肠道菌群在肠易激综合征治疗中的研究进展
Research Progress on Gut Microbiota in the Treatment of Irritable Bowel Syndrome
DOI: 10.12677/TCM.2024.131002, PDF, HTML, XML, 下载: 111  浏览: 202 
作者: 王思颖:黑龙江中医药大学,第一临床医学院,黑龙江 哈尔滨
关键词: 肠易激综合征肠道菌群粪菌移植低FODMAPs饮食Irritable Bowel Syndrome Gut Microbiota Fecal Microbiota Transplantation Low FODMAPs Diet
摘要: 肠易激综合征(IBS)是临床常见的功能性胃肠病(FGIDs)之一,由于目前没有完全理解其发病机制,治疗方向以缓解症状为主,而肠道菌群在肠易激综合征的治疗中日益受到关注。近期研究揭示了肠道菌群与肠易激综合征之间的紧密联系,菌群失调可能引发免疫和神经系统的异常,进而导致肠易激综合征症状的发生。治疗方面,针对菌群的调整成为一项重要策略,以恢复肠道微生态平衡。本文阐述了肠道菌群在肠易激综合征治疗中的研究进展。
Abstract: Irritable Bowel Syndrome (IBS) is a common functional gastrointestinal disorder (FGID) in clinical practice. Due to the incomplete understanding of its pathogenesis, the treatment approach mainly focuses on symptom relief. Recently, there has been increasing attention on the role of gut microbi-ota in the treatment of Irritable Bowel Syndrome. Recent research has revealed a close connection between gut microbiota and Irritable Bowel Syndrome, suggesting that dysbiosis may trigger ab-normalities in the immune and nervous systems, leading to the occurrence of IBS symptoms. In terms of treatment, adjusting the microbiota has become a crucial strategy to restore the balance of the gut microbiota. This article elaborates on the research progress of gut microbiota in the treat-ment of Irritable Bowel Syndrome.
文章引用:王思颖. 肠道菌群在肠易激综合征治疗中的研究进展[J]. 中医学, 2024, 13(1): 9-14. https://doi.org/10.12677/TCM.2024.131002

1. 引言

肠易激综合征(IBS)是一种功能性胃肠病,其特征包括腹痛或腹部不适,伴随着排便习惯的变化,如便秘、腹泻或交替出现。这一症状集合通常持续数月,对患者的生活质量产生显著影响,但无明显的器质性异常或病理学上的特征 [1] 。目前最为常见的IBS诊断标准为罗马IV标准,依据临床症状的不同将IBS细分为四种亚型,分别是便秘型(IBS-C)、腹泻型(IBS-D)、混合型(IBS-M)和不定型(IBS-U) [2] 。罗马基金会通过互联网调查了来自26个国家的73,000多人,这项于2020年发表的流行病学报告显示,依据罗马IV标准的国家中IBS发病率在1.3%~7.6%,在功能性肠病中占比为4.1%;而在同时使用罗马III和罗马IV标准的国家,按照罗马III标准,IBS的发病率为10.1%,按照罗马IV标准为3.8% [3] 。

2. IBS的发病机制

IBS的发病涉及社会心理学、遗传学、神经学和免疫学等,其病理生理机制十分复杂,确切病因尚不清楚,但毋庸置疑有多种因素参与其中,包括肠道运动的异常、肠道敏感度增加、神经–内分泌–免疫系统的失衡以及心理社会因素 [4] [5] [6] 。随着对脑–肠交互作用理解逐渐加深,人们提出了脑–肠轴这一概念,它整合了大脑和肠道之间的神经、激素和免疫信号,并为肠道微生物菌群及其代谢物提供了一条潜在的进入大脑的途径 [7] 。由于脑–肠交流系统的双向性,不仅大脑能够影响胃肠功能以及免疫功能,胃肠道功能出现异常亦会作用于大脑。3项来自不同国家的流行病学报告显示,约半数IBS患者首先出现胃肠道症状,而后才发展为情绪障碍 [8] [9] [10] ,这表明原发性肠道疾病可能是导致部分患者出现情绪障碍的潜在因素。

