Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

Tspan8 and Tspan8/CD151 knockout mice unravel the contribution of tumor and host exosomes to tumor progression

Authors: Kun Zhao, Zhe Wang, Thilo Hackert, Claudia Pitzer, Margot Zöller

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

The tetraspanins Tspan8 and CD151 promote metastasis, exosomes (Exo) being suggested to be important in the crosstalk between tumor and host. The contribution of Tspan8 and CD151 to host versus tumor-derived exosome (TEX) activities being not defined, we approached the questions using 3-methylcholanthrene-induced (MCA) tumors from wt, Tspan8ko, CD151ko and Tspan8/CD151 (db)ko mice, implanted into tetraspanin-competent and deficient hosts.

Methods

Tumor growth and dissemination, hematopoiesis and angiogenesis were surveyed in wild type (wt), Tspan8ko, CD151ko and dbko mice bearing tetraspanin-competent and -deficient MCA tumors. In vitro studies using tumor cells, bone marrow cells (BMC) and endothelial cells (EC) elaborated the mechanism of serum (s)Exo- and TEX-induced target modulation.

Results

Tumors grew in autochthonous and syngeneic hosts differing in Tspan8- and/or CD151-competence. However, Tspan8ko- and/or CD151ko-tumor cell dissemination and settlement in metastatic organs was significantly reduced in the autochthonous host, and less severely in the wt-host. Impaired wt-MCA tumor dissemination in the ko-host confirmed a contribution of host- and tumor-Tspan8/-CD151 to tumor cell dissemination, delivery of sExo and TEX being severely impaired by a Tspan8ko/CD151ko. Coculturing tumor cells, BMC and EC with sExo and TEX revealed minor defects in epithelial mesenchymal transition and apoptosis resistance of ko tumors. Strongly reduced migratory and invasive capacity of Tspan8ko/CD151ko-MCA relies on distorted associations with integrins and CAM and missing Tspan8/CD151-promoted recruitment of proteases. The defects, differing between Tspan8ko- and CD151ko-MCA, were rescued by wt-TEX and, less efficiently Tspan8ko- and CD151ko-TEX. Minor defects in hematopoietic progenitor maturation were based on the missing association of hematopoietic growth factors /− receptors with CD151 and, less pronounced, Tspan8. Rescue of impaired angiogenesis in ko mice by wt-sExo and promotion of angiogenesis by TEX depended on the association of Tspan8 and CD151 with GPCR and RTK in EC and tumor cells.

Conclusions

Tspan8-/CD151-TEX play central roles in tumor progression. Tspan8-/CD151-sExo and TEX contribute by stimulating angiogenesis. Tspan8 and CD151 fulfill these tasks by associating with function-relevant proteins, the additive impact of Tspan8 and CD151 relying on differences in preferred associations. The distinct Tspan8 and CD151 contributions suggest a blockade of TEX-Tspan8 and -CD151 promising for therapeutic intervention.
Appendix
Available only for authorised users
Literature
1.
go back to reference McAllister SS, Weinberg RA. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cell Biol. 2014;16:717–27.PubMedPubMedCentralCrossRef McAllister SS, Weinberg RA. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cell Biol. 2014;16:717–27.PubMedPubMedCentralCrossRef
2.
go back to reference Gacche RN, Meshram RJ. Targeting tumor micro-environment for design and development of novel anti-angiogenic agents arresting tumor growth. Prog Biophys Mol Biol. 2013;113:333–54.PubMedCrossRef Gacche RN, Meshram RJ. Targeting tumor micro-environment for design and development of novel anti-angiogenic agents arresting tumor growth. Prog Biophys Mol Biol. 2013;113:333–54.PubMedCrossRef
3.
go back to reference Saleem SN, Abdel-Mageed AB. Tumor-derived exosomes in oncogenic reprogramming and cancer progression. Cell Mol Life Sci. 2015;72:1–10.PubMedCrossRef Saleem SN, Abdel-Mageed AB. Tumor-derived exosomes in oncogenic reprogramming and cancer progression. Cell Mol Life Sci. 2015;72:1–10.PubMedCrossRef
6.
go back to reference Milane L, Singh A, Mattheolabakis G, Suresh M, Amiji MM. Exosome mediated communication within the tumor microenvironment. J Control Release. 2015;219:278–94.PubMedCrossRef Milane L, Singh A, Mattheolabakis G, Suresh M, Amiji MM. Exosome mediated communication within the tumor microenvironment. J Control Release. 2015;219:278–94.PubMedCrossRef
7.
go back to reference Kowal J, Tkach M, Théry C. Biogenesis and secretion of exosomes. Curr Opin Cell Biol. 2014;29:116–25.PubMedCrossRef Kowal J, Tkach M, Théry C. Biogenesis and secretion of exosomes. Curr Opin Cell Biol. 2014;29:116–25.PubMedCrossRef
8.
go back to reference French KC, Antonyak MA, Cerione RA. Extracellular vesicle docking at the cellular port: Extracellular vesicle binding and uptake. Semin Cell Dev Biol. 2017;67:48–55.PubMedPubMedCentralCrossRef French KC, Antonyak MA, Cerione RA. Extracellular vesicle docking at the cellular port: Extracellular vesicle binding and uptake. Semin Cell Dev Biol. 2017;67:48–55.PubMedPubMedCentralCrossRef
9.
go back to reference Xu R, Greening DW, Zhu HJ, Takahashi N, Simpson RJ. Extracellular vesicle isolation and characterization: toward clinical application. J Clin Invest. 2016;126:1152–62.PubMedPubMedCentralCrossRef Xu R, Greening DW, Zhu HJ, Takahashi N, Simpson RJ. Extracellular vesicle isolation and characterization: toward clinical application. J Clin Invest. 2016;126:1152–62.PubMedPubMedCentralCrossRef
11.
go back to reference Record M, Carayon K, Poirot M, Silvente-Poirot S. Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochim Biophys Acta. 2014;1841:108–20.PubMedCrossRef Record M, Carayon K, Poirot M, Silvente-Poirot S. Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies. Biochim Biophys Acta. 2014;1841:108–20.PubMedCrossRef
12.
13.
go back to reference Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.PubMedCrossRef Colombo M, Raposo G, Théry C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 2014;30:255–89.PubMedCrossRef
14.
go back to reference Mazurov D, Barbashova L, Filatov A. Tetraspanin protein CD9 interacts with metalloprotease CD10 and enhances its release via exosomes. FEBS J. 2013;280:1200–13.PubMedCrossRef Mazurov D, Barbashova L, Filatov A. Tetraspanin protein CD9 interacts with metalloprotease CD10 and enhances its release via exosomes. FEBS J. 2013;280:1200–13.PubMedCrossRef
15.
go back to reference Rana S, Yue S, Stadel D, Zöller M. Toward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection. Int J Biochem Cell Biol. 2012;44:1574–84.PubMedCrossRef Rana S, Yue S, Stadel D, Zöller M. Toward tailored exosomes: the exosomal tetraspanin web contributes to target cell selection. Int J Biochem Cell Biol. 2012;44:1574–84.PubMedCrossRef
16.
go back to reference Stipp CS, Kolesnikova TV, Hemler ME. Functional domains in tetraspanin proteins. Trends Biochem Sci. 2003;28:106–12.PubMedCrossRef Stipp CS, Kolesnikova TV, Hemler ME. Functional domains in tetraspanin proteins. Trends Biochem Sci. 2003;28:106–12.PubMedCrossRef
17.
go back to reference Hemler ME. Tetraspanin functions and associated microdomains. Nat Rev Mol Cell Biol. 2005;6:801–11.PubMedCrossRef Hemler ME. Tetraspanin functions and associated microdomains. Nat Rev Mol Cell Biol. 2005;6:801–11.PubMedCrossRef
18.
