Radiation-induced extracellular vesicles from cancer-associated fibroblasts drive oesophageal squamous cell carcinoma metastasis via the miR-193a-3p/PTEN/Akt pathway

Yechun Pang , Tiantian Guo , Yue Zhou , Shanshan Jiang , Yida Li , Jianjiao Ni , Xiao Chu , Li Chu , Fangyu Chen , Xi Yang , Zhengfei Zhu

Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (10) : e70483

PDF
Clinical and Translational Medicine ›› 2025, Vol. 15 ›› Issue (10) : e70483 DOI: 10.1002/ctm2.70483
RESEARCH ARTICLE

Radiation-induced extracellular vesicles from cancer-associated fibroblasts drive oesophageal squamous cell carcinoma metastasis via the miR-193a-3p/PTEN/Akt pathway

Author information +
History +
PDF

Abstract

Background: The recurrence and metastasis of oesophageal squamous cell cancer (ESCC) following radiation therapy are major treatment challenges. Cancer-associated fibroblasts (CAFs) are key in the ESCC microenvironment, yet their role in post-radiation recurrence remains unclear.

Materials and Methods: KYSE150 ESCC cells were co-implanted with non-irradiated (0 Gy) or irradiated (8 Gy) CAFs in nude mice. CAF-derived extracellular vesicles (EVs) were isolated via differential centrifugation and analysed by electron microscopy and immunoblotting. Transwell assays evaluated EVs' effects on ESCC cell migration and invasion in vitro. RNA sequencing identified differentially expressed microRNAs, and functional experiments verified the role of miR-193a-3p. Plasma samples from 32 ESCC patients and tissue samples from 76 ESCC patients were analysed for miR-193a-3p expression.

Results: Irradiated CAFs promoted the lung metastasis of ESCC cells in vivo, and their EVs enhanced ESCC cell invasion, migration and metastasis. Elevated miR-193a-3p levels in EVs from irradiated CAFs increased miR-193a-3p expression in ESCC cells. This effect was effectively attenuated by RNase and Triton X-100 (degrading microRNAs encapsulated in EVs), or GW4869 (inhibiting EVs biogenesis and secretion)—indicating that miR-193a-3p functions in an EV-dependent manner. Knockdown of miR-193a-3p diminished the invasion, migration and epithelial–mesenchymal transition (EMT)-promoting activities of CAF-derived EVs. Luciferase assays confirmed PTEN as a target of miR-193a-3p; miR-193a-3p overexpression decreased PTEN and increased p-Akt expression. In vivo, coinjection of miR-193a-3p-knockdown CAFs with KYSE150 ESCC cells resulted in smaller tumours, fewer lung metastases, increased PTEN and E-cadherin, and decreased p-Akt and Snail expression. Clinically, radiation increased plasma exosomal miR-193a-3p levels, and high miR-193a-3p expression was correlated with shorter survival, identifying miR-193a-3p as an independent predictor of poor prognosis in ESCC patients.

Conclusion: EVs from irradiated CAFs promote ESCC metastasis via the miR-193a-3p-mediated PTEN/Akt signalling pathway. Targeting this EVs-mediated interaction represents a promising strategy for improving ESCC radiotherapy outcomes.

Keywords

ESCC / extracellular vesicles / metastasis / radiation

Cite this article

Download citation ▾
Yechun Pang, Tiantian Guo, Yue Zhou, Shanshan Jiang, Yida Li, Jianjiao Ni, Xiao Chu, Li Chu, Fangyu Chen, Xi Yang, Zhengfei Zhu. Radiation-induced extracellular vesicles from cancer-associated fibroblasts drive oesophageal squamous cell carcinoma metastasis via the miR-193a-3p/PTEN/Akt pathway. Clinical and Translational Medicine, 2025, 15(10): e70483 DOI:10.1002/ctm2.70483

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021; 71: 209-249.

[2]

Obermannová R, Alsina M, Cervantes A, et al. Oesophageal cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol. 2022; 33: 992-1004.

[3]

Ishikawa H, Sakurai H, Yamakawa M, et al. Clinical outcomes and prognostic factors for patients with early esophageal squamous cell carcinoma treated with definitive radiation therapy alone. J Clin Gastroenterol. 2005; 39: 495-500.

