Skip to main content
Top
Published in: Cancer and Metastasis Reviews 2/2010

Open Access 01-06-2010

Senescent cells as a source of inflammatory factors for tumor progression

Authors: Albert R. Davalos, Jean-Philippe Coppe, Judith Campisi, Pierre-Yves Desprez

Published in: Cancer and Metastasis Reviews | Issue 2/2010

Login to get access

Abstract

Cellular senescence, which is associated with aging, is a process by which cells enter a state of permanent cell cycle arrest, therefore constituting a potent tumor suppressive mechanism. Recent studies show that, despite the beneficial effects of cellular senescence, senescent cells can also exert harmful effects on the tissue microenvironment. The most significant of these effects is the acquisition of a senescent-associated secretory phenotype (SASP), which entails a striking increase in the secretion of pro-inflammatory cytokines. Here, we summarize our knowledge of the SASP and the impact it has on tissue microenvironments and ability to stimulate tumor progression.
Literature
2.
3.
go back to reference Campisi, J. (2001). Cellular senescence as a tumor-suppressor mechanism. Trends in Cell Biology, 11(11), 27–31.CrossRef Campisi, J. (2001). Cellular senescence as a tumor-suppressor mechanism. Trends in Cell Biology, 11(11), 27–31.CrossRef
4.
go back to reference Wright, W. E., & Shay, J. W. (2001). Cellular senescence as a tumor-protection mechanism: The essential role of counting. Current Opinion in Genetics and Development, 11, 98–103.PubMedCrossRef Wright, W. E., & Shay, J. W. (2001). Cellular senescence as a tumor-protection mechanism: The essential role of counting. Current Opinion in Genetics and Development, 11, 98–103.PubMedCrossRef
5.
go back to reference Ben-Porath, I., & Weinberg, R. A. (2004). When cells get stressed: An integrative view of cellular senescence. Journal of Clinical Investigation, 113, 8–13.PubMed Ben-Porath, I., & Weinberg, R. A. (2004). When cells get stressed: An integrative view of cellular senescence. Journal of Clinical Investigation, 113, 8–13.PubMed
6.
go back to reference Collins, C. J., & Sedivy, J. M. (2003). Involvement of the INK4a/Arf gene locus in senescence. Aging Cell, 2, 145–150.PubMedCrossRef Collins, C. J., & Sedivy, J. M. (2003). Involvement of the INK4a/Arf gene locus in senescence. Aging Cell, 2, 145–150.PubMedCrossRef
7.
go back to reference Lowe, S. W., & Sherr, C. J. (2003). Tumor suppression by Ink4a-Arf: Progress and puzzles. Current Opinion in Genetics and Development, 13, 77–83.PubMedCrossRef Lowe, S. W., & Sherr, C. J. (2003). Tumor suppression by Ink4a-Arf: Progress and puzzles. Current Opinion in Genetics and Development, 13, 77–83.PubMedCrossRef
8.
go back to reference Ohtani, N., et al. (2004). The p16INK4a-RB pathway: Molecular link between cellular senescence and tumor suppression. Journal of Medical Investigation, 51, 146–153.PubMedCrossRef Ohtani, N., et al. (2004). The p16INK4a-RB pathway: Molecular link between cellular senescence and tumor suppression. Journal of Medical Investigation, 51, 146–153.PubMedCrossRef
9.
go back to reference Gil, J., & Peters, G. (2006). Regulation of the INK4b–ARF–INK4a tumour suppressor locus: All for one or one for all. Nature Reviews. Molecular Cell Biology, 7, 667–677.PubMedCrossRef Gil, J., & Peters, G. (2006). Regulation of the INK4b–ARF–INK4a tumour suppressor locus: All for one or one for all. Nature Reviews. Molecular Cell Biology, 7, 667–677.PubMedCrossRef
10.
go back to reference Braig, M., & Schmitt, C. A. (2006). Oncogene-induced senescence: Putting the brakes on tumor development. Cancer Research, 66, 2881–2884.PubMedCrossRef Braig, M., & Schmitt, C. A. (2006). Oncogene-induced senescence: Putting the brakes on tumor development. Cancer Research, 66, 2881–2884.PubMedCrossRef
11.
go back to reference Shay, J. W., & Roninson, I. B. (2004). Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene, 23, 2919–2933.PubMedCrossRef Shay, J. W., & Roninson, I. B. (2004). Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene, 23, 2919–2933.PubMedCrossRef
12.
go back to reference Ventura, A., et al. (2007). Restoration of p53 function leads to tumour regression in vivo. Nature, 445, 661–665.PubMedCrossRef Ventura, A., et al. (2007). Restoration of p53 function leads to tumour regression in vivo. Nature, 445, 661–665.PubMedCrossRef
13.
go back to reference Xue, W., et al. (2007). Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature, 445, 656–660.PubMedCrossRef Xue, W., et al. (2007). Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature, 445, 656–660.PubMedCrossRef
14.
go back to reference Campisi, J. (2005). Senescent cells, tumor suppression and organismal aging: Good citizens, bad neighbors. Cell, 120, 513–522.PubMedCrossRef Campisi, J. (2005). Senescent cells, tumor suppression and organismal aging: Good citizens, bad neighbors. Cell, 120, 513–522.PubMedCrossRef
15.
go back to reference Jeyapalan, J. C., et al. (2007). Accumulation of senescent cells in mitotic tissue of aging primates. Mechanisms of Ageing and Development, 128, 36–44.PubMedCrossRef Jeyapalan, J. C., et al. (2007). Accumulation of senescent cells in mitotic tissue of aging primates. Mechanisms of Ageing and Development, 128, 36–44.PubMedCrossRef
16.
go back to reference Castro, P., et al. (2003). Cellular senescence in the pathogenesis of benign prostatic hyperplasia. Prostate, 55, 30–38.PubMedCrossRef Castro, P., et al. (2003). Cellular senescence in the pathogenesis of benign prostatic hyperplasia. Prostate, 55, 30–38.PubMedCrossRef
17.
go back to reference Michaloglou, C., et al. (2005). BRAFE600-associated senescence-like cell cycle arrest of human nevi. Nature, 436, 720–724.PubMedCrossRef Michaloglou, C., et al. (2005). BRAFE600-associated senescence-like cell cycle arrest of human nevi. Nature, 436, 720–724.PubMedCrossRef
18.
go back to reference Krtolica, A., et al. (2001). Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging. Proceedings of the National Academy of Sciences of the United States of America, 98, 12072–12077.PubMedCrossRef Krtolica, A., et al. (2001). Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging. Proceedings of the National Academy of Sciences of the United States of America, 98, 12072–12077.PubMedCrossRef
19.