近年来,随着微生态学和基因测序技术的不断进步,对肠道菌群的研究变得更加精密。由于肠道菌群的数量庞大、种类繁多,以及对机体生理和病理过程的不可或缺的影响,它在各种疾病的发病机制中受到广泛探讨。在这一趋势下,人们对肠道菌群与IBS之间的关系也投入了更多关注。在生理状态下,数量庞大的肠道菌群具有保护肠道粘膜完整性、防止病原体侵入、调节免疫系统、改善胃肠道功能和产生代谢产物等主要作用 [11] [12] [13] ,这些微生物与宿主之间保持着和谐共生的关系;而病理状态下,肠道菌群失调,有益菌与有害菌的平衡被打乱,这种改变不仅会导致肠道局部病变,还会波及人体多个系统,是导致包括IBS在内许多疾病发生发展的重要因素。

3. IBS的常见治疗方案

在治疗方案上,由于疾病机制不明确和病因的复杂多样,IBS主要是对症治疗,如何改善症状一直是各项研究的重点方向。目前临床一线治疗包括调整饮食结构、食用可溶性纤维、服用抗痉挛药物等。而对于症状严重的患者,治疗中可应用三环类抗抑郁药、肠道促分泌剂、作用于阿片或5-羟色胺受体的药物、抗生素和心理疗法 [14] [15] 。然而,药物干预并不适用于所有患者,且其中部分药物的副作用也十分明显,包括头痛、头晕、口干和失眠等 [16] ,甚至可能会发生严重的不良事件,如心血管疾病和缺血性结肠炎 [17] 。尽管心理干预(CBT、冥想等)没有任何严重副作用,但很难持续地进行长期治疗 [18] 。现阶段IBS的治疗方案仍有局限性,需要多种干预措施相结合以达到更好的疗效,因此,通过调节肠道菌群治疗肠易激综合征的方法也得到了越来越多的研究。

4. IBS肠道菌群与健康菌群

以前的研究认为,健康人与患有IBS的个体在肠道微生物方面存在显著差异 [19] [20] ,然而,最近的一些证据表明,虽然肠道微生物可以用于区分不同类型的IBS,但健康对照组与IBS患者在微生物组成方面并没有显著差异 [21] [22] [23] ,只有IBS的症状严重程度与肠道微生物失调有关 [23] 。在一项随机招募的符合罗马IV标准的瑞典人群研究中,基于376例乙状结肠活检标本(63例IBS病例)和185例粪便样本(32例IBS病例)研究IBS的特征菌群,结论仍旧是在IBS患者中未发现明显的微生物特征 [24] 。但也有研究者指出,这项研究的IBS病例数量太少,因此可能会存在统计方面的问题,正如以前报告中提到微生物群结构随着IBS严重程度变化而变化,小样本量和相对较轻的症状可能是未检测到IBS特征菌群的原因之一 [25] 。

尽管上述报告存在着各种互相矛盾的结论,仍然无法否定肠道菌群在IBS发病机制中的重要性。有研究应用抗生素、感染或内毒素注射的方式作用于肠道菌群诱导了内脏过敏 [26] ,有研究将IBS患者的粪便微生物群转移到小鼠身上,导致其内脏过敏 [27] ,并且出现肠道功能和行为的改变 [28] ,这一从人到小鼠的转移从侧面支持了以肠道菌群为导向治疗IBS的可行性。

