Back to Journals » Journal of Inflammation Research » Volume 18
The Role of Gut Microbiota on Intestinal Fibrosis in Inflammatory Bowel Disease and Traditional Chinese Medicine Intervention
Authors Fang L, Peng H , Tan Z , Deng N, Peng X
Received 13 November 2024
Accepted for publication 10 April 2025
Published 7 May 2025 Volume 2025:18 Pages 5951—5967
DOI https://doi.org/10.2147/JIR.S504827
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Professor Ning Quan
Leyao Fang,1– 3 Huiyi Peng,1– 3 Zhoujin Tan,2,3 Na Deng,2,3 Xinxin Peng1,2
1The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China; 2The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Changsha, People’s Republic of China; 3School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
Correspondence: Na Deng, School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, People’s Republic of China, Email [email protected] Xinxin Peng, The First Hospital of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, People’s Republic of China, Email [email protected]
Abstract: Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder of the intestine, frequently complicated by intestinal fibrosis. As fibrosis progresses, it can result in luminal stricture and compromised intestinal function, significantly diminishing patients’ quality of life. Emerging evidence suggests that gut microbiota and their metabolites contribute to the pathogenesis of IBD-associated intestinal fibrosis by influencing inflammation and modulating immune responses. This review systematically explores the mechanistic link between gut microbiota and intestinal fibrosis in IBD and evaluates the therapeutic potential of traditional Chinese medicine (TCM) interventions. Relevant studies were retrieved from PubMed, Web of Science, Embase, Scopus, CNKI, Wanfang, and VIP databases. Findings indicate that TCM, including Chinese herbal prescriptions and bioactive constituents, can modulate gut microbiota composition and microbial metabolites, ultimately alleviating intestinal fibrosis through anti-inflammatory, immunemodulatory, and anti-fibrotic mechanisms. These insights highlight the potential of TCM as a promising strategy for targeting gut microbiota in the management of IBD-associated fibrosis.
Keywords: inflammatory bowel disease, intestinal fibrosis, intestinal inflammation, gut microbiota, traditional Chinese medicine
Introduction
Inflammatory bowel disease (IBD), encompassing Crohn’s disease (CD) and ulcerative colitis (UC), is a chronic inflammatory disorder of the intestine. Although its exact cause remains unclear, genetic susceptibility, environmental factors, and immune dysregulation are known to contribute to its onset.1 The gut microbiota plays a vital role in maintaining intestinal homeostasis, supporting gut barrier integrity, and regulating systemic inflammation. Numerous studies have demonstrated that the intestinal microbiota can exert either detrimental or beneficial effects on disease progression.2,3 Gut microbiota dysbiosis can promote intestinal inflammation, impaire barrier function, and stimulate the overgrowth of pathogenic bacteria, all of which may contribute to gastrointestinal disorders. The gut microbiota and its metabolites directly or indirectly interact with the intestinal epithelium, influencing intestinal immunity, inflammatory responses, and mucosal barrier function. This dysbiosis leads to intestinal dysfunction and plays a role in the onset and progression of intestinal diseases. Furthermore, the intestinal microbiota of patients with intestinal fibrosis differs significantly from that of healthy individuals.4 Animal models have also demonstrated that specific intestinal bacteria can induce intestinal fibrosis.5 Intestinal microbiota metabolites influence intestinal function and are closely associated with IBD.6 While intestinal microbiota and their metabolites contribute to the onset and advancement of intestinal fibrosis, certain microbiota elements might be beneficial.7
Intestinal fibrosis is the main manifestation of structural damage in IBD patients.8 IBD-associated intestinal fibrosis primarily occurs in CD but is less commonly seen in UC, which is typically considered an inflammatory process confined to the mucosal and submucosal layers. However, recent research has shown that chronic inflammation in UC can lead to localized fibrosis.9 A study of 706 UC tissue cross-sections identified submucosal fibrosis exclusively in inflamed regions, with colectomy samples showing a strong correlation between fibrosis and inflammation.10 These findings suggest that submucosal fibrosis and thickening of the muscularis mucosae are prevalent pathological features of progressive UC. Patients who underwent surgery to relieve intestinal stricture still experienced recurrence of stenosis in 70% of cases, with more than half requiring additional surgery. This recurrent issue significantly impacts their quality of life.11,12 Excessive accumulation of extracellular matrix (ECM) is a hallmark of intestinal fibrosis and a primary cause of structural damage in IBD.13 The pathogenesis of intestinal fibrosis in IBD is complex, involving chronic inflammation, microbial factors, and even dietary influences that provide the body with persistent harmful stimuli. In response to tissue injury, the body initiates a self-repair process.14 Intestinal microbiota dysbiosis, impaired barrier function, and the activation of cytokines and growth factors, along with the triggering of immune responses, all contribute to excessive ECM deposition and metabolic imbalance (Figure 1). Currently, there are no clinically effective therapies for intestinal fibrosis, making prevention the most viable strategy.14 Although IBD treatments like aminosalicylates and immunosuppressants can reduce inflammation, they do not relieve fibrostenotic obstruction. Biologics, such as anti-tumor necrosis factor, may promote ulcer healing but could also contribute to fibrostenosis.15 Therefore, urgent research is needed to find effective treatments for IBD-related intestinal fibrosis. Emerging evidence suggests that modulating gut microbiota balance holds promise as a potential therapeutic approach.
Traditional Chinese Medicine (TCM), rooted in the principles of holism and syndrome differentiation, has been widely used to treat gastrointestinal disorders with proven therapeutic efficacy. The ancient Chinese medical texts record numerous formulas that are still used in clinical practice today. With the development of modern research techniques, the mechanisms by which Chinese medicine exerts its effects are gradually being explained, and many bioactive compounds from Chinese herbs have been extracted and applied. Recent studies have demonstrated that TCM formulations and bioactive compounds possess therapeutic potential for various fibrotic diseases. Several studies suggest that TCM interventions modulate intestinal microbiota and their metabolites, which are closely linked to inflammation and fibrosis. Beyond microbiota regulation, other strategies for managing IBD-related fibrosis include inhibiting profibrotic signaling pathways and modulating immune responses. This review aims to explore the role of gut microbiota in the pathogenesis of intestinal fibrosis in IBD and evaluate the therapeutic potential of TCM in addressing this complex condition.
Methods
To ensure a thorough and systematic review of TCM in modulating gut microbiota and its metabolites for the treatment of intestinal fibrosis in IBD, we searched multiple electronic databases, including PubMed, Web of Science, Embase, Scopus, China National Knowledge Infrastructure (CNKI), and Wanfang. The search was conducted up to September 2024, with no language constraints. The following search terms and combinations were used: (“inflammatory bowel disease” OR “IBD” OR “Crohn’s disease” OR “ulcerative colitis”) AND (“intestinal fibrosis” OR “fibrosis” OR “stenosis”) AND (“traditional Chinese medicine” OR “Chinese herbal medicine” OR “Chinese medicine formula” OR “TCM monomers” OR “bioactive components”) AND (“gut microbiota” OR “intestinal microbiota” OR “microbial metabolites”). Inclusion Criteria: Research on the effects of TCM, Chinese herbal formulations, or active components of Chinese herbs on intestinal fibrosis in IBD; Original research publications, covering in vitro, in vivo, and clinical trials. Systematic reviews and meta-analyses on the topic. Exclusion criteria: The exclusion criteria include studies that are unrelated to IBD-associated intestinal fibrosis; Case studies, conference abstracts, letters, editorials, and non-peer-reviewed papers; Studies with insufficient experimental data or mechanistic insights. Two reviewers conducted the selection procedure independently in order to verify accuracy and minimize bias. Discrepancies were handled by discussing or consulting with a third reviewer. Furthermore, the references of selected research were manually checked for potentially relevant literature.
Intestinal Fibrosis and the Role of the Gut Microbiota
Intestinal Microbiota Dysbiosis in IBD
Microbiota in the human intestine have an important effect on both health and disease.16 Alterations in the diversity and composition of the gut microbiota are major contributors to IBD.17,18 The mucosal barrier in the non-inflamed ileum of UC patients becomes impaired, leading to reduced mucus secretion, enhanced bacterial infiltration, and a reduction in bacterial diversity due to mucosal barrier defects and aberrant bacterial colonization.19 The microbiological, chemical, mechanical, and immunological barriers constituting the mucosal layer of the intestine collaboratively contribute to the development and prevention of IBD (Figure 2). Intestinal barrier failure is a major cause of illness recurrence in individuals with IBD.20 The gut microbiota acts as a biological barrier and regulates the production of important chemical barriers, such as mucin and antimicrobial peptides (AMPs).21 The intestinal microbiota compositions of healthy individuals and those with IBD are different. Studies have shown that individuals with IBD have a higher percentage of pathogenic bacteria such as Bacteroides fragilis, Escherichia coli, and Clostridium clostridioforme, and a lower proportion of beneficial bacteria, such as Bifidobacterium and Lactobacillus.22–24 Research on the potentially harmful microbiota associated with intestinal fibrosis has been ongoing for many years, with initial tests demonstrating that bacterial suspensions applied to the intestinal wall elevate tissue levels of transforming growth factor-β1 (TGF-β1) and collagen.25 Myofibroblasts and fibrocytes which respond to flagellin, a bacterial component, secrete large amounts of ECM.26 However, some bacteria can exert beneficial effects. Lactobacillus acidophilus can reduce α-smooth muscle actin (α-SMA) levels and collagen I accumulation.27 The microbiota plays a critical role in the progression of IBD, with its metabolites potentially acting as key regulators of the disease’s pathogenesis.28 The intestinal microbiota produces several metabolites, including lipopolysaccharide (LPS), bile acids (BAs), tryptophan, and short-chain fatty acids (SCFAs), which significantly influence intestinal inflammation and fibrosis. Individuals with IBD exhibit disruption in BA metabolism. According to previous research, IBD patients have a different fecal BA pool than healthy controls, caused by an increase in primary bile acids and a reduction in secondary bile acids (SBAs).22 Supplementation with SBAs can reduce intestinal inflammation in murine models of colitis.29 In patients with IBD, inflammatory cells and dysbacteriosis are elevated, along with reduced levels of SCFAs.30 Enema fluid containing butyrate or glutamine reduces fibrosis and inflammation in experimental diverted colitis.31 Alterations in tryptophan metabolism are associated with gut microbial dysbiosis and the severity of illness.32 LPS activates intestinal fibroblasts, leading to increased collagen synthesis.33 Therefore, the gut microbiota and its metabolites are closely associated with the pathogenesis of IBD.