go back to reference Liu, D., & Hornsby, P. J. (2007). Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Research, 67, 3117–3126.PubMedCrossRef Liu, D., & Hornsby, P. J. (2007). Senescent human fibroblasts increase the early growth of xenograft tumors via matrix metalloproteinase secretion. Cancer Research, 67, 3117–3126.PubMedCrossRef
20.
go back to reference Bavik, C., et al. (2006). The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms. Cancer Research, 66, 794–802.PubMedCrossRef Bavik, C., et al. (2006). The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms. Cancer Research, 66, 794–802.PubMedCrossRef
21.
go back to reference Parrinello, S., et al. (2005). Stromal–epithelial interactions in aging and cancer: Senescent fibroblasts alter epithelial cell differentiation. Journal of Cell Science, 118(Pt 3), 485–496.PubMedCrossRef Parrinello, S., et al. (2005). Stromal–epithelial interactions in aging and cancer: Senescent fibroblasts alter epithelial cell differentiation. Journal of Cell Science, 118(Pt 3), 485–496.PubMedCrossRef
22.
go back to reference Coppe, J. P., et al. (2008). Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biology, 6(12), 2853–2868.PubMedCrossRef Coppe, J. P., et al. (2008). Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biology, 6(12), 2853–2868.PubMedCrossRef
23.
go back to reference Coppe, J. P., et al. (2010). A human-like senescence-associated secretory phenotype is conserved in mouse cells dependent on physiological oxygen. PLoS ONE, 5(2), e9188.PubMedCrossRef Coppe, J. P., et al. (2010). A human-like senescence-associated secretory phenotype is conserved in mouse cells dependent on physiological oxygen. PLoS ONE, 5(2), e9188.PubMedCrossRef
24.
go back to reference Campisi, J., & d'Adda di Fagagna, F. (2007). Cellular senescence: When bad things happen to good cells. Nature Reviews. Molecular Cell Biology, 8, 729–740.PubMedCrossRef Campisi, J., & d'Adda di Fagagna, F. (2007). Cellular senescence: When bad things happen to good cells. Nature Reviews. Molecular Cell Biology, 8, 729–740.PubMedCrossRef
25.
26.
go back to reference Hayflick, L. (1965). The limited in vitro lifetime of human diploid cell strains. Experimental Cell Research, 37, 614–636.PubMedCrossRef Hayflick, L. (1965). The limited in vitro lifetime of human diploid cell strains. Experimental Cell Research, 37, 614–636.PubMedCrossRef
27.
go back to reference d'Adda di Fagagna, F., et al. (2003). A DNA damage checkpoint response in telomere-initiated senescence. Nature, 426, 194–198.PubMedCrossRef d'Adda di Fagagna, F., et al. (2003). A DNA damage checkpoint response in telomere-initiated senescence. Nature, 426, 194–198.PubMedCrossRef
28.
go back to reference Dimri, G. P., et al. (1995). A novel biomarker identifies senescent human cells in culture and in aging skin in vivo. Proceedings of the National Academy of Sciences of the United States of America, 92, 9363–9367.PubMedCrossRef Dimri, G. P., et al. (1995). A novel biomarker identifies senescent human cells in culture and in aging skin in vivo. Proceedings of the National Academy of Sciences of the United States of America, 92, 9363–9367.PubMedCrossRef
29.
go back to reference Ben-Porath, I., & Weinberg, R. A. (2005). The signals and pathways activating cellular senescence. International Journal of Biochemistry and Cell Biology, 37(5), 961–976.PubMedCrossRef Ben-Porath, I., & Weinberg, R. A. (2005). The signals and pathways activating cellular senescence. International Journal of Biochemistry and Cell Biology, 37(5), 961–976.PubMedCrossRef
30.
go back to reference Schmitt, C. A. (2003). Senescence, apoptosis and therapy—Cutting the lifelines of cancer. \Nature Reviews. Cancer, 3(4), 286–295.PubMedCrossRef Schmitt, C. A. (2003). Senescence, apoptosis and therapy—Cutting the lifelines of cancer. \Nature Reviews. Cancer, 3(4), 286–295.PubMedCrossRef
31.
go back to reference Martin, G. M. (2005). Genetic modulation of senescent phenotypes in Homo sapiens. Cell, 120, 523–532.PubMedCrossRef Martin, G. M. (2005). Genetic modulation of senescent phenotypes in Homo sapiens. Cell, 120, 523–532.PubMedCrossRef
32.
go back to reference Chien, K. R., & Karsenty, G. (2005). Longevity and lineages: Toward the integrative biology of degenerative diseases in heart, muscle, and bone. Cell, 120(4), 533–544.PubMedCrossRef Chien, K. R., & Karsenty, G. (2005). Longevity and lineages: Toward the integrative biology of degenerative diseases in heart, muscle, and bone. Cell, 120(4), 533–544.PubMedCrossRef
33.
34.
go back to reference Balaban, R. S., Nemoto, S., & Finkel, T. (2005). Mitochondria, oxidants, and aging. Cell, 120, 483–495.PubMedCrossRef Balaban, R. S., Nemoto, S., & Finkel, T. (2005). Mitochondria, oxidants, and aging. Cell, 120, 483–495.PubMedCrossRef
35.
go back to reference Collado, M., & Serrano, M. (2006). The power and the promise of oncogene-induced senescence markers. Nature Reviews. Cancer, 6, 472–476.PubMedCrossRef Collado, M., & Serrano, M. (2006). The power and the promise of oncogene-induced senescence markers. Nature Reviews. Cancer, 6, 472–476.PubMedCrossRef
36.
go back to reference Hill, R., et al. (2005). Selective evolution of stromal mesenchyme with p53 loss in response to epithelial tumorigenesis. Cell, 123, 1001–1011.PubMedCrossRef Hill, R., et al. (2005). Selective evolution of stromal mesenchyme with p53 loss in response to epithelial tumorigenesis. Cell, 123, 1001–1011.PubMedCrossRef
37.
go back to reference Najjar, S. S., Scuteri, A., & Lakatta, E. G. (2005). Arterial aging: Is it an immutable cardiovascular risk factor? Hypertension, 46(3), 454–462.PubMedCrossRef Najjar, S. S., Scuteri, A., & Lakatta, E. G. (2005). Arterial aging: Is it an immutable cardiovascular risk factor? Hypertension, 46(3), 454–462.PubMedCrossRef
38.
go back to reference Kunieda, T., et al. (2006). Angiotensin II induces premature senescence of vascular smooth muscle cells and accelerates the development of atherosclerosis via a p21-dependent pathway. Circulation, 114(9), 953–960.PubMedCrossRef Kunieda, T., et al. (2006). Angiotensin II induces premature senescence of vascular smooth muscle cells and accelerates the development of atherosclerosis via a p21-dependent pathway. Circulation, 114(9), 953–960.PubMedCrossRef
39.
