Modifying the Tumour Microenvironment: Challenges and Future Perspectives for Anticancer Plasma Treatments
Abstract
:1. Introduction
2. Plasma-Derived ROS, Cell Death, and the ECM
3. The Tumour Microenvironment (TME)
3.1. Cellular Components of the Tumour Microenvironment
3.2. Acellular Components of the Tumour Microenvironment
4. Mechanisms of Cell Communication
4.1. Cell-to-Cell Communication
4.2. Cell-to-ECM Communication
5. Novel 3D in vitro Models to Explore the Effect of Plasma on the TME
5.1. Spheroids
5.2. Organoids
5.3. Scaffolds
5.4. Microfluidics-Based Tumour Models—Tumour-on-a-Chip
5.5. 3D Bioprinted Tumour Model
6. Perspectives and Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Roma-Rodrigues, C.; Mendes, R.; Baptista, P.V.; Fernandes, A.R. Targeting tumor microenvironment for cancer therapy. Int. J. Mol. Sci. 2019, 20, 840. [Google Scholar] [CrossRef]
- Conrads, H.; Schmidt, M. Plasma generation and plasma sources. Plasma Sources Sci. Technol. 2000, 9, 441–454. [Google Scholar] [CrossRef]
- Moreau, M.; Orange, N.; Feuilloley, M.G. Non-thermal plasma technologies: New tools for bio-decontamination. Biotechnol. Adv. 2008, 26, 610–617. [Google Scholar] [CrossRef] [PubMed]
- Fridman, A.A. Plasma Chemistry; Cambridge University Press: Cambridge, UK; New York, NY, USA, 2008; p. xlii. 978p. [Google Scholar]
- O’Connell, D.; Cox, L.J.; Hyland, W.B.; McMahon, S.J.; Reuter, S.; Graham, W.G.; Gans, T.; Currell, F.J. Cold atmospheric pressure plasma jet interactions with plasmid DNA. Appl. Phys. Lett. 2011, 98, 043701. [Google Scholar] [CrossRef]
- Moisan, M.; Barbeau, J.; Crevier, M.C.; Pelletier, J.; Philip, N.; Saoudi, B. Plasma sterilization. Methods mechanisms. Pure Appl. Chem. 2002, 74, 349–358. [Google Scholar] [CrossRef]
- Graves, D.B. The emerging role of reactive oxygen and nitrogen species in redox biology and some implications for plasma applications to medicine and biology. J. Phys. D Appl. Phys. 2012, 45, 263001. [Google Scholar] [CrossRef]
- Fridman, A.; Chirokov, A.; Gutsol, A. Non-thermal atmospheric pressure discharges. J. Phys. D Appl. Phys. 2005, 38, R1–R24. [Google Scholar] [CrossRef]
- Reuter, S.; von Woedtke, T.; Weltmann, K.D. The kINPen-a review on physics and chemistry of the atmospheric pressure plasma jet and its applications. J. Phys. D Appl. Phys. 2018, 51. [Google Scholar] [CrossRef]
- Wende, K.; von Woedtke, T.; Weltmann, K.D.; Bekeschus, S. Chemistry and biochemistry of cold physical plasma derived reactive species in liquids. Biol. Chem. 2019, 400, 19–38. [Google Scholar] [CrossRef]
- Girard, F.; Peret, M.; Dumont, N.; Badets, V.; Blanc, S.; Gazeli, K.; Noel, C.; Belmonte, T.; Marlin, L.; Cambus, J.P.; et al. Correlations between gaseous and liquid phase chemistries induced by cold atmospheric plasmas in a physiological buffer. Phys. Chem. Chem. Phys. 2018, 20, 9198–9210. [Google Scholar] [CrossRef]
- Gorbanev, Y.; O’Connell, D.; Chechik, V. Non-Thermal Plasma in Contact with Water: The Origin of Species. Chem. Eur. J. 2016, 22, 3496–3505. [Google Scholar] [CrossRef] [PubMed]
- Gorbanev, Y.; Privat-Maldonado, A.; Bogaerts, A. Analysis of Short-Lived Reactive Species in Plasma-Air-Water Systems: The Dos and the Do Nots. Anal Chem. 2018, 90, 13151–13158. [Google Scholar] [CrossRef] [PubMed]
- Weltmann, K.D.; von Woedtke, T. Plasma medicine—Current state of research and medical application. Plasma Phys. Control. F 2017, 59, 014031. [Google Scholar] [CrossRef]
- Graves, D.B. Low temperature plasma biomedicine: A tutorial review. Phys. Plasmas 2014, 21. [Google Scholar] [CrossRef]
- Lin, A.; Truong, B.; Patel, S.; Kaushik, N.; Choi, E.H.; Fridman, G.; Fridman, A.; Miller, V. Nanosecond-pulsed DBD plasma-generated reactive oxygen species trigger immunogenic cell death in A549 lung carcinoma cells through intracellular oxidative stress. Int. J. Mol. Sci. 2017, 18, 966. [Google Scholar] [CrossRef]
- Oehmigen, K.; Hahnel, M.; Brandenburg, R.; Wilke, C.; Weltmann, K.D.; von Woedtke, T. The Role of Acidification for Antimicrobial Activity of Atmospheric Pressure Plasma in Liquids. Plasma Process. Polym. 2010, 7, 250–257. [Google Scholar] [CrossRef]
- Weidinger, A.; Kozlov, A.V. Biological activities of reactive oxygen and nitrogen species: Oxidative stress versus signal transduction. Biomolecules 2015, 5, 472–484. [Google Scholar] [CrossRef]
- Privat-Maldonado, A.; Schmidt, A.; Lin, A.; Weltmann, K.-D.; Wende, K.; Bogaerts, A.; Bekeschus, S. ROS from physical plasmas: Redox chemistry for biomedical therapy. Oxid. Med. Cell. Longev. 2019, 2019, 29. [Google Scholar] [CrossRef]
- Kim, J.; Kim, J.; Bae, J.S. ROS homeostasis and metabolism: A critical liaison for cancer therapy. Exp. Mol. Med. 2016, 48, e269. [Google Scholar] [CrossRef]
- Movahed, Z.G.; Rastegari-Pouyani, M.; Mohammadi, M.H.; Mansouri, K. Cancer cells change their glucose metabolism to overcome increased ROS: One step from cancer cell to cancer stem cell? Biomed. Pharmacother. 2019, 112. [Google Scholar] [CrossRef]
- Trachootham, D.; Alexandre, J.; Huang, P. Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach? Nat. Rev. Drug Discov. 2009, 8, 579–591. [Google Scholar] [CrossRef] [PubMed]
- Riley, P.A. Free radicals in biology: Oxidative stress and the effects of ionizing radiation. Int. J. Radiat. Biol. 1994, 65, 27–33. [Google Scholar] [CrossRef] [PubMed]
- Castaldo, S.A.; Freitas, J.R.; Conchinha, N.V.; Madureira, P.A. The Tumorigenic Roles of the Cellular REDOX Regulatory Systems. Oxid. Med. Cell. Longev. 2016, 2016, 8413032. [Google Scholar] [CrossRef] [PubMed]
- Danhier, P.; De Saedeleer, C.J.; Karroum, O.; De Preter, G.; Porporato, P.E.; Jordan, B.F.; Gallez, B.; Sonveaux, P. Optimization of tumor radiotherapy with modulators of cell metabolism: Toward clinical applications. Semin. Radiat. Oncol. 2013, 23, 262–272. [Google Scholar] [CrossRef]
- Liedtke, K.R.; Diedrich, S.; Pati, O.; Freund, E.; Flieger, R.; Heidecke, C.D.; Partecke, L.I.; Bekeschus, S. Cold Physical Plasma Selectively Elicits Apoptosis in Murine Pancreatic Cancer Cells In Vitro and In Ovo. Anticancer Res. 2018, 38, 5655–5663. [Google Scholar] [CrossRef]
- Liu, J.; Wang, Z. Increased oxidative stress as a selective anticancer therapy. Oxid. Med. Cell. Longev. 2015, 2015, 294303. [Google Scholar] [CrossRef]
- Privat-Maldonado, A.; Gorbanev, Y.; Dewilde, S.; Smits, E.; Bogaerts, A. Reduction of Human Glioblastoma Spheroids Using Cold Atmospheric Plasma: The Combined Effect of Short- and Long-Lived Reactive Species. Cancers 2018, 10, 394. [Google Scholar] [CrossRef]
- Dubuc, A.; Monsarrat, P.; Virard, F.; Merbahi, N.; Sarrette, J.P.; Laurencin-Dalicieux, S.; Cousty, S. Use of cold-atmospheric plasma in oncology: A concise systematic review. Ther. Adv. Med. Oncol. 2018, 10, 1–12. [Google Scholar] [CrossRef]
- Vandamme, M.; Robert, E.; Dozias, S.; Sobilo, J.; Lerondel, S.; Le Pape, A.; Pouvesle, J.-M. Response of human glioma U87 xenografted on mice to non thermal plasma treatment. Plasma Med. 2011, 1, 27. [Google Scholar] [CrossRef]
- Vandamme, M.; Robert, E.; Lerondel, S.; Sarron, V.; Ries, D.; Dozias, S.; Sobilo, J.; Gosset, D.; Kieda, C.; Legrain, B.; et al. ROS implication in a new antitumor strategy based on non-thermal plasma. Int. J. Cancer 2012, 130, 2185–2194. [Google Scholar] [CrossRef]
- Brulle, L.; Vandamme, M.; Ries, D.; Martel, E.; Robert, E.; Lerondel, S.; Trichet, V.; Richard, S.; Pouvesle, J.M.; Le Pape, A. Effects of a non thermal plasma treatment alone or in combination with gemcitabine in a MIA PaCa2-luc orthotopic pancreatic carcinoma model. PLoS ONE 2012, 7, e52653. [Google Scholar] [CrossRef] [PubMed]
- Lin, L.; Wang, L.; Liu, Y.; Xu, C.; Tu, Y.; Zhou, J. Nonthermal plasma inhibits tumor growth and proliferation and enhances the sensitivity to radiation in vitro and in vivo. Oncol. Rep. 2018, 40, 3405–3415. [Google Scholar] [CrossRef] [PubMed]
- Mizuno, K.; Yonetamari, K.; Shirakawa, Y.; Akiyama, T.; Ono, R. Anti-tumor immune response induced by nanosecond pulsed streamer discharge in mice. J. Phys. D Appl. Phys. 2017, 50, 12LT01. [Google Scholar] [CrossRef]
- Freund, E.; Liedtke, K.R.; van der Linde, J.; Metelmann, H.R.; Heidecke, C.D.; Partecke, L.I.; Bekeschus, S. Physical plasma-treated saline promotes an immunogenic phenotype in CT26 colon cancer cells in vitro and in vivo. Sci. Rep. 2019, 9, 634. [Google Scholar] [CrossRef] [PubMed]
- Tsai, M.J.; Chang, W.A.; Huang, M.S.; Kuo, P.L. Tumor microenvironment: A new treatment target for cancer. ISRN Biochem. 2014, 2014, 351959. [Google Scholar] [CrossRef] [PubMed]
- Lu, X.; Reuter, S.; Laroussi, M.; Liu, D. Nonequilibrium Atmospheric Pressure Plasma Jets: Fundamentals, Diagnostics, and Medical Applications; CRC Press, Taylor & Francis Group: Boca Raton, FL, USA, 2019; pp. 307–347. [Google Scholar]
- Van der Paal, J.; Neyts, E.C.; Verlackt, C.C.W.; Bogaerts, A. Effect of lipid peroxidation on membrane permeability of cancer and normal cells subjected to oxidative stress. Chem. Sci. 2016, 7, 489–498. [Google Scholar] [CrossRef] [Green Version]
- Van der Paal, J.; Verheyen, C.; Neyts, E.C.; Bogaerts, A. Hampering Effect of Cholesterol on the Permeation of Reactive Oxygen Species through Phospholipids Bilayer: Possible Explanation for Plasma Cancer Selectivity. Sci. Rep. 2017, 7, 39526. [Google Scholar] [CrossRef]
- Shaw, P.; Kumar, N.; Hammerschmid, D.; Privat-Maldonado, A.; Dewilde, S.; Bogaerts, A. Synergistic effects of melittin and plasma treatment: A promising approach for cancer therapy. Cancers 2019, 11, 1109. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Chen, X.; Gueydan, C.; Han, J. Plasma membrane changes during programmed cell deaths. Cell Res. 2018, 28, 9–21. [Google Scholar] [CrossRef]
- Dezest, M.; Chavatte, L.; Bourdens, M.; Quinton, D.; Camus, M.; Garrigues, L.; Descargues, P.; Arbault, S.; Burlet-Schiltz, O.; Casteilla, L.; et al. Mechanistic insights into the impact of cold atmospheric pressure plasma on human epithelial cell lines. Sci. Rep. 2017, 7, 41163. [Google Scholar] [CrossRef] [Green Version]
- Yan, D.; Talbot, A.; Nourmohammadi, N.; Sherman, J.H.; Cheng, X.; Keidar, M. Toward understanding the selective anticancer capacity of cold atmospheric plasma—A model based on aquaporins (Review). Biointerphases 2015, 10, 040801. [Google Scholar] [CrossRef] [PubMed]
- Poltavets, V.; Kochetkova, M.; Pitson, S.M.; Samuel, M.S. The role of the extracellular matrix and its molecular and cellular regulators in cancer cell plasticity. Front. Oncol. 2018, 8, 431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hirst, A.M.; Simms, M.S.; Mann, V.M.; Maitland, N.J.; O’Connell, D.; Frame, F.M. Low-temperature plasma treatment induces DNA damage leading to necrotic cell death in primary prostate epithelial cells. Br. J. Cancer 2015, 112, 1536–1545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, X.; Klas, M.; Liu, Y.Y.; Stack, M.S.; Ptasinska, S. DNA damage in oral cancer cells induced by nitrogen atmospheric pressure plasma jets. Appl. Phys. Lett. 2013, 102. [Google Scholar] [CrossRef]
