Skip to main content
Top
Published in: BMC Cancer 1/2017

Open Access 01-12-2017 | Research article

Lysosome trafficking is necessary for EGF-driven invasion and is regulated by p38 MAPK and Na+/H+ exchangers

Authors: Samantha S. Dykes, Joshua J. Steffan, James A. Cardelli

Published in: BMC Cancer | Issue 1/2017

Login to get access

Abstract

Background

Tumor invasion through a basement membrane is one of the earliest steps in metastasis, and growth factors, such as Epidermal Growth Factor (EGF) and Hepatocyte Growth Factor (HGF), stimulate this process in a majority of solid tumors. Basement membrane breakdown is one of the hallmarks of invasion; therefore, tumor cells secrete a variety of proteases to aid in this process, including lysosomal proteases. Previous studies demonstrated that peripheral lysosome distribution coincides with the release of lysosomal cathepsins.

Methods

Immunofluorescence microscopy, western blot, and 2D and 3D cell culture techniques were performed to evaluate the effects of EGF on lysosome trafficking and cell motility and invasion.

Results

EGF-mediated lysosome trafficking, protease secretion, and invasion is regulated by the activity of p38 mitogen activated protein kinase (MAPK) and sodium hydrogen exchangers (NHEs). Interestingly, EGF stimulates anterograde lysosome trafficking through a different mechanism than previously reported for HGF, suggesting that there are redundant signaling pathways that control lysosome positioning and trafficking in tumor cells.