go back to reference Krishnamurthy, J., et al. (2004). Ink4a/Arf expression is a biomarker of aging. Journal of Clinical Investigation, 114, 1299–1307.PubMed Krishnamurthy, J., et al. (2004). Ink4a/Arf expression is a biomarker of aging. Journal of Clinical Investigation, 114, 1299–1307.PubMed
40.
go back to reference Herbig, U., et al. (2004). Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Molecular Cell, 14(4), 501–513.PubMedCrossRef Herbig, U., et al. (2004). Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21(CIP1), but not p16(INK4a). Molecular Cell, 14(4), 501–513.PubMedCrossRef
41.
go back to reference Narita, M., et al. (2003). Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell, 113, 703–716.PubMedCrossRef Narita, M., et al. (2003). Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell, 113, 703–716.PubMedCrossRef
42.
go back to reference Collado, M., & Serrano, M. (2010). Senescence in tumours: Evidence from mice and humans. Nature Reviews. Cancer, 10(1), 51–57.PubMedCrossRef Collado, M., & Serrano, M. (2010). Senescence in tumours: Evidence from mice and humans. Nature Reviews. Cancer, 10(1), 51–57.PubMedCrossRef
43.
go back to reference Coppe, J. P., et al. (2010). The senescence-associated secretory phenotype: The dark side of tumor suppression. Annual Review of Pathology: Mechanisms of Disease, 5, 99–118.CrossRef Coppe, J. P., et al. (2010). The senescence-associated secretory phenotype: The dark side of tumor suppression. Annual Review of Pathology: Mechanisms of Disease, 5, 99–118.CrossRef
44.
go back to reference Coppe, J. P., et al. (2006). Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence. Journal of Biological Chemistry, 281(40), 29568–29574.PubMedCrossRef Coppe, J. P., et al. (2006). Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence. Journal of Biological Chemistry, 281(40), 29568–29574.PubMedCrossRef
45.
go back to reference Lu, S. Y., et al. (2006). Ripe areca nut extract induces G1 phase arrests and senescence-associated phenotypes in normal human oral keratinocyte. Carcinogenesis, 27(6), 1273–1284.PubMedCrossRef Lu, S. Y., et al. (2006). Ripe areca nut extract induces G1 phase arrests and senescence-associated phenotypes in normal human oral keratinocyte. Carcinogenesis, 27(6), 1273–1284.PubMedCrossRef
46.
go back to reference Sarkar, D., et al. (2004). Human polynucleotide phosphorylase (hPNPaseold-35): A potential link between aging and inflammation. Cancer Research, 64(20), 7473–7478.PubMedCrossRef Sarkar, D., et al. (2004). Human polynucleotide phosphorylase (hPNPaseold-35): A potential link between aging and inflammation. Cancer Research, 64(20), 7473–7478.PubMedCrossRef
47.
go back to reference Kuilman, T., et al. (2008). Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell, 133, 1019–1031.PubMedCrossRef Kuilman, T., et al. (2008). Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell, 133, 1019–1031.PubMedCrossRef
48.
go back to reference Garfinkel, S., et al. (1994). Post-transcriptional regulation of interleukin 1 alpha in various strains of young and senescent human umbilical vein endothelial cells. Proceedings of the National Academy of Sciences of the United States of America, 91(4), 1559–1563.PubMedCrossRef Garfinkel, S., et al. (1994). Post-transcriptional regulation of interleukin 1 alpha in various strains of young and senescent human umbilical vein endothelial cells. Proceedings of the National Academy of Sciences of the United States of America, 91(4), 1559–1563.PubMedCrossRef
49.
go back to reference McLachlan, J. A., et al. (1995). Immunological functions of aged human monocytes. Pathobiology, 63(3), 148–159.PubMedCrossRef McLachlan, J. A., et al. (1995). Immunological functions of aged human monocytes. Pathobiology, 63(3), 148–159.PubMedCrossRef
50.
go back to reference Maier, J. A. M., et al. (1990). Extension of the life-span of human endothelial cells by an interleukin-1a antisense oligomer. Science, 249, 1570–1574.PubMedCrossRef Maier, J. A. M., et al. (1990). Extension of the life-span of human endothelial cells by an interleukin-1a antisense oligomer. Science, 249, 1570–1574.PubMedCrossRef
51.
go back to reference Kumar, S., Millis, A. J., & Baglioni, C. (1992). Expression of interleukin 1-inducible genes and production of interleukin 1 by aging human fibroblasts. Proceedings of the National Academy of Sciences of the United States America, 89(10), 4683–4687.CrossRef Kumar, S., Millis, A. J., & Baglioni, C. (1992). Expression of interleukin 1-inducible genes and production of interleukin 1 by aging human fibroblasts. Proceedings of the National Academy of Sciences of the United States America, 89(10), 4683–4687.CrossRef
52.
go back to reference Palmieri, D., Watson, J. M., & Rinehart, C. A. (1999). Age-related expression of PEDF/EPC-1 in human endometrial stromal fibroblasts: Implications for interactive senescence. Experimental Cell Research, 247(1), 142–147.PubMedCrossRef Palmieri, D., Watson, J. M., & Rinehart, C. A. (1999). Age-related expression of PEDF/EPC-1 in human endometrial stromal fibroblasts: Implications for interactive senescence. Experimental Cell Research, 247(1), 142–147.PubMedCrossRef
53.
go back to reference Chang, B. D., et al. (2002). Molecular determinants of terminal growth arrest induced in tumor cells by a chemotherapeutic agent. Proceedings of the National Academy of Sciences of the United States of America, 99, 389–394.PubMedCrossRef Chang, B. D., et al. (2002). Molecular determinants of terminal growth arrest induced in tumor cells by a chemotherapeutic agent. Proceedings of the National Academy of Sciences of the United States of America, 99, 389–394.PubMedCrossRef
54.
go back to reference Bode-Boger, S. M., Scalera, F., & Martens-Lobenhoffer, J. (2005). Asymmetric dimethylarginine (ADMA) accelerates cell senescence. Vascular Medicine, 10(Suppl 1), S65–S71.PubMedCrossRef Bode-Boger, S. M., Scalera, F., & Martens-Lobenhoffer, J. (2005). Asymmetric dimethylarginine (ADMA) accelerates cell senescence. Vascular Medicine, 10(Suppl 1), S65–S71.PubMedCrossRef
55.
go back to reference Wang, S., et al. (1996). Characterization of IGFBP-3, PAI-1 and SPARC mRNA expression in senescent fibroblasts. Mechanisms of Ageing and Development, 92(2–3), 121–132.PubMedCrossRef Wang, S., et al. (1996). Characterization of IGFBP-3, PAI-1 and SPARC mRNA expression in senescent fibroblasts. Mechanisms of Ageing and Development, 92(2–3), 121–132.PubMedCrossRef
56.