- Dickey, J.S.; Redon, C.E.; Nakamura, A.J.; Baird, B.J.; Sedelnikova, O.A.; Bonner, W.M. H2AX: Functional roles and potential applications. Chromosoma 2009, 118, 683–692. [Google Scholar] [CrossRef] [Green Version]
- Ahn, H.J.; Kim, K.I.; Kim, G.; Moon, E.; Yang, S.S.; Lee, J.S. Atmospheric-pressure plasma jet induces apoptosis involving mitochondria via generation of free radicals. PLoS ONE 2011, 6, 7. [Google Scholar] [CrossRef] [Green Version]
- Joh, H.M.; Kim, S.J.; Chung, T.H.; Leem, S.H. Reactive oxygen species-related plasma effects on the apoptosis of human bladder cancer cells in atmospheric pressure pulsed plasma jets. Appl. Phys. Lett. 2012, 101, 5. [Google Scholar] [CrossRef]
- Keidar, M. Plasma for cancer treatment. Plasma Sources Sci. Technol. 2015, 24, 20. [Google Scholar] [CrossRef]
- Kim, C.H.; Bahn, J.H.; Lee, S.H.; Kim, G.Y.; Jun, S.I.; Lee, K.; Baek, S.J. Induction of cell growth arrest by atmospheric non-thermal plasma in colorectal cancer cells. J. Biotechnol. 2010, 150, 530–538. [Google Scholar] [CrossRef]
- Kim, S.J.; Chung, T.H.; Bae, S.H.; Leem, S.H. Induction of apoptosis in human breast cancer cells by a pulsed atmospheric pressure plasma jet. Appl. Phys. Lett. 2010, 97, 3. [Google Scholar] [CrossRef]
- Ratovitski, E.A.; Cheng, X.Q.; Yan, D.Y.; Sherman, J.H.; Canady, J.; Trink, B.; Keidar, M. Anti-cancer therapies of 21st century: Novel approach to treat human cancers using cold atmospheric plasma. Plasma Process. Polym. 2014, 11, 1128–1137. [Google Scholar] [CrossRef]
- Yan, D.Y.; Sherman, J.H.; Keidar, M. Cold atmospheric plasma, a novel promising anti-cancer treatment modality. Oncotarget 2017, 8, 15977–15995. [Google Scholar] [CrossRef] [PubMed]
- Arjunan, K.P.; Friedman, G.; Fridman, A.; Clyne, A.M. Non-thermal dielectric barrier discharge plasma induces angiogenesis through reactive oxygen species. J. R. Soc. Interface 2012, 9, 147–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ishaq, M.; Kumar, S.; Varinli, H.; Han, Z.J.; Rider, A.E.; Evans, M.D.M.; Murphy, A.B.; Ostrikov, K. Atmospheric gas plasma-induced ROS production activates TNF-ASK1 pathway for the induction of melanoma cancer cell apoptosis. Mol. Biol. Cell 2014, 25, 1523–1531. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.J.; Chung, T.H. Cold atmospheric plasma jet-generated RONS and their selective effects on normal and carcinoma cells. Sci. Rep. 2016, 6, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bauer, G.; Graves, D.B. Mechanisms of selective antitumor action of cold atmospheric plasma-derived reactive oxygen and nitrogen species. Plasma Process. Polym. 2016, 13, 1157–1178. [Google Scholar] [CrossRef]
- Bauer, G.; Sersenová, D.; Graves, D.B.; Machala, Z. Cold atmospheric plasma and plasma-activated medium trigger RONS-based tumor cell apoptosis. Sci. Rep. 2019, 9, 14210. [Google Scholar] [CrossRef]
- Bauer, G.; Sersenová, D.; Graves, D.B.; Machala, Z. Dynamics of singlet Oxygen-triggered, RONS-based apoptosis induction after treatment of tumor cells with cold atmospheric plasma or plasma-activated medium. Sci. Rep. 2019, 9, 13931. [Google Scholar] [CrossRef] [Green Version]
- Adachi, T.; Nonomura, S.; Horiba, M.; Hirayama, T.; Kamiya, T.; Nagasawa, H.; Hara, H. Iron stimulates plasma-activated medium-induced A549 cell injury. Sci. Rep. 2016, 6, 12. [Google Scholar] [CrossRef] [Green Version]
- Guerrero-Preston, R.; Ogawa, T.; Uemura, M.; Shumulinsky, G.; Valle, B.L.; Pirini, F.; Ravi, R.; Sidransky, D.; Keidar, M.; Trink, B. Cold atmospheric plasma treatment selectively targets head and neck squamous cell carcinoma cells. Int. J. Mol. Med. 2014, 34, 941–946. [Google Scholar] [CrossRef] [Green Version]
- Iseki, S.; Nakamura, K.; Hayashi, M.; Tanaka, H.; Kondo, H.; Kajiyama, H.; Kano, H.; Kikkawa, F.; Hori, M. Selective killing of ovarian cancer cells through induction of apoptosis by nonequilibrium atmospheric pressure plasma. Appl. Phys. Lett. 2012, 100, 4. [Google Scholar] [CrossRef]
- Ishaq, M.; Evans, M.D.M.; Ostrikov, K. Atmospheric pressure gas plasma-induced colorectal cancer cell death is mediated by Nox2-ASK1 apoptosis pathways and oxidative stress is mitigated by Srx-Nrf2 anti-oxidant system. Biochim. Biophys. Acta Mol. Cell Res. 2014, 1843, 2827–2837. [Google Scholar] [CrossRef] [PubMed]
- Kaushik, N.K.; Kaushik, N.; Park, D.; Choi, E.H. Altered antioxidant system stimulates dielectric barrier discharge plasma-induced cell death for solid tumor cell treatment. PLoS ONE 2014, 9, 11. [Google Scholar] [CrossRef] [PubMed]
- Keidar, M.; Walk, R.; Shashurin, A.; Srinivasan, P.; Sandler, A.; Dasgupta, S.; Ravi, R.; Guerrero-Preston, R.; Trink, B. Cold plasma selectivity and the possibility of a paradigm shift in cancer therapy. Br. J. Cancer 2011, 105, 1295–1301. [Google Scholar] [CrossRef] [PubMed]
- Kumar, N.; Park, J.H.; Jeon, S.N.; Park, B.S.; Choi, E.H.; Attri, P. The action of microsecond-pulsed plasma-activated media on the inactivation of human lung cancer cells. J. Phys. D Appl. Phys. 2016, 49, 9. [Google Scholar] [CrossRef]
- Kurake, N.; Tanaka, H.; Ishikawa, K.; Kondo, T.; Sekine, M.; Nakamura, K.; Kajiyama, H.; Kikkawa, F.; Mizuno, M.; Hori, M. Cell survival of glioblastoma grown in medium containing hydrogen peroxide and/or nitrite, or in plasma-activated medium. Arch. Biochem. Biophys. 2016, 605, 102–108. [Google Scholar] [CrossRef]
- Siu, A.; Volotskova, O.; Cheng, X.Q.; Khalsa, S.S.; Bian, K.; Murad, F.; Keidar, M.; Sherman, J.H. Differential effects of cold atmospheric plasma in the treatment of malignant glioma. PLoS ONE 2015, 10, 14. [Google Scholar] [CrossRef]
- Utsumi, F.; Kajiyama, H.; Nakamura, K.; Tanaka, H.; Hori, M.; Kikkawa, F. Selective cytotoxicity of indirect nonequilibrium atmospheric pressure plasma against ovarian clear-cell carcinoma. SpringerPlus 2014, 3, 9. [Google Scholar] [CrossRef] [Green Version]
- Biscop, E.; Lin, A.; Boxem, W.V.; Loenhout, J.V.; Backer, J.; Deben, C.; Dewilde, S.; Smits, E.; Bogaerts, A.A. Influence of cell type and culture medium on determining cancer selectivity of cold atmospheric plasma treatment. Cancers 2019, 11, 1287. [Google Scholar] [CrossRef] [Green Version]
- Du, J.; Liu, J.; Smith, B.J.; Tsao, M.S.; Cullen, J.J. Role of Rac1-dependent NADPH oxidase in the growth of pancreatic cancer. Cancer Gene Ther. 2011, 18, 135–143. [Google Scholar] [CrossRef] [Green Version]
- Irani, K.; Goldschmidt-Clermont, P.J. Ras, superoxide and signal transduction. Biochem. Pharmacol. 1998, 55, 1339–1346. [Google Scholar] [PubMed]
- Irani, K.; Xia, Y.; Zweier, J.L.; Sollott, S.J.; Der, C.J.; Fearon, E.R.; Sundaresan, M.; Finkel, T.; Goldschmidt-Clermont, P.J. Mitogenic signaling mediated by oxidants in ras-transformed fibroblasts. Science 1997, 275, 1649–1652. [Google Scholar] [CrossRef] [PubMed]
- Kamata, T. Roles of Nox1 and other Nox isoforms in cancer development. Cancer Sci. 2009, 100, 1382–1388. [Google Scholar] [CrossRef] [PubMed]
- Lambeth, J.D. Nox enzymes, ROS, and chronic disease: An example of antagonistic pleiotropy. Free Radic. Biol. Med. 2007, 43, 332–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Laurent, E.; McCoy, J.W.; Macina, R.A.; Liu, W.H.; Cheng, G.J.; Robine, S.; Papkoff, J.; Lambeth, J.D. Nox1 is over-expressed in human colon cancers and correlates with activating mutations in K-Ras. Int. J. Cancer 2008, 123, 100–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, Q.; Cavallin, L.E.; Yan, B.; Zhu, S.K.; Duran, E.M.; Wang, H.L.; Hale, L.P.; Dong, C.M.; Cesarman, E.; Mesri, E.A.; et al. Antitumorigenesis of antioxidants in a transgenic Rac1 model of Kaposi’s sarcoma. PNAS 2009, 106, 8683–8688. [Google Scholar] [CrossRef] [Green Version]
- Mitsushita, J.; Lambeth, J.D.; Kamata, T. The superoxide-generating oxidase Nox1 is functionally required for Ras oncogene transformation. Cancer Res. 2004, 64, 3580–3585. [Google Scholar] [CrossRef] [Green Version]
- Suh, Y.A.; Arnold, R.S.; Lassegue, B.; Shi, J.; Xu, X.X.; Sorescu, D.; Chung, A.B.; Griendling, K.K.; Lambeth, J.D. Cell transformation by the superoxide-generating oxidase Mox1. Nature 1999, 401, 79–82. [Google Scholar] [CrossRef]
- Tominaga, K.; Kawahara, T.; Sano, T.; Toida, K.; Kuwano, Y.; Sasaki, H.; Kawai, T.; Teshima-Kondo, S.; Rokutan, K. Evidence for cancer-associated expression of NADPH oxidase 1 (Nox1)-based oxidase system in the human stomach. Free Radic. Biol. Med. 2007, 43, 1627–1638. [Google Scholar] [CrossRef]
- Weinberg, F.; Chandel, N.S. Reactive oxygen species-dependent signaling regulates cancer. Cell. Mol. Life Sci. 2009, 66, 3663–3673. [Google Scholar] [CrossRef]
- Yang, J.Q.; Li, S.J.; Domann, F.E.; Buettner, G.R.; Oberley, L.W. Superoxide generation in v-Ha-ras-transduced human keratinocyte HaCaT cells. Mol. Carcinog. 1999, 26, 180–188. [Google Scholar] [CrossRef]
- Bauer, G. Tumor cell-protective catalase as a novel target for rational therapeutic approaches based on specific intercellular ROS signaling. Anticancer Res. 2012, 32, 2599–2624. [Google Scholar] [PubMed]
- Bauer, G. Targeting extracellular ROS signaling of tumor cells. Anticancer Res. 2014, 34, 1467–1482. [Google Scholar] [PubMed]
- Engelmann, I.; Dormann, S.; Saran, M.; Bauer, G. Transformed target cell-derived superoxide anions drive apoptosis induction by myeloperoxidase. Redox Rep. 2000, 5, 207–214. [Google Scholar] [CrossRef]
- Herdener, M.; Heigold, S.; Saran, M.; Bauer, G. Target cell-derived superoxide anions cause efficiency and selectivity of intercellular induction of apoptosis. Free Radic. Biol. Med. 2000, 29, 1260–1271. [Google Scholar] [CrossRef]
- Heigold, S.; Sers, C.; Bechtel, W.; Ivanovas, B.; Schafer, R.; Bauer, G. Nitric oxide mediates apoptosis induction selectively in transformed fibroblasts compared to nontransformed fibroblasts. Carcinogenesis 2002, 23, 929–941. [Google Scholar] [CrossRef]
- Semenza, G.L. Regulation of cancer cell metabolism by hypoxia-inducible factor 1. Semin. Cancer Biol. 2009, 19, 12–16. [Google Scholar] [CrossRef]
- Bechtel, W.; Bauer, G. Catalase protects tumor cells from apoptosis induction by intercellular ROS signaling. Anticancer Res. 2009, 29, 4541–4557. [Google Scholar]
- Bohm, B.; Heinzelmann, S.; Motz, M.; Bauer, G. Extracellular localization of catalase is associated with the transformed state of malignant cells. Biol. Chem. 2015, 396, 1339–1356. [Google Scholar] [CrossRef] [Green Version]
