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

Open Access 01-12-2017 | Research

Bi-directional exosome-driven intercommunication between the hepatic niche and cancer cells

Authors: Nikolina Dioufa, Amanda M. Clark, Bo Ma, Colin H. Beckwitt, Alan Wells

Published in: Molecular Cancer | Issue 1/2017

Login to get access

Abstract

Background

Our understanding of the multiple roles exosomes play during tumor progression is still very poor and the contribution of the normal tissue derived exosomes in distant seeding and tumor outgrowth has also not been widely appreciated.

Methods

Using our all-human liver microphysiological system (MPS) platform as a model to closely recapitulate the early metastatic events, we isolated exosomes from both tumor cells and liver microenvironment.

Results

We observed that while priming of the hepatic niche (HepN) with MDA-231 breast cancer derived exosomes facilitated seeding of the cancer cells in the liver, subsequent tumor outgrowth was diminished; this was consistent with increased entry into dormancy. We found that hepatic niche (HepN) derived exosomes contribute significantly to the exosome pool and are distinguished from cancer derived exosomes based on their size, protein and miRNA content. By Ingenuity Pathway Analysis (IPA) of the miRNA content of the HepN, MDA-231/HepN and MDA-231 cells we showed that the HepN derived exosomes affect the breast cancer cells by suppressing pathways involved in cancer cell proliferation and invasion. More importantly exposure of MDA-231 and MDA-468 cells to purified normal HepN derived exosomes, induced changes in the cells consistent with a Mesenchymal to Epithelial reverting Transition (MErT). miRNA arrays performed on MDA-231 treated with Hum Hep/NPC derived exosomes showed significant changes in the levels of a select number of miRNAs involved in epithelial cell differentiation and miRNAs, such as miR186, miR23a and miR205, from our top and bottom bins have previously been reported to regulate E-cadherin transcription and MErT induction in various cancer types. Consistently HepN derived exosome treatment of breast and prostate cancer lines lead to a transient induction of E-cadherin and ZO-1 at the protein level and a more epithelial-like morphology of the cells.

