Skip to main content
Top
Published in: Breast Cancer Research 1/2015

Open Access 01-12-2015 | Research article

Role of ADAM17 in the non-cell autonomous effects of oncogene-induced senescence

Authors: Beatriz Morancho, Águeda Martínez-Barriocanal, Josep Villanueva, Joaquín Arribas

Published in: Breast Cancer Research | Issue 1/2015

Login to get access

Abstract

Introduction

Cellular senescence is a terminal cell proliferation arrest that can be triggered by oncogenes. One of the traits of oncogene-induced senescence (OIS) is the so-called senescence-associated secretory phenotype or senescence secretome. Depending on the context, the non-cell autonomous effects of OIS may vary from tumor suppression to promotion of metastasis. Despite being such a physiological and pathologically relevant effector, the mechanisms of generation of the senescence secretome are largely unknown.

Methods

We analyzed by label-free proteomics the secretome of p95HER2-induced senescent cells and compared the levels of the membrane-anchored proteins with their transcript levels. Then, protein and RNA levels of ADAM17 were evaluated by using Western blot and reverse transcription-polymerase chain reaction, its localization by using biotin labeling and immunofluorescence, and its activity by using alkaline phosphatase-tagged substrates. The p95HER2-expressing cell lines, senescent MCF7 and proliferating MCF10A, were analyzed to study ADAM17 regulation. Finally, we knocked down ADAM17 to determine its contribution to the senescence-associated secretome. The effect of this secretome was evaluated in migration assays in vitro and in nude mice by assessing the metastatic ability of orthotopically co-injected non-senescent cells.

Results

Using breast cancer cells expressing p95HER2, a constitutively active fragment of the proto-oncogene HER2 that induces OIS, we show that the extracellular domains of a variety of membrane-bound proteins form part of the senescence secretome. We determine that these proteins are regulated transcriptionally and, in addition, that their shedding is limited by the protease ADAM17. The activity of the sheddase is constrained, at least in part, by the accumulation of cellular cholesterol. The blockade of ADAM17 abrogates several prometastatic effects of the p95HER2-induced senescence secretome, both in vitro and in vivo.