Mechanisms Underlying Gut Microbiota-Mediated Intestinal Fibrosis
Intestinal microbiota plays a crucial role in the initiation and progression of intestinal fibrosis by influencing barrier function, immune responses, and fibroblast activation.18 An imbalance in the host‘s intestinal flora, characterized by a significant increase in pathogenic bacteria, leads to the production of harmful molecules that directly damage the intestinal cells. The intestinal mucosa and epithelial cells are severely damaged, making them more susceptible to microbial damage. These processes involve the activation of fibroblasts and the excessive accumulation of extracellular matrix. Research on intestinal fibrosis commonly uses bacteria-induced IBD models, such as Adherent-invasive Escherichia coli (AIEC) and Salmonella enterica serovar infection mouse models.34 AIEC infection has been shown to cause persistent intestinal inflammation and fibrosis in mice.35 AIEC may suppress exosomal miRNA lethal-7b (let-7b) produced by intestinal epithelial cells, thereby facilitating the fibrotic phenotype of intestinal macrophages and, consequently, fibrosis.36 Elimination of the myeloid differentiation primary response gene 88 in a Salmonella-induced colitis model reduces intestinal fibrosis.37 Creeping fat is closely associated with intestinal fibrosis.38 Clostridium innocuum in mesenteric adipose tissue induces M2 macrophages, which secrete pro-fibrotic factors like TGF-β.39 The intestinal microbiome directly and indirectly activates fibroblasts via epithelial or immune cells.40 Certain molecules from the microbiota can affect fibroblast transcription, leading to intestinal fibrosis.37,41 Myofibroblasts and fibrocytes that react to flagellin, a bacterial component, secrete large amounts of ECM. Consequently, it appears that potential pathogens contribute significantly to intestinal fibrosis. Furthermore, symbiotic bacteria can transform into pathogenic forms, penetrating the epithelium and triggering immune responses that disrupt junctional proteins, thereby increasing permeability and promoting a “leaky gut”.42 “Leaky gut” is associated with intestinal inflammation, which exposes mesenchymal cells to luminal components and inflammatory mediators.43 Prolonged exposure of mesenchymal cells to this inflammatory environment can lead to their activation and contribute to the development of intestinal fibrosis. In addition, the induction of cytokines may contribute to microbiota-driven intestinal fibrosis.44 Cytokine induction contributes to microbiota-driven fibrosis, with TL1A-mediated fibroblast activation correlating with specific bacterial presence. Th17 cells produce amphiregulin, further driving fibrosis.45,46 The formation of multiple ECMs proteins is significantly induced by TGF-β1. As an essential part of the TGF-β/Smad signaling pathway, the Smad protein family acts as a signal converter and is located downstream of TGF-β. Matrix metalloproteinases (MMPs) and tissue inhibitors of matrix metalloproteinases (TIMPs) are among the pro- and anti-fibrotic factors that regulate intestinal degradation and repair.47 To a substantial extent, the ECM can be broken down by MMPs and TIMPs control MMPs’ activity. TGF-β may affect the balance between MMPs and TIMPs. Endogenous antimicrobial peptides suppress TGF-β1, insulin-like growth factor-1 (IGF-I), and collagen production through mitogen-activated protein kinase (MAPK)-dependent pathways.48,49
Microbial metabolites also play a key role. LPS stimulates the nuclear factor κ-B (NF-κB) pathway via Toll-like receptor 4 (TLR4), while TLR4 deficiency reduces fibrosis, TGF-β levels, and collagen synthesis.50 High levels of serum succinate and succinate receptor 1 (SUCNR1) were found in excised intestinal tissue and intestinal fibroblasts from CD patients. SUCNR1 signaling has been shown to exacerbateinflammation and fibroblast activation in patients with CD.51 The link between CD and reduced hydrogen sulfide (H2S) synthesis, as well as enhanced intestinal microbial hydrogen sulfide production, is mediated by assimilatory sulfate reduction (ASR). The microbial ASR-mediated metabolism of dietary sulfate is crucial for colitis.52 Low H2S levels reduce inflammation, whereas excessive H2S disrupts mucus integrity.53 Although some metabolites are associated with intestinal fibrosis in IBD, research indicates that specific metabolites can mitigate intestinal inflammation and alleviate fibrosis. Beneficial metabolites like indoleacrylic acid (IA) from Peptostreptococcus enhance epithelial barrier integrity, while aryl hydrocarbon receptor (AHR) activation by tryptophan and SCFAs influences inflammation.54,55 Regulatory T (Treg) cells play a key role in maintaining immune balance in the gut and controlling excessive pro-inflammatory responses. Normal BAs metabolism is essential for the regulation of TH17 and Treg differentiation.56 Indole-3-propionic acid (IPA) inhibits pregnane X receptor (PXR) to reduce inflammation,57 while SCFAs enhance forkhead transcription factor p-3 (Foxp3) expression and Treg function via G protein-coupled receptor 43 (GPR43) protecting against colitis.58,59 IBD fibrosis results from interactions among metabolites and pathways, encompassing pro- and anti-inflammatory as well as pro- and anti-fibrotic mechanisms at various stages of the disease.60 The microbiota and its metabolites influence the intestinal barrier, activate the inflammatory response, and disrupt the immune system, leading to fibroblast activation and excessive ECM production (Figure 3). Potential treatments may involve modulating microbial metabolites by enhancing beneficial ones, reducing harmful ones, or targeting their receptors. Further research is needed to optimize microbiota-based therapies for IBD fibrosis.
Regulating the Microbiota to Alleviate Intestinal Fibrosis
Current evidence suggests that diet plays a dual role in IBD, influencing disease risk and activity by modulating gut microbiota and intestinal inflammation. Due to impaired immune response, intestinal permeability, and changes in the mucosal layer, diet may cause microbial dysbiosis and intestinal inflammation.61 Western diets, rich in sugar, fat, and processed ingredients, contribute to microbial dysbiosis, increased intestinal permeability, and fibrosis progression.62–65 In contrast, specific dietary interventions, such as the Crohn’s Disease Exclusion Diet (CDED), Crohn’s disease-TREAT (CD-TREAT) diet,66 Ulcerative Colitis Exclusion Diet (UCED),67 and the Mediterranean diet,68 have been shown to restore microbial balance and support intestinal health. Suskind et al found that switching to a low-“fermentable oligo-, di-, and mono-saccharides and polyols” (FODMAP) diet drastically affected the microbiota composition.69 Additionally, nutrients with anti-fibrotic properties can target key receptors like γ (PPARγ),70 AhR,71 and VDR,72 offering potential therapeutic benefits. While unhealthy diets may exacerbate intestinal fibrosis, dietary modifications can help mitigate these effects by reshaping gut microbiota and its metabolites.
A decrease in beneficial microbiota is more strongly linked to the onset of IBD than an increase in pathogenic microbiota. Beneficial microbes, including those involved in butyrate generation, such as Faecalibacterium, Ruminococcus Christensenellaceae, Methanobrevibacter, and Butyricicoccus, were much less abundant in CD patients than in normal controls.23,73 Butyrate preserves epithelial cell integrity and intestinal homeostasis, which are essential for preventing intestinal diseases.74,75 Akkermansia muciniphila, crucial for gut barrier integrity, has emerged as a potential therapeutic target.76 Probiotics like Lactobacillus plantarum LC27 and Bifidobacterium longum LC67 can restore microbial balance, reduce inflammation, and enhance IBD therapy outcomes.77 Bifidobacterium breve (B. breve) H4-2 and H9-3 can alleviate colitis induced by dextran sulfate sodium (DSS).78 Prebiotics, including oligosaccharides and inulin,79,80 support beneficial bacteria, while synbiotics facilitate probiotic colonization.81 Research indicates that fecal microbiota transplantation (FMT) is an efficacious intervention for IBD worsened by Clostridium difficile infection and reinstates a variety of intestinal microbiota.82 By promoting short-chain fatty acid production, modulating inflammatory responses, and reinforcing the intestinal barrier, probiotics, prebiotics, and synbiotics present promising strategies for alleviating intestinal inflammation and fibrosis.83
Several pharmaceuticals have shown anti-fibrotic effects in animal studies (Table 1). The Lactococcus lactis-derived Hsp65 antigen reduced IL-6 and TGF-α while increasing IL-10 in chronic intestinal inflammation models, suggesting potential therapeutic benefits.84 TL1A activates fibroblasts and regulates the intestinal immune response and tissue remodeling, while TL1A-induced intestinal fibrosis relies on the intestinal microbiome.45 The anti-TL1A antibody (PRA023) has demonstrated higher endoscopic response and clinical remission rates in moderate-to-severe Crohn’s disease. Nicotinamide mononucleotide (NMN) therapy modulates gut microbiota and mitigates radiation-induced intestinal fibrosis.85 Fibroblast growth factor 19 (FGF19) deficiency in Crohn’s disease affects microbiome composition and inflammation, while FGF19-M52, via FXR, alters bile acid metabolism and protects against intestinal inflammation.86 Lactobacillus johnsonii extracellular vesicles (EVs) suppress NLRP3 activation, contributing to antifibrotic effects. Exploring gut microbiota-related antifibrotic therapies remains a promising research direction.87 Therefore, exploring antifibrotic therapies targeting gut microbiota and its metabolic byproducts offers a promising and feasible approach for treating IBD-associated intestinal fibrosis.