go back to reference Grillari, J., et al. (2000). Subtractive hybridization of mRNA from early passage and senescent endothelial cells. Experimental Gerontology, 35(2), 187–197.PubMedCrossRef Grillari, J., et al. (2000). Subtractive hybridization of mRNA from early passage and senescent endothelial cells. Experimental Gerontology, 35(2), 187–197.PubMedCrossRef
57.
go back to reference Lopez-Bermejo, A., et al. (2000). Characterization of insulin-like growth factor-binding protein-related proteins (IGFBP-rPs) 1, 2, and 3 in human prostate epithelial cells: Potential roles for IGFBP-rP1 and 2 in senescence of the prostatic epithelium. Endocrinology, 141(11), 4072–4080.PubMedCrossRef Lopez-Bermejo, A., et al. (2000). Characterization of insulin-like growth factor-binding protein-related proteins (IGFBP-rPs) 1, 2, and 3 in human prostate epithelial cells: Potential roles for IGFBP-rP1 and 2 in senescence of the prostatic epithelium. Endocrinology, 141(11), 4072–4080.PubMedCrossRef
58.
go back to reference Kim, K. H., et al. (2004). Expression of connective tissue growth factor, a biomarker in senescence of human diploid fibroblasts, is up-regulated by a transforming growth factor-beta-mediated signaling pathway. Biochemical and Biophysical Research Communications, 318(4), 819–825.PubMedCrossRef Kim, K. H., et al. (2004). Expression of connective tissue growth factor, a biomarker in senescence of human diploid fibroblasts, is up-regulated by a transforming growth factor-beta-mediated signaling pathway. Biochemical and Biophysical Research Communications, 318(4), 819–825.PubMedCrossRef
59.
go back to reference West, M. D., Pereira-Smith, O. M., & Smith, J. R. (1989). Replicative senescence of human skin fibroblasts correlates with a loss of regulation and overexpression of collagenase activity. Experimental Cell Research, 184, 138–147.PubMedCrossRef West, M. D., Pereira-Smith, O. M., & Smith, J. R. (1989). Replicative senescence of human skin fibroblasts correlates with a loss of regulation and overexpression of collagenase activity. Experimental Cell Research, 184, 138–147.PubMedCrossRef
60.
go back to reference Millis, A. J. T., et al. (1992). Differential expression of metalloproteinase and tissue inhibitor of metalloproteinase genes in diploid human fibroblasts. Experimental Cell Research, 201, 373–379.PubMedCrossRef Millis, A. J. T., et al. (1992). Differential expression of metalloproteinase and tissue inhibitor of metalloproteinase genes in diploid human fibroblasts. Experimental Cell Research, 201, 373–379.PubMedCrossRef
61.
go back to reference Zeng, G., & Millis, A. J. (1996). Differential regulation of collagenase and stromelysin mRNA in late passage cultures of human fibroblasts. Experimental Cell Research, 222(1), 150–156.PubMedCrossRef Zeng, G., & Millis, A. J. (1996). Differential regulation of collagenase and stromelysin mRNA in late passage cultures of human fibroblasts. Experimental Cell Research, 222(1), 150–156.PubMedCrossRef
62.
go back to reference Hornebeck, W., & Maquart, F. X. (2003). Proteolyzed matrix as a template for the regulation of tumor progression. Biomedicine and Pharmacotherapy, 57(5–6), 223–230.CrossRef Hornebeck, W., & Maquart, F. X. (2003). Proteolyzed matrix as a template for the regulation of tumor progression. Biomedicine and Pharmacotherapy, 57(5–6), 223–230.CrossRef
63.
go back to reference McQuibban, G. A., et al. (2002). Matrix metalloproteinase processing of monocyte chemoattractant proteins generates CC chemokine receptor antagonists with anti-inflammatory properties in vivo. Blood, 100(4), 1160–1167.PubMed McQuibban, G. A., et al. (2002). Matrix metalloproteinase processing of monocyte chemoattractant proteins generates CC chemokine receptor antagonists with anti-inflammatory properties in vivo. Blood, 100(4), 1160–1167.PubMed
64.
go back to reference Blasi, F., & Carmeliet, P. (2002). uPAR: A versatile signalling orchestrator. Nature Reviews. Molecular Cell Biology, 3(12), 932–943.PubMedCrossRef Blasi, F., & Carmeliet, P. (2002). uPAR: A versatile signalling orchestrator. Nature Reviews. Molecular Cell Biology, 3(12), 932–943.PubMedCrossRef
65.
go back to reference Sato, I., et al. (1993). Reduction of nitric oxide producing activity associated with in vitro aging in cultured human umbilical vein endothelial cell. Biochemical and Biophysical Research Communications, 195(2), 1070–1076.PubMedCrossRef Sato, I., et al. (1993). Reduction of nitric oxide producing activity associated with in vitro aging in cultured human umbilical vein endothelial cell. Biochemical and Biophysical Research Communications, 195(2), 1070–1076.PubMedCrossRef
66.
go back to reference Lee, A. C., et al. (1999). Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species. Journal of Biological Chemistry, 274(12), 7936–7940.PubMedCrossRef Lee, A. C., et al. (1999). Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species. Journal of Biological Chemistry, 274(12), 7936–7940.PubMedCrossRef
67.
go back to reference van der Loo, B., et al. (2000). Enhanced peroxynitrite formation is associated with vascular aging. Journal of Experimental Medicine, 192(12), 1731–1744.PubMedCrossRef van der Loo, B., et al. (2000). Enhanced peroxynitrite formation is associated with vascular aging. Journal of Experimental Medicine, 192(12), 1731–1744.PubMedCrossRef
68.
go back to reference Macip, S., et al. (2002). Inhibition of p21-mediated ROS accumulation can rescue p21-induced senescence. EMBO Journal, 21, 2180–2188.PubMedCrossRef Macip, S., et al. (2002). Inhibition of p21-mediated ROS accumulation can rescue p21-induced senescence. EMBO Journal, 21, 2180–2188.PubMedCrossRef
69.
go back to reference Xin, M. G., et al. (2003). Senescence-enhanced oxidative stress is associated with deficiency of mitochondrial cytochrome c oxidase in vascular endothelial cells. Mechanisms of Ageing and Development, 124(8–9), 911–919.PubMedCrossRef Xin, M. G., et al. (2003). Senescence-enhanced oxidative stress is associated with deficiency of mitochondrial cytochrome c oxidase in vascular endothelial cells. Mechanisms of Ageing and Development, 124(8–9), 911–919.PubMedCrossRef
70.
