1
|
Tian J, Wang W, Liu Y, Zhang X, Zhao H, Qu H. Role of endoscopic ultrasound as a predictor of histological healing in ulcerative colitis. Ann Med 2025; 57:2499961. [PMID: 40305512 PMCID: PMC12044909 DOI: 10.1080/07853890.2025.2499961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory bowel disease with rising global prevalence.Histological healing (HH) is a key treatment target associated with better long-term outcomes. Although endoscopic ultrasound (EUS) is known to be related to both clinical and endoscopic activity in UC, its role in defining HH remains unclear. Therefore, this study was aimed at investigating the association between EUS and histological activity (HA), as well as the predictive potential of EUS for HH. METHOD In this cross-sectional analysis, 68 UC adults underwent EUS and colonoscopy with biopsies. We used the Mayo Endoscopic Score (MES) for endoscopic activity, the Nancy Index (NI) for biopsy grading, and the Endoscopic Ultrasound-Ulcerative Colitis (EUS-UC) score for EUS analysis, defining endoscopic remission as MES ≤ 1 and HH as NI ≤ 1.A receiver operating characteristic (ROC) curve was employed to evaluate the ability of the indices to predict HH. RESULTS Totally 23 patients (33.80%) achieved HH, while 45 (66.20%) showed HA. The EUS-UC scores were significantly lower in the HH group (p < 0.001) and correlated strongly with NI (ρ = 0.73). EUS-UC score was an independent risk factor for HH (adjusted OR = 1.918, 95% CI: 1.195-3.080, p = 0.007). The EUS-UC score demonstrated a strong predictive value for HH, with an AUC of 0.840, a sensitivity of 75.56%, and a specificity of 78.26%. CONCLUSION The EUS-UC score has a significant correlation with histological outcomes and shows strong potential as a reliable, invasive predictor of HH in UC, with implications for improved disease monitoring.
Collapse
Affiliation(s)
- Jin Tian
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Wei Wang
- Department of Gastroenterology, the First Affiliated Hospital of Shandong Second Medical University, Weifang People’s Hospital, Weifang, Shandong, China
| | - Yongshuai Liu
- Department of Gastroenterology, the First Affiliated Hospital of Shandong Second Medical University, Weifang People’s Hospital, Weifang, Shandong, China
| | - Xin Zhang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Hanqing Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
| | - Hongmei Qu
- Department of Gastroenterology, the First Affiliated Hospital of Shandong Second Medical University, Weifang People’s Hospital, Weifang, Shandong, China
| |
Collapse
|
2
|
Sano M, Kanatani Y, Ueda T, Nemoto S, Miyake Y, Tomita N, Suzuki H. Explainable artificial intelligence for prediction of refractory ulcerative colitis: analysis of a Japanese Nationwide Registry. Ann Med 2025; 57:2499960. [PMID: 40323686 PMCID: PMC12054586 DOI: 10.1080/07853890.2025.2499960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 05/07/2025] Open
Abstract
OBJECTIVE Ulcerative colitis (UC) is a chronic inflammatory bowel disease for which remission is dependent on corticosteroid (CS) treatment. The diversity of disease pathophysiology necessitates optimal case-specific treatment selection. This study aimed to identify prognostic factors for refractory UC using a machine learning model based on nationwide registry data. METHODS The study included 4003 patients with UC with a Mayo score of ≥3 at the time of registration who had been using CS since their entry out of 79,096 newly registered UC cases in a nationwide registry from April 2003 to March 2012 (before the widespread use of biologic agents in Japan) with 3-year data. A pointwise linear (PWL) model was used for machine learning. RESULTS A PWL model, which was developed to predict long-term remission (lasting >3 years), had an area-under-the-curve (AUC), precision rate, recall rate, and F-value of 0.774, 0.55, 0.70, 0.62, respectively, in the test dataset from the time of registration to 2 years later. Furthermore, the presence of pseudopolyps at the time of registration was significantly and negatively correlated with remission, highlighting its importance as a prognostic factor. CONCLUSIONS In this study, we constructed a highly accurate prognosis prediction model for UC, in which inflammation persists for an extensive period, by training a machine learning model for long-term disease progression. The results showed that machine learning can be used to determine the factors affecting remission during the treatment of refractory UC.
Collapse
Affiliation(s)
- Masaya Sano
- Department of Gastroenterology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Yasuhiro Kanatani
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Takashi Ueda
- Department of Gastroenterology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Shota Nemoto
- Industrial & Digital Business Unit, Hitachi Ltd, Chiyoda-ku, Tokyo, Japan
| | - Yurin Miyake
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Naoko Tomita
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Hidekazu Suzuki
- Department of Gastroenterology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| |
Collapse
|
3
|
Khokhar RK, Nashwan AJ. Gut virome and its emerging role in inflammatory bowel disease. World J Methodol 2025; 15:100534. [DOI: 10.5662/wjm.v15.i3.100534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/20/2025] [Accepted: 02/06/2025] [Indexed: 03/06/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a progressive multifactorial inflammatory disease of the gut. The cause of IBD is yet unknown. Some researchers have shown that genetic factors, environmental factors, and the gut microbiome are significant considerations. Our gut contains gut virome and gut bacteria, which vary among individuals due to some factors. The gut virome is a substantial component of the microbiome. This editorial explores the emerging role of gut virome in IBD.
Collapse
Affiliation(s)
- Rahat Khatoon Khokhar
- Department of Medicine, People’s University of Medical and Health Sciences, Nawabshah 67450, Pakistan
| | - Abdulqadir J Nashwan
- Department of Nursing & Midwifery Research, Hamad Medical Corporation, Doha 3050, Qatar
| |
Collapse
|
4
|
Min S, Fang Y, Zhang M, Shen H, Zhu L. The potential mechanism of co-administration of Scutellaria baicalensis Georgi and Rubia cordifolia L ameliorating ulcerative colitis: Integration of metabolomics, network pharmacology, and molecular docking. J Pharm Biomed Anal 2025; 263:116948. [PMID: 40344969 DOI: 10.1016/j.jpba.2025.116948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/27/2025] [Accepted: 05/03/2025] [Indexed: 05/11/2025]
Abstract
Scutellaria baicalensis Georgi (HQ) and Rubia cordifolia L. (QC) are clinically effective against ulcerative colitis (UC), but their synergistic mechanisms remain unclear. This study evaluated the therapeutic effects of HQ, QC, and their combination (HQ-QC) in dextran sulfate sodium (DSS)-induced colitis and explored underlying mechanisms. Mice were divided into normal, model, HQ, QC, HQ-QC, and 5-ASA groups. Colon tissue metabolomics was performed using UPLC-Q-TOF-MS, while network pharmacology and molecular docking identified potential anti-UC targets. The HQ-QC combination significantly alleviated weight loss, colon shortening, and histopathological damage, and improved intestinal barrier function and inflammation compared to single herbs. Metabolomics revealed xanthine, cytosine, and N-octanoylsphingosine-1-phosphate as key metabolites enriched in vascular smooth muscle contraction and fatty acid metabolism pathways. Network pharmacology identified 62 potential targets, with wogonin and xyloidone as major active compounds. KEGG analysis highlighted platelet activation as a key pathway, and molecular docking confirmed strong binding of wogonin and xyloidone to platelet activation-related targets including PIK3CG, NOS3, PTGS1, and MAPK14. These findings suggest HQ-QC exerts synergistic effects in treating DSS-induced UC by regulating vascular smooth muscle contraction and platelet activation, providing mechanistic insight into its clinical potential.
Collapse
Affiliation(s)
- Shichen Min
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yulai Fang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Mengyuan Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Hong Shen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| | - Lei Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.
| |
Collapse
|
5
|
Zou J, Ding W, Chen Y, Pan X, Fu X, Zheng S, Lin Y, Xia C, Ma Y, Zhang X, Wu S, Gao F. Incorporating zinc coordination driven nanozyme into chitosan and hyaluronic acid based nanoplatform for scavenging H 2S/ROS in managing inflammatory bowel disease. Carbohydr Polym 2025; 361:123501. [PMID: 40368544 DOI: 10.1016/j.carbpol.2025.123501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/25/2025] [Accepted: 03/12/2025] [Indexed: 05/16/2025]
Abstract
Inflammatory bowel disease (IBD) causes damage to patients, and therapy remains a crucial challenge owing to the presence of excessive reactive oxygen species (ROS) and hydrogen sulfide (H2S) in colonic inflammatory circulation. Herein, we report a shikonin (SK) and zinc (Zn) driven coordination nanozyme (SK-Zn, S-Z), which effectively scavenged ROS and H2S. Furthermore, a pH responsive targeted polymer nanoplatform (S-Z@ChF), consisting of S-Z, chitosan, hyaluronic acid and poly ferulic acid, was developed for the oral treatment of colitis. In vivo experiments revealed that the nanoplatform could target colonic inflammatory lesions, relieve inflammatory index and restore the colonic mechanical barrier. The transcriptomics and pharmacodynamic mechanism analysis revealed that S-Z@ChF scavenged ROS, inhibited nuclear factor kappa-B (NF-κB) and pyruvate kinase isozyme type M2 (PKM2) and signal transducer and activator of transcription 3 (STAT3) pathways, balancing the level of macrophage polarization and pro-inflammatory enzyme. Additionally, S-Z@ChF reliably regulated the gut microbiota during H2S scavenging. In conclusion, S-Z@ChF, which provides multiple anti-inflammatory pathways and microenvironment reprogramming support for blocking inflammatory circulation, was found to be biologically safe, with significant potential for clinical application in IBD.
Collapse
Affiliation(s)
- Jiafeng Zou
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Wenxing Ding
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - You Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xier Pan
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiuzhi Fu
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Shulei Zheng
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yiting Lin
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Chuanhe Xia
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Ying Ma
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaochun Zhang
- Clinical Traditional Chinese Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Shuang Wu
- Clinical Traditional Chinese Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China.
| | - Feng Gao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Pharmaceutical Engineering and Process of Chemical Engineering Research Center of Ministry of Education, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
6
|
Song T, Zhuang M, Wong ZYD, Xu G, Tang ML, Kou B, Sun X. Discovery of a deuterated TNF-α small molecule modulator for potential treatment of ulcerative colitis. Eur J Med Chem 2025; 291:117616. [PMID: 40239485 DOI: 10.1016/j.ejmech.2025.117616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Tumor necrosis factor-alpha (TNF-α) is an important target for the treatment of inflammatory diseases. Targeting TNF-α inhibition, such as antibody drug infliximab and adalimumab, has emerged as an effective therapeutic strategy for managing the most difficult-to-treat chronic ulcerative colitis (UC). So far, there are no small molecule TNF-α inhibitors available on the market for the treatment of UC. Previously, we reported an indanone analogue (R)-STU104 showed considerable inhibitory activity on TNF-α production in both acute and chronic mouse models of UC with a favorable safety profile. However, further development potential of this compound was greatly limited due to its poor metabolic stability in human liver microsomes and suboptimal pharmacokinetic profiles in mice. Herein, we discovered a deuterated TNF-α small molecule modulator (R)-104-6D-01, which demonstrated promising clinical potential for the treatment of UC. This new compound exhibited enhanced oral bioavailability and exposure in pharmacokinetic studies, as well as superior anti-UC efficacy in a DSS-induced mouse UC model, compared with (R)-STU104 at a dosage of 30 mg/kg/d. Collectively, (R)-104-6D-01 proves to be a promising candidate of potential use in treating UC as an oral TNF-α small molecule modulator.
Collapse
Affiliation(s)
- Tong Song
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Mengxiao Zhuang
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | | | - Ge Xu
- Jiangsu Angeltech Pharmaceuticals Co., Ltd, 4F, 19 South Taiping Road, Chengxiang Town, Taicang City, Suzhou, Jiangsu Province, 215400, China
| | - Mei-Lin Tang
- Shanghai Pudong Hospital, Pharmacophenomics Laboratory, Human Phenome Institute, Fudan University, Shanghai, 201203, China
| | - Buyu Kou
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China
| | - Xun Sun
- School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai, 201203, China; The Institutes of Integrative Medicine of Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China.
| |
Collapse
|
7
|
Su G, Chai L, Chen Y, Xia H, Li Y, Zhang Z, Liu G, Wang S, Yang C, Jin J, Wu L, Li Y. Anti-inflammatory withanolides from Physalis minima and their therapeutic potential against ulcerative colitis in mice. PHYTOCHEMISTRY 2025; 235:114451. [PMID: 39986410 DOI: 10.1016/j.phytochem.2025.114451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Eleven undescribed withanolides, phyminimolides I-XI (1-11), along with eight known derivatives (12-19), were isolated from the whole plant of Physalis minima Linn. Their structures were elucidated by extensive spectroscopic methods, including 1D-NMR, 2D-NMR, HRESIMS, IR, experimental electronic circular dichroism, quantum chemical calculation, and X-ray crystallography. The anti-inflammatory potential of these compounds was assessed in vitro via their capacity to inhibit nuclear factor kappa-B signaling in lipopolysaccharide-stimulated 293T/NF-κB-luciferase reporter cells, revealing IC50 values of 9.95, 3.15, 1.16, 9.73, and 11.74 μM for compounds 4-6, 13, and 15, respectively. Notably, compounds 4 and 13 also demonstrated inhibitory effects on interleukin-6 pathway activation, with IC50 values of 1.61 and 7.56 μM, further evidencing their anti-inflammatory mechanism. In vivo assessment using dextran sulfate sodium-induced mice model of ulcerative colitis confirmed the translational efficacy of these findings. Compound 15, administered at a dose of 25 mg/kg, significantly ameliorated disease symptoms, as evidenced by a decreased Disease Activity Index score (P < 0.05).
