Artículo

Estamos trabajando para incorporar este artículo al repositorio
Consulte el artículo en la página del editor
Consulte la política de Acceso Abierto del editor

Abstract:

IL-6 is a pleiotropic cytokine with multiple pathophysiological functions. As a key factor of the senescence secretome, it can not only promote tumorigenesis and cell proliferation but also exert tumor suppressive functions, depending on the cellular context. IL-6, as do other cytokines, plays important roles in the function, growth and neuroendocrine responses of the anterior pituitary gland. The multiple actions of IL-6 on normal and adenomatous pituitary function, cell proliferation, angiogenesis and extracellular matrix remodeling indicate its importance in the regulation of the anterior pituitary. Pituitary tumors are mostly benign adenomas with low mitotic index and rarely became malignant. Premature senescence occurs in slow-growing benign tumors, like pituitary adenomas. The dual role of IL-6 in senescence and tumorigenesis is well represented in pituitary tumor development, as it has been demonstrated that effects of paracrine IL-6 may allow initial pituitary cell growth, whereas autocrine IL-6 in the same tumor triggers senescence and restrains aggressive growth and malignant transformation. IL-6 is instrumental in promotion and maintenance of the senescence program in pituitary adenomas. © 2017 Society for Endocrinology.

Registro:

Documento: Artículo
Título:Programmed cell senescence: Role of IL-6 in the pituitary
Autor:Sapochnik, M.; Fuertes, M.; Arzt, E.
Filiación:Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), CONICET-Partner Institute of the Max Planck Society, Buenos Aires, Argentina
Departamento de Fisiología y Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
Palabras clave:Autocrine; IL-6; Pituitary tumor; Senescence; interleukin 6; IL6 protein, human; interleukin 6; mitogen activated protein kinase kinase; phosphatidylinositol 3 kinase; STAT protein; carcinogenesis; cell aging; cell growth; human; hypophysis; hypophysis adenoma; hypophysis cell; hypophysis tumor; malignant transformation; molecular pathology; nonhuman; paracrine signaling; priority journal; protein function; Review; adenohypophysis; adenoma; animal; autocrine effect; cell aging; cell cycle; cell proliferation; extracellular matrix; gene expression regulation; genetics; metabolism; neovascularization (pathology); pathology; Adenoma; Animals; Autocrine Communication; Cell Cycle; Cell Proliferation; Cellular Senescence; Extracellular Matrix; Gene Expression Regulation; Humans; Interleukin-6; Mitogen-Activated Protein Kinase Kinases; Neovascularization, Pathologic; Paracrine Communication; Phosphatidylinositol 3-Kinases; Pituitary Gland, Anterior; Pituitary Neoplasms; STAT Transcription Factors
Año:2017
Volumen:58
Número:4
Página de inicio:R241
Página de fin:R253
DOI: http://dx.doi.org/10.1530/JME-17-0026
Título revista:Journal of Molecular Endocrinology
Título revista abreviado:J. Mol. Endocrinol.
ISSN:09525041
CODEN:JMLEE
CAS:mitogen activated protein kinase kinase, 142805-58-1; phosphatidylinositol 3 kinase, 115926-52-8; IL6 protein, human; Interleukin-6; Mitogen-Activated Protein Kinase Kinases; Phosphatidylinositol 3-Kinases; STAT Transcription Factors
Registro:https://bibliotecadigital.exactas.uba.ar/collection/paper/document/paper_09525041_v58_n4_pR241_Sapochnik

Referencias:

  • Acosta, J.C., O’Loghlen, A., Banito, A., Guijarro, M.V., Augert, A., Raguz, S., Fumagalli, M., Popov, N., Chemokine signaling via the CXCR2 receptor reinforces senescence (2008) Cell, 133, pp. 1006-1018
  • Alexandraki, K.I., Munayem Khan, M., Chahal, H.S., Dalantaeva, N.S., Trivellin, G., Berney, D.M., Caron, P., Jordan, S., Oncogene-induced senescence in pituitary adenomas and carcinomas (2012) Hormones, 11, pp. 297-307
  • Ancrile, B., Lim, K.H., Counter, C.M., Oncogenic Ras-induced secretion of IL6 is required for tumorigenesis (2007) Genes and Development, 21, pp. 1714-1719
  • Andoniadou, C.L., Matsushima, D., Mousavy Gharavy, S.N., Signore, M., Mackintosh, A.I., Schaeffer, M., Gaston-Massuet, C., Le Tissier, P., Sox2(+) stem/progenitor cells in the adult mouse pituitary support organ homeostasis and have tumor-inducing potential (2013) Cell Stem Cell, 13, pp. 433-445
  • Andus, T., Geiger, T., Hirano, T., Northoff, H., Ganter, U., Bauer, J., Kishimoto, T., Heinrich, P.C., Recombinant human B cell stimulatory factor 2 (BSF-2/IFN-beta 2) regulates beta-fibrinogen and albumin mRNA levels in Fao-9 cells (1987) FEBS Letters, 221, pp. 18-22
  • Arzt, E., Gp130 cytokine signaling in the pituitary gland: A paradigm for cytokine-neuro-endocrine pathways (2001) Journal of Clinical Investigation, 108, pp. 1729-1733