5. 粪菌移植

粪菌移植(FMT)是一种通过重建肠道菌群来治疗疾病的方法,有随机对照试验证明FMT治疗IBS的疗效确切 [29] [30] ,疗效在接受移植3个月后最为显著,到12个月再随访时效果则不明显 [29] ,在该实验基础上进行元基因组测序,观察到经FMT有良好反应的患者肠道微生物组成与粪便样本捐赠者的相似性增加,这表明FMT可能引发肠道菌群的长期变化 [31] 。另有一项在165名IBS患者中进行的随机对照实验,通过较大的样本量、包括了三种IBS亚型、以及单一明确的粪便捐赠者,同样证实了FMT的疗效 [30] 。值得一提的是,该研究团队进行了为期3年的随访,证明了FMT疗法的长期效果以及安全性。研究中发现,FMT组患者经治疗后生物失调指数(DI)降低,且DI与IBS-SSS和Fas总分呈正相关 [32] ,以上结果同样可以支持肠道微生物失调的程度与IBS症状严重程度相关这一观点。亦有研究结论表明FMT对IBS症状缓解没有明显作用 [33] ,产生这种相互矛盾结果的原因可能有很多,例如患者队列大小、研究所使用的方案、粪便移植的方式和剂量等各种差异,还有不可忽视的粪便捐赠者的不同。即便研究结果存在争议,FMT疗法的前景依旧被看好,通过移植健康个体的肠道菌群来重建患者的肠道菌群,改善菌群失调的状态,这种方法在理论和实践中都被证明可行,只是需要更多相对完善的临床试验来确定最佳的移植方案。

6. 低FODMAPs饮食

FODMAPs饮食是一种特殊的饮食策略,旨在减少一组在肠道中发酵产生气体、引起肠道不适的碳水化合物的摄入。这些碳水化合物包括可发酵低聚糖、二糖、单糖和多元醇为特征的食物。研究发现IBS患者摄入FODMAPs饮食后,会出现严重且痛苦的胃肠道症状,而与之相对的低FODMAPs饮食则对IBS症状改善有益。将高FODMAPs饮食喂给大鼠后,观察到大鼠粪便中革兰氏阴性菌增加、脂多糖升高,并引发了炎症反应、屏障功能障碍和内脏敏感等肠道病理学变化。然而,这些变化可以通过采用低FODMAPs饮食得以逆转 [34] 。粪便内毒素增加介导了肠道屏障功能障碍和由于微生物失调而导致的内脏敏感,这是FODMAPs饮食造成IBS症状加剧的机制,与之相对的,通过调节肠道微生物、预防肠道炎症,低FODMAPs饮食被认为是一种可靠的缓解症状的治疗方法。研究表明,饮食干预通过增加拟杆菌、减少病原菌改变了IBS患者的肠道菌群组成 [35] ,然而长期低FODMAPs饮食会造成肠道中双歧杆菌减少,尽管可以通过益生菌恢复 [36] [37] ,这仍旧使得低FODMAPs饮食无法被推荐作为长期治疗方法。此外,无法寻找到高质量和富有营养的低FODMAPs食品作为传统饮食的替代品,也是IBS患者在长期应用该饮食疗法时依从性差的原因之一 [38] 。总而言之,低FODMAPs饮食对IBS症状改善的确有益处,但其可能产生的长期的负面后果值得重视,相关研究还应继续着眼于FODMAPs饮食与肠道菌群的关联,以期寻找到有价值的微生物生物标记物,便于更有针对性地筛选出可能从饮食疗法中获益的IBS患者。

7. 小结与展望

通过采用不同的治疗策略,如菌群移植、低FODMAPs饮食等,可以调整肠道微生物的平衡,从而改善IBS患者的症状。菌群移植作为一种较新的治疗手段,通过将健康捐赠者的肠道菌群引入患者的肠道,以重建正常微生物群落。通过减少与高FODMAPs食物相关的不适感,低FODMAPs饮食可以减少对这些碳水化合物敏感的菌群的增殖,使失衡的肠道菌群恢复正常,显著提高IBS患者的生活质量,使其更好地应对日常生活。

