Colorectal Cancer Cell-Derived Extracellular Vesicles Promote Angiogenesis Through JAK/STAT3/VEGFA Signaling
Simple Summary
Abstract
1. Introduction
2. Materials and Methods
2.1. Cell Lines and Cell Culture
2.2. Isolation and Characterization of Extracellular Vesicles
2.3. Uptake of HT29-EVs
2.4. Proliferation, Migration, and Tube Formation Assays
2.5. Animal Studies
2.6. Immunohistochemistry
2.7. Western Blotting Analysis
2.8. Immunofluorescence
2.9. F-Actin Cytoskeleton Staining Assay
2.10. Bioinformatics Analysis
2.11. Statistical Analysis
3. Results
3.1. Isolation and Characterization of HT29-EVs
3.2. HT29-EVs Promote the Growth and Angiogenesis of HT-29 Colorectal Cancer Tumors In Vivo
3.3. HT29-EVs Are Taken Up by HUVECs and Promote Their Proliferation, Migration, and Tube Formation
3.4. HT29-EVs Regulate Angiogenesis Possibly Through the JAK/STAT3 Pathway
3.5. The Promoting Effect of CRC-EVs on HUVECs Was Attenuated by Inhibiting the JAK/STAT3 Signaling Pathway
4. Discussion
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.-L.; Chen, H.-H.; Zheng, L.-L.; Sun, L.-P.; Shi, L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct. Target. Ther. 2023, 8, 198. [Google Scholar] [CrossRef]
- Xi, Y.; Xu, P. Global colorectal cancer burden in 2020 and projections to 2040. Transl. Oncol. 2021, 14, 101174. [Google Scholar] [CrossRef]
- Cao, M.; Wang, Y.; Lu, G.; Qi, H.; Li, P.; Dai, X.; Lu, J. Classical Angiogenic Signaling Pathways and Novel Anti-Angiogenic Strategies for Colorectal Cancer. Curr. Issues Mol. Biol. 2022, 44, 4447–4471. [Google Scholar] [CrossRef] [PubMed]
- Folkman, J. Tumor angiogenesis: Therapeutic implications. N. Engl. J. Med. 1971, 285, 1182–1186. [Google Scholar] [CrossRef]
- Kumar, M.A.; Baba, S.K.; Sadida, H.Q.; Marzooqi, S.A.; Jerobin, J.; Altemani, F.H.; Algehainy, N.; Alanazi, M.A.; Abou-Samra, A.B.; Kumar, R.; et al. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct. Target. Ther. 2024, 9, 27. [Google Scholar] [CrossRef] [PubMed]
- Takahashi, Y.; Takakura, Y. Extracellular vesicle-based therapeutics: Extracellular vesicles as therapeutic targets and agents. Pharmacol. Ther. 2023, 242, 108352. [Google Scholar] [CrossRef]
- Kalluri, R.; McAndrews, K.M. The role of extracellular vesicles in cancer. Cell 2023, 186, 1610–1626. [Google Scholar] [CrossRef]
- Maacha, S.; Bhat, A.A.; Jimenez, L.; Raza, A.; Haris, M.; Uddin, S.; Grivel, J.C. Extracellular vesicles-mediated intercellular communication: Roles in the tumor microenvironment and anti-cancer drug resistance. Mol. Cancer 2019, 18, 55. [Google Scholar] [CrossRef]
- Herbert, S.P.; Stainier, D.Y.R. Molecular control of endothelial cell behaviour during blood vessel morphogenesis. Nat. Rev. Mol. Cell Biol. 2011, 12, 551–564. [Google Scholar] [CrossRef]