  • Arzt, E., Buric, R., Stelzer, G., Stalla, J., Sauer, J., Renner, U., Stalla, G.K., Interleukin involvement in anterior pituitary cell growth regulation: Effects of IL-2 and IL-6 (1993) Endocrinology, 132, pp. 459-467
  • Arzt, E., Pereda, M.P., Castro, C.P., Pagotto, U., Renner, U., Stalla, G.K., Pathophysiological role of the cytokine network in the anterior pituitary gland (1999) Frontiers in Neuroendocrinology, 20, pp. 71-95
  • Arzt, E., Chesnokova, V., Stalla, G.K., Melmed, S., Pituitary adenoma growth: A model for cellular senescence and cytokine action (2009) Cell Cycle, 8, pp. 677-678
  • Baker, D.J., Sedivy, J.M., Probing the depths of cellular senescence (2013) Journal of Cell Biology, 202, pp. 11-13
  • Barrett, E., Brothers, S., Wahlestedt, C., Beurel, E., I-BET151 selectively regulates IL-6 production (2014) Biochimica Et Biophysica Acta, 1842, pp. 1549-1555
  • Bethin, K.E., Vogt, S.K., Muglia, L.J., Interleukin-6 is an essential, corticotropin-releasing hormone-independent stimulator of the adrenal axis during immune system activation (2000) PNAS, 97, pp. 9317-9322
  • Borg, S.A., Kerry, K.E., Baxter, L., Royds, J.A., Jones, T.H., Expression of interleukin-6 and its effects on growth of HP75 human pituitary tumor cells (2003) Journal of Clinical Endocrinology and Metabolism, 88, pp. 4938-4944
  • Bowman, T., Garcia, R., Turkson, J., Jove, R., STATs in oncogenesis (2000) Oncogene, 19, pp. 2474-2488
  • Braig, M., Lee, S., Loddenkemper, C., Rudolph, C., Peters, A.H., Schlegelberger, B., Stein, H., Schmitt, C.A., Oncogene-induced senescence as an initial barrier in lymphoma development (2005) Nature, 436, pp. 660-665
  • Castro, C.P., Giacomini, D., Nagashima, A.C., Onofri, C., Graciarena, M., Kobayashi, K., Paez-Pereda, M., Arzt, E., Reduced expression of the cytokine transducer gp130 inhibits hormone secretion, cell growth, and tumor development of pituitary lactosomatotrophic GH3 cells (2003) Endocrinology, 144, pp. 693-700
  • Chalaris, A., Garbers, C., Rabe, B., Rose-John, S., Scheller, J., The soluble Interleukin 6 receptor: Generation and role in inflammation and cancer (2011) European Journal of Cell Biology, 90, pp. 484-494
  • Chen, Z., Trotman, L.C., Shaffer, D., Lin, H.K., Dotan, Z.A., Niki, M., Koutcher, J.A., Gerald, W., Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis (2005) Nature, 436, pp. 725-730
  • Chesnokova, V., Kovacs, K., Castro, A.V., Zonis, S., Melmed, S., Pituitary hypoplasia in Pttg-/-mice is protective for Rb+/-pituitary tumorigenesis (2005) Molecular Endocrinology, 19, pp. 2371-2379
  • Chesnokova, V., Zonis, S., Rubinek, T., Yu, R., Ben-Shlomo, A., Kovacs, K., Wawrowsky, K., Melmed, S., Senescence mediates pituitary hypoplasia and restrains pituitary tumor growth (2007) Cancer Research, 67, pp. 10564-10572
  • Chesnokova, V., Zonis, S., Kovacs, K., Ben-Shlomo, A., Wawrowsky, K., Bannykh S & Melmed S 2008 p21(Cip1) restrains pituitary tumor growth PNAS, 105, pp. 17498-17503
  • Chesnokova, V., Zonis, S., Zhou, C., Ben-Shlomo, A., Wawrowsky, K., Toledano, Y., Tong, Y., Melmed, S., Lineage-specific restraint of pituitary gonadotroph cell adenoma growth (2011) Plos ONE, 6
  • Cichowski, K., Hahn, W.C., Unexpected pieces to the senescence puzzle (2008) Cell, 133, pp. 958-961
  • Clayton, R.N., Farrell, W.E., Pituitary tumour clonality revisited (2004) Frontiers of Hormone Research, 32, pp. 186-204
  • Colao, A., Ochoa, A.S., Auriemma, R.S., Faggiano, A., Pivonello, R., Lombardi, G., Pituitary carcinomas (2010) Frontiers of Hormone Research, 38, pp. 94-108
  • Collado, M., Gil, J., Efeyan, A., Guerra, C., Schuhmacher, A.J., Barradas, M., Benguria, A., Barbacid, M., Tumour biology: Senescence in premalignant tumours (2005) Nature, 436, p. 642
  • Coppe, J.P., Patil, C.K., Rodier, F., Sun, Y., Munoz, D.P., Goldstein, J., Nelson, P.S., Campisi, J., Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor (2008) Plos Biology, 6, pp. 2853-2868
  • Coppe, J.P., Boysen, M., Sun, C.H., Wong, B.J., Kang, M.K., Park, N.H., Desprez, P.Y., Krtolica, A., A role for fibroblasts in mediating the effects of tobacco-induced epithelial cell growth and invasion (2008) Molecular Cancer Research, 6, pp. 1085-1098
  • Coppe, J.P., Desprez, P.Y., Krtolica, A., Campisi, J., The senescence-associated secretory phenotype: The dark side of tumor suppression (2010) Annual Review of Pathology, 5, pp. 99-118
  • Courtois-Cox, S., Genther Williams, S.M., Reczek, E.E., Johnson, B.W., McGillicuddy, L.T., Johannessen, C.M., Hollstein, P.E., Cichowski, K., A negative feedback signaling network underlies oncogene-induced senescence (2006) Cancer Cell, 10, pp. 459-472
  • D’Adda Di Fagagna, F., Living on a break: Cellular senescence as a DNA-damage response (2008) Nature Reviews Cancer, 8, pp. 512-522
  • Darnell, J.E., Jr., STATs and gene regulation (1997) Science, 277, pp. 1630-1635
  • Dinarello, C.A., Historical insights into cytokines (2007) European Journal of Immunology, 37, pp. S34-S45
  • Donangelo, I., Gutman, S., Horvath, E., Kovacs, K., Wawrowsky, K., Mount, M., Melmed, S., Pituitary tumor transforming gene overexpression facilitates pituitary tumor development (2006) Endocrinology, 147, pp. 4781-4791
  • Donangelo, I., Ren, S.G., Eigler, T., Svendsen, C., Melmed, S., Sca1(+) murine pituitary adenoma cells show tumor-growth advantage (2014) Endocrine-Related Cancer, 21, pp. 203-216
  • Dou, L., Zhao, T., Wang, L., Huang, X., Jiao, J., Gao, D., Zhang, H., Wang, S., 2013 miR-200s contribute to interleukin-6 (IL-6)-induced insulin resistance in hepatocytes Journal of Biological Chemistry, 288, pp. 22596-22606
  • Dranoff, G., Cytokines in cancer pathogenesis and cancer therapy (2004) Nature Reviews Cancer, 4, pp. 11-22
  • Dworakowska, D., Grossman, A.B., The pathophysiology of pituitary adenomas (2009) Best Practice and Research Clinical Endocrinology and Metabolism, 23, pp. 525-541
  • Dworakowska, D., Grossman, A.B., The molecular pathogenesis of pituitary tumors: Implications for clinical management (2012) Minerva Endocrinologica, 37, pp. 157-172
  • Farnoud, M.R., Kujas, M., Derome, P., Racadot, J., Peillon, F., Li, J.Y., Interactions between normal and tumoral tissues at the boundary of human anterior pituitary adenomas. An immunohistochemical study (1994) Virchows Archiv, 424, pp. 75-82
  • Farrell, W.E., Pituitary tumours: Findings from whole genome analyses (2006) Endocrine-Related Cancer, 13, pp. 707-716
  • Farrell, W.E., Clayton, R.N., Molecular genetics of pituitary tumours (1998) Trends in Endocrinology and Metabolism, 9, pp. 20-26
  • Fauquier, T., Rizzoti, K., Dattani, M., Lovell-Badge, R., Robinson, I.C., SOX2-expressing progenitor cells generate all of the major cell types in the adult mouse pituitary gland (2008) PNAS, 105, pp. 2907-2912
  • Florio, T., Adult pituitary stem cells: From pituitary plasticity to adenoma development (2011) Neuroendocrinology, 94, pp. 265-277
  • Garcia-Lavandeira, M., Quereda, V., Flores, I., Saez, C., Diaz-Rodriguez, E., Japon, M.A., Ryan, A.K., Malumbres, M., A GRFa2/Prop1/stem (GPS) cell niche in the pituitary (2009) Plos ONE, 4
  • Garcia-Lavandeira, M., Diaz-Rodriguez, E., Bahar, D., Garcia-Rendueles, A.R., Rodrigues, J.S., Dieguez, C., Alvarez, C.V., Pituitary cell turnover: From adult stem cell recruitment through differentiation to death (2015) Neuroendocrinology, 101, pp. 175-192
  • Garg, M., Potter, J.A., Abrahams, V.M., Identification of microRNAs that regulate TLR2-mediated trophoblast apoptosis and inhibition of IL-6 mRNA (2013) Plos ONE, 8
  • Gaston-Massuet, C., Oniadou, C.L., Signore, M., Jayakody, S.A., Charolidi, N., Kyeyune, R., Vernay, B., Le Tissier, P., Increased Wingless (Wnt) signaling in pituitary progenitor/ stem cells gives rise to pituitary tumors in mice and humans (2011) PNAS, 108, pp. 11482-11487
  • Gerlo, S., Haegeman, G., Vanden Berghe, W., Transcriptional regulation of autocrine IL-6 expression in multiple myeloma cells (2008) Cellular Signalling, 20, pp. 1489-1496
  • Gleiberman, A.S., Michurina, T., Encinas, J.M., Roig, J.L., Krasnov, P., Balordi, F., Fishell, G., Enikolopov, G., Genetic approaches identify adult pituitary stem cells (2008) PNAS, 105, pp. 6332-6337
  • Gloddek, J., Pagotto, U., Paez Pereda, M., Arzt, E., Stalla, G.K., Renner, U., Pituitary adenylate cyclase-activating polypeptide, interleukin-6 and glucocorticoids regulate the release of vascular endothelial growth factor in pituitary folliculostellate cells (1999) Journal of Endocrinology, 160, pp. 483-490
  • Goel, V.K., Ibrahim, N., Jiang, G., Singhal, M., Fee, S., Flotte, T., Westmoreland, S., Haluska, F.G., Melanocytic nevus-like hyperplasia and melanoma in transgenic BRAFV600E mice (2009) Oncogene, 28, pp. 2289-2298
  • Graciarena, M., Carbia-Nagashima, A., Onofri, C., Perez-Castro, C., Giacomini, D., Renner, U., Stalla, G.K., Arzt, E., Involvement of the gp130 cytokine transducer in MtT/S pituitary somatotroph tumour development in an autocrine-paracrine model (2004) European Journal of Endocrinology, 151, pp. 595-604
  • Green, M.R., Senescence: Not just for tumor suppression (2008) Cell, 134, pp. 562-564
  • Gruys, E., Toussaint, M.J., Niewold, T.A., Koopmans, S.J., Acute phase reaction and acute phase proteins (2005) Journal of Zheijang University Science B, 6, pp. 1045-1056
  • Hanisch, A., Dieterich, K.D., Dietzmann, K., Ludecke, K., Buchfelder, M., Fahlbusch, R., Lehnert, H., Expression of members of the interleukin-6 family of cytokines and their receptors in human pituitary and pituitary adenomas (2000) Journal of Clinical Endocrinology and Metabolism, 85, pp. 4411-4414
  • He, Y., Sun, X., Huang, C., Long, X.R., Lin, X., Zhang, L., Lv, X.W., Li, J., MiR-146a regulates IL-6 production in lipopolysaccharide-induced RAW264.7 macrophage cells by inhibiting Notch1 (2014) Inflammation, 37, pp. 71-82
  • Heinrich, P.C., Behrmann, I., Haan, S., Hermanns, H.M., Muller-Newen, G., Schaper, F., Principles of interleukin (IL)-6-type cytokine signalling and its regulation (2003) Biochemical Journal, 374, pp. 1-20
  • Hirano, T., Revisiting the 1986 molecular cloning of interleukin 6 (2014) Frontiers in Immunology, 5, p. 456
  • Hirano, T., Yasukawa, K., Harada, H., Taga, T., Watanabe, Y., Matsuda, T., Kashiwamura, S., Iwamatsu, A., Complementary DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce immunoglobulin (1986) Nature, 324, pp. 73-76
  • Hirano, T., Nakajima, K., Hibi, M., Signaling mechanisms through gp130: A model of the cytokine system (1997) Cytokine and Growth Factor Reviews, 8, pp. 241-252
  • Hirano, T., Ishihara, K., Hibi, M., Roles of STAT3 in mediating the cell growth, differentiation and survival signals relayed through the IL-6 family of cytokine receptors (2000) Oncogene, 19, pp. 2548-2556
  • Hodes, G.E., Pfau, M.L., Leboeuf, M., Golden, S.A., Christoffel, D.J., Bregman, D., Rebusi, N., Warren, B.L., Individual differences in the peripheral immune system promote resilience versus susceptibility to social stress (2014) PNAS, 111, pp. 16136-16141
  • Hofler, H., Walter, G.F., Denk, H., Immunohistochemistry of folliculo-stellate cells in normal human adenohypophyses and in pituitary adenomas (1984) Acta Neuropathologica, 65, pp. 35-40
  • Hosoyama, T., Nishijo, K., Garcia, M.M., Schaffer, B.S., Ohshima-Hosoyama, S., Prajapati, S.I., Davis, M.D., Marks, D.L., A postnatal Pax7 progenitor gives rise to pituitary adenomas (2010) Genes and Cancer, 1, pp. 388-402
  • Hunter, C.A., Jones, S.A., IL-6 as a keystone cytokine in health and disease (2015) Nature Immunology, 16, pp. 448-457
  • Iwasaki, H., Takeuchi, O., Teraguchi, S., Matsushita, K., Uehata, T., Kuniyoshi, K., Satoh, T., Standley, D.M., The IkappaB kinase complex regulates the stability of cytokine-encoding mRNA induced by TLR-IL-1R by controlling degradation of regnase-1 (2011) Nature Immunology, 12, pp. 1167-1175
  • Jiang, X., Zhang, X., The molecular pathogenesis of pituitary adenomas: An update (2013) Endocrinology and Metabolism, 28, pp. 245-254
  • Jones, T.H., Daniels, M., James, R.A., Justice, S.K., McCorkle, R., Price, A., Kendall-Taylor, P., Weetman, A.P., Production of bioactive and immunoreactive interleukin-6 (IL-6) and expression of IL-6 messenger ribonucleic acid by human pituitary adenomas (1994) Journal of Clinical Endocrinology and Metabolism, 78, pp. 180-187
  • Jones, S.A., Horiuchi, S., Topley, N., Yamamoto, N., Fuller, G.M., The soluble interleukin 6 receptor: Mechanisms of production and implications in disease (2001) FASEB Journal, 15, pp. 43-58
  • Jones, S.A., Scheller, J., Rose-John, S., Therapeutic strategies for the clinical blockade of IL-6/gp130 signaling (2011) Journal of Clinical Investigation, 121, pp. 3375-3383
  • Kim, D., Pemberton, H., Stratford, A.L., Buelaert, K., Watkinson, J.C., Lopes, V., Franklyn, J.A., McCabe, C.J., Pituitary tumour transforming gene (PTTG) induces genetic instability in thyroid cells (2005) Oncogene, 24, pp. 4861-4866
  • Kim, D.S., Franklyn, J.A., Smith, V.E., Stratford, A.L., Pemberton, H.N., Warfield, A., Watkinson, J.C., McCabe, C.J., Securin induces genetic instability in colorectal cancer by inhibiting double-stranded DNA repair activity (2007) Carcinogenesis, 28, pp. 749-759
  • Kishimoto, T., Interleukin-6: From basic science to medicine-40 years in immunology (2005) Annual Review of Immunology, 23, pp. 1-21
  • Klimpel, G.R., Soluble factor(S) from LPS-activated macrophages induce cytotoxic T cell differentiation from alloantigen-primed spleen cells (1980) Journal of Immunology, 125, pp. 1243-1249
  • Kojima, H., Inoue, T., Kunimoto, H., Nakajima, K., IL-6-STAT3 signaling and premature senescence (2013) JAK-STAT, 2
  • Kopczak, A., Renner, U., Karl Stalla, G., Advances in understanding pituitary tumors (2014) F1000prime Reports, 6, p. 5
  • Kovacs, E., Investigation of interleukin-6 (IL-6), soluble IL-6 receptor (sIL-6R) and soluble gp130 (sgp130) in sera of cancer patients (2001) Biomedicine and Pharmacotherapy, 55, pp. 391-396
  • Kraakman, M.J., Kammoun, H.L., Allen, T.L., Deswaerte, V., Henstridge, D.C., Estevez, E., Matthews, V.B., Murphy, A.J., Blocking IL-6 trans-signaling prevents high-fat diet-induced adipose tissue macrophage recruitment but does not improve insulin resistance (2015) Cell Metabolism, 21, pp. 403-416