Collapse
Affiliation(s)
- Guozhu Su
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Lisha Chai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Yu Chen
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Huimin Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Ying Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Zhen Zhang
- Pharmacy Department, Linyi People's Hospital, Shandong, Linyi, 276003, People's Republic of China
| | - Guosheng Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Shangyi Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Chengshuo Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Jing Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Lianqiu Wu
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China.
| | - Yong Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, People's Republic of China.
| |
Collapse
|
8
|
Long S, Wang L, Zeng Q, Li Y, Su J, Chen Y, Zhou G. Exochorda racemosa attenuates DSS-induced colitis in C57BL/6 J mice by regulating inflammatory factors, reducing oxidative stress, and modulating intestinal flora. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156768. [PMID: 40250030 DOI: 10.1016/j.phymed.2025.156768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/27/2025] [Accepted: 04/13/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Exochorda racemosa is a member of the genus Exochorda in the Rosaceae family. Its tender leaves and buds are favored as a unique wild vegetable by people in central China. PURPOSE This study systematically evaluated the pharmacological safety and anti-inflammatory efficacy of E. racemosa extracts, with concurrent identification and characterization of their primary bioactive components. METHODS The chemical composition of E. racemosa extract (ERE) was analyzed using HPLC and LC-MS techniques. The safety and in vivo anti-inflammatory effects of ERE were evaluated using the maximum tolerated dose (MTD) in ICR mice and a dextran sodium sulfate-induced ulcerative colitis model in C57BL/6 J mice. RESULTS HPLC and LC-MS analyses revealed that ERE contained abundant flavonoid active ingredients. MTD study confirmed that ERE exhibited good safety. The symptoms of persistent weight loss, DAI, and shortened colon length in UC mice were suppressed by ERE. Pathological damage in colon tissues was attenuated by ERE, with a considerable reduction in histopathological scores and a substantial increase in the number of goblet cells. The levels of IL-6, IL-1β, and TNF-α in serum were significantly decreased following ERE treatment. Moreover, nitric oxide (NO) levels and myeloperoxidase (MPO) activity in colon tissues decreased, whereas glutathione (GSH) levels and superoxide dismutase (SOD) activity in colon tissues increased after ERE treatment. Furthermore, ERE could regulate the intestinal microbial composition and maintain intestinal flora homeostasis, thereby inhibiting inflammatory responses. CONCLUSION ERE exhibited a favorable safety profile and alleviated UC through multiple mechanisms. It is expected to serve as a promising low-toxicity natural product for adjuvant treatment in UC.
Collapse
Affiliation(s)
- Sha Long
- Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, PR China
| | - Lu Wang
- Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, PR China
| | - Qi Zeng
- Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, PR China
| | - Yaoyao Li
- Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, PR China
| | - Jiangtao Su
- Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, PR China
| | - Yuxin Chen
- Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, PR China
| | - Gao Zhou
- Hubei Key Laboratory of Industrial Microbiology, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Life and Health Sciences, Hubei University of Technology, Wuhan 430068, PR China; Postdoctoral Research Center of Mayinglong Pharmaceutical Group Co., Ltd., Wuhan, Hubei 430064, PR China; Jiangxi Province Key Laboratory of Sustainable Utilization of Traditional Chinese Medicine Resources, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang 330115, PR China.
| |
Collapse
|
9
|
Zheng BX, Yi Y, Wang XW, Li CY, Zhao Y, Tian JZ, Wang LM, Han JY, Pan C, Liu SY, Liu CY, Qin SS, Tang X, Liu MT, Liang AH. Geniposide via enema alleviates colitis by modulating intestinal flora and bile acid metabolites, inhibiting S100A8/S100A9/NF-κB, and promoting TGR5 inhibition of NLRP3 inflammasome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156791. [PMID: 40279965 DOI: 10.1016/j.phymed.2025.156791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/12/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Geniposide (GE) has potential efficacy in treating ulcerative colitis (UC). However, its reactivity can be affected by rapid degradation after oral administration. Furthermore, increasing oral doses may lead to hepatotoxicity. Thus, We used enema administration, characterized by smaller dose and higher localized concentration in the lesion, to improve the above situation. PURPOSE We aimed to confirm that enema administration is a better modality than oral administration for GE against UC and to explore its mechanism. STUDY DESIGN/METHOD We established UC mouse model, monitoring Disease Activity Index (DAI), inflammatory cytokines levels, and histopathology. Macrogenomics and bile acid (BAs) metabolomics analysed the major intestinal flora and BAs. Simultaneouslly, we conducted quantitative proteomics analysis and screened core proteins and pathway. In vitro validation was taken by qPCR, immunofluorescence and immunoblotting experiments. RESULTS GE via enema alleviate UC by inhibiting inflammatory factor production through downregulating S100A8/S100A9/NF-κB pathway. Analysis of the intestinal flora and BAs revealed that the enhanced abundance of Lachnospiraceae, which improves the ratio of primary to secondary BAs, and the reduced abundance of Provocaceae, which increases intestinal permeability and promotes inflammation, favored the restoration of the intestinal barrier. In addition, in vitro experiments confirmed that the key BA metabolites (mainly UDCA, DCA, and LCA) stimulated TGR5 signal to inhibit the assembly of the NLRP3 inflammasome and alleviated inflammation. CONCLUSION We firstly confirmed that GE alleviates UC via the enema route in a better manner than the oral route, through enhancing the intestinal barrier, restoring intestinal flora and BAs homeostasis, and inhibiting inflammatory injury. This study initially revealed that GE can alleviate UC through elevating UDCA, DCA, and LCA levels at the colonic site to activate TGR5 receptor for inhibiting the NLRP3 inflammasome, in addition to downregulating the S100A8/S100A9/-TLR4-NF-κB pathway related inflammatory response directly. The evidences offer a promising strategy and profround meaning for UC treatment.
Collapse
Affiliation(s)
- Bao-Xin Zheng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yan Yi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Xing-Wen Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Chun-Ying Li
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Yong Zhao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Jing-Zhuo Tian
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Lian-Mei Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Jia-Yin Han
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Chen Pan
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Su-Yan Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Chen-Yue Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Sha-Sha Qin
- Chongqing University Of Chinese Medicine, Chongging 400060, China.
| | - Xuan Tang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Mei-Ting Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Ai-Hua Liang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
10
|
Qi F, Shen Z, Zhou S, Zhang Y, Zhang Y, Wang H, Du Y, Xie Z, Li D, Ge H. Tea residue protein-derived oligopeptides attenuate DSS-induced acute colitis complicated with hepatic injury in C57BL/6J mice by regulating the gut-microbiome-liver axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156792. [PMID: 40311592 DOI: 10.1016/j.phymed.2025.156792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/10/2025] [Accepted: 04/19/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Impairment of the intestinal mucosal barrier is a prevalent feature of acute colitis, and untreated acute colitis can lead to extra-intestinal manifestations, including hepatic injury. Previous research has demonstrated that large-leaf yellow tea residue protein-derived oligopeptides (TPP) can alleviate ulcerative colitis symptoms and hepatic injury in mice. However, the underlying regulatory mechanisms by which TPP improves colitis complicated with liver injury are unknown. PURPOSE To explore the potential mechanism by which TPP alleviates acute colitis complicated with hepatic injury. METHODS Acute colitis with hepatic injury was induced in mice using 3.5 % dextran sodium sulfate. Both 16S rRNA sequencing and transcriptomic analyses were utilized to investigate the impact of TPP on mitigating symptoms in mice. RESULTS It indicated that TPP administration effectively reduced inflammatory symptoms in the colon and liver, enhanced the secretion of mucin occluding, claudin-1, ZO-1, and MUC-2, decreased intestinal mucosal permeability, and restored homeostasis within the gut microbiome of mice. Moreover, transcriptomic analysis has evidenced the effectiveness of TPP in mitigating liver-related effects. RNA-seq KEGG enrichment and RT-qPCR analyses validated TPP could modulate the "gut-microbiome-liver" axis, and participate in signaling pathways related to inflammatory regulation, as well as bile acid metabolism and synthesis. CONCLUSION These findings suggest that TPP administration is a promising novel approach for preventing and treating acute colitis complicated with hepatic injury.
Collapse
Affiliation(s)
- Fengxue Qi
- School of Food and Nutrition, Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei Anhui 230036, PR China
| | - Ziyi Shen
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Anhui Agricultural University, Hefei Anhui 230036, PR China
| | - Simeng Zhou
- School of Food and Nutrition, Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei Anhui 230036, PR China
| | - Yuan Zhang
- School of Food and Nutrition, Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei Anhui 230036, PR China
| | - Yaru Zhang
- School of Food and Nutrition, Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei Anhui 230036, PR China
| | - Hongyan Wang
- School of Food and Nutrition, Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei Anhui 230036, PR China
| | - Yiqun Du
- School of Food and Nutrition, Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei Anhui 230036, PR China
| | - Zhongwen Xie
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Anhui Agricultural University, Hefei Anhui 230036, PR China
| | - Daxiang Li
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Anhui Agricultural University, Hefei Anhui 230036, PR China.
| | - Huifang Ge
- School of Food and Nutrition, Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei Anhui 230036, PR China; National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, Anhui Agricultural University, Hefei Anhui 230036, PR China.
| |
Collapse
|
11
|
Huang H, Yang C, Li S, Zhan H, Tan J, Chen C, Liu J, Wang M, Li H. Lizhong decoction alleviates experimental ulcerative colitis via regulating gut microbiota-SCFAs-Th17/Treg axis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119958. [PMID: 40350047 DOI: 10.1016/j.jep.2025.119958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/26/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lizhong decoction (LZD), a Traditional Chinese Medicine formula, is widely utilized to treat gastrointestinal diseases, including ulcerative colitis in China for thousands of years. AIM OF THE STUDY To investigate whether the protective effect of LZD on ulcerative colitis is dependent on gut microbiota and T-cell immune homeostasis. MATERIAL AND METHODS The preventive effects of LZD on dextran sodium sulfate (DSS)-induced colitis mice were evaluated through the measurement of body weight, disease activity index, colon length and hematoxylin-eosin staining. Flow cytometry was used to detect the ratio of Th17/Treg cells. Pseudo sterile mice and fecal transplantation experiments were used to investigate whether the preventive effect of LZD was dependent on the gut microbiota. The alterations of gut microbiota were identified by the 16S rDNA sequencing. The content of intestinal short-chain fatty acids (SCFAs) was detected by LC-MS/MS analysis. The downstream signal pathways of SCFAs were detected by the immunoblotting. RESULTS LZD administration significantly alleviated weight loss and intestinal injury in DSS-induced colitis mice. LZD administration also promotes the balance of Th17/Treg cells. Moreover, LZD administration relies on gut microbiota to alleviate ulcerative colitis and regulate Th17/Treg cell balance. LZD administration significantly improves gut microbial composition in colitis mice, elevating the abundance of SCFAs producing bacterium such as lachnospiraceae_nk4a136_group and Akkermansia. LZD treatment further increases the abundance of SCFAs and promotes activation of free fatty acid activated receptor 2 (FFAR2). CONCLUSION LZD administration promotes Th17/Treg cell balance in a gut microbiota-SCFAs dependent manner, which in turn ameliorates ulcerative colitis.
Collapse
MESH Headings
- Animals
- Gastrointestinal Microbiome/drug effects
- Colitis, Ulcerative/drug therapy
- Colitis, Ulcerative/immunology
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/microbiology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Dextran Sulfate
- Male
- Mice
- Mice, Inbred C57BL
- Fatty Acids, Volatile/metabolism
- Disease Models, Animal
- Colon/drug effects
- Colon/pathology
- Colon/microbiology
- Fecal Microbiota Transplantation
Collapse
Affiliation(s)
- Hengjun Huang
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China.
| | - Chengyu Yang
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
| | - Silu Li
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
| | - Huang Zhan
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
| | - Jinlong Tan
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
| | - Congcong Chen
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
| | - Jian Liu
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
| | - Maolin Wang
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China
| | - Hui Li
- Jiangxi Province Key Laboratory of Traditional Chinese Medicine Pharmacology, Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, 330115, China; Jiangxi Health Industry Institute of Traditional Chinese Medicine, Nanchang, 330115, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
12
|
Luo L, Liu Q, Zhang Y, Yu X, Wang L, Sun W, Li T, Xu B, Zhang K, Yu Y, Cui C, Li C, Mei L. Precisely edited gut microbiota by tungsten-doped Prussian blue nanoparticles for the treatment of inflammatory bowel disease. J Control Release 2025; 382:113755. [PMID: 40258476 DOI: 10.1016/j.jconrel.2025.113755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025]
Abstract
Inflammatory bowel disease (IBD) is characterized by recurring gastrointestinal inflammation, accompanied by a significant rise in global prevalence and disease severity. The overaccumulation of reactive oxygen and nitrogen species (RONS) in the intestinal environment disrupts redox homeostasis and drives pathological overgrowth of Escherichia coli, which are central to IBD pathogenesis. Herein, we designed a multifunctional nanozyme (W-PB) to enable sustained and targeted regulation of intestinal homeostasis through dual mechanisms: specific inhibition of E. coli overgrowth during colitis and efficient RONS clearance. To ensure colon-specific delivery, W-PB was encapsulated in an electrostatically crosslinked hydrogel composed of alginate and chitosan. This formulation protects W-PB from degradation in harsh gastrointestinal conditions and releases the nanoparticles selectively under weakly alkaline intestinal pH. The released tungsten ions suppress E. coli growth via competitive displacement of molybdenum in the molybdopterin cofactor, while W-PB simultaneously neutralizes excess RONS to shield intestinal cells from oxidative damage. In DSS-induced colitis models, the W-PB gel demonstrated significant therapeutic efficacy, achieved through intestinal microbiota remodeling and oxidative stress mitigation.
Collapse
Affiliation(s)
- Lingpeng Luo
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Qingyun Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Yushi Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Xuya Yu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Ling Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, PR China
| | - Weiting Sun
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Tingxuan Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Bin Xu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Kai Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Yongkang Yu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore.
| | - Chunhui Cui
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.
| | - Chen Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China.
| | - Lin Mei
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China; Furong Laboratory, Central South University, Changsha 410008, PR China.
| |
Collapse
|
13
|
Wu B, Yan W, Lu Y, Xiao Y. Diagnostic values of CD27, CD20 and MPO in pediatric ulcerative colitis. Gene 2025; 952:149415. [PMID: 40089083 DOI: 10.1016/j.gene.2025.149415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Inflammatory bowel disease (IBD), including ulcerative colitis (UC), is a chronic inflammatory disorder with a rising incidence in pediatric populations. Immune factors play important roles in the pathogenesis of UC. This study aimed to explore the relationships of intestinal immune molecules CD27, CD20 and myeloperoxidase (MPO) with pediatric UC and their diagnostic values. In this study, gene expression data of 206 new-onset UC children and 20 non-IBD controls obtained from the NCBI Gene Expression Omnibus public database and immunohistochemistry analysis were used to evaluate CD27, CD20 and MPO expression in diseased intestinal tissues of UC children. And the diagnostic potentials of them for UC were analyzed using receiver operating characteristic curve and area under the curve (AUC). We found that CD27, CD20 and MPO mRNA and protein expressions were increased in the diseased intestinal tissues of UC children. CD27, CD20 and MPO showed good diagnostic potential for UC in children, with an AUC of 0.95 for CD27, 0.79 for CD20 and 0.92 for MPO, and combination of them had better diagnostic performance with an AUC of 0.98. Besides, they were associated with immune-related biological processes and pathways, and correlated with genes related to immune factors, intestinal epithelial barrier function, and intestinal fibrosis. In conclusion, our findings demonstrated that CD27, CD20 and MPO were increased in diseased intestinal tissues of UC children, and had good diagnostic performance for UC in children.
Collapse
Affiliation(s)
- Bo Wu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weihui Yan
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Ying Lu
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; Shanghai Institute of Pediatric Research, Shanghai, China
| | - Yongtao Xiao
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China; Shanghai Institute of Pediatric Research, Shanghai, China.
| |
Collapse
|
14
|
Li L, Lam I, Wang J, Yu H, Chan C, Cai S. Epigenetic mechanism of iPSC-MSC-EVs in colonic epithelial cell pyroptosis in ulcerative colitis cell models via modulation of ELF3/miR-342-3p/KDM6B axis and histone methylation. Int Immunopharmacol 2025; 157:114704. [PMID: 40315630 DOI: 10.1016/j.intimp.2025.114704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/09/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease in the colon and rectum. Mesenchymal stem cell-derived extracellular vesicles (MSCs-EVs) have emerged as promising cell-free therapeutics for UC, leveraging their immunomodulatory and tissue-protective properties. However, the specific epigenetic mechanisms by which EVs regulate pyroptosis (an inflammatory cell death pathway) remain poorly understood. This study explores how EVs derived from induced pluripotent stem cell-derived mesenchymal stem cells (iPSC-MSCs) regulate pyroptosis in colonic epithelial cells of UC by targeting the histone-modifying protein KDM6B, aiming to provide new therapeutic insights for UC. iPSCs were differentiated into MSCs, and their EVs were isolated and characterized. EVs were engineered to carry the circular RNA circ-CCND1 and co-cultured with UC model cells induced by DSS. Cell viability, inflammatory cytokine levels, and key molecular markers related to pyroptosis (NLRP3, cleaved Caspase-1, GSDMD-N) were measured. The molecular mechanism was dissected using RNA-protein binding assays and gene expression analysis, focusing on the circ-CCND1/KDM6B/ELF3/miR-342-3p signaling axis. EV treatment reduced pyroptosis in UC model cells, with enhanced efficacy when EVs carried circ-CCND1. Mechanistically, circ-CCND1 in EVs entered cells and bound to KDM6B, inhibiting its activation of the ELF3 gene, leading to increased miR-342-3p, which in turn suppressed KDM6B expression, forming a feedback loop that dampened pyroptosis. In conclusions, iPSC-MSC-derived EVs inhibit inflammatory cell death in colonic epithelial cells by regulating histone modification-related pathways, highlighting their potential as a novel therapeutic strategy for UC.
Collapse
Affiliation(s)
- Lixuan Li
- Department of Gastroenterology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510000, China.
| | - Ienghou Lam
- Department of Gastroenterology, Kiang Wu Hospital, Macau SAR 999078, China
| | - Jintao Wang
- Department of Gastroenterology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510000, China
| | - Honho Yu
- Department of Gastroenterology, Kiang Wu Hospital, Macau SAR 999078, China
| | - Chonin Chan
- Department of Gastroenterology, Kiang Wu Hospital, Macau SAR 999078, China
| | - Shaowei Cai
- Department of Gastroenterology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510000, China
| |
Collapse
|
15
|
Noor NM, Parkes M, Raine T. U-ACTIVATE long-term efficacy and safety outcomes for upadacitinib in ulcerative colitis. Lancet Gastroenterol Hepatol 2025; 10:492-493. [PMID: 40347952 DOI: 10.1016/s2468-1253(25)00046-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 05/14/2025]
Affiliation(s)
- Nurulamin M Noor
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK.
| | - Miles Parkes
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SP, UK
| | - Tim Raine
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
16
|
Mandal A, Banerjee S, Ghosh S, Biswas S, Bagchi A, Sil PC. α-ketoglutarate ameliorates colitis through modulation of inflammation, ER stress, and apoptosis. Toxicol Rep 2025; 14:101897. [PMID: 39886045 PMCID: PMC11780160 DOI: 10.1016/j.toxrep.2025.101897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 02/01/2025] Open
Abstract
Colitis is an inflammatory disorder of the gastrointestinal tract. A widely consumed dietary nutrient, α-ketoglutarate (α-KG) is known to play a crucial role in cellular metabolism and provide protection to intestinal epithelium under various pathophysiological conditions. In this study, 2,4,6-trinitrobenzenesulfonic acid (TNBS) was used to induce colitis in Wistar rats. After 36 hours of TNBS administration, the rats were orally treated with a solution of α-KG at 1 g/kg body weight for 5 days. Development of colitis was confirmed by observable physical symptoms of repeated loose blood-mixed stool, apathy for food and weight loss. Macroscopic inspection revealed an inflamed colonic surface with ulcerations. Histopathological observations included alterations in crypts-structure and disruption in both epithelial and mucosal layers of colon in colitis induced rats. Colitis resulted in elevated levels of pro-inflammatory cytokines, ER stress-mediated cell death and intrinsic apoptosis pathway. The ameliorative effects of α-KG against TNBS-mediated toxicity were confirmed through molecular technics and docking analysis. Additionally, there were no instances of toxicity of α-KG. Therefore, α-KG can be considered as a valuable therapeutic agent for further comprehensive research.
Collapse
Affiliation(s)
- Ankita Mandal
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| | - Sharmistha Banerjee
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| | - Sumit Ghosh
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| | - Sima Biswas
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, Kalyani, West Bengal 741235, India
| | - Angshuman Bagchi
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, Kalyani, West Bengal 741235, India
| | - Parames C. Sil
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata, West Bengal 700054, India
| |
Collapse
|
17
|
Panaccione R, Vermeire S, Danese S, Higgins PDR, Lichtenstein GR, Nakase H, Glover S, Colombel JF, Eccleston J, Kujawski M, Remple V, Yao X, Geng Z, Palac H, Sharma D, Suravaram S, Schreiber S. Long-term efficacy and safety of upadacitinib in patients with moderately to severely active ulcerative colitis: an interim analysis of the phase 3 U-ACTIVATE long-term extension study. Lancet Gastroenterol Hepatol 2025; 10:507-519. [PMID: 40347957 DOI: 10.1016/s2468-1253(25)00017-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND The U-ACTIVATE long-term extension study aims to evaluate the long-term efficacy and safety of upadacitinib in patients with moderately to severely active ulcerative colitis. Here, we report interim results after 3 years of total treatment. METHODS U-ACTIVATE is an ongoing, 288-week, phase 3, long-term extension study done at 307 centres across 43 countries (active sites on Dec 31, 2021, are presented as part of this interim analysis) and began on Jan 31, 2017. In brief, patients aged 16-75 years with a confirmed diagnosis of moderately to severely active ulcerative colitis for 90 days or more, an adapted Mayo score of 5-9, and an endoscopic subscore of 2 or 3 were eligible for the upadacitinib induction studies; patients who had a clinical response in the induction studies were eligible to enter the U-ACHIEVE maintenance study. Individuals who completed the U-ACHIEVE maintenance study were subsequently eligible for inclusion in the efficacy population of this long-term extension study. Patients in clinical remission per adapted Mayo score at week 52 of the maintenance study could continue their double-masked treatment upon entering the long-term extension study. Patients not in clinical remission originally randomly assigned to upadacitinib 15 mg were eligible to escalate to upadacitinib 30 mg, those originally randomly assigned to upadacitinib 30 mg continued on upadacitinib 30 mg, and those originally assigned to placebo were eligible to escalate to upadacitinib 15 mg in a masked way. We present data from weeks 48 and 96 of the long-term extension period. Key efficacy outcomes were clinical remission (per adapted Mayo score), endoscopic remission, maintenance of clinical remission, and maintenance of endoscopic remission, and are presented for those patients who had a clinical response after 8 weeks of upadacitinib 45 mg induction, completed 52 weeks of maintenance (U-ACHIEVE maintenance), and subsequently entered the long-term extension. Safety outcomes were treatment-emergent adverse events and adverse events of special interest, which were prespecified and were recorded in two populations: one comprising patients who received at least one dose of study drug in the long-term extension study and the other comprising all patients in the maintenance or long-term extension studies. Our primary approach for efficacy analysis was as-observed (ie, all observed data were used without imputation for missing data until patients switched to a different dose during the long-term extension study). This study is registered with ClinicalTrials.gov (NCT03006068). FINDINGS 414 patients from the phase 3 upadacitinib U-ACHIEVE maintenance study were eligible to enter this long-term extension study for assessment of efficacy endpoints following treatment with upadacitinib. Of these individuals, 369 patients (231 [63%] male individuals and 138 [37%] female individuals) were treated with upadacitinib in the long-term extension study: 142 patients with upadacitinib 15 mg and 227 with upadacitinib 30 mg. In the as-observed population, 84 (71%) of 118 patients receiving upadacitinib 15 mg were in clinical remission at week 48, as were 130 (67%) of 193 receiving upadacitinib 30 mg; by week 96, 69 (76%) of 91 patients receiving upadacitinib 15 mg and 104 (74%) of 141 of those receiving upadacitinib 30 mg were in clinical remission. Most patients who entered the long-term extension in clinical remission maintained it in the as-observed analysis (week 48 upadacitinib 15 mg 62 [81%] of 77 and upadacitinib 30 mg 90 [81%] of 111; week 96 upadacitinib 15 mg 50 [78%] of 64 and upadacitinib 30 mg 69 [84%] of 82). In the as-observed population, 60 (49%) of 123 patients receiving upadacitinib 15 mg and 93 (46%) of 202 receiving upadacitinib 30 mg were in endoscopic remission at week 48; by week 96, 45 (47%) of 95 patients receiving upadacitinib 15 mg and 69 (45%) of 153 receiving upadacitinib 30 mg were in endoscopic remission. Most patients who entered the long-term extension in endoscopic remission maintained it in the as-observed analysis (week 48 upadacitinib 15 mg 28 [70%] of 40 and upadacitinib 30 mg 51 [76%] of 67; week 96 upadacitinib 15 mg 20 [65%] of 31 and upadacitinib 30 mg 37 [73%] of 51). In the long-term extension-only safety analysis, we assessed data from 467 patients, representing 1027·9 patient-years of continuous long-term extension exposure on a consistent upadacitinib dose. Treatment-emergent adverse events were recorded at 238·5 events per 100 patient-years for upadacitinib 15 mg and 233·4 events per 100 patient-years for upadacitinib 30 mg. Event rates of serious treatment-emergent adverse events were 11·7 events per 100 patient-years for upadacitinib 15 mg and 12·4 events per 100 patient-years for upadacitinib 30 mg. The most common adverse events of special interest were hepatic disorder, lymphopenia, creatine phosphokinase elevation, serious infection, neutropenia, and herpes zoster. Three treatment-emergent adverse events leading to death were reported in the long-term extension-only safety population. INTERPRETATION This interim analysis supports the positive long-term risk-benefit profile for upadacitinib 15 mg and 30 mg among patients with moderately to severely active ulcerative colitis. FUNDING AbbVie.
Collapse
Affiliation(s)
- Remo Panaccione
- Division of Gastroenterology and Hepatology, University of Calgary, Calgary, AB, Canada
| | - Séverine Vermeire
- Department of Gastroenterology and Hepatology, University Hospital Leuven, Leuven, Belgium
| | - Silvio Danese
- Gastroenterology and Endoscopy, Istituto di Ricovero e Cura a Carattere Scientifico Ospedale San Raffaele and University Vita-Salute San Raffaele, Milan, Italy
| | - Peter D R Higgins
- Department of Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI, USA
| | | | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Sarah Glover
- Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Jean-Frédéric Colombel
- Henry Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | | | | | | | | | | | | | | | | | - Stefan Schreiber
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, Christian Albrecht University of Kiel, Kiel, Germany.
| |
Collapse
|
18
|
Acherman YIZ, Arebi N, Arthurs E, Bemelman WA, van der Bilt JDW, Braat H, Brink MA, Brookes M, Brown JPY, Brown SR, Buskens CJ, Consten ECJ, Cooney R, Crolla RMPH, Davies RJ, Depla ACTM, D'Haens GR, Dijkgraaf MGW, Doherty G, van Duijvendijk P, Duijvestein M, Eshuis EJ, Evans JP, Faiz OD, Fong SCM, Gerhards MF, Grainger J, Grimes CE, Handley K, Heuthorst L, Hompes R, Iqbal TH, Jansen J, Kaur M, Magill L, Mallant-Hent RC, Mannaerts GHH, Moran G, Nicholson GA, Pathmakanthan S, Pierik EGJM, Pinkney TD, Ponsioen CY, Raine T, Reilly I, Sahami S, Seenan JP, Seerden TCJ, Shabbir J, Shaw SM, Singh B, Stellingwerf ME, Stokkers PCF, Visser E, Vlug MS, Vrouenraets BC, West R, Wiggers JK, Wildenberg ME, Winter D, Yassin NA, van der Zanden EPM. Appendicectomy plus standard medical therapy versus standard medical therapy alone for maintenance of remission in ulcerative colitis (ACCURE): a pragmatic, open-label, international, randomised trial. Lancet Gastroenterol Hepatol 2025; 10:550-561. [PMID: 40228513 PMCID: PMC12062198 DOI: 10.1016/s2468-1253(25)00026-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND The appendix might have an immunomodulatory role in ulcerative colitis. Appendicectomy has been suggested as a potentially therapeutic intervention to maintain remission in ulcerative colitis. We aimed to evaluate the clinical effectiveness of laparoscopic appendicectomy in maintaining remission in patients with ulcerative colitis. METHODS We did a pragmatic, open-label, international, randomised controlled superiority trial in 22 centres across the Netherlands, Ireland, and the UK. Patients with established ulcerative colitis who were in remission but had been treated for disease relapse within the preceding 12 months were randomly assigned (1:1) to undergo appendicectomy plus continued maintenance medical therapy (intervention group) or to continue maintenance medical therapy alone (control group). Randomisation was done with a central, computer-generated allocation concealment, stratified by disease extent. Patients and treating physicians were unmasked to group allocation. The prespecified primary outcome was the proportion of patients with a disease relapse within 1 year, predefined as a total Mayo score of 5 or higher with an endoscopic subscore of 2 or 3, or, in absence of endoscopy, based on a centrally independent masked review by a critical event committee as an exacerbation of abdominal symptoms (eg, elevated stool frequency subscore of ≥1 point from baseline) with a rectal bleeding subscore of ≥1 or faecal calprotectin level above 150 μg/g or necessitating treatment intensification other than mesalazine. Analyses were done on an intention-to-treat principle. This trial is complete and was registered with the Netherlands Trial Register (NTR2883) and ISRCTN (ISRCTN60945764). FINDINGS Between Sept 20, 2012, and Sept 21, 2022, 1386 patients were screened. 201 patients were randomly assigned to the appendicectomy group (n=101) or the control group (n=100). After exclusion of four patients due to eligibility violations (three had active disease and one received biological agents at time of randomisation), 99 patients in the appendicectomy group and 98 patients in the control group were included in the intention-to-treat analyses. The 1-year relapse rate was significantly lower in the appendicectomy group than in the control group (36 [36%] of 99 patients vs 55 [56%] of 98 patients; relative risk 0·65 [95% CI 0·47-0·89]; p=0·005; adjusted p=0·002). Adverse events occurred in 11 (11%) of 96 patients in the appendicectomy group and 10 (10%) of 101 patients in the control group. The most frequently reported adverse events were postoperative temporary self-limiting abdominal pain in the appendicectomy group (three [3%] patients) and skin rash in the control group (three [3%] patients). Two cases (2%) of low-grade appendiceal mucinous neoplasm were incidentally found in resected appendix specimens in the appendicectomy group. Serious adverse events occurred in two (2%) of 96 patients who underwent appendicectomy and none in the control group. There were no deaths. INTERPRETATION Appendicectomy is superior to standard medical therapy alone in maintaining remission in patients with ulcerative colitis. FUNDING Fonds Nuts-Ohra and National Institute for Health Research Efficacy and Mechanism Evaluation.
Collapse
|
19
|
Ryszkiewicz P, Malinowska B, Schlicker E. Polypharmacology: new drugs in 2023-2024. Pharmacol Rep 2025; 77:543-560. [PMID: 40095348 PMCID: PMC12066383 DOI: 10.1007/s43440-025-00715-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Polypharmacology is an emerging approach to drug design and development that involves the use of multi-target-directed ligands (MTDLs), agents capable of interacting with multiple biological targets simultaneously. The effective treatment of chronic and multifactorial conditions, driven by the dysregulation of multiple interconnected pathways, such as cancer, autoimmune and metabolic disorders, cardiovascular and neurodegenerative diseases, is one of the most substantial challenges in contemporary pharmacology. 'Traditional' single-target-based treatment frequently shows limited effectiveness, as resistance to therapy develops or relapses occur. The rational use of MTDLs seems therefore a promising way to address the complexity of biological systems, feedback mechanisms, crosstalk, and molecular pathways. Many MTDLs have been successfully marketed to date. Moreover, plenty of them offer an additional benefit in comparison to 'traditional' treatment approaches. To assess whether the polypharmacological trend remains prevalent, we thoroughly analysed drugs approved in the years of 2023-2024 in Germany. Among 73 newly introduced substances, 18 are in line with the polypharmacology concept, including 10 antitumor agents, 5 drugs indicated for autoimmune disorders, 1 indicated for hand eczema, 1 antidiabetic (and anti-obesity) drug, and 1 modified corticosteroid.
Collapse
Affiliation(s)
- Piotr Ryszkiewicz
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Białystok, 15-222, Poland.
| | - Barbara Malinowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Białystok, Białystok, 15-222, Poland
| | - Eberhard Schlicker
- Department of Pharmacology and Toxicology, University of Bonn, Venusberg Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
20
|
Tian S, Hu Y, Zhou Y, Li J, Liu J, Zhou Q. Low-intensity pulsed ultrasound irradiation ameliorates gut inflammation and neuroinflammation in mice with DSS-induced colitis. APL Bioeng 2025; 9:026117. [PMID: 40385988 PMCID: PMC12085233 DOI: 10.1063/5.0263732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025] Open
Abstract
Ulcerative colitis (UC) is a lifelong autoimmune disease associated with a high prevalence of mental disorders. An abnormal gut-brain axis plays a key role in UC. Low-intensity pulsed ultrasound (LIPUS) has been reported to alleviate neuroinflammation. This study aimed to evaluate the therapeutic effects of LIPUS in an experimental colitis model. A mouse model of colitis induced by dextran sulfate sodium (DSS) was established to evaluate the therapeutic effects of LIPUS irradiation. Intestinal inflammation and the mucosal barrier were detected using reverse transcriptase polymerase chain reaction and immunofluorescence staining. The key proteins of the NLRP3 inflammatory pathway in different groups were measured by western blotting. 16S rRNA sequencing and ultra-performance liquid-chromatography tandem mass spectrometry were applied for the detection of bacteria, metabolites, and neurotransmitters. LIPUS irradiation significantly improved the mucosal barrier in DSS-induced colitis mice and relieved intestinal inflammation and neuroinflammation by downregulating the NLRP3 inflammatory pathway. 16S rRNA sequencing showed that LIPUS irradiation significantly upregulated the abundance of Bacteroides and glutamate metabolism. Metabolic analysis revealed that the most significant metabolites between the DSS + LIPUS and DSS groups were mostly involved in alanine, aspartate, and glutamate metabolism. The detection of neurotransmitters revealed that the levels of gamma-amino butyric acid (GABA) were significantly upregulated in DSS-induced colitis mice after LIPUS irradiation, and correlation analysis revealed a positive correlation between GABA and Bacteroides. LIPUS irradiation not only alleviated gut inflammation and neuroinflammation by regulating the NLRP3 inflammatory pathway in DSS-induced colitis mice but also increased GABA levels by upregulating Bacteroides, indicating that LIPUS might be a promising therapeutic technology for UC treatment.
Collapse
Affiliation(s)
| | - Yugang Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, People's Republic of China
| | - Yanxiang Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, People's Republic of China
| | - Jiao Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, People's Republic of China
| | - Jian Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, People's Republic of China
| | - Qing Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, People's Republic of China
| |
Collapse
|
21
|
Chen J, Yuan Q, Yu C, Fu J, Wang P, Tang S, Lin X, Shou Q, Fu H. Integrating 16srRNA sequencing, non-targeted metabolomics, and transcriptome sequencing to explore the mechanism of Total glucosides of paeony alleviating ulcerative colitis. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1259:124600. [PMID: 40286482 DOI: 10.1016/j.jchromb.2025.124600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
Total glucosides of paeony (TGP), an active ingredient extracted from the dried root of Paeonia lactiflora Pall., has been approved in China for the treatment of various autoimmune diseases. However, the role and mechanism of TGP in UC have yet to be fully elucidated. This study aims to investigate the regulatory effects and underlying mechanisms of TGP on intestinal homeostasis disruption and immune imbalance in a mouse model of dextran sulfate sodium (DSS)-induced colitis. The results showed that TGP alleviated DSS induced body weight loss, colonic shortening and histopathological changes in mice, and also enhanced the integrity of the intestinal barrier by up-regulating the expression of ZO-1, Occludin and tight junction protein in the colon. The results of 16S and antibiotic cocktail (ABX) experiments showed that TGP alleviated colitis by inhibiting Th17 cell differentiation by correcting intestinal microbial imbalance in UC mice. Mechanism studies showed that TGP inhibited the activation of JAK2/STAT3 signaling pathway in UC mice, and decreased the levels of inflammatory factors in colon supernatant and serum. Importantly, TGP regulates JAK2/STAT3 to inhibit Th17 cell differentiation depending on gut flora. In addition, TGP can also improve the metabolic imbalance in UC mice, especially purine metabolism. In conclusion, TGP promotes the normalization of purine metabolism and relies on gut microbiota to regulate JAK2/STAT3 pathway, inhibit Th17 cell differentiation, and alleviate colitis. Our findings highlight TGP as a promising treatment candidate for ulcerative colitis.
Collapse
Affiliation(s)
- Jianglin Chen
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qi Yuan
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Cui Yu
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Jie Fu
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Penghao Wang
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Shuiyan Tang
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xiaochen Lin
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Qiyang Shou
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Huiying Fu
- Second Clinical Medical School, Jinhua Academy, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| |
Collapse
|
22
|
Lei H, Liu Y, Li J, Chen J, Chen L, Liu Y, Liu H, Li W, Jiang Z, Li Z, Su X. Colon-targeted dual-coating MOF nanoparticles for the delivery of curcumin with anti-inflammatory properties in the treatment of ulcerative colitis. Colloids Surf B Biointerfaces 2025; 250:114545. [PMID: 39908958 DOI: 10.1016/j.colsurfb.2025.114545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
The inflammatory response is the core mechanism of the pathogenesis and symptoms of ulcerative colitis (UC), and inhibiting inflammation is a promising therapeutic approach to improving UC. Curcumin is considered a potential treatment for UC due to its significant anti-inflammatory and antioxidant effects. However, its bioavailability in the post-oral administration is limited. Therefore, the stability, sustained release, and colon targeting of curcumin in the treatment of UC have become a challenge. Herein, curcumin was efficiently filled in the porous structure of University of Oslo 66 (UiO-66). Amino-functionalized UiO-66 (MOF) was bound to hyaluronic acid (HA) via chemical crosslinking and electrostatic interactions. Polydopamine (PDA) layer was then applied to form Cur@MOF@HA-PDA NPs for colon targeting for UC treatment. Cur@MOF@HA-PDA NPs not only enhanced the stability of curcumin but also possessed acid resistance and reactive oxygen species (ROS) responsive properties, enabling it to be effectively delivered to the UC lesion site for curcumin release after oral administration, thereby enhancing the therapeutic effect. In vitro studies revealed that Cur@MOF@HA-PDA NPs possessed the ability to eliminate intracellular ROS, inhibit inflammatory (M1) polarization, and promote anti-inflammatory (M2) polarization. Additionally, in vivo experiments demonstrated that Cur@MOF@HA-PDA NPs could effectively alleviate the intestinal inflammatory symptoms of UC mice, promoting intestinal tissue repair. Furthermore, it was also confirmed that Cur@MOF@HA-PDA NPs achieved the treatment of UC by inhibiting inflammatory responses, modulating intestinal immune functions, and promoting the polarization of M2 macrophages. In short, Cur@MOF@HA-PDA NPs, as colon-targeted drug delivery nanosystems, offer a promising therapeutic strategy for the treatment of UC.
Collapse
Affiliation(s)
- Haoqiang Lei
- Huangpu People's Hospital of Zhongshan, Zhongshan, Guangdong 528429, PR China
| | - Yipeng Liu
- The Second People's Hospital of Foshan, Foshan, Guangdong 528000, PR China
| | - Jing Li
- Sunshine Lake Pharma Co., Ltd., Dongguan, Guangdong 523871, PR China
| | - Junyuan Chen
- Huangpu People's Hospital of Zhongshan, Zhongshan, Guangdong 528429, PR China
| | - Liji Chen
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong 523710, PR China
| | - Ying Liu
- Guangdong Huayi Biomedical Science and Technology Center, Guangzhou, Guangdong 511450, PR China
| | - Hongsheng Liu
- Guangdong Huayi Biomedical Science and Technology Center, Guangzhou, Guangdong 511450, PR China
| | - Wenqiang Li
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, PR China
| | - Zhuofei Jiang
- Dong Guan Maternal and Child Health Care Hospital, Dongguan, Guangdong 523808, PR China.
| | - Zhidong Li
- Guangdong Medical University, Dongguan, Guangdong 523808, PR China.
| | - Xiaohua Su
- The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong 523710, PR China; Guangdong Medical University, Dongguan, Guangdong 523808, PR China.
| |
Collapse
|
23
|
Sun H, Hu L, Hao P, Liu Y, Tian Y. BAICALIN INHIBITS CELL APOPTOSIS, INFLAMMATION, AND FERROPTOSIS IN ULCERATIVE COLITIS BY INFLUENCING SP1-MEDIATED TRANSCRIPTION OF SLC6A14. Shock 2025; 63:900-907. [PMID: 40138728 DOI: 10.1097/shk.0000000000002587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
ABSTRACT Background: Baicalin is considered to be able to alleviate the progression of ulcerative colitis (UC), but the underlying molecular mechanism needs to be further elucidated. Methods: TNF-α-induced human normal colorectal mucosa cells (FHC) were used to mimic UC models in vitro , and trinitrobenzene sulfonic acid (TNBS)-injected rats were used to construct UC models in vivo . Cell proliferation and apoptosis were determined by CCK8 assay, EdU assay and flow cytometry. Inflammation factors were examined by ELISA, and ferroptosis-related markers were detected by corresponding kit. The mRNA and protein levels of solute carrier family 6 member 14 (SLC6A14) and specific protein 1 (SP1) were analyzed by qRT-PCR and western blot. The interaction between SP1 and SLC6A14 promoter was verified by ChIP assay and dual-luciferase reporter assay. Results: Baicalin enhanced proliferation, while repressed apoptosis, inflammation, and ferroptosis in TNF-α-induced FHC cells. SLC6A14 was upregulated in UC patients, and baicalin could decrease SLC6A14 expression. SLC6A14 overexpression reversed the inhibitory effect of baicalin on TNF-α-induced FHC cell injury. SP1 could bind to SLC6A14 promoter region to upregulate its expression, and ectopic expression of SLC6A14 also abolished the suppressive effect of SP1 knockdown on TNF-α-induced FHC cell injury. Baicalin reduced SP1 expression to downregulate SLC6A14. In addition, baicalin alleviated UC process in vivo via repressing inflammation, and ferroptosis. Conclusion: Baicalin repressed SP1-mediated transcription of SLC6A14 to restrain cell apoptosis, inflammation, and ferroptosis, thus alleviating UC progression.
Collapse
Affiliation(s)
- Huifang Sun
- Department of Internal Medicine, Luquan District People's Hospital, Shijiazhuang, Hebei, China
| | - Lijuan Hu
- Department of Emergency, Hebei Yiling Hospital (Affiliated Hospital of Hebei Medical University), Shijiazhuang, Hebei, China
| | - Peipei Hao
- Department of Pediatrics, Renqiu People's Hospital, Cangzhou, Hebei, China
| | - Yawei Liu
- Department of endocrinology, Renqiu People's Hospital, Cangzhou, Hebei, China
| | - Ying Tian
- Department of Internal Medicine, Guangyang District People'S Hospital, Langfang, Hebei China
| |
Collapse
|
24
|
Deng B, Wang K, He H, Xu M, Li J, He P, Liu Y, Ma J, Zhang J, Dong W. Biochanin A mitigates colitis by inhibiting ferroptosis-mediated intestinal barrier dysfunction, oxidative stress, and inflammation via the JAK2/STAT3 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156699. [PMID: 40215818 DOI: 10.1016/j.phymed.2025.156699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory bowel disease marked by intestinal barrier dysfunction, oxidative stress, and inflammation. Biochanin A (BCA) is a natural flavonoid found in various plants, known for its anti-inflammatory, antioxidant, and anticancer properties, while its therapeutic role in UC and potential mechanism remains unexplored. PURPOSE To explores the therapeutic potential of BCA in alleviating UC, focusing on its effects on ferroptosis and the JAK2/STAT3 signaling pathway. METHODS The BCA's therapeutic effects on Dextran sulfate sodium (DSS)-induced colitis model in C57BL/6J mice was investigated. Subsequently, a comprehensive range of techniques was performed to investigate the impact of BCA on intestinal barrier integrity, oxidative stress, inflammation. Besides, the RNA sequencing was performed to explore the potential mechanism. The role of ferroptosis inhibition in BCA's effects in vitro and in vivo was explored by co-treating with the ferroptosis activator Erastin. RESULTS Treatment of colitis mice with BCA significantly improved DAI scores and histopathological damage scores, reduced oxidative stress, enhanced intestinal barrier function, and suppress inflammatory responses. RNA sequencing result found that BCA could lead to the inhibition of ferroptosis in mice colon tissue. Moreover, erastin co-treatment negated BCA's effects in vitro and vivo. Mechanistically, BCA exerts these effects by suppressing the JAK2/STAT3 pathway, which plays a pivotal role in ferroptosis and inflammation. Molecular docking studies further confirm the direct binding of BCA to both GPX4 and STAT3. CONCLUSION These results establish BCA as a promising natural compound for UC treatment, offering a novel therapeutic strategy by specifically targeting ferroptosis and its associated molecular pathways, thereby addressing key gaps in current UC management and advancing potential clinical applications.
Collapse
Affiliation(s)
- Beiying Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Digestive System Disease, Wuhan, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Kunpeng Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Laboratory of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Haodong He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Miao Xu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Digestive System Disease, Wuhan, China
| | - Jiao Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pengzhan He
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yinghui Liu
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingjing Ma
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jixiang Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
25
|
Wang W, Wen Y, Luo J, Miao Y, Zhang F, Niu J. Heat shock transcription factor 2 reduces mitochondrial pathway apoptosis in intestinal epithelial cells by inhibiting the increase in mitochondrial membrane permeability in ulcerative colitis. PLoS One 2025; 20:e0325275. [PMID: 40440295 PMCID: PMC12121780 DOI: 10.1371/journal.pone.0325275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 05/08/2025] [Indexed: 06/02/2025] Open
Abstract
The destruction of intestinal mucosal mechanical barrier homeostasis caused by excessive apoptosis of intestinal epithelial cells (IECs) is an important reason for the occurrence and development of ulcerative colitis (UC). The increase in mitochondrial membrane permeability caused by the opening of the mitochondrial membrane permeability transition pore (mPTP) is a key link in the initiation of mitochondrial pathway apoptosis. Our previous studies revealed that heat shock transcription factor 2 (HSF2), which is highly expressed in the intestinal mucosa of UC patients, can inhibit the expression of the cytochrome C (Cyto-C)/Caspase-9/Caspase-3 proteins in the mitochondrial pathway of apoptosis, but the regulatory mechanism is unknown. It has been reported that heat shock proteins regulated by heat shock transcription factors are closely related to mPTP opening. Therefore, we hypothesized that HSF2 affects mitochondrial pathway apoptosis in IECs by regulating mPTP opening. In this study, we altered the level of HSF2 in Caco-2 cells by lentivirus transfection to explore the changes in the mitochondrial membrane permeability of Caco-2 cells in an inflammatory environment. Subsequently, the mPTP agonist atractylorhizin (Atr) and inhibitor cyclosporine A (CsA) were used to clarify the regulatory effects of HSF2 on mPTP and the Cyto-C/Caspase-9/Caspase-3 pathways. Our study confirmed for the first time that HSF2 plays a protective role in UC by inhibiting mPTP opening, the increase in mitochondrial membrane permeability and the activation of the mitochondrial-mediated apoptosis pathway in IECs.