Conclusions

Considering these findings, we conclude that ectodomain shedding is tightly regulated in oncogene-induced senescent cells by integrating transcription of the shedding substrates with limiting ADAM17 activity. The remaining activity of ADAM17 contributes to the non-cell autonomous protumorigenic effects of p95HER2-induced senescent cells. Because ADAM17 is druggable, these results represent an approximation to the pharmacological regulation of the senescence secretome.
Appendix
Available only for authorised users
Literature
1.
go back to reference Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014;15:482–96.CrossRefPubMed Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014;15:482–96.CrossRefPubMed
2.
go back to reference Angelini PD, Zacarias Fluck MF, Pedersen K, Parra-Palau JL, Guiu M, Bernadó Morales C, et al. Constitutive HER2 signaling promotes breast cancer metastasis through cellular senescence. Cancer Res. 2013;73:450–8.CrossRefPubMed Angelini PD, Zacarias Fluck MF, Pedersen K, Parra-Palau JL, Guiu M, Bernadó Morales C, et al. Constitutive HER2 signaling promotes breast cancer metastasis through cellular senescence. Cancer Res. 2013;73:450–8.CrossRefPubMed
3.
go back to reference Kang T-W, Yevsa T, Woller N, Hoenicke L, Wuestefeld T, Dauch D, et al. Senescence surveillance of pre-malignanthepatocytes limits liver cancer development. Nature. 2011;479:547–51.CrossRefPubMed Kang T-W, Yevsa T, Woller N, Hoenicke L, Wuestefeld T, Dauch D, et al. Senescence surveillance of pre-malignanthepatocytes limits liver cancer development. Nature. 2011;479:547–51.CrossRefPubMed
4.
go back to reference Toso A, Revandkar A, Di Mitri D, Guccini I, Proietti M, Sarti M, et al. Enhancing chemotherapy efficacy in Pten-Deficient prostate tumors by activating the senescence-associated antitumor immunity. Cell Rep. 2014;9:75–89.CrossRefPubMed Toso A, Revandkar A, Di Mitri D, Guccini I, Proietti M, Sarti M, et al. Enhancing chemotherapy efficacy in Pten-Deficient prostate tumors by activating the senescence-associated antitumor immunity. Cell Rep. 2014;9:75–89.CrossRefPubMed
5.
go back to reference Reddy JP, Peddibhotla S, Bu W, Zhao J, Haricharan S, Du YC, et al. Defining the ATM-mediated barrier to tumorigenesis in somatic mammary cells following ErbB2 activation. Proc Natl Acad Sci U S A. 2010;107:3728–33.CrossRefPubMedPubMedCentral Reddy JP, Peddibhotla S, Bu W, Zhao J, Haricharan S, Du YC, et al. Defining the ATM-mediated barrier to tumorigenesis in somatic mammary cells following ErbB2 activation. Proc Natl Acad Sci U S A. 2010;107:3728–33.CrossRefPubMedPubMedCentral
6.
go back to reference Xue W, Zender L, Miething C, Dickins RA, Hernando E, Krizhanovsky V, et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature. 2007;445:656–60.CrossRefPubMedPubMedCentral Xue W, Zender L, Miething C, Dickins RA, Hernando E, Krizhanovsky V, et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature. 2007;445:656–60.CrossRefPubMedPubMedCentral
7.
8.
go back to reference Zacarias-Fluck M, Morancho B, Vicario R, Luque-García A, Ramón CF, Escorihuela M, et al. Effect of cellular senescence on the growth of HER2-positive breast cancers. J Natl Cancer Inst. 2015;107. doi:10.1093/jnci/djv020. Zacarias-Fluck M, Morancho B, Vicario R, Luque-García A, Ramón CF, Escorihuela M, et al. Effect of cellular senescence on the growth of HER2-positive breast cancers. J Natl Cancer Inst. 2015;107. doi:10.​1093/​jnci/​djv020.
9.
go back to reference Schelter F, Kobuch J, Moss ML, Becherer JD, Comoglio PM, Boccaccio C, et al. A disintegrin and metalloproteinase-10 (ADAM-10) mediates DN30 antibody-induced shedding of the met surface receptor. J Biol Chem. 2010;285:26335–40.CrossRefPubMedPubMedCentral Schelter F, Kobuch J, Moss ML, Becherer JD, Comoglio PM, Boccaccio C, et al. A disintegrin and metalloproteinase-10 (ADAM-10) mediates DN30 antibody-induced shedding of the met surface receptor. J Biol Chem. 2010;285:26335–40.CrossRefPubMedPubMedCentral