  • Krtolica, A., Parrinello, S., Lockett, S., Desprez, P.Y., Campisi, J., Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging (2001) PNAS, 98, pp. 12072-12077
  • Kuilman, T., Peeper, D.S., Senescence-messaging secretome: SMS-ing cellular stress (2009) Nature Reviews Cancer, 9, pp. 81-94
  • Kuilman, T., Michaloglou, C., Vredeveld, L.C., Douma, S., van Doorn, R., Desmet, C.J., Aarden, L.A., Peeper, D.S., Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network (2008) Cell, 133, pp. 1019-1031
  • Kumari, N., Dwarakanath, B.S., Das, A., Bhatt, A.N., Role of interleukin-6 in cancer progression and therapeutic resistance (2016) Tumour Biology, 37, pp. 11553-11572
  • Lake, M.G., Krook, L.S., Cruz, S.V., Pituitary adenomas: An overview (2013) American Family Physician, 88, pp. 319-327
  • Lazzerini Denchi, E., Helin, K., E2F1 is crucial for E2F-dependent apoptosis (2005) EMBO Reports, 6, pp. 661-668
  • Lecot, P., Alimirah, F., Desprez, P.Y., Campisi, J., Wiley, C., Context-dependent effects of cellular senescence in cancer development (2016) British Journal of Cancer, 114, pp. 1180-1184
  • Lee, K.A., Hai, T.Y., Sivaraman, L., Thimmappaya, B., Hurst, H.C., Jones, N.C., Green, M.R., A cellular protein, activating transcription factor, activates transcription of multiple E1A-inducible adenovirus early promoters (1987) PNAS, 84, pp. 8355-8359
  • Lehmann, U., Schmitz, J., Weissenbach, M., Sobota, R.M., Hortner, M., Friederichs, K., Behrmann, I., Schneider-Mergener, J., SHP2 and SOCS3 contribute to Tyr-759-dependent attenuation of interleukin-6 signaling through gp130 (2003) Journal of Biological Chemistry, 278, pp. 661-671
  • Levy, A., Lightman, S., Molecular defects in the pathogenesis of pituitary tumours (2003) Frontiers in Neuroendocrinology, 24, pp. 94-127
  • Liu, B., Liao, J., Rao, X., Kushner, S.A., Chung, C.D., Chang, D.D., Shuai, K., Inhibition of Stat1-mediated gene activation by PIAS1 (1998) PNAS, 95, pp. 10626-10631
  • Masuda, K., Ripley, B., Nishimura, R., Mino, T., Takeuchi, O., Shioi, G., Kiyonari, H., Kishimoto, T., Arid5a controls IL-6 mRNA stability, which contributes to elevation of IL-6 level in vivo (2013) PNAS, 110, pp. 9409-9414
  • Matsumoto, H., Ishibashi, Y., Ohtaki, T., Hasegawa, Y., Koyama, C., Inoue, K., Newly established murine pituitary folliculo-stellate-like cell line (TtT/GF) secretes potent pituitary glandular cell survival factors, one of which corresponds to metalloproteinase inhibitor (1993) Biochemical and Biophysical Research Communications, 194, pp. 909-915
  • Matsusaka, T., Fujikawa, K., Nishio, Y., Mukaida, N., Matsushima, K., Kishimoto, T., Akira, S., Transcription factors NF-IL6 and NF-kappa B synergistically activate transcription of the inflammatory cytokines, interleukin 6 and interleukin 8 (1993) PNAS, 90, pp. 10193-10197
  • McFarland-Mancini, M.M., Funk, H.M., Paluch, A.M., Zhou, M., Giridhar, P.V., Mercer, C.A., Kozma, S.C., Drew, A.F., Differences in wound healing in mice with deficiency of IL-6 versus IL-6 receptor (2010) Journal of Immunology, 184, pp. 7219-7228
  • McInnes, I.B., Schett, G., Cytokines in the pathogenesis of rheumatoid arthritis (2007) Nature Reviews Immunology, 7, pp. 429-442
  • Melmed, S., Update in pituitary disease (2008) Journal of Clinical Endocrinology and Metabolism, 93, pp. 331-338
  • Melmed, S., Pathogenesis of pituitary tumors (2011) Nature Reviews Endocrinology, 7, pp. 257-266
  • Melmed, S., Pituitary tumors (2015) Endocrinology Metabolism Clinics of North America, 44, pp. 1-9
  • Mertens, F., Gremeaux, L., Chen, J., Fu, Q., Willems, C., Roose, H., Govaere, O., Becu-Villalobos, D., Pituitary tumors contain a side population with tumor stem cell-associated characteristics (2015) Endocrine-Related Cancer, 22, pp. 481-504
  • Michaloglou, C., Vredeveld, L.C., Soengas, M.S., Denoyelle, C., Kuilman, T., van Der Horst, C.M., Majoor, D.M., Peeper, D.S., BRAFE600-associated senescence-like cell cycle arrest of human naevi (2005) Nature, 436, pp. 720-724
  • Mihara, M., Hashizume, M., Yoshida, H., Suzuki, M., Shiina, M., IL-6/IL-6 receptor system and its role in physiological and pathological conditions (2012) Clinical Science, 122, pp. 143-159
  • Muñoz-Espín, D., Serrano, M., Cellular senescence: From physiology to pathology (2014) Nature Reviews Molecular Cell Biology, 15, pp. 482-496
  • Naka, T., Narazaki, M., Hirata, M., Matsumoto, T., Minamoto, S., Aono, A., Nishimoto, N., Yoshizaki, K., Structure and function of a new STAT-induced STAT inhibitor (1997) Nature, 387, pp. 924-929
  • Nakajima, K., Yamanaka, Y., Nakae, K., Kojima, H., Ichiba, M., Kiuchi, N., Kitaoka, T., Hirano, T., A central role for Stat3 in IL-6-induced regulation of growth and differentiation in M1 leukemia cells (1996) EMBO Journal, 15, pp. 3651-3658
  • Narita, M., Lowe, S.W., Senescence comes of age (2005) Nature Medicine, 11, pp. 920-922
  • Paez-Pereda, M., Giacomini, D., Refojo, D., Nagashima, A.C., Hopfner, U., Grubler, Y., Chervin, A., Hentges, S.T., Involvement of bone morphogenetic protein 4 (BMP-4) in pituitary prolactinoma pathogenesis through a Smad/estrogen receptor crosstalk (2003) PNAS, 100, pp. 1034-1039
  • Paez-Pereda, M., Kuchenbauer, F., Arzt, E., Stalla, G.K., Regulation of pituitary hormones and cell proliferation by components of the extracellular matrix (2005) Brazilian Journal of Medical and Biological Research, 38, pp. 1487-1494
  • Pei, L., Melmed, S., Isolation and characterization of a pituitary tumor-transforming gene (PTTG) (1997) Molecular Endocrinology, 11, pp. 433-441
  • Pereda, M.P., Goldberg, V., Chervin, A., Carrizo, G., Molina, A., Rada, J., Sauer, J., Arzt, E., Interleukin-2 (IL-2) and IL-6 regulate c-fos protooncogene expression in human pituitary adenoma explants (1996) Molecular and Cellular Endocrinology, 124, pp. 33-42
  • Pereda, M.P., Lohrer, P., Kovalovsky, D., Perez Castro, C., Goldberg, V., Losa, M., Chervin, A., Stalla, G.K., Interleukin-6 is inhibited by glucocorticoids and stimulates ACTH secretion and POMC expression in human corticotroph pituitary adenomas (2000) Experimental and Clinical Endocrinology and Diabetes, 108, pp. 202-207
  • Perez Castro, C., Nagashima, A.C., Pereda, M.P., Goldberg, V., Chervin, A., Largen, P., Renner, U., Arzt, E., The gp130 cytokines interleukin-11 and ciliary neurotropic factor regulate through specific receptors the function and growth of lactosomatotropic and folliculostellate pituitary cell lines (2000) Endocrinology, 141, pp. 1746-1753
  • Perez Castro, C., Carbia Nagashima, A., Paez Pereda, M., Goldberg, V., Chervin, A., Carrizo, G., Molina, H., Arzt, E., Effects of the gp130 cytokines ciliary neurotropic factor (CNTF) and interleukin-11 on pituitary cells: CNTF receptors on human pituitary adenomas and stimulation of prolactin and GH secretion in normal rat anterior pituitary aggregate cultures (2001) Journal of Endocrinology, 169, pp. 539-547
  • Perez-Castro, C., Renner, U., Haedo, M.R., Stalla, G.K., Arzt, E., Cellular and molecular specificity of pituitary gland physiology (2012) Physiological Reviews, 92, pp. 1-38
  • Peters, M., Jacobs, S., Ehlers, M., Vollmer, P., Mullberg, J., Wolf, E., Brem, G., Rose-John, S., The function of the soluble interleukin 6 (IL-6) receptor in vivo: Sensitization of human soluble IL-6 receptor transgenic mice towards IL-6 and prolongation of the plasma half-life of IL-6 (1996) Journal of Experimental Medicine, 183, pp. 1399-1406
  • Ray, D., Melmed, S., Pituitary cytokine and growth factor expression and action (1997) Endocrine Reviews, 18, pp. 206-228
  • Reincke, M., Sbiera, S., Hayakawa, A., Theodoropoulou, M., Osswald, A., Beuschlein, F., Meitinger, T., Saeki, Y., Mutations in the deubiquitinase gene USP8 cause Cushing’s disease (2015) Nature Genetics, 47, pp. 31-38
  • Renner, U., Pagotto, U., Arzt, E., Stalla, G.K., Autocrine and paracrine roles of polypeptide growth factors, cytokines and vasogenic substances in normal and tumorous pituitary function and growth: A review (1996) European Journal of Endocrinology, 135, pp. 515-532
  • Renner, U., Gloddek, J., Arzt, E., Inoue, K., Stalla, G.K., Interleukin-6 is an autocrine growth factor for folliculostellate-like TtT/GF mouse pituitary tumor cells (1997) Experimental and Clinical Endocrinology and Diabetes, 105, pp. 345-352
  • Renner, U., Gloddek, J., Pereda, M.P., Arzt, E., Stalla, G.K., Regulation and role of intrapituitary IL-6 production by folliculostellate cells (1998) Domestic Animal Endocrinology, 15, pp. 353-362
  • Rohleder, N., Aringer, M., Boentert, M., Role of interleukin-6 in stress, sleep, and fatigue (2012) Annals of the New York Academy of Sciences, 1261, pp. 88-96
  • Rose-John, S., IL-6 trans-signaling via the soluble IL-6 receptor: Importance for the pro-inflammatory activities of IL-6 (2012) International Journal of Biological Sciences, 8, pp. 1237-1247
  • Rose-John, S., Heinrich, P.C., Soluble receptors for cytokines and growth factors: Generation and biological function (1994) Biochemical Journal, 300, pp. 281-290
  • Rose-John, S., Scheller, J., Elson, G., Jones, S.A., Interleukin-6 biology is coordinated by membrane-bound and soluble receptors: Role in inflammation and cancer (2006) Journal of Leukocyte Biology, 80, pp. 227-236
  • Rossato, M., Curtale, G., Tamassia, N., Castellucci, M., Mori, L., Gasperini, S., Mariotti, B., Cassatella, M.A., IL-10-induced microRNA-187 negatively regulates TNF-alpha, IL-6, and IL-12p40 production in TLR4-stimulated monocytes (2012) PNAS, 109, pp. E3101-E3110
  • Ruggeri, R.M., Villari, D., Simone, A., Scarfi, R., Attard, M., Orlandi, F., Barresi, G., Benvenga, S., Co-expression of interleukin-6 (IL-6) and interleukin-6 receptor (IL-6R) in thyroid nodules is associated with co-expression of CD30 ligand/CD30 receptor (2002) Journal of Endocrinological Investigation, 25, pp. 959-966
  • Sapochnik, M., Haedo, M.R., Fuertes, M., Ajler, P., Carrizo, G., Cervio, A., Sevlever, G., Arzt, E., Autocrine IL-6 mediates pituitary tumor senescence (2016) Oncotarget, 8, pp. 4690-4702
  • Scheithauer, B.W., Gaffey, T.A., Lloyd, R.V., Sebo, T.J., Kovacs, K.T., Horvath, E., Yapicier, O., Kuroki, T., Pathobiology of pituitary adenomas and carcinomas (2006) Neurosurgery, 59, pp. 341-353. , discussion 341-353
  • Scheller, J., Rose-John, S., Interleukin-6 and its receptor: From bench to bedside (2006) Medical Microbiology and Immunology, 195, pp. 173-183
  • Scheller, J., Chalaris, A., Schmidt-Arras, D., Rose-John, S., The pro-and anti-inflammatory properties of the cytokine interleukin-6 (2011) Biochimica Et Biophysica Acta, 1813, pp. 878-888
  • Schett, G., Elewaut, D., McInnes, I.B., Dayer, J.M., Neurath, M.F., How cytokine networks fuel inflammation: Toward a cytokine-based disease taxonomy (2013) Nature Medicine, 19, pp. 822-824
  • Simonetti, S., Serrano, C., Hernandez-Losa, J., Bague, S., Orellana, R., Valverde, C., Lleonart, M.E., Ramon Y Cajal, S., Schwannomas, benign tumors with a senescent phenotype (2014) Histology and Histopathology, 29, pp. 721-730
  • Spangelo, B.L., Judd, A.M., Isakson, P.C., Macleod, R.M., Interleukin-1 stimulates interleukin-6 release from rat anterior pituitary cells in vitro (1991) Endocrinology, 128, pp. 2685-2692
  • Sparmann, A., Bar-Sagi, D., Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis (2004) Cancer Cell, 6, pp. 447-458
  • Spooren, A., Kooijman, R., Lintermans, B., van Craenenbroeck, K., Vermeulen, L., Haegeman, G., Gerlo, S., Cooperation of NFkappaB and CREB to induce synergistic IL-6 expression in astrocytes (2010) Cellular Signalling, 22, pp. 871-881
  • Starr, R., Willson, T.A., Viney, E.M., Murray, L.J., Rayner, J.R., Jenkins, B.J., Gonda, T.J., Nicola, N.A., A family of cytokine-inducible inhibitors of signalling (1997) Nature, 387, pp. 917-921
  • Sun, Y., Sun, J., Tomomi, T., Nieves, E., Mathewson, N., Tamaki, H., Evers, R., Reddy, P., PU.1-dependent transcriptional regulation of miR-142 contributes to its hematopoietic cell-specific expression and modulation of IL-6 (2013) Journal of Immunology, 190, pp. 4005-4013
  • Tichomirowa, M., Theodoropoulou, M., Lohrer, P., Schaaf, L., Losa, M., Uhl, E., Lange, M., Renner, U., Bacterial endotoxin (Lipopolysaccharide) stimulates interleukin-6 production and inhibits growth of pituitary tumour cells expressing the toll-like receptor 4 (2005) Journal of Neuroendocrinology, 17, pp. 152-160
  • Trivellin, G., Daly, A.F., Faucz, F.R., Yuan, B., Rostomyan, L., Larco, D.O., Schernthaner-Reiter, M.H., Caberg, J.H., Gigantism and acromegaly due to Xq26 microduplications and GPR101 mutation (2014) New England Journal of Medicine, 371, pp. 2363-2374
  • Ueta, Y., Levy, A., Chowdrey, H.S., Lightman, S.L., S-100 antigen-positive folliculostellate cells are not the source of IL-6 gene expression in human pituitary adenomas (1995) Journal of Neuroendocrinology, 7, pp. 467-474
  • Vajtai, I., Kappeler, A., Sahli, R., Folliculo-stellate cells of ‘true dendritic’ type are involved in the inflammatory microenvironment of tumor immunosurveillance of pituitary adenomas (2007) Diagnostic Pathology, 2, p. 20
  • Vandeva, S., Jaffrain-Rea, M.L., Daly, A.F., Tichomirowa, M., Zacharieva, S., Beckers, A., The genetics of pituitary adenomas (2010) Best Practice and Research Clinical Endocrinology and Metabolism, 24, pp. 461-476
  • Vankelecom, H., Gremeaux, L., Stem cells in the pituitary gland: A burgeoning field (2010) General and Comparative Endocrinology, 166, pp. 478-488
  • Vankelecom, H., Carmeliet, P., van Damme, J., Billiau, A., Denef, C., Production of interleukin-6 by folliculo-stellate cells of the anterior pituitary gland in a histiotypic cell aggregate culture system (1989) Neuroendocrinology, 49, pp. 102-106
  • Ventura, A., Kirsch, D.G., McLaughlin, M.E., Tuveson, D.A., Grimm, J., Lintault, L., Newman, J., Jacks, T., Restoration of p53 function leads to tumour regression in vivo (2007) Nature, 445, pp. 661-665
  • Vierimaa, O., Georgitsi, M., Lehtonen, R., Vahteristo, P., Kokko, A., Raitila, A., Tuppurainen, K., Paschke, R., Pituitary adenoma predisposition caused by germline mutations in the AIP gene (2006) Science, 312, pp. 1228-1230
  • Vizioli, M.G., Santos, J., Pilotti, S., Mazzoni, M., Anania, M.C., Miranda, C., Pagliardini, S., Greco, A., Oncogenic RAS-induced senescence in human primary thyrocytes: Molecular effectors and inflammatory secretome involved (2014) Oncotarget, 5, pp. 8270-8283
  • Vlotides, G., Eigler, T., Melmed, S., Pituitary tumor-transforming gene: Physiology and implications for tumorigenesis (2007) Endocrine Reviews, 28, pp. 165-186
  • Wolf, J., Rose-John, S., Garbers, C., Interleukin-6 and its receptors: A highly regulated and dynamic system (2014) Cytokine, 70, pp. 11-20
  • Woloski, B.M., Fuller, G.M., Identification and partial characterization of hepatocyte-stimulating factor from leukemia cell lines: Comparison with interleukin 1 (1985) PNAS, 82, pp. 1443-1447
  • Xiang, M., Birkbak, N.J., Vafaizadeh, V., Walker, S.R., Yeh, J.E., Liu, S., Kroll, Y., Groner, B., STAT3 induction of miR-146b forms a feedback loop to inhibit the NF-kappaB to IL-6 signaling axis and STAT3-driven cancer phenotypes (2014) Science Signaling, 7, pp. ra11
  • Xue, W., Zender, L., Miething, C., Dickins, R.A., Hernando, E., Krizhanovsky, V., Cordon-Cardo, C., Lowe, S.W., Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas (2007) Nature, 445, pp. 656-660
  • Yang, X., Liang, L., Zhang, X.F., Jia, H.L., Qin, Y., Zhu, X.C., Gao, X.M., Sheng, Y.Y., MicroRNA-26a suppresses tumor growth and metastasis of human hepatocellular carcinoma by targeting interleukin-6-Stat3 pathway (2013) Hepatology, 58, pp. 158-170
  • Yao, X., Huang, J., Zhong, H., Shen, N., Faggioni, R., Fung, M., Yao, Y., Targeting interleukin-6 in inflammatory autoimmune diseases and cancers (2014) Pharmacology and Therapeutics, 141, pp. 125-139
  • Yasukawa, K., Hirano, T., Watanabe, Y., Muratani, K., Matsuda, T., Nakai, S., Kishimoto, T., Structure and expression of human B cell stimulatory factor-2 (BSF-2/IL-6) gene (1987) EMBO Journal, 6, pp. 2939-2945
  • Yoshida, K., Taga, T., Saito, M., Suematsu, S., Kumanogoh, A., Tanaka, T., Fujiwara, H., Nakahata, T., Targeted disruption of gp130, a common signal transducer for the interleukin 6 family of cytokines, leads to myocardial and hematological disorders (1996) PNAS, 93, pp. 407-411
  • Yoshizaki, K., Nakagawa, T., Fukunaga, K., Tseng, L.T., Yamamura, Y., Kishimoto, T., Isolation and characterization of B cell differentiation factor (BCDF) secreted from a human B lymphoblastoid cell line (1984) Journal of Immunology, 132, pp. 2948-2954
  • Young, A.R., Narita, M., Ferreira, M., Kirschner, K., Sadaie, M., Darot, J.F., Tavare, S., Watt, F.M., Autophagy mediates the mitotic senescence transition (2009) Genes and Development, 23, pp. 798-803
  • Young, A.R., Narita, M., Narita, M., Cell senescence as both a dynamic and a static phenotype (2013) Methods in Molecular Biology, 965, pp. 1-13
  • Yu, R., Melmed, S., Pathogenesis of pituitary tumors (2010) Progress in Brain Research, 182, pp. 207-227
  • Yu, H., Pardoll, D., Jove, R., STATs in cancer inflammation and immunity: A leading role for STAT3 (2009) Nature Reviews Cancer, 9, pp. 798-809
  • Yun, U.J., Park, S.E., Jo, Y.S., Kim, J., Shin, D.Y., DNA damage induces the IL-6/STAT3 signaling pathway, which has anti-senescence and growth-promoting functions in human tumors (2012) Cancer Letters, 323, pp. 155-160
  • Zhang, X., Horwitz, G.A., Prezant, T.R., Valentini, A., Nakashima, M., Bronstein, M.D., Melmed, S., Structure, expression, and function of human pituitary tumor-transforming gene (PTTG) (1999) Molecular Endocrinology, 13, pp. 156-166
  • Zhang, T., Zhao, B., Li, J., Zhang, C., Li, H., Wu, J., Zhang, S., Hui, G., Pituitary gene expression differs in D-galactose-induced cell senescence and steroid-induced prolactinomas (2015) Molecular Medicine Reports, 11, pp. 3027-3032

Citas:

---------- APA ----------
Sapochnik, M., Fuertes, M. & Arzt, E. (2017) . Programmed cell senescence: Role of IL-6 in the pituitary. Journal of Molecular Endocrinology, 58(4), R241-R253.
http://dx.doi.org/10.1530/JME-17-0026
---------- CHICAGO ----------
Sapochnik, M., Fuertes, M., Arzt, E. "Programmed cell senescence: Role of IL-6 in the pituitary" . Journal of Molecular Endocrinology 58, no. 4 (2017) : R241-R253.
http://dx.doi.org/10.1530/JME-17-0026
---------- MLA ----------
Sapochnik, M., Fuertes, M., Arzt, E. "Programmed cell senescence: Role of IL-6 in the pituitary" . Journal of Molecular Endocrinology, vol. 58, no. 4, 2017, pp. R241-R253.
http://dx.doi.org/10.1530/JME-17-0026
---------- VANCOUVER ----------
Sapochnik, M., Fuertes, M., Arzt, E. Programmed cell senescence: Role of IL-6 in the pituitary. J. Mol. Endocrinol. 2017;58(4):R241-R253.
http://dx.doi.org/10.1530/JME-17-0026