自噬在菌群相关肿瘤中的研究进展

王宇 刘超 杜江 周旋

王宇, 刘超, 杜江, 周旋. 自噬在菌群相关肿瘤中的研究进展[J]. 中国肿瘤临床, 2022, 49(1): 37-41. doi: 10.12354/j.issn.1000-8179.2022.20211224
引用本文: 王宇, 刘超, 杜江, 周旋. 自噬在菌群相关肿瘤中的研究进展[J]. 中国肿瘤临床, 2022, 49(1): 37-41. doi: 10.12354/j.issn.1000-8179.2022.20211224
Yu Wang, Chao Liu, Jiang Du, Xuan Zhou. Research progress of autophagy in microbiota-associated tumors[J]. CHINESE JOURNAL OF CLINICAL ONCOLOGY, 2022, 49(1): 37-41. doi: 10.12354/j.issn.1000-8179.2022.20211224
Citation: Yu Wang, Chao Liu, Jiang Du, Xuan Zhou. Research progress of autophagy in microbiota-associated tumors[J]. CHINESE JOURNAL OF CLINICAL ONCOLOGY, 2022, 49(1): 37-41. doi: 10.12354/j.issn.1000-8179.2022.20211224

自噬在菌群相关肿瘤中的研究进展

doi: 10.12354/j.issn.1000-8179.2022.20211224
基金项目: 本文课题受国家自然科学基金面上项目 (编号:82002892,81872206,82073010)和天津市教委资助项目 (编号:2019KJ188)资助
详细信息
    作者简介:

    王宇:专业方向为头颈肿瘤临床和基础转化性研究

    通讯作者:

    周旋 xuanzhou@tmu.edu.cn

Research progress of autophagy in microbiota-associated tumors

Funds: This work was supported by the National Natural Science Foundation of China General Project (No.82002892,No. 81872206,No. 82073010), Tianjin Municipal Education Commission Support Grant (No. 2019KJ188)
More Information
  • 摘要: 自噬是真核生物中高度保守的分解代谢过程,其实质是溶酶体依赖的蛋白质降解途径。自噬调节异常与许多肿瘤的发生发展密切相关,其既能促进肿瘤存活,亦能促进肿瘤消亡。研究表明菌群失调可通过促进黏膜炎症或引起全身疾病直接或间接导致肿瘤发生发展,并在调节免疫系统对肿瘤的反应中发挥作用。菌群失衡和自噬过程可通过多种分子机制联系在一起并通过这些途径影响肿瘤进展。本文旨在对自噬在菌群相关肿瘤中的关键作用进行综述,并探讨了自噬调节这一复杂生物学过程的分子机制;此外,本文还分析了以菌群/自噬生物轴为靶点的多种分子药物治疗肿瘤的可能性,对以自噬或菌群为靶点的临床试验进行总结并针对其作为抗癌策略的治疗潜力进行评价。

     

  • [1] Li XH, He SK, Ma BY. Autophagy and autophagy-related proteins in cancer[J]. Mol Cancer, 2020, 19(1):12. doi: 10.1186/s12943-020-1138-4
    [2] Onorati AV, Dyczynski M, Ojha R, et al. Targeting autophagy in cancer[J]. Cancer, 2018, 124(16):3307-3318. doi: 10.1002/cncr.31335
    [3] Garrettw S. Cancer and the microbiota[J]. Science, 2015, 348(6230):80-86.
    [4] Cho TJ, Wee SW, Woo VH, et al. Porphyromonas gingivalis-induced autophagy suppresses cell proliferation through G1 arrest in oral cancer cells[J]. Arch Oral Biol, 2014, 59(4):370-378. doi: 10.1016/j.archoralbio.2014.01.001
    [5] Liu Y, Baba Y, Ishimoto T, et al. Fusobacterium nucleatum confers chemoresistance by modulating autophagy in oesophageal squamous cell carcinoma[J]. Br J Cancer, 2021, 124(5):963-974. doi: 10.1038/s41416-020-01198-5
    [6] Liu S, Zhou X, Peng X, et al. Porphyromonas gingivalis promotes immunoevasion of oral cancer by protecting cancer from macrophage attack[J]. J Immunol, 2020, 205(1):282-289. doi: 10.4049/jimmunol.1901138
    [7] Si H, Yang Q, Hu H, et al. Colorectal cancer occurrence and treatment based on changes in intestinal flora[J]. Semin Cancer Biol, 2021, 70:3-10. doi: 10.1016/j.semcancer.2020.05.004
    [8] Gao L, Xu TS, Huang G, et al. Oral microbiomes: more and more importance in oral cavity and whole body[J]. Protein Cell, 2018, 9(5):488-500. doi: 10.1007/s13238-018-0548-1
    [9] Qi YJ, Jiao YL, Chen P, et al. Porphyromonas gingivalis promotes progression of esophageal squamous cell cancer via TGFβ-dependent Smad/YAP/TAZ signaling[J]. PLoS Biol, 2020, 18(9):e3000825. doi: 10.1371/journal.pbio.3000825
    [10] Yamamura K, Baba Y, Nakagawa S, et al. Human microbiome Fusobacterium nucleatum in esophageal cancer tissue is associated with prognosis[J]. Clin Cancer Res, 2016, 22(22):5574-5581. doi: 10.1158/1078-0432.CCR-16-1786
    [11] Chen XH, Wang A, Chu AN, et al. Mucosa-associated microbiota in gastric cancer tissues compared with non-cancer tissues[J]. Front Microbiol, 2019, 10:1261. doi: 10.3389/fmicb.2019.01261
    [12] Wu J, Xu S, Xiang CJ, et al. Tongue coating microbiota community and risk effect on gastric cancer[J]. J Cancer, 2018, 9(21):4039-4048. doi: 10.7150/jca.25280
    [13] Castellarin M, Warren RL, Freeman JD, et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma[J]. Genome Res, 2012, 22(2):299-306. doi: 10.1101/gr.126516.111
    [14] Shen JJ, Zhai J, You Q, et al. Cancer-associated fibroblasts-derived VCAM1 induced by H. pylori infection facilitates tumor invasion in gastric cancer[J]. Oncogene, 2020, 39(14):2961-2974. doi: 10.1038/s41388-020-1197-4
    [15] Sears CL, Garrett WS. Microbes, microbiota, and colon cancer[J]. Cell Host Microbe, 2014, 15(3):317-328. doi: 10.1016/j.chom.2014.02.007
    [16] Amaravadi R, KimmelmanAC, White E. Recent insights into the function of autophagy in cancer[J]. Genes Dev, 2016, 30(17):1913-1930. doi: 10.1101/gad.287524.116
    [17] Amaravadi RK, KimmelmanAC, Debnath J. Targeting autophagy in cancer: recent advances and future directions[J]. Cancer Discov, 2019, 9(9):1167-1181. doi: 10.1158/2159-8290.CD-19-0292
    [18] Yun CW, Lee SH. The roles of autophagy in cancer[J]. Int J Mol Sci, 2018, 19(11):3466. doi: 10.3390/ijms19113466
    [19] Wang K, DuBY, Xu B, et al. JMJD6-STAT3Y705ph axis promotes autophagy in osteosarcoma cancer cells by regulating ATG[J]. Biofactors, 2020, 46(5):839-848.
    [20] Liu L, Yang M, Kang R, et al. HMGB1-induced autophagy promotes chemotherapy resistance in leukemia cells[J]. Leukemia, 2011, 25(1):23-31. doi: 10.1038/leu.2010.225
    [21] Cai J, Li R, Xu X, et al. CK1α suppresses lung tumour growth by stabilizing PTEN and inducing autophagy[J]. Nat Cell Biol, 2018, 20(4):465-478. doi: 10.1038/s41556-018-0065-8
    [22] Raju D, Hussey S, Ang M, et al. Vacuolating cytotoxin and variants in Atg16L1 that disrupt autophagy promote Helicobacter pylori infection in humans[J]. Gastroenterology, 2012, 142(5):1160-1171. doi: 10.1053/j.gastro.2012.01.043
    [23] Eslami M, Yousefi B, Kokhaei P, et al. Current information on the association of Helicobacter pylori with autophagy and gastric cancer[J]. J Cell Physiol, 2019, 234(10):17127-17143. doi: 10.1002/jcp.28473
    [24] Xie C, Li NS, Wang H, et al. Inhibition of autophagy aggravates DNA damage response and gastric tumorigenesis via Rad51 ubiquitination in response to H. pylori infection[J]. Gut Microbes, 2020, 11(6):1567-1589. doi: 10.1080/19490976.2020.1774311
    [25] Ponzetto A, Figura N. Colon cancer risk and vacA toxin of Helicobacter pylori[J]. Gastroenterology, 2019, 156(8):2356.
    [26] Mima K, Nishihara R, Qian ZR, et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis[J]. Gut, 2016, 65(12):1973-1980. doi: 10.1136/gutjnl-2015-310101
    [27] Li MH, Chen WD, Wang YD. The roles of the gut microbiota-miRNA interaction in the host pathophysiology[J]. Mol Med, 2020, 26(1):101. doi: 10.1186/s10020-020-00234-7
    [28] Shi CS, Kehrl JH. MyD88 and trif target beclin 1 to trigger autophagy in macrophages[J]. J Biol Chem, 2008, 283(48):33175-33182. doi: 10.1074/jbc.M804478200
    [29] Erkisa M, Aydinlik S, Cevatemre B, et al. A promising therapeutic combination for metastatic prostate cancer: Chloroquine as autophagy inhibitor and palladium(Ⅱ) barbiturate complex[J]. Biochimie, 2020, 175:159-172. doi: 10.1016/j.biochi.2020.05.010
    [30] Jiang RY, Pei HL, Gu WD, et al. Autophagic inhibitor attenuates rapamycin-induced inhibition of proliferation in cultured A549 lung cancer cells[J]. Eur Rev Med Pharmacol Sci, 2014, 18(6):806-810.
    [31] Liu WD, Sun W, Hua YQ, et al. Effect of rapamycin and chloroquine on osteosarcoma[J]. Chin J Oncol, 2017, 97(19):1510-1514.
    [32] Qi L, Ye JW, Xue WH, et al. The mechanism of aspirin combined with metformin induced apoptosis of thyroid cancer TPC-1 cells[J]. Chin J Oncol, 2019, 41(4):276-281.
    [33] Hu R, Li T, Hui KY, et al. Apatinib sensitizes chemoresistant NSCLC cells to doxetaxel via regulating autophagy and enhances the therapeutic efficacy in advanced and refractory/recurrent NSCLC[J]. Mol Med Rep, 2020, 22(5):3935-3943.
    [34] Ostenfeld MS, Høyer-Hansen M, Bastholm L, et al. Anti-cancer agent siramesine is a lysosomotropic detergent that induces cytoprotective autophagosome accumulation[J]. Autophagy, 2008, 4(4):487-499. doi: 10.4161/auto.5774
  • 加载中
计量
  • 文章访问数:  121
  • HTML全文浏览量:  23
  • PDF下载量:  26
  • 被引次数: 0
出版历程
  • 收稿日期:  2021-08-01
  • 网络出版日期:  2022-02-11

目录

    /

    返回文章
    返回