10.
go back to reference Arribas J, Massague J. Transforming growth factor-alpha and beta-amyloid precursor protein share a secretory mechanism. J Cell Biol. 1995;128:433–41.CrossRefPubMed Arribas J, Massague J. Transforming growth factor-alpha and beta-amyloid precursor protein share a secretory mechanism. J Cell Biol. 1995;128:433–41.CrossRefPubMed
11.
go back to reference Schnell U, Kuipers J, Mueller JL, Veenstra-Algra A, Sivagnanam M, Giepmans BN. Absence of cell-surface EpCAM in congenital tufting enteropathy. Hum Mol Genet. 2013;22:2566–71.CrossRefPubMedPubMedCentral Schnell U, Kuipers J, Mueller JL, Veenstra-Algra A, Sivagnanam M, Giepmans BN. Absence of cell-surface EpCAM in congenital tufting enteropathy. Hum Mol Genet. 2013;22:2566–71.CrossRefPubMedPubMedCentral
12.
go back to reference Guaiquil V, Swendeman S, Yoshida T, Chavala S, Campochiaro PA, Blobel CP. ADAM9 is involved in pathological retinal neovascularization. Mol Cell Biol. 2009;29:2694–703.CrossRefPubMedPubMedCentral Guaiquil V, Swendeman S, Yoshida T, Chavala S, Campochiaro PA, Blobel CP. ADAM9 is involved in pathological retinal neovascularization. Mol Cell Biol. 2009;29:2694–703.CrossRefPubMedPubMedCentral
13.
go back to reference Reiss K, Saftig P. The “a disintegrin and metalloprotease” (ADAM) family of sheddases: physiological and cellular functions. Semin Cell Dev Biol. 2009;20:126–37.CrossRefPubMed Reiss K, Saftig P. The “a disintegrin and metalloprotease” (ADAM) family of sheddases: physiological and cellular functions. Semin Cell Dev Biol. 2009;20:126–37.CrossRefPubMed
14.
go back to reference Effenberger T, Von Der Heyde J, Bartsch K, Garbers C, Schulze-Osthoff K, Chalaris A, et al. Senescence-associated release of transmembrane proteins involves proteolytic processing by ADAM17 and microvesicle shedding. FASEB J. 2014;28:4847–56.CrossRefPubMed Effenberger T, Von Der Heyde J, Bartsch K, Garbers C, Schulze-Osthoff K, Chalaris A, et al. Senescence-associated release of transmembrane proteins involves proteolytic processing by ADAM17 and microvesicle shedding. FASEB J. 2014;28:4847–56.CrossRefPubMed
15.
go back to reference Pedersen K, Angelini PD, Laos S, Bach-Faig A, Cunningham MP, Ferrer-Ramón C, et al. A naturally occurring HER2 carboxy-terminal fragment promotes mammary tumor growth and metastasis. Mol Cell Biol. 2009;29:3319–31.CrossRefPubMedPubMedCentral Pedersen K, Angelini PD, Laos S, Bach-Faig A, Cunningham MP, Ferrer-Ramón C, et al. A naturally occurring HER2 carboxy-terminal fragment promotes mammary tumor growth and metastasis. Mol Cell Biol. 2009;29:3319–31.CrossRefPubMedPubMedCentral
17.
go back to reference Meerbrey KL, Hu G, Kessler JD, Roarty K, Li MZ, Fang JE, et al. The pINDUCER lentiviral toolkit for inducible RNA interference in vitro and in vivo. Proc Natl Acad Sci U S A. 2011;108:3665–70.CrossRefPubMedPubMedCentral Meerbrey KL, Hu G, Kessler JD, Roarty K, Li MZ, Fang JE, et al. The pINDUCER lentiviral toolkit for inducible RNA interference in vitro and in vivo. Proc Natl Acad Sci U S A. 2011;108:3665–70.CrossRefPubMedPubMedCentral
18.
go back to reference Sahin U, Weskamp G, Kelly K, Zhou H-M, Higashiyama S, Peschon J, et al. Distinct roles for ADAM10 and ADAM17 in ectodomain shedding of six EGFR ligands. J Cell Biol. 2004;164:769–79.CrossRefPubMedPubMedCentral Sahin U, Weskamp G, Kelly K, Zhou H-M, Higashiyama S, Peschon J, et al. Distinct roles for ADAM10 and ADAM17 in ectodomain shedding of six EGFR ligands. J Cell Biol. 2004;164:769–79.CrossRefPubMedPubMedCentral
19.
go back to reference Merlos-Suárez AA, Fernández-Larrea JJ, Reddy PP, Baselga JJ, Arribas JJ. Pro-tumor necrosis factor-alpha processing activity is tightly controlled by a component that does not affect notch processing. J Biol Chem. 1998;273:24955–62.CrossRefPubMed Merlos-Suárez AA, Fernández-Larrea JJ, Reddy PP, Baselga JJ, Arribas JJ. Pro-tumor necrosis factor-alpha processing activity is tightly controlled by a component that does not affect notch processing. J Biol Chem. 1998;273:24955–62.CrossRefPubMed