[4]

Minsky BD, Pajak TF, Ginsberg RJ, et al. INT 0123 (Radiation Therapy Oncology Group 94-05) phase III trial of combined-modality therapy for esophageal cancer: high-dose versus standard-dose radiation therapy. J Clin Oncol. 2002; 20: 1167-1174.

[5]

Barker HE, Paget JT, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer. 2015; 15: 409-425.

[6]

Ji J, Ding K, Cheng B, et al. Radiotherapy-induced astrocyte senescence promotes an immunosuppressive microenvironment in glioblastoma to facilitate tumor regrowth. Adv Sci (Weinh). 2024; 11: e2304609.

[7]

Jiang W, Chan CK, Weissman IL, Kim BYS, Hahn SM. Immune priming of the tumor microenvironment by radiation. Trends Cancer. 2016; 2: 638-645.

[8]

Chen M, Xiao L, Jia H, et al. Stereotactic ablative radiotherapy and FAPα-based cancer vaccine suppresses metastatic tumor growth in 4T1 mouse breast cancer. Radiother Oncol. 2023; 189: 109946.

[9]

Raaijmakers KTPM, Adema GJ, Bussink J, Ansems M. Cancer-associated fibroblasts, tumor and radiotherapy: interactions in the tumor micro-environment. J Exp Clin Cancer Res. 2024; 43: 323.

[10]

Ragunathan K, Upfold NLE, Oksenych V. Interaction between fibroblasts and immune cells following DNA damage induced by ionizing radiation. Int J Mol Sci. 2020; 21: 8635.

[11]

Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov. 2019; 18: 99-115.

[12]

Li X, Yong T, Wei Z, et al. Reversing insufficient photothermal therapy-induced tumor relapse and metastasis by regulating cancer-associated fibroblasts. Nat Commun. 2022; 13: 2794.

[13]

Liang T, Tao T, Wu K, et al. Cancer-associated fibroblast-induced remodeling of tumor microenvironment in recurrent bladder cancer. Adv Sci (Weinh). 2023; 10: e2303230.

[14]

Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006; 6: 392-401.

[15]

Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016; 16: 582-598.

[16]

Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013; 19: 1423-1437.

[17]

Swartz MA, Iida N, Roberts EW, et al. Tumor microenvironment complexity: emerging roles in cancer therapy. Cancer Res. 2012; 72: 2473-2480.

[18]

Lavie D, Ben-Shmuel A, Erez N, Scherz-Shouval R. Cancer-associated fibroblasts in the single-cell era. Nat Cancer. 2022; 3: 793-807.

[19]

Shi Z, Jiang T, Cao B, Sun X, Liu J. CAF-derived exosomes deliver LINC01410 to promote epithelial-mesenchymal transition of esophageal squamous cell carcinoma. Exp Cell Res. 2022; 412: 113033.

[20]

Che Y, Wang J, Li Y, et al. Cisplatin-activated PAI-1 secretion in the cancer-associated fibroblasts with paracrine effects promoting esophageal squamous cell carcinoma progression and causing chemoresistance. Cell Death Dis. 2018; 9: 759.

[21]

Zhang H, Yue J, Jiang Z, et al. CAF-secreted CXCL1 conferred radioresistance by regulating DNA damage response in a ROS-dependent manner in esophageal squamous cell carcinoma. Cell Death Dis. 2017; 8: e2790.

[22]

Chen QY, Li YN, Wang XY, et al. Tumor fibroblast-derived FGF2 regulates expression of SPRY1 in esophageal tumor-infiltrating T cells and plays a role in T-cell exhaustion. Cancer Res. 2020; 80: 5583-5596.

[23]

Tommelein J, De Vlieghere E, Verset L, et al. Radiotherapy-activated cancer-associated fibroblasts promote tumor progression through paracrine IGF1R activation. Cancer Res. 2018; 78: 659-670.

[24]

Becker A, Thakur BK, Weiss JM, Kim HS, Peinado H, Lyden D. Extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell. 2016; 30: 836-848.

[25]

Ortiz A. Extracellular vesicles in cancer progression. Semin Cancer Biol. 2021; 76: 139-142.

[26]

Lazar S, Goldfinger LE. Platelets and extracellular vesicles and their cross talk with cancer. Blood. 2021; 137: 3192-3200.

[27]

Xu R, Greening DW, Zhu HJ, Takahashi N, Simpson RJ. Extracellular vesicle isolation and characterization: toward clinical application. J Clin Invest. 2016; 126: 1152-1162.

[28]

Zhang C, Qin C, Dewanjee S, et al. Tumor-derived small extracellular vesicles in cancer invasion and metastasis: molecular mechanisms, and clinical significance. Mol Cancer. 2024; 23: 18.

[29]

Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020; 367: eaau6977.

[30]

Qi R, Bai Y, Li K, et al. Cancer-associated fibroblasts suppress ferroptosis and induce gemcitabine resistance in pancreatic cancer cells by secreting exosome-derived ACSL4-targeting miRNAs. Drug Resist Updat. 2023; 68: 100960.

[31]

Zhuang J, Shen L, Li M, et al. Cancer-associated fibroblast-derived miR-146a-5p generates a niche that promotes bladder cancer stemness and chemoresistance. Cancer Res. 2023; 83: 1611-1627.

[32]

Chen B, Sang Y, Song X, et al. Exosomal miR-500a-5p derived from cancer-associated fibroblasts promotes breast cancer cell proliferation and metastasis through targeting USP28. Theranostics. 2021; 11: 3932-3947.

[33]

Zhao H, Yang L, Baddour J, et al. Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism. Elife. 2016; 5: e10250.

[34]

Chen X, Liu Y, Zhang Q, et al. Exosomal miR-590-3p derived from cancer-associated fibroblasts confers radioresistance in colorectal cancer. Mol Ther Nucleic Acids. 2020; 24: 113-126.

[35]

Zhao S, Mi Y, Zheng B, et al. Highly-metastatic colorectal cancer cell released miR-181a-5p-rich extracellular vesicles promote liver metastasis by activating hepatic stellate cells and remodelling the tumour microenvironment. J Extracell Vesicles. 2022; 11: e12186.

[36]

Mori MA, Ludwig RG, Garcia-Martin R, Brandão BB, Kahn CR. Extracellular miRNAs: from biomarkers to mediators of physiology and disease. Cell Metab. 2019; 30: 656-673.

[37]

Yang N, Hellevik T, Berzaghi R, Martinez-Zubiaurre I. Radiation-induced effects on TGF-β and PDGF receptor signaling in cancer-associated fibroblasts. Cancer Rep (Hoboken). 2024; 7(3): e2018.

[38]

Lyu G, Guan Y, Zhang C, et al. TGF-β signaling alters H4K20me3 status via miR-29 and contributes to cellular senescence and cardiac aging. Nat Commun. 2018; 9(1): 2560.

[39]

Ahmed KM, Nantajit D, Fan M, Murley JS, Grdina DJ, Li JJ. Coactivation of ATM/ERK/NF-kappaB in the low-dose radiation-induced radioadaptive response in human skin keratinocytes. Free Radic Biol Med. 2009; 46(11): 1543-1550.

[40]

Wang J, Wang Y, Wang Y, Ma Y, Lan Y, Yang X. Transforming growth factor β-regulated microRNA-29a promotes angiogenesis through targeting the phosphatase and tensin homolog in endothelium. J Biol Chem. 2013; 288(15): 10418-10426.

[41]

Paroo Z, Ye X, Chen S, Liu Q. Phosphorylation of the human microRNA-generating complex mediates MAPK/Erk signaling. Cell. 2009; 139(1): 112-122.

[42]

Li JH, Liu S, Zhou H, Qu LH, Yang JH. starBase v2.0: decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 2014; 42: D92-7.

[43]

Olena AF, Patton JG. Genomic organization of microRNAs. J Cell Physiol. 2010; 222(3): 540-545.

[44]

Wang JX, Gao J, Ding SL, et al. Oxidative modification of miR-184 enables it to target Bcl-xL and Bcl-w. Mol Cell. 2015; 59(1): 50-61.

[45]

Desgagné V, Bouchard L, Guérin R. microRNAs in lipoprotein and lipid metabolism: from biological function to clinical application. Clin Chem Lab Med. 2017; 55(5): 667-686.

[46]

Pfeifer M, Grau M, Lenze D, et al. PTEN loss defines a PI3K/AKT pathway-dependent germinal center subtype of diffuse large B-cell lymphoma. Proc Natl Acad Sci USA. 2013; 110: 12420-12425.

[47]

Sheng S, Qiao M, Pardee AB. Metastasis and AKT activation. J Cell Physiol. 2009; 218: 451-454.

[48]

Xu QL, Li H, Zhu YJ, Xu G. The treatments and postoperative complications of esophageal cancer: a review. J Cardiothorac Surg. 2020; 15: 163.

[49]

Yang X, Li Y, Zou L, Zhu Z. Role of exosomes in crosstalk between cancer-associated fibroblasts and cancer cells. Front Oncol. 2019; 9: 356.

[50]

Ha SY, Yeo SY, Xuan YH, Kim SH. The prognostic significance of cancer-associated fibroblasts in esophageal squamous cell carcinoma. PLoS ONE. 2014; 9: e99955.

[51]

Chen Y, Li X, Yang H, et al. Expression of basic fibroblast growth factor, CD31, and α-smooth muscle actin and esophageal cancer recurrence after definitive chemoradiation. Tumour Biol. 2014; 35: 7275-7282.

[52]

Barcellos-Hoff MH, Ravani SA. Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Res. 2000; 60: 1254-1260.

[53]

Ohuchida K, Mizumoto K, Murakami M, et al. Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res. 2004; 64: 3215-3222.

[54]

Kamochi N, Nakashima M, Aoki S, et al. Irradiated fibroblast-induced bystander effects on invasive growth of squamous cell carcinoma under cancer-stromal cell interaction. Cancer Sci. 2008; 99: 2417-2427.

[55]

Tsai KK, Stuart J, Chuang YY, Little JB, Yuan ZM. Low-dose radiation-induced senescent stromal fibroblasts render nearby breast cancer cells radioresistant. Radiat Res. 2009; 172: 306-313.

[56]

Patel ZS, Grugan KD, Rustgi AK, Cucinotta FA, Huff JL. Ionizing radiation enhances esophageal epithelial cell migration and invasion through a paracrine mechanism involving stromal-derived hepatocyte growth factor. Radiat Res. 2012; 177: 200-208.

[57]

Chargari C, Clemenson C, Martins I, Perfettini JL, Deutsch E. Understanding the functions of tumor stroma in resistance to ionizing radiation: emerging targets for pharmacological modulation. Drug Resist Updat. 2013; 16: 10-21.

[58]

Bao CH, Wang XT, Ma W, et al. Irradiated fibroblasts promote epithelial-mesenchymal transition and HDGF expression of esophageal squamous cell carcinoma. Biochem Biophys Res Commun. 2015; 458: 441-447.

[59]

Li D, Qu C, Ning Z, et al. Radiation promotes epithelial-to-mesenchymal transition and invasion of pancreatic cancer cell by activating carcinoma-associated fibroblasts. Am J Cancer Res. 2016; 6: 2192-2206.

[60]

Gupta A, Probst HC, Vuong V, et al. Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. J Immunol. 2012; 189(2): 558-566.

[61]

Klug F, Prakash H, Huber PE, et al. Low-dose irradiation programs macrophage differentiation to an iNOS⁺/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell. 2013; 24(5): 589-602.

[62]

Sheng Y, Zhang B, Xing B, et al. Cancer-associated fibroblasts exposed to high-dose ionizing radiation promote M2 polarization of macrophages, which induce radiosensitivity in cervical cancer. Cancers (Basel). 2023; 15(5): 1620.

[63]

Ma T, Li H, Yang W, Liu Q, Yan H. Over-expression of miR-193a-3p regulates the apoptosis of colorectal cancer cells by targeting PAK3. Am J Transl Res. 2022; 14: 1361-1375.

[64]

Liu X, Min S, Wu N, et al. miR-193a-3p inhibition of the Slug activator PAK4 suppresses non-small cell lung cancer aggressiveness via the p53/Slug/L1CAM pathway. Cancer Lett. 2019; 447: 56-65.

[65]

Yu M, Liu Z, Liu Y, et al. PTP1B markedly promotes breast cancer progression and is regulated by miR-193a-3p. FEBS J. 2019; 286: 1136-1153.

[66]

Lv L, Li Y, Deng H, et al. MiR-193a-3p promotes the multi-chemoresistance of bladder cancer by targeting the HOXC9 gene. Cancer Lett. 2015; 357: 105-113.

[67]

Yi Y, Chen J, Jiao C, et al. Upregulated miR-193a-3p as an oncogene in esophageal squamous cell carcinoma regulating cellular proliferation, migration and apoptosis. Oncol Lett. 2016; 12: 4779-4784.