![]() |
Table 1 Anti-Intestinal Fibrosis Drugs Based on Microbiota |
The Effect of TCM on Gut Microbiota and Intestinal Fibrosis
The Intestinal Microbiota as a Target in TCM for Treating Organ Fibrosis
As research on anti-fibrosis treatments has progressed, the benefits and unique aspects of TCM in the treatment of fibrotic disorders have become increasingly apparent. TCM believes that chronic inflammation is accompanied by latent pathogenic factors that hinder the flow of qi, blood, and water, leading to the formation of pathological products. Qi stagnation, blood stasis, and phlegm-dampness are key pathological factors in fibrotic diseases, and the elimination of these factors has become a central concept in TCM for the treatment of such disorders.88 Cryptotanshinone (CPT), a major biologically active component in the roots of Salvia miltiorrhiza, modulates radiation-induced intestinal microbiota and BAs metabolism in mice, reducing pulmonary inflammation and fibrosis.89 Certain compounds present in herbs, such as polyphenols and polysaccharides, can assist the body in producing beneficial metabolites like SCFAs and tryptophan, thereby alleviating liver fibrosis and inflammation90 Danggui Shaoyao San has been shown to help treat CCl4-induced hepatic fibrosis by influencing the intestinal microbiota and its metabolites (SCFAs and BAs).91 Ethanol extracts of Ganoderma lucidum ameliorated hepatic fibrosis and enhanced the interaction between metabolites and the intestinal microbiota through the NF-κB and TGF-β1/Smad pathways.92 Betanin decreased streptozotocin-induced fibrosis in a diabetic nephropathy model by altering the expression of EMT-associated markers.93 Ootheca mantidis reduced renal fibrosis in mice by suppressing apoptosis and boosting the intestinal bacteria Akkermansia muciniphila while modifying glutamine metabolism.94 The occurrence and progression of fibrotic disorders are directly linked to the intestinal microbiota. From single herbs to compounds, and from crude extracts to monomer components, the biological foundations of TCM’s anti-fibrotic activity are gradually being uncovered.
The Impact of TCM Prescriptions on Intestinal Flora and Fibrosis
Traditional Chinese medicine compounds, single herbs, and monomers can alleviate IBD and intestinal fibrosis by modulating gut microbiota and their metabolites.95,96 Table 2 summarizes the specific mechanisms of various TCM formulas and their effects on gut microbiota and metabolic products. Gut microbiota imbalance plays a crucial role in the onset and progression of intestinal fibrosis in IBD. TCM formulas can restore gut microbiota balance by inhibiting harmful bacteria and promoting beneficial bacteria. In addition, traditional Chinese medicine can exert therapeutic effects by modulating the levels of microbial metabolites.97 Kuijieyuan Decoction (KJYD) decreases Escherichia-Shigella and Desulfovibrio, while increasing Alloprevotella, Treponema, and Prevotella, leading to the suppression of the PI3K/AKT/NF-κB pathway and the alleviation of colitis.98 Similarly, Huanglian Jiedu Decoction (HLJD) suppresses Alistipes, Prevotella, and Bacteroides, while increasing Oscillibacter and Lactobacillus, thereby restoring microbial homeostasis and modulating Csf1r/Src signaling to mitigate inflammation.99,100 Additionally, Yiyi Fuzi Baijiang Powder (YFBP) reduces Prevotella_9, while enhancing Lachnospiraceae_NK4A136_group, contributing to the inhibition of TLR4/NF-κB/NLRP3 and the restoration of the intestinal barrier.101 Several TCM formulas exert protective effects by increasing beneficial bacteria, which promote SCFA production and regulate immune balance. For example, Gegen Qinlian Decoction (GGQL) increases Lactobacillus and Lachnospiraceae_NK4A136_group, leading to higher SCFA levels and improved Treg/Th17 balance, which helps alleviate experimental colitis.102 Similarly, Rhubarb Peony Decoction (RPD) upregulates Butyricicoccus pullicaecorum, restoring SCFA levels and regulating the Th17/Treg ratio, thereby exerting anti-inflammatory and intestinal barrier-protective effects.103 Anchang Yuyang Decoction (AYYD) increases Romboutsia and Monoglobus, contributing to higher SCFA levels and improved intestinal barrier function.104 Beyond modulating gut microbiota composition, some formulas exert their effects by regulating bile acid metabolism and LPS production. Baitouweng Tang (BTW) enhances Lactobacillus and Clostridium, increasing FXR and TGR5 expression and regulating bile acid metabolism, which contributes to gut microbiota balance and symptom relief in DSS-induced colitis.105 Furthermore, San Wu Huangqin Decoction (SWHQD) lowers Escherichia-Shigella abundance and reduces LPS production, thereby restoring epithelial MUC2 secretion and colonic tight junction integrity.106 In addition to microbiota modulation, some TCM formulas directly regulate fibrosis-related pathways. Qingchang Wenzhong Decoction (QCWZD) alleviates fibrosis by inhibiting IL-17, NF-κB, and TLR4, reducing inflammation and fibrosis progression.107 Meanwhile, Xue-jie-San (XJS) inhibits Notch1 and FGL-1 signaling, thereby suppressing autophagy, endothelial-to-mesenchymal transition (EndoMT), and EMT, mechanisms implicated in Crohn’s disease-related fibrosis.108,109 Similarly, Wumei Wan (WMW) mitigates fibrosis by downregulating α-SMA, collagen I, and MMP-9, slowing fibrosis progression110 Qingchang Tongluo Decoction (QTD) inhibits intestinal fibroblast proliferation and collagen deposition via suppression of the TGF-β1/Smad pathway, making it a promising treatment for CD-related intestinal fibrosis.111 Taken together, these findings suggest that TCM interventions can modulate gut microbiota balance, as well as influence metabolites such as SCFAs, bile acids, and LPS production. Additionally, they directly regulate fibroblast activity and fibrosis-related pathways, collectively contributing to the prevention and treatment of IBD-associated intestinal fibrosis.
![]() |
Table 2 Chinese Medicine Compound in the Treatment of Intestinal Fibrosis |
Monomers of TCM Alleviate Intestinal Fibrosis by Modulating the Intestinal Microbiota
Natural plant chemicals have long served as sources of medicinal compounds and structural foundations for the development of pharmaceuticals. Evidence suggests that Chinese medicinal monomers can modulate fibroblast activity and decrease the progression of intestinal fibrosis through fibrosis-related molecular signals, inflammatory cytokines, and other mechanisms. These TCM monomers can also alleviate IBD-associated intestinal fibrosis by regulating the intestinal microbiota and its metabolites (Table 3). For instance, the sesquiterpene atractylenolides from Atractylodes macrocephala can modulate the intestinal microbiota by increasing beneficial bacteria such as Lactobacillus and restoring the intestinal barrier, offering potential treatment for UC.112 Rhein, a bioactive compound primarily found in Rheum palmatum, has garnered increasing attention for its potential anti-fibrotic properties across various organs.113,114 Notably, Rhein affects the intestinal microbiota, indirectly influencing colonic purine metabolism and alleviating colitis.115 Puerarin, found in Pueraria lobatae Radix, exerts strong anti-inflammatory effects by reducing Akkermansia muciniphila, inhibiting macrophage M1 polarization, and thereby alleviating colitis in UC-like animal models.116 Ginsenoside Rg1, derived from Panax ginseng, aids in treating colitis and repairing the intestinal mucosal barrier by regulating the microbiota and inflammatory cytokine levels.117 Polysaccharides from Dendrobium officinale increase the variety of microbes in the intestine and stimulate the colon to produce SCFAs.118 Poria cocos polysaccharide (PCP) increases small-intestinal SCFAs and strengthens the intestinal barrier.119 Schisandra chinensis polysaccharide (SCP) can balance intestinal microbiota and boost SCFAs levels.120 Patrinia Scabiosifolia extract can restore gut microbiota balance, reduce the expression of β-defensin 2, and decrease the expression of TGF-β and α-SMA, thereby exerting a therapeutic effect on TNBS-induced intestinal fibrosis.121 TCM monomers influence signaling pathways, reduce inflammatory factors, and decrease collagen production, thus mitigating intestinal fibrosis in IBD patients. Nrf2 activators, for example, can be used to maintain this molecule during the early stages of IBD, preventing the onset of cancer and fibrosis. Maggot extracts alleviate inflammation-induced intestinal fibrosis by enhancing Nrf2 expression and inhibiting the TGF-β1/SMADs pathway.122
![]() |
Table 3 Chinese Medicine Monomer in the Treatment of Intestinal Fibrosis |
Recent research has also focused on the improving efficacy of active substances in traditional Chinese medicine. For example, MIR2911 from Lonicera japonica can be generated in host small extracellular vesicles and absorbed through diet, directly influencing intestinal bacteria, reducing Escherichia -Shigella, and improving colitis symptoms.123 TGF-β, a key factor in EMT, plays a significant role in fibrosis induction124,125 Calycosin, by inhibiting the TGF-β/Smad pathway, prevents intestinal fibrosis.126 Furthermore, curcumin has been shown to reduce intestinal fibrosis in a TNBS-induced rat model by elevating PPAR-γ and E-cadherin levels and altering the MMP-3/TIMP-1 ratio, while also decreasing the levels of α-SMA and fibronectin.127,128 Plant-derived exosome-like nanoparticles (GENs) derived from Panax ginseng may improve colitis by controlling pro-inflammatory cytokines, balancing the intestinal microbiota, and enhancing the intestinal barrier.129 Additionally, the total flavone from Abelmoschus manihot can correct the Th17/Treg imbalance, decrease α-SMA, TGF-β, and TIMP-1 production, enhance MMPs, and prevent ECM deposition.130 Chinese medicine components with anti-fibrotic activity are becoming increasingly recognized due to expanding research and technological advancements, leading to a broader understanding of their mechanisms of action.