go back to reference Finkel, T., & Holbrook, N. J. (2000). Oxidants, oxidative stress and the biology of ageing. Nature, 408, 239–247.PubMedCrossRef Finkel, T., & Holbrook, N. J. (2000). Oxidants, oxidative stress and the biology of ageing. Nature, 408, 239–247.PubMedCrossRef
71.
go back to reference Finkel, T., Serrano, M., & Blasco, M. A. (2007). The common biology of cancer and ageing. Nature, 448, 767–774.PubMedCrossRef Finkel, T., Serrano, M., & Blasco, M. A. (2007). The common biology of cancer and ageing. Nature, 448, 767–774.PubMedCrossRef
72.
go back to reference Funayama, R., & Ishikawa, F. (2007). Cellular senescence and chromatin structure. Chromosoma, 116(5), 431–440.PubMedCrossRef Funayama, R., & Ishikawa, F. (2007). Cellular senescence and chromatin structure. Chromosoma, 116(5), 431–440.PubMedCrossRef
73.
go back to reference Mehta, I. S., et al. (2007). Alterations to nuclear architecture and genome behavior in senescent cells. Annals of the New York Academy of Sciences, 1100, 250–263.PubMedCrossRef Mehta, I. S., et al. (2007). Alterations to nuclear architecture and genome behavior in senescent cells. Annals of the New York Academy of Sciences, 1100, 250–263.PubMedCrossRef
74.
go back to reference Narita, M. (2007). Cellular senescence and chromatin organisation. British Journal of Cancer, 96(5), 686–691.PubMedCrossRef Narita, M. (2007). Cellular senescence and chromatin organisation. British Journal of Cancer, 96(5), 686–691.PubMedCrossRef
76.
go back to reference Acosta, J. C., et al. (2008). Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell, 133(6), 1006–1018.PubMedCrossRef Acosta, J. C., et al. (2008). Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell, 133(6), 1006–1018.PubMedCrossRef
77.
go back to reference Rodier, F., et al. (2009). Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nature Cell Biology, 11(8), 973–979.PubMedCrossRef Rodier, F., et al. (2009). Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion. Nature Cell Biology, 11(8), 973–979.PubMedCrossRef
78.
go back to reference d'Adda di Fagagna, F. (2008). Living on a break: Cellular senescence as a DNA-damage response. Nature Reviews. Cancer, 8(7), 512–22.PubMedCrossRef d'Adda di Fagagna, F. (2008). Living on a break: Cellular senescence as a DNA-damage response. Nature Reviews. Cancer, 8(7), 512–22.PubMedCrossRef
79.
go back to reference Hiscott, J., et al. (1993). Characterization of a functional NF-kappa B site in the human interleukin 1 beta promoter: Evidence for a positive autoregulatory loop. Molecular and Cellular Biology, 13(10), 6231–6240.PubMed Hiscott, J., et al. (1993). Characterization of a functional NF-kappa B site in the human interleukin 1 beta promoter: Evidence for a positive autoregulatory loop. Molecular and Cellular Biology, 13(10), 6231–6240.PubMed
80.
go back to reference Niu, J., et al. (2004). Identification of an autoregulatory feedback pathway involving interleukin-1alpha in induction of constitutive NF-kappaB activation in pancreatic cancer cells. Journal of Biological Chemistry, 279(16), 16452–16462.PubMedCrossRef Niu, J., et al. (2004). Identification of an autoregulatory feedback pathway involving interleukin-1alpha in induction of constitutive NF-kappaB activation in pancreatic cancer cells. Journal of Biological Chemistry, 279(16), 16452–16462.PubMedCrossRef
81.
go back to reference Orjalo, A. V., et al. (2009). Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proceedings of the National Academy of Sciences of the United States of America, 106(40), 17031–17036.PubMedCrossRef Orjalo, A. V., et al. (2009). Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network. Proceedings of the National Academy of Sciences of the United States of America, 106(40), 17031–17036.PubMedCrossRef
82.
go back to reference Bhaumik, D., et al. (2009). MicroRNAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL-8. Aging, 1(4), 402–411.PubMed Bhaumik, D., et al. (2009). MicroRNAs miR-146a/b negatively modulate the senescence-associated inflammatory mediators IL-6 and IL-8. Aging, 1(4), 402–411.PubMed
83.
go back to reference Taganov, K. D., et al. (2006). NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proceedings of the National Academy of Sciences of the United States of America, 103(33), 12481–12486.PubMedCrossRef Taganov, K. D., et al. (2006). NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proceedings of the National Academy of Sciences of the United States of America, 103(33), 12481–12486.PubMedCrossRef
84.
go back to reference Adams, P. D. (2007). Remodeling of chromatin structure in senescent cells and its potential impact on tumor suppression and aging. Gene, 397(1–2), 84–93.PubMedCrossRef Adams, P. D. (2007). Remodeling of chromatin structure in senescent cells and its potential impact on tumor suppression and aging. Gene, 397(1–2), 84–93.PubMedCrossRef
85.
go back to reference Bianchi, M. E., & Agresti, A. (2005). HMG proteins: Dynamic players in gene regulation and differentiation. Current Opinion in Genetics and Development, 15(5), 496–506.PubMedCrossRef Bianchi, M. E., & Agresti, A. (2005). HMG proteins: Dynamic players in gene regulation and differentiation. Current Opinion in Genetics and Development, 15(5), 496–506.PubMedCrossRef
86.
go back to reference Lotze, M. T., & Tracey, K. J. (2005). High-mobility group box 1 protein (HMGB1): Nuclear weapon in the immune arsenal. Nature Reviews. Immunology, 5(4), 331–342.PubMedCrossRef Lotze, M. T., & Tracey, K. J. (2005). High-mobility group box 1 protein (HMGB1): Nuclear weapon in the immune arsenal. Nature Reviews. Immunology, 5(4), 331–342.PubMedCrossRef
87.
go back to reference Park, J. S., et al. (2003). Activation of gene expression in human neutrophils by high mobility group box 1 protein. American Journal of Physiology Cell Physiology, 284(4), C870–C879.PubMed Park, J. S., et al. (2003). Activation of gene expression in human neutrophils by high mobility group box 1 protein. American Journal of Physiology Cell Physiology, 284(4), C870–C879.PubMed
88.
go back to reference Stros, M., et al. (2004). High-affinity binding of tumor-suppressor protein p53 and HMGB1 to hemicatenated DNA loops. Biochemistry, 43(22), 7215–7225.PubMedCrossRef Stros, M., et al. (2004). High-affinity binding of tumor-suppressor protein p53 and HMGB1 to hemicatenated DNA loops. Biochemistry, 43(22), 7215–7225.PubMedCrossRef
89.
