- -

Stress hormones promote growth of B16-F10 melanoma metastases: an interleukin 6-and glutathione-dependent mechanism

RiuNet: Repositorio Institucional de la Universidad Politécnica de Valencia

Compartir/Enviar a

Citas

Estadísticas

  • Estadisticas de Uso

Stress hormones promote growth of B16-F10 melanoma metastases: an interleukin 6-and glutathione-dependent mechanism

Mostrar el registro completo del ítem

Valles, SL.; Benlloch, M.; Rodriguez, ML.; Mena-Mollá, S.; Pellicer, JA.; Asensi-Miralles, MÁ.; Obrador, E.... (2013). Stress hormones promote growth of B16-F10 melanoma metastases: an interleukin 6-and glutathione-dependent mechanism. Journal of Translational Medicine. 11:1-14. https://doi.org/10.1186/1479-5876-11-72

Por favor, use este identificador para citar o enlazar este ítem: http://hdl.handle.net/10251/81330

Ficheros en el ítem

Metadatos del ítem

Título: Stress hormones promote growth of B16-F10 melanoma metastases: an interleukin 6-and glutathione-dependent mechanism
Autor: Valles, Soraya L. Benlloch, Maria Rodriguez , María Lucía Mena-Mollá, Salvador Pellicer, Jose A Asensi-Miralles, Miguel Ángel Obrador, Elena Estrela, José María
Entidad UPV: Universitat Politècnica de València. Instituto de Reconocimiento Molecular y Desarrollo Tecnológico - Institut de Reconeixement Molecular i Desenvolupament Tecnològic
Fecha difusión:
Resumen:
[EN] Background: Interleukin (IL)-6 (mainly of tumor origin) activates glutathione (GSH) release from hepatocytes and its interorgan transport to B16-F10 melanoma metastatic foci. We studied if this capacity to overproduce ...[+]
Palabras clave: Metastases , Glutathione , Interleukin 6 , Stress hormones , Apoptosis
Derechos de uso: Reserva de todos los derechos
Fuente:
Journal of Translational Medicine. (issn: 1479-5876 ) (eissn: 23517603 )
DOI: 10.1186/1479-5876-11-72
Editorial:
BioMed Central
Versión del editor: http://doi.org/10.1186/1479-5876-11-72
Código del Proyecto:
info:eu-repo/grantAgreement/MICINN//SAF2009-07729/ES/Terapia Anti-Bcl-2 Y Deplecion De Gsh: Sensibilizacion Del Melanoma Maligno Humano A La Quimioradioterapia/
info:eu-repo/grantAgreement/MICINN//IPT-010000-2010-021/ES/NANOCOMPLEJOS DE ARN SINTETICO COMO NUEVA TERAPIA CONTRA CÁNCERES AGRESIVOS PARA LOS QUE NO SE DISPONE DE TRATAMIENTO EFECTIVO/
Agradecimientos:
This research was supported by grant (SAF2009-07729 and IPT-010000-2010-21) from the Ministerio de Economia y Competitividad (http://www.idi.mineco.gob.es), Spain.
Tipo: Artículo

References

Meister, A. (1983). Selective modification of glutathione metabolism. Science, 220(4596), 472-477. doi:10.1126/science.6836290

Estrela, J. M., Ortega, A., & Obrador, E. (2006). Glutathione in Cancer Biology and Therapy. Critical Reviews in Clinical Laboratory Sciences, 43(2), 143-181. doi:10.1080/10408360500523878

Obrador, E., Benlloch, M., Pellicer, J. A., Asensi, M., & Estrela, J. M. (2011). Intertissue Flow of Glutathione (GSH) as a Tumor Growth-promoting Mechanism. Journal of Biological Chemistry, 286(18), 15716-15727. doi:10.1074/jbc.m110.196261 [+]
Meister, A. (1983). Selective modification of glutathione metabolism. Science, 220(4596), 472-477. doi:10.1126/science.6836290

Estrela, J. M., Ortega, A., & Obrador, E. (2006). Glutathione in Cancer Biology and Therapy. Critical Reviews in Clinical Laboratory Sciences, 43(2), 143-181. doi:10.1080/10408360500523878

Obrador, E., Benlloch, M., Pellicer, J. A., Asensi, M., & Estrela, J. M. (2011). Intertissue Flow of Glutathione (GSH) as a Tumor Growth-promoting Mechanism. Journal of Biological Chemistry, 286(18), 15716-15727. doi:10.1074/jbc.m110.196261

Meister, A. (1991). Glutathione deficiency produced by inhibition of its synthesis, and its reversal; Applications in research and therapy. Pharmacology & Therapeutics, 51(2), 155-194. doi:10.1016/0163-7258(91)90076-x

Hanigan, M. H. (1995). Expression of gamma-glutamyl transpeptidase provides tumor cells with a selective growth advantage at physiologic concentrations of cyst(e)ine. Carcinogenesis, 16(2), 181-185. doi:10.1093/carcin/16.2.181

Obrador, E. (2002). γ-Glutamyl transpeptidase overexpression increases metastatic growth of B16 melanoma cells in the mouse liver. Hepatology, 35(1), 74-81. doi:10.1053/jhep.2002.30277

Ballatori, N., & Rebbeor, J. (1998). Roles of MRP2 and oatp1 in Hepatocellular Export of Reduced Glutathione. Seminars in Liver Disease, 18(04), 377-387. doi:10.1055/s-2007-1007171

Hodge, D. R., Hurt, E. M., & Farrar, W. L. (2005). The role of IL-6 and STAT3 in inflammation and cancer. European Journal of Cancer, 41(16), 2502-2512. doi:10.1016/j.ejca.2005.08.016

Barton, B. E. (2005). Interleukin-6 and new strategies for the treatment of cancer, hyperproliferative diseases and paraneoplastic syndromes. Expert Opinion on Therapeutic Targets, 9(4), 737-752. doi:10.1517/14728222.9.4.737

Rose-John, S., Waetzig, G. H., Scheller, J., Grötzinger, J., & Seegert, D. (2007). The IL-6/sIL-6R complex as a novel target for therapeutic approaches. Expert Opinion on Therapeutic Targets, 11(5), 613-624. doi:10.1517/14728222.11.5.613

Ara, T., & DeClerck, Y. A. (2010). Interleukin-6 in bone metastasis and cancer progression. European Journal of Cancer, 46(7), 1223-1231. doi:10.1016/j.ejca.2010.02.026

Emmenegger, U., & Kerbel, R. S. (2010). Chemotherapy counteracted. Nature, 468(7324), 637-638. doi:10.1038/468637a

Wang, Y., Niu, X. L., Qu, Y., Wu, J., Zhu, Y. Q., Sun, W. J., & Li, L. Z. (2010). Autocrine production of interleukin-6 confers cisplatin and paclitaxel resistance in ovarian cancer cells. Cancer Letters, 295(1), 110-123. doi:10.1016/j.canlet.2010.02.019

Sternberg, E. M. (1997). Neural-immune interactions in health and disease. Journal of Clinical Investigation, 100(11), 2641-2647. doi:10.1172/jci119807

Reiche, E. M. V., Nunes, S. O. V., & Morimoto, H. K. (2004). Stress, depression, the immune system, and cancer. The Lancet Oncology, 5(10), 617-625. doi:10.1016/s1470-2045(04)01597-9

Besedovsky, H. O., Del Rey, A., Klusman, I., Furukawa, H., Monge Arditi, G., & Kabiersch, A. (1991). Cytokines as modulators of the hypothalamus-pituitary-adrenal axis. The Journal of Steroid Biochemistry and Molecular Biology, 40(4-6), 613-618. doi:10.1016/0960-0760(91)90284-c

Bethin, K. E., Vogt, S. K., & Muglia, L. J. (2000). Interleukin-6 is an essential, corticotropin-releasing hormone-independent stimulator of the adrenal axis during immune system activation. Proceedings of the National Academy of Sciences, 97(16), 9317-9322. doi:10.1073/pnas.97.16.9317

Herr, I., & Pfitzenmaier, J. (2006). Glucocorticoid use in prostate cancer and other solid tumours: implications for effectiveness of cytotoxic treatment and metastases. The Lancet Oncology, 7(5), 425-430. doi:10.1016/s1470-2045(06)70694-5

Bernabé, D. G., Tamae, A. C., Biasoli, É. R., & Oliveira, S. H. P. (2011). Stress hormones increase cell proliferation and regulates interleukin-6 secretion in human oral squamous cell carcinoma cells. Brain, Behavior, and Immunity, 25(3), 574-583. doi:10.1016/j.bbi.2010.12.012

Antoni, M. H., Lutgendorf, S. K., Cole, S. W., Dhabhar, F. S., Sephton, S. E., McDonald, P. G., … Sood, A. K. (2006). The influence of bio-behavioural factors on tumour biology: pathways and mechanisms. Nature Reviews Cancer, 6(3), 240-248. doi:10.1038/nrc1820

Yang, E. V., Kim, S., Donovan, E. L., Chen, M., Gross, A. C., Webster Marketon, J. I., … Glaser, R. (2009). Norepinephrine upregulates VEGF, IL-8, and IL-6 expression in human melanoma tumor cell lines: Implications for stress-related enhancement of tumor progression. Brain, Behavior, and Immunity, 23(2), 267-275. doi:10.1016/j.bbi.2008.10.005