- Deichman, G.I. Natural selection and early changes of phenotype of tumor cells in vivo: Acquisition of new defense mechanisms. Biochem. 2000, 65, 78–94. [Google Scholar]
- Deichman, G.I. Early phenotypic changes of in vitro transformed cells during in vivo progression: Possible role of the host innate immunity. Semin. Cancer Biol. 2002, 12, 317–326. [Google Scholar] [CrossRef]
- Deichman, G.I.; Matveeva, V.A.; Kashkina, L.M.; Dyakova, N.A.; Uvarova, E.N.; Nikiforov, M.A.; Gudkov, A.V. Cell transforming genes and tumor progression: In vivo unified secondary phenotypic cell changes. Int. J. Cancer 1998, 75, 277–283. [Google Scholar] [CrossRef]
- Heinzelmann, S.; Bauer, G. Multiple protective functions of catalase against intercellular apoptosis-inducing ROS signaling of human tumor cells. Biol. Chem. 2010, 391, 675–693. [Google Scholar] [CrossRef] [PubMed]
- Bauer, G. The synergistic effect between hydrogen peroxide and nitrite, two long-lived molecular species from cold atmospheric plasma, triggers tumor cells to induce their own cell death. Redox Biol. 2019, 26. [Google Scholar] [CrossRef] [PubMed]
- Bauer, G. Intercellular singlet oxygen-mediated bystander signaling triggered by long-lived species of cold atmospheric plasma and plasma-activated medium. Redox Biol. 2019, 26. [Google Scholar] [CrossRef]
- Escobar, J.A.; Rubio, M.A.; Lissi, E.A. SOD and catalase inactivation by singlet oxygen and peroxyl radicals. Free Radic. Biol. Med. 1996, 20, 285–290. [Google Scholar] [CrossRef]
- Bauer, G. Increasing the endogenous NO level causes catalase inactivation and reactivation of intercellular apoptosis signaling specifically in tumor cells. Redox Biol. 2015, 6, 353–371. [Google Scholar] [CrossRef] [Green Version]
- Di Mascio, P.; Bechara, E.J.H.; Medeiros, M.H.G.; Briviba, K.; Sies, H. Singlet molecular-oxygen production in the reaction of peroxynitrite with hydrogen-peroxide. FEBS Lett. 1994, 355, 287–289. [Google Scholar] [CrossRef] [Green Version]
- Riethmuller, M.; Burger, N.; Bauer, G. Singlet oxygen treatment of tumor cells triggers extracellular singlet oxygen generation, catalase inactivation and reactivation of intercellular apoptosis-inducing signaling. Redox Biol. 2015, 6, 157–168. [Google Scholar] [CrossRef] [Green Version]
- Scheit, K.; Bauer, G. Direct and indirect inactivation of tumor cell protective catalase by salicylic acid and anthocyanidins reactivates intercellular ROS signaling and allows for synergistic effects. Carcinogenesis 2015, 36, 400–411. [Google Scholar] [CrossRef] [Green Version]
- Bengtson, C.; Bogaerts, A. Catalase dependence of cell signaling pathways underlying selectivity of cancer treatment by cold atmospheric plasma. Cancers 2019. submitted. [Google Scholar]
- Balkwill, F.R.; Capasso, M.; Hagemann, T. The tumor microenvironment at a glance. J. Cell Sci. 2012, 125, 5591–5596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rybalchenko, O.V.; Orlova, O.G.; Astaf’ev, A.M.; Pinchuk, M.E.; Stepanova, O.M.; Pariyskaya, E.N.; Zakharova, L.B. Morphological changes in infected wounds under the influence of non-thermal atmospheric pressure plasma. Res. J. Pharm Biol. Chem. Sci. 2018, 9, 1746–1753. [Google Scholar]
- Arndt, S.; Unger, P.; Berneburg, M.; Bosserhoff, A.K.; Karrer, S. Cold atmospheric plasma (CAP) activates angiogenesis-related molecules in skin keratinocytes, fibroblasts and endothelial cells and improves wound angiogenesis in an autocrine and paracrine mode. J. Dermatol. Sci. 2018, 89, 181–190. [Google Scholar] [CrossRef] [PubMed]
- Brun, P.; Pathak, S.; Castagliuolo, I.; Palu, G.; Brun, P.; Zuin, M.; Cavazzana, R.; Martines, E. Helium generated cold plasma finely regulates activation of human fibroblast-like primary cells. PLoS ONE 2014, 9, e104397. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.R.; Xu, G.M.; Shi, X.M.; Zhang, G.J. Low temperature plasma promoting fibroblast proliferation by activating the NF-κβ pathway and increasing cyclinD1 expression. Sci. Rep. 2017, 7. [Google Scholar] [CrossRef] [Green Version]
- Arndt, S.; Unger, P.; Wacker, E.; Shimizu, T.; Heinlin, J.; Li, Y.F.; Thomas, H.M.; Morfill, G.E.; Zimmermann, J.L.; Bosserhoff, A.K.; et al. Cold atmospheric plasma (CAP) changes gene expression of key molecules of the wound healing machinery and improves wound healing in vitro and in vivo. PLoS ONE 2013, 8, e79325. [Google Scholar] [CrossRef] [Green Version]
- Denton, A.E.; Roberts, E.W.; Fearon, D.T. Stromal cells in the tumor microenvironment. Adv. Exp. Med. Biol. 2018, 1060, 99–114. [Google Scholar] [CrossRef]
- Luga, V.; Zhang, L.; Viloria-Petit, A.M.; Ogunjimi, A.A.; Inanlou, M.R.; Chiu, E.; Buchanan, M.; Hosein, A.N.; Basik, M.; Wrana, J.L. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell 2012, 151, 1542–1556. [Google Scholar] [CrossRef] [Green Version]
- Zhou, W.Y.; Fong, M.Y.; Min, Y.F.; Somlo, G.; Liu, L.; Palomares, M.R.; Yu, Y.; Chow, A.; O’Connor, S.T.F.; Chin, A.R.; et al. Cancer-secreted miR-105 destroys vascular endothelial barriers to promote metastasis. Cancer Cell 2014, 25, 501–515. [Google Scholar] [CrossRef] [Green Version]
- Boelens, M.C.; Wu, T.J.; Nabet, B.Y.; Xu, B.; Qiu, Y.; Yoon, T.; Azzam, D.J.; Victor, C.T.S.; Wiemann, B.Z.; Ishwaran, H.; et al. Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell 2014, 159, 499–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, X.M.; Cai, J.F.; Xu, G.M.; Ren, H.B.; Chen, S.L.; Chang, Z.S.; Liu, J.R.; Huang, C.Y.; Zhang, G.J.; Wu, X.L. Effect of cold plasma on cell viability and collagen synthesis in cultured murine fibroblasts. Plasma Sci. Technol. 2016, 18, 353–359. [Google Scholar] [CrossRef]
- Bourdens, M.; Jeanson, Y.; Taurand, M.; Juin, N.; Carriere, A.; Clement, F.; Casteilla, L.; Bulteau, A.L.; Planat-Benard, V. Short exposure to cold atmospheric plasma induces senescence in human skin fibroblasts and adipose mesenchymal stromal cells. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Virard, F.; Cousty, S.; Cambus, J.P.; Valentin, A.; Kemoun, P.; Clement, F. Cold atmospheric plasma induces a predominantly necrotic cell death via the microenvironment. PLoS ONE 2015, 10, e0133120. [Google Scholar] [CrossRef]
- Hida, K.; Maishi, N.; Annan, D.A.; Hida, Y. Contribution of tumor endothelial cells in cancer progression. Int. J. Mol. Sci. 2018, 19, 1272. [Google Scholar] [CrossRef] [Green Version]
- Koumoutsakos, P.; Pivkin, I.; Milde, F. The fluid mechanics of cancer and its therapy. Annu. Rev. Fluid Mech. 2013, 45, 325–355. [Google Scholar] [CrossRef] [Green Version]
- Leunig, M.; Yuan, F.; Menger, M.D.; Boucher, Y.; Goetz, A.E.; Messmer, K.; Jain, R.K. Angiogenesis, microvascular architecture, microhemodynamics, and interstitial fluid pressure during early growth of human adenocarcinoma Ls174t in Scid mice. Cancer Res. 1992, 52, 6553–6560. [Google Scholar]
- Swartz, M.A.; Lund, A.W. Lymphatic and interstitial flow in the tumour microenvironment: Linking mechanobiology with immunity. Nat. Rev. Cancer 2012, 12, 210–219. [Google Scholar] [CrossRef]
- Gweon, B.; Kim, H.; Kim, K.; Kim, M.; Shim, E.; Kim, S.; Choe, W.; Shin, J.H. Suppression of angiogenesis by atmospheric pressure plasma in human aortic endothelial cells. Appl. Phys. Lett. 2014, 104. [Google Scholar] [CrossRef] [Green Version]
- Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 2018, 32, 1267–1284. [Google Scholar] [CrossRef] [Green Version]
- Evans, R.A.; Diamond, M.S.; Rech, A.J.; Chao, T.; Richardson, M.W.; Lin, J.H.; Bajor, D.L.; Byrne, K.T.; Stanger, B.Z.; Riley, J.L.; et al. Lack of immunoediting in murine pancreatic cancer reversed with neoantigen. JCI Insight 2016, 1. [Google Scholar] [CrossRef] [PubMed]
- Liedtke, K.R.; Freund, E.; Hackbarth, C.; Heidecke, C.D.; Partecke, L.I.; Bekeschus, S. A myeloid and lymphoid infiltrate in murine pancreatic tumors exposed to plasma-treated medium. Clin. Plasma Med. 2018, 11, 10–17. [Google Scholar] [CrossRef]
- Bekeschus, S.; Roder, K.; Fregin, B.; Otto, O.; Lippert, M.; Weltmann, K.D.; Wende, K.; Schmidt, A.; Gandhirajan, R.K. Toxicity and immunogenicity in murine melanoma following exposure to physical plasma-derived oxidants. Oxid. Med. Cell. Longev. 2017. [Google Scholar] [CrossRef] [PubMed]
- Bekeschus, S.; Clemen, R.; Metelmann, H.R. Potentiating anti-tumor immunity with physical plasma. Clin. Plasma Med. 2018, 12, 17–22. [Google Scholar] [CrossRef]
- Yuen, G.J.; Demissie, E.; Pillai, S. B lymphocytes and cancer: A love-hate relationship. Trends Cancer 2016, 2, 747–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andreu, P.; Johansson, M.; Affara, N.I.; Pucci, F.; Tan, T.; Junankar, S.; Korets, L.; Lam, J.; Tawfik, D.; DeNardo, D.G.; et al. FcRgamma activation regulates inflammation-associated squamous carcinogenesis. Cancer Cell 2010, 17, 121–134. [Google Scholar] [CrossRef] [Green Version]
- Mu, X.; Shi, W.; Xu, Y.; Xu, C.; Zhao, T.; Geng, B.; Yang, J.; Pan, J.; Hu, S.; Zhang, C.; et al. Tumor-derived lactate induces M2 macrophage polarization via the activation of the ERK/STAT3 signaling pathway in breast cancer. Cell Cycle 2018, 17, 428–438. [Google Scholar] [CrossRef]
- Khabipov, A.; Kading, A.; Liedtke, K.R.; Freund, E.; Partecke, L.I.; Bekeschus, S. RAW 264.7 macrophage polarization by pancreatic cancer cells—A model for studying tumour-promoting macrophages. Anticancer Res. 2019, 39, 2871–2882. [Google Scholar] [CrossRef]
- Freund, E.; Moritz, J.; Stope, M.; Seebauer, C.; Schmidt, A.; Bekeschus, S. Plasma-derived reactive species shape a differentiation profile in human monocytes. Appl. Sci. 2019, 9, 2530. [Google Scholar] [CrossRef] [Green Version]
- Rodder, K.; Moritz, J.; Miller, V.; Weltmann, K.D.; Metelmann, H.R.; Gandhirajan, R.; Bekeschus, S. Activation of murine immune cells upon co-culture with plasma-treated B16F10 melanoma cells. Appl. Sci. 2019, 9, 660. [Google Scholar] [CrossRef] [Green Version]
- Bekeschus, S.; Scherwietes, L.; Freund, E.; Liedtke, K.R.; Hackbarth, C.; von Woedtke, T.; Partecke, L.I. Plasma-treated medium tunes the inflammatory profile in murine bone marrow-derived macrophages. Clin. Plasma Med. 2018, 11, 1–9. [Google Scholar] [CrossRef]
- Kaushik, N.K.; Kaushik, N.; Adhikari, M.; Ghimire, B.; Linh, N.N.; Mishra, Y.K.; Lee, S.J.; Choi, E.H. Preventing the solid cancer progression via release of anticancer-cytokines in co-culture with cold plasma-stimulated macrophages. Cancers 2019, 11, 842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaushik, N.K.; Kaushik, N.; Min, B.; Choi, K.H.; Hong, Y.J.; Miller, V.; Fridman, A.; Choi, E.H. Cytotoxic macrophage-released tumour necrosis factor-alpha (TNF-alpha) as a killing mechanism for cancer cell death after cold plasma activation. J. Phys. D Appl. Phys. 2016, 49. [Google Scholar] [CrossRef]