go back to reference Jayaraman, L., et al. (1998). High mobility group protein-1 (HMG-1) is a unique activator of p53. Genes and Development, 12(4), 462–472.PubMedCrossRef Jayaraman, L., et al. (1998). High mobility group protein-1 (HMG-1) is a unique activator of p53. Genes and Development, 12(4), 462–472.PubMedCrossRef
90.
go back to reference Scaffidi, P., Misteli, T., & Bianchi, M. E. (2002). Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature, 418(6894), 191–195.PubMedCrossRef Scaffidi, P., Misteli, T., & Bianchi, M. E. (2002). Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature, 418(6894), 191–195.PubMedCrossRef
91.
go back to reference Kokkola, R., et al. (2005). RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scandinavian Journal of Immunology, 61(1), 1–9.PubMedCrossRef Kokkola, R., et al. (2005). RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scandinavian Journal of Immunology, 61(1), 1–9.PubMedCrossRef
92.
go back to reference Rouhiainen, A., et al. (2007). Pivotal advance: Analysis of proinflammatory activity of highly purified eukaryotic recombinant HMGB1 (amphoterin). Journal of Leukocyte Biology, 81(1), 49–58.PubMedCrossRef Rouhiainen, A., et al. (2007). Pivotal advance: Analysis of proinflammatory activity of highly purified eukaryotic recombinant HMGB1 (amphoterin). Journal of Leukocyte Biology, 81(1), 49–58.PubMedCrossRef
93.
go back to reference Bianchi, M.E., (2009). HMGB1 loves company. J Leukoc Biol. Bianchi, M.E., (2009). HMGB1 loves company. J Leukoc Biol.
94.
go back to reference Sha, Y., et al. (2008). HMGB1 develops enhanced proinflammatory activity by binding to cytokines. Journal of Immunology, 180(4), 2531–2537. Sha, Y., et al. (2008). HMGB1 develops enhanced proinflammatory activity by binding to cytokines. Journal of Immunology, 180(4), 2531–2537.
95.
go back to reference Kokkola, R., et al. (2003). Successful treatment of collagen-induced arthritis in mice and rats by targeting extracellular high mobility group box chromosomal protein 1 activity. Arthritis and Rheumatism, 48(7), 2052–2058.PubMedCrossRef Kokkola, R., et al. (2003). Successful treatment of collagen-induced arthritis in mice and rats by targeting extracellular high mobility group box chromosomal protein 1 activity. Arthritis and Rheumatism, 48(7), 2052–2058.PubMedCrossRef
96.
go back to reference Elliott, M. R., et al. (2009). Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature, 461(7261), 282–286.PubMedCrossRef Elliott, M. R., et al. (2009). Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature, 461(7261), 282–286.PubMedCrossRef
97.
go back to reference Krizhanovsky, V., et al. (2008). Senescence of activated stellate cells limits liver fibrosis. Cell, 134(4), 657–667.PubMedCrossRef Krizhanovsky, V., et al. (2008). Senescence of activated stellate cells limits liver fibrosis. Cell, 134(4), 657–667.PubMedCrossRef
98.
go back to reference Fages, C., et al. (2000). Regulation of cell migration by amphoterin. Journal of Cell Science, 113(Pt 4), 611–620.PubMed Fages, C., et al. (2000). Regulation of cell migration by amphoterin. Journal of Cell Science, 113(Pt 4), 611–620.PubMed
99.
go back to reference Schlueter, C., et al. (2005). Angiogenetic signaling through hypoxia: HMGB1: An angiogenetic switch molecule. American Journal of Pathology, 166(4), 1259–1263.PubMed Schlueter, C., et al. (2005). Angiogenetic signaling through hypoxia: HMGB1: An angiogenetic switch molecule. American Journal of Pathology, 166(4), 1259–1263.PubMed
100.
go back to reference Taguchi, A., et al. (2000). Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature, 405(6784), 354–360.PubMedCrossRef Taguchi, A., et al. (2000). Blockade of RAGE-amphoterin signalling suppresses tumour growth and metastases. Nature, 405(6784), 354–360.PubMedCrossRef
101.
go back to reference Brennan, F. M., Maini, R. N., & Feldmann, M. (1995). Cytokine expression in chronic inflammatory disease. British Medical Bulletin, 51(2), 368–384.PubMed Brennan, F. M., Maini, R. N., & Feldmann, M. (1995). Cytokine expression in chronic inflammatory disease. British Medical Bulletin, 51(2), 368–384.PubMed
102.
go back to reference Brod, S. A. (2000). Unregulated inflammation shortens human functional longevity. Inflammation Research, 49(11), 561–570.PubMedCrossRef Brod, S. A. (2000). Unregulated inflammation shortens human functional longevity. Inflammation Research, 49(11), 561–570.PubMedCrossRef
103.
go back to reference Caruso, C., et al. (2004). Aging, longevity, inflammation, and cancer. Annals of the New York Academy of Sciences, 1028, 1–13.PubMedCrossRef Caruso, C., et al. (2004). Aging, longevity, inflammation, and cancer. Annals of the New York Academy of Sciences, 1028, 1–13.PubMedCrossRef
104.
go back to reference Tsai, K. K., et al. (2005). Cellular mechanisms for low-dose ionizing radiation-induced perturbation of the breast tissue microenvironment. Cancer Research, 65, 6734–6744.PubMedCrossRef Tsai, K. K., et al. (2005). Cellular mechanisms for low-dose ionizing radiation-induced perturbation of the breast tissue microenvironment. Cancer Research, 65, 6734–6744.PubMedCrossRef
105.
go back to reference Sun, P., et al. (2007). PRAK is essential for ras-induced senescence and tumor suppression. Cell, 128, 295–308.PubMedCrossRef Sun, P., et al. (2007). PRAK is essential for ras-induced senescence and tumor suppression. Cell, 128, 295–308.PubMedCrossRef
106.
go back to reference Choi, J., et al. (2000). Expression of senescence-associated beta-galactosidase in enlarged prostates from men with benign prostatic hyperplasia. Urology, 56, 160–166.PubMedCrossRef Choi, J., et al. (2000). Expression of senescence-associated beta-galactosidase in enlarged prostates from men with benign prostatic hyperplasia. Urology, 56, 160–166.PubMedCrossRef
107.
go back to reference Ohuchida, K., et al. (2004). Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor–stromal interactions. Cancer Research, 64(9), 3215–3222.PubMedCrossRef Ohuchida, K., et al. (2004). Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor–stromal interactions. Cancer Research, 64(9), 3215–3222.PubMedCrossRef
108.
go back to reference Coppe, J. P., et al. (2008). A role for fibroblasts in mediating the effects of tobacco-induced epithelial cell growth and invasion. Molecular Cancer Research, 6(7), 1085–1098.PubMedCrossRef Coppe, J. P., et al. (2008). A role for fibroblasts in mediating the effects of tobacco-induced epithelial cell growth and invasion. Molecular Cancer Research, 6(7), 1085–1098.PubMedCrossRef
109.