Conclusions

Collectively our data revealed a novel mechanism of regulation of the metastatic cascade, showing a well-orchestrated, timely controlled crosstalk between the cancer cells and the HepN and implicating for the first time the normal tissue/HepN derived exosomes in enabling seeding and entry into dormancy of the cancer cells at the metastatic site.
Appendix
Available only for authorised users
Literature
1.
go back to reference Pickren J, Tsukada Y, Lane W. In: Weiss L, Gilbert L, editors. Liver metastasis. Boston, MA: G.K. Hall Medical Publishers; 1982. Pickren J, Tsukada Y, Lane W. In: Weiss L, Gilbert L, editors. Liver metastasis. Boston, MA: G.K. Hall Medical Publishers; 1982.
2.
go back to reference Yates C, Shepard CR, Papworth G, Dash A, Beer Stolz D, Tannenbaum S, Griffith L, Wells A. Novel three-dimensional organotypic liver bioreactor to directly visualize early events in metastatic progression. Adv Cancer Res. 2007;97:225–46.CrossRefPubMed Yates C, Shepard CR, Papworth G, Dash A, Beer Stolz D, Tannenbaum S, Griffith L, Wells A. Novel three-dimensional organotypic liver bioreactor to directly visualize early events in metastatic progression. Adv Cancer Res. 2007;97:225–46.CrossRefPubMed
3.
go back to reference Chao Y, Wu Q, Acquafondata M, Dhir R, Wells A. Partial mesenchymal to epithelial reverting transition in breast and prostate cancer metastases. Cancer Microenviron. 2012;5(1):19–28.CrossRefPubMed Chao Y, Wu Q, Acquafondata M, Dhir R, Wells A. Partial mesenchymal to epithelial reverting transition in breast and prostate cancer metastases. Cancer Microenviron. 2012;5(1):19–28.CrossRefPubMed
4.
go back to reference Gunasinghe NP, Wells A, Thompson EW, Hugo HJ. Mesenchymal-epithelial transition (MET) as a mechanism for metastatic colonisation in breast cancer. Cancer Metastasis Rev. 2012;31(3–4):469–78.CrossRefPubMed Gunasinghe NP, Wells A, Thompson EW, Hugo HJ. Mesenchymal-epithelial transition (MET) as a mechanism for metastatic colonisation in breast cancer. Cancer Metastasis Rev. 2012;31(3–4):469–78.CrossRefPubMed
5.
go back to reference Wells A, Yates C, Shepard CR. E-cadherin as an indicator of mesenchymal to epithelial reverting transitions during the metastatic seeding of disseminated carcinomas. Clin Exp Metastasis. 2008;25(6):621–8.CrossRefPubMedPubMedCentral Wells A, Yates C, Shepard CR. E-cadherin as an indicator of mesenchymal to epithelial reverting transitions during the metastatic seeding of disseminated carcinomas. Clin Exp Metastasis. 2008;25(6):621–8.CrossRefPubMedPubMedCentral
6.
go back to reference Bukholm IK, Nesland JM, Børresen-Dale AL. Re-expression of E-cadherin, alpha-catenin and beta-catenin, but not of gamma-catenin, in metastatic tissue from breast cancer patients. J Pathol. 2000;190(1):15–9.CrossRefPubMed Bukholm IK, Nesland JM, Børresen-Dale AL. Re-expression of E-cadherin, alpha-catenin and beta-catenin, but not of gamma-catenin, in metastatic tissue from breast cancer patients. J Pathol. 2000;190(1):15–9.CrossRefPubMed
7.
go back to reference Paul J. Kowalski, mark a Rubin, and Celina G Kleer. E-cadherin expression in primary carcinomas of the breast and its distant metastases. Breast Cancer Res. 2003;5(6):R217–22.CrossRef Paul J. Kowalski, mark a Rubin, and Celina G Kleer. E-cadherin expression in primary carcinomas of the breast and its distant metastases. Breast Cancer Res. 2003;5(6):R217–22.CrossRef
8.
go back to reference Chao YL, Shepard CR, Wells A. Breast carcinoma cells re-express E-cadherin during mesenchymal to epithelial reverting transition. Mol Cancer. 2010;9:179.CrossRefPubMedPubMedCentral Chao YL, Shepard CR, Wells A. Breast carcinoma cells re-express E-cadherin during mesenchymal to epithelial reverting transition. Mol Cancer. 2010;9:179.CrossRefPubMedPubMedCentral
9.