go back to reference Bassani S, Cingolani LA. Tetraspanins: Interactions and interplay with integrins. Int J Biochem Cell Biol. 2012;44:703–8.PubMedCrossRef Bassani S, Cingolani LA. Tetraspanins: Interactions and interplay with integrins. Int J Biochem Cell Biol. 2012;44:703–8.PubMedCrossRef
19.
go back to reference Berditchevski F, Odintsova E. Tetraspanins as regulators of protein trafficking. Traffic. 2007;8:89–96.PubMedCrossRef Berditchevski F, Odintsova E. Tetraspanins as regulators of protein trafficking. Traffic. 2007;8:89–96.PubMedCrossRef
20.
go back to reference Yáñez-Mó M, Gutiérrez-López MD, Cabañas C. Functional interplay between tetraspanins and proteases. Cell Mol Life Sci. 2011;68:3323–35.PubMedCrossRef Yáñez-Mó M, Gutiérrez-López MD, Cabañas C. Functional interplay between tetraspanins and proteases. Cell Mol Life Sci. 2011;68:3323–35.PubMedCrossRef
21.
go back to reference Yue S, Zhao K, Erb U, Rana S, Zöller M. Joint features and complementarities of Tspan8 and CD151 revealed in knockdown and knockout models. Biochem Soc Trans. 2017;45:437–47.PubMedCrossRef Yue S, Zhao K, Erb U, Rana S, Zöller M. Joint features and complementarities of Tspan8 and CD151 revealed in knockdown and knockout models. Biochem Soc Trans. 2017;45:437–47.PubMedCrossRef
22.
go back to reference Sadej R, Grudowska A, Turczyk L, Kordek R, Romanska HM. CD151 in cancer progression and metastasis: a complex scenario. Lab Investig. 2014;94:41–51.PubMedCrossRef Sadej R, Grudowska A, Turczyk L, Kordek R, Romanska HM. CD151 in cancer progression and metastasis: a complex scenario. Lab Investig. 2014;94:41–51.PubMedCrossRef
23.
go back to reference Zöller M. Tetraspanins: push and pull in suppressing and promoting metastasis. Nat Rev Cancer. 2009;9:40–55.PubMedCrossRef Zöller M. Tetraspanins: push and pull in suppressing and promoting metastasis. Nat Rev Cancer. 2009;9:40–55.PubMedCrossRef
24.
go back to reference Hasegawa M, Furuya M, Kasuya Y, Nishiyama M, Sugiura T, Nikaido T, et al. CD151 dynamics in carcinoma-stroma interaction: integrin expression, adhesion strength and proteolytic activity. Lab Investig. 2007;87:882–92.PubMedCrossRef Hasegawa M, Furuya M, Kasuya Y, Nishiyama M, Sugiura T, Nikaido T, et al. CD151 dynamics in carcinoma-stroma interaction: integrin expression, adhesion strength and proteolytic activity. Lab Investig. 2007;87:882–92.PubMedCrossRef
25.
go back to reference Ke AW, Shi GM, Zhou J, Huang XY, Shi YH, Ding ZB, et al. CD151 amplifies signaling by integrin α6β1 to PI3K and induces the epithelial-mesenchymal transition in HCC cells. Gastroenterology. 2011;140:1629–41.CrossRefPubMed Ke AW, Shi GM, Zhou J, Huang XY, Shi YH, Ding ZB, et al. CD151 amplifies signaling by integrin α6β1 to PI3K and induces the epithelial-mesenchymal transition in HCC cells. Gastroenterology. 2011;140:1629–41.CrossRefPubMed
26.
go back to reference Berditchevski F. Complexes of tetraspanins with integrins: more than meets the eye. J Cell Sci. 2001;114:4143–51.PubMed Berditchevski F. Complexes of tetraspanins with integrins: more than meets the eye. J Cell Sci. 2001;114:4143–51.PubMed
27.
go back to reference Novitskaya V, Romanska H, Kordek R, Potemski P, Kusińska R, Parsons M, et al. Integrin α3β1-CD151 complex regulates dimerization of ErbB2 via RhoA. Oncogene. 2014;33:2779–89.PubMedCrossRef Novitskaya V, Romanska H, Kordek R, Potemski P, Kusińska R, Parsons M, et al. Integrin α3β1-CD151 complex regulates dimerization of ErbB2 via RhoA. Oncogene. 2014;33:2779–89.PubMedCrossRef
28.
go back to reference Deng X, Li Q, Hoff J, Novak M, Yang H, Jin H, et al. Integrin-associated CD151 drives ErbB2-evoked mammary tumor onset and metastasis. Neoplasia. 2012;14:678–89.PubMedPubMedCentralCrossRef Deng X, Li Q, Hoff J, Novak M, Yang H, Jin H, et al. Integrin-associated CD151 drives ErbB2-evoked mammary tumor onset and metastasis. Neoplasia. 2012;14:678–89.PubMedPubMedCentralCrossRef
29.
go back to reference Li Q, Yang XH, Xu F, Sharma C, Wang HX, Knoblich K, et al. Tetraspanin CD151 plays a key role in skin squamous cell carcinoma. Oncogene. 2013;32:1772–83.PubMedCrossRef Li Q, Yang XH, Xu F, Sharma C, Wang HX, Knoblich K, et al. Tetraspanin CD151 plays a key role in skin squamous cell carcinoma. Oncogene. 2013;32:1772–83.PubMedCrossRef
30.
31.
go back to reference Toya SP, Wary KK, Mittal M, Li F, Toth PT, Park C, et al. Integrin α6β1 Expressed in ESCs Instructs the Differentiation to Endothelial Cells. Stem Cells. 2015;33:1719–29.PubMedPubMedCentralCrossRef Toya SP, Wary KK, Mittal M, Li F, Toth PT, Park C, et al. Integrin α6β1 Expressed in ESCs Instructs the Differentiation to Endothelial Cells. Stem Cells. 2015;33:1719–29.PubMedPubMedCentralCrossRef
32.
go back to reference Akiel MA, Santhekadur PK, Mendoza RG, Siddiq A, Fisher PB, Sarkar D. Tetraspanin 8 mediates AEG-1-induced invasion and metastasis in hepatocellular carcinoma cells. FEBS Lett. 2016;590:2700–8.PubMedPubMedCentralCrossRef Akiel MA, Santhekadur PK, Mendoza RG, Siddiq A, Fisher PB, Sarkar D. Tetraspanin 8 mediates AEG-1-induced invasion and metastasis in hepatocellular carcinoma cells. FEBS Lett. 2016;590:2700–8.PubMedPubMedCentralCrossRef
33.
go back to reference Fang T, Lin J, Wang Y, Chen G, Huang J, Chen J, et al. Tetraspanin-8 promotes hepatocellular carcinoma metastasis by increasing ADAM12m expression. Oncotarget. 2016;7:40630–43.PubMedPubMedCentral Fang T, Lin J, Wang Y, Chen G, Huang J, Chen J, et al. Tetraspanin-8 promotes hepatocellular carcinoma metastasis by increasing ADAM12m expression. Oncotarget. 2016;7:40630–43.PubMedPubMedCentral
34.
go back to reference Park CS, Kim TK, Kim HG, Kim YJ, Jeoung MH, Lee WR, et al. Therapeutic targeting of tetraspanin8 in epithelial ovarian cancer invasion and metastasis. Oncogene. 2016;35:4540–8.PubMedCrossRef Park CS, Kim TK, Kim HG, Kim YJ, Jeoung MH, Lee WR, et al. Therapeutic targeting of tetraspanin8 in epithelial ovarian cancer invasion and metastasis. Oncogene. 2016;35:4540–8.PubMedCrossRef
35.
go back to reference Pan SJ, Wu YB, Cai S, Pan YX, Liu W, Bian LG, et al. Over-expression of tetraspanin 8 in malignant glioma regulates tumor cell progression. Biochem Biophys Res Commun. 2015;458:476–82.PubMedCrossRef Pan SJ, Wu YB, Cai S, Pan YX, Liu W, Bian LG, et al. Over-expression of tetraspanin 8 in malignant glioma regulates tumor cell progression. Biochem Biophys Res Commun. 2015;458:476–82.PubMedCrossRef
36.