Carretero, J., Obrador, E., Anasagasti, M. J., Martin, J. J., Vidal-Vanaclocha, F., & Estrela, J. M. (1999). Clinical and Experimental Metastasis, 17(7), 567-574. doi:10.1023/a:1006725226078

Lachize, S., Apostolakis, E. M., van der Laan, S., Tijssen, A. M. I., Xu, J., de Kloet, E. R., & Meijer, O. C. (2009). Steroid receptor coactivator-1 is necessary for regulation of corticotropin-releasing hormone by chronic stress and glucocorticoids. Proceedings of the National Academy of Sciences, 106(19), 8038-8042. doi:10.1073/pnas.0812062106

Veenema, A. H., Reber, S. O., Selch, S., Obermeier, F., & Neumann, I. D. (2008). Early Life Stress Enhances the Vulnerability to Chronic Psychosocial Stress and Experimental Colitis in Adult Mice. Endocrinology, 149(6), 2727-2736. doi:10.1210/en.2007-1469

Asensi, M., Sastre, J., Pallardo, F. V., Delaasuncion, J. G., Estrela, J. M., & Vina, J. (1994). A High-Performance Liquid Chromatography Method for Measurement of Oxidized Glutathione in Biological Samples. Analytical Biochemistry, 217(2), 323-328. doi:10.1006/abio.1994.1126

Ortega, A. L., Carretero, J., Obrador, E., Gambini, J., Asensi, M., Rodilla, V., & Estrela, J. M. (2003). Tumor Cytotoxicity by Endothelial Cells. Journal of Biological Chemistry, 278(16), 13888-13897. doi:10.1074/jbc.m207140200

SAKAKIBARA, H., KOYANAGI, A., SUZUKI, T., SUZUKI, A., LING, L., & SHIMOI, K. (2010). Effects of Animal Care Procedures on Plasma Corticosterone Levels in Group-Housed Mice during the Nocturnal Active Phase. Experimental Animals, 59(5), 637-642. doi:10.1538/expanim.59.637

Lucot, J. B., Jackson, N., Bernatova, I., & Morris, M. (2005). Measurement of plasma catecholamines in small samples from mice. Journal of Pharmacological and Toxicological Methods, 52(2), 274-277. doi:10.1016/j.vascn.2004.11.004

Dobos, J., Kenessey, I., Tímár, J., & Ladányi, A. (2011). Glucocorticoid Receptor Expression and Antiproliferative Effect of Dexamethasone on Human Melanoma Cells. Pathology & Oncology Research, 17(3), 729-734. doi:10.1007/s12253-011-9377-8

Tsuji, M., Kuno, T., Tanaka, C., Ichihashi, M., & Mishima, Y. (1983). Beta-adrenergic receptors of B16 melanoma cell. Archives of Dermatological Research, 275(6), 415-416. doi:10.1007/bf00417345

Im, A., & Appleman, L. J. (2010). Mifepristone: pharmacology and clinical impact in reproductive medicine, endocrinology and oncology. Expert Opinion on Pharmacotherapy, 11(3), 481-488. doi:10.1517/14656560903535880

Smoak, K. A., & Cidlowski, J. A. (2004). Mechanisms of glucocorticoid receptor signaling during inflammation. Mechanisms of Ageing and Development, 125(10-11), 697-706. doi:10.1016/j.mad.2004.06.010

Cole, S. W., & Sood, A. K. (2011). Molecular Pathways: Beta-Adrenergic Signaling in Cancer: Figure 1. Clinical Cancer Research, 18(5), 1201-1206. doi:10.1158/1078-0432.ccr-11-0641

Matsusaka, T., Fujikawa, K., Nishio, Y., Mukaida, N., Matsushima, K., Kishimoto, T., & Akira, S. (1993). Transcription factors NF-IL6 and NF-kappa B synergistically activate transcription of the inflammatory cytokines, interleukin 6 and interleukin 8. Proceedings of the National Academy of Sciences, 90(21), 10193-10197. doi:10.1073/pnas.90.21.10193

McEwen, B. S. (2007). Physiology and Neurobiology of Stress and Adaptation: Central Role of the Brain. Physiological Reviews, 87(3), 873-904. doi:10.1152/physrev.00041.2006

Lee, J.-H., Yoo, S. B., Kim, N. Y., Cha, M. J., & Jahng, J. W. (2008). Interleukin-6 and the Hypothalamic-Pituitary-Adrenal Activation in a Tumor Bearing Mouse. International Journal of Neuroscience, 118(3), 355-364. doi:10.1080/00207450701592915

Li, Y.-F., He, R.-R., Tsoi, B., Li, X.-D., Li, W.-X., Abe, K., & Kurihara, H. (2012). Anti-Stress Effects of Carnosine on Restraint-Evoked Immunocompromise in Mice through Spleen Lymphocyte Number Maintenance. PLoS ONE, 7(4), e33190. doi:10.1371/journal.pone.0033190