- Han, B.; Zhang, H.; Tian, R.; Liu, H.; Wang, Z.; Wang, Z.; Tian, J.; Cui, Y.; Ren, S.; Zuo, X.; et al. Exosomal EPHA2 derived from highly metastatic breast cancer cells promotes angiogenesis by activating the AMPK signaling pathway through Ephrin A1-EPHA2 forward signaling. Theranostics 2022, 12, 4127–4146. [Google Scholar] [CrossRef] [PubMed]
- Kornilov, R.; Puhka, M.; Mannerström, B.; Hiidenmaa, H.; Peltoniemi, H.; Siljander, P.; Seppänen-Kaijansinkko, R.; Kaur, S. Efficient ultrafiltration-based protocol to deplete extracellular vesicles from fetal bovine serum. J. Extracell. Vesicles 2018, 7, 1422674. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Mahairaki, V.; Bai, H.; Ding, Z.; Li, J.; Witwer, K.W.; Cheng, L. Highly Purified Human Extracellular Vesicles Produced by Stem Cells Alleviate Aging Cellular Phenotypes of Senescent Human Cells. Stem Cells 2019, 37, 779–790. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Cao, Z.; Wei, Q.; Ma, K.; Hu, W.; Huang, Q.; Su, J.; Li, H.; Zhang, C.; Fu, X. VH298-loaded extracellular vesicles released from gelatin methacryloyl hydrogel facilitate diabetic wound healing by HIF-1α-mediated enhancement of angiogenesis. Acta Biomater. 2022, 147, 342–355. [Google Scholar] [CrossRef]
- Jiang, Y.; Wang, L.; Yang, B.; Ma, G.; Chen, Z.; Ma, J.; Chang, X.; Fang, L.; Wang, Z. Bifidobacterium-derived membrane vesicles inhibit triple-negative breast cancer growth by inducing tumor cell apoptosis. Mol. Biol. Rep. 2023, 50, 7547–7556. [Google Scholar] [CrossRef]
- Nowak-Sliwinska, P.; Alitalo, K.; Allen, E.; Anisimov, A.; Aplin, A.C.; Auerbach, R.; Augustin, H.G.; Bates, D.O.; van Beijnum, J.R.; Bender, R.H.F.; et al. Consensus guidelines for the use and interpretation of angiogenesis assays. Angiogenesis 2018, 21, 425–532. [Google Scholar] [CrossRef]
- He, L.; Zhu, W.; Chen, Q.; Yuan, Y.; Wang, Y.; Wang, J.; Wu, X. Ovarian cancer cell-secreted exosomal miR-205 promotes metastasis by inducing angiogenesis. Theranostics 2019, 9, 8206–8220. [Google Scholar] [CrossRef]
- Alhasan, L.; Addai, Z. Allicin-induced modulation of angiogenesis in lung cancer cells (A549). Trop. J. Pharm. Res. 2018, 17, 2129–2134. [Google Scholar] [CrossRef]
- Barrett, T.; Wilhite, S.E.; Ledoux, P.; Evangelista, C.; Kim, I.F.; Tomashevsky, M.; Marshall, K.A.; Phillippy, K.H.; Sherman, P.M.; Holko, M.; et al. NCBI GEO: Archive for functional genomics data sets--update. Nucleic Acids Res. 2013, 41, D991–D995. [Google Scholar] [CrossRef]
- Chin, C.-H.; Chen, S.-H.; Wu, H.-H.; Ho, C.-W.; Ko, M.-T.; Lin, C.-Y. cytoHubba: Identifying hub objects and sub-networks from complex interactome. BMC Syst. Biol. 2014, 8, S11. [Google Scholar] [CrossRef]
- Yu, G.; Wang, L.G.; Han, Y.; He, Q.Y. clusterProfiler: An R package for comparing biological themes among gene clusters. Omics 2012, 16, 284–287. [Google Scholar] [CrossRef] [PubMed]