Conclusions

These data suggest that EGF stimulation induces peripheral (anterograde) lysosome trafficking, which is critical for EGF-mediated invasion and protease release, through the activation of p38 MAPK and NHEs. Taken together, this report demonstrates that anterograde lysosome trafficking is necessary for EGF-mediated tumor invasion and begins to characterize the molecular mechanisms required for EGF-stimulated lysosome trafficking.
Appendix
Available only for authorised users
Literature
1.
go back to reference Odental J, Takes R, Friedl P. Plasticity of tumor cell invasion: governance by growth factors and cytokines. Carcinogenesis. 2016;37(12):1117–28. Odental J, Takes R, Friedl P. Plasticity of tumor cell invasion: governance by growth factors and cytokines. Carcinogenesis. 2016;37(12):1117–28.
3.
go back to reference Hynes NE, Lane HA. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer. 2005;5(5):341–54.CrossRefPubMed Hynes NE, Lane HA. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer. 2005;5(5):341–54.CrossRefPubMed
4.
go back to reference Di Lorenzo G, Tortora G, D'Armiento FP, De Rosa G, Staibano S, Autorino R, D'Armiento M, De Laurentiis M, De Placido S, Catalano G, et al. Expression of epidermal growth factor receptor correlates with disease relapse and progression to androgen-independence in human prostate cancer. Clin Cancer Res. 2002;8(11):3438–44.PubMed Di Lorenzo G, Tortora G, D'Armiento FP, De Rosa G, Staibano S, Autorino R, D'Armiento M, De Laurentiis M, De Placido S, Catalano G, et al. Expression of epidermal growth factor receptor correlates with disease relapse and progression to androgen-independence in human prostate cancer. Clin Cancer Res. 2002;8(11):3438–44.PubMed
5.
go back to reference Normanno N, De Luca A, Bianco C, Strizzi L, Mancino M, Maiello MR, Carotenuto A, De Feo G, Caponigro F, Salomon DS. Epidermal growth factor receptor (EGFR) signaling in cancer. Gene. 2006;366(1):2–16.CrossRefPubMed Normanno N, De Luca A, Bianco C, Strizzi L, Mancino M, Maiello MR, Carotenuto A, De Feo G, Caponigro F, Salomon DS. Epidermal growth factor receptor (EGFR) signaling in cancer. Gene. 2006;366(1):2–16.CrossRefPubMed
6.
go back to reference Authier F, Chauvet G. In vitro endosome-lysosome transfer of dephosphorylated EGF receptor and Shc in rat liver. FEBS Lett. 1999;461(1–2):25–31.CrossRefPubMed Authier F, Chauvet G. In vitro endosome-lysosome transfer of dephosphorylated EGF receptor and Shc in rat liver. FEBS Lett. 1999;461(1–2):25–31.CrossRefPubMed
7.
go back to reference Authier F, Metioui M, Bell AW, Mort JS. Negative regulation of epidermal growth factor signaling by selective proteolytic mechanisms in the endosome mediated by cathepsin B. J Biol Chem. 1999;274(47):33723–31.CrossRefPubMed Authier F, Metioui M, Bell AW, Mort JS. Negative regulation of epidermal growth factor signaling by selective proteolytic mechanisms in the endosome mediated by cathepsin B. J Biol Chem. 1999;274(47):33723–31.CrossRefPubMed
8.
go back to reference Sorkin A, Krolenko S, Kudrjavtceva N, Lazebnik J, Teslenko L, Soderquist AM, Nikolsky N. Recycling of epidermal growth factor-receptor complexes in A431 cells: identification of dual pathways. J Cell Biol. 1991;112(1):55–63.CrossRefPubMed Sorkin A, Krolenko S, Kudrjavtceva N, Lazebnik J, Teslenko L, Soderquist AM, Nikolsky N. Recycling of epidermal growth factor-receptor complexes in A431 cells: identification of dual pathways. J Cell Biol. 1991;112(1):55–63.CrossRefPubMed
9.
go back to reference Friedl P, Wolf K. Tube travel: the role of proteases in individual and collective cancer cell invasion. Cancer Res. 2008;68(18):7247–9.CrossRefPubMed Friedl P, Wolf K. Tube travel: the role of proteases in individual and collective cancer cell invasion. Cancer Res. 2008;68(18):7247–9.CrossRefPubMed
10.
go back to reference Koblinski JE, Ahram M, Sloane BF. Unraveling the role of proteases in cancer. Clin Chim Acta. 2000;291(2):113–35.CrossRefPubMed Koblinski JE, Ahram M, Sloane BF. Unraveling the role of proteases in cancer. Clin Chim Acta. 2000;291(2):113–35.CrossRefPubMed
11.
go back to reference Sabeh F, Shimizu-Hirota R, Weiss SJ. Protease-dependent versus -independent cancer cell invasion programs: three-dimensional amoeboid movement revisited. J Cell Biol. 2009;185(1):11–9.CrossRefPubMedPubMedCentral Sabeh F, Shimizu-Hirota R, Weiss SJ. Protease-dependent versus -independent cancer cell invasion programs: three-dimensional amoeboid movement revisited. J Cell Biol. 2009;185(1):11–9.CrossRefPubMedPubMedCentral
12.
go back to reference Jordens I, Fernandez-Borja M, Marsman M, Dusseljee S, Janssen L, Calafat J, Janssen H, Wubbolts R, Neefjes J. The Rab7 effector protein RILP controls lysosomal transport by inducing the recruitment of dynein-dynactin motors. Curr Biol. 2001;11(21):1680–5.CrossRefPubMed Jordens I, Fernandez-Borja M, Marsman M, Dusseljee S, Janssen L, Calafat J, Janssen H, Wubbolts R, Neefjes J. The Rab7 effector protein RILP controls lysosomal transport by inducing the recruitment of dynein-dynactin motors. Curr Biol. 2001;11(21):1680–5.CrossRefPubMed
13.
go back to reference Tanaka Y, Kanai Y, Okada Y, Nonaka S, Takeda S, Harada A, Hirokawa N. Targeted disruption of mouse conventional kinesin heavy chain, kif5B, results in abnormal perinuclear clustering of mitochondria. Cell. 1998;93(7):1147–58.CrossRefPubMed Tanaka Y, Kanai Y, Okada Y, Nonaka S, Takeda S, Harada A, Hirokawa N. Targeted disruption of mouse conventional kinesin heavy chain, kif5B, results in abnormal perinuclear clustering of mitochondria. Cell. 1998;93(7):1147–58.CrossRefPubMed
14.
go back to reference Steffan JJ, Cardelli JA. Thiazolidinediones induce Rab7-RILP-MAPK-dependent juxtanuclear lysosome aggregation and reduce tumor cell invasion. Traffic. 2010;11(2):274–86.CrossRefPubMed Steffan JJ, Cardelli JA. Thiazolidinediones induce Rab7-RILP-MAPK-dependent juxtanuclear lysosome aggregation and reduce tumor cell invasion. Traffic. 2010;11(2):274–86.CrossRefPubMed
15.
go back to reference Steffan JJ, Dykes SS, Coleman DT, Adams LK, Rogers D, Carroll JL, Williams BJ, Cardelli JA. Supporting a Role for the GTPase Rab7 in Prostate Cancer Progression. PLoS One. 2014;9(2):e87882.CrossRefPubMedPubMedCentral Steffan JJ, Dykes SS, Coleman DT, Adams LK, Rogers D, Carroll JL, Williams BJ, Cardelli JA. Supporting a Role for the GTPase Rab7 in Prostate Cancer Progression. PLoS One. 2014;9(2):e87882.CrossRefPubMedPubMedCentral
16.
go back to reference Steffan JJ, Snider JL, Skalli O, Welbourne T, Cardelli JA. Na+/H+ Exchangers and RhoA Regulate Acidic Extracellular pH-Induced Lysosome Trafficking in Prostate Cancer Cells. Traffic. 2009;10(6):737–53.CrossRefPubMed Steffan JJ, Snider JL, Skalli O, Welbourne T, Cardelli JA. Na+/H+ Exchangers and RhoA Regulate Acidic Extracellular pH-Induced Lysosome Trafficking in Prostate Cancer Cells. Traffic. 2009;10(6):737–53.CrossRefPubMed
17.
go back to reference Steffan JJ, Williams BC, Welbourne T, Cardelli JA. HGF-induced invasion by prostate tumor cells requires anterograde lysosome trafficking and activity of Na+−H+ exchangers. J Cell Sci. 2010;123(Pt 7):1151–9.CrossRefPubMedPubMedCentral Steffan JJ, Williams BC, Welbourne T, Cardelli JA. HGF-induced invasion by prostate tumor cells requires anterograde lysosome trafficking and activity of Na+−H+ exchangers. J Cell Sci. 2010;123(Pt 7):1151–9.CrossRefPubMedPubMedCentral
18.
go back to reference Tu C, Ortega-Cava CF, Chen G, Fernandes ND, Cavallo-Medved D, Sloane BF, Band V, Band H. Lysosomal cathepsin B participates in the podosome-mediated extracellular matrix degradation and invasion via secreted lysosomes in v-Src fibroblasts. Cancer Res. 2008;68(22):9147–56.CrossRefPubMedPubMedCentral Tu C, Ortega-Cava CF, Chen G, Fernandes ND, Cavallo-Medved D, Sloane BF, Band V, Band H. Lysosomal cathepsin B participates in the podosome-mediated extracellular matrix degradation and invasion via secreted lysosomes in v-Src fibroblasts. Cancer Res. 2008;68(22):9147–56.CrossRefPubMedPubMedCentral
19.
go back to reference Miyake H, Hara I, Eto H. Serum level of cathepsin B and its density in men with prostate cancer as novel markers of disease progression. Anticancer Res. 2004;24(4):2573–7.PubMed Miyake H, Hara I, Eto H. Serum level of cathepsin B and its density in men with prostate cancer as novel markers of disease progression. Anticancer Res. 2004;24(4):2573–7.PubMed
20.
go back to reference Victor BC, Anbalagan A, Mohamed MM, Sloane BF, Cavallo-Medved D. Inhibition of cathepsin B activity attenuates extracellular matrix degradation and inflammatory breast cancer invasion. Breast Cancer Res. 2011;13(6):R115.CrossRefPubMedPubMedCentral Victor BC, Anbalagan A, Mohamed MM, Sloane BF, Cavallo-Medved D. Inhibition of cathepsin B activity attenuates extracellular matrix degradation and inflammatory breast cancer invasion. Breast Cancer Res. 2011;13(6):R115.CrossRefPubMedPubMedCentral
21.
go back to reference Withana NP, Blum G, Sameni M, Slaney C, Anbalagan A, Olive MB, Bidwell BN, Edgington L, Wang L, Moin K, et al. Cathepsin B inhibition limits bone metastasis in breast cancer. Cancer Res. 2012;72(5):1199–209.CrossRefPubMedPubMedCentral Withana NP, Blum G, Sameni M, Slaney C, Anbalagan A, Olive MB, Bidwell BN, Edgington L, Wang L, Moin K, et al. Cathepsin B inhibition limits bone metastasis in breast cancer. Cancer Res. 2012;72(5):1199–209.CrossRefPubMedPubMedCentral
22.
go back to reference Jedeszko C, Sameni M, Olive MB, Moin K, Sloane BF. Visualizing protease activity in living cells: from two dimensions to four dimensions. Curr Protoc Cell Biol 2008;Chapter 4:Unit. 4:20. Jedeszko C, Sameni M, Olive MB, Moin K, Sloane BF. Visualizing protease activity in living cells: from two dimensions to four dimensions. Curr Protoc Cell Biol 2008;Chapter 4:Unit. 4:20.
23.
go back to reference Glunde K, Guggino SE, Solaiyappan M, Pathak AP, Ichikawa Y, Bhujwalla ZM. Extracellular acidification alters lysosomal trafficking in human breast cancer cells. Neoplasia. 2003;5(6):533–45.CrossRefPubMedPubMedCentral Glunde K, Guggino SE, Solaiyappan M, Pathak AP, Ichikawa Y, Bhujwalla ZM. Extracellular acidification alters lysosomal trafficking in human breast cancer cells. Neoplasia. 2003;5(6):533–45.CrossRefPubMedPubMedCentral
24.
go back to reference Snider JL, Allison C, Bellaire BH, Ferrero RL, Cardelli JA. The beta1 integrin activates JNK independent of CagA, and JNK activation is required for Helicobacter pylori CagA+−induced motility of gastric cancer cells. J Biol Chem. 2008;283(20):13952–63.CrossRefPubMedPubMedCentral Snider JL, Allison C, Bellaire BH, Ferrero RL, Cardelli JA. The beta1 integrin activates JNK independent of CagA, and JNK activation is required for Helicobacter pylori CagA+−induced motility of gastric cancer cells. J Biol Chem. 2008;283(20):13952–63.CrossRefPubMedPubMedCentral
25.
go back to reference Coleman DT, Bigelow R, Cardelli JA. Inhibition of fatty acid synthase by luteolin post-transcriptionally down-regulates c-Met expression independent of proteosomal/lysosomal degradation. Mol Cancer Ther. 2009;8(1):214–24.CrossRefPubMedPubMedCentral Coleman DT, Bigelow R, Cardelli JA. Inhibition of fatty acid synthase by luteolin post-transcriptionally down-regulates c-Met expression independent of proteosomal/lysosomal degradation. Mol Cancer Ther. 2009;8(1):214–24.CrossRefPubMedPubMedCentral
26.
go back to reference Duhon D, Bigelow RL, Coleman DT, Steffan JJ, Yu C, Langston W, Kevil CG, Cardelli JA. The polyphenol epigallocatechin-3-gallate affects lipid rafts to block activation of the c-Met receptor in prostate cancer cells. Mol Carcinog. 2010;49(8):739–49.PubMed Duhon D, Bigelow RL, Coleman DT, Steffan JJ, Yu C, Langston W, Kevil CG, Cardelli JA. The polyphenol epigallocatechin-3-gallate affects lipid rafts to block activation of the c-Met receptor in prostate cancer cells. Mol Carcinog. 2010;49(8):739–49.PubMed
27.
go back to reference Gan Y, Shi C, Inge L, Hibner M, Balducci J, Huang Y. Differential roles of ERK and Akt pathways in regulation of EGFR-mediated signaling and motility in prostate cancer cells. Oncogene. 2010;29(35):4947–58.CrossRefPubMed Gan Y, Shi C, Inge L, Hibner M, Balducci J, Huang Y. Differential roles of ERK and Akt pathways in regulation of EGFR-mediated signaling and motility in prostate cancer cells. Oncogene. 2010;29(35):4947–58.CrossRefPubMed
28.
go back to reference Vlahos CJ, Matter WF, Hui KY, Brown RF. A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002). J Biol Chem. 1994;269(7):5241–8.PubMed Vlahos CJ, Matter WF, Hui KY, Brown RF. A specific inhibitor of phosphatidylinositol 3-kinase, 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002). J Biol Chem. 1994;269(7):5241–8.PubMed
29.
go back to reference Favata MF, Horiuchi KY, Manos EJ, Daulerio AJ, Stradley DA, Feeser WS, Van Dyk DE, Pitts WJ, Earl RA, Hobbs F, et al. Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J Biol Chem. 1998;273(29):18623–32.CrossRefPubMed Favata MF, Horiuchi KY, Manos EJ, Daulerio AJ, Stradley DA, Feeser WS, Van Dyk DE, Pitts WJ, Earl RA, Hobbs F, et al. Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J Biol Chem. 1998;273(29):18623–32.CrossRefPubMed
30.
go back to reference Lee JC, Laydon JT, McDonnell PC, Gallagher TF, Kumar S, Green D, McNulty D, Blumenthal MJ, Heys JR, Landvatter SW, et al. A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature. 1994;372(6508):739–46.CrossRefPubMed Lee JC, Laydon JT, McDonnell PC, Gallagher TF, Kumar S, Green D, McNulty D, Blumenthal MJ, Heys JR, Landvatter SW, et al. A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature. 1994;372(6508):739–46.CrossRefPubMed
31.
32.
go back to reference KP X, FS Y. Cross talk between c-Met and epidermal growth factor receptor during retinal pigment epithelial wound healing. Invest Ophthalmol Vis Sci. 2007;48(5):2242–8.CrossRef KP X, FS Y. Cross talk between c-Met and epidermal growth factor receptor during retinal pigment epithelial wound healing. Invest Ophthalmol Vis Sci. 2007;48(5):2242–8.CrossRef
34.
go back to reference Sattler M, Pride YB, Ma P, Gramlich JL, Chu SC, Quinnan LA, Shirazian S, Liang C, Podar K, Christensen JG, et al. A novel small molecule met inhibitor induces apoptosis in cells transformed by the oncogenic TPR-MET tyrosine kinase. Cancer Res. 2003;63(17):5462–9.PubMed Sattler M, Pride YB, Ma P, Gramlich JL, Chu SC, Quinnan LA, Shirazian S, Liang C, Podar K, Christensen JG, et al. A novel small molecule met inhibitor induces apoptosis in cells transformed by the oncogenic TPR-MET tyrosine kinase. Cancer Res. 2003;63(17):5462–9.PubMed
35.
go back to reference Yaish P, Gazit A, Gilon C, Levitzki A. Blocking of EGF-dependent cell proliferation by EGF receptor kinase inhibitors. Science. 1988;242(4880):933–5.CrossRefPubMed Yaish P, Gazit A, Gilon C, Levitzki A. Blocking of EGF-dependent cell proliferation by EGF receptor kinase inhibitors. Science. 1988;242(4880):933–5.CrossRefPubMed
36.
go back to reference Coaxum SD, Garnovskaya MN, Gooz M, Baldys A, Raymond JR. Epidermal growth factor activates Na(+/)H(+) exchanger in podocytes through a mechanism that involves Janus kinase and calmodulin. Biochim Biophys Acta. 2009;1793(7):1174–81.CrossRefPubMedPubMedCentral Coaxum SD, Garnovskaya MN, Gooz M, Baldys A, Raymond JR. Epidermal growth factor activates Na(+/)H(+) exchanger in podocytes through a mechanism that involves Janus kinase and calmodulin. Biochim Biophys Acta. 2009;1793(7):1174–81.CrossRefPubMedPubMedCentral
37.
go back to reference De Giusti VC, Nolly MB, Yeves AM, Caldiz CI, Villa-Abrille MC, Chiappe de Cingolani GE, Ennis IL, Cingolani HE, Aiello EA. Aldosterone stimulates the cardiac Na(+)/H(+) exchanger via transactivation of the epidermal growth factor receptor. Hypertension. 2011;58(5):912–9.CrossRefPubMed De Giusti VC, Nolly MB, Yeves AM, Caldiz CI, Villa-Abrille MC, Chiappe de Cingolani GE, Ennis IL, Cingolani HE, Aiello EA. Aldosterone stimulates the cardiac Na(+)/H(+) exchanger via transactivation of the epidermal growth factor receptor. Hypertension. 2011;58(5):912–9.CrossRefPubMed
38.
go back to reference Vergarajauregui S, San Miguel A, Puertollano R. Activation of p38 mitogen-activated protein kinase promotes epidermal growth factor receptor internalization. Traffic. 2006;7(6):686–98.CrossRefPubMedPubMedCentral Vergarajauregui S, San Miguel A, Puertollano R. Activation of p38 mitogen-activated protein kinase promotes epidermal growth factor receptor internalization. Traffic. 2006;7(6):686–98.CrossRefPubMedPubMedCentral
39.
go back to reference Grandal MV, Grovdal LM, Henriksen L, Andersen MH, Holst MR, Madshus IH, van Deurs B. Differential Roles of Grb2 and AP-2 in p38 MAPK- and EGF-Induced EGFR Internalization. Traffic. 2012;13(4):576–85.CrossRefPubMed Grandal MV, Grovdal LM, Henriksen L, Andersen MH, Holst MR, Madshus IH, van Deurs B. Differential Roles of Grb2 and AP-2 in p38 MAPK- and EGF-Induced EGFR Internalization. Traffic. 2012;13(4):576–85.CrossRefPubMed
40.
go back to reference Winograd-Katz SE, Levitzki A. Cisplatin induces PKB/Akt activation and p38(MAPK) phosphorylation of the EGF receptor. Oncogene. 2006;25(56):7381–90.CrossRefPubMed Winograd-Katz SE, Levitzki A. Cisplatin induces PKB/Akt activation and p38(MAPK) phosphorylation of the EGF receptor. Oncogene. 2006;25(56):7381–90.CrossRefPubMed
41.
go back to reference Zwang Y, Yarden Y. p38 MAP kinase mediates stress-induced internalization of EGFR: implications for cancer chemotherapy. EMBO J. 2006;25(18):4195–206.CrossRefPubMedPubMedCentral Zwang Y, Yarden Y. p38 MAP kinase mediates stress-induced internalization of EGFR: implications for cancer chemotherapy. EMBO J. 2006;25(18):4195–206.CrossRefPubMedPubMedCentral
42.
44.
go back to reference Stommel JMKA, Ying H, et al. Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science. 2007;318:287–90.CrossRefPubMed Stommel JMKA, Ying H, et al. Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science. 2007;318:287–90.CrossRefPubMed
45.
go back to reference Milligan SA, Burke P, Coleman DT, Bigelow RL, Steffan JJ, Carroll JL, Williams BJ, Cardelli JA. The Green Tea Polyphenol EGCG Potentiates the Antiproliferative Activity of c-Met and Epidermal Growth Factor Receptor Inhibitors in Non-small Cell Lung Cancer Cells. Clin Cancer Res. 2009;15(15):4885–94.CrossRefPubMedPubMedCentral Milligan SA, Burke P, Coleman DT, Bigelow RL, Steffan JJ, Carroll JL, Williams BJ, Cardelli JA. The Green Tea Polyphenol EGCG Potentiates the Antiproliferative Activity of c-Met and Epidermal Growth Factor Receptor Inhibitors in Non-small Cell Lung Cancer Cells. Clin Cancer Res. 2009;15(15):4885–94.CrossRefPubMedPubMedCentral
46.
go back to reference Cardone RA, Casavola V, Reshkin SJ. The role of disturbed pH dynamics and the Na+/H+ exchanger in metastasis. Nat Rev Cancer. 2005;5(10):786–95.CrossRefPubMed Cardone RA, Casavola V, Reshkin SJ. The role of disturbed pH dynamics and the Na+/H+ exchanger in metastasis. Nat Rev Cancer. 2005;5(10):786–95.CrossRefPubMed
47.
go back to reference Cardone RA, Greco MR, Zeeberg K, Zaccagnino A, Saccomano M, Bellizzi A, Bruns P, Menga M, Pilarsky C, Schwab A, et al. A Novel NHE1-Centered Signaling Cassette Drives Epidermal Growth Factor Receptor-Dependent Pancreatic Tumor Metastasis and Is a Target for Combination Therapy. Neoplasia. 2015;17(2):155–66.CrossRefPubMedPubMedCentral Cardone RA, Greco MR, Zeeberg K, Zaccagnino A, Saccomano M, Bellizzi A, Bruns P, Menga M, Pilarsky C, Schwab A, et al. A Novel NHE1-Centered Signaling Cassette Drives Epidermal Growth Factor Receptor-Dependent Pancreatic Tumor Metastasis and Is a Target for Combination Therapy. Neoplasia. 2015;17(2):155–66.CrossRefPubMedPubMedCentral
48.
go back to reference Dykes SS, Gao C, Songock WK, Bigelow RL, Woude GV, Bodily JM, Cardelli JA. Zinc finger E-box binding homeobox-1 (Zeb1) drives anterograde lysosome trafficking and tumor cell invastion via upregualtion of Na+/H+ Exchanger-1 (NHE1). Mol Carcinog. 2017;56(2):722–34.CrossRefPubMed Dykes SS, Gao C, Songock WK, Bigelow RL, Woude GV, Bodily JM, Cardelli JA. Zinc finger E-box binding homeobox-1 (Zeb1) drives anterograde lysosome trafficking and tumor cell invastion via upregualtion of Na+/H+ Exchanger-1 (NHE1). Mol Carcinog. 2017;56(2):722–34.CrossRefPubMed
49.
go back to reference Denker SP, Huang DC, Orlowski J, Furthmayr H, Barber DL. Direct binding of the Na--H exchanger NHE1 to ERM proteins regulates the cortical cytoskeleton and cell shape independently of H(+) translocation. Mol Cell. 2000;6(6):1425–36.CrossRefPubMed Denker SP, Huang DC, Orlowski J, Furthmayr H, Barber DL. Direct binding of the Na--H exchanger NHE1 to ERM proteins regulates the cortical cytoskeleton and cell shape independently of H(+) translocation. Mol Cell. 2000;6(6):1425–36.CrossRefPubMed
50.
go back to reference Magalhaes MA, Larson DR, Mader CC, Bravo-Cordero JJ, Gil-Henn H, Oser M, Chen X, Koleske AJ, Condeelis J. Cortactin phosphorylation regulates cell invasion through a pH-dependent pathway. J Cell Biol. 2011;195(5):903–20.CrossRefPubMedPubMedCentral Magalhaes MA, Larson DR, Mader CC, Bravo-Cordero JJ, Gil-Henn H, Oser M, Chen X, Koleske AJ, Condeelis J. Cortactin phosphorylation regulates cell invasion through a pH-dependent pathway. J Cell Biol. 2011;195(5):903–20.CrossRefPubMedPubMedCentral
51.
go back to reference Nishimura Y, Itoh K, Yoshioka K, Uehata M, Himeno M. Small guanosine triphosphatase Rho/Rho-associated kinase as a novel regulator of intracellular redistribution of lysosomes in invasive tumor cells. Cell Tissue Res. 2000;301(3):341–51.CrossRefPubMed Nishimura Y, Itoh K, Yoshioka K, Uehata M, Himeno M. Small guanosine triphosphatase Rho/Rho-associated kinase as a novel regulator of intracellular redistribution of lysosomes in invasive tumor cells. Cell Tissue Res. 2000;301(3):341–51.CrossRefPubMed
52.
go back to reference Nishimura Y, Itoh K, Yoshioka K, Tokuda K, Himeno M. Overexpression of ROCK in human breast cancer cells: evidence that ROCK activity mediates intracellular membrane traffic of lysosomes. Pathol Oncol Res. 2003;9(2):83–95.CrossRefPubMed Nishimura Y, Itoh K, Yoshioka K, Tokuda K, Himeno M. Overexpression of ROCK in human breast cancer cells: evidence that ROCK activity mediates intracellular membrane traffic of lysosomes. Pathol Oncol Res. 2003;9(2):83–95.CrossRefPubMed
53.
go back to reference Greco MR, Antelmi E, Busco G, Guerra L, Rubino R, Casavola V, Reshkin SJ, Cardone RA. Protease activity at invadopodial focal digestive areas is dependent on NHE1-driven acidic pHe. Oncol Rep. 2014;31(2):940–6.CrossRefPubMed Greco MR, Antelmi E, Busco G, Guerra L, Rubino R, Casavola V, Reshkin SJ, Cardone RA. Protease activity at invadopodial focal digestive areas is dependent on NHE1-driven acidic pHe. Oncol Rep. 2014;31(2):940–6.CrossRefPubMed
54.
go back to reference Busco G, Cardone RA, Greco MR, Bellizzi A, Colella M, Antelmi E, Mancini MT, Dell'Aquila ME, Casavola V, Paradiso A, et al. NHE1 promotes invadopodial ECM proteolysis through acidification of the peri-invadopodial space. FASEB J. 2010;24(10):3903–15.CrossRefPubMed Busco G, Cardone RA, Greco MR, Bellizzi A, Colella M, Antelmi E, Mancini MT, Dell'Aquila ME, Casavola V, Paradiso A, et al. NHE1 promotes invadopodial ECM proteolysis through acidification of the peri-invadopodial space. FASEB J. 2010;24(10):3903–15.CrossRefPubMed
55.
go back to reference Damaghi M, Tafreshi NK, Lloyd MC, Sprung R, Estrella V, Wojtkowiak JW, Morse DL, Koomen JM, Bui MM, Gatenby RA, et al. Chronic acidosis in the tumour microenvironment selects for overexpression of LAMP2 in the plasma membrane. Nat Comms. 2015;6:8752.CrossRef Damaghi M, Tafreshi NK, Lloyd MC, Sprung R, Estrella V, Wojtkowiak JW, Morse DL, Koomen JM, Bui MM, Gatenby RA, et al. Chronic acidosis in the tumour microenvironment selects for overexpression of LAMP2 in the plasma membrane. Nat Comms. 2015;6:8752.CrossRef
56.
go back to reference Derijard B, Raingeaud J, Barrett T, IH W, Han J, Ulevitch RJ, Davis RJ. Independent human MAP-kinase signal transduction pathways defined by MEK and MKK isoforms. Science. 1995;267(5198):682–5.CrossRefPubMed Derijard B, Raingeaud J, Barrett T, IH W, Han J, Ulevitch RJ, Davis RJ. Independent human MAP-kinase signal transduction pathways defined by MEK and MKK isoforms. Science. 1995;267(5198):682–5.CrossRefPubMed
57.
go back to reference Raingeaud J, Whitmarsh AJ, Barrett T, Derijard B, Davis RJ. MKK3- and MKK6-regulated gene expression is mediated by the p38 mitogen-activated protein kinase signal transduction pathway. Mol Cell Biol. 1996;16(3):1247–55.CrossRefPubMedPubMedCentral Raingeaud J, Whitmarsh AJ, Barrett T, Derijard B, Davis RJ. MKK3- and MKK6-regulated gene expression is mediated by the p38 mitogen-activated protein kinase signal transduction pathway. Mol Cell Biol. 1996;16(3):1247–55.CrossRefPubMedPubMedCentral
58.
go back to reference Zohn IE, Li Y, Skolnik EY, Anderson KV, Han J, Niswander L. p38 and a p38-interacting protein are critical for downregulation of E-cadherin during mouse gastrulation. Cell. 2006;125(5):957–69.CrossRefPubMed Zohn IE, Li Y, Skolnik EY, Anderson KV, Han J, Niswander L. p38 and a p38-interacting protein are critical for downregulation of E-cadherin during mouse gastrulation. Cell. 2006;125(5):957–69.CrossRefPubMed
59.
go back to reference Kim MS, Lee EJ, Kim HR, Moon A. p38 kinase is a key signaling molecule for H-Ras-induced cell motility and invasive phenotype in human breast epithelial cells. Cancer Res. 2003;63(17):5454–61.PubMed Kim MS, Lee EJ, Kim HR, Moon A. p38 kinase is a key signaling molecule for H-Ras-induced cell motility and invasive phenotype in human breast epithelial cells. Cancer Res. 2003;63(17):5454–61.PubMed
60.
go back to reference Rosenthal DT, Iyer H, Escudero S, Bao L, Wu Z, Ventura AC, Kleer CG, Arruda EM, Garikipati K, Merajver SD. p38gamma promotes breast cancer cell motility and metastasis through regulation of RhoC GTPase, cytoskeletal architecture, and a novel leading edge behavior. Cancer Res. 2011;71(20):6338–49.CrossRefPubMedPubMedCentral Rosenthal DT, Iyer H, Escudero S, Bao L, Wu Z, Ventura AC, Kleer CG, Arruda EM, Garikipati K, Merajver SD. p38gamma promotes breast cancer cell motility and metastasis through regulation of RhoC GTPase, cytoskeletal architecture, and a novel leading edge behavior. Cancer Res. 2011;71(20):6338–49.CrossRefPubMedPubMedCentral
61.
go back to reference Garcia MC, Ray DM, Lackford B, Rubino M, Olden K, Roberts JD. Arachidonic acid stimulates cell adhesion through a novel p38 MAPK-RhoA signaling pathway that involves heat shock protein 27. J Biol Chem. 2009;284(31):20936–45.CrossRefPubMedPubMedCentral Garcia MC, Ray DM, Lackford B, Rubino M, Olden K, Roberts JD. Arachidonic acid stimulates cell adhesion through a novel p38 MAPK-RhoA signaling pathway that involves heat shock protein 27. J Biol Chem. 2009;284(31):20936–45.CrossRefPubMedPubMedCentral
62.
go back to reference Morfini GA, Bosco DA, Brown H, Gatto R, Kaminska A, Song Y, Molla L, Baker L, Marangoni MN, Berth S, et al. Inhibition of fast axonal transport by pathogenic SOD1 involves activation of p38 MAP kinase. PLoS One. 2013;8(6):e65235.CrossRefPubMedPubMedCentral Morfini GA, Bosco DA, Brown H, Gatto R, Kaminska A, Song Y, Molla L, Baker L, Marangoni MN, Berth S, et al. Inhibition of fast axonal transport by pathogenic SOD1 involves activation of p38 MAP kinase. PLoS One. 2013;8(6):e65235.CrossRefPubMedPubMedCentral
64.
go back to reference Bagshaw RD, Callahan JW, Mahuran DJ. The Arf-family protein, Arl8b, is involved in the spatial distribution of lysosomes. Biochem Biophys Res Commun. 2006;344(4):1186–91.CrossRefPubMed Bagshaw RD, Callahan JW, Mahuran DJ. The Arf-family protein, Arl8b, is involved in the spatial distribution of lysosomes. Biochem Biophys Res Commun. 2006;344(4):1186–91.CrossRefPubMed
65.
go back to reference Dykes SS, Gray AL, Coleman DT, Saxena M, Stephens CA, Carroll JL, Pruitt K, Cardelli JA. The Arf-like GTPase Arl8b is essential for three-dimensional invasive growth of prostate cancer in vitro and xenograft formation and growth in vivo. Oncotarget. 2016;7(21):31037–52.CrossRefPubMedPubMedCentral Dykes SS, Gray AL, Coleman DT, Saxena M, Stephens CA, Carroll JL, Pruitt K, Cardelli JA. The Arf-like GTPase Arl8b is essential for three-dimensional invasive growth of prostate cancer in vitro and xenograft formation and growth in vivo. Oncotarget. 2016;7(21):31037–52.CrossRefPubMedPubMedCentral
66.
go back to reference Artym VV, Zhang Y, Seillier-Moiseiwitsch F, Yamada KM, Mueller SC. Dynamic interactions of cortactin and membrane type 1 matrix metalloproteinase at invadopodia: defining the stages of invadopodia formation and function. Cancer Res. 2006;66(6):3034–43.CrossRefPubMed Artym VV, Zhang Y, Seillier-Moiseiwitsch F, Yamada KM, Mueller SC. Dynamic interactions of cortactin and membrane type 1 matrix metalloproteinase at invadopodia: defining the stages of invadopodia formation and function. Cancer Res. 2006;66(6):3034–43.CrossRefPubMed
67.
go back to reference Sung BH, Zhu X, Kaverina I, Weaver AM. Cortactin controls cell motility and lamellipodial dynamics by regulating ECM secretion. Curr Biol. 2011;21(17):1460–9.CrossRefPubMedPubMedCentral Sung BH, Zhu X, Kaverina I, Weaver AM. Cortactin controls cell motility and lamellipodial dynamics by regulating ECM secretion. Curr Biol. 2011;21(17):1460–9.CrossRefPubMedPubMedCentral
68.
go back to reference Ritter AT, Asano Y, Stinchcombe JC, Dieckmann NM, Chen BC, Gawden-Bone C, van Engelenburg S, Legant W, Gao L, Davidson MW, et al. Actin depletion initiates events leading to granule secretion at the immunological synapse. Immunity. 2015;42(5):864–76.CrossRefPubMedPubMedCentral Ritter AT, Asano Y, Stinchcombe JC, Dieckmann NM, Chen BC, Gawden-Bone C, van Engelenburg S, Legant W, Gao L, Davidson MW, et al. Actin depletion initiates events leading to granule secretion at the immunological synapse. Immunity. 2015;42(5):864–76.CrossRefPubMedPubMedCentral
69.
go back to reference Wheeler DL, Dunn EF, Harari PM. Understanding resistance to EGFR inhibitors-impact on future treatment strategies. Nat Rev Clin Oncol. 2010;7(9):493–507.CrossRefPubMedPubMedCentral Wheeler DL, Dunn EF, Harari PM. Understanding resistance to EGFR inhibitors-impact on future treatment strategies. Nat Rev Clin Oncol. 2010;7(9):493–507.CrossRefPubMedPubMedCentral
Metadata
Title
Lysosome trafficking is necessary for EGF-driven invasion and is regulated by p38 MAPK and Na+/H+ exchangers
Authors
Samantha S. Dykes
Joshua J. Steffan
James A. Cardelli
Publication date
01-12-2017
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2017
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-017-3660-3

Other articles of this Issue 1/2017

BMC Cancer 1/2017 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