Sarabdjitsingh, R. A., Kofink, D., Karst, H., de Kloet, E. R., & Joëls, M. (2012). Stress-Induced Enhancement of Mouse Amygdalar Synaptic Plasticity Depends on Glucocorticoid and ß-Adrenergic Activity. PLoS ONE, 7(8), e42143. doi:10.1371/journal.pone.0042143

Moreno-Smith, M., Lutgendorf, S. K., & Sood, A. K. (2010). Impact of stress on cancer metastasis. Future Oncology, 6(12), 1863-1881. doi:10.2217/fon.10.142

Tissing, W. J. E., Meijerink, J. P. P., den Boer, M. L., & Pieters, R. (2003). Molecular determinants of glucocorticoid sensitivity and resistance in acute lymphoblastic leukemia. Leukemia, 17(1), 17-25. doi:10.1038/sj.leu.2402733

Anderer, G., Schrappe, M., Brechlin, A. M., Lauten, M., Muti, P., Welte, K., & Stanulla, M. (2000). Polymorphisms within glutathione S-transferase genes and initial response to glucocorticoids in childhood acute lymphoblastic leukaemia. Pharmacogenetics, 10(8), 715-726. doi:10.1097/00008571-200011000-00006

Thaker, P. H., & Sood, A. K. (2008). Neuroendocrine influences on cancer biology. Seminars in Cancer Biology, 18(3), 164-170. doi:10.1016/j.semcancer.2007.12.005

Takeda, T., Kurachi, H., Yamamoto, T., Nishio, Y., Nakatsuji, Y., Morishige, K., … Murata, Y. (1998). Crosstalk between the interleukin-6 (IL-6)-JAK-STAT and the glucocorticoid-nuclear receptor pathway: synergistic activation of IL-6 response element by IL-6 and glucocorticoid. Journal of Endocrinology, 159(2), 323-330. doi:10.1677/joe.0.1590323

Rodriguez-Rocha, H., Garcia Garcia, A., Zavala-Flores, L., Li, S., Madayiputhiya, N., & Franco, R. (2012). Glutaredoxin 1 Protects Dopaminergic Cells by Increased Protein Glutathionylation in Experimental Parkinson’s Disease. Antioxidants & Redox Signaling, 17(12), 1676-1693. doi:10.1089/ars.2011.4474

Tome, M. E., Jaramillo, M. C., & Briehl, M. M. (2011). Hydrogen peroxide signaling is required for glucocorticoid-induced apoptosis in lymphoma cells. Free Radical Biology and Medicine, 51(11), 2048-2059. doi:10.1016/j.freeradbiomed.2011.09.002

Lázár-Molnár, E., Hegyesi, H., Tóth, S., & Falus, A. (2000). AUTOCRINE AND PARACRINE REGULATION BY CYTOKINES AND GROWTH FACTORS IN MELANOMA. Cytokine, 12(6), 547-554. doi:10.1006/cyto.1999.0614

Sansone, P., & Bromberg, J. (2012). Targeting the Interleukin-6/Jak/Stat Pathway in Human Malignancies. Journal of Clinical Oncology, 30(9), 1005-1014. doi:10.1200/jco.2010.31.8907

Arrigo, A.-P. (1999). Gene expression and the thiol redox state. Free Radical Biology and Medicine, 27(9-10), 936-944. doi:10.1016/s0891-5849(99)00175-6

Antelmann, H., & Helmann, J. D. (2011). Thiol-Based Redox Switches and Gene Regulation. Antioxidants & Redox Signaling, 14(6), 1049-1063. doi:10.1089/ars.2010.3400

Leibowitz, B., & Yu, J. (2010). Mitochondrial signaling in cell death via the Bcl-2 family. Cancer Biology & Therapy, 9(6), 417-422. doi:10.4161/cbt.9.6.11392

Powe, D. G., Voss, M. J., Habashy, H. O., Zänker, K. S., Green, A. R., Ellis, I. O., & Entschladen, F. (2011). Alpha- and beta-adrenergic receptor (AR) protein expression is associated with poor clinical outcome in breast cancer: an immunohistochemical study. Breast Cancer Research and Treatment, 130(2), 457-463. doi:10.1007/s10549-011-1371-z

Powe, D. G., & Entschladen, F. (2011). Using β-blockers to inhibit breast cancer progression. Nature Reviews Clinical Oncology, 8(9), 511-512. doi:10.1038/nrclinonc.2011.123

[-]

recommendations

 

Este ítem aparece en la(s) siguiente(s) colección(ones)

Mostrar el registro completo del ítem