随着技术的进步,如高通量测序技术的应用,将有望更全面地揭示肠道微生物的多样性和功能,更深入地探索肠道菌群与IBS之间的关系,理解微生物组成的变化对患者症状的影响,并提高治疗方法的针对性。此外,个体化治疗的研究也将是未来的重要方向,因为不同个体对于菌群调节的反应可能存在差异。通过深入研究IBS患者的微生物组成,有望发现更为精准、有效的调节策略,为IBS患者提供个体化的治疗方案。

参考文献

[1] Buono, J.L., Carson, R.T. and Flores, N.M. (2017) Health-Related Quality of Life, Work Productivity, and Indirect Costs among Patients with Irritable Bowel Syndrome with Diarrhea. Health and Quality of Life Outcomes, 15, Article No. 35.
https://doi.org/10.1186/s12955-017-0611-2
[2] Lacy, B.E. and Patel, N.K. (2017) Rome Criteria and a Diagnostic Approach to Irritable Bowel Syndrome. Journal of Clinical Medicine, 6, Article 99.
https://doi.org/10.3390/jcm6110099
[3] Sperber, A.D., Bangdiwala, S.I., Drossman, D.A., et al. (2021) World-wide Prevalence and Burden of Functional Gastrointestinal Disorders, Results of Rome Foundation Global Study. Gas-troenterology, 160, 99-114.E3.
[4] Vasant, D.H., Paine, P.A., Black, C.J., et al. (2021) British Society of Gastroenter-ology Guidelines on the Management of Irritable Bowel Syndrome. Gut, 70, 1214-1240.
https://doi.org/10.1136/gutjnl-2021-324598
[5] 杨芳, 严晶, 刘丽娜, 等. 肠易激综合征病因及发病机制研究的新进展[J]. 河北医科大学学报, 2020, 41(8): 987-992.
[6] Tian, S., Zhang, H., Chen, S., et al. (2023) Global Re-search Progress of Visceral Hypersensitivity and Irritable Bowel Syndrome: Bibliometrics and Visualized Analysis. Frontiers in Pharmacology, 14, Article 1175057.
https://doi.org/10.3389/fphar.2023.1175057
[7] Mayer, E.A. (2011) Gut Feelings: The Emerging Biology of Gut-Brain Communication. Nature Reviews Neuroscience, 12, 453-466.
https://doi.org/10.1038/nrn3071
[8] Mawdsley, J.E. and Rampton, D.S. (2005) Psychological Stress in IBD: New Insights into Pathogenic and Therapeutic Implications. Gut, 54, 1481-1491.
https://doi.org/10.1136/gut.2005.064261
[9] Koloski, N.A., Jones, M., Kalantar, J., et al. (2012) The Brain—Gut Pathway in Functional Gastrointestinal Disorders Is Bidirectional: A 12-Year Prospective Population-Based Study. Gut, 61, 1284-1290.
https://doi.org/10.1136/gutjnl-2011-300474
[10] Koloski, N.A., Jones, M. and Talley, N.J. (2016) Evidence That Independent Gut-to-Brain and Brain-to-Gut Pathways Operate in the Irritable Bowel Syndrome and Functional Dyspep-sia: A 1-Year Population-Based Prospective Study. Alimentary Pharmacology & Therapeutics, 44, 592-600.
https://doi.org/10.1111/apt.13738
[11] Sender, R., Fuchs, S. and Milo, R. (2016) Are We Really Vastly Outnum-bered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell, 164, 337-340.
https://doi.org/10.1016/j.cell.2016.01.013
[12] De Vos, W.M., Tilg, H., Van Hul, M. and Cani, P.D. (2022) Gut Microbiome and Health: Mechanistic Insights. Gut, 71, 1020-1032.
https://doi.org/10.1136/gutjnl-2021-326789
[13] Leonardi, I., Gao, I.H., Lin, W.Y., et al. (2022) Mucosal Fungi Promote Gut Barrier Function and Social Behavior via Type 17 Immunity. Cell, 185, 831-846.E14.
https://doi.org/10.1016/j.cell.2022.01.017
[14] Hookway, C., Buckner, S., Crosland, P., et al. (2015) Irritable Bowel Syndrome in Adults in Primary Care: Summary of Updated NICE Guidance. BMJ, 350, h701.
https://doi.org/10.1136/bmj.h701
[15] Li, C.Y. and Li, S.C. (2015) Treatment of Irritable Bowel Syndrome in Chi-na: A Review. World Journal of Gastroenterology, 21, 2315-2322.
https://doi.org/10.3748/wjg.v21.i8.2315
[16] Chang, L., Lembo, A. and Sultan, S. (2014) American Gastroentero-logical Association Institute Technical Review on the Pharmacological Management of Irritable Bowel Syndrome. Gas-troenterology, 147, 1149-1172.E2.
https://doi.org/10.1053/j.gastro.2014.09.002
[17] Nam, Y., Min, Y.S. and Sohn, U.D. (2018) Recent Advances in Pharmacological Research on the Management of Irritable Bowel Syndrome. Archives of Pharmacal Research, 41, 955-966.
https://doi.org/10.1007/s12272-018-1068-5
[18] Naliboff, B.D., Frese, M.P. and Rapgay, L. (2008) Mind/Body Psychological Treatments for Irritable Bowel Syndrome. Evidence-Based Complementary and Alternative Medicine, 5, Article ID: 919413.
https://doi.org/10.1093/ecam/nem046
[19] Jeffery, I.B., Quigley, E.M., Ohman, L., et al. (2012) The Microbiota Link to Irritable Bowel Syndrome: An Emerging Story. Gut Microbes, 3, 572-576.
https://doi.org/10.4161/gmic.21772
[20] Parkes, G.C., Rayment, N.B., Hudspith, B.N., et al. (2012) Distinct Mi-crobial Populations Exist in the Mucosa-Associated Microbiota of Sub-Groups of Irritable Bowel Syndrome. Neurogas-troenterology & Motility, 24, 31-39.
https://doi.org/10.1111/j.1365-2982.2011.01803.x
[21] Jeffery, I.B., O’toole, P.W., Öhman, L., et al. (2012) An Irritable Bowel Syndrome Subtype Defined by Species-Specific Alterations in Faecal Microbiota. Gut, 61, 997-1006.
https://doi.org/10.1136/gutjnl-2011-301501
[22] Labus, J.S., Hollister, E.B., Jacobs, J., et al. (2017) Differences in Gut Microbial Composition Correlate with Regional Brain Volumes in Irritable Bowel Syndrome. Microbiome, 5, Article No. 49.
https://doi.org/10.1186/s40168-017-0260-z
[23] Tap, J., Derrien, M., Tornblom, H., et al. (2017) Identification of an Intestinal Microbiota Signature Associated with Severity of Irritable Bowel Syndrome. Gastroenterology, 152, 111-123.E8.
https://doi.org/10.1053/j.gastro.2016.09.049
[24] Hugerth, L.W., Andreasson, A., Talley, N.J., et al. (2020) No Distinct Microbiome Signature of Irritable Bowel Syndrome Found in a Swedish Random Population. Gut, 69, 1076-1084.
https://doi.org/10.1136/gutjnl-2019-318717
[25] Jeffery, I.B., O’herlihy, E., Shanahan, F. and O’Toole, P.W. (2020) Microbiome Alterations in IBS. Gut, 69, 2263-2264.
https://doi.org/10.1136/gutjnl-2020-320919
[26] Kennedy, P.J., Cryan, J.F., Dinan, T.G. and Clarke, G. (2014) Ir-ritable Bowel Syndrome: A Microbiome-Gut-Brain Axis Disorder? World Journal of Gastroenterology, 20, 14105-14125.
https://doi.org/10.3748/wjg.v20.i39.14105
[27] Crouzet, L., Gaultier, E., Del’homme, C., et al. (2013) The Hypersensitivity to Colonic Distension of IBS Patients Can Be Transferred to Rats through Their Fecal Mi-crobiota. Neurogastroenterology & Motility, 25, e272-e282.