go back to reference Pan SJ, Zhan SK, Pan YX, Liu W, Bian LG, Sun B, et al. Tetraspanin 8-rictor-integrin α3 complex is required for glioma cell migration. Int J Mol Sci. 2015;16:5363–74.PubMedPubMedCentralCrossRef Pan SJ, Zhan SK, Pan YX, Liu W, Bian LG, Sun B, et al. Tetraspanin 8-rictor-integrin α3 complex is required for glioma cell migration. Int J Mol Sci. 2015;16:5363–74.PubMedPubMedCentralCrossRef
37.
go back to reference Ailane N, Greco C, Zhu Y, Sala-Valdés M, Billard M, Casal I, et al. Effect of an anti-human Co-029/tspan8 mouse monoclonal antibody on tumor growth in a nude mouse model. Front Physiol. 2014;5:364.PubMedPubMedCentralCrossRef Ailane N, Greco C, Zhu Y, Sala-Valdés M, Billard M, Casal I, et al. Effect of an anti-human Co-029/tspan8 mouse monoclonal antibody on tumor growth in a nude mouse model. Front Physiol. 2014;5:364.PubMedPubMedCentralCrossRef
38.
go back to reference Claas C, Seiter S, Claas A, Savelyeva L, Schwab M, Zöller M. Association between the rat homologue of CO-029, a metastasis-associated tetraspanin molecule and consumption coagulopathy. J Cell Biol. 1998;141:267–80.PubMedPubMedCentralCrossRef Claas C, Seiter S, Claas A, Savelyeva L, Schwab M, Zöller M. Association between the rat homologue of CO-029, a metastasis-associated tetraspanin molecule and consumption coagulopathy. J Cell Biol. 1998;141:267–80.PubMedPubMedCentralCrossRef
39.
go back to reference Gesierich S, Berezovskiy I, Ryschich E, Zöller M. Systemic induction of the angiogenesis switch by the tetraspanin D6.1A/CO-029. Cancer Res. 2006;66:7083–94.PubMedCrossRef Gesierich S, Berezovskiy I, Ryschich E, Zöller M. Systemic induction of the angiogenesis switch by the tetraspanin D6.1A/CO-029. Cancer Res. 2006;66:7083–94.PubMedCrossRef
40.
go back to reference Yue S, Mu W, Erb U, Zöller M. The tetraspanins CD151 and Tspan8 are essential exosome components for the crosstalk between cancer initiating cells and their surrounding. Oncotarget. 2015;6:2366–84.PubMed Yue S, Mu W, Erb U, Zöller M. The tetraspanins CD151 and Tspan8 are essential exosome components for the crosstalk between cancer initiating cells and their surrounding. Oncotarget. 2015;6:2366–84.PubMed
41.
go back to reference Zhao K, Erb U, Hackert T, Zöller M, Yue S. Distorted leukocyte migration, angiogenesis, wound repair and metastasis in Tspan8 and Tspan8/CD151 double knockout mice indicate complementary activities of Tspan8 and CD51. Biochim Biophys Acta. 2018;1865:379–91.CrossRef Zhao K, Erb U, Hackert T, Zöller M, Yue S. Distorted leukocyte migration, angiogenesis, wound repair and metastasis in Tspan8 and Tspan8/CD151 double knockout mice indicate complementary activities of Tspan8 and CD51. Biochim Biophys Acta. 2018;1865:379–91.CrossRef
42.
go back to reference Wright MD, Geary SM, Fitter S, Moseley GW, Lau LM, Sheng KC, et al. Characterization of mice lacking the tetraspanin superfamily member CD151. Mol Cell Biol. 2004;24:5978–88.PubMedPubMedCentralCrossRef Wright MD, Geary SM, Fitter S, Moseley GW, Lau LM, Sheng KC, et al. Characterization of mice lacking the tetraspanin superfamily member CD151. Mol Cell Biol. 2004;24:5978–88.PubMedPubMedCentralCrossRef
44.
go back to reference Berditchevski F, Odintsova E. ErbB receptors and tetraspanins: Casting the net wider. Int J Biochem Cell Biol. 2016;77(Pt A):68–71.PubMedCrossRef Berditchevski F, Odintsova E. ErbB receptors and tetraspanins: Casting the net wider. Int J Biochem Cell Biol. 2016;77(Pt A):68–71.PubMedCrossRef
45.
go back to reference Detchokul S, Newell B, Williams ED, Frauman AG. CD151 is associated with prostate cancer cell invasion and lymphangiogenesis in vivo. Oncol Rep. 2014;31:241–7.PubMedCrossRef Detchokul S, Newell B, Williams ED, Frauman AG. CD151 is associated with prostate cancer cell invasion and lymphangiogenesis in vivo. Oncol Rep. 2014;31:241–7.PubMedCrossRef
46.
47.
go back to reference Iwasaki T, Takeda Y, Maruyama K, Yokosaki Y, Tsujino K, Tetsumoto S, et al. Deletion of tetraspanin CD9 diminishes lymphangiogenesis in vivo and in vitro. J Biol Chem. 2013;288:2118–31.PubMedCrossRef Iwasaki T, Takeda Y, Maruyama K, Yokosaki Y, Tsujino K, Tetsumoto S, et al. Deletion of tetraspanin CD9 diminishes lymphangiogenesis in vivo and in vitro. J Biol Chem. 2013;288:2118–31.PubMedCrossRef
48.
go back to reference Oh HH, Park KJ, Kim N, Park SY, Park YL, Oak CY, et al. Impact of KITENIN on tumor angiogenesis and lymphangiogenesis in colorectal cancer. Oncol Rep. 2016;35:253–60.PubMedCrossRef Oh HH, Park KJ, Kim N, Park SY, Park YL, Oak CY, et al. Impact of KITENIN on tumor angiogenesis and lymphangiogenesis in colorectal cancer. Oncol Rep. 2016;35:253–60.PubMedCrossRef
50.
go back to reference Boyiadzis M, Whiteside TL. The emerging roles of tumor-derived exosomes in hematological malignancies. Leukemia. 2017;31:1259–68.PubMedCrossRef Boyiadzis M, Whiteside TL. The emerging roles of tumor-derived exosomes in hematological malignancies. Leukemia. 2017;31:1259–68.PubMedCrossRef
52.
go back to reference Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883–91.PubMedPubMedCentralCrossRef Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18:883–91.PubMedPubMedCentralCrossRef
53.
54.
go back to reference Colak S, Medema JP. Cancer stem cells--important players in tumor therapy resistance. FEBS J. 2014;281:4779–91.PubMedCrossRef Colak S, Medema JP. Cancer stem cells--important players in tumor therapy resistance. FEBS J. 2014;281:4779–91.PubMedCrossRef
55.
go back to reference Frączek N, Bronisz I, Pietryka M, Kępińska D, Strzała P, Mielnicka K, et al. An outline of main factors of drug resistance influencing cancer therapy. J Chemother. 2016;28:457–64.PubMedCrossRef Frączek N, Bronisz I, Pietryka M, Kępińska D, Strzała P, Mielnicka K, et al. An outline of main factors of drug resistance influencing cancer therapy. J Chemother. 2016;28:457–64.PubMedCrossRef
57.
go back to reference Herr MJ, Kotha J, Hagedorn N, Smith B, Jennings LK. Tetraspanin CD9 promotes the invasive phenotype of human fibrosarcoma cells via upregulation of matrix metalloproteinase-9. PLoS One. 2013;8:e67766.PubMedPubMedCentralCrossRef Herr MJ, Kotha J, Hagedorn N, Smith B, Jennings LK. Tetraspanin CD9 promotes the invasive phenotype of human fibrosarcoma cells via upregulation of matrix metalloproteinase-9. PLoS One. 2013;8:e67766.PubMedPubMedCentralCrossRef
58.
go back to reference Li PY, Lv J, Qi WW, Zhao SF, Sun LB, Liu N, et al. Tspan9 inhibits the proliferation, migration and invasion of human gastric cancer SGC7901 cells via the ERK1/2 pathway. Oncol Rep. 2016;36:448–54.PubMedCrossRef Li PY, Lv J, Qi WW, Zhao SF, Sun LB, Liu N, et al. Tspan9 inhibits the proliferation, migration and invasion of human gastric cancer SGC7901 cells via the ERK1/2 pathway. Oncol Rep. 2016;36:448–54.PubMedCrossRef
59.
go back to reference Ranjan A, Bane SM, Kalraiya RD. Glycosylation of the laminin receptor (α3β1) regulates its association with tetraspanin CD151: Impact on cell spreading, motility, degradation and invasion of basement membrane by tumor cells. Exp Cell Res. 2014;322:249–64.PubMedCrossRef Ranjan A, Bane SM, Kalraiya RD. Glycosylation of the laminin receptor (α3β1) regulates its association with tetraspanin CD151: Impact on cell spreading, motility, degradation and invasion of basement membrane by tumor cells. Exp Cell Res. 2014;322:249–64.PubMedCrossRef
60.
go back to reference Schröder HM, Hoffmann SC, Hecker M, Korff T, Ludwig T. The tetraspanin network modulates MT1-MMP cell surface trafficking. Int J Biochem Cell Biol. 2013;45:1133–44.PubMedCrossRef Schröder HM, Hoffmann SC, Hecker M, Korff T, Ludwig T. The tetraspanin network modulates MT1-MMP cell surface trafficking. Int J Biochem Cell Biol. 2013;45:1133–44.PubMedCrossRef
61.
go back to reference Gu H, Chen C, Hao X, Wang C, Zhang X, Li Z, et al. Sorting protein VPS33B regulates exosomal autocrine signaling to mediate hematopoiesis and leukemogenesis. J Clin Invest. 2016;126:4537–53.PubMedPubMedCentralCrossRef Gu H, Chen C, Hao X, Wang C, Zhang X, Li Z, et al. Sorting protein VPS33B regulates exosomal autocrine signaling to mediate hematopoiesis and leukemogenesis. J Clin Invest. 2016;126:4537–53.PubMedPubMedCentralCrossRef
62.
go back to reference Bohlius J, Tonia T, Nüesch E, Jüni P, Fey MF, Egger M, et al. Effects of erythropoiesis-stimulating agents on fatigue- and anaemia-related symptoms in cancer patients: systematic review and meta-analyses of published and unpublished data. Br J Cancer. 2014;111:33–45.PubMedPubMedCentralCrossRef Bohlius J, Tonia T, Nüesch E, Jüni P, Fey MF, Egger M, et al. Effects of erythropoiesis-stimulating agents on fatigue- and anaemia-related symptoms in cancer patients: systematic review and meta-analyses of published and unpublished data. Br J Cancer. 2014;111:33–45.PubMedPubMedCentralCrossRef
63.
go back to reference Liang R, Ghaffari S. Mitochondria and FOXO3 in stem cell homeostasis, a window into hematopoietic stem cell fate determination. J Bioenerg Biomembr. 2017;49:343–6.PubMedCrossRefPubMedCentral Liang R, Ghaffari S. Mitochondria and FOXO3 in stem cell homeostasis, a window into hematopoietic stem cell fate determination. J Bioenerg Biomembr. 2017;49:343–6.PubMedCrossRefPubMedCentral
64.
go back to reference Yukawa H, Suzuki K, Aoki K, Arimoto T, Yasui T, Kaji N, et al. Imaging of angiogenesis of human umbilical vein endothelial cells by uptake of exosomes secreted from hepatocellular carcinoma cells. Sci Rep. 2018;8:6765.PubMedPubMedCentralCrossRef Yukawa H, Suzuki K, Aoki K, Arimoto T, Yasui T, Kaji N, et al. Imaging of angiogenesis of human umbilical vein endothelial cells by uptake of exosomes secreted from hepatocellular carcinoma cells. Sci Rep. 2018;8:6765.PubMedPubMedCentralCrossRef
65.
go back to reference Greening DW, Gopal SK, Mathias RA, Liu L, Sheng J, Zhu HJ, et al. Emerging roles of exosomes during epithelial-mesenchymal transition and cancer progression. Semin Cell Dev Biol. 2015;40:60–71.PubMedCrossRef Greening DW, Gopal SK, Mathias RA, Liu L, Sheng J, Zhu HJ, et al. Emerging roles of exosomes during epithelial-mesenchymal transition and cancer progression. Semin Cell Dev Biol. 2015;40:60–71.PubMedCrossRef
66.
go back to reference Maecker HT, Todd SC, Levy S. The tetraspanin superfamily: molecular facilitators. FASEB J. 1997;11:428–42.PubMedCrossRef Maecker HT, Todd SC, Levy S. The tetraspanin superfamily: molecular facilitators. FASEB J. 1997;11:428–42.PubMedCrossRef
67.
go back to reference Charrin S, le Naour F, Silvie O, Milhiet PE, Boucheix C, Rubinstein E. Lateral organization of membrane proteins: tetraspanins spin their web. Biochem J. 2009;420:133–54.PubMedCrossRef Charrin S, le Naour F, Silvie O, Milhiet PE, Boucheix C, Rubinstein E. Lateral organization of membrane proteins: tetraspanins spin their web. Biochem J. 2009;420:133–54.PubMedCrossRef
68.
go back to reference Bailey RL, Herbert JM, Khan K, Heath VL, Bicknell R, Tomlinson MG. The emerging role of tetraspanin microdomains on endothelial cells. Biochem Soc Trans. 2011;39:1667–73.PubMedCrossRef Bailey RL, Herbert JM, Khan K, Heath VL, Bicknell R, Tomlinson MG. The emerging role of tetraspanin microdomains on endothelial cells. Biochem Soc Trans. 2011;39:1667–73.PubMedCrossRef
69.
go back to reference Wang HX, Li Q, Sharma C, Knoblich K, Hemler ME. Tetraspanin protein contributions to cancer. Biochem Soc Trans. 2011;39:547–52.PubMedCrossRef Wang HX, Li Q, Sharma C, Knoblich K, Hemler ME. Tetraspanin protein contributions to cancer. Biochem Soc Trans. 2011;39:547–52.PubMedCrossRef
70.
go back to reference Nazarenko I, Rana S, Baumann A, McAlear J, Hellwig A, Trendelenburg M, et al. Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer Res. 2010;70:1668–78.PubMedCrossRef Nazarenko I, Rana S, Baumann A, McAlear J, Hellwig A, Trendelenburg M, et al. Cell surface tetraspanin Tspan8 contributes to molecular pathways of exosome-induced endothelial cell activation. Cancer Res. 2010;70:1668–78.PubMedCrossRef
71.
go back to reference Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci U S A. 2016;113:E968–77.PubMedPubMedCentralCrossRef Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, et al. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci U S A. 2016;113:E968–77.PubMedPubMedCentralCrossRef
72.
go back to reference Rana S, Claas C, Kretz CC, Nazarenko I, Zoeller M. Activation-induced internalization differs for the tetraspanins CD9 and Tspan8: Impact on tumor cell motility. Int J Biochem Cell Biol. 2011;43:106–19.PubMedCrossRef Rana S, Claas C, Kretz CC, Nazarenko I, Zoeller M. Activation-induced internalization differs for the tetraspanins CD9 and Tspan8: Impact on tumor cell motility. Int J Biochem Cell Biol. 2011;43:106–19.PubMedCrossRef
73.
go back to reference Sachs N, Secades P, van Hulst L, Song JY, Sonnenberg A. Reduced susceptibility to two-stage skin carcinogenesis in mice with epidermis-specific deletion of CD151. J Invest Dermatol. 2014;134:221–8.PubMedCrossRef Sachs N, Secades P, van Hulst L, Song JY, Sonnenberg A. Reduced susceptibility to two-stage skin carcinogenesis in mice with epidermis-specific deletion of CD151. J Invest Dermatol. 2014;134:221–8.PubMedCrossRef
74.
go back to reference Klosek SK, Nakashiro K, Hara S, Shintani S, Hasegawa H, Hamakawa H. CD151 forms a functional complex with c-Met in human salivary gland cancer cells. Biochem Biophys Res Commun. 2005;336:408–16.PubMedCrossRef Klosek SK, Nakashiro K, Hara S, Shintani S, Hasegawa H, Hamakawa H. CD151 forms a functional complex with c-Met in human salivary gland cancer cells. Biochem Biophys Res Commun. 2005;336:408–16.PubMedCrossRef
75.
go back to reference Kariya Y, Kariya Y, Gu J. Roles of laminin-332 and alpha6beta4 integrin in tumor progression. Mini Rev Med Chem. 2009;9:1284–91.PubMedCrossRef Kariya Y, Kariya Y, Gu J. Roles of laminin-332 and alpha6beta4 integrin in tumor progression. Mini Rev Med Chem. 2009;9:1284–91.PubMedCrossRef
76.
go back to reference Novitskaya V, Romanska H, Dawoud M, Jones JL, Berditchevski F. Tetraspanin CD151 regulates growth of mammary epithelial cells in three-dimensional extracellular matrix: implication for mammary ductal carcinoma in situ. Cancer Res. 2010;70:4698–708.PubMedPubMedCentralCrossRef Novitskaya V, Romanska H, Dawoud M, Jones JL, Berditchevski F. Tetraspanin CD151 regulates growth of mammary epithelial cells in three-dimensional extracellular matrix: implication for mammary ductal carcinoma in situ. Cancer Res. 2010;70:4698–708.PubMedPubMedCentralCrossRef
77.
go back to reference Romanska HM, Berditchevski F. Tetraspanins in human epithelial malignancies. J Pathol. 2011;223:4–14.PubMedCrossRef Romanska HM, Berditchevski F. Tetraspanins in human epithelial malignancies. J Pathol. 2011;223:4–14.PubMedCrossRef
78.
go back to reference Ramovs V, Te Molder L, Sonnenberg A. The opposing roles of laminin-binding integrins in cancer. Matrix Biol. 2017;57-58:213–43.PubMedCrossRef Ramovs V, Te Molder L, Sonnenberg A. The opposing roles of laminin-binding integrins in cancer. Matrix Biol. 2017;57-58:213–43.PubMedCrossRef
79.
go back to reference Paoli P, Giannoni E, Chiarugi P. Anoikis molecular pathways and its role in cancer progression. Biochim Biophys Acta. 2013;1833:3481–98.PubMedCrossRef Paoli P, Giannoni E, Chiarugi P. Anoikis molecular pathways and its role in cancer progression. Biochim Biophys Acta. 2013;1833:3481–98.PubMedCrossRef
80.
go back to reference Benvenuti S, Comoglio PM. The MET receptor tyrosine kinase in invasion and metastasis. J Cell Physiol. 2007;213:316–25.PubMedCrossRef Benvenuti S, Comoglio PM. The MET receptor tyrosine kinase in invasion and metastasis. J Cell Physiol. 2007;213:316–25.PubMedCrossRef
81.
84.
go back to reference Gawlik-Rzemieniewska N, Bednarek I. The role of NANOG transcriptional factor in the development of malignant phenotype of cancer cells. Cancer Biol Ther. 2016;17:1–10.PubMedCrossRef Gawlik-Rzemieniewska N, Bednarek I. The role of NANOG transcriptional factor in the development of malignant phenotype of cancer cells. Cancer Biol Ther. 2016;17:1–10.PubMedCrossRef
86.
go back to reference Wang Z, von Au A, Schnölzer M, Hackert T, Zöller M. CD44v6-competent tumor exosomes promote motility, invasion and cancer-initiating cell marker expression in pancreatic and colorectal cancer cells. Oncotarget. 2016;7:55409–36.PubMedPubMedCentral Wang Z, von Au A, Schnölzer M, Hackert T, Zöller M. CD44v6-competent tumor exosomes promote motility, invasion and cancer-initiating cell marker expression in pancreatic and colorectal cancer cells. Oncotarget. 2016;7:55409–36.PubMedPubMedCentral
87.
88.
go back to reference Longo N, Yáñez-Mó M, Mittelbrunn M, de la Rosa G, Muñoz ML, Sánchez-Madrid F, et al. Regulatory role of tetraspanin CD9 in tumor-endothelial cell interaction during transendothelial invasion of melanoma cells. Blood. 2001;98:3717–26.PubMedCrossRef Longo N, Yáñez-Mó M, Mittelbrunn M, de la Rosa G, Muñoz ML, Sánchez-Madrid F, et al. Regulatory role of tetraspanin CD9 in tumor-endothelial cell interaction during transendothelial invasion of melanoma cells. Blood. 2001;98:3717–26.PubMedCrossRef
89.
go back to reference Lin Q, Peng S, Yang Y. Inhibition of CD9 expression reduces the metastatic capacity of human hepatocellular carcinoma cell line MHCC97-H. Int J Oncol. 2018;53:266–74.PubMed Lin Q, Peng S, Yang Y. Inhibition of CD9 expression reduces the metastatic capacity of human hepatocellular carcinoma cell line MHCC97-H. Int J Oncol. 2018;53:266–74.PubMed
90.
go back to reference Gilsanz A, Sánchez-Martín L, Gutiérrez-López MD, Ovalle S, Machado-Pineda Y, Reyes R, et al. ALCAM/CD166 adhesive function is regulated by the tetraspanin CD9. Cell Mol Life Sci. 2013;70:475–93.PubMedCrossRef Gilsanz A, Sánchez-Martín L, Gutiérrez-López MD, Ovalle S, Machado-Pineda Y, Reyes R, et al. ALCAM/CD166 adhesive function is regulated by the tetraspanin CD9. Cell Mol Life Sci. 2013;70:475–93.PubMedCrossRef
91.
go back to reference Gutiérrez-López MD, Gilsanz A, Yáñez-Mó M, Ovalle S, Lafuente EM, Domínguez C, et al. The sheddase activity of ADAM17/TACE is regulated by the tetraspanin CD9. Cell Mol Life Sci. 2011;68:3275–92.PubMedCrossRef Gutiérrez-López MD, Gilsanz A, Yáñez-Mó M, Ovalle S, Lafuente EM, Domínguez C, et al. The sheddase activity of ADAM17/TACE is regulated by the tetraspanin CD9. Cell Mol Life Sci. 2011;68:3275–92.PubMedCrossRef
92.
go back to reference Cook GA, Longhurst CM, Grgurevich S, Cholera S, Crossno JT Jr, Jennings LK. Identification of CD9 extracellular domains important in regulation of CHO cell adhesion to fibronectin and fibronectin pericellular matrix assembly. Blood. 2002;100:4502–11.PubMedCrossRef Cook GA, Longhurst CM, Grgurevich S, Cholera S, Crossno JT Jr, Jennings LK. Identification of CD9 extracellular domains important in regulation of CHO cell adhesion to fibronectin and fibronectin pericellular matrix assembly. Blood. 2002;100:4502–11.PubMedCrossRef
94.
go back to reference Tang QH, Liu ZY, Zuo HJ, Liu ZX. Involvement of activation of C-met signaling pathway in CD151-induced HUVECs angiogenesis. J Huazhong Univ Sci Technolog Med Sci. 2015;35:35–41.PubMedCrossRef Tang QH, Liu ZY, Zuo HJ, Liu ZX. Involvement of activation of C-met signaling pathway in CD151-induced HUVECs angiogenesis. J Huazhong Univ Sci Technolog Med Sci. 2015;35:35–41.PubMedCrossRef
95.
go back to reference Cortese K, Sahores M, Madsen CD, Tacchetti C, Blasi F. Clathrin and LRP-1-independent constitutive endocytosis and recycling of uPAR. PLoS One. 2008;3:e3730.PubMedPubMedCentralCrossRef Cortese K, Sahores M, Madsen CD, Tacchetti C, Blasi F. Clathrin and LRP-1-independent constitutive endocytosis and recycling of uPAR. PLoS One. 2008;3:e3730.PubMedPubMedCentralCrossRef
96.
go back to reference Herkenne S, Paques C, Nivelles O, Lion M, Bajou K, Pollenus T, et al. The interaction of uPAR with VEGFR2 promotes VEGF-induced angiogenesis. Sci Signal. 2015;8:ra117.PubMedCrossRef Herkenne S, Paques C, Nivelles O, Lion M, Bajou K, Pollenus T, et al. The interaction of uPAR with VEGFR2 promotes VEGF-induced angiogenesis. Sci Signal. 2015;8:ra117.PubMedCrossRef
97.
go back to reference Rao Malla R, Gopinath S, Alapati K, Gorantla B, Gondi CS, Rao JS. Knockdown of cathepsin B and uPAR inhibits CD151 and α3β1 integrin-mediated cell adhesion and invasion in glioma. Mol Carcinog. 2013;52:777–90.PubMed Rao Malla R, Gopinath S, Alapati K, Gorantla B, Gondi CS, Rao JS. Knockdown of cathepsin B and uPAR inhibits CD151 and α3β1 integrin-mediated cell adhesion and invasion in glioma. Mol Carcinog. 2013;52:777–90.PubMed
98.
go back to reference Heiler S, Mu W, Zöller M, Thuma F. The importance of claudin-7 palmitoylation on membrane subdomain localization and metastasis-promoting activities. Cell Commun Signal. 2015;13:29.PubMedPubMedCentralCrossRef Heiler S, Mu W, Zöller M, Thuma F. The importance of claudin-7 palmitoylation on membrane subdomain localization and metastasis-promoting activities. Cell Commun Signal. 2015;13:29.PubMedPubMedCentralCrossRef
100.
101.
go back to reference Turunen SP, Tatti-Bugaeva O, Lehti K. Membrane-type matrix metalloproteases as diverse effectors of cancer progression. Biochim Biophys Acta. 2017;1864(11 Pt A):1974–88.CrossRef Turunen SP, Tatti-Bugaeva O, Lehti K. Membrane-type matrix metalloproteases as diverse effectors of cancer progression. Biochim Biophys Acta. 2017;1864(11 Pt A):1974–88.CrossRef
102.
go back to reference Zhou P, Erfani S, Liu Z, Jia C, Chen Y, Xu B, et al. CD151-α3β1 integrin complexes are prognostic markers of glioblastoma and cooperate with EGFR to drive tumor cell motility and invasion. Oncotarget. 2015;6:29675–93.PubMedPubMedCentral Zhou P, Erfani S, Liu Z, Jia C, Chen Y, Xu B, et al. CD151-α3β1 integrin complexes are prognostic markers of glioblastoma and cooperate with EGFR to drive tumor cell motility and invasion. Oncotarget. 2015;6:29675–93.PubMedPubMedCentral
103.
go back to reference Sceneay J, Smyth MJ, Möller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32:449–64.PubMedCrossRef Sceneay J, Smyth MJ, Möller A. The pre-metastatic niche: finding common ground. Cancer Metastasis Rev. 2013;32:449–64.PubMedCrossRef
104.
go back to reference Kosaka N, Yoshioka Y, Tominaga N, Hagiwara K, Katsuda T, Ochiya T. Dark side of the exosome: the role of the exosome in cancer metastasis and targeting the exosome as a strategy for cancer therapy. Future Oncol. 2014;10:671–81.PubMedCrossRef Kosaka N, Yoshioka Y, Tominaga N, Hagiwara K, Katsuda T, Ochiya T. Dark side of the exosome: the role of the exosome in cancer metastasis and targeting the exosome as a strategy for cancer therapy. Future Oncol. 2014;10:671–81.PubMedCrossRef
105.
go back to reference Wang J, De Veirman K, De Beule N, Maes K, De Bruyne E, Van Valckenborgh E, et al. The bone marrow microenvironment enhances multiple myeloma progression by exosome-mediated activation of myeloid-derived suppressor cells. Oncotarget. 2015;6:43992–4004.PubMedPubMedCentral Wang J, De Veirman K, De Beule N, Maes K, De Bruyne E, Van Valckenborgh E, et al. The bone marrow microenvironment enhances multiple myeloma progression by exosome-mediated activation of myeloid-derived suppressor cells. Oncotarget. 2015;6:43992–4004.PubMedPubMedCentral
106.
go back to reference Ng YH, Rome S, Jalabert A, Forterre A, Singh H, Hincks CL, et al. Endometrial exosomes/microvesicles in the uterine microenvironment: a new paradigm for embryo-endometrial cross talk at implantation. PLoS One. 2013;8:e58502.PubMedPubMedCentralCrossRef Ng YH, Rome S, Jalabert A, Forterre A, Singh H, Hincks CL, et al. Endometrial exosomes/microvesicles in the uterine microenvironment: a new paradigm for embryo-endometrial cross talk at implantation. PLoS One. 2013;8:e58502.PubMedPubMedCentralCrossRef
107.
go back to reference Desterke C, Martinaud C, Guerton B, Pieri L, Bogani C, Clay D, et al. Tetraspanin CD9 participates in dysmegakaryopoiesis and stromal interactions in primary myelofibrosis. Haematologica. 2015;100:757–67.PubMedPubMedCentralCrossRef Desterke C, Martinaud C, Guerton B, Pieri L, Bogani C, Clay D, et al. Tetraspanin CD9 participates in dysmegakaryopoiesis and stromal interactions in primary myelofibrosis. Haematologica. 2015;100:757–67.PubMedPubMedCentralCrossRef
108.
go back to reference Clay D, Rubinstein E, Mishal Z, Anjo A, Prenant M, Jasmin C, et al. CD9 and megakaryocyte differentiation. Blood. 2001;97:1982–9.PubMedCrossRef Clay D, Rubinstein E, Mishal Z, Anjo A, Prenant M, Jasmin C, et al. CD9 and megakaryocyte differentiation. Blood. 2001;97:1982–9.PubMedCrossRef
109.
go back to reference Oritani K, Wu X, Medina K, Hudson J, Miyake K, Gimble JM, et al. Antibody ligation of CD9 modifies production of myeloid cells in long-term cultures. Blood. 1996;87:2252–61.PubMed Oritani K, Wu X, Medina K, Hudson J, Miyake K, Gimble JM, et al. Antibody ligation of CD9 modifies production of myeloid cells in long-term cultures. Blood. 1996;87:2252–61.PubMed
110.
go back to reference Spring FA, Griffiths RE, Mankelow TJ, Agnew C, Parsons SF, Chasis JA, et al. Tetraspanins CD81 and CD82 facilitate α4β1-mediated adhesion of human erythroblasts to vascular cell adhesion molecule-1. PLoS One. 2013;8:e62654.PubMedPubMedCentralCrossRef Spring FA, Griffiths RE, Mankelow TJ, Agnew C, Parsons SF, Chasis JA, et al. Tetraspanins CD81 and CD82 facilitate α4β1-mediated adhesion of human erythroblasts to vascular cell adhesion molecule-1. PLoS One. 2013;8:e62654.PubMedPubMedCentralCrossRef
111.