Discussion
Inflammatory bowel disease arises from the complex interplay of genetic predisposition, environmental influences, and immune dysregulation, with gut microbiota playing an important role in disease progression131,132 An imbalance in the immune response of the intestinal mucosa can occur due to microbiota dysregulation, metabolite changes, and pathogenic bacterial invasion, all of which disrupt the normal intestinal mucosal barrier.133,134 This immune activation promotes the secretion of cytokines and molecular mediators, leading to the differentiation of mesenchymal-like cells and ECM deposition, which ultimately contributes to intestinal fibrosis. Despite its significant impact on patient prognosis, intestinal fibrosis remains an unmet clinical challenge, as no clinically approved treatment options are available for intestinal fibrosis. Existing therapeutic strategies primarily focus on inhibiting myofibroblast proliferation, reducing ECM accumulation, and targeting pro-fibrotic signaling pathways. While some experimental antifibrotic agents have demonstrated efficacy in animal models and early-phase clinical trials, their translation into effective therapies is limited by concerns regarding efficacy, safety, and long-term outcomes. Novel approaches, such as gene regulation and stem cell therapy, show promise but require further validation, particularly concerning cost-effectiveness and patient safety. Patients with IBD are increasingly pursuing complementary and alternative therapies. Given the growing recognition of the gut microbiota in IBD-related fibrosis, targeting the gut microbiota has emerged as a potential therapeutic strategy. Several interventions, including dietary modifications and probiotic supplementation, have been proposed to restore microbial balance and mitigate fibrosis. However, there remain critical gaps in understanding how specific bacterial species and their metabolites directly influence fibrotic pathways. Additionally, the bidirectional relationship between gut microbiota and pharmacological interventions is increasingly acknowledged. The gut microbiota can affect drug metabolism and efficacy, while certain drugs can reshape microbial composition and function, underscoring the need for microbiota-informed precision medicine.
Traditional Chinese medicine has gained attention for its multicomponent and multitarget therapeutic properties, offering a promising approach to modulating gut microbiota and ameliorating intestinal fibrosis. Evidence suggests that TCM can restore microbial homeostasis by increasing beneficial bacteria, reducing pathogenic species, and influencing microbial-derived metabolites, thereby enhancing intestinal barrier integrity and improving intestinal inflammation, all of which have demonstrated antifibrotic effects. Additionally, TCM preparations regulate fibroblast activity and suppress pro-fibrotic signaling pathways, thus alleviating IBD-related fibrosis. Although there is limited direct evidence on the role of TCM in regulating gut microbiota for the treatment of intestinal fibrosis, we hypothesize that TCM formulas and bioactive components of Chinese medicine may alleviate IBD-associated intestinal fibrosis through modulation of the intestinal microbiota. Three key factors support this hypothesis: (1) TCM can improve intestinal microbiota dysbiosis by increasing beneficial bacteria, decreasing pathogenic bacteria, and enhancing gut barrier integrity. A key factor in the persistence of inflammation and fibrosis in IBD is the disruption of the intestinal barrier. (2) TCM regulates intestinal microbiota metabolites, some of which play a role in the pathogenesis of IBD-related fibrosis, while others have direct therapeutic potential. (3) TCM has the potential to modulate immune responses via the gut microbiota. The interaction between the gut microbiota and host immunity plays a pivotal role in the onset and progression of inflammatory disorders. The gut microbiota and immune system can activate signaling pathways, generate cytokines, influence fibroblasts, alter the ECM, and produce anti-fibrotic substances.
Although TCM-based therapies show potential, several challenges hinder their clinical application: (1) Limited mechanistic studies. While preclinical evidence supports the antifibrotic effects of TCM, the precise molecular mechanisms remain incompletely understood. Future research should focus on targeted validation of microbiota-mediated pathways and their interactions with TCM compounds. (2) Heterogeneity in TCM formulations. The lack of standardized nomenclature and inconsistent dosage compositions affect reproducibility and hinder cross-study comparisons. Establishing standardized formulations and dose optimization is essential for advancing TCM-based treatments. (3) Limited high-quality clinical evidence. Most studies on TCM interventions for intestinal fibrosis rely on animal models, with minimal evidence from well-designed randomized controlled trials (RCTs). High-quality clinical research is needed to validate the efficacy and safety of TCM formulas in human patients. To bridge these gaps, future research should prioritize multidisciplinary approaches that integrate microbiome analysis, omics technologies (metabolomics, transcriptomics) to elucidate microbiota-driven fibrotic mechanisms. Additionally, well-designed RCTs evaluating the efficacy of microbiota-targeted therapies, including TCM, are crucial for translating preclinical insights into clinical applications. In summary, intestinal microbiota and their metabolites play a notable role in IBD-associated intestinal fibrosis. Strategies that modulate gut microbiota represent a promising therapeutic avenue, with TCM offering unique advantages in restoring microbial balance and mitigating fibrosis (Figure 4). However, rigorous research is necessary to optimize TCM interventions, address current limitations, and establish a robust foundation for future clinical applications.
Conclusion
Intestinal fibrosis is a common and challenging complication of IBD, with gut microbiota dysbiosis playing a crucial role in its development. Accumulating evidence suggests that alterations in gut microbiota composition and its metabolites contribute to intestinal fibrosis through multiple mechanisms. Targeting the gut microbiota and its metabolites has emerged as a promising strategy for managing intestinal fibrosis in IBD. TCM has shown efficacy in regulating the intestinal microbiota, influencing the production of intestinal microbiota metabolites, and providing notable benefits in alleviating intestinal fibrosis. TCM formulas, single herbs, and monomers exert beneficial effects by enriching beneficial bacteria, inhibiting pathogenic species, and regulating key metabolic pathways. These findings highlight the potential of TCM as a therapeutic approach for IBD-associated fibrosis. Further studies are warranted to elucidate the precise mechanisms and optimize microbiota-targeted interventions for improved clinical outcomes.
Abbreviations
IBD, Inflammatory Bowel Disease; CD, Crohn’s Disease; UC, Ulcerative Colitis; ECM, extracellular matrix; TGF-β1, transforming growth factor-β1; AIEC, Adherent-invasive Escherichia coli; α-SMA, α-smooth muscle actin; AMPs, antimicrobial peptides; LPS, lipopolysaccharide; Bas, bile acids; SCFAs, short-chain fatty acids; SBAs, secondary bile acids; IA, indoleacrylic acid; AHR, aryl hydrocarbon receptor; MMPs, matrix metalloproteinases; TIMPs, tissue inhibitors of matrix metalloproteinases; IGF-I, insulin-like growth factor-1; MAPK, mitogen-activated protein kinase; let-7b, exosomal miRNA lethal-7b; SUCNR1, succinate receptor 1; NF-κB, nuclear factor κ-B; TLR4, Toll-like receptor 4; H2S, hydrogen sulfide; ASR, sulfate reduction; Treg, regulatory T; TH17, T helper cells 17; IPA, Indole-3-propionic acid; PXR, pregnane X receptor; Foxp3, forkhead transcription factor p-3; GPR43, G protein-coupled receptor 43; HFD, high-fat diets; CDED, Crohn’s Disease Exclusion Diet; PEN, partial enteral nutrition; CD-TREAT, Crohn’s Disease-TREAT; EEN, Exclusive Enteral Nutrition; UCED, Ulcerative colitis exclusion diet; FODMAP, fermentable oligo-, di-, and mono-saccharides and polyols; PPARγ, peroxisome proliferator-activated receptor γ; DSS, dextran sulfate sodium-induced; FMT, fecal microbiota transplantation; NMN, nicotinamide mononucleotide; FGF19, fibroblast growth factor 19; EVs, extracellular vesicles; NLRP3, NOD like receptor thermal protein domain associated protein 3; FGL-1, fibrinogen-like protein 1; EndoMT, endothelial-to-mesenchymal transition; GENs, exosome-like nanoparticles; RCTs, randomized controlled trials.
Acknowledgments
We gratefully acknowledge the support from the Domestic First-class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine (4901-020000200207), and the Hunan Province “Double First-Class” Discipline Construction Project of Pediatrics of Traditional Chinese Medicine at Hunan University of Chinese Medicine (Approved No. [2022]14, issued by the Department of Education of Hunan Province). We also thank all the authors for their dedication to this study.
Funding
This study was supported by the Domestic First-class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine (4901-020000200207), and the Hunan Province “Double First-Class” Discipline Construction Project of Pediatrics of Traditional Chinese Medicine at Hunan University of Chinese Medicine(Approved No. [2022]14, issued by the Department of Education of Hunan Province).
Disclosure
The authors report no conflicts of interest in this work.
References
1. Ciorba MA, Konnikova L, Hirota SA, et al. Challenges in IBD research 2024: preclinical human IBD mechanisms. Inflamm Bowel Dis. 2024;30(2):S5–s18. doi:10.1093/ibd/izae081
2. Liu S, Zhao W, Lan P, Mou X. The microbiome in inflammatory bowel diseases: from pathogenesis to therapy. Protein Cell. 2021;12(5):331–345. doi:10.1007/s13238-020-00745-3
3. Ungaro F, Massimino L, D’Alessio S, Danese S. The gut virome in inflammatory bowel disease pathogenesis: from metagenomics to novel therapeutic approaches. United Eur Gastroenterol J. 2019;7(8):999–1007. doi:10.1177/2050640619876787
4. Lu Q, Yang MF, Liang YJ, et al. Immunology of inflammatory bowel disease: molecular mechanisms and therapeutics. J Inflamm Res. 2022;15:1825–1844. doi:10.2147/jir.S353038
5. Ni J, Wu GD, Albenberg L, Tomov VT. Gut microbiota and IBD: causation or correlation? Nat Rev Gastroenterol Hepatol. 2017;14(10):573–584. doi:10.1038/nrgastro.2017.88
6. Lavelle A, Sokol H. Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2020;17(4):223–237. doi:10.1038/s41575-019-0258-z
7. Akutko K, Stawarski A. Probiotics, prebiotics and synbiotics in inflammatory bowel diseases. J Clin Med. 2021;10(11):2466. doi:10.3390/jcm10112466
8. Leong RWL. The significance of granulomas in Crohn’s disease and inflammatory bowel disease epidemiology in Asia. J Gastroenterol Hepatol. 2020;35(4):523–524. doi:10.1111/jgh.15018
9. Gordon IO, Abushamma S, Kurowski JA, et al. Paediatric ulcerative colitis is a fibrotic disease and is linked with chronicity of inflammation. J Crohns Colitis. 2022;16(5):804–821. doi:10.1093/ecco-jcc/jjab216
10. Gordon IO, Agrawal N, Willis E, et al. Fibrosis in ulcerative colitis is directly linked to severity and chronicity of mucosal inflammation. Aliment Pharmacol Ther. 2018;47(7):922–939. doi:10.1111/apt.14526
11. Scharl M, Huber N, Lang S, Fürst A, Jehle E, Rogler G. Hallmarks of epithelial to mesenchymal transition are detectable in Crohn’s disease associated intestinal fibrosis. Clin Transl Med. 2015;4:1. doi:10.1186/s40169-015-0046-5
12. Ng SC, Kaplan GG, Tang W, et al. Population density and risk of inflammatory bowel disease: a prospective population-based study in 13 countries or regions in Asia-Pacific. Am J Gastroenterol. 2019;114(1):107–115. doi:10.1038/s41395-018-0233-2
13. Rieder F, Fiocchi C, Rogler G. Mechanisms, management, and treatment of fibrosis in patients with inflammatory bowel diseases. Gastroenterology. 2017;152(2):340–350.e6. doi:10.1053/j.gastro.2016.09.047
14. D’Alessio S, Ungaro F, Noviello D, Lovisa S, Peyrin-Biroulet L, Danese S. Revisiting fibrosis in inflammatory bowel disease: the gut thickens. Nat Rev Gastroenterol Hepatol. 2022;19(3):169–184. doi:10.1038/s41575-021-00543-0
15. Lu C, Baraty B, Lee Robertson H, et al. Systematic review: medical therapy for fibrostenosing Crohn’s disease. Aliment Pharmacol Ther. 2020;51(12):1233–1246. doi:10.1111/apt.15750
16. Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol. 2019;16(1):35–56. doi:10.1038/s41575-018-0061-2
17. Lavelle A, Hoffmann TW, Pham HP, Langella P, Guédon E, Sokol H. Baseline microbiota composition modulates antibiotic-mediated effects on the gut microbiota and host. Microbiome. 2019;7(1):111. doi:10.1186/s40168-019-0725-3
18. Schirmer M, Garner A, Vlamakis H, Xavier RJ. Microbial genes and pathways in inflammatory bowel disease. Nat Rev Microbiol. 2019;17(8):497–511. doi:10.1038/s41579-019-0213-6
19. Alipour M, Zaidi D, Valcheva R, et al. Mucosal barrier depletion and loss of bacterial diversity are primary abnormalities in paediatric ulcerative colitis. J Crohns Colitis. 2016;10(4):462–471. doi:10.1093/ecco-jcc/jjv223
20. Mehandru S, Colombel JF. The intestinal barrier, an arbitrator turned provocateur in IBD. Nat Rev Gastroenterol Hepatol. 2021;18(2):83–84. doi:10.1038/s41575-020-00399-w
21. Paone P, Cani PD. Mucus barrier, mucins and gut microbiota: the expected slimy partners? Gut. 2020;69(12):2232–2243. doi:10.1136/gutjnl-2020-322260
22. Franzosa EA, Sirota-Madi A, Avila-Pacheco J, et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat Microbiol. 2019;4(2):293–305. doi:10.1038/s41564-018-0306-4
23. Ott SJ, Musfeldt M, Wenderoth DF, et al. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut. 2004;53(5):685–693. doi:10.1136/gut.2003.025403
24. Vich Vila A, Imhann F, Collij V, et al. Gut microbiota composition and functional changes in inflammatory bowel disease and irritable bowel syndrome. Sci Transl Med. 2018;10(472):eaap8914. doi:10.1126/scitranslmed.aap8914
25. Mourelle M, Salas A, Guarner F, Crespo E, García-Lafuente A, Malagelada JR. Stimulation of transforming growth factor beta1 by enteric bacteria in the pathogenesis of rat intestinal fibrosis. Gastroenterology. 1998;114(3):519–526. doi:10.1016/s0016-5085(98)70535-9
26. Imai J, Kitamoto S, Sugihara K, et al. Flagellin-mediated activation of IL-33-ST2 signaling by a pathobiont promotes intestinal fibrosis. Mucosal Immunol. 2019;12(3):632–643. doi:10.1038/s41385-019-0138-4
27. Park JS, Choi JW, Jhun J, et al. Lactobacillus acidophilus improves intestinal inflammation in an acute colitis mouse model by regulation of Th17 and treg cell balance and fibrosis development. J Med Food. 2018;21(3):215–224. doi:10.1089/jmf.2017.3990
28. Bernardi F, D’Amico F, Bencardino S, et al. Gut microbiota metabolites: unveiling their role in inflammatory bowel diseases and fibrosis. Pharmaceuticals. 2024;17(3):347. doi:10.3390/ph17030347
29. Sinha SR, Haileselassie Y, Nguyen LP, et al. Dysbiosis-induced secondary bile acid deficiency promotes intestinal inflammation. Cell Host Microbe. 2020;27(4):659–670.e5. doi:10.1016/j.chom.2020.01.021
30. Gonçalves P, Araújo JR, Di Santo JP. A cross-talk between microbiota-derived short-chain fatty acids and the host mucosal immune system regulates intestinal homeostasis and inflammatory bowel disease. Inflamm Bowel Dis. 2018;24(3):558–572. doi:10.1093/ibd/izx029
31. Handa O, Miura H, Gu T, et al. Reduction of butyric acid-producing bacteria in the ileal mucosa-associated microbiota is associated with the history of abdominal surgery in patients with Crohn’s disease. Redox Rep. 2023;28(1):2241615. doi:10.1080/13510002.2023.2241615
32. Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun. 2018;9(1):3294. doi:10.1038/s41467-018-05470-4
33. Burke JP, Cunningham MF, Watson RW, Docherty NG, Coffey JC, O’Connell PR. Bacterial lipopolysaccharide promotes profibrotic activation of intestinal fibroblasts. Br J Surg. 2010;97(7):1126–1134. doi:10.1002/bjs.7045
34. Rieder F, Kessler S, Sans M, Fiocchi C. Animal models of intestinal fibrosis: new tools for the understanding of pathogenesis and therapy of human disease. Am J Physiol Gastrointest Liver Physiol. 2012;303(7):G786–801. doi:10.1152/ajpgi.00059.2012
35. Small CL, Reid-Yu SA, McPhee JB, Coombes BK. Persistent infection with Crohn’s disease-associated adherent-invasive Escherichia coli leads to chronic inflammation and intestinal fibrosis. Nat Commun. 2013;4:1957. doi:10.1038/ncomms2957
36. Xu Y, Qian W, Huang L, et al. Crohn’s disease-associated AIEC inhibiting intestinal epithelial cell-derived exosomal let-7b expression regulates macrophage polarization to exacerbate intestinal fibrosis. Gut Microbes. 2023;15(1):2193115. doi:10.1080/19490976.2023.2193115
37. Zhao S, Dejanovic D, Yao P, et al. Selective deletion of MyD88 signaling in α-SMA positive cells ameliorates experimental intestinal fibrosis via post-transcriptional regulation. Mucosal Immunol. 2020;13(4):665–678. doi:10.1038/s41385-020-0259-9
38. Suau R, Pardina E, Domènech E, Lorén V, Manyé J. The complex relationship between microbiota, immune response and creeping fat in Crohn’s disease. J Crohns Colitis. 2022;16(3):472–489. doi:10.1093/ecco-jcc/jjab159
39. Ha CWY, Martin A, Sepich-Poore GD, et al. Translocation of viable gut microbiota to mesenteric adipose drives formation of creeping fat in humans. Cell. 2020;183(3):666–683.e17. doi:10.1016/j.cell.2020.09.009
40. Watanabe D, Kamada N. Contribution of the gut microbiota to intestinal fibrosis in Crohn’s disease. Front Med Lausanne. 2022;9:826240. doi:10.3389/fmed.2022.826240
41. Otte JM, Rosenberg IM, Podolsky DK. Intestinal myofibroblasts in innate immune responses of the intestine. Gastroenterology. 2003;124(7):1866–1878. doi:10.1016/s0016-5085(03)00403-7
42. Yu S, Sun Y, Shao X, et al. Leaky gut in IBD: intestinal barrier-gut microbiota interaction. J Microbiol Biotechnol. 2022;32(7):825–834. doi:10.4014/jmb.2203.03022
43. Wang J, Yang B, Chandra J, Ivanov A, Brown JM, Rieder F. Preventing fibrosis in IBD: update on immune pathways and clinical strategies. Expert Rev Clin Immunol. 2024;20(7):727–734. doi:10.1080/1744666x.2024.2330604
44. Jacob N, Jacobs JP, Kumagai K, et al. Inflammation-independent TL1A-mediated intestinal fibrosis is dependent on the gut microbiome. Mucosal Immunol. 2018;11(5):1466–1476. doi:10.1038/s41385-018-0055-y
45. Xu WD, Li R, Huang AF. Role of TL1A in inflammatory autoimmune diseases: a comprehensive review. Front Immunol. 2022;13:891328. doi:10.3389/fimmu.2022.891328
46. Zhao X, Yang W, Yu T, et al. Th17 cell-derived amphiregulin promotes colitis-associated intestinal fibrosis through activation of mTOR and MEK in intestinal myofibroblasts. Gastroenterology. 2023;164(1):89–102. doi:10.1053/j.gastro.2022.09.006
47. Valatas V, Filidou E, Drygiannakis I, Kolios G. Stromal and immune cells in gut fibrosis: the myofibroblast and the scarface. Ann Gastroenterol. 2017;30(4):393–404. doi:10.20524/aog.2017.0146
48. Rieder F, Schleder S, Wolf A, et al. Association of the novel serologic anti-glycan antibodies anti-laminarin and anti-chitin with complicated Crohn’s disease behavior. Inflamm Bowel Dis. 2010;16(2):263–274. doi:10.1002/ibd.21046
49. Yoo JH, Ho S, Tran DH, et al. Anti-fibrogenic effects of the anti-microbial peptide cathelicidin in murine colitis-associated fibrosis. Cell Mol Gastroenterol Hepatol. 2015;1(1):55–74.e1. doi:10.1016/j.jcmgh.2014.08.001
50. Jun YK, Kwon SH, Yoon HT, et al. Toll-like receptor 4 regulates intestinal fibrosis via cytokine expression and epithelial-mesenchymal transition. Sci Rep. 2020;10(1):19867. doi:10.1038/s41598-020-76880-y
51. Macias-Ceja DC, Ortiz-Masiá D, Salvador P, et al. Succinate receptor mediates intestinal inflammation and fibrosis. Mucosal Immunol. 2019;12(1):178–187. doi:10.1038/s41385-018-0087-3
52. Luo W, Zhao M, Dwidar M, et al. Microbial assimilatory sulfate reduction-mediated H(2)S: an overlooked role in Crohn’s disease development. Microbiome. 2024;12(1):152. doi:10.1186/s40168-024-01873-2
53. Blachier F, Andriamihaja M, Larraufie P, Ahn E, Lan A, Kim E. Production of hydrogen sulfide by the intestinal microbiota and epithelial cells and consequences for the colonic and rectal mucosa. Am J Physiol Gastrointest Liver Physiol. 2021;320(2):G125–g135. doi:10.1152/ajpgi.00261.2020
54. Wlodarska M, Luo C, Kolde R, et al. Indoleacrylic acid produced by commensal peptostreptococcus species suppresses inflammation. Cell Host Microbe. 2017;22(1):25–37.e6. doi:10.1016/j.chom.2017.06.007
55. Pernomian L, Duarte-Silva M, de Barros Cardoso CR. The aryl hydrocarbon receptor (AHR) as a potential target for the control of intestinal inflammation: insights from an immune and bacteria sensor receptor. Clin Rev Allergy Immunol. 2020;59(3):382–390. doi:10.1007/s12016-020-08789-3
56. Song X, Sun X, Oh SF, et al. Microbial bile acid metabolites modulate gut RORγ(+) regulatory T cell homeostasis. Nature. 2020;577(7790):410–415. doi:10.1038/s41586-019-1865-0
57. Flannigan KL, Nieves KM, Szczepanski HE, et al. The pregnane X receptor and indole-3-propionic acid shape the intestinal mesenchyme to restrain inflammation and fibrosis. Cell Mol Gastroenterol Hepatol. 2023;15(3):765–795. doi:10.1016/j.jcmgh.2022.10.014
58. Arpaia N, Campbell C, Fan X, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–455. doi:10.1038/nature12726
59. Smith PM, Howitt MR, Panikov N, et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341(6145):569–573. doi:10.1126/science.1241165
60. Cicchinelli S, Gemma S, Pignataro G, et al. Intestinal fibrogenesis in inflammatory bowel diseases: exploring the potential role of gut microbiota metabolites as modulators. Pharmaceuticals. 2024;17(4):490. doi:10.3390/ph17040490
61. Sasson AN, Ananthakrishnan AN, Raman M. Diet in treatment of inflammatory bowel diseases. Clin Gastroenterol Hepatol. 2021;19(3):425–435.e3. doi:10.1016/j.cgh.2019.11.054
62. Fiorindi C, Russo E, Balocchini L, Amedei A, Giudici F. Inflammatory bowel disease and customized nutritional intervention focusing on gut microbiome balance. Nutrients. 2022;14(19):4117. doi:10.3390/nu14194117
63. Rizzello F, Spisni E, Giovanardi E, et al. Implications of the westernized diet in the onset and progression of IBD. Nutrients. 2019;11(5):1033. doi:10.3390/nu11051033
64. Amamou A, Rouland M, Yaker L, et al. Dietary salt exacerbates intestinal fibrosis in chronic TNBS colitis via fibroblasts activation. Sci Rep. 2021;11(1):15055. doi:10.1038/s41598-021-94280-8
65. Gonza I, Goya-Jorge E, Douny C, et al. Food additives impair gut microbiota from healthy individuals and IBD patients in a colonic in vitro fermentation model. Food Res Int. 2024;182:114157. doi:10.1016/j.foodres.2024.114157
66. Levine A, Wine E, Assa A, et al. Crohn’s disease exclusion diet plus partial enteral nutrition induces sustained remission in a randomized controlled trial. Gastroenterology. 2019;157(2):440–450.e8. doi:10.1053/j.gastro.2019.04.021
67. Sarbagili-Shabat C, Albenberg L, Van Limbergen J, et al. A novel UC exclusion diet and antibiotics for treatment of mild to moderate pediatric ulcerative colitis: a prospective open-label pilot study. Nutrients. 2021;13(11):3736. doi:10.3390/nu13113736
68. De Filippis F, Pellegrini N, Vannini L, et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut. 2016;65(11):1812–1821. doi:10.1136/gutjnl-2015-309957
69. Suskind DL, Cohen SA, Brittnacher MJ, et al. Clinical and fecal microbial changes with diet therapy in active inflammatory bowel disease. J Clin Gastroenterol. 2018;52(2):155–163. doi:10.1097/mcg.0000000000000772
70. Speca S, Rousseaux C, Dubuquoy C, et al. Novel PPARγ modulator GED-0507-34 levo ameliorates inflammation-driven intestinal fibrosis. Inflamm Bowel Dis. 2016;22(2):279–292. doi:10.1097/mib.0000000000000618
71. Monteleone I, Zorzi F, Marafini I, et al. Aryl hydrocarbon receptor-driven signals inhibit collagen synthesis in the gut. Eur J Immunol. 2016;46(4):1047–1057. doi:10.1002/eji.201445228
72. Yu M, Wu H, Wang J, et al. Vitamin D receptor inhibits EMT via regulation of the epithelial mitochondrial function in intestinal fibrosis. J Biol Chem. 2021;296:100531. doi:10.1016/j.jbc.2021.100531
73. Pascal V, Pozuelo M, Borruel N, et al. A microbial signature for Crohn’s disease. Gut. 2017;66(5):813–822. doi:10.1136/gutjnl-2016-313235
74. Litvak Y, Byndloss MX, Bäumler AJ. Colonocyte metabolism shapes the gut microbiota. Science. 2018;362(6418):eaat9076. doi:10.1126/science.aat9076
75. Furusawa Y, Obata Y, Fukuda S, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446–450. doi:10.1038/nature12721
76. Zheng M, Han R, Yuan Y, et al. The role of Akkermansia muciniphila in inflammatory bowel disease: current knowledge and perspectives. Front Immunol. 2022;13:1089600. doi:10.3389/fimmu.2022.1089600
77. Jang SE, Jeong JJ, Kim JK, Han MJ, Kim DH. Simultaneous amelioratation of colitis and liver injury in mice by bifidobacterium longum LC67 and lactobacillus plantarum LC27. Sci Rep. 2018;8(1):7500. doi:10.1038/s41598-018-25775-0
78. Niu MM, Guo HX, Cai JW, Meng XC. Bifidobacterium breve alleviates DSS-induced colitis in mice by maintaining the mucosal and epithelial barriers and modulating gut microbes. Nutrients. 2022;14(18):3671. doi:10.3390/nu14183671
79. Sivananthan K, Petersen AM. Review of Saccharomyces boulardii as a treatment option in IBD. Immunopharmacol Immunotoxicol. 2018;40(6):465–475. doi:10.1080/08923973.2018.1469143
80. Rasmussen HE, Hamaker BR. Prebiotics and inflammatory bowel disease. Gastroenterol Clin North Am. 2017;46(4):783–795. doi:10.1016/j.gtc.2017.08.004
81. Pandey KR, Naik SR, Vakil BV. Probiotics, prebiotics and synbiotics- a review. J Food Sci Technol. 2015;52(12):7577–7587. doi:10.1007/s13197-015-1921-1
82. Borody TJ, Clancy A. Fecal microbiota transplantation for ulcerative colitis-where to from here? Transl Gastroenterol Hepatol. 2019;4:48. doi:10.21037/tgh.2019.06.04
83. Jadhav A, Jagtap S, Vyavahare S, Sharbidre A, Kunchiraman B. Reviewing the potential of probiotics, prebiotics and synbiotics: advancements in treatment of ulcerative colitis. Front Cell Infect Microbiol. 2023;13:1268041. doi:10.3389/fcimb.2023.1268041
84. Gomes-Santos AC, de Oliveira RP, Moreira TG, et al. Hsp65-producing Lactococcus lactis prevents inflammatory intestinal disease in mice by IL-10- and TLR2-dependent pathways. Front Immunol. 2017;8:30. doi:10.3389/fimmu.2017.00030
85. Zhao X, Ji K, Zhang M, et al. NMN alleviates radiation-induced intestinal fibrosis by modulating gut microbiota. Int J Radiat Biol. 2023;99(5):823–834. doi:10.1080/09553002.2023.2145029
86. Gadaleta RM, Garcia-Irigoyen O, Cariello M, et al. Fibroblast growth factor 19 modulates intestinal microbiota and inflammation in presence of farnesoid X receptor. EBioMedicine. 2020;54:102719. doi:10.1016/j.ebiom.2020.102719
87. Tao S, Fan J, Li J, et al. Extracellular vesicles derived from Lactobacillus johnsonii promote gut barrier homeostasis by enhancing M2 macrophage polarization. J Adv Res. 2024;69:545–563. doi:10.1016/j.jare.2024.03.011
88. Yang J, Li Y, Zeng L, Zhao FR. Traditional Chinese medicine against fibrotic diseases: a review. Chin J Exp Traditional Med Formulae. 2024;1–9. doi:10.13422/j.cnki.syfjx.20231837
89. Li Z, Shen Y, Xin J, et al. Cryptotanshinone alleviates radiation-induced lung fibrosis via modulation of gut microbiota and bile acid metabolism. Phytother Res. 2023;37(10):4557–4571. doi:10.1002/ptr.7926
90. Xue X, Zhou H, Gao J, et al. The impact of traditional Chinese medicine and dietary compounds on modulating gut microbiota in hepatic fibrosis: a review. Heliyon. 2024;10(19):e38339. doi:10.1016/j.heliyon.2024.e38339
91. Zhao Y, Zhao M, Zhang Y, et al. Bile acids metabolism involved in the beneficial effects of Danggui Shaoyao San via gut microbiota in the treatment of CCl(4) induced hepatic fibrosis. J Ethnopharmacol. 2024;319(Pt 3):117383. doi:10.1016/j.jep.2023.117383
92. Zhang J, Wang W, Cui X, et al. Ganoderma lucidum ethanol extracts ameliorate hepatic fibrosis and promote the communication between metabolites and gut microbiota g_Ruminococcus through the NF-κB and TGF-β1/Smads pathways. J Ethnopharmacol. 2024;322:117656. doi:10.1016/j.jep.2023.117656
93. Sutariya B, Saraf M. Betanin, isolated from fruits of Opuntia elatior Mill attenuates renal fibrosis in diabetic rats through regulating oxidative stress and TGF-β pathway. J Ethnopharmacol. 2017;198:432–443. doi:10.1016/j.jep.2016.12.048
94. Wang J, Guo X, Zou Z, et al. Ootheca mantidis mitigates renal fibrosis in mice by the suppression of apoptosis via increasing the gut microbe Akkermansia muciniphila and modulating glutamine metabolism. Biomed Pharmacother. 2023;166:115434. doi:10.1016/j.biopha.2023.115434
95. Zhu M, Song Y, Xu Y, Xu H. Manipulating microbiota in inflammatory bowel disease treatment: clinical and natural product interventions explored. Int J Mol Sci. 2023;24(13):11004. doi:10.3390/ijms241311004
96. Yuan S, Li Y, Li J, et al. Traditional Chinese medicine and natural products: potential approaches for inflammatory bowel disease. Front Pharmacol. 2022;13:892790. doi:10.3389/fphar.2022.892790
97. Guo M, Wu Y, Peng M, Xiao N, Lei Z, Tan Z. Decreasing of trimethylamine N-oxide by cecal microbiota and choline-trimethylamine lyase are associated with sishen pill on diarrhea with kidney-yang deficiency syndrome. J Inflamm Res. 2024;17:7275–7294. doi:10.2147/jir.S470254
98. Liu B, Piao X, Niu W, et al. Kuijieyuan decoction improved intestinal barrier injury of ulcerative colitis by affecting TLR4-dependent PI3K/AKT/NF-κB oxidative and inflammatory signaling and gut microbiota. Front Pharmacol. 2020;11:1036. doi:10.3389/fphar.2020.01036
99. Yuan Z, Yang L, Zhang X, Ji P, Wei Y. Therapeutic effect of n-butanol fraction of Huang-lian-Jie-du Decoction on ulcerative colitis and its regulation on intestinal flora in colitis mice. Biomed Pharmacother. 2020;121:109638. doi:10.1016/j.biopha.2019.109638
100. Su S, Liu T, Zheng JY, et al. Huang Lian Jie Du decoction attenuated colitis via suppressing the macrophage Csf1r/Src pathway and modulating gut microbiota. Front Immunol. 2024;15:1375781. doi:10.3389/fimmu.2024.1375781
101. Yang J, Miao L, Xue Y, Wang X. Yiyi Fuzi Baijiang powder alleviates dextran sulfate sodium-induced ulcerative colitis in rats via inhibiting the TLR4/NF-κB/NLRP3 inflammasome signaling pathway to repair the intestinal epithelial barrier, and modulating intestinal microbiota. Oxid Med Cell Longev. 2023;2023:3071610. doi:10.1155/2023/3071610
102. Wang Y, Zhang J, Xu L, et al. Modified gegen qinlian decoction regulates Treg/Th17 balance to ameliorate DSS-induced acute experimental colitis in mice by altering the gut microbiota. Front Pharmacol. 2021;12:756978. doi:10.3389/fphar.2021.756978
103. Luo S, Wen R, Wang Q, et al. Rhubarb Peony Decoction ameliorates ulcerative colitis in mice by regulating gut microbiota to restoring Th17/Treg balance. J Ethnopharmacol. 2019;231:39–49. doi:10.1016/j.jep.2018.08.033
104. Wei X, Liang J, Liu J, et al. Anchang Yuyang Decoction inhibits experimental colitis-related carcinogenesis by regulating PPAR signaling pathway and affecting metabolic homeostasis of host and microbiota. J Ethnopharmacol. 2024;326:117995. doi:10.1016/j.jep.2024.117995
105. Hua YL, Jia YQ, Zhang XS, et al. Baitouweng Tang ameliorates DSS-induced ulcerative colitis through the regulation of the gut microbiota and bile acids via pathways involving FXR and TGR5. Biomed Pharmacother. 2021;137:111320. doi:10.1016/j.biopha.2021.111320