go back to reference Clark AM, Ma B, Taylor DL, Griffith L, Wells A. Liver metastases: microenvironments and ex-vivo models. Exp Biol Med (Maywood). 2016;241(15):1639–52.CrossRef Clark AM, Ma B, Taylor DL, Griffith L, Wells A. Liver metastases: microenvironments and ex-vivo models. Exp Biol Med (Maywood). 2016;241(15):1639–52.CrossRef
10.
go back to reference Wheeler SE, Clark AM, Taylor DP, Young CL, Pillai VC, Stolz DB, Venkataramanan R, Lauffenburger D, Griffith L, Wells A. Spontaneous dormancy of metastatic breast cancer cells in an all human liver microphysiologic system. Br J Cancer. 2014;111(12):2342–50.CrossRefPubMedPubMedCentral Wheeler SE, Clark AM, Taylor DP, Young CL, Pillai VC, Stolz DB, Venkataramanan R, Lauffenburger D, Griffith L, Wells A. Spontaneous dormancy of metastatic breast cancer cells in an all human liver microphysiologic system. Br J Cancer. 2014;111(12):2342–50.CrossRefPubMedPubMedCentral
11.
go back to reference Clark AM, Wheeler SE, Young CL, Stockdale L, Shepard Neiman J, Zhao W, Stolz DB, Venkataramanan R, Lauffenburger D, Griffithc L, Wells A. A liver microphysiological system of tumor cell dormancy and inflammatory responsiveness is affected by scaffold properties. Lab Chip. 2017;17:156–68.CrossRef Clark AM, Wheeler SE, Young CL, Stockdale L, Shepard Neiman J, Zhao W, Stolz DB, Venkataramanan R, Lauffenburger D, Griffithc L, Wells A. A liver microphysiological system of tumor cell dormancy and inflammatory responsiveness is affected by scaffold properties. Lab Chip. 2017;17:156–68.CrossRef
13.
go back to reference Azmi AS, Bao B, Sarkar FH. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metas Rev. 2013;32:623–42.CrossRef Azmi AS, Bao B, Sarkar FH. Exosomes in cancer development, metastasis, and drug resistance: a comprehensive review. Cancer Metas Rev. 2013;32:623–42.CrossRef
14.
go back to reference Kahlert C, Kalluri R. Exosomes in tumor microenvironment influence cancer progression and metastasis. J Mol Med (Berl). 2013;91(4):431–7.CrossRef Kahlert C, Kalluri R. Exosomes in tumor microenvironment influence cancer progression and metastasis. J Mol Med (Berl). 2013;91(4):431–7.CrossRef
15.
go back to reference Syn N, Wang L, Sethi G, Thiery JP, Goh BC. Exosome-mediated metastasis: from epithelial-Mesenchymal transition to escape from Immunosurveillance. Trends Pharmacol Sci. 2016;37(7):606–17.CrossRefPubMed Syn N, Wang L, Sethi G, Thiery JP, Goh BC. Exosome-mediated metastasis: from epithelial-Mesenchymal transition to escape from Immunosurveillance. Trends Pharmacol Sci. 2016;37(7):606–17.CrossRefPubMed
16.
17.
go back to reference Abd-Elmageed ZY, Yang Y, Thomas R, Ranjan M, Mondal D, Moroz K, Fang Z, Rezk BM, Moparty K, Sikka SC, Sartor O, Abdel-Mageed AB. Neoplastic reprogramming of patient-derived adipose stem cells by prostate cancer cell-associated exosomes. Stem Cells. 2014;32:983–97.CrossRefPubMedPubMedCentral Abd-Elmageed ZY, Yang Y, Thomas R, Ranjan M, Mondal D, Moroz K, Fang Z, Rezk BM, Moparty K, Sikka SC, Sartor O, Abdel-Mageed AB. Neoplastic reprogramming of patient-derived adipose stem cells by prostate cancer cell-associated exosomes. Stem Cells. 2014;32:983–97.CrossRefPubMedPubMedCentral
18.
go back to reference Grahovac J, Becker D, Wells A. Melanoma cell invasiveness is promoted at least in part by the epidermal growth factor-like repeats of tenascin-C. J Invest Dermatol. 2013;133(1):210–20.CrossRefPubMed Grahovac J, Becker D, Wells A. Melanoma cell invasiveness is promoted at least in part by the epidermal growth factor-like repeats of tenascin-C. J Invest Dermatol. 2013;133(1):210–20.CrossRefPubMed
19.
go back to reference Théry C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;Chapter 3:Unit 3.22.PubMed Théry C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol. 2006;Chapter 3:Unit 3.22.PubMed
20.
go back to reference Hwa AJ, Fry RC, Sivaraman A, So PT, Samson LD, Stolz DB, Griffith LG. Rat liver sinusoidal endothelial cells survive without exogenous VEGF in 3D perfused co-cultures with hepatocytes. FASEB J. 2007;21(10):2564–79.CrossRefPubMed Hwa AJ, Fry RC, Sivaraman A, So PT, Samson LD, Stolz DB, Griffith LG. Rat liver sinusoidal endothelial cells survive without exogenous VEGF in 3D perfused co-cultures with hepatocytes. FASEB J. 2007;21(10):2564–79.CrossRefPubMed
21.
go back to reference Wheeler SE, Borenstein JT, Clark AM, Ebrahimkhani M, Fox I, Griffith L, Inman W, Lauffenburger D, Nguyen T, Prantil-Baun R, Stolz D, Taylor D, Ulrich T, Venkateswaran C, Venkataramanan R, Wells A and C Young. All-human microphysical model of metastasis therapy. Stem Cell Research and Therapy. 2013;4:S11. Wheeler SE, Borenstein JT, Clark AM, Ebrahimkhani M, Fox I, Griffith L, Inman W, Lauffenburger D, Nguyen T, Prantil-Baun R, Stolz D, Taylor D, Ulrich T, Venkateswaran C, Venkataramanan R, Wells A and C Young. All-human microphysical model of metastasis therapy. Stem Cell Research and Therapy. 2013;4:S11.
22.
go back to reference Tian T, Wang Y, Wang H, Zhu Z, Xiao Z. Visualizing of the cellular uptake and intracellular trafficking of Exosomes by live-cell microscopy. J Cell Biochem. 2010;111:488–96.CrossRefPubMed Tian T, Wang Y, Wang H, Zhu Z, Xiao Z. Visualizing of the cellular uptake and intracellular trafficking of Exosomes by live-cell microscopy. J Cell Biochem. 2010;111:488–96.CrossRefPubMed
23.
go back to reference Tian T, Zhu YL, Hu FH, Wang YY, Huang NP, Xiao ZD. Dynamics of exosome internalization and trafficking. J Cell Physiol. 2013;228(7):1487–95.CrossRefPubMed Tian T, Zhu YL, Hu FH, Wang YY, Huang NP, Xiao ZD. Dynamics of exosome internalization and trafficking. J Cell Physiol. 2013;228(7):1487–95.CrossRefPubMed
24.
go back to reference Chao Y, Wu Q, Shepard C, Wells A. Hepatocyte induced re-expression of E-cadherin in breast and prostate cancer cells increases chemoresistance. Clin Exp Metastasis. 2012;29:39–50.CrossRefPubMed Chao Y, Wu Q, Shepard C, Wells A. Hepatocyte induced re-expression of E-cadherin in breast and prostate cancer cells increases chemoresistance. Clin Exp Metastasis. 2012;29:39–50.CrossRefPubMed
25.
go back to reference Mincy C, Eapen A. Exosome: potential biomarker for cancer. World J Res Rev (WJRR). 2016;2(4):01–4. Mincy C, Eapen A. Exosome: potential biomarker for cancer. World J Res Rev (WJRR). 2016;2(4):01–4.
26.
go back to reference Chevillet JR, Kang Q, Ruf IK, Briggs HA, Vojtech LN, Hughes SM, Cheng HH, Arroyo JD, Meredith EK, Gallichotte EN, Pogosova-Agadjanyan EL, Morrissey C, Stirewalt DL, Hladik F, Yu EY, Higano CS, Tewari M. Quantitative and stoichiometric analysis of the microRNA content of exosomes. Proc Natl Acad Sci (USA). 2014;111:14888–93. Chevillet JR, Kang Q, Ruf IK, Briggs HA, Vojtech LN, Hughes SM, Cheng HH, Arroyo JD, Meredith EK, Gallichotte EN, Pogosova-Agadjanyan EL, Morrissey C, Stirewalt DL, Hladik F, Yu EY, Higano CS, Tewari M. Quantitative and stoichiometric analysis of the microRNA content of exosomes. Proc Natl Acad Sci (USA). 2014;111:14888–93.
27.
go back to reference Hui X, Jiang J, Li Z, Ding L, Wang Y. MiR-186 acts as a tumor suppressor by targeting TWIST1/2 and regulating the epithelial-mesenchymal transition in glioblastoma multiforme. Int J Clin Exp Pathol. 2016;9(9):9706–14. Hui X, Jiang J, Li Z, Ding L, Wang Y. MiR-186 acts as a tumor suppressor by targeting TWIST1/2 and regulating the epithelial-mesenchymal transition in glioblastoma multiforme. Int J Clin Exp Pathol. 2016;9(9):9706–14.
28.
go back to reference Yao K, He L, Gan Y, Zeng Q, Dai Y, Tan J. MiR-186 suppresses the growth and metastasis of bladder cancer by targeting NSBP1. Diagn Pathol. 2015;10:146.CrossRefPubMedPubMedCentral Yao K, He L, Gan Y, Zeng Q, Dai Y, Tan J. MiR-186 suppresses the growth and metastasis of bladder cancer by targeting NSBP1. Diagn Pathol. 2015;10:146.CrossRefPubMedPubMedCentral
29.
go back to reference Dong Y, Jin X, Sun Z, Zhao Y, Song X. MiR-186 inhibited migration of NSCLC via targeting cdc42 and effecting EMT process. Mol Cells. 2017;40(3):195–201.PubMedPubMedCentral Dong Y, Jin X, Sun Z, Zhao Y, Song X. MiR-186 inhibited migration of NSCLC via targeting cdc42 and effecting EMT process. Mol Cells. 2017;40(3):195–201.PubMedPubMedCentral
30.
go back to reference Zhu X, Shen H, Yin X, Long L, Xie C, Liu Y, Hui L, Lin X, Fang Y, Cao Y, Xu Y, Li M, Xu W, Li Y. miR-186 regulation of Twist1 and ovarian cancer sensitivity to cisplatin. Oncogene. 2016;35(3):323–32.CrossRefPubMed Zhu X, Shen H, Yin X, Long L, Xie C, Liu Y, Hui L, Lin X, Fang Y, Cao Y, Xu Y, Li M, Xu W, Li Y. miR-186 regulation of Twist1 and ovarian cancer sensitivity to cisplatin. Oncogene. 2016;35(3):323–32.CrossRefPubMed
31.
go back to reference Yanjiao M, Shixiu W, Ruping Z, Qinghua D. MiR-205 promotes proliferation, migration and invasion of nasopharyngeal carcinoma cells by activation of AKT signaling. J Int Med Res. 2016;44(2):231–40.CrossRef Yanjiao M, Shixiu W, Ruping Z, Qinghua D. MiR-205 promotes proliferation, migration and invasion of nasopharyngeal carcinoma cells by activation of AKT signaling. J Int Med Res. 2016;44(2):231–40.CrossRef
32.
go back to reference Li J, Yang S, Yan W, Yang J, Qin YJ, Lin XL, Xie RY, Wang SC, Jin W, Gao F, Shi JW, Zhao WT, Jia JS, Shen HF, Ke JR, Liu B, Zhao YQ, Huang WH, Yao KT, Li DJ, Xiao D. MicroRNA-19 triggers epithelial-Mesenchymal transition of lung cancer cells accompanied by growth inhibition. Lab Investig. 2015;95(9):1056–70.CrossRefPubMed Li J, Yang S, Yan W, Yang J, Qin YJ, Lin XL, Xie RY, Wang SC, Jin W, Gao F, Shi JW, Zhao WT, Jia JS, Shen HF, Ke JR, Liu B, Zhao YQ, Huang WH, Yao KT, Li DJ, Xiao D. MicroRNA-19 triggers epithelial-Mesenchymal transition of lung cancer cells accompanied by growth inhibition. Lab Investig. 2015;95(9):1056–70.CrossRefPubMed
34.
go back to reference Melo SA, Sugimoto H, O'Connell JT, Kato N, Villanueva A, Vidal A, Qiu L, Vitkin E, Perelman LT, Melo CA, Lucci A, Ivan C, Calin GA, Kalluri R. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell. 2014;26(5):707–21.CrossRefPubMedPubMedCentral Melo SA, Sugimoto H, O'Connell JT, Kato N, Villanueva A, Vidal A, Qiu L, Vitkin E, Perelman LT, Melo CA, Lucci A, Ivan C, Calin GA, Kalluri R. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell. 2014;26(5):707–21.CrossRefPubMedPubMedCentral
35.
go back to reference Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011;71:3792–801.CrossRefPubMed Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011;71:3792–801.CrossRefPubMed
36.
go back to reference Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Redondo M, Williams C, García-Santos G, Ghajar C, Nitadori-Hoshino A, Hoffman C, Badal K, Garcia BA, Callahan MK, Yuan J, Martins VR, Skog J, Kaplan RN, Brady MS, Wolchok JD, Chapman PB, Kang Y, Bromberg J, Lyden D. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18(6):883–91.CrossRefPubMedPubMedCentral Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Redondo M, Williams C, García-Santos G, Ghajar C, Nitadori-Hoshino A, Hoffman C, Badal K, Garcia BA, Callahan MK, Yuan J, Martins VR, Skog J, Kaplan RN, Brady MS, Wolchok JD, Chapman PB, Kang Y, Bromberg J, Lyden D. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18(6):883–91.CrossRefPubMedPubMedCentral
37.
go back to reference Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, Becker A, Hoshino A, Mark MT, Molina H, Xiang J, Zhang T, Theilen TM, García-Santos G, Williams C, Ararso Y, Huang Y, Rodrigues G, Shen TL, Labori KJ, Lothe IM, Kure EH, Hernandez J, Doussot A, Ebbesen SH, Grandgenett PM, Hollingsworth MA, Jain M, Mallya K, Batra SK, Jarnagin WR, Schwartz RE, Matei I, Peinado H, Stanger BZ, Bromberg J, Lyden D. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17(6):816–26.CrossRefPubMed Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, Becker A, Hoshino A, Mark MT, Molina H, Xiang J, Zhang T, Theilen TM, García-Santos G, Williams C, Ararso Y, Huang Y, Rodrigues G, Shen TL, Labori KJ, Lothe IM, Kure EH, Hernandez J, Doussot A, Ebbesen SH, Grandgenett PM, Hollingsworth MA, Jain M, Mallya K, Batra SK, Jarnagin WR, Schwartz RE, Matei I, Peinado H, Stanger BZ, Bromberg J, Lyden D. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat Cell Biol. 2015;17(6):816–26.CrossRefPubMed