go back to reference Barcellos-Hoff, M. H., & Ravani, S. A. (2000). Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Research, 60, 1254–1260.PubMed Barcellos-Hoff, M. H., & Ravani, S. A. (2000). Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Research, 60, 1254–1260.PubMed
110.
go back to reference Yang, F., et al. (2005). Stromal expression of connective tissue growth factor promotes angiogenesis and prostate cancer tumorigenesis. Cancer Research, 65(19), 8887–8895.PubMedCrossRef Yang, F., et al. (2005). Stromal expression of connective tissue growth factor promotes angiogenesis and prostate cancer tumorigenesis. Cancer Research, 65(19), 8887–8895.PubMedCrossRef
111.
go back to reference Lehmann, B. D., et al. (2008). Senescence-associated exosome release from human prostate cancer cells. Cancer Research, 68, 7864–7871.PubMedCrossRef Lehmann, B. D., et al. (2008). Senescence-associated exosome release from human prostate cancer cells. Cancer Research, 68, 7864–7871.PubMedCrossRef
112.
go back to reference Dilley, T. K., Bowden, G. T., & Chen, Q. M. (2003). Novel mechanisms of sublethal oxidant toxicity: Induction of premature senescence in human fibroblasts confers tumor promoter activity. Experimental Cell Research, 290, 38–48.PubMedCrossRef Dilley, T. K., Bowden, G. T., & Chen, Q. M. (2003). Novel mechanisms of sublethal oxidant toxicity: Induction of premature senescence in human fibroblasts confers tumor promoter activity. Experimental Cell Research, 290, 38–48.PubMedCrossRef
113.
go back to reference Dhawan, P., & Richmond, A. (2002). Role of CXCL1 in tumorigenesis of melanoma. Journal of Leukocyte Biology, 72(1), 9–18.PubMed Dhawan, P., & Richmond, A. (2002). Role of CXCL1 in tumorigenesis of melanoma. Journal of Leukocyte Biology, 72(1), 9–18.PubMed
114.
go back to reference Balentien, E., et al. (1991). Effects of MGSA/GRO alpha on melanocyte transformation. Oncogene, 6(7), 1115–1124.PubMed Balentien, E., et al. (1991). Effects of MGSA/GRO alpha on melanocyte transformation. Oncogene, 6(7), 1115–1124.PubMed
115.
go back to reference Schadendorf, D., et al. (1993). IL-8 produced by human malignant melanoma cells in vitro is an essential autocrine growth factor. Journal of Immunology, 151(5), 2667–2675. Schadendorf, D., et al. (1993). IL-8 produced by human malignant melanoma cells in vitro is an essential autocrine growth factor. Journal of Immunology, 151(5), 2667–2675.
116.
go back to reference Bernardini, G., et al. (2000). I-309 binds to and activates endothelial cell functions and acts as an angiogenic molecule in vivo. Blood, 96(13), 4039–4045.PubMed Bernardini, G., et al. (2000). I-309 binds to and activates endothelial cell functions and acts as an angiogenic molecule in vivo. Blood, 96(13), 4039–4045.PubMed
117.
go back to reference Salcedo, R., et al. (2001). Eotaxin (CCL11) induces in vivo angiogenic responses by human CCR3+ endothelial cells. Journal of Immunology, 166(12), 7571–7578. Salcedo, R., et al. (2001). Eotaxin (CCL11) induces in vivo angiogenic responses by human CCR3+ endothelial cells. Journal of Immunology, 166(12), 7571–7578.
118.
119.
go back to reference Sparmann, A., & Bar-Sagi, D. (2004). Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell, 6(5), 447–458.PubMedCrossRef Sparmann, A., & Bar-Sagi, D. (2004). Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell, 6(5), 447–458.PubMedCrossRef
120.
go back to reference Frey, A. B. (2006). Myeloid suppressor cells regulate the adaptive immune response to cancer. Journal of Clinical Investigation, 116(10), 2587–2590.PubMedCrossRef Frey, A. B. (2006). Myeloid suppressor cells regulate the adaptive immune response to cancer. Journal of Clinical Investigation, 116(10), 2587–2590.PubMedCrossRef
121.
go back to reference Birchmeier, C., et al. (2003). Met, metastasis, motility and more. Nature Reviews. Molecular Cell Biology, 4(12), 915–925.PubMedCrossRef Birchmeier, C., et al. (2003). Met, metastasis, motility and more. Nature Reviews. Molecular Cell Biology, 4(12), 915–925.PubMedCrossRef
122.
go back to reference Camphausen, K., et al. (2001). Radiation therapy to a primary tumor accelerates metastatic growth in mice. Cancer Research, 61(5), 2207–2211.PubMed Camphausen, K., et al. (2001). Radiation therapy to a primary tumor accelerates metastatic growth in mice. Cancer Research, 61(5), 2207–2211.PubMed
123.
go back to reference Qian, L. W., et al. (2002). Radiation-induced increase in invasive potential of human pancreatic cancer cells and its blockade by a matrix metalloproteinase inhibitor, CGS27023. Clinical Cancer Research, 8(4), 1223–1227.PubMed Qian, L. W., et al. (2002). Radiation-induced increase in invasive potential of human pancreatic cancer cells and its blockade by a matrix metalloproteinase inhibitor, CGS27023. Clinical Cancer Research, 8(4), 1223–1227.PubMed
124.
go back to reference Strieter, R. M., et al. (2006). Cancer CXC chemokine networks and tumour angiogenesis. European Journal of Cancer, 42(6), 768–778.PubMedCrossRef Strieter, R. M., et al. (2006). Cancer CXC chemokine networks and tumour angiogenesis. European Journal of Cancer, 42(6), 768–778.PubMedCrossRef
125.
go back to reference Orr, F. W., & Wang, H. H. (2001). Tumor cell interactions with the microvasculature: A rate-limiting step in metastasis. Surgical Oncology Clinics of North America, 10(2), 357–81. ix−x.PubMed Orr, F. W., & Wang, H. H. (2001). Tumor cell interactions with the microvasculature: A rate-limiting step in metastasis. Surgical Oncology Clinics of North America, 10(2), 357–81. ix−x.PubMed
126.
go back to reference Nickoloff, B. J., et al. (2004). Tumor suppressor maspin is up-regulated during keratinocyte senescence, exerting a paracrine antiangiogenic activity. Cancer Research, 64(9), 2956–2961.PubMedCrossRef Nickoloff, B. J., et al. (2004). Tumor suppressor maspin is up-regulated during keratinocyte senescence, exerting a paracrine antiangiogenic activity. Cancer Research, 64(9), 2956–2961.PubMedCrossRef
127.
go back to reference Mantovani, A. (2004). Chemokines in neoplastic progression. Seminars in Cancer Biology, 14(3), 147–148.PubMedCrossRef Mantovani, A. (2004). Chemokines in neoplastic progression. Seminars in Cancer Biology, 14(3), 147–148.PubMedCrossRef
128.
go back to reference Homey, B., Muller, A., & Zlotnik, A. (2002). Chemokines: Agents for the immunotherapy of cancer? Nature Reviews. Immunology, 2(3), 175–184.PubMedCrossRef Homey, B., Muller, A., & Zlotnik, A. (2002). Chemokines: Agents for the immunotherapy of cancer? Nature Reviews. Immunology, 2(3), 175–184.PubMedCrossRef
129.