- Miller, V.; Lin, A.; Fridman, G.; Dobrynin, D.; Fridman, A. Plasma stimulation of migration of macrophages. Plasma Process. Polym. 2014, 11, 1193–1197. [Google Scholar] [CrossRef]
- Tran Janco, J.M.; Lamichhane, P.; Karyampudi, L.; Knutson, K.L. Tumor-infiltrating dendritic cells in cancer pathogenesis. J. Immunol. 2015, 194, 2985–2991. [Google Scholar] [CrossRef] [Green Version]
- Michielsen, A.J.; Hogan, A.E.; Marry, J.; Tosetto, M.; Cox, F.; Hyland, J.M.; Sheahan, K.D.; O’Donoghue, D.P.; Mulcahy, H.E.; Ryan, E.J.; et al. Tumour tissue microenvironment can inhibit dendritic cell maturation in colorectal cancer. PLoS ONE 2011, 6, e27944. [Google Scholar] [CrossRef] [Green Version]
- Van Loenhout, J.; Flieswasser, T.; Freire Boullosa, L.; de Waele, J.; Van Audenaerde, J.; Marcq, E.; Jacobs, J.; Lin, A.; Lion, E.; Dewitte, H.; et al. Cold atmospheric plasma-treated PBS eliminates immunosuppressive pancreatic stellate cells and induces immunogenic cell death of pancreatic cancer cells. Cancers 2019, 11, 1597. [Google Scholar] [CrossRef] [Green Version]
- Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [Green Version]
- Cools-Lartigue, J.; Spicer, J.; Najmeh, S.; Ferri, L. Neutrophil extracellular traps in cancer progression. Cell Mol. Life Sci. 2014, 71, 4179–4194. [Google Scholar] [CrossRef]
- Bekeschus, S.; Winterbourn, C.C.; Kolata, J.; Masur, K.; Hasse, S.; Broker, B.M.; Parker, H.A. Neutrophil extracellular trap formation is elicited in response to cold physical plasma. J. Leukoc. Biol. 2016, 100, 791–799. [Google Scholar] [CrossRef]
- Chen, Y.Q.; He, Y.F.; Wang, X.C.; Lu, F.; Gao, J.H. Adipose-derived mesenchymal stem cells exhibit tumor tropism and promote tumorsphere formation of breast cancer cells. Oncol. Rep. 2019, 41, 2126–2136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spaeth, E.; Klopp, A.; Dembinski, J.; Andreeff, M.; Marini, F. Inflammation and tumor microenvironments: Defining the migratory itinerary of mesenchymal stem cells. Gene Ther. 2008, 15, 730–738. [Google Scholar] [CrossRef] [PubMed]
- Mazini, L.; Rochette, L.; Amine, M.; Malka, G. Regenerative capacity of adipose derived stem cells (ADSCs), comparison with mesenchymal stem cells (MSCs). Int. J. Mol. Sci. 2019, 20, 2523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kang, S.U.; Kim, H.J.; Kim, D.H.; Han, C.H.; Lee, Y.S.; Kim, C.H. Nonthermal plasma treated solution inhibits adipocyte differentiation and lipogenesis in 3T3-L1 preadipocytes via ER stress signal suppression. Sci. Rep. 2018, 8. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Byrne, K.T.; Yan, F.; Yamazoe, T.; Chen, Z.; Baslan, T.; Richman, L.P.; Lin, J.H.; Sun, Y.H.; Rech, A.J.; et al. Tumor cell-intrinsic factors underlie heterogeneity of immune cell infiltration and response to immunotherapy. Immunity 2018, 49, 178–193 e177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walker, C.; Mojares, E.; Hernandez, A.D. Role of extracellular matrix in development and cancer progression. Int. J. Mol. Sci. 2018, 19, 3028. [Google Scholar] [CrossRef] [Green Version]
- Handorf, A.M.; Zhou, Y.; Halanski, M.A.; Li, W.J. Tissue stiffness dictates development, homeostasis, and disease progression. Organogenesis 2015, 11, 1–15. [Google Scholar] [CrossRef] [Green Version]
- Coulson, R.; Liew, S.H.; Connelly, A.A.; Yee, N.S.; Deb, S.; Kumar, B.; Vargas, A.C.; O’Toole, S.A.; Parslow, A.C.; Poh, A.; et al. The angiotensin receptor blocker, Losartan, inhibits mammary tumor development and progression to invasive carcinoma. Oncotarget 2017, 8, 18640–18656. [Google Scholar] [CrossRef] [Green Version]
- Diop-Frimpong, B.; Chauhan, V.P.; Krane, S.; Boucher, Y.; Jain, R.K. Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. PNAS 2011, 108, 2909–2914. [Google Scholar] [CrossRef] [Green Version]
- Cassinelli, G.; Lanzi, C.; Tortoreto, M.; Cominetti, D.; Petrangolini, G.; Favini, E.; Zaffaroni, N.; Pisano, C.; Penco, S.; Vlodavsky, I.; et al. Antitumor efficacy of the heparanase inhibitor SST0001 alone and in combination with antiangiogenic agents in the treatment of human pediatric sarcoma models. Biochem. Pharmacol. 2013, 85, 1424–1432. [Google Scholar] [CrossRef]
- Keyvani, A.; Atyabi, S.M.; Sardari, S.; Norouzian, D.; Madanchi. Effects of cold atmospheric plasma jet on collagen structure in different treatment times. Basic Res. J. Med. Clin. Sci. 2017, 6, 84–90. [Google Scholar]
- Dong, X.; Chen, M.; Wang, Y.; Yu, Q. A mechanistic study of plasma treatment effects on demineralized dentin surfaces for improved adhesive/dentin interface bonding. Clin. Plasma Med. 2014, 2, 11–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kang, S.U.; Kim, Y.S.; Kim, Y.E.; Park, J.K.; Lee, Y.S.; Kang, H.Y.; Jang, J.W.; Ryeo, J.B.; Lee, Y.; Shin, Y.S.; et al. Opposite effects of non-thermal plasma on cell migration and collagen production in keloid and normal fibroblasts. PLoS ONE 2017, 12, e0187978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sari, D.H.; Ningsih, S.S.; Antarianto, R.D.; Sadikin, M.; Hardiany, N.S.; Jusman, S.W.A. mRNA relative expression of cancer associated fibroblasts markers in keloid scars. Adv. Sci. Lett. 2017, 23, 6893–6895. [Google Scholar] [CrossRef]
- Eisenhauer, P.; Chernets, N.; Song, Y.; Dobrynin, D.; Pleshko, N.; Steinbeck, M.J.; Freeman, T.A. Chemical modification of extracellular matrix by cold atmospheric plasma-generated reactive species affects chondrogenesis and bone formation. J. Tissue Eng. Regen. Med. 2016, 10, 772–782. [Google Scholar] [CrossRef] [Green Version]
- Choi, J.H.; Lee, H.W.; Lee, J.K.; Hong, J.W.; Kim, G.C. Low-temperature atmospheric plasma increases the expression of anti-aging genes of skin cells without causing cellular damages. Arch. Dermatol. Res. 2013, 305, 133–140. [Google Scholar] [CrossRef]
- Metelmann, H.R.; Seebauer, C.; Miller, V.; Fridman, A.; Bauer, G.; Graves, D.B.; Pouvesle, J.M.; Rutkowski, R.; Schuster, M.; Bekeschus, S.; et al. Clinical experience with cold plasma in the treatment of locally advanced head and neck cancer. Clin. Plasma Med. 2018, 9, 6–13. [Google Scholar] [CrossRef]
- Metelmann, H.R.; Nedrelow, D.S.; Seebauer, C.; Schuster, M.; von Woedtke, T.; Weltmann, K.D.; Kindler, S.; Metelmann, P.H.; Finkelstein, S.E.; Von Hoff, D.D.; et al. Head and neck cancer treatment and physical plasma. Clin. Plasma Med. 2015, 3, 17–23. [Google Scholar] [CrossRef]
- Whatcott, C.J.; Diep, C.H.; Jiang, P.; Watanabe, A.; LoBello, J.; Sima, C.; Hostetter, G.; Shepard, H.M.; Von Hoff, D.D.; Han, H.Y. Desmoplasia in primary tumors and metastatic lesions of pancreatic cancer. Clin. Cancer Res. 2015, 21, 3561–3568. [Google Scholar] [CrossRef] [Green Version]
- Han, X.; Li, Y.; Xu, Y.; Zhao, X.; Zhang, Y.; Yang, X.; Wang, Y.; Zhao, R.; Anderson, G.J.; Zhao, Y.; et al. Reversal of pancreatic desmoplasia by re-educating stellate cells with a tumour microenvironment-activated nanosystem. Nat. Commun. 2018, 9, 3390. [Google Scholar] [CrossRef] [Green Version]
- Tammi, R.H.; Kultti, A.H.; Kosma, V.-M.; Pirinen, R.; Auvinen, P.; Tammi, M.I. Hyaluronan in human tumors: Importance of stromal and cancer cell-associated hyaluronan. In Hyaluronan in Cancer Biology; Stern, R., Ed.; Academic Press: San Diego, CA, USA, 2009; pp. 257–284. [Google Scholar] [CrossRef]
- McCarthy, J.B.; El-Ashry, D.; Turley, E.A. Hyaluronan, cancer-associated fibroblasts and the tumor microenvironment in malignant progression. Front. Cell Dev. Biol. 2018, 6. [Google Scholar] [CrossRef]
- Soltes, L.; Mendichi, R.; Kogan, G.; Schiller, J.; Stankovska, M.; Arnhold, J. Degradative action of reactive oxygen species on hyaluronan. Biomacromolecules 2006, 7, 659–668. [Google Scholar] [CrossRef] [PubMed]
- Degendorfer, G.; Chuang, C.Y.; Kawasaki, H.; Hammer, A.; Malle, E.; Yamakura, F.; Davies, M.J. Peroxynitrite-mediated oxidation of plasma fibronectin. Free Radic. Biol. Med. 2016, 97, 602–615. [Google Scholar] [CrossRef] [PubMed]
- Misra, S.; Hascall, V.C.; Markwald, R.R.; Ghatak, S. Interactions between hyaluronan and its receptors (CD44, RHAMM) regulate the activities of inflammation and cancer. Front. Immunol. 2015, 6, 201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wolf, K.; Friedl, P. Extracellular matrix determinants of proteolytic and non-proteolytic cell migration. Trends Cell Biol. 2011, 21, 736–744. [Google Scholar] [CrossRef] [PubMed]
- Choi, B.B.R.; Choi, J.H.; Ji, J.; Song, K.W.; Lee, H.J.; Kim, G.C. Increment of growth factors in mouse skin treated with non-thermal plasma. Int. J. Med. Sci. 2018, 15, 1203. [Google Scholar] [CrossRef]
- Morry, J.; Ngamcherdtrakul, W.; Yantasee, W. Oxidative stress in cancer and fibrosis: Opportunity for therapeutic intervention with antioxidant compounds, enzymes, and nanoparticles. Redox Biol. 2017, 11, 240–253. [Google Scholar] [CrossRef]
- Durbeej, M. Laminins. Cell Tissue Res. 2010, 339, 259–268. [Google Scholar] [CrossRef]
- Shao, P.-L.; Liao, J.-D.; Wong, T.-W.; Wang, Y.-C.; Leu, S.; Yip, H.-K. Enhancement of wound healing by non-thermal N2/Ar micro-plasma exposure in mice with fractional-CO2-laser-induced wounds. PLoS ONE 2016, 11, e0156699. [Google Scholar] [CrossRef]
- Sahebalzamani, M.A.; Khorasani, M.T.; Joupari, M.D. Enhancement of fibroblasts outgrowth onto polycaprolactone nanofibrous grafted by laminin protein using carbon dioxide plasma treatment. Nano Biomed. Eng. 2017, 9, 191–198. [Google Scholar] [CrossRef] [Green Version]
- Degendorfer, G.; Chuang, C.Y.; Hammer, A.; Malle, E.; Davies, M.J. Peroxynitrous acid induces structural and functional modifications to basement membranes and its key component, laminin. Free Radic. Biol. Med. 2015, 89, 721–733. [Google Scholar] [CrossRef] [PubMed]
- Nybo, T.; Dieterich, S.; Gamon, L.F.; Chuang, C.Y.; Hammer, A.; Hoefler, G.; Malle, E.; Rogowska-Wrzesinska, A.; Davies, M.J. Chlorination and oxidation of the extracellular matrix protein laminin and basement membrane extracts by hypochlorous acid and myeloperoxidase. Redox Biol. 2019, 20, 496–513. [Google Scholar] [CrossRef] [PubMed]
- Lorentzen, L.G.; Chuang, C.Y.; Rogowska-Wrzesinska, A.; Davies, M.J. Identification and quantification of sites of nitration and oxidation in the key matrix protein laminin and the structural consequences of these modifications. Redox Biol. 2019, 24, 101226. [Google Scholar] [CrossRef] [PubMed]
- Vanichkitrungruang, S.; Chuang, C.Y.; Hawkins, C.L.; Hammer, A.; Hoefler, G.; Malle, E.; Davies, M.J. Oxidation of human plasma fibronectin by inflammatory oxidants perturbs endothelial cell function. Free Radic. Biol. Med. 2019, 136, 118–134. [Google Scholar] [CrossRef] [PubMed]