go back to reference Han P, Guo X, Story C. Role of beta(1)-integrins and their associated tetraspanin molecules in fibronectin-enhanced megakaryopoiesis. Cytotherapy. 2004;6:465–75.PubMedCrossRef Han P, Guo X, Story C. Role of beta(1)-integrins and their associated tetraspanin molecules in fibronectin-enhanced megakaryopoiesis. Cytotherapy. 2004;6:465–75.PubMedCrossRef
113.
go back to reference Azab AK, Quang P, Azab F, Pitsillides C, Thompson B, Chonghaile T, et al. P-selectin glycoprotein ligand regulates the interaction of multiple myeloma cells with the bone marrow microenvironment. Blood. 2012;119:1468–78.PubMedPubMedCentralCrossRef Azab AK, Quang P, Azab F, Pitsillides C, Thompson B, Chonghaile T, et al. P-selectin glycoprotein ligand regulates the interaction of multiple myeloma cells with the bone marrow microenvironment. Blood. 2012;119:1468–78.PubMedPubMedCentralCrossRef
114.
go back to reference Krause DS, Lazarides K, Lewis JB, von Andrian UH, Van Etten RA. Selectins and their ligands are required for homing and engraftment of BCR-ABL1+ leukemic stem cells in the bone marrow niche. Blood. 2014;123:1361–71.PubMedPubMedCentralCrossRef Krause DS, Lazarides K, Lewis JB, von Andrian UH, Van Etten RA. Selectins and their ligands are required for homing and engraftment of BCR-ABL1+ leukemic stem cells in the bone marrow niche. Blood. 2014;123:1361–71.PubMedPubMedCentralCrossRef
115.
go back to reference Gros SJ, Kurschat N, Drenckhan A, Dohrmann T, Forberich E, Effenberger K, et al. Involvement of CXCR4 chemokine receptor in metastastic HER2-positive esophageal cancer. PLoS One. 2012;7:e47287.PubMedPubMedCentralCrossRef Gros SJ, Kurschat N, Drenckhan A, Dohrmann T, Forberich E, Effenberger K, et al. Involvement of CXCR4 chemokine receptor in metastastic HER2-positive esophageal cancer. PLoS One. 2012;7:e47287.PubMedPubMedCentralCrossRef
116.
go back to reference Mühlethaler-Mottet A, Liberman J, Ascenção K, Flahaut M, Balmas-Bourloud K, Yan P, et al. The CXCR4/CXCR7/CXCL12 Axis Is Involved in a Secondary but Complex Control of Neuroblastoma Metastatic Cell Homing. PLoS One. 2015;10:e0125616.PubMedPubMedCentralCrossRef Mühlethaler-Mottet A, Liberman J, Ascenção K, Flahaut M, Balmas-Bourloud K, Yan P, et al. The CXCR4/CXCR7/CXCL12 Axis Is Involved in a Secondary but Complex Control of Neuroblastoma Metastatic Cell Homing. PLoS One. 2015;10:e0125616.PubMedPubMedCentralCrossRef
117.
go back to reference Arnaud MP, Vallée A, Robert G, Bonneau J, Leroy C, Varin-Blank N, et al. CD9, a key actor in the dissemination of lymphoblastic leukemia, modulating CXCR4-mediated migration via RAC1 signaling. Blood. 2015;126:1802–12.PubMedCrossRef Arnaud MP, Vallée A, Robert G, Bonneau J, Leroy C, Varin-Blank N, et al. CD9, a key actor in the dissemination of lymphoblastic leukemia, modulating CXCR4-mediated migration via RAC1 signaling. Blood. 2015;126:1802–12.PubMedCrossRef
118.
go back to reference Yoshida T, Kawano Y, Sato K, Ando Y, Aoki J, Miura Y, et al. A CD63 mutant inhibits T-cell tropic human immunodeficiency virus type 1 entry by disrupting CXCR4 trafficking to the plasma membrane. Traffic. 2008;9:540–58.PubMedCrossRef Yoshida T, Kawano Y, Sato K, Ando Y, Aoki J, Miura Y, et al. A CD63 mutant inhibits T-cell tropic human immunodeficiency virus type 1 entry by disrupting CXCR4 trafficking to the plasma membrane. Traffic. 2008;9:540–58.PubMedCrossRef
119.
go back to reference Head BP, Patel HH, Insel PA. Interaction of membrane/lipid rafts with the cytoskeleton: impact on signaling and function: membrane/lipid rafts, mediators of cytoskeletal arrangement and cell signaling. Biochim Biophys Acta. 2013;1838:532–45.PubMedCrossRef Head BP, Patel HH, Insel PA. Interaction of membrane/lipid rafts with the cytoskeleton: impact on signaling and function: membrane/lipid rafts, mediators of cytoskeletal arrangement and cell signaling. Biochim Biophys Acta. 2013;1838:532–45.PubMedCrossRef
121.
go back to reference Verweij FJ, Bebelman MP, Jimenez CR, Garcia-Vallejo JJ, Janssen H, Neefjes J, et al. Quantifying exosome secretion from single cells reveals a modulatory role for GPCR signaling. J Cell Biol. 2018;217:1129–42.PubMedPubMedCentralCrossRef Verweij FJ, Bebelman MP, Jimenez CR, Garcia-Vallejo JJ, Janssen H, Neefjes J, et al. Quantifying exosome secretion from single cells reveals a modulatory role for GPCR signaling. J Cell Biol. 2018;217:1129–42.PubMedPubMedCentralCrossRef
122.
go back to reference Ramchandani D, Weber GF. Interactions between osteopontin and vascular endothelial growth factor: Implications for cancer. Biochim Biophys Acta. 2015;1855:202–22.PubMed Ramchandani D, Weber GF. Interactions between osteopontin and vascular endothelial growth factor: Implications for cancer. Biochim Biophys Acta. 2015;1855:202–22.PubMed
123.
go back to reference Shibuya M. Vascular endothelial growth factor and its receptor system: physiological functions in angiogenesis and pathological roles in various diseases. J Biochem. 2013;153:13–9.PubMedCrossRef Shibuya M. Vascular endothelial growth factor and its receptor system: physiological functions in angiogenesis and pathological roles in various diseases. J Biochem. 2013;153:13–9.PubMedCrossRef
124.
go back to reference Cheng N, Brantley DM, Chen J. The ephrins and Eph receptors in angiogenesis. Cytokine Growth Factor Rev. 2002;13:75–85.PubMedCrossRef Cheng N, Brantley DM, Chen J. The ephrins and Eph receptors in angiogenesis. Cytokine Growth Factor Rev. 2002;13:75–85.PubMedCrossRef
125.
go back to reference Barreiro O, Zamai M, Yáñez-Mó M, Tejera E, López-Romero P, Monk PN, et al. Endothelial adhesion receptors are recruited to adherent leukocytes by inclusion in preformed tetraspanin nanoplatforms. J Cell Biol. 2008;183:527–42.PubMedPubMedCentralCrossRef Barreiro O, Zamai M, Yáñez-Mó M, Tejera E, López-Romero P, Monk PN, et al. Endothelial adhesion receptors are recruited to adherent leukocytes by inclusion in preformed tetraspanin nanoplatforms. J Cell Biol. 2008;183:527–42.PubMedPubMedCentralCrossRef
126.
go back to reference Wadkin JCR, Patten DA, Kamarajah SK, Shepherd EL, Novitskaya V, Berditchevski F, et al. CD151 supports VCAM-1-mediated lymphocyte adhesion to liver endothelium and is upregulated in chronic liver disease and hepatocellular carcinoma. Am J Physiol Gastrointest Liver Physiol. 2017;313:G138–49.PubMedPubMedCentralCrossRef Wadkin JCR, Patten DA, Kamarajah SK, Shepherd EL, Novitskaya V, Berditchevski F, et al. CD151 supports VCAM-1-mediated lymphocyte adhesion to liver endothelium and is upregulated in chronic liver disease and hepatocellular carcinoma. Am J Physiol Gastrointest Liver Physiol. 2017;313:G138–49.PubMedPubMedCentralCrossRef
127.