https://doi.org/10.1111/nmo.12103
[28] De Palma, G., Lynch, M.D.J., Lu, J., et al. (2017) Transplantation of Fecal Microbiota from Patients with Irritable Bowel Syndrome Alters Gut Function and Behavior in Recipient Mice. Science Translational Medicine, 9, eaaf6397.
[29] Johnsen, P.H., Hilpusch, F., Cavanagh, J.P., et al. (2018) Faecal Microbiota Transplantation versus Placebo for Moderate-to-Severe Irritable Bowel Syndrome: A Double-Blind, Randomised, Place-bo-Controlled, Parallel-Group, Single-Centre Trial. The Lancet Gastroenterology and Hepatology, 3, 17-24.
https://doi.org/10.1016/S2468-1253(17)30338-2
[30] El-Salhy, M., Hatlebakk, J.G., Gilja, O.H., et al. (2020) Ef-ficacy of Faecal Microbiota Transplantation for Patients with Irritable Bowel Syndrome in a Randomised, Double-Blind, Placebo-Controlled Study. Gut, 69, 859-867.
https://doi.org/10.1136/gutjnl-2019-319630
[31] Goll, R., Johnsen, P.H., Hjerde, E., et al. (2020) Effects of Fecal Microbiota Transplantation in Subjects with Irritable Bowel Syndrome Are Mirrored by Changes in Gut Microbiome. Gut Microbes, 12, Article ID: 1794263.
https://doi.org/10.1080/19490976.2020.1794263
[32] El-Salhy, M., Winkel, R., Casen, C., et al. (2022) Efficacy of Fecal Microbiota Transplantation for Patients with Irritable Bowel Syndrome at 3 Years after Transplantation. Gastroen-terology, 163, 982-994.E14.
https://doi.org/10.1053/j.gastro.2022.06.020
[33] Halkjaer, S.I., Christensen, A.H., Lo, B.Z.S., et al. (2018) Faecal Microbiota Transplantation Alters Gut Microbiota in Patients with Irritable Bowel Syndrome: Results from a Randomised, Double-Blind Placebo-Controlled Study. Gut, 67, 2107-2115.
https://doi.org/10.1136/gutjnl-2018-316434
[34] Zhou, S.Y., Gillilland III., M., Wu, X., et al. (2018) FODMAP Diet Modulates Visceral Nociception by Lipopolysaccharide-Mediated Intestinal Inflammation and Barrier Dysfunction. Journal of Clinical Investigation, 128, 267-280.
https://doi.org/10.1172/JCI92390
[35] Vervier, K., Moss, S., Kumar, N., et al. (2022) Two Microbiota Subtypes Identified in Irritable Bowel Syndrome with Distinct Responses to the Low FODMAP Diet. Gut, 71, 1821-1830.
https://doi.org/10.1136/gutjnl-2021-325177
[36] Mcintosh, K., Reed, D.E., Schneider, T., et al. (2017) FODMAPs Alter Symptoms and the Metabolome of Patients with IBS: A Randomised Controlled Trial. Gut, 66, 1241-1251.
https://doi.org/10.1136/gutjnl-2015-311339
[37] Staudacher, H.M., Lomer, M.C.E., Farquharson, F.M., et al. (2017) A Diet Low in FODMAPs Reduces Symptoms in Patients with Irritable Bowel Syndrome and a Probiotic Re-stores Bifidobacterium Species: A Randomized Controlled Trial. Gastroenterology, 153, 936-947.
https://doi.org/10.1053/j.gastro.2017.06.010
[38] Ispiryan, L., Zannini, E. and Arendt, E.K. (2022) FODMAP Modulation as a Dietary Therapy for IBS: Scientific and Market Perspective. Comprehensive Reviews in Food Science and Food Safety, 21, 1491-1516.
https://doi.org/10.1111/1541-4337.12903