- Bisaria, A.; Hayer, A.; Garbett, D.; Cohen, D.; Meyer, T. Membrane-proximal F-actin restricts local membrane protrusions and directs cell migration. Science 2020, 368, 1205–1210. [Google Scholar] [CrossRef] [PubMed]
- Hicklin, D.J.; Ellis, L.M. Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J. Clin. Oncol. 2005, 23, 1011–1027. [Google Scholar] [CrossRef] [PubMed]
- Ogata-Kawata, H.; Izumiya, M.; Kurioka, D.; Honma, Y.; Yamada, Y.; Furuta, K.; Gunji, T.; Ohta, H.; Okamoto, H.; Sonoda, H.; et al. Circulating exosomal microRNAs as biomarkers of colon cancer. PLoS ONE 2014, 9, e92921. [Google Scholar] [CrossRef]
- Xue, C.; Xie, J.; Zhao, D.; Lin, S.; Zhou, T.; Shi, S.; Shao, X.; Lin, Y.; Zhu, B.; Cai, X. The JAK/STAT3 signalling pathway regulated angiogenesis in an endothelial cell/adipose-derived stromal cell co-culture, 3D gel model. Cell Prolif. 2017, 50, e12307. [Google Scholar] [CrossRef]
- Liang, L.; Hui, K.; Hu, C.; Wen, Y.; Yang, S.; Zhu, P.; Wang, L.; Xia, Y.; Qiao, Y.; Sun, W.; et al. Autophagy inhibition potentiates the anti-angiogenic property of multikinase inhibitor anlotinib through JAK2/STAT3/VEGFA signaling in non-small cell lung cancer cells. J. Exp. Clin. Cancer Res. 2019, 38, 71. [Google Scholar] [CrossRef]
- Yang, E.; Wang, X.; Gong, Z.; Yu, M.; Wu, H.; Zhang, D. Exosome-mediated metabolic reprogramming: The emerging role in tumor microenvironment remodeling and its influence on cancer progression. Signal Transduct. Target. Ther. 2020, 5, 242. [Google Scholar] [CrossRef]
- Dai, X.; Xie, Y.; Dong, M. Cancer-associated fibroblasts derived extracellular vesicles promote angiogenesis of colorectal adenocarcinoma cells through miR-135b-5p/FOXO1 axis. Cancer Biol. Ther. 2022, 23, 76–88. [Google Scholar] [CrossRef] [PubMed]
- Huang, M.; Liu, M.; Huang, D.; Ma, Y.; Ye, G.; Wen, Q.; Li, Y.; Deng, L.; Qi, Q.; Liu, T.; et al. Tumor perivascular cell-derived extracellular vesicles promote angiogenesis via the Gas6/Axl pathway. Cancer Lett. 2022, 524, 131–143. [Google Scholar] [CrossRef]
- Li, X.; Peng, X.; Zhang, C.; Bai, X.; Li, Y.; Chen, G.; Guo, H.; He, W.; Zhou, X.; Gou, X. Bladder Cancer-Derived Small Extracellular Vesicles Promote Tumor Angiogenesis by Inducing HBP-Related Metabolic Reprogramming and SerRS O-GlcNAcylation in Endothelial Cells. Adv. Sci. 2022, 9, e2202993. [Google Scholar] [CrossRef]
- Folkman, J. Anti-angiogenesis: New concept for therapy of solid tumors. Ann. Surg. 1972, 175, 409–416. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Kang, G.; Wang, T.; Huang, H. Tumor angiogenesis and anti-angiogenic gene therapy for cancer. Oncol. Lett. 2018, 16, 687–702. [Google Scholar] [CrossRef] [PubMed]
- Siveen, K.S.; Prabhu, K.; Krishnankutty, R.; Kuttikrishnan, S.; Tsakou, M.; Alali, F.Q.; Dermime, S.; Mohammad, R.M.; Uddin, S. Vascular Endothelial Growth Factor (VEGF) Signaling in Tumour Vascularization: Potential and Challenges. Curr. Vasc. Pharmacol. 2017, 15, 339–351. [Google Scholar] [CrossRef]