Collapse
Affiliation(s)
- Wen Wang
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming, Yunnan, China
| | - Yunling Wen
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming, Yunnan, China
| | - Juan Luo
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming, Yunnan, China
| | - Yinglei Miao
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming, Yunnan, China
| | - Fengrui Zhang
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming, Yunnan, China
| | - Junkun Niu
- Department of Gastroenterology, First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming, Yunnan, China
| |
Collapse
|
26
|
Zhang S, Qian Y, Li N, Zhu Q, Zhang S, Wen P, Xiao Y, Yan C, Lin Z, Zhong J, Ma J, Wu X, Zhuang G, Zhang K. Specific MSI2 deletion maintains intestinal barrier integrity by down-regulating ILC3s-derived IL-17 a in mice with colitis. Int Immunopharmacol 2025; 156:114717. [PMID: 40279942 DOI: 10.1016/j.intimp.2025.114717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is an inflammatory bowel disease with an unknown cause. Previous studies have shown that Group 3 innate lymphoid cells (ILC3s) are crucial for maintaining intestinal mucosal immune homeostasis by producing key cytokines such as IL-22 and IL-17 A. While the RNA-binding protein Musashi-2 (MSI2) is recognized as essential for promoting intestinal epithelial regeneration post-injury, its impact on immune regulation remains unclear. Therefore, we aim to investigate the protective mechanisms associated with ILC3s-specific MSI2 deletion in a mouse model of ulcerative colitis. METHODS Dextran sulfate sodium (DSS) was used to induce a mouse colitis model. Colitis severity was evaluated through weight loss, diarrhea, fecal traits, colon length, and pathological scoring. Transcriptome sequencing was utilized to identify differentially expressed genes in colon tissues. Flow cytometry was employed to measure the quantity and functionality of ILC3s. Western blot was conducted to analyze protein expression, while real-time polymerase chain reaction and enzyme-linked immunosorbent assay were employed to quantify inflammatory factors. Additionally, immunofluorescence, AB-PAS staining, and immunohistochemistry were employed to evaluate the integrity of the intestinal barrier. RESULTS Following DSS treatment, colon damage was milder in Msi2∆Rorc mice than in Msi2fl/fl mice. Transcriptomic analysis revealed the down-regulation of cytokines and pro-inflammatory factors in the colon tissue of Msi2∆Rorc mice. Flow cytometry showed that specific deletion of MSI2 reduced the infiltration of ILC3s in the intestinal lamina propria of Msi2∆Rorc mice and decreased IL-17 A production. The reduction of IL-17 A-mediated immune responses lessened inflammatory damage to the intestinal barrier, thereby reducing colitis severity. CONCLUSIONS Specific deletion of MSI2 alleviates DSS-induced colitis in mice by reducing ILC3s infiltration and IL-17 A secretion in the lamina propria of the colon. This decrease in inflammatory mediators and cell infiltration dampens the inflammatory response in the intestinal mucosa, helping to maintain the integrity of the intestinal barrier in mice with colitis. These findings enhance our understanding of UC pathogenesis and offer novel avenues for clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Shuaishuai Zhang
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Yunyun Qian
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Nengneng Li
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Qiang Zhu
- Department of General Surgery, First General Hospital of Fuzhou, Fujian Medical University, 350005 Fuzhou, Fujian, China
| | - Shiying Zhang
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Peizhen Wen
- Department of General Surgery, Changzheng Hospital, Navy Medical University, 415 Fengyang Road, 200003 Shanghai, China
| | - Yi Xiao
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Changxiu Yan
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Zeyang Lin
- Department of Pathology, Zhongshan Hospital, Xiamen University, 361001 Xiamen, Fujian, China
| | - Jianfa Zhong
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Jingmiao Ma
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Xia Wu
- Department of Organ Transplantation, Xiang'an Hospital, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China; Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China
| | - Guohong Zhuang
- Organ Transplantation Institute of Xiamen University, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, 361102 Xiamen, Fujian, China.
| | - Kun Zhang
- Department of General Surgery, First General Hospital of Fuzhou, Fujian Medical University, 350005 Fuzhou, Fujian, China.
| |
Collapse
|
27
|
Wang Q, Li L, Zhuang S, Huang M, Xiang Y. Efficacy of polyphenols in adjuvant treating ulcerative colitis: A meta-analysis of randomized controlled trials. Medicine (Baltimore) 2025; 104:e41985. [PMID: 40419878 DOI: 10.1097/md.0000000000041985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND The effectiveness and safety of polyphenols in treating ulcerative colitis remain controversial. This study aimed to evaluate the efficacy and safety of polyphenols in the treatment of ulcerative colitis. METHODS This study followed the preferred reporting items for systematic reviews and meta-analyses 2020 guidelines. A systematic search was conducted across PubMed, Embase, Web of Science, and Cochrane databases to identify relevant articles. The random-effects model was employed to calculate the odds ratio (OR) and corresponding 95% confidence intervals (CIs). RESULTS A total of 13 trials involving 742 participants were included in the meta-analysis. The clinical remission rates were higher in the polyphenol group compared to the control group, as demonstrated by both the intention-to-treat (ITT) (OR: 4.71; 95% CI: 2.02-10.99; P = .000) and per-protocol (PP) analysis (OR: 7.14; 95% CI: 3.11-16.39; P = .000). Similarly, the clinical response rate was higher in the polyphenol group compared to the control group, according to the ITT (OR: 5.40; 95% CI: 2.60-11.24; P = .000) and PP analysis (OR: 9.14; 95% CI: 4.25-19.64; P = .000). Moreover, the total endoscopic remission rate was superior in the polyphenol group, as indicated by the ITT (OR: 3.16; 95% CI: 1.20-8.37; P = .020) and PP analysis (OR: 4.92; 95% CI: 2.03-11.93; P = .000). No significant differences were observed regarding side effects between the 2 groups, according to the ITT (OR: 0.99; 95% CI: 0.56-1.76; P = .973) and PP analysis (OR: 0.99; 95% CI: 0.54-1.80; P = .971). CONCLUSION Polyphenols demonstrated effectiveness in inducing clinical remission, clinical response, and endoscopic remission in patients with ulcerative colitis. Furthermore, there was no evidence of a higher incidence of adverse effects associated with their use.
Collapse
Affiliation(s)
- Qiuxiang Wang
- Department of Traditional Chinese Medicine, Guangyuan Central Hospital, Sichuan Province, China
| | - Liuying Li
- Department of Traditional Chinese Medicine, Zigong First People's Hospital, Sichuan Province, China
| | - Shuhan Zhuang
- Zhejiang Chinese Medical University, Zhejiang Province, China
| | - Mei Huang
- The Second Hospital of Traditional Chinese Medicine of Yibin City, Sichuan Province, China
| | - Yongguo Xiang
- Department of Traditional Chinese Medicine, Guangyuan Central Hospital, Sichuan Province, China
| |
Collapse
|
28
|
Kuriakose Kuzhiyanjal AJ, Rhodes S, Liu E, Limdi JK. Endoscopic Scoring in Ulcerative Colitis: Evaluating Practice Patterns and Role of Educational Interventions. Br J Hosp Med (Lond) 2025; 86:1-12. [PMID: 40405852 DOI: 10.12968/hmed.2024.0859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Aims/Background Endoscopic scoring systems are recommended internationally for assessing disease activity, response to therapy and mucosal healing. However, their real-world application remains inconsistent. This study aimed to evaluate the impact of an educational intervention on endoscopic scoring documentation and identify factors influencing its use. Methods A retrospective observational study was conducted at four hospital sites in Greater Manchester, UK. Data from endoscopies performed on ulcerative colitis (UC) patients were compared before and after an educational intervention. Logistic regression was used to analyse factors affecting documentation rates. Results Endoscopic score documentation increased from 39% (pre-intervention) to 46% (post-intervention) (p = 0.162). Nurse endoscopists had the highest documentation rates (83%), while surgeons had the lowest (8%). Attendance at educational sessions significantly increased documentation rates (29% vs. 74-80%, p < 0.001). Conclusion Educational interventions modestly improved endoscopic scoring documentation. Further targeted training and standardised reporting templates are needed to enhance adherence and patient outcomes in UC management.
Collapse
Affiliation(s)
| | - Sarah Rhodes
- Centre for Biostatistics, Division of Population Health, Health Services Research and Primary Care, University of Manchester, Manchester, UK
| | - Eleanor Liu
- Division of Gastroenterology-Section of IBD, Northern Care Alliance NHS Foundation Trust, Manchester, UK
| | - Jimmy K Limdi
- Division of Gastroenterology-Section of IBD, Northern Care Alliance NHS Foundation Trust, Manchester, UK
- Manchester Academic Health Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
29
|
Doggrell SA. With guselkumab, does the dual mechanisms to inhibit IL-23, help in ulcerative colitis? Expert Opin Biol Ther 2025:1-6. [PMID: 40394835 DOI: 10.1080/14712598.2025.2508836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 05/16/2025] [Indexed: 05/22/2025]
Abstract
INTRODUCTION Ulcerative colitis (UC) includes a dysregulated immune response. The conventional therapy includes immunosuppressants, and biologics targeting inflammatory mediators, but these are often inadequate, or subjects become unable to tolerate them. AREAS COVERED QUASAR: the induction and maintenance components of the phase 3 trial of guselkumab, which inhibits IL-23 by dual mechanisms, in subjects with moderate-to-severe UC. QUASAR enrolled those that had an inadequate response and/or intolerance to corticosteroids, immunosuppressants, biologics, or Janus kinase (JAK) inhibitors. In both parts of the trial, guselkumab improved clinical remission with no excess of adverse events. EXPERT OPINION For those enrolled throughout, after the maintenance part, the benefit with guselkumab on clinical remission was 24% percentage points (45 vs 21%), which is relatively small. There is no direct comparison of guselkumab with other IL-23 inhibitors in UC. Indirectly comparing trials suggests the clinical remission rates at the end of the trials was higher with guselkumab than with the other approved IL-23, inhibitors, mirikizumab or risankizumab (17 or 15% points, respectively). Thus, guselkumab may be more efficacious than the other 1 L-23 antagonists, possibly due to its additional action to block the CD64 receptor. However, this needs to be tested in a direct comparison trial.
Collapse
Affiliation(s)
- Sheila A Doggrell
- School of Pharmacy and Medical Sciences, Gold Coast Campus, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
30
|
Pereira CH, Kittaka H, Ouille V EJ, Almeida JFQ, Pelaez A, Keshavarzian A, Blatter LA, Banach K. Colitis induced ventricular alternans increases the risk for ventricular arrhythmia. J Mol Cell Cardiol 2025:S0022-2828(25)00086-0. [PMID: 40409406 DOI: 10.1016/j.yjmcc.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 05/08/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
Inflammatory bowel disease was linked to an increased risk for conduction defects and ventricular arrhythmia. It coincides with dysregulation of gut microbiota, increased inflammation, and deregulation of the renin-angiotensin system. In this study, we aimed to determine the mechanism of colitis-induced electrophysiological remodeling that increases the risk for ventricular arrhythmia. In a mouse model of dextran sulfate sodium induced active colitis (3.5 %, 7 days) cardiac electrophysiological properties were quantified during active inflammation. Electrocardiographic recordings exhibited a prolonged QT duration in mice with active colitis compared to control. Field potential (FP) recordings of Langendorff perfused colitis-hearts exhibited increased FP dispersion, a reduced threshold for ventricular alternans, and an increased propensity for spatially discordant alternans. The increased propensity for alternans was also reflected in isolated ventricular myocytes where Ca2+ transient alternans occurred at lower pacing frequencies and increased alternans ratios. The action potential was unchanged during colitis but myocytes exhibited a prolonged Ca2+ transient duration that corresponded with attenuated phospholamban phosphorylation. Stimulating cellular SERCA activity (Istaroxime), normalized the propensity for alternans. Serum levels of Angiotensin II (AngII) were increased during colitis and Angiotensin-converting enzyme (ACE) inhibitor or AngII receptor type 1 blocker prevented the increased alternans inducibility in isolated myocytes and hearts. Our data demonstrate that active colitis promotes reversible remodeling of ventricular Ca2+ handling properties and increases the propensity for alternans and arrythmia. The changes can be prevented by ACE or AT1R inhibition supporting a cardiac benefit for controlling RAS signaling in patients with active colitis.
Collapse
Affiliation(s)
- Carlos H Pereira
- Department of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL 60612, USA
| | - Hiroki Kittaka
- Department of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL 60612, USA.
| | - Edward J Ouille V
- Department of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL 60612, USA.
| | - Jonathas F Q Almeida
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, 1220 E. Broad St, Richmond, VA 23298, USA.
| | - Andres Pelaez
- Department of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL 60612, USA.
| | - Ali Keshavarzian
- Rush University Medical Center, 1725 W. Harrison St., Chicago, IL 60612, USA.
| | - Lothar A Blatter
- Dept. of Physiology & Biophysics, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL 60612, USA.
| | - Kathrin Banach
- Department of Internal Medicine/Cardiology, Rush University Medical Center, 1750 W. Harrison St., Chicago, IL 60612, USA.
| |
Collapse
|
31
|
Zhou P, Tang T, Zhao P, Wang Q, Hu X, Si J, Yang T, Zhou S, An W, Jiang Y. Unveiling the hidden dance: SPP1 + macrophages identified in ulcerative colitis reveal crosstalk with CHI3L1 + fibroblasts. J Transl Med 2025; 23:567. [PMID: 40399882 PMCID: PMC12093798 DOI: 10.1186/s12967-025-06565-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/30/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by persistent inflammation of the colon. The specific cause of UC is still not fully understood, but this condition is believed to arise from a combination of environmental, genetic, microbial, and immune factors. This study aimed to explore the specific roles of macrophages and fibroblasts in UC pathogenesis, focusing on their interactions and contributions to disease progression. METHODS We utilized single-cell RNA sequencing (scRNA-seq) to analyze macrophages and fibroblasts in peripheral blood and colon biopsy samples from UC patients. Bulk RNA sequencing and spatial transcriptomic data from the Gene Expression Omnibus (GEO) database and flow cytometry and multiplex immunohistochemistry (mIHC) data were used for validation. Statistical analyses were performed to assess the correlation between cell abundance and disease severity. RESULTS Macrophages and fibroblasts were identified as key communication hubs in UC; specifically, SPP1 + macrophages and CHI3L1 + fibroblasts were significantly enriched at the sites of inflammation. These cells are strongly correlated with disease severity and orchestrate inflammatory responses within the intestinal immune microenvironment, contributing to UC-associated colorectal cancer. CONCLUSIONS Our study identified SPP1 + macrophages and CHI3L1 + fibroblasts as key contributors to UC pathogenesis. These cells are enriched in inflammatory sites, are correlated with disease severity, and play a role in UC-associated colorectal cancer, providing new insights into UC mechanisms.