20.
go back to reference Fu H-L, Valiathan RR, Arkwright R, Sohail A, Mihai C, Kumarasiri M, et al. Discoidin domain receptors: unique receptor tyrosine kinases in collagen-mediated signaling. J Biol Chem. 2013;288:7430–7.CrossRefPubMedPubMedCentral Fu H-L, Valiathan RR, Arkwright R, Sohail A, Mihai C, Kumarasiri M, et al. Discoidin domain receptors: unique receptor tyrosine kinases in collagen-mediated signaling. J Biol Chem. 2013;288:7430–7.CrossRefPubMedPubMedCentral
21.
go back to reference Weskamp G, Mendelson K, Swendeman S, Le Gall S, Ma Y, Lyman S, et al. Pathological neovascularization is reduced by inactivation of ADAM17 in endothelial cells but not in pericytes. Circ Res. 2010;106:932–40.CrossRefPubMedPubMedCentral Weskamp G, Mendelson K, Swendeman S, Le Gall S, Ma Y, Lyman S, et al. Pathological neovascularization is reduced by inactivation of ADAM17 in endothelial cells but not in pericytes. Circ Res. 2010;106:932–40.CrossRefPubMedPubMedCentral
22.
go back to reference Nath D, Williamson NJ, Jarvis R, Murphy G. Shedding of c-Met is regulated by crosstalk between a G-protein coupled receptor and the EGF receptor and is mediated by a TIMP-3 sensitive metalloproteinase. J Cell Sci. 2001;114:1213–20.PubMed Nath D, Williamson NJ, Jarvis R, Murphy G. Shedding of c-Met is regulated by crosstalk between a G-protein coupled receptor and the EGF receptor and is mediated by a TIMP-3 sensitive metalloproteinase. J Cell Sci. 2001;114:1213–20.PubMed
23.
go back to reference Maetzel D, Denzel S, Mack B, Canis M, Went P, Benk M, et al. Nuclear signalling by tumour-associated antigen EpCAM. Nat Cell Biol. 2009;11:162–71.CrossRefPubMed Maetzel D, Denzel S, Mack B, Canis M, Went P, Benk M, et al. Nuclear signalling by tumour-associated antigen EpCAM. Nat Cell Biol. 2009;11:162–71.CrossRefPubMed
24.
go back to reference Borroto A, Ruiz-Paz S, de la Torre TV, Borrell-Pages M, Merlos-Suarez A, Pandiella A, et al. Impaired trafficking and activation of tumor necrosis factor-alpha-converting enzyme in cell mutants defective in protein ectodomain shedding. J Biol Chem. 2003;278:25933–9.CrossRefPubMed Borroto A, Ruiz-Paz S, de la Torre TV, Borrell-Pages M, Merlos-Suarez A, Pandiella A, et al. Impaired trafficking and activation of tumor necrosis factor-alpha-converting enzyme in cell mutants defective in protein ectodomain shedding. J Biol Chem. 2003;278:25933–9.CrossRefPubMed
25.
go back to reference Matthews V, Schuster B, Schütze S, Bussmeyer I, Ludwig A, Hundhausen C, et al. Cellular cholesterol depletion triggers shedding of the human interleukin-6 receptor by ADAM10 and ADAM17 (TACE). J Biol Chem. 2003;278:38829–39.CrossRefPubMed Matthews V, Schuster B, Schütze S, Bussmeyer I, Ludwig A, Hundhausen C, et al. Cellular cholesterol depletion triggers shedding of the human interleukin-6 receptor by ADAM10 and ADAM17 (TACE). J Biol Chem. 2003;278:38829–39.CrossRefPubMed
26.
go back to reference Murai T, Maruyama Y, Mio K, Nishiyama H, Suga M, Sato C. Low cholesterol triggers membrane microdomain-dependent CD44 shedding and suppresses tumor cell migration. Biol Chem. 2011;286:1999–2007.CrossRef Murai T, Maruyama Y, Mio K, Nishiyama H, Suga M, Sato C. Low cholesterol triggers membrane microdomain-dependent CD44 shedding and suppresses tumor cell migration. Biol Chem. 2011;286:1999–2007.CrossRef
27.
go back to reference von Tresckow B, Kallen KJ, von Strandmann EP, Borchmann P, Lange H, Engert A, et al. Depletion of cellular cholesterol and lipid rafts increases shedding of CD30. J Immunol. 2004;172:4324–31.CrossRef von Tresckow B, Kallen KJ, von Strandmann EP, Borchmann P, Lange H, Engert A, et al. Depletion of cellular cholesterol and lipid rafts increases shedding of CD30. J Immunol. 2004;172:4324–31.CrossRef
28.