- Al Kawas, H.; Saaid, I.; Jank, P.; Westhoff, C.C.; Denkert, C.; Pross, T.; Weiler, K.B.S.; Karsten, M.M. How VEGF-A and its splice variants affect breast cancer development—Clinical implications. Cell Oncol. 2022, 45, 227–239. [Google Scholar] [CrossRef]
- Zhao, C.; Wang, H.; Xiong, C.; Liu, Y. Hypoxic glioblastoma release exosomal VEGF-A induce the permeability of blood-brain barrier. Biochem. Biophys. Res. Commun. 2018, 502, 324–331. [Google Scholar] [CrossRef]
- Zhang, Z.; Shuai, Y.; Zhou, F.; Yin, J.; Hu, J.; Guo, S.; Wang, Y.; Liu, W. PDLSCs Regulate Angiogenesis of Periodontal Ligaments via VEGF Transferred by Exosomes in Periodontitis. Int. J. Med. Sci. 2020, 17, 558–567. [Google Scholar] [CrossRef] [PubMed]
- Heck, K.A.; Lindholm, H.T.; Niederdorfer, B.; Tsirvouli, E.; Kuiper, M.; Flobak, Å.; Lægreid, A.; Thommesen, L. Characterisation of Colorectal Cancer Cell Lines through Proteomic Profiling of Their Extracellular Vesicles. Proteomes 2023, 11, 3. [Google Scholar] [CrossRef]
- Chen, K.; Wang, Q.; Liu, X.; Wang, F.; Yang, Y.; Tian, X. Hypoxic pancreatic cancer derived exosomal miR-30b-5p promotes tumor angiogenesis by inhibiting GJA1 expression. Int. J. Biol. Sci. 2022, 18, 1220–1237. [Google Scholar] [CrossRef]
- Shang, D.; Xie, C.; Hu, J.; Tan, J.; Yuan, Y.; Liu, Z.; Yang, Z. Pancreatic cancer cell-derived exosomal microRNA-27a promotes angiogenesis of human microvascular endothelial cells in pancreatic cancer via BTG2. J. Cell Mol. Med. 2020, 24, 588–604. [Google Scholar] [CrossRef]
- Muthyalaiah, Y.S.; Jonnalagadda, B.; John, C.M.; Arockiasamy, S. Impact of Advanced Glycation End products (AGEs) and its receptor (RAGE) on cancer metabolic signaling pathways and its progression. Glycoconj. J. 2021, 38, 717–734. [Google Scholar] [CrossRef]
- Farhan, M.; Silva, M.; Xingan, X.; Huang, Y.; Zheng, W. Role of FOXO Transcription Factors in Cancer Metabolism and Angiogenesis. Cells 2020, 9, 1586. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Tan, J.; Xing, Y.; Kan, N.; Ling, J.; Dong, G.; Liu, G.; Chen, H. p43 induces IP-10 expression through the JAK-STAT signaling pathway in HMEC-1 cells. Int. J. Mol. Med. 2016, 38, 1217–1224. [Google Scholar] [CrossRef] [PubMed]
- Rajakumar, T.; Pugalendhi, P. Allyl isothiocyanate inhibits invasion and angiogenesis in breast cancer via EGFR-mediated JAK-1/STAT-3 signaling pathway. Amino Acids 2023, 55, 981–992. [Google Scholar] [CrossRef] [PubMed]
- Chiyomaru, T.; Yamamura, S.; Fukuhara, S.; Hidaka, H.; Majid, S.; Saini, S.; Arora, S.; Deng, G.; Shahryari, V.; Chang, I.; et al. Genistein up-regulates tumor suppressor microRNA-574-3p in prostate cancer. PLoS ONE 2013, 8, e58929. [Google Scholar] [CrossRef]
- Wang, Z.Z.; Luo, Y.R.; Du, J.; Yu, Y.; Yang, X.Z.; Cui, Y.J.; Jin, X.F. MiR-296-5p inhibits cell invasion and migration of esophageal squamous cell carcinoma by downregulating STAT3 signaling. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 5206–5214. [Google Scholar] [CrossRef] [PubMed]