106. Zhou Y, Feng Y, Cen R, et al. San-Wu-Huang-Qin decoction attenuates tumorigenesis and mucosal barrier impairment in the AOM/DSS model by targeting gut microbiome. Phytomedicine. 2022;98:153966. doi:10.1016/j.phymed.2022.153966
107. Cheng Y, Li J, Zhang X, et al. Protective effect of qingchang wenzhong decoction on colitis and colitis-related carcinogenesis by regulating inflammation and intestinal fibrosis. J Inflamm Res. 2023;16:1479–1495. doi:10.2147/jir.S402395
108. Gao Y, Lu LJ, Zhang ZZ, et al. Xue-Jie-San prevents the early development of colitis-associated intestinal fibrosis by blocking Notch1 and FGL1 signaling pathways. J Ethnopharmacol. 2023;315:116678. doi:10.1016/j.jep.2023.116678
109. Gao Y, Zhang Z, Du J, et al. Xue-Jie-San restricts ferroptosis in Crohn’s disease via inhibiting FGL1/NF-κB/STAT3 positive feedback loop. Front Pharmacol. 2023;14:1148770. doi:10.3389/fphar.2023.1148770
110. Duan ZL, Wang YJ, Lu ZH, et al. Wumei Wan attenuates angiogenesis and inflammation by modulating RAGE signaling pathway in IBD: network pharmacology analysis and experimental evidence. Phytomedicine. 2023;111:154658. doi:10.1016/j.phymed.2023.154658
111. Li Y, Hu J, Au R, et al. Therapeutic effects of qingchang tongluo decoction on intestinal fibrosis in crohn’s disease: network pharmacology, molecular docking and experiment validation. Drug Des Devel Ther. 2024;18:3269–3293. doi:10.2147/dddt.S458811
112. Qian H, Ye Z, Hu Y, et al. Molecular targets associated with ulcerative colitis and the benefits of atractylenolides-based therapy. Front Pharmacol. 2024;15:1398294. doi:10.3389/fphar.2024.1398294
113. Luo Y, Jiang J, Cheng J, et al. Inhibitory effects of Rhein on renal interstitial fibrosis via the SHH-Gli1 signal pathway. Evid Based Complement Alternat Med. 2022;2022:4398265. doi:10.1155/2022/4398265
114. Barbosa DM, Fahlbusch P, Herzfeld de Wiza D, et al. Rhein, a novel Histone Deacetylase (HDAC) inhibitor with antifibrotic potency in human myocardial fibrosis. Sci Rep. 2020;10(1):4888. doi:10.1038/s41598-020-61886-3
115. Wu J, Wei Z, Cheng P, et al. Rhein modulates host purine metabolism in intestine through gut microbiota and ameliorates experimental colitis. Theranostics. 2020;10(23):10665–10679. doi:10.7150/thno.43528
116. Tao Q, Liang Q, Fu Y, et al. Puerarin ameliorates colitis by direct suppression of macrophage M1 polarization in DSS mice. Phytomedicine. 2024;135:156048. doi:10.1016/j.phymed.2024.156048
117. Long J, Liu XK, Kang ZP, et al. Ginsenoside Rg1 ameliorated experimental colitis by regulating the balance of M1/M2 macrophage polarization and the homeostasis of intestinal flora. Eur J Pharmacol. 2022;917:174742. doi:10.1016/j.ejphar.2022.174742
118. Zhang Y, Wu Z, Liu J, et al. Identification of the core active structure of a Dendrobium officinale polysaccharide and its protective effect against dextran sulfate sodium-induced colitis via alleviating gut microbiota dysbiosis. Food Res Int. 2020;137:109641. doi:10.1016/j.foodres.2020.109641
119. Duan Y, Huang J, Sun M, et al. Poria cocos polysaccharide improves intestinal barrier function and maintains intestinal homeostasis in mice. Int J Biol Macromol. 2023;249:125953. doi:10.1016/j.ijbiomac.2023.125953
120. Su L, Mao C, Wang X, et al. The anti-colitis effect of schisandra chinensis polysaccharide is associated with the regulation of the composition and metabolism of gut microbiota. Front Cell Infect Microbiol. 2020;10:519479. doi:10.3389/fcimb.2020.519479
121. Zhao HQ, Lu NH, Su YD, Jin ZHY, Fu YJ, Li ZP. Investigation of the mechanism of patrinia scabiosifolia on intestinal fibrosis in rats with IBD based on intestinal flora regulation of β-defensin 2. Acta Neuropharmacologica. 2023;13(01):8–15+29.
122. Wang R, Wang D, Wang H, et al. Therapeutic targeting of Nrf2 signaling by maggot extracts ameliorates inflammation-associated intestinal fibrosis in chronic DSS-induced colitis. Front Immunol. 2021;12:670159. doi:10.3389/fimmu.2021.670159
123. Li W, Ding J, Chen S, et al. Alleviation of colitis by honeysuckle MIR2911 via direct regulation of gut microbiota. J Control Release. 2024;376:123–137. doi:10.1016/j.jconrel.2024.09.050
124. Lee JH, Massagué J. TGF-β in developmental and fibrogenic EMTs. Semin Cancer Biol. 2022;86(Pt 2):136–145. doi:10.1016/j.semcancer.2022.09.004
125. Peng D, Fu M, Wang M, Wei Y, Wei X. Targeting TGF-β signal transduction for fibrosis and cancer therapy. Mol Cancer. 2022;21(1):104. doi:10.1186/s12943-022-01569-x
126. Liu J, Deng T, Wang Y, et al. Calycosin inhibits intestinal fibrosis on CCD-18Co cells via modulating transforming growth factor-β/smad signaling pathway. Pharmacology. 2019;104(1–2):81–89. doi:10.1159/000500186
127. Fontani F, Marcucci T, Picariello L, Tonelli F, Vincenzini MT, Iantomasi T. Redox regulation of MMP-3/TIMP-1 ratio in intestinal myofibroblasts: effect of N-acetylcysteine and curcumin. Exp Cell Res. 2014;323(1):77–86. doi:10.1016/j.yexcr.2014.02.019
128. Xu S, Jiang B, Wang H, Shen C, Chen H, Zeng L. Curcumin suppresses intestinal fibrosis by inhibition of PPARγ-mediated epithelial-mesenchymal transition. Evid Based Complement Alternat Med. 2017;2017:7876064. doi:10.1155/2017/7876064
129. Kim J, Zhang S, Zhu Y, Wang R, Wang J. Amelioration of colitis progression by ginseng-derived exosome-like nanoparticles through suppression of inflammatory cytokines. J Ginseng Res. 2023;47(5):627–637. doi:10.1016/j.jgr.2023.01.004
130. Qiao L, Fang L, Zhu J, et al. Total flavone of abelmoschus manihot ameliorates TNBS-induced colonic fibrosis by regulating Th17/treg balance and reducing extracellular matrix. Front Pharmacol. 2021;12:769793. doi:10.3389/fphar.2021.769793
131. Glassner KL, Abraham BP, Quigley EMM. The microbiome and inflammatory bowel disease. J Allergy Clin Immunol. 2020;145(1):16–27. doi:10.1016/j.jaci.2019.11.003
132. Rieder F, Mukherjee PK, Massey WJ, Wang Y, Fiocchi C. Fibrosis in IBD: from pathogenesis to therapeutic targets. Gut. 2024;73(5):854–866. doi:10.1136/gutjnl-2023-329963
133. Xie S, Deng N, Fang L, Shen J, Tan Z, Cai Y. TMAO is involved in kidney-yang deficiency syndrome diarrhea by mediating the “gut-kidney axis”. Heliyon. 2024;10(15):e35461. doi:10.1016/j.heliyon.2024.e35461
134. Mirsepasi-Lauridsen HC, Vallance BA, Krogfelt KA, Petersen AM. Escherichia coli pathobionts associated with inflammatory bowel disease. Clin Microbiol Rev. 2019;32(2):100–128. doi:10.1128/cmr.00060-18
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles

The Role of Gut Microbiota in the Progression of Parkinson’s Disease and the Mechanism of Intervention by Traditional Chinese Medicine
Huan P, Wang L, He Z, He J
Neuropsychiatric Disease and Treatment 2022, 18:1507-1520
Published Date: 26 July 2022

Mesenchymal Stem Cells Ameliorate DSS-Induced Experimental Colitis by Modulating the Gut Microbiota and MUC-1 Pathway
Wang H, Sun Y, Xiao FJ, Zhao X, Zhang WY, Xia YJ, Wang LS
Journal of Inflammation Research 2023, 16:2023-2039
Published Date: 11 May 2023

Gut Microbiota and Aging: Traditional Chinese Medicine and Modern Medicine
Li J, Li D, Chen Y, Chen W, Xu J, Gao L
Clinical Interventions in Aging 2023, 18:963-986
Published Date: 17 June 2023
Drug Repurposing in Crohn’s Disease Using Danish Real-World Data
Shakibfar S, Allin KH, Jess T, Barbieri MA, Battini V, Simoncic E, Kirchgesner J, Ulven T, Sessa M
Pragmatic and Observational Research 2024, 15:17-29
Published Date: 21 February 2024
Microbial Disruptions in Inflammatory Bowel Disease: A Comparative Analysis
Ma J, Wang K, Wang J, Zeng Q, Liu K, Zheng S, Chen Y, Yao J
International Journal of General Medicine 2024, 17:1355-1367
Published Date: 6 April 2024