[68]

Xue J, Xiao P, Yu X, Zhang X. A positive feedback loop between AlkB homolog 5 and miR-193a-3p promotes growth and metastasis in esophageal squamous cell carcinoma. Hum Cell. 2021; 34: 502-514.

[69]

Meng F, Qian L, Lv L, et al. miR-193a-3p regulation of chemoradiation resistance in oesophageal cancer cells via the PSEN1 gene. Gene. 2016; 579: 139-145.

[70]

Liu A, Zhu Y, Chen W, Merlino G, Yu Y. PTEN dual lipid- and protein-phosphatase function in tumor progression. Cancers (Basel). 2022; 14: 3666.

[71]

Zhu Z, Yu W, Fu X, et al. Phosphorylated Akt1 is associated with poor prognosis in esophageal squamous cell carcinoma. J Exp Clin Cancer Res. 2015; 34: 95.

[72]

Saenz-Pipaon G, Dichek DA. Targeting and delivery of microRNA-targeting antisense oligonucleotides in cardiovascular diseases. Atherosclerosis. 2023; 374: 44-54.

[73]

Dhuri K, Bechtold C, Quijano E, et al. Antisense oligonucleotides: an emerging area in drug discovery and development. J Clin Med. 2020; 9(6): 2004.

[74]

Okamoto K, Murawaki Y. The therapeutic potential of RNA interference: novel approaches for cancer treatment. Curr Pharm Biotechnol. 2012; 13(11): 2235-2247.

[75]

Zhang Z, Huang Y, Li J, et al. Antitumor activity of anti-miR-75. Delivered through lipid nanoparticles. Adv Healthc Mater. 2023; 12(6): e2202412.

[76]

Griveau A, Bejaud J, Anthiya S, Avril S, Autret D, Garcion E. Silencing of miR-21 by locked nucleic acid-lipid nanocapsule complexes sensitize human glioblastoma cells to radiation-induced cell death. Int J Pharm. 2013; 454(2): 765-774.

[77]

Alix-Panabières C, Pantel K. Clinical applications of circulating tumor cells and circulating tumor DNA as liquid biopsy. Cancer Discov. 2016; 6: 479-491.

[78]

Liu T, Du LT, Wang YS, et al. Development of a novel serum exosomal microRNA nomogram for the preoperative prediction of lymph node metastasis in esophageal squamous cell carcinoma. Front Oncol. 2020; 10: 573501.

[79]

Luo A, Zhou X, Shi X, et al. Exosome-derived miR-339-5p mediates radiosensitivity by targeting Cdc25A in locally advanced esophageal squamous cell carcinoma. Oncogene. 2019; 38: 4990-5006.

[80]

Chen F, Xu B, Li J, et al. Hypoxic tumour cell-derived exosomal miR-340-5p promotes radioresistance of oesophageal squamous cell carcinoma via KLF10. J Exp Clin Cancer Res. 2021; 40: 38.

[81]

Wang WT, Guo CQ, Cui GH, Zhao S. Correlation of plasma miR-21 and miR-93 with radiotherapy and chemotherapy efficacy and prognosis in patients with esophageal squamous cell carcinoma. World J Gastroenterol. 2019; 25(37): 5604-5618.

[82]

Lawson JD, Otto K, Grist W, Johnstone PA. Frequency of esophageal stenosis after simultaneous modulated accelerated radiation therapy and chemotherapy for head and neck cancer. Am J Otolaryngol. 2008; 29: 13-19.

[83]

Ghosh-Laskar S, Mummudi N, Kumar S, et al. Definitive radiation therapy with dose escalation is beneficial for patients with squamous cell cancer of the esophagus. J Cancer Res Ther. 2022; 18: S285-S292.

[84]

Guo W, Zhou B, Dou L, et al. Single-cell RNA sequencing and spatial transcriptomics of esophageal squamous cell carcinoma with lymph node metastases. Exp Mol Med. 2025; 57(1): 59-71.

[85]

Zheng S, Liu B, Guan X. The role of tumor microenvironment in invasion and metastasis of esophageal squamous cell carcinoma. Front Oncol. 2022; 12: 911285.

RIGHTS & PERMISSIONS

2025 The Author(s). Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

10

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/