38.
go back to reference Peinado H, Lavotshkin S, Lyden D. The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Semin Cancer Biol. 2011;21(2):139–46.CrossRefPubMed Peinado H, Lavotshkin S, Lyden D. The secreted factors responsible for pre-metastatic niche formation: old sayings and new thoughts. Semin Cancer Biol. 2011;21(2):139–46.CrossRefPubMed
39.
go back to reference Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, MacDonald DD, Jin DK, Shido K, Kerns SA, Zhu Z, Hicklin D, Wu Y, Port JL, Altorki N, Port ER, Ruggero D, Shmelkov SV, Jensen KK, Rafii S, Lyden D. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438(7069):820–7.CrossRefPubMedPubMedCentral Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, MacDonald DD, Jin DK, Shido K, Kerns SA, Zhu Z, Hicklin D, Wu Y, Port JL, Altorki N, Port ER, Ruggero D, Shmelkov SV, Jensen KK, Rafii S, Lyden D. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438(7069):820–7.CrossRefPubMedPubMedCentral
40.
go back to reference Lin KK, Rossi L, Boles NC, Hall BE, George TC, Goodell MA. CD81 is essential for the re-entry of hematopoietic stem cells to quiescence following stress-induced proliferation via deactivation of the Akt pathway. PLoS Biol. 2011 Sep;9(9):e1001148.CrossRefPubMedPubMedCentral Lin KK, Rossi L, Boles NC, Hall BE, George TC, Goodell MA. CD81 is essential for the re-entry of hematopoietic stem cells to quiescence following stress-induced proliferation via deactivation of the Akt pathway. PLoS Biol. 2011 Sep;9(9):e1001148.CrossRefPubMedPubMedCentral
41.
go back to reference Glinskii OV, Huxley VH, Glinsky GV, Pienta KJ, Raz A, Glinsky VV. Mechanical entrapment is insufficient and intercellular adhesion is essential for metastatic cell arrest in distant organs. Neoplasia. 2005;7:522–7.CrossRefPubMedPubMedCentral Glinskii OV, Huxley VH, Glinsky GV, Pienta KJ, Raz A, Glinsky VV. Mechanical entrapment is insufficient and intercellular adhesion is essential for metastatic cell arrest in distant organs. Neoplasia. 2005;7:522–7.CrossRefPubMedPubMedCentral
42.
go back to reference Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002;3:415–28.CrossRefPubMed Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002;3:415–28.CrossRefPubMed
43.
go back to reference Strathdee G. Epigenetic versus genetic alterations in the inactivation of E-cadherin seminars. Cancer Biol Ther. 2002;12:373–9. Sumadi Lukman AnwarCrossRef Strathdee G. Epigenetic versus genetic alterations in the inactivation of E-cadherin seminars. Cancer Biol Ther. 2002;12:373–9. Sumadi Lukman AnwarCrossRef
44.
go back to reference Anwar SL, Lehmann U. DNA methylation, microRNAs, and their crosstalk as potential biomarkers in hepatocellular carcinoma. World J Gastroenterol. 2014 Jun 28;20(24):7894–913.CrossRefPubMedPubMedCentral Anwar SL, Lehmann U. DNA methylation, microRNAs, and their crosstalk as potential biomarkers in hepatocellular carcinoma. World J Gastroenterol. 2014 Jun 28;20(24):7894–913.CrossRefPubMedPubMedCentral
45.
go back to reference Qian Z, Shen Q, Yang X, Qiu Y, Zhang W. The role of extracellular vesicles: an epigenetic view of the cancer microenvironment. Biomed Res Int. 2015;2015:649161.PubMedPubMedCentral Qian Z, Shen Q, Yang X, Qiu Y, Zhang W. The role of extracellular vesicles: an epigenetic view of the cancer microenvironment. Biomed Res Int. 2015;2015:649161.PubMedPubMedCentral
Metadata
Title
Bi-directional exosome-driven intercommunication between the hepatic niche and cancer cells
Authors
Nikolina Dioufa
Amanda M. Clark
Bo Ma
Colin H. Beckwitt
Alan Wells
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2017
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-017-0740-6

Other articles of this Issue 1/2017

Molecular 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