M. E. (2015) Combination effects of AhR agonists and Wnt/β-catenin modulators in zebrafish embryos: Implications for physiological and toxicological AhR functions. Toxicology and applied pharmacology 284, 163-179 121.Procházková, J., Kabátková, M., Bryja, V., Umannová, L., Bernatík, O., Kozubík, A., Machala, M., and Vondráček, J. (2011) The Interplay of the Aryl Hydrocarbon Receptor and β-Catenin Alters Both AhR-Dependent Transcription and Wnt/β-Catenin Signaling in Liver Progenitors. Toxicological Sciences 122, 349-360 122.Kawajiri, K., Kobayashi, Y., Ohtake, F., Ikuta, T., Matsushima, Y., Mimura, J., Pettersson, S., Pollenz, R. S., Sakaki, T., and Hirokawa, T. (2009) Aryl hydrocarbon receptor suppresses intestinal carcinogenesis in ApcMin/+ mice with natural ligands. Proceedings of the National Academy of Sciences 106, 13481-13486 123.Al-Dhfyan, A., Alhoshani, A., and Korashy, H. M. (2017) Aryl hydrocarbon receptor/cytochrome P450 1A1 pathway mediates breast cancer stem cells expansion through PTEN inhibition and β-Catenin and Akt activation. Molecular cancer 16, 14 124.Lau, M., Klausen, C., and Leung, P. (2011) E-cadherin inhibits tumor cell growth by suppressing PI3K/Akt signaling via [beta]-catenin-Egr1-mediated PTEN expression. Oncogene 30, 2753
|