- Nybo, T.; Cai, H.; Chuang, C.Y.; Gamon, L.F.; Rogowska-Wrzesinska, A.; Davies, M.J. Chlorination and oxidation of human plasma fibronectin by myeloperoxidase-derived oxidants, and its consequences for smooth muscle cell function. Redox Biol. 2018, 19, 388–400. [Google Scholar] [CrossRef] [PubMed]
- Malandrino, A.; Mak, M.; Kamm, R.D.; Moeendarbary, E. Complex mechanics of the heterogeneous extracellular matrix in cancer. Extreme Mech. Lett. 2018, 21, 25–34. [Google Scholar] [CrossRef]
- Myllyharju, J.; Schipani, E. Extracellular matrix genes as hypoxia-inducible targets. Cell Tissue Res. 2010, 339, 19–29. [Google Scholar] [CrossRef] [Green Version]
- Degendorfer, G.; Chuang, C.Y.; Mariotti, M.; Hammer, A.; Hoefler, G.; Hägglund, P.; Malle, E.; Wise, S.G.; Davies, M.J. Exposure of tropoelastin to peroxynitrous acid gives high yields of nitrated tyrosine residues, di-tyrosine cross-links and altered protein structure and function. Free Radic. Biol. Med. 2018, 115, 219–231. [Google Scholar] [CrossRef] [Green Version]
- Qin, J.; Vinogradova, O.; Plow, E.F. Integrin bidirectional signaling: A molecular view. PLoS Biol. 2004, 2, e169. [Google Scholar] [CrossRef] [Green Version]
- Volotskova, O.; Stepp, M.A.; Keidar, M. Integrin activation by a cold atmospheric plasma jet. New J. Phys. 2012, 14, 053019. [Google Scholar] [CrossRef]
- Haertel, B.; Strassenburg, S.; Oehmigen, K.; Wende, K.; von Woedtke, T.; Lindequist, U. Differential influence of components resulting from atmospheric-pressure plasma on integrin expression of human HaCaT keratinocytes. Biomed. Res. Int. 2013, 2013, 761451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haertel, B.; Wende, K.; von Woedtke, T.; Weltmann, K.D.; Lindequist, U. Non-thermal atmospheric-pressure plasma can influence cell adhesion molecules on HaCaT-keratinocytes. Exp. Derm. 2011, 20, 282–284. [Google Scholar] [CrossRef] [PubMed]
- Shashurin, A.; Stepp, M.A.; Hawley, T.S.; Pal-Ghosh, S.; Brieda, L.; Bronnikov, S.; Jurjus, R.A.; Keidar, M. Influence of cold plasma atmospheric jet on surface integrin expression of living cells. Plasma Process. Polym. 2010, 7, 294–300. [Google Scholar] [CrossRef]
- Schmidt, A.; Bekeschus, S.; Wende, K.; Vollmar, B.; von Woedtke, T. A cold plasma jet accelerates wound healing in a murine model of full-thickness skin wounds. Exp. Dermatol. 2017, 26, 156–162. [Google Scholar] [CrossRef]
- Basuroy, S.; Dunagan, M.; Sheth, P.; Seth, A.; Rao, R.K. Hydrogen peroxide activates focal adhesion kinase and c-Src by a phosphatidylinositol 3 kinase-dependent mechanism and promotes cell migration in Caco-2 cell monolayers. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 299, G186–G195. [Google Scholar] [CrossRef] [Green Version]
- Wu, C.; Dedhar, S. Integrin-linked kinase (ILK) and its interactors: A new paradigm for the coupling of extracellular matrix to actin cytoskeleton and signaling complexes. J. Cell Biol. 2001, 155, 505–510. [Google Scholar] [CrossRef]
- Im, M.; Dagnino, L. Protective role of integrin-linked kinase against oxidative stress and in maintenance of genomic integrity. Oncotarget 2018, 9, 13637–13651. [Google Scholar] [CrossRef] [Green Version]
- Zhang, C.; Wang, H.J.; Bao, Q.C.; Wang, L.; Guo, T.K.; Chen, W.L.; Xu, L.L.; Zhou, H.S.; Bian, J.L.; Yang, Y.R.; et al. NRF2 promotes breast cancer cell proliferation and metastasis by increasing RhoA/ROCK pathway signal transduction. Oncotarget 2016, 7, 73593–73606. [Google Scholar] [CrossRef]
- Walko, G.; Castanon, M.J.; Wiche, G. Molecular architecture and function of the hemidesmosome. Cell Tissue Res. 2015, 360, 529–544. [Google Scholar] [CrossRef] [Green Version]
- Laval, S.; Laklai, H.; Fanjul, M.; Pucelle, M.; Laurell, H.; Billon-Gales, A.; Le Guellec, S.; Delisle, M.B.; Sonnenberg, A.; Susini, C.; et al. Dual roles of hemidesmosomal proteins in the pancreatic epithelium: The phosphoinositide 3-kinase decides. Oncogene 2014, 33, 1934–1944. [Google Scholar] [CrossRef]
- Haralambiev, L.; Wien, L.; Gelbrich, N.; Kramer, A.; Mustea, A.; Burchardt, M.; Ekkernkamp, A.; Stope, M.B.; Gumbel, D. Effects of cold atmospheric plasma on the expression of chemokines, growth factors, TNF superfamily members, interleukins, and cytokines in human osteosarcoma cells. Anticancer Res. 2019, 39, 151–157. [Google Scholar] [CrossRef] [PubMed]
- Bekeschus, S.; Wulf, C.P.; Freund, E.; Koensgen, D.; Mustea, A.; Weltmann, K.-D.; Stope, M.B. Plasma treatment of ovarian cancer cells mitigates their immuno-modulatory products active on THP-1 monocytes. Plasma 2018, 1, 18. [Google Scholar] [CrossRef] [Green Version]
- Krutovskikh, V.A.; Piccoli, C.; Yamasaki, H. Gap junction intercellular communication propagates cell death in cancerous cells. Oncogene 2002, 21, 1989–1999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, R.G.; Chen, Z.T.; Keidar, M.; Leng, Y.S. The impact of radicals in cold atmospheric plasma on the structural modification of gap junction: A reactive molecular dynamics study. Int. J. Smart Nano Mater. 2019, 10, 144–155. [Google Scholar] [CrossRef]
- Pouget, J.P.; Georgakilas, A.G.; Ravanat, J.L. Targeted and off-target (bystander and abscopal) effects of radiation therapy: Redox mechanisms and risk/benefit analysis. Antioxid. Redox Signal. 2018, 29, 1447–1487. [Google Scholar] [CrossRef]
- Lin, A.; Gorbanev, Y.; Cos, P.; Smits, E.; Bogaerts, A. Plasma elicits immunogenic death in melanoma cells. Clin. Plasma Med. 2018, 9, 9. [Google Scholar] [CrossRef]
- Lin, A.G.; Xiang, B.; Merlino, D.J.; Baybutt, T.R.; Sahu, J.; Fridman, A.; Snook, A.E.; Miller, V. Non-thermal plasma induces immunogenic cell death in vivo in murine CT26 colorectal tumors. OncoImmunology 2018, 7, e1484978. [Google Scholar] [CrossRef] [Green Version]
- Lin, A.; Gorbanev, Y.; De Backer, J.; Van Loenhout, J.; Van Boxem, W.; Lemiere, F.; Cos, P.; Dewilde, S.; Smits, E.; Bogaerts, A. Non-thermal plasma as a unique delivery system of short-lived reactive oxygen and nitrogen species for immunogenic cell death in melanoma cells. Adv. Sci. 2019, 6. [Google Scholar] [CrossRef] [Green Version]
- Jabalee, J.; Towle, R.; Garnis, C. The role of extracellular vesicles in cancer: Cargo, function, and therapeutic implications. Cells 2018, 7, 93. [Google Scholar] [CrossRef] [Green Version]
- Sasaki, S.; Kanzaki, M.; Kaneko, T. Calcium influx through TRP channels induced by short-lived reactive species in plasma-irradiated solution. Sci. Rep. 2016, 6. [Google Scholar] [CrossRef] [Green Version]
- Schneider, C.; Gebhardt, L.; Arndt, S.; Karrer, S.; Zimmermann, J.L.; Fischer, M.J.M.; Bosserhoff, A.K. Cold atmospheric plasma causes a calcium influx in melanoma cells triggering CAP-induced senescence. Sci. Rep. 2018, 8. [Google Scholar] [CrossRef] [PubMed]
- Akbarali, H.I. Oxidative Stress and Ion Channels. In Systems Biology of Free Radicals and Antioxidants; Laher, I., Ed.; Springer: Berlin/Heidelberg, Germany, 2014; pp. 355–373. [Google Scholar] [CrossRef]
- Xie, Z.; Barski, O.A.; Cai, J.; Bhatnagar, A.; Tipparaju, S.M. Catalytic reduction of carbonyl groups in oxidized PAPC by Kvbeta2 (AKR6). Chem. Biol. Interact. 2011, 191, 255–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Halestrap, A.P. Calcium, mitochondria and reperfusion injury: A pore way to die. Biochem. Soc. Trans. 2006, 34, 232–237. [Google Scholar] [CrossRef] [PubMed]
- Yamamoto, S.; Takahashi, N.; Mori, Y. Chemical physiology of oxidative stress-activated TRPM2 and TRPC5 channels. Prog. Biophys. Mol. Biol. 2010, 103, 18–27. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.J.; Men, R.T.; Wen, M.Y.; Hu, X.L.; Liu, X.J.; Yang, L. Blockage of TRPM7 channel induces hepatic stellate cell death through endoplasmic reticulum stress-mediated apoptosis. Life Sci. 2014, 94, 37–44. [Google Scholar] [CrossRef] [PubMed]
- Jiang, J.X.; Penuela, S. Connexin and pannexin channels in cancer. BMC Cell Biol. 2016, 17 (Suppl. 1), 12. [Google Scholar] [CrossRef] [Green Version]
- Boyd-Tressler, A.; Penuela, S.; Laird, D.W.; Dubyak, G.R. Chemotherapeutic drugs induce ATP release via caspase-gated pannexin-1 channels and a caspase/pannexin-1-independent mechanism. J. Biol. Chem. 2014, 289, 27246–27263. [Google Scholar] [CrossRef] [Green Version]
- Freeman, T.J.; Sayedyahossein, S.; Johnston, D.; Sanchez-Pupo, R.E.; O’Donnell, B.; Huang, K.; Lakhani, Z.; Nouri-Nejad, D.; Barr, K.J.; Harland, L.; et al. Inhibition of pannexin 1 reduces the tumorigenic properties of human melanoma cells. Cancers 2019, 11, 102. [Google Scholar] [CrossRef] [Green Version]
- Poornima, V.; Vallabhaneni, S.; Mukhopadhyay, M.; Bera, A.K. Nitric oxide inhibits the pannexin 1 channel through a cGMP-PKG dependent pathway. Nitric Oxide 2015, 47, 77–84. [Google Scholar] [CrossRef]
- Patel, G.K.; Zubair, H.; Khan, M.A.; Srivastava, S.K.; Ahmad, A.; Patton, M.C.; Singh, S.; Khushman, M.d.; Singh, A.P. Exosomes: Key supporters of tumor metastasis. In Diagnostic and Therapeutic Applications of Exosomes in Cancer; Amiji, M., Ramesh, R., Eds.; Academic Press: Cambridge, MA, USA, 2018; pp. 261–283. [Google Scholar] [CrossRef]
- Burrello, J.; Monticone, S.; Gai, C.; Gomez, Y.; Kholia, S.; Camussi, G. Stem cell-derived extracellular vesicles and immune-modulation. Front. Cell Dev. Biol. 2016, 4, 83. [Google Scholar] [CrossRef] [Green Version]
- Bekeschus, S.; Moritz, J.; Schmidt, A.; Wende, K. Redox regulation of leukocyte-derived microparticle release and protein content in response to cold physical plasma-derived oxidants. Clin. Plasma Med. 2017, 7–8, 24–35. [Google Scholar] [CrossRef]
- Partecke, L.I.; Evert, K.; Haugk, J.; Doering, F.; Normann, L.; Diedrich, S.; Weiss, F.U.; Evert, M.; Huebner, N.O.; Guenther, C.; et al. Tissue tolerable plasma (TTP) induces apoptosis in pancreatic cancer cells in vitro and in vivo. BMC Cancer 2012, 12, 473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stoffels, E.; Roks, A.J.M.; Deelmm, L.E. Delayed effects of cold atmospheric plasma on vascular cells. Plasma Process. Polym. 2008, 5, 599–605. [Google Scholar] [CrossRef]
- Bodega, G.; Alique, M.; Puebla, L.; Carracedo, J.; Ramirez, R.M. Microvesicles: ROS scavengers and ROS producers. J. Extracell. Vesicles 2019, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stillwell, W. Membrane Transport. In An Introduction to Biological Membranes, 2nd ed.; Stillwell, W., Ed.; Elsevier: Amsterdam, The Netherlands, 2016; pp. 423–451. [Google Scholar] [CrossRef]
- Prise, K.M.; O’Sullivan, J.M. Radiation-induced bystander signalling in cancer therapy. Nat. Rev. Cancer 2009, 9, 351–360. [Google Scholar] [CrossRef] [PubMed]
- Aasen, T.; Leithe, E.; Graham, S.V.; Kameritsch, P.; Mayan, M.D.; Mesnil, M.; Pogoda, K.; Tabernero, A. Connexins in cancer: Bridging the gap to the clinic. Oncogene 2019, 38, 4429–4451. [Google Scholar] [CrossRef] [Green Version]
- Choi, J.H.; Nam, S.H.; Song, Y.S.; Lee, H.W.; Lee, H.J.; Song, K.; Hong, J.W.; Kim, G.C. Treatment with low-temperature atmospheric pressure plasma enhances cutaneous delivery of epidermal growth factor by regulating E-cadherin-mediated cell junctions. Arch. Dermatol. Res. 2014, 306, 635–643. [Google Scholar] [CrossRef]
- Retamal, M.A. Connexin and pannexin hemichannels are regulated by redox potential. Front. Physiol. 2014, 5, 80. [Google Scholar] [CrossRef] [Green Version]
- Sahu, P.; Jena, S.R.; Samanta, L. Tunneling nanotubes: A versatile target for cancer therapy. Curr. Cancer Drug Targets 2018, 18, 514–521. [Google Scholar] [CrossRef]
- Zhang, L.; Zhang, Y. Tunneling nanotubes between rat primary astrocytes and C6 glioma cells alter proliferation potential of glioma cells. Neurosci. Bull. 2015, 31, 371–378. [Google Scholar] [CrossRef] [Green Version]
- Arkwright, P.D.; Luchetti, F.; Tour, J.; Roberts, C.; Ayub, R.; Morales, A.P.; Rodriguez, J.J.; Gilmore, A.; Canonico, B.; Papa, S.; et al. Fas stimulation of T lymphocytes promotes rapid intercellular exchange of death signals via membrane nanotubes. Cell Res. 2010, 20, 72–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.; Gerdes, H.H. Transfer of mitochondria via tunneling nanotubes rescues apoptotic PC12 cells. Cell Death Differ. 2015, 22, 1181–1191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ranzinger, J.; Rustom, A.; Heide, D.; Morath, C.; Schemmer, P.; Nawroth, P.P.; Zeier, M.; Schwenger, V. The receptor for advanced glycation end-products (RAGE) plays a key role in the formation of nanotubes (NTs) between peritoneal mesothelial cells and in murine kidneys. Cell Tissue Res. 2014, 357, 667–679. [Google Scholar] [CrossRef] [PubMed]
- Zihni, C.; Mills, C.; Matter, K.; Balda, M.S. Tight junctions: From simple barriers to multifunctional molecular gates. Nat. Rev. Mol. Cell. Biol. 2016, 17, 564–580. [Google Scholar] [CrossRef]
- Hoentsch, M.; von Woedtke, T.; Weltmann, K.D.; Nebe, J.B. Time-dependent effects of low-temperature atmospheric-pressure argon plasma on epithelial cell attachment, viability and tight junction formation in vitro. J. Phys. D Appl. Phys. 2012, 45. [Google Scholar] [CrossRef]
- Rao, R. Oxidative stress-induced disruption of epithelial and endothelial tight junctions. Front. Biosci. 2008, 13, 7210–7226. [Google Scholar] [CrossRef] [Green Version]
- Intercellular Junctions. Cell Biology, 3rd ed.; Pollard, T.D., Earnshaw, W.C., Lippincott-Schwartz, J., Johnson, G.T., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 543–553. [Google Scholar] [CrossRef]
- Schmidt, A.; von Woedtke, T.; Bekeschus, S. Periodic exposure of keratinocytes to cold physical plasma: An in vitro model for redox-related diseases of the skin. Oxid. Med. Cell. Longev. 2016, 2016, 9816072. [Google Scholar] [CrossRef] [Green Version]
- Overgaard, C.E.; Daugherty, B.L.; Mitchell, L.A.; Koval, M. Claudins: Control of Barrier Function and Regulation in Response to Oxidant Stress. Antioxid. Redox Signal. 2011, 15, 1179–1193. [Google Scholar] [CrossRef] [Green Version]
- Chen-Quay, S.C.; Eiting, K.T.; Li, A.W.A.; Lamharzi, N.; Quay, S.C. Identification of tight junction modulating lipids. J. Pharm. Sci. 2009, 98, 606–619. [Google Scholar] [CrossRef]
- Oshima, T.; Sasaki, M.; Kataoka, H.; Miwa, H.; Takeuchi, T.; Joh, T. Wip1 protects hydrogen peroxide-induced colonic epithelial barrier dysfunction. Cell. Mol. Life Sci. 2007, 64, 3139–3147. [Google Scholar] [CrossRef]
- Cummins, P.M. Occludin: One protein, many forms. Mol. Cell. Biol. 2012, 32, 242–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lochhead, J.J.; McCaffrey, G.; Quigley, C.E.; Finch, J.; DeMarco, K.M.; Nametz, N.; Davis, T.P. Oxidative stress increases blood-brain barrier permeability and induces alterations in occludin during hypoxia-reoxygenation. J. Cereb. Blood F Metab. 2010, 30, 1625–1636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Blasig, I.E.; Bellmann, C.; Cording, J.; del Vecchio, G.; Zwanziger, D.; Huber, O.; Haseloff, R.F. Occludin protein family: Oxidative stress and reducing conditions. Antioxid. Redox Signal. 2011, 15, 1195–1219. [Google Scholar] [CrossRef] [PubMed]
- Lischper, M.; Beuck, S.; Thanabalasundaram, G.; Pieper, C.; Galla, H.J. Metalloproteinase mediated occludin cleavage in the cerebral microcapillary endothelium under pathological conditions. Brain Res. 2010, 1326, 114–127. [Google Scholar] [CrossRef] [PubMed]
- Knights, A.J.; Funnell, A.P.; Crossley, M.; Pearson, R.C. Holding tight: Cell junctions and cancer spread. Trends Cancer Res. 2012, 8, 61–69. [Google Scholar] [PubMed]
- Lee, H.Y.; Choi, J.H.; Hong, J.W.; Kim, G.C.; Lee, H.J. Comparative study of the Ar and He atmospheric pressure plasmas on E-cadherin protein regulation for plasma-mediated transdermal drug delivery. J. Phys. D Appl. Phys. 2018, 51. [Google Scholar] [CrossRef]
- Hung, Y.W.; Lee, L.T.; Peng, Y.C.; Chang, C.T.; Wong, Y.K.; Tung, K.C. Effect of a nonthermal-atmospheric pressure plasma jet on wound healing: An animal study. J. Chin. Med. Assoc. 2016, 79, 320–328. [Google Scholar] [CrossRef] [Green Version]
- Schmelz, M. Selective disruption of cadherin/catenin complexes by oxidative stress in precision-cut mouse liver slices. Toxicol. Sci. 2001, 61, 389–394. [Google Scholar] [CrossRef]
- Haidari, M.; Zhang, W.; Wakame, K. Disruption of endothelial adherens junction by invasive breast cancer cells is mediated by reactive oxygen species and is attenuated by AHCC. Life Sci. 2013, 93, 994–1003. [Google Scholar] [CrossRef]
- Goitre, L.; Pergolizzi, B.; Ferro, E.; Trabalzini, L.; Retta, S.F. Molecular crosstalk between integrins and cadherins: Do reactive oxygen species set the talk? J. Signal Transduct. 2012, 2012, 807682. [Google Scholar] [CrossRef]
- Jiang, J.; Wang, K.; Chen, Y.; Chen, H.; Nice, E.C.; Huang, C. Redox regulation in tumor cell epithelial-mesenchymal transition: Molecular basis and therapeutic strategy. Signal Transduct. Target. Ther. 2017, 2, 17036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Delva, E.; Tucker, D.K.; Kowalczyk, A.P. The desmosome. Cold Spring Harb. Perspect. Biol. 2009, 1, a002543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Neuber, S.; Jager, S.; Meyer, M.; Wischmann, V.; Koch, P.J.; Moll, R.; Schmidt, A. c-Src mediated tyrosine phosphorylation of plakophilin 3 as a new mechanism to control desmosome composition in cells exposed to oxidative stress. Cell Tissue Res. 2015, 359, 799–816. [Google Scholar] [CrossRef] [PubMed]
- Zhou, G.; Yang, L.; Gray, A.; Srivastava, A.K.; Li, C.; Zhang, G.; Cui, T. The role of desmosomes in carcinogenesis. Onco Targets Ther. 2017, 10, 4059–4063. [Google Scholar] [CrossRef] [Green Version]
- Sisakhtnezhad, S.; Khosravi, L. Emerging physiological and pathological implications of tunneling nanotubes formation between cells. Eur. J. Cell Biol. 2015, 94, 429–443. [Google Scholar] [CrossRef] [PubMed]
- Rustom, A. The missing link: Does tunnelling nanotube-based supercellularity provide a new understanding of chronic and lifestyle diseases? Open Biol. 2016, 6, 160057. [Google Scholar] [CrossRef] [Green Version]
- Semenza, G.L. Hypoxia-inducible factors: Mediators of cancer progression and targets for cancer therapy. Trends Pharmacol. Sci. 2012, 33, 207–214. [Google Scholar] [CrossRef] [Green Version]
- Gilkes, D.M.; Semenza, G.L.; Wirtz, D. Hypoxia and the extracellular matrix: Drivers of tumour metastasis. Nat. Rev. Cancer 2014, 14, 430. [Google Scholar] [CrossRef] [Green Version]
- Malik, R.; Lelkes, P.I.; Cukierman, E. Biomechanical and biochemical remodeling of stromal extracellular matrix in cancer. Trends Biotechnol. 2015, 33, 230–236. [Google Scholar] [CrossRef] [Green Version]
- Erler, J.T.; Bennewith, K.L.; Nicolau, M.; Dornhöfer, N.; Kong, C.; Le, Q.-T.; Chi, J.-T.A.; Jeffrey, S.S.; Giaccia, A.J. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature 2006, 440, 1222. [Google Scholar] [CrossRef]
- Keith, B.; Johnson, R.S.; Simon, M.C. HIF1α and HIF2α: Sibling rivalry in hypoxic tumour growth and progression. Nat. Rev. Cancer 2012, 12, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barker, H.E.; Cox, T.R.; Erler, J.T. The rationale for targeting the LOX family in cancer. Nat. Rev. Cancer 2012, 12, 540. [Google Scholar] [CrossRef] [PubMed]
- Bekeschus, S.; Schmidt, A.; Weltmann, K.-D.; von Woedtke, T. The plasma jet kINPen–A powerful tool for wound healing. Clin. Plasma Med. 2016, 4, 19–28. [Google Scholar] [CrossRef]
- Chatraie, M.; Torkaman, G.; Khani, M.; Salehi, H.; Shokri, B. In vivo study of non-invasive effects of non-thermal plasma in pressure ulcer treatment. Sci. Rep. 2018, 8, 5621. [Google Scholar] [CrossRef] [PubMed]
- Lugano, R.; Vemuri, K.; Yu, D.; Bergqvist, M.; Smits, A.; Essand, M.; Johansson, S.; Dejana, E.; Dimberg, A. CD93 promotes β1 integrin activation and fibronectin fibrillogenesis during tumor angiogenesis. J. Clin. Investig. 2018, 128. [Google Scholar] [CrossRef] [Green Version]
- Han, Z.; Lu, Z.-R. Targeting fibronectin for cancer imaging and therapy. J. Mater. Chem. B 2017, 5, 639–654. [Google Scholar] [CrossRef] [Green Version]
- Sethi, T.; Rintoul, R.C.; Moore, S.M.; MacKinnon, A.C.; Salter, D.; Choo, C.; Chilvers, E.R.; Dransfield, I.; Donnelly, S.C.; Strieter, R. Extracellular matrix proteins protect small cell lung cancer cells against apoptosis: A mechanism for small cell lung cancer growth and drug resistance in vivo. Nat. Med. 1999, 5, 662. [Google Scholar] [CrossRef]
- Pontiggia, O.; Sampayo, R.; Raffo, D.; Motter, A.; Xu, R.; Bissell, M.J.; de Kier Joffé, E.B.; Simian, M. The tumor microenvironment modulates tamoxifen resistance in breast cancer: A role for soluble stromal factors and fibronectin through β1 integrin. Breast Cancer Res. Treat. 2012, 133, 459–471. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Z.; Qutaish, M.; Han, Z.; Schur, R.M.; Liu, Y.; Wilson, D.L.; Lu, Z.-R. MRI detection of breast cancer micrometastases with a fibronectin-targeting contrast agent. Nat. Commun. 2015, 6, 7984. [Google Scholar] [CrossRef] [Green Version]
- Jiang, K.; Song, X.; Yang, L.; Li, L.; Wan, Z.; Sun, X.; Gong, T.; Lin, Q.; Zhang, Z. Enhanced antitumor and anti-metastasis efficacy against aggressive breast cancer with a fibronectin-targeting liposomal doxorubicin. J. Control. Release 2018, 271, 21–30. [Google Scholar] [CrossRef]
- Sun, Y.; Kim, H.S.; Saw, P.E.; Jon, S.; Moon, W.K. Targeted therapy for breast cancer stem cells by liposomal delivery of siRNA against fibronectin EDB. Adv. Healthc. Mater. 2015, 4, 1675–1680. [Google Scholar] [CrossRef] [PubMed]
- Sponziello, M.; Rosignolo, F.; Celano, M.; Maggisano, V.; Pecce, V.; De Rose, R.F.; Lombardo, G.E.; Durante, C.; Filetti, S.; Damante, G. Fibronectin-1 expression is increased in aggressive thyroid cancer and favors the migration and invasion of cancer cells. Mol. Cell. Endocrinol. 2016, 431, 123–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yusupov, M.; Lackmann, J.W.; Razzokov, J.; Kumar, S.; Stapelmann, K.; Bogaerts, A. Impact of plasma oxidation on structural features of human epidermal growth factor. Plasma Process. Polym. 2018, 15, 1800022. [Google Scholar] [CrossRef]
- Razzokov, J.; Yusupov, M.; Bogaerts, A. Oxidation destabilizes toxic amyloid beta peptide aggregation. Sci. Rep. 2019, 9, 5476. [Google Scholar] [CrossRef] [PubMed]
- De Backer, J.; Razzokov, J.; Hammerschmid, D.; Mensch, C.; Hafideddine, Z.; Kumar, N.; van Raemdonck, G.; Yusupov, M.; Van Doorslaer, S.; Johannessen, C. The effect of reactive oxygen and nitrogen species on the structure of cytoglobin: A potential tumor suppressor. Redox Biol. 2018, 19, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Yusupov, M.; Razzokov, J.; Cordeiro, R.M.; Bogaerts, A. Transport of reactive oxygen and nitrogen species across aquaporin: A molecular level picture. Oxid. Med. Cell. Longev. 2019, 2019, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brucher, B.L.; Jamall, I.S. Cell-cell communication in the tumor microenvironment, carcinogenesis, and anticancer treatment. Cell. Physiol. Biochem. 2014, 34, 213–243. [Google Scholar] [CrossRef]
- Langhans, S.A. Three-dimensional in vitro cell culture models in drug discovery and drug repositioning. Front. Pharmacol. 2018, 9, 6. [Google Scholar] [CrossRef]
- Nath, S.; Devi, G.R. Three-dimensional culture systems in cancer research: Focus on tumor spheroid model. Pharmacol. Ther. 2016, 163, 94–108. [Google Scholar] [CrossRef] [Green Version]
- Gupta, N.; Liu, J.R.; Patel, B.; Solomon, D.E.; Vaidya, B.; Gupta, V. Microfluidics-based 3D cell culture models: Utility in novel drug discovery and delivery research. Bioeng. Transl. Med. 2016, 1, 63–81. [Google Scholar] [CrossRef]
- Huang, Z.; Tunnacliffe, A. Response of human cells to desiccation: Comparison with hyperosmotic stress response. J. Physiol. 2004, 558, 181–191. [Google Scholar] [CrossRef] [PubMed]
- Koppenol, W.H.; Bounds, P.L.; Dang, C.V. Otto Warburg’s contributions to current concepts of cancer metabolism. Nat. Rev. Cancer 2011, 11, 325–337. [Google Scholar] [CrossRef] [PubMed]
- Judee, F.; Fongia, C.; Ducommun, B.; Yousfi, M.; Lobjois, V.; Merbahi, N. Short and long time effects of low temperature plasma activated media on 3D multicellular tumor spheroids. Sci. Rep. 2016, 6, 21421. [Google Scholar] [CrossRef]
- Plewa, J.-M.; Yousfi, M.; Frongia, C.; Eichwald, O.; Ducommun, B.; Merbahi, N.; Lobjois, V. Low-temperature plasma-induced antiproliferative effects on multi-cellular tumor spheroids. New J. Phys. 2014, 16, 043027. [Google Scholar] [CrossRef]
- Chauvin, J.; Judee, F.; Merbahi, N.; Vicendo, P. Effects of plasma activated medium on head and neck FaDu cancerous cells: Comparison of 3D and 2D response. Anticancer Agents Med. Chem. 2018, 18, 776–783. [Google Scholar] [CrossRef]
- Chauvin, J.; Gibot, L.; Griseti, E.; Golzio, M.; Rols, M.P.; Merbahi, N.; Vicendo, P. Elucidation of in vitro cellular steps induced by antitumor treatment with plasma-activated medium. Sci. Rep. 2019, 9. [Google Scholar] [CrossRef]
- Bekeschus, S.; Freund, E.; Spadola, C.; Privat-Maldonado, A.; Hackbarth, C.; Bogaerts, A.; Schmidt, A.; Wende, K.; Weltmann, K.D.; von Woedtke, T.; et al. Risk assessment of kINPen plasma treatment of four human pancreatic cancer cell lines with respect to metastasis. Cancers 2019, 11, 1237. [Google Scholar] [CrossRef] [Green Version]
- Sagwal, S.K.; Pasqual-Melo, G.; Bodnar, Y.; Gandhirajan, R.K.; Bekeschus, S. Combination of chemotherapy and physical plasma elicits melanoma cell death via upregulation of SLC22A16. Cell Death Dis. 2018, 9. [Google Scholar] [CrossRef]
- Xu, H.; Lyu, X.; Yi, M.; Zhao, W.; Song, Y.; Wu, K. Organoid technology and applications in cancer research. J. Hematol. Oncol. 2018, 11, 116. [Google Scholar] [CrossRef]
- Yuan, H.; Xing, K.; Hsu, H.Y. Trinity of three-dimensional (3D) scaffold, vibration, and 3d printing on cell culture application: A systematic review and indicating future direction. Bioengineering 2018, 5, 57. [Google Scholar] [CrossRef] [Green Version]
- Labay, C.; Hamouda, I.; Tampieri, F.; Ginebra, M.P.; Canal, C. Production of reactive species in alginate hydrogels for cold atmospheric plasma-based therapies. Sci. Rep. 2019, 9, 16160. [Google Scholar] [CrossRef] [PubMed]
- Hoshiba, T. Decellularized extracellular matrix for cancer research. Materials 2019, 12, 1311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Przekora, A. Current Trends in Fabrication of Biomaterials for Bone and Cartilage Regeneration: Materials Modifications and Biophysical Stimulations. Int. J. Mol. Sci. 2019, 20, 435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhushan, B.; Kumar, R. Plasma treated and untreated thermoplastic biopolymers/biocomposites in tissue engineering and biodegradable implants. In Materials for Biomedical Engineering: Hydrogels and Polymer-Based Scaffolds; Holban, A.-M., Grumezescu, A., Eds.; Elsevier: Amsterdam, The Netherlands, 2019. [Google Scholar]
- Sontheimer-Phelps, A.; Hassell, B.A.; Ingber, D.E. Modelling cancer in microfluidic human organs-on-chips. Nat. Rev. Cancer 2019, 19, 65–81. [Google Scholar] [CrossRef]
- Tsai, H.F.; Trubelja, A.; Shen, A.Q.; Bao, G. Tumour-on-a-chip: Microfluidic models of tumour morphology, growth and microenvironment. J. R. Soc. Interface 2017, 14. [Google Scholar] [CrossRef] [Green Version]
- Langer, E.M.; Allen-Petersen, B.L.; King, S.M.; Kendsersky, N.D.; Turnidge, M.A.; Kuziel, G.M.; Riggers, R.; Samatham, R.; Amery, T.S.; Jacques, S.L.; et al. Modeling tumor phenotypes in vitro with three-dimensional bioprinting. Cell Rep. 2019, 26, 608–623 e606. [Google Scholar] [CrossRef] [Green Version]
- Albritton, J.L.; Miller, J.S. 3D bioprinting: Improving in vitro models of metastasis with heterogeneous tumor microenvironments. Dis. Models Mech. 2017, 10, 3–14. [Google Scholar] [CrossRef] [Green Version]
- Ahangar, P.; Cooke, M.E.; Weber, M.H.; Rosenzweig, D.H. Current biomedical applications of 3D printing and additive manufacturing. Appl. Sci. 2019, 9, 1713. [Google Scholar] [CrossRef] [Green Version]
- Adimora, N.J.; Jones, D.P.; Kemp, M.L. A model of redox kinetics implicates the thiol proteome in cellular hydrogen peroxide responses. Antioxid. Redox Signal. 2010, 13, 731–743. [Google Scholar] [CrossRef] [Green Version]
- Benfeitas, R.; Selvaggio, G.; Antunes, F.; Coelho, P.; Salvador, A. Hydrogen peroxide metabolism and sensing in human erythrocytes: A validated kinetic model and reappraisal of the role of peroxiredoxin II. Free Radic. Biol. Med. 2014, 74, 35–49. [Google Scholar] [CrossRef]
- Chen, B.; Deen, W.M. Analysis of the effects of cell spacing and liquid depth on nitric oxide and its oxidation products in cell cultures. Chem. Res. Toxicol. 2001, 14, 135–147. [Google Scholar] [CrossRef] [PubMed]
- Chen, B.; Deen, W.M. Effect of liquid depth on the synthesis and oxidation of nitric oxide in macrophage cultures. Chem. Res. Toxicol. 2002, 15, 490–496. [Google Scholar] [CrossRef] [PubMed]
- Chen, B.; Keshive, M.; Deen, W.M. Diffusion and reaction of nitric oxide in suspension cell cultures. Biophys. J. 1998, 75, 745–754. [Google Scholar] [CrossRef] [Green Version]
- Chin, M.P.; Deen, W.M. Prediction of nitric oxide concentrations in melanomas. Nitric Oxide Biol. Chem. 2010, 23, 319–326. [Google Scholar] [CrossRef] [PubMed]
- Chin, M.P.; Schauer, D.B.; Deen, W.M. Nitric oxide, oxygen, and superoxide formation and consumption in macrophages and colonic epithelial cells. Chem. Res. Toxicol. 2010, 23, 778–787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, T.M.; Hayton, W.L.; Mallery, S.R. Kinetic modeling of nitric-oxide-associated reaction network. Pharm. Res. 2006, 23, 1702–1711. [Google Scholar] [CrossRef] [PubMed]
- Komalapriya, C.; Kaloriti, D.; Tillmann, A.T.; Yin, Z.K.; Herrero-de-Dios, C.; Jacobsen, M.D.; Belmonte, R.C.; Cameron, G.; Haynes, K.; Grebogi, C.; et al. Integrative model of oxidative stress adaptation in the fungal pathogen candida albicans. PLoS ONE 2015, 10, 32. [Google Scholar] [CrossRef] [Green Version]
- Lim, C.H.; Dedon, P.C.; Deen, W.A. Kinetic analysis of intracellular concentrations of reactive nitrogen species. Chem. Res. Toxicol. 2008, 21, 2134–2147. [Google Scholar] [CrossRef] [Green Version]
- Lim, J.B.; Huang, B.K.; Deen, W.M.; Sikes, H.D. Analysis of the lifetime and spatial localization of hydrogen peroxide generated in the cytosol using a reduced kinetic model. Free Radic. Biol. Med. 2015, 89, 47–53. [Google Scholar] [CrossRef]
- Nalwaya, N.; Deen, W.M. Analysis of the effects of nitric oxide and oxygen on nitric oxide production by macrophages. J. Theor. Biol. 2004, 226, 409–419. [Google Scholar] [CrossRef]
- Nalwaya, N.; Deen, W.M. Peroxynitrite exposure of cells cocultured with macrophages. Ann. Biomed. Eng. 2004, 32, 664–676. [Google Scholar] [CrossRef] [PubMed]
- Bauer, G. siRNA-based analysis of the abrogation of the protective function of membrane-associated catalase of tumor cells. Anticancer Res. 2017, 37, 567–581. [Google Scholar] [CrossRef] [PubMed]
- Bauer, G. Targeting Protective Catalase of Tumor Cells with Cold Atmospheric Plasma-Activated Medium (PAM). Anti-Cancer Agents Med. Chem. 2018, 18, 784–804. [Google Scholar] [CrossRef] [PubMed]
Molecule | Physiological Role(s) | Reported Response to Plasma | Redox Changes and Functional Consequences |
---|---|---|---|
ECM components | |||
Hyaluronan | Regulates cell behaviour interacting with CD44 [167]. Auto- and paracrine signalling between CAF and tumour cells [163] | Unknown | ROS can depolymerize and fragment HA aggregates [165] |
Collagen (COL) | Major ECM component, supports cell movement through ECM [168] | Oxidized amino acids, broke H-bonds [153], and loosen COL I structure [154]. Fibroblast activation to produce COL [169] | ROS induce overproduction, deposition and remodelling of COL to generate stiffer tissue ECM in tumours [104,170] |
Laminin (LM) | Glycoprotein, important in cell differentiation, migration, and adhesion [171]. Cell–cell and cell-ECM interaction | Increased expression in wound bed region in mice [172]. Enhanced adhesion, growth, and viability in plasma-treated LM-modified PCL* [173] | ONOOH and HOCl induce nitration, oxidation, and chlorination of LM residues [174,175,176]. Modification of self-polymerization and cell adhesion sites, which modulates the ECM structure |