130.
go back to reference Ben-Baruch, A. (2006). Inflammation-associated immune suppression in cancer: The roles played by cytokines, chemokines and additional mediators. Seminars in Cancer Biology, 16(1), 38–52.PubMedCrossRef Ben-Baruch, A. (2006). Inflammation-associated immune suppression in cancer: The roles played by cytokines, chemokines and additional mediators. Seminars in Cancer Biology, 16(1), 38–52.PubMedCrossRef
131.
go back to reference Potempa, S., & Ridley, A. J. (1998). Activation of both MAP kinase and phosphatidylinositide 3-kinase by Ras is required for hepatocyte growth factor/scatter factor-induced adherens junction disassembly. Molecular Biology of the Cell, 9(8), 2185–2200.PubMed Potempa, S., & Ridley, A. J. (1998). Activation of both MAP kinase and phosphatidylinositide 3-kinase by Ras is required for hepatocyte growth factor/scatter factor-induced adherens junction disassembly. Molecular Biology of the Cell, 9(8), 2185–2200.PubMed
132.
go back to reference Paumelle, R., et al. (2002). Hepatocyte growth factor/scatter factor activates the ETS1 transcription factor by a RAS–RAF–MEK–ERK signaling pathway. Oncogene, 21(15), 2309–2319.PubMedCrossRef Paumelle, R., et al. (2002). Hepatocyte growth factor/scatter factor activates the ETS1 transcription factor by a RAS–RAF–MEK–ERK signaling pathway. Oncogene, 21(15), 2309–2319.PubMedCrossRef
133.
go back to reference Thiery, J. P. (2002). Epithelial–mesenchymal transitions in tumour progression. Nature Reviews. Cancer, 2(6), 442–454.PubMedCrossRef Thiery, J. P. (2002). Epithelial–mesenchymal transitions in tumour progression. Nature Reviews. Cancer, 2(6), 442–454.PubMedCrossRef
134.
go back to reference Tonini, T., Rossi, F., & Claudio, P. P. (2003). Molecular basis of angiogenesis and cancer. Oncogene, 22(42), 6549–6556.PubMedCrossRef Tonini, T., Rossi, F., & Claudio, P. P. (2003). Molecular basis of angiogenesis and cancer. Oncogene, 22(42), 6549–6556.PubMedCrossRef
135.
go back to reference Nesbit, M., et al. (2001). Low-level monocyte chemoattractant protein-1 stimulation of monocytes leads to tumor formation in nontumorigenic melanoma cells. Journal of Immunology, 166(11), 6483–6490. Nesbit, M., et al. (2001). Low-level monocyte chemoattractant protein-1 stimulation of monocytes leads to tumor formation in nontumorigenic melanoma cells. Journal of Immunology, 166(11), 6483–6490.
136.
go back to reference Schaider, H., et al. (2003). Differential response of primary and metastatic melanomas to neutrophils attracted by IL-8. International Journal of Cancer, 103(3), 335–343.CrossRef Schaider, H., et al. (2003). Differential response of primary and metastatic melanomas to neutrophils attracted by IL-8. International Journal of Cancer, 103(3), 335–343.CrossRef
137.
go back to reference Sica, A., et al. (2006). Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: Potential targets of anti-cancer therapy. European Journal of Cancer, 42(6), 717–727.PubMedCrossRef Sica, A., et al. (2006). Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: Potential targets of anti-cancer therapy. European Journal of Cancer, 42(6), 717–727.PubMedCrossRef
138.
go back to reference Selivanova, G., et al. (1997). Restoration of the growth suppression function of mutant p53 by a synthetic peptide derived from the p53 C-terminal domain. Nature Medicine, 3(6), 632–638.PubMedCrossRef Selivanova, G., et al. (1997). Restoration of the growth suppression function of mutant p53 by a synthetic peptide derived from the p53 C-terminal domain. Nature Medicine, 3(6), 632–638.PubMedCrossRef
139.
go back to reference Foster, B. A., et al. (1999). Pharmacological rescue of mutant p53 conformation and function. Science, 286(5449), 2507–2510.PubMedCrossRef Foster, B. A., et al. (1999). Pharmacological rescue of mutant p53 conformation and function. Science, 286(5449), 2507–2510.PubMedCrossRef
140.
go back to reference Chene, P. (2003). Inhibiting the p53–MDM2 interaction: An important target for cancer therapy. Nature Reviews. Cancer, 3(2), 102–109.PubMedCrossRef Chene, P. (2003). Inhibiting the p53–MDM2 interaction: An important target for cancer therapy. Nature Reviews. Cancer, 3(2), 102–109.PubMedCrossRef
141.
go back to reference Selivanova, G., & Wiman, K. G. (2007). Reactivation of mutant p53: Molecular mechanisms and therapeutic potential. Oncogene, 26(15), 2243–2254.PubMedCrossRef Selivanova, G., & Wiman, K. G. (2007). Reactivation of mutant p53: Molecular mechanisms and therapeutic potential. Oncogene, 26(15), 2243–2254.PubMedCrossRef
142.
go back to reference Barnes, P. J., & Karin, M. (1997). Nuclear factor-kappaB: A pivotal transcription factor in chronic inflammatory diseases. New England Journal of Medicine, 336(15), 1066–1071.PubMedCrossRef Barnes, P. J., & Karin, M. (1997). Nuclear factor-kappaB: A pivotal transcription factor in chronic inflammatory diseases. New England Journal of Medicine, 336(15), 1066–1071.PubMedCrossRef
Metadata
Title
Senescent cells as a source of inflammatory factors for tumor progression
Authors
Albert R. Davalos
Jean-Philippe Coppe
Judith Campisi
Pierre-Yves Desprez
Publication date
01-06-2010
Publisher
Springer US
Published in
Cancer and Metastasis Reviews / Issue 2/2010
Print ISSN: 0167-7659
Electronic ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-010-9220-9

Other articles of this Issue 2/2010

Cancer and Metastasis Reviews 2/2010 Go to the issue

EditorialNotes

Preface

Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine