留言板

尊敬的读者、作者、审稿人, 关于本刊的投稿、审稿、编辑和出版的任何问题, 您可以本页添加留言。我们将尽快给您答复。谢谢您的支持!

姓名
邮箱
手机号码
标题
留言内容
验证码

肠道菌群通过诱导上皮细胞-间充质转化参与肛瘘发病的机制探讨

叶倩倩 柳奕诚 罗春生 徐进 汪庆明

叶倩倩, 柳奕诚, 罗春生, 徐进, 汪庆明. 肠道菌群通过诱导上皮细胞-间充质转化参与肛瘘发病的机制探讨[J]. 中华全科医学, 2024, 22(1): 121-125. doi: 10.16766/j.cnki.issn.1674-4152.003346
引用本文: 叶倩倩, 柳奕诚, 罗春生, 徐进, 汪庆明. 肠道菌群通过诱导上皮细胞-间充质转化参与肛瘘发病的机制探讨[J]. 中华全科医学, 2024, 22(1): 121-125. doi: 10.16766/j.cnki.issn.1674-4152.003346
YE Qianqian, LIU Yicheng, LUO Chunsheng, XU Jin, WANG Qingming. Discussion on the mechanism of intestinal flora participating in the pathogenesis of anal fistula by inducing EMT[J]. Chinese Journal of General Practice, 2024, 22(1): 121-125. doi: 10.16766/j.cnki.issn.1674-4152.003346
Citation: YE Qianqian, LIU Yicheng, LUO Chunsheng, XU Jin, WANG Qingming. Discussion on the mechanism of intestinal flora participating in the pathogenesis of anal fistula by inducing EMT[J]. Chinese Journal of General Practice, 2024, 22(1): 121-125. doi: 10.16766/j.cnki.issn.1674-4152.003346

肠道菌群通过诱导上皮细胞-间充质转化参与肛瘘发病的机制探讨

doi: 10.16766/j.cnki.issn.1674-4152.003346
基金项目: 

上海申康医院发展中心临床三年行动计划资助项目 SHDC2020CR3094B

上海市青年科技英才扬帆计划资助项目 21YF1433300

上海市2023年度“科技创新行动计划”医学创新研究专项 23Y11921800

“十四五”中医特色专科和中医急诊能力提升项目 ZYTSZK1-8

详细信息
    通讯作者:

    汪庆明,E-mail:davidwqm@163.com

  • 中图分类号: R657.16

Discussion on the mechanism of intestinal flora participating in the pathogenesis of anal fistula by inducing EMT

  • 摘要: 肛瘘是肛肠疾病中的常见病、多发病,以肛周流脓、疼痛、瘙痒为主要症状,男性青壮年为主要发病人群。目前肛瘘发病机制尚不明确,治疗仍以手术为主,手术预后与术者对于内口的把握以及括约肌的保护有密切的关系。近年来,肠道菌群的研究被推上热门,其在多种疾病中的作用也逐渐被证实,已有研究表示菌群变化与肠道微环境的紊乱参与肛瘘的发生,具体表现在菌群丰度多样性的增加和肠源性细菌的感染。上皮细胞-间充质转化(EMT)是癌症等疾病重要的表型变化,随着研究的深入,已被证明参与肛瘘的发生发展,尤以克罗恩病多见。本文基于肛瘘解剖学特征和主流发病机制,通过梳理国内外相关研究,总结肠道菌群-EMT-肛瘘之间存在的相关联系,从肠道菌群与肛瘘、肠道菌群与EMT、EMT与肛瘘三个方面阐述肛瘘发病机制,并得出肛门瘘管的形成可能是由于肠道菌群变化影响肠黏膜屏障稳态,进而诱导EMT发生并参与炎症诱导纤维化和瘘管的发展,由于EMT与隐腺肛瘘的研究较少,此机制仍需在隐腺肛瘘中进一步验证。该机制提示未来可以通过调节患者肠道菌群,减轻炎症反应和干预EMT进程参与肛瘘的预防和治疗,以期为肛瘘提供非手术的治疗新途径,达到减轻患者诊疗痛苦的目的。

     

  • 图  1  肛瘘发病机制

    Figure  1.  Pathogenesis of anal fistula

    表  1  肠道菌群与EMT的相关研究

    Table  1.   The correlation between intestinal flora and EMT

    第一作者 发表年份 涉及指标 具体机制
    CHANDRAKESAN P[40] 2014 Wnt/β-catenin、Notch、TGF-β通路 肠道病原体通过诱导Wnt/β-catenin、Notch和TGF-β通路激活促进了EMT发生。
    WAN G S[38] 2018 IL-6、TNF-α 外周IL-6和TNF-α升高随后促进结直肠癌的EMT过程,并参与癌症的进展和转移。
    XU S Y[39] 2019 IL-6、TNF-α 肠道生态失调通过分泌IL-6和TNF-α刺激卵巢癌中肿瘤相关巨噬细胞活化,最终诱导EMT的发展。
    SONG X L[41] 2022 VEGFR2 菌群代谢物脱氧胆酸通过VEGFR2活化促进肿瘤血管生成拟态形成和EMT,进一步加剧了肠道癌变。
    下载: 导出CSV

    表  2  EMT与肛瘘的修改意见相关研究

    Table  2.   Study on modification of EMT and anal fistula

    第一作者 发表年份 涉及指标 具体机制
    RATTO C[11] 2016 IL-1β、IL-8 隐腺瘘中存在慢性炎症,瘘管两端的炎症模式不同,但都存在EMT。细胞因子IL-1β和IL-8可能在瘘管形成中发挥作用。
    BRUCKNER R S[48] 2021 CD3+CD8- T细胞、CD3+CD8+ T细胞、TNF-α CD3+CD8-和CD3+CD8+ T两个细胞亚群都以时间依赖性方式促进EMT相关基因表达和TNF-α的产生,从而在克罗恩病肛周瘘中起重要作用。
    OSMAN N H[47] 2023 Vimentin、Snail、α-SMA、E-cadherin、Vimentin、TGF-β1、miR-200b、Zeb-1、PERK、NF-κB E-cadherin和miR-200b仅在正常肛门黏膜中表达,PERK和NF-κB在瘘管中表达较强,Snail和α-SMA在克罗恩病患者中高表达,所有IBD患者Vimentin和TGF-β1均高表达。
    MCGREGOR C G C[46] 2023 MMP、TNF-α、IL-13、TGF-β 严重的肠道炎症导致细胞因子TNF-α的分泌,IL-13和TGF-β,TGF-β是最有效的EMT诱导剂。MMP的过表达利于瘘管的形成。
    下载: 导出CSV
  • [1] 中国医师协会肛肠医师分会临床指南工作委员会. 肛瘘诊治中国专家共识(2020版)[J]. 中华胃肠外科杂志, 2020, 23(12): 1123-1130. doi: 10.3760/cma.j.cn.441530-20200925-00537

    Clinical Guidelines Working Committee of the Anorectal Physician Branch of the Chinese Medical Doctor Association. Chinese expert consensus on the diagnosis and treatment of anal fistula (2020 edition)[J]. Chinese Journal of Gastroenterology, 2020, 23(12): 1123-1130. doi: 10.3760/cma.j.cn.441530-20200925-00537
    [2] 程议乐, 武永连, 李万里, 等. 国内肛肠疾病流行病学调查研究进展[J]. 中国肛肠病杂志, 2022, 42(6): 74-76. https://www.cnki.com.cn/Article/CJFDTOTAL-GCBZ202206031.htm

    CHENG Y L, WU Y L, LI W L, et al. Progress in domestic epidemiological investigation of anorectal diseases[J]. Chinese Journal of Coloproctology, 2022, 42(6): 74-76. https://www.cnki.com.cn/Article/CJFDTOTAL-GCBZ202206031.htm
    [3] PORTINCASA P, BONFRATE L, VACCA M, et al. Gut microbiota and short chain fatty acids: implications in glucose homeostasis[J]. Int J Mol Sci, 2022, 23(3): 1105. doi: 10.3390/ijms23031105
    [4] 卢燕, 刘占利. 肠道菌群和癫痫相关性研究进展[J]. 中华全科医学, 2018, 16(9): 1550-1553, 1569. doi: 10.16766/j.cnki.issn.1674-4152.000422

    LU Y, LIU Z L. Research progress on the correlation between intestinal flora and epileps[J]. Chinese Journal of General Practice, 2018, 16(9): 1550-1553, 1569. doi: 10.16766/j.cnki.issn.1674-4152.000422
    [5] SETTANNI C R, BIBBÒ S, IANIRO G, et al. Gastrointestinal involvement of autism spectrum disorder: focus on gut microbiota[J]. Expert Rev Gastroenterol Hepatol, 2021, 15(6): 599-622. doi: 10.1080/17474124.2021.1869938
    [6] LI Z Z, ZHANG B, WANG N, et al. A novel peptide protects against diet-induced obesity by suppressing appetite and modulating the gut microbiota[J]. Gut, 2023, 72(4): 686-698. doi: 10.1136/gutjnl-2022-328035
    [7] MATSON V, CHERVIN C S, GAJEWSKI T F. Cancer and the microbiome-influence of the commensal microbiota on cancer, immune responses, and immunotherapy[J]. Gastroenterology, 2021, 160(2): 600-613. doi: 10.1053/j.gastro.2020.11.041
    [8] CHOO J M, MURPHY K J, WADE A T, et al. Interactions between mediterranean diet supplemented with dairy foods and the gut microbiota influence cardiovascular health in an Australian population[J]. Nutrients, 2023, 15(16): 3645. doi: 10.3390/nu15163645
    [9] CAPARRÓS E, WIEST R, SCHARL M, et al. Dysbiotic microbiota interactions in Crohn's disease[J]. Gut Microbes, 2021, 13(1): 1949096. DOI: 10.1080/19490976.2021.1949096.
    [10] MOLINA-CASTRO S E, TIFFON C, GIRAUD J, et al. The hippo kinase LATS2 controls helicobacter pylori-induced epithelial-mesenchymal transition and intestinal metaplasia in gastric mucosa[J]. Cell Mol Gastroenterol Hepatol, 2020, 9(2): 257-276. doi: 10.1016/j.jcmgh.2019.10.007
    [11] RATTO C, LITTA F, LUCCHETTI D, et al. Immunopathological characterization of cryptoglandular anal fistula: a pilot study investigating its pathogenesis[J]. Colorectal Dis, 2016, 18(12): O436-O444. DOI: 10.1111/codi.13527.
    [12] 赵冬安, 史学文, 朱孟琳. 肛瘘发病机制的研究现状[J]. 中国肛肠病杂志, 2018, 38(2): 73-74. doi: 10.3969/j.issn.1000-1174.2018.02.031

    ZHAO D A, SHI X W, ZHU M L. Research status of the pathogenesis of anal fistula[J]. Chinese Journal of Coloproctology, 2018, 38(2): 73-74. doi: 10.3969/j.issn.1000-1174.2018.02.031
    [13] 陈伟, 郑雪平. 肛瘘的中西医病因病机(理)研究概况[J]. 中医药临床杂志, 2018, 30(5): 978-981. https://www.cnki.com.cn/Article/CJFDTOTAL-AHLC201805062.htm

    CHEN W, ZHENG X P. Overview of research on the etiology and pathogenesis of anal fistula in traditional Chinese and Western medicine[J]. Clinical Journal of Traditional Chinese Medicine, 2018, 30(5): 978-981. https://www.cnki.com.cn/Article/CJFDTOTAL-AHLC201805062.htm
    [14] JAISWAL P, SHARMA S, PRATAP A, et al. Significant presence of biofilm-producing gut-derived bacteria in anal fistula of chronic duration[J]. Int Wound J, 2021, 18(4): 519-524. doi: 10.1111/iwj.13551
    [15] LITTA F, PAPAIT A, LUCCHETTI D, et al. The pathogenesis of cryptoglandular anal fistula: new insight into the immunological profile[J]. Colorectal Dis, 2022, 24(12): 1567-1575. doi: 10.1111/codi.16290
    [16] SANCHEZ-HARO E, VELA E, CLERIES M, et al. Clinical characterization of patients with anal fistula during follow-up of anorectal abscess: a large population-based study[J]. Tech Coloproctol, 2023, 27(10): 897-907. doi: 10.1007/s10151-023-02840-z
    [17] 徐飞, 陈燕, 唐迎春, 等. 锌指蛋白521基因多态性对肛周脓肿合并感染患者创面愈合及预后的影响[J]. 中华医院感染学杂志, 2022, 32(19): 2974-2977. https://www.cnki.com.cn/Article/CJFDTOTAL-ZHYY202219020.htm

    XU F, CHEN Y, TANG Y C, et al. Effects of zinc finger protein 521 gene polymorphism on wound healing and prognosis in patients with perianal abscess and infection[J]. Chinese Journal of Nosocomiology, 2022, 32(19): 2974-2977. https://www.cnki.com.cn/Article/CJFDTOTAL-ZHYY202219020.htm
    [18] YAMADA S, KANDA Y. Evaluation of barrier functions in human iPSC-derived intestinal epithelium[J]. Methods Mol Biol, 2021, 2367: 27-35.
    [19] UNTERSMAYR E, BRANDT A, KOIDL L, et al. The intestinal barrier dysfunction as driving factor of inflammaging[J]. Nutrients, 2022, 14(5): 949. doi: 10.3390/nu14050949
    [20] 邢肖伟, 陶金华, 江曙, 等. 肠道菌群影响黏膜屏障结构与功能的研究进展[J]. 中国微生态学杂志, 2018, 30(6): 725-730. https://www.cnki.com.cn/Article/CJFDTOTAL-ZGWS201806026.htm

    XING X W, TAO J H, JIANG S, et al. Research progress on the influence of intestinal flora on mucosal barrier structure and function[J]. Chinese Journal of Microbiology, 2018, 30(6): 725-730. https://www.cnki.com.cn/Article/CJFDTOTAL-ZGWS201806026.htm
    [21] ROGERS A P, MILETO S J, LYRAS D. Impact of enteric bacterial infections at and beyond the epithelial barrier[J]. Nat Rev Microbiol, 2023, 21(4): 260-274. doi: 10.1038/s41579-022-00794-x
    [22] 李丹阳, 周瑶, 陶亮. 肠黏膜屏障与病原细菌感染[J]. 生理科学进展, 2022, 53(6): 416-421. doi: 10.3969/j.issn.0559-7765.2022.06.005

    LI D Y, ZHOU Y, TAO L. Intestinal mucosal barrier and pathogenic bacterial infection[J]. Progress in Physiological Sciences, 2022, 53(6): 416-421. doi: 10.3969/j.issn.0559-7765.2022.06.005
    [23] 王倩, 邹贤军. 克罗恩病肛瘘的外科诊治现状[J]. 临床外科杂志, 2023, 31(6): 521-523. https://www.cnki.com.cn/Article/CJFDTOTAL-LCWK202306006.htm

    WANG Q, ZOU X J. Current status of surgical diagnosis and treatment of anal fistula in Crohn's disease[J]. Journal of Clinical Surgery, 2023, 31(6): 521-523. https://www.cnki.com.cn/Article/CJFDTOTAL-LCWK202306006.htm
    [24] CAO S Y, COLONNA M, DEEPAK P. Pathogenesis of perianal fistulising Crohn's disease: current knowledge, gaps in understanding, and future research directions[J]. J Crohns Colitis, 2023, 17(6): 1010-1022. doi: 10.1093/ecco-jcc/jjad008
    [25] CHEN Y W, MAI Q D, CHEN Z X, et al. Dietary palmitoleic acid reprograms gut microbiota and improves biological therapy against colitis[J]. Gut Microbes, 2023, 15(1): 2211501. DOI: 10.1080/19490976.2023.2211501.
    [26] 万仁辉, 翟骁, 鲁瑾, 等. 嗜黏蛋白阿克曼菌与肥胖的关系[J]. 第二军医大学学报, 2019, 40(2): 203-208. https://www.cnki.com.cn/Article/CJFDTOTAL-DEJD201902014.htm

    WAN R H, ZHAI X, LU J, et al. The relationship between Akkermansia muciniphila and obesity[J]. Journal of Second Military Medical University, 2019, 40(2): 203-208. https://www.cnki.com.cn/Article/CJFDTOTAL-DEJD201902014.htm
    [27] WADE H, PAN K, DUAN Q H, et al. Akkermansia muciniphila and its membrane protein ameliorates intestinal inflammatory stress and promotes epithelial wound healing via CREBH and miR-143/145[J]. J Biomed Sci, 2023, 30(1): 38. doi: 10.1186/s12929-023-00935-1
    [28] 杜佳琦, 张卫刚. 探讨肠道菌群与肛瘘的相关性[J]. 上海中医药杂志, 2017, 51(S1): 8-9. https://www.cnki.com.cn/Article/CJFDTOTAL-SHZZ2017S1003.htm

    DU J Q, ZHANG W G. Exploring the correlation between intestinal flora and anal fistula[J]. Shanghai Journal of Traditional Chinese Medicine, 2017, 51(S1): 8-9. https://www.cnki.com.cn/Article/CJFDTOTAL-SHZZ2017S1003.htm
    [29] 梅婷婷. 肛瘘患者肠道微生物多样性的检测和分析[D]. 杭州: 浙江中医药大学, 2021.

    MEI T T. Detection and analysis of intestinal microbial diversity in patients with anal fistula[D]. Hangzhou: Zhejiang University of Traditional Chinese Medicine, 2021.
    [30] 裘建明, 杨关根, 王东, 等. 复杂性肛瘘患者肠道微生态检测及分析[J]. 中华胃肠外科杂志, 2022, 25(9): 792-797. doi: 10.3760/cma.j.cn441530-20220412-00142

    QIU J M, YANG G G, WANG D, et al. Detection and analysis of intestinal microecology in patients with complex anal fistula[J]. Chinese Journal of Gastroenterology, 2022, 25(9): 792-797. doi: 10.3760/cma.j.cn441530-20220412-00142
    [31] 黄进, 褚怡霏, 汪庆明, 等. 基于16S rDNA测序分析湿热下注型肛瘘的局部菌群[J]. 现代医学, 2022, 50(6): 731-735. https://www.cnki.com.cn/Article/CJFDTOTAL-TDYX202206013.htm

    HUANG J, CHU Y F, WANG Q M, et al. Analysis of local bacterial flora of moist-heat injection-type anal fistula based on 16S rDNA sequencing[J]. Modern Medicine, 2022, 50(6): 731-735. https://www.cnki.com.cn/Article/CJFDTOTAL-TDYX202206013.htm
    [32] 朱静怡, 张玲, 柳奕诚, 等. 黄连解毒汤对湿热下注型肛瘘患者肠道菌群的影响[J]. 上海中医药大学学报, 2022, 36(4): 12-18, 25. https://www.cnki.com.cn/Article/CJFDTOTAL-SHZD202204002.htm

    ZHU J Y, ZHANG L, LIU Y C, et al. Effect of Huanglian Jiedu Decoction on intestinal flora in patients with damp-heat anal fistula[J]. Journal of Shanghai University of Traditional Chinese Medicine, 2022, 36(4): 12-18, 25. https://www.cnki.com.cn/Article/CJFDTOTAL-SHZD202204002.htm
    [33] 刘道成, 陈金明, 杨关根, 等. Vimentin表达水平与肛瘘感染细菌谱以及患者临床特征的关系研究[J]. 结直肠肛门外科, 2019, 25(1): 43-47. https://www.cnki.com.cn/Article/CJFDTOTAL-DCGM201901013.htm

    LIU D C, CHEN J M, YANG G G, et al. Study on the relationship between vimentin expression level, bacterial spectrum of anal fistula infection and clinical characteristics of patients[J]. Colorectal and anal surgery, 2019, 25(1): 43-47. https://www.cnki.com.cn/Article/CJFDTOTAL-DCGM201901013.htm
    [34] CAI P, RONG H, ZHU Q Q, et al. The potential roles of gut microbiome in anal fistula[J]. AMB Express, 2023, 13(1): 58. doi: 10.1186/s13568-023-01560-9
    [35] WŁODARCZYK M, WŁODARCZYK J, SOBOLEWSKA-WŁODAR CZYK A, et al. Current concepts in the pathogenesis of cryptoglandular perianal fistula[J]. J Int Med Res, 2021, 49(2): 30006052098 6669. DOI: 10.1177/0300060520986669.
    [36] SUGRUE J, NORDENSTAM J, ABCARIAN H, et al. Pathogenesis and persistence of cryptoglandular anal fistula: a systematic review[J]. Tech Coloproctol, 2017, 21(6): 425-432.
    [37] JAISWAL P, SHARMA S, PRATAP A, et al. Significant presence of biofilm-producing gut-derived bacteria in anal fistula of chronic duration[J]. Int Wound J, 2021, 18(4): 519-524.
    [38] WAN G S, XIE M L, YU H J, et al. Intestinal dysbacteriosis activates tumor-associated macrophages to promote epithelial-mesenchymal transition of colorectal cancer[J]. Innate Immun, 2018, 24(8): 480-489.
    [39] XU S Y, LIU Z Z, LV M H, et al. Intestinal dysbiosis promotes epithelial-mesenchymal transition by activating tumor-associated macrophages in ovarian cancer[J]. Pathog Dis, 2019, 77(2): ftz019. DOI: 10.1093/femspd/ftz019.
    [40] CHANDRAKESAN P, ROY B, JAKKULA L U, et al. Utility of a bacterial infection model to study epithelial-mesenchymal transition, mesenchymal-epithelial transition or tumorigenesis[J]. Oncogene, 2014, 33(20): 2639-2654.
    [41] SONG X L, AN Y P, CHEN D F, et al. Microbial metabolite deoxycholic acid promotes vasculogenic mimicry formation in intestinal carcinogenesis[J]. Cancer Sci, 2022, 113(2): 459-477.
    [42] WARYAH C, CURSONS J, FOROUTAN M, et al. Synthetic epigenetic reprogramming of mesenchymal to epithelial states using the CRISPR/dCas9 platform in triple negative breast cancer[J]. Adv Sci (Weinh), 2023, 10(22): e2301802. DOI: 10.1002/advs.202301802.
    [43] DONGRE A, WEINBERG R A. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer[J]. Nat Rev Mol Cell Biol, 2019, 20(2): 69-84.
    [44] ZHANG N, NG A S, CAI S J, et al. Novel therapeutic strategies: targeting epithelial-mesenchymal transition in colorectal cancer[J]. Lancet Oncol, 2021, 22(8): e358-e368.
    [45] JAYACHANDRAN J, SRINIVASAN H, MANI K P. Molecular mechanism involved in epithelial to mesenchymal transition[J]. Arch Biochem Biophys, 2021, 710: 108984. DOI: 10.1016/j.abb.2021.108984.
    [46] MCGREGOR C G C, TANDON R, SIMMONS A. Pathogenesis of fistulating Crohn's disease: a review[J]. Cell Mol Gastroenterol Hepatol, 2023, 15(1): 1-11.
    [47] OSMAN N H, JAILANI R F, RAHMAN H A, et al. Gene and protein expression of epithelial to mesenchymal transition for intestinal and anal fistula: a systematic review[J]. Ann Coloproctol, 2023, 39(2): 106-114.
    [48] BRUCKNER R S, SPALINGER M R, BARNHOORN M C, et al. Contribution of CD3+CD8- and CD3+CD8+ T cells to TNF-α overexpression in Crohn disease-associated perianal fistulas and induction of epithelial-mesenchymal transition in HT-29 cells[J]. Inflamm Bowel Dis, 2021, 27(4): 538-549.
  • 加载中
图(1) / 表(2)
计量
  • 文章访问数:  68
  • HTML全文浏览量:  33
  • PDF下载量:  5
  • 被引次数: 0
出版历程
  • 收稿日期:  2023-07-28
  • 网络出版日期:  2024-03-09

目录

    /

    返回文章
    返回