Fibronectin (FN) | Involved in cell adhesion, growth, migration, and differentiation | Increased FN expression in THP-1 cells by kINPen [131]. Induced FN formation in activated fibroblasts [169] | ROS oxidize FN [166]. HOCl causes FN oxidation, which modulates cell adhesion, proliferation, and mRNA expression [177,178] |
Elastin | Entropic elastic behaviour, its stretch is limited by its association with collagen [179] | Fibroblast activation in mouse skin to produce elastin [169] | ROS destroy elastin network integrity, influence production of ECM proteins [180]. ONOOH induces tyrosine nitration and crosslinking in topoelastin, alters its structure, function, and changes matrix assembly [181] |
Adhesion to ECM | |||
Focal adhesions (FA) | Complex protein assemblies, bind cells with ECM via actin/integrin links. Mediates mechanical and biochemical “outside-in” and “inside-out” signalling [182]. Cell-ECM and cell–cell adhesion | Increased FA size in WTDF3 mouse fibroblasts [183], expression of α2-integrin/CD49b, β1-integrin/CD29 [184], β1-integrin [185] and FA proteins in HaCaT cells [42]. Activated β1-integrin in WTDF3 mouse fibroblasts [183]. Reduced α5- and β1-integrin in fibroblasts, PAM cells [186,187] | Oxidative stress activates FA kinase, accelerates cell migration [188]. Integrin-linked kinase (ILK) signalling via PKB/Akt can suppress apoptosis and anoikis [189]. ILK required to maintain redox balance [190]. NRF2 mediated oxidative stress response [191] |
Hemidesmosomes (HD) | Integrin-based adhesive junction [192]. Cell-ECM and cell–cell adhesion | Upregulation of proteins related to HD assembly by plasma [42] | HD disruption facilitates cell detachment and migration [193]. |
Catalytic enzymes | |||
Catalase | Membrane-associated catalase protects cancer cell from apoptosis-inducing signalling pathways | Inactivation through reaction of 1O2 with histidine in the active site [59] | Generation of •OH leads to lipid peroxidation and induction of apoptosis. Generation of 1O2 inactivates catalase at adjacent cells [59] |
Superoxide dismutase (SOD) | Membrane-associated SOD protects catalase from inactivation by O2•‒ | Inactivation through reaction of 1O2 with histidine in the active site [59] | Increased concentration of O2•‒ and subsequent enhanced generation of •OH [59] |
Peroxidase (POD) | Pathogenic resistance | Unknown | N/A |
NADPH oxidase 1 (NOX1) | Cell proliferation | Unknown | N/A |
Molecule | Physiological Role(s) | Reported Response to Plasma | Redox Changes and Functional Consequences |
---|---|---|---|
Communication junctions | |||
Ion channels | Ca2+-permeable and voltage-independent cation channels. Include transient receptor potential (TRP) channels. Auto- and paracrine cell–cell communication | Activated intracellular Ca2+ influx through TRP channels [203]. Induced Ca2+ release by ER* and mitochondria needed to induce senescence in melanoma cells [204] | ROS affect channel function, structure and downstream signalling pathways [205]. Can sense lipid oxidation [206]. Increased intracellular [Ca2+] by TRPC3 and TRPC4 leads to cell death upon oxidative stress [207]. H2O2 oxidizes TRPM2 and induces chemokine production [208]. TRP7 blockade induces apoptosis [209] |
Pannexins (Panx) | Transmembrane proteins, form channels for the release of ATP and other metabolites [210]. Auto- and paracrine cell–cell communication | Unknown | Oxidative stress regulates Panx channel activation; ATP, ADP, and AMP release for apoptotic cell clearance [210]. Overexpression of Panx1 in cancer [211], its inhibition reduces tumour growth and invasiveness [212]. NO may inhibit Panx1 current [213] |
Extracellular vesicles (EVs) | Secreted exosomes, microvesicles and apoptotic bodies [214]. Interact with adjacent or distant cells [215]. Para-, auto-, exo- and endocrine cell–cell communication | Increased number of EVs released by THP-1 and PMN* [216]. Less EVs produced by plasma-treated OVCAR-3 and SKOV-3 ovarian cancer cells [195]. Induced formation of apoptotic bodies [26,217,218] | Tumour cells produce high number of EVs with altered redox balance and ROS levels. EVs can scavenge/produce ROS and modify ROS content in target cells [219]. EVs involved in tumour development and metastasis [214] |
Gap junctions (GJs) | Connect cells for electrical and metabolic (sugars, ions, amino acids, nucleotides) communication [220]. Auto- and paracrine communication | Plasma-generated ROS and intracellular ROS produced upon plasma treatment triggered bystander effect and damaged GJs [197] | Bystander effect: GJs can transmit ROS and cell death signals to neighbouring cells [196,221] |
Connexins | Form gap junctions, transfer ions, small messengers, and metabolites. Forms hemichannels that communicate intra- and extracellular spaces [222] | Destroyed structure of connexins’ N-terminal tail [197]. Temporary loss of cell–cell contact [223]. Reduced Cx43 connexin expression in epithelial cells, transient increase of Cx43 in fibroblasts [187] | Redox status modulates the opening/closing and permeability of connexin hemichannels to NO and large molecules [224] |
Tunnelling nanotubes (TnTs) | Long, filamentous, actin-based structures, connect cells to transfer drugs, organelles, nucleic acids, and proteins [225]. Cell–cell communication | Unknown | High H2O2 levels induce TnTs formation [226]. Propagation of death signal Fas ligand through TnTs between T cells [227]. TnTs mediate mitochondria transfer to rescue cells on oxidative stress [228]. Increased number of TnTs upon high oxidative stress [229] |
Tight junctions (TJs) | Restrict diffusion based on size and charge to maintain homeostasis. TJs maintain cell surface polarity [230]. Cell–cell communication | Disrupted tight junctions in epithelial cells and caused retraction of Zonula occludens ZO-1 protein from cell membrane [231] | High doses of NO and H2O2 increases paracellular permeability in epithelial cells [232] |
Claudins | Main structural TJs proteins. Block lipid and protein diffusion, ease transference of small ions [233] | Downregulated expression by repetitive exposure to plasma-treated medium [234] | ONOO‒ could interfere with claudin function [235]. Lipid peroxidation [236] and H2O2 can disrupt tight junctions [237] |
Occludins | Contribute to TJ stability and barrier function [238] | Downregulated expression by repetitive exposure to plasma-treated medium [234] | Oxidative stress reduces occludin oligomerization [239], interaction with other TJ proteins and barrier tightness [240]. H2O2 induces occludins cleavage [241]; NO abolishes its immunoreactivity and redirects it to cytoplasm [240] |
Anchoring junctions | |||
Adherens | Homophilic lateral cell-to-cell adhesion via cadherin/catenin complex, transmit mechanical forces between cells, regulate signalling and transcription [242]. Required for TJs assembly [233] | Decreased E-cad expression [185,187]; function modulation, internalization in HaCaT cells [243]. Decreased E-cad in mice epidermis cells [243]. Increased E-cad expression in wounds of rats [244] and β-catenin expression in keratinocytes [234] | ROS selectively disrupts cadherin/catenin complexes [245,246], modulate receptors involved in cell-matrix and cell–cell adhesion [247]. Loss of E-cadherin activates EMT [248]. |
Desmosomes | Intercellular junctions, link cells and stabilize the tissue structure [249]. Cell–cell adhesion | Increased the number of desmosomes in wounds [105] | ROS induce PKP3 phosphorylation, pPKP3 release from desmosome and desmosome instability [250]. Desmosomes are intracellular signal transducers in Wnt pathway [251] |
3D Culture Models | Main Feature | Advantages | Disadvantages | Suitable Plasma Treatment |
---|---|---|---|---|
Spheroids | Self assembly | Formed from cell lines | Simple architecture | Direct and indirect |
Recreates gradients of nutrients/oxygen | Not all cell lines form spheroids | |||
Easy to generate | Static conditions | |||
Uniform size Reproducible | ||||
High-throughput | Plasma treatment in presence of liquid | |||
Allows co-cultures | ||||
Organoids | Capable of self-renewal and self-organization | Formed from primary cells | Require validation to identify outgrow of unwanted cells (normal/cancer cells) | Direct and indirect |
Requires small amount of tissue | Requires access to human samples | |||
Resembles complexity, architecture, gene expression from in vivo tumours | More expensive | |||
Can be transplanted into mice | Static conditions | |||
Allows co-cultures | Plasma treatment in presence of liquid | |||
Scaffolds | Naturally-derived ECM components or synthetic polymers | Formed from primary cells or cell lines | Batch-to-batch variability of natural matrixes | Direct and indirect |
Resemble mechanical forces in tumours | ||||
Versatile | Synthetic matrixes can be expensive | |||
Diffusion gradients | ||||
Very reproducible | Might require complex cell retrieval/imaging methods | |||
Allow co-cultures | ||||
Tumour-on-a-chip | Spatiotemporal control of chemical/physical properties | Formed from primary cells or cell lines | More expensive | Indirect only |
Resembles diffusion gradients/perfusion | Requires special equipment | |||
Highly sensitive | ||||
Vascularized | Difficult to scale up | |||
Allow co-cultures | ||||
3D-bioprinted tumours | Precise control of biomaterials, cells, and biological factors | Formed from primary cells or cell lines | More expensive | Direct and indirect |
Recreates natural function and structure | Requires special equipment | |||
High-throughput | ||||
Vascularized | Needs optimization | |||
Allow co-cultures |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Privat-Maldonado, A.; Bengtson, C.; Razzokov, J.; Smits, E.; Bogaerts, A. Modifying the Tumour Microenvironment: Challenges and Future Perspectives for Anticancer Plasma Treatments. Cancers 2019, 11, 1920. https://doi.org/10.3390/cancers11121920
Privat-Maldonado A, Bengtson C, Razzokov J, Smits E, Bogaerts A. Modifying the Tumour Microenvironment: Challenges and Future Perspectives for Anticancer Plasma Treatments. Cancers. 2019; 11(12):1920. https://doi.org/10.3390/cancers11121920
Chicago/Turabian StylePrivat-Maldonado, Angela, Charlotta Bengtson, Jamoliddin Razzokov, Evelien Smits, and Annemie Bogaerts. 2019. "Modifying the Tumour Microenvironment: Challenges and Future Perspectives for Anticancer Plasma Treatments" Cancers 11, no. 12: 1920. https://doi.org/10.3390/cancers11121920
APA StylePrivat-Maldonado, A., Bengtson, C., Razzokov, J., Smits, E., & Bogaerts, A. (2019). Modifying the Tumour Microenvironment: Challenges and Future Perspectives for Anticancer Plasma Treatments. Cancers, 11(12), 1920. https://doi.org/10.3390/cancers11121920