go back to reference Nomura S, Iwata S, Hatano R, Komiya E, Dang NH, Iwao N, et al. Inhibition of VEGF-dependent angiogenesis by the anti-CD82 monoclonal antibody 4F9 through regulation of lipid raft microdomains. Biochem Biophys Res Commun. 2016;474:111–7.PubMedCrossRef Nomura S, Iwata S, Hatano R, Komiya E, Dang NH, Iwao N, et al. Inhibition of VEGF-dependent angiogenesis by the anti-CD82 monoclonal antibody 4F9 through regulation of lipid raft microdomains. Biochem Biophys Res Commun. 2016;474:111–7.PubMedCrossRef
128.
go back to reference Tugues S, Honjo S, König C, Padhan N, Kroon J, Gualandi L, et al. Tetraspanin CD63 promotes vascular endothelial growth factor receptor 2-β1 integrin complex formation, thereby regulating activation and downstream signaling in endothelial cells in vitro and in vivo. J Biol Chem. 2013;288:19060–71.PubMedPubMedCentralCrossRef Tugues S, Honjo S, König C, Padhan N, Kroon J, Gualandi L, et al. Tetraspanin CD63 promotes vascular endothelial growth factor receptor 2-β1 integrin complex formation, thereby regulating activation and downstream signaling in endothelial cells in vitro and in vivo. J Biol Chem. 2013;288:19060–71.PubMedPubMedCentralCrossRef
129.
go back to reference Klein-Soyer C, Azorsa DO, Cazenave JP, Lanza F. CD9 participates in endothelial cell migration during in vitro wound repair. Arterioscler Thromb Vasc Biol. 2000;20:360–9.PubMedCrossRef Klein-Soyer C, Azorsa DO, Cazenave JP, Lanza F. CD9 participates in endothelial cell migration during in vitro wound repair. Arterioscler Thromb Vasc Biol. 2000;20:360–9.PubMedCrossRef
130.
go back to reference Colin S, Guilmain W, Creoff E, Schneider C, Steverlynck C, Bongaerts M, et al. A truncated form of CD9-partner 1 (CD9P-1), GS-168AT2, potently inhibits in vivo tumour-induced angiogenesis and tumour growth. Br J Cancer. 2011;105:1002–11.PubMedPubMedCentralCrossRef Colin S, Guilmain W, Creoff E, Schneider C, Steverlynck C, Bongaerts M, et al. A truncated form of CD9-partner 1 (CD9P-1), GS-168AT2, potently inhibits in vivo tumour-induced angiogenesis and tumour growth. Br J Cancer. 2011;105:1002–11.PubMedPubMedCentralCrossRef
131.
go back to reference Peng D, Zuo H, Liu Z, Qin J, Zhou Y, Li P, et al. The tetraspanin CD151-ARSA mutant inhibits angiogenesis via the YRSL sequence. Mol Med Rep. 2013;7:836–42.PubMedCrossRef Peng D, Zuo H, Liu Z, Qin J, Zhou Y, Li P, et al. The tetraspanin CD151-ARSA mutant inhibits angiogenesis via the YRSL sequence. Mol Med Rep. 2013;7:836–42.PubMedCrossRef
132.
go back to reference Peddibhotla SS, Brinkmann BF, Kummer D, Tuncay H, Nakayama M, Adams RH, et al. Tetraspanin CD9 links junctional adhesion molecule-A to αvβ3 integrin to mediate basic fibroblast growth factor-specific angiogenic signaling. Mol Biol Cell. 2013;24:933–44.PubMedPubMedCentralCrossRef Peddibhotla SS, Brinkmann BF, Kummer D, Tuncay H, Nakayama M, Adams RH, et al. Tetraspanin CD9 links junctional adhesion molecule-A to αvβ3 integrin to mediate basic fibroblast growth factor-specific angiogenic signaling. Mol Biol Cell. 2013;24:933–44.PubMedPubMedCentralCrossRef
133.
go back to reference Takeda Y, Kazarov AR, Butterfield CE, Hopkins BD, Benjamin LE, Kaipainen A, et al. Deletion of tetraspanin Cd151 results in decreased pathologic angiogenesis in vivo and in vitro. Blood. 2007;109:1524–32.PubMedPubMedCentralCrossRef Takeda Y, Kazarov AR, Butterfield CE, Hopkins BD, Benjamin LE, Kaipainen A, et al. Deletion of tetraspanin Cd151 results in decreased pathologic angiogenesis in vivo and in vitro. Blood. 2007;109:1524–32.PubMedPubMedCentralCrossRef
134.
go back to reference Wang J, Huang Y, Zhang J, Wei Y, Mahoud S, Bakheet AM, et al. Pathway-related molecules of VEGFC/D-VEGFR3/NRP2 axis in tumor lymphangiogenesis and lymphatic metastasis. Clin Chim Acta. 2016;461:165–71.PubMedCrossRef Wang J, Huang Y, Zhang J, Wei Y, Mahoud S, Bakheet AM, et al. Pathway-related molecules of VEGFC/D-VEGFR3/NRP2 axis in tumor lymphangiogenesis and lymphatic metastasis. Clin Chim Acta. 2016;461:165–71.PubMedCrossRef
135.
go back to reference Touat M, Ileana E, Postel-Vinay S, André F, Soria JC. Targeting FGFR Signaling in Cancer. Clin Cancer Res. 2015;21:2684–94.PubMedCrossRef Touat M, Ileana E, Postel-Vinay S, André F, Soria JC. Targeting FGFR Signaling in Cancer. Clin Cancer Res. 2015;21:2684–94.PubMedCrossRef
136.
go back to reference Chen PH, Bendris N, Hsiao YJ, Reis CR, Mettlen M, Chen HY, et al. Crosstalk between CLCb/Dyn1-Mediated Adaptive Clathrin-Mediated Endocytosis and Epidermal Growth Factor Receptor Signaling Increases Metastasis. Dev Cell. 2017;40:278–88.PubMedPubMedCentralCrossRef Chen PH, Bendris N, Hsiao YJ, Reis CR, Mettlen M, Chen HY, et al. Crosstalk between CLCb/Dyn1-Mediated Adaptive Clathrin-Mediated Endocytosis and Epidermal Growth Factor Receptor Signaling Increases Metastasis. Dev Cell. 2017;40:278–88.PubMedPubMedCentralCrossRef
137.
go back to reference Delos-Santos RC, Garay C, Antonescu CN. Charming neighborhoods on the cell surface: plasma membrane microdomains regulate receptor tyrosine kinase signaling. Cell Signal. 2015;27:1963–76.PubMedCrossRef Delos-Santos RC, Garay C, Antonescu CN. Charming neighborhoods on the cell surface: plasma membrane microdomains regulate receptor tyrosine kinase signaling. Cell Signal. 2015;27:1963–76.PubMedCrossRef
138.
go back to reference Pitulescu ME, Adams RH. Regulation of signaling interactions and receptor endocytosis in growing blood vessels. Cell Adhes Migr. 2014;8:366–77.CrossRef Pitulescu ME, Adams RH. Regulation of signaling interactions and receptor endocytosis in growing blood vessels. Cell Adhes Migr. 2014;8:366–77.CrossRef
140.
142.
go back to reference Thuma F, Zöller M. Outsmart tumor exosomes to steal the cancer initiating cell its niche. Semin Cancer Biol. 2014;28:39–50.PubMedCrossRef Thuma F, Zöller M. Outsmart tumor exosomes to steal the cancer initiating cell its niche. Semin Cancer Biol. 2014;28:39–50.PubMedCrossRef
Metadata
Title
Tspan8 and Tspan8/CD151 knockout mice unravel the contribution of tumor and host exosomes to tumor progression
Authors
Kun Zhao
Zhe Wang
Thilo Hackert
Claudia Pitzer
Margot Zöller
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0961-6

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 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