- Rébé, C.; Végran, F.; Berger, H.; Ghiringhelli, F. STAT3 activation: A key factor in tumor immunoescape. Jakstat 2013, 2, e23010. [Google Scholar] [CrossRef]
- Sgrignani, J.; Garofalo, M.; Matkovic, M.; Merulla, J.; Catapano, C.V.; Cavalli, A. Structural Biology of STAT3 and Its Implications for Anticancer Therapies Development. Int. J. Mol. Sci. 2018, 19, 1591. [Google Scholar] [CrossRef]
- Zhang, Q.; Xu, F.; Shi, Y.; Chen, Y.-W.; Wang, H.-P.; Yu, X.; Li, Y. C-X-C motif chemokine receptor 4 promotes tumor angiogenesis in gastric cancer via activation of JAK2/STAT3. Cell Biol. Int. 2017, 41, 854–862. [Google Scholar] [CrossRef]
- Schust, J.; Sperl, B.; Hollis, A.; Mayer, T.U.; Berg, T. Stattic: A small-molecule inhibitor of STAT3 activation and dimerization. Chem. Biol. 2006, 13, 1235–1242. [Google Scholar] [CrossRef]
- Wang, H.; Liu, Z.; Guan, L.; Li, J.; Chen, S.; Yu, W.; Lai, M. LYW-6, a novel cryptotanshinone derived STAT3 targeting inhibitor, suppresses colorectal cancer growth and metastasis. Pharmacol. Res. 2020, 153, 104661. [Google Scholar] [CrossRef]
- Fong, S.S.; Foo, Y.Y.; Saw, W.S.; Leo, B.F.; Teo, Y.Y.; Chung, I.; Goh, B.T.; Misran, M.; Imae, T.; Chang, C.C.; et al. Chitosan-Coated-PLGA Nanoparticles Enhance the Antitumor and Antimigration Activity of Stattic—A STAT3 Dimerization Blocker. Int. J. Nanomed. 2022, 17, 137–150. [Google Scholar] [CrossRef] [PubMed]
- Masoumi-Dehghi, S.; Babashah, S.; Sadeghizadeh, M. microRNA-141-3p-containing small extracellular vesicles derived from epithelial ovarian cancer cells promote endothelial cell angiogenesis through activating the JAK/STAT3 and NF-κB signaling pathways. J. Cell Commun. Signal 2020, 14, 233–244. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Long, Y.; Dan, Y.; Jiang, Y.; Ma, J.; Zhou, T.; Fang, L.; Wang, Z. Colorectal Cancer Cell-Derived Extracellular Vesicles Promote Angiogenesis Through JAK/STAT3/VEGFA Signaling. Biology 2024, 13, 873. https://doi.org/10.3390/biology13110873
Long Y, Dan Y, Jiang Y, Ma J, Zhou T, Fang L, Wang Z. Colorectal Cancer Cell-Derived Extracellular Vesicles Promote Angiogenesis Through JAK/STAT3/VEGFA Signaling. Biology. 2024; 13(11):873. https://doi.org/10.3390/biology13110873
Chicago/Turabian StyleLong, Yuqing, Yuxi Dan, Yao Jiang, Jing Ma, Tao Zhou, Liaoqiong Fang, and Zhibiao Wang. 2024. "Colorectal Cancer Cell-Derived Extracellular Vesicles Promote Angiogenesis Through JAK/STAT3/VEGFA Signaling" Biology 13, no. 11: 873. https://doi.org/10.3390/biology13110873
APA StyleLong, Y., Dan, Y., Jiang, Y., Ma, J., Zhou, T., Fang, L., & Wang, Z. (2024). Colorectal Cancer Cell-Derived Extracellular Vesicles Promote Angiogenesis Through JAK/STAT3/VEGFA Signaling. Biology, 13(11), 873. https://doi.org/10.3390/biology13110873