Collapse
Affiliation(s)
- Peiwen Zhou
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Tongyu Tang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Pingwei Zhao
- Department of Gastrointestinal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Quan Wang
- Department of Gastrointestinal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xintong Hu
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Junzhuo Si
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Tianshi Yang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shuai Zhou
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Wenyan An
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yanfang Jiang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
32
|
Liao Z, Liu X, Li L, Li S, Xing X, Zheng X, Song W, Gui P, Liu Q, Rong G, Shao Y, Zou M, Liao H, Wu X. Mechanism of the Proprietary Chinese Medicine "JiuLiWan" to Treat Ulcerative Colitis Revealed by Network Pharmacology, Molecular Docking, and Experimental Verification In Vitro. ACS OMEGA 2025; 10:19598-19613. [PMID: 40415848 PMCID: PMC12096223 DOI: 10.1021/acsomega.5c00261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/14/2025] [Accepted: 03/25/2025] [Indexed: 05/27/2025]
Abstract
JiuLiWan (JLW), as a classic traditional Chinese medicine formula, has been clinically used against ulcerative colitis (UC). However, the exact mechanism of its therapeutic effect remains unclear. This study aims to explore and validate the main components and pharmacological mechanism of JLW in the treatment of UC through network pharmacology, molecular docking, and cell experiments. Network pharmacology analyses indicated a total of 107 main components and 286 core targets of JLW against UC. Pathway enrichment analysis demonstrated the involvement of PI3K-AKT, MAPK, Ras, Rap1, TNF, T cell receptor, HIF-1, C-type lectin receptor, VEGF, and Th17 cell differentiation signal pathways in the efficacy of the formula. The molecular docking results indicated that the prominent components (ailanthone (AIL), butylidenephthalide, honokiol, dehydrocostuslactone, ganoderic acid A, atractylenolide I, neokurarinol, glycyrrhetinic acid, palmatine, tangeretin, and bruceine A) could bind to core targets AKT1, P53, STAT3, c-JUN, and ERK1. Subsequently, AIL was used as a representative compound to conduct cell experiments to verify its role and mechanism in anti-inflammation and immunomodulation. Interestingly, AIL could switch Jurkat T cells into a quiescence state without activating the inflammatory and immune status. However, AIL could significantly decrease the levels of interleukin-2 (IL-2) and interferon-gamma (IFN-γ), as well as the expression of surface activation markers CD69 and CD25, in PMA/ionomycin-activated Jurkat T cells by suppressing the RAF/ERK/STAT3 signaling pathway and increasing the phosphorylation of p53. This study combines network pharmacology prediction with experimental verification in vitro to demonstrate the mechanism of JLW in treating UC and provides an effective, safe, and inexpensive strategy for UC treatment.
Collapse
Affiliation(s)
- Zhifang Liao
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Xiao Liu
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Linxuan Li
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
- Interdisciplinary
Science Research Center of Western Guangdong, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| | - Sikai Li
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Xingxing Xing
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Xiwen Zheng
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Wenyu Song
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Pin Gui
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Qi Liu
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Guanghong Rong
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
| | - Yiming Shao
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| | - Mingzhi Zou
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
- Interdisciplinary
Science Research Center of Western Guangdong, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| | - Hongbo Liao
- Guangdong
Provincial Key Laboratory of Research and Development of Natural Drugs,
School of Pharmacy, Guangdong Medical University, Zhanjiang, Guangdong Province524023, P.R. of
China
| | - Xin Wu
- Dongguan
Key Laboratory of Characteristic Research and Achievement Transformation
of Integrated Chinese and Western Medicine for Prevention and Treatment
to Common Diseases, The First Dongguan Affiliated
Hospital of Guangdong Medical University, Guangdong Medical University, Dongguan, Guangdong Province523000, P. R. of
China
- The Key Laboratory
of Sepsis Translational Medicine, The Second
Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong Province524003, P.R. of
China
| |
Collapse
|
33
|
Dong L, Wang W, Zheng H, Sun Y, Han S. Construction of Mn 2+-Polyphenol Nanoparticles and Its Application in the Treatment of Ulcerative Colitis. ACS APPLIED BIO MATERIALS 2025; 8:4367-4382. [PMID: 40340318 DOI: 10.1021/acsabm.5c00471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease characterized by chronic inflammation and ulcerative erosion of the colonic mucosa. Because of the complex causes, UC is often difficult to completely cure, greatly increasing the risk of bowel cancer and other diseases. In the UC environment, excessive production of reactive oxygen species (ROS) and high levels of inflammatory factors lead to the continuous deterioration of inflammation. We developed Mn-PEGCG nanoparticles (NPs) based on the metal-polyphenol network for the preventive treatment of UC. Mn-PEGCG NPs were synthesized by the polymerization of epigallocatechin gallate (EGCG) and further complexation with Mn2+. After oral administration, Mn-PEGCG NPs are more likely to reach the inflammation site of the colon through electrostatic interaction, effectively clear ROS, reduce the production of pro-inflammatory cytokines, exert antioxidant and anti-inflammatory effects, and protect colon cells from oxidative stress damage. In addition, it can play the role of EGCG in promoting the expression of tight junction protein and enhancing the intestinal epithelial barrier. In a mouse model of UC, oral administration of Mn-PEGCG NPs reduced intestinal inflammation, alleviated pathological structural damage in mice, and promoted mucus secretion and tight junction protein expression, thereby strengthening the intestinal barrier. Mn-PEGCG NPs can effectively alleviate inflammation and repair the intestinal barrier to maintain the stability of the intestinal environment, which is an ideal treatment strategy for UC.
Collapse
Affiliation(s)
- Liangyu Dong
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, People's Republic of China
| | - Wenyu Wang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, People's Republic of China
| | - Huapeng Zheng
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, People's Republic of China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, People's Republic of China
| | - Shangcong Han
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266071, People's Republic of China
| |
Collapse
|
34
|
Cassaro F, Impellizzeri P, Romeo C, Arena S. Comparative outcomes of laparoscopic and open surgery in inflammatory bowel disease in pediatric and young adult patients: a systematic review and meta-analysis. J Gastrointest Surg 2025; 29:102085. [PMID: 40398665 DOI: 10.1016/j.gassur.2025.102085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/07/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Pediatric inflammatory bowel disease (IBD), encompassing Crohn's disease, ulcerative colitis, and indeterminate colitis, often necessitates surgical intervention in cases of severe or refractory disease. Although biologic therapies have significantly reduced the need for surgery, operative management remains essential for certain patients. The choice between laparoscopic (laparoscopy group [LG]) and open conventional surgery (open group [OG]) continues to be a subject of debate. This meta-analysis aimed to compare the postoperative outcomes of LG and OG in pediatric patients with IBD. METHODS We conducted a meta-analysis of observational studies comparing LG and OG outcomes in pediatric patients with IBD. Key outcomes analyzed included major and minor postoperative complications, reoperations, readmissions, operative time, and length of hospital stay. RESULTS Seven studies met the inclusion criteria, analyzing 3417 patients, with 1399 (41%) undergoing OG and 2018 (59%) undergoing LG. Our analysis revealed no significant differences in major postoperative complications, reoperation, and readmissions between LG and OG (P = .114, P = .082, and P = .641, respectively). However, LG was associated with shorter hospital stays (6.04 vs 8.35 days; P < .05). Conversions from LG to open surgery amounted to a total of 153 (7.57%). Open surgery had a significantly shorter operative time (173.8 vs 195.5 min; P = .005). CONCLUSION Both laparoscopic and open conventional surgeries are safe, effective, and reliable in managing pediatric IBD. Although open surgery offers shorter operative times, laparoscopy reduces hospital stay and minor postoperative complications. The choice of approach depends on the surgeon's experience and patient-specific factors.
Collapse
Affiliation(s)
- Fabiola Cassaro
- Unit of Pediatric Surgery, Department of Human Pathology of Adult and Childhood "Gaetano Barresi," University of Messina, Messina, Italy; Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, Messina, Italy
| | - Pietro Impellizzeri
- Unit of Pediatric Surgery, Department of Human Pathology of Adult and Childhood "Gaetano Barresi," University of Messina, Messina, Italy
| | - Carmelo Romeo
- Unit of Pediatric Surgery, Department of Human Pathology of Adult and Childhood "Gaetano Barresi," University of Messina, Messina, Italy
| | - Salvatore Arena
- Unit of Pediatric Surgery, Department of Human Pathology of Adult and Childhood "Gaetano Barresi," University of Messina, Messina, Italy.
| |
Collapse
|
35
|
Li L, He Y, Chen Y, Zhou X. cGAS-STING Pathway's Impact on Intestinal Barrier. J Gastroenterol Hepatol 2025. [PMID: 40377214 DOI: 10.1111/jgh.16974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/09/2025] [Accepted: 04/03/2025] [Indexed: 05/18/2025]
Abstract
Intestinal inflammation and increased permeability have been linked to metabolic dysregulation in patients with compromised intestinal barrier function. Among the pathways, garnering attention is the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. Upon binding to double-stranded DNA (dsDNA), cGAS catalyzes the conversion of ATP and GTP into cyclic GMP-AMP (cGAMP). Subsequently, cGAMP binds to STING, triggering the activation of tank-binding kinase 1 (TBK1), which activates interferon regulatory factor 3 (IRF3), thus inducing the production of type I interferon. Activated TBK1 can also induce the activation of nuclear factor κB (NF-κB), thus mediating the production of proinflammatory cytokines. The effects of this process vary among innate and adaptive immune cells, as well as intestinal epithelial cells (IECs). This review aims to elucidate the impact and role of the cGAS-STING pathway on intestinal barrier function.
Collapse
Affiliation(s)
- Liqi Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yingge He
- Department of Thyroid and Breast Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu Chen
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Xiaoshu Zhou
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
36
|
Zhou J, Guo Y, Tian Z, Lv Z, Jiang S, Zhang W. Aberrant formation of the neutrophil extracellular trap and the expression of the PLEKHA1 in systemic lupus erythematosus and ulcerative colitis. Mol Cell Biochem 2025:10.1007/s11010-025-05300-4. [PMID: 40379887 DOI: 10.1007/s11010-025-05300-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 04/27/2025] [Indexed: 05/19/2025]
Abstract
Systemic lupus erythematosus (SLE) and ulcerative colitis (UC) are both chronic autoimmune diseases with unclear shared mechanisms, largely due to limited mechanistic studies and clinical research cohorts. Transcriptome datasets from the Gene Expression Omnibus (GEO) database were analyzed for SLE and UC, identifying differentially expressed genes (DEGs). Weighted gene co-expression network analysis (WGCNA) identified significant module genes, including PLEKHA1. The diagnostic potential of PLEKHA1 was confirmed using machine-learning algorithms and real-time fluorescence quantitative PCR (RT-PCR) in clinical samples. Additionally, the study explored the link between PLEKHA1 and neutrophil extracellular trap (NET) formation. Our analyses identified transcriptional signatures associated with neutrophil degranulation and NET formation pathways in the peripheral blood of both SLE and UC, a perspective not previously explored. PLEKHA1 was identified as a promising biomarker that may impact NET formation. Pathway enrichment analyses indicated that PLEKHA1 plays a regulatory role in NET formation in both diseases. This study provides novel transcriptional evidence by proposing neutrophil degranulation and NET formation as common pathways in SLE and UC, with PLEKHA1 acting as a shared diagnostic gene. PLEKHA1 may regulate neutrophil activation and immune response, influencing NET formation and neutrophil degranulation in SLE patients' peripheral blood.
Collapse
Affiliation(s)
- Jieyu Zhou
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- Department of Medical Laboratory Science, Xiangya Medical College, Central South University, Changsha, Hunan, China
| | - Yilin Guo
- Department of Blood Transfusion, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan, China
| | - Ziying Tian
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- Department of Medical Laboratory Science, Xiangya Medical College, Central South University, Changsha, Hunan, China
| | - Zihan Lv
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- Department of Medical Laboratory Science, Xiangya Medical College, Central South University, Changsha, Hunan, China
| | - Su Jiang
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
- Department of Medical Laboratory Science, Xiangya Medical College, Central South University, Changsha, Hunan, China
| | - Wenling Zhang
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
- Department of Medical Laboratory Science, Xiangya Medical College, Central South University, Changsha, Hunan, China.
| |
Collapse
|
37
|
Chen H, Sun W, Li C, Wang Q, Wang X, Du Y, Chen W, Wang M, Huang C, Wang R. Inflammatory targeted nanoplatform incorporated with antioxidative nano iron oxide to attenuate ulcerative colitis progression. iScience 2025; 28:112448. [PMID: 40343277 PMCID: PMC12059676 DOI: 10.1016/j.isci.2025.112448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/07/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
Antioxidative nanomaterials with reactive oxygen species (ROS) scavenging capabilities hold promise for the treatment of ulcerative colitis (UC). However, their clinical application is limited by rapid diffusion, susceptibility to inactivation, and insufficient targeting of inflammatory sites. This study focuses on developing a nanoplatform by integrating iron oxide nanoparticles (IONPs) into zeolitic imidazolate frameworks-8 (ZIF-8), termed as ZIF-8@IONPs. ZIF-8@IONPs exhibited good biocompatibility and effective ROS scavenging capabilities in RAW 264.7 cells. To enhance inflammatory targeting, HA@ZIF-8@IONPs were generated through hyaluronic acid (HA) surface modification. HA@ZIF-8@IONPs effectively reduced damage to intestinal tissues in the UC mouse model. Mechanistic revealed that HA@ZIF-8@IONPs exhibited antioxidant and anti-inflammatory activities by eliminating endogenous ROS, activating the Nrf2 signaling pathway, and inhibiting the NF-κB signaling pathway. This study highlights the nanoplatform's potential as a promising candidate for UC treatment due to its great targeting of inflammatory microenvironments and efficient ROS scavenging.
Collapse
Affiliation(s)
- Haojun Chen
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Wei Sun
- Department of Orthopedics, The Jiangyin Clinical College of Xuzhou Medical University, 163 Shoushan Road, Jiangyin 214400, Jiangsu, P.R. China
| | - Can Li
- School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qiuyang Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xucai Wang
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Yingjie Du
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Wenbo Chen
- Department of Wood Science, The University of British Columbia, 2424 Main Mall, Vancouver, BC V6T 1Z4, Canada
| | - Min Wang
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University & Jiangsu Province Hospital, Nanjing 210000, China
| | - Caoxing Huang
- Co-Innovation Center for Efficient Processing and Utilization of Forest Resources, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Rong Wang
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, Jiangsu, China
| |
Collapse
|
38
|
Yang X, Yang J, Wang A, Zheng Y, Lin J, Kong Z, Tian Y, Dong H, Zhang Z, Song R. Saikosaponin A ameliorates ulcerative colitis by targeting the CH25H/25-OHC axis to inhibit NLRP3 inflammasome in macrophages. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156844. [PMID: 40414048 DOI: 10.1016/j.phymed.2025.156844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/02/2025] [Accepted: 05/11/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) has demonstrated an escalating global incidence and prevalence, thereby posing substantial challenges to public health. Despite recent advancements in therapeutic interventions, the clinical management of UC remains suboptimal, underscoring the urgent need for novel treatment strategies. Saikosaponin A (SSa), a bioactive compound derived from the traditional Chinese herb Radix Bupleuri (RB), exhibits potent anti-inflammatory and immunomodulatory effects. However, its effects on UC and the underlying molecular mechanisms remain to be thoroughly explored. PURPOSE This study aims to elucidate the underlying mechanisms of SSa in ameliorating UC and establish a pharmacological foundation for developing novel treatment modalities to address unmet clinical needs in UC treatment. METHODS The protective effects of SSa against DSS-induced acute colitis were evaluated in a 3 % DSS-treated mouse model. Histological (H&E staining) and molecular analyses (RT-qPCR, ELISA, Western blotting, and flow cytometry) were performed to assess colonic tissue damage and inflammatory responses. Macrophage depletion via tail vein injection of clodronate liposomes confirmed the pivotal role of macrophages in UC pathogenesis and SSa's anti-inflammatory effects. The inhibitory effects of SSa on NLRP3 inflammasome activation were analyzed in vivo and in LPS/ATP-stimulated bone marrow-derived macrophages (BMDMs) using RT-qPCR, ELISA, and Western blotting. Bioinformatics analysis, targeted LC-MS/MS, and molecular docking were employed to identify potential molecular targets and mechanisms of SSa. Drug affinity responsive target stability (DARTS), cellular thermal shift assay (CETSA), and CH25H siRNA knockdown assays were used to validate CH25H as the direct target of SSa. RESULTS SSa effectively attenuated DSS-induced colitis in mice by alleviating colonic inflammation, preserving intestinal barrier integrity, reducing LPS translocation, and mitigating systemic organ injury in the liver and spleen. The inflammatory response of macrophages and the production of IL-1β were identified as key pathogenic components in colitis, and the clearance of macrophages significantly ameliorated colitis progression while SSa administration post-macrophage clearance did not further alter the disease phenotype. Mechanistically, SSa inhibited the NLRP3 inflammasome activation-mediated IL-1β secretion in macrophages. The sterol metabolism played a crucial role in the activation of the NLRP3 inflammasome. SSa also restored the disturbed sterol homeostasis in the colon under inflammatory conditions, especially promoted the synthesis of 25-hydroxycholesterol (25-OHC). Further investigation revealed that SSa primarily exerts its therapeutic effects by directly targeting cholesterol 25-hydroxylase (CH25H), which promotes the production of 25-OHC and inhibits macrophage NLRP3 inflammasome activation. CONCLUSION This pioneering study demonstrated the therapeutic effect of SSa on DSS-induced colitis by targeting CH25H to enhance 25-OHC biosynthesis, which subsequently inhibited NLRP3 inflammasome activation in IL-1β-producing macrophages. These findings reveal a novel mechanism of SSa in UC treatment through cholesterol metabolism-regulated cascade immune modulation, providing strong pharmacological support for its development as a potential UC therapy.
Collapse
Affiliation(s)
- Xue Yang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Jinni Yang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Anhui Wang
- Institute of Materia Medica Chinese Academy of Medical Science, Beijing 100050, China
| | - Yuan Zheng
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Jiachun Lin
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Ziwen Kong
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan Tian
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Haijuan Dong
- The Public Laboratory Platform of China Pharmaceutical University, Nanjing, 210009, China
| | - Zunjian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
| | - Rui Song
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
39
|
Lin Z, Wang J, Luo H, Huang L, Pan Z, Yang S, Zhong C, Shan NC, Ye Z, Tan H, Yang X, Zhang B, Huang C, Zhang H. Changdiqing decoction (CDQD) ameliorates colitis via suppressing inflammatory macrophage activation and modulating gut microbiota. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156856. [PMID: 40412060 DOI: 10.1016/j.phymed.2025.156856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 05/11/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is a non-specific inflammatory bowel disease. Unlike any single form of cell death reported previously, macrophage PANoptosis, a unique programmed cell death characterized by inflammation and necrosis, plays a crucial role in the pathogenesis of colitis. Changdiqing Decoction (CDQD), an empirical hospital prescription enema, has been used to treat UC for decades. This study aimed to investigate the multi-target anti-colitic effects of CDQD by examining its impact on intestinal homeostasis and its anti-inflammatory properties. METHODS A dextran sulfate sodium (DSS)-induced mouse model of acute colitis was employed. Interferon-gamma (IFN-γ) and KPT-330 were used to induce macrophage PANoptosis. Ultra-high-performance liquid chromatography-high-resolution mass spectrometry (UHPLCHRMS) was utilized to identify the chemical constituents of CDQD. Multi-omics analysis and fecal microbiota transplantation (FMT) were used to explore the therapeutic targets and gut microbiota alterations induced by CDQD. RESULTS CDQD treatment significantly alleviated colitis symptoms in mice, with a dose-dependent therapeutic effect. The decoction mitigated PANoptosis in colon tissues and bone marrow-derived macrophages (BMDMs). 16S rRNA sequencing analysis and metabonomics revealed that CDQD administration significantly altered the gut microbiota composition and metabolite profiles. Notably, CDQD-modulated gut microbiota exhibited anti-colitic effects through FMT. Integrated transcriptomics and network pharmacology analysis revealed that CDQD significantly downregulated the PI3K/Akt signaling pathway in colitis. This finding was further validated using the inhibitors LY294002 and MK2206. CONCLUSIONS CDQD alleviates colitis by suppressing inflammatory macrophage activation and modulating the gut microbiota. Our research provides a novel traditional Chinese medicine strategy for the treatment of UC via enema administration.
Collapse
Affiliation(s)
- Zelong Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, China
| | - Jun Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment for Refractory Chronic Diseases, China
| | - Huishan Luo
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Linwen Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Zhaoyu Pan
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Shilong Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Cailing Zhong
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Ng Chong Shan
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Ziwen Ye
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huishi Tan
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Xiaobo Yang
- Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, China.
| | - Beiping Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, China.
| | - Chongyang Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China.
| | - Haiyan Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, China.
| |
Collapse
|
40
|
Pirolli NH, Raufman JP, Jay SM. Therapeutic Potential and Translational Challenges for Bacterial Extracellular Vesicles in Inflammatory Bowel Disease. Inflamm Bowel Dis 2025:izaf107. [PMID: 40357729 DOI: 10.1093/ibd/izaf107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Indexed: 05/15/2025]
Abstract
Despite the availability of numerous new immune-directed therapeutics, the major constituents of inflammatory bowel disease (IBD)-ulcerative colitis (UC) and Crohn's disease (CD)-continue to afflict millions worldwide, resulting in significant morbidity and long-term health risks. IBD results from a triad of immune, environmental (eg, gut microbiome), and genetic (including epigenetic) mechanisms, and therefore has been subject to a wide variety of therapeutic strategies. Among these, the administration of probiotics, particularly Gram-positive lactic acid bacteria (LAB), targeting both immune and environmental factors, has shown promising potential for efficacy in selected populations in early clinical trials. However, knowledge gaps and inconsistent efficacy currently prevent recommendations for the use of probiotics in larger IBD patient populations. The inconsistent efficacy of probiotics is likely due to variable cell viability and potency after administration, further exacerbated by IBD patient heterogeneity. Thus, an alternative to live probiotics for IBD has emerged in the form of bacterial extracellular vesicles (BEVs)-cell-secreted nanovesicles containing abundant bioactive cargo that, like live probiotics, can regulate immune and environmental factors but with fewer viability limitations and safety concerns. In this review, we summarize the work done to date establishing the potential of BEVs to provide the therapeutic benefits in IBD and discuss the hurdles BEVs must overcome to achieve clinical translation. We also consider future directions for BEV therapeutics, especially treatment potential for necrotizing enterocolitis (NEC), which shares similarities in pathophysiology with IBD.
Collapse
Affiliation(s)
- Nicholas H Pirolli
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
| | - Jean-Pierre Raufman
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
- Biomedical Laboratory Research and Development Service, Veterans Affairs Maryland Healthcare System, Baltimore, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
| | - Steven M Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
- Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
41
|
Fang Y, Min S, Wu Y, Xu F, Chen H, Li Y, Lu Y, Hu J, Zhu L, Shen H. Integration of Multi-Omics and Network Pharmacology Analysis Reveals the Mechanism of Qingchang Huashi Jianpi Bushen Formula in Repairing the Epithelial Barrier of Ulcerative Colitis. J Inflamm Res 2025; 18:6167-6189. [PMID: 40386180 PMCID: PMC12083493 DOI: 10.2147/jir.s510966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/30/2025] [Indexed: 05/20/2025] Open
Abstract
Purpose Derivation of Qingchang Huashi formula, named Qingchang Huashi Jianpi Bushen (QCHS_JPBS) formula, has shown significant therapeutic effect on patients with ulcerative colitis (UC). In this study, the potential mechanism of QCHS_JPBS formula in repairing mucosal damage was explored from the perspective of intestinal stem cell (ISCs) differentiation, and potential targets of the QCHS_JPBS formula to improve UC were predicted using network pharmacology analysis. Methods The therapeutic efficacy of QCHS_JPBS formula was evaluated in a mouse model of 2.5% dextran sulfate sodium (DSS) induced colitis. The effect of this formula on the ISC differentiation was evaluated using tissue transmission electron microscopy, immunofluorescence, and RT-qPCR. The cecal contents were subjected to 16s RNA sequencing analysis and non-target metabolomics analysis using LC-MS/MS. The fecal microbiota transplantation method verified the essential role of gut microbiota in promoting ISC differentiation and repairing