go back to reference Tellier E, Canault M, Poggi M, Bonardo B, Nicolay A, Alessi M-C, et al. HDLs activate ADAM17-dependent shedding. J Cell Physiol. 2008;214:687–93.CrossRefPubMed Tellier E, Canault M, Poggi M, Bonardo B, Nicolay A, Alessi M-C, et al. HDLs activate ADAM17-dependent shedding. J Cell Physiol. 2008;214:687–93.CrossRefPubMed
29.
go back to reference Ohlig S, Farshi P, Pickhinke U, van den Boom J, Höing S, Jakuschev S, et al. Sonic hedgehog shedding results in functional activation of the solubilized protein. Dev Cell. 2011;20:764–74.CrossRefPubMed Ohlig S, Farshi P, Pickhinke U, van den Boom J, Höing S, Jakuschev S, et al. Sonic hedgehog shedding results in functional activation of the solubilized protein. Dev Cell. 2011;20:764–74.CrossRefPubMed
30.
go back to reference Maretzky T, Schulte M, Ludwig A, Rose-John S, Blobel C, Hartmann D, et al. L1 is sequentially processed by two differently activated metalloproteases and presenilin/gamma-secretase and regulates neural cell adhesion, cell migration, and neurite outgrowth. Mol Cell Biol. 2005;25:9040–53.CrossRefPubMedPubMedCentral Maretzky T, Schulte M, Ludwig A, Rose-John S, Blobel C, Hartmann D, et al. L1 is sequentially processed by two differently activated metalloproteases and presenilin/gamma-secretase and regulates neural cell adhesion, cell migration, and neurite outgrowth. Mol Cell Biol. 2005;25:9040–53.CrossRefPubMedPubMedCentral
31.
go back to reference Sene A, Khan AA, Cox D, Nakamura REI, Santeford A, Kim BM, et al. Impaired cholesterol efflux in senescent macrophages promotes age-related macular degeneration. Cell Metab. 2013;17:549–61.CrossRefPubMedPubMedCentral Sene A, Khan AA, Cox D, Nakamura REI, Santeford A, Kim BM, et al. Impaired cholesterol efflux in senescent macrophages promotes age-related macular degeneration. Cell Metab. 2013;17:549–61.CrossRefPubMedPubMedCentral
32.
go back to reference Cadenas C, Vosbeck S, Hein EM, Hellwig B, Langer A, Hayen H, et al. Glycerophospholipid profile in oncogene-induced senescence. Biochim Biophys Acta. 2012;1821:1256–68.CrossRefPubMed Cadenas C, Vosbeck S, Hein EM, Hellwig B, Langer A, Hayen H, et al. Glycerophospholipid profile in oncogene-induced senescence. Biochim Biophys Acta. 2012;1821:1256–68.CrossRefPubMed
33.
go back to reference Peschon JJ, Slack JL, Reddy P, Stocking KL, Sunnarborg SW, Lee DC, et al. An essential role for ectodomain shedding in mammalian development. Science. 1998;282:1281–4.CrossRefPubMed Peschon JJ, Slack JL, Reddy P, Stocking KL, Sunnarborg SW, Lee DC, et al. An essential role for ectodomain shedding in mammalian development. Science. 1998;282:1281–4.CrossRefPubMed
34.
go back to reference Campisi J. Cellular senescence: putting the paradoxes in perspective. Curr Opin Genet Dev. 2011;21:107–12.CrossRefPubMed Campisi J. Cellular senescence: putting the paradoxes in perspective. Curr Opin Genet Dev. 2011;21:107–12.CrossRefPubMed
35.
36.
go back to reference Arribas J, Esselens C. ADAM17 as a therapeutic target in multiple diseases. Curr Pharm Des. 2009;15:2319–35.CrossRefPubMed Arribas J, Esselens C. ADAM17 as a therapeutic target in multiple diseases. Curr Pharm Des. 2009;15:2319–35.CrossRefPubMed
37.
go back to reference Richards FM, Tape CJ, Jodrell DI, Murphy G. Anti-tumour effects of a specific anti-ADAM17 antibody in an ovarian cancer model in vivo. PLoS One. 2012;7, e40597.CrossRefPubMedPubMedCentral Richards FM, Tape CJ, Jodrell DI, Murphy G. Anti-tumour effects of a specific anti-ADAM17 antibody in an ovarian cancer model in vivo. PLoS One. 2012;7, e40597.CrossRefPubMedPubMedCentral
Metadata
Title
Role of ADAM17 in the non-cell autonomous effects of oncogene-induced senescence
Authors
Beatriz Morancho
Águeda Martínez-Barriocanal
Josep Villanueva
Joaquín Arribas
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2015
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-015-0619-7

Other articles of this Issue 1/2015

Breast Cancer Research 1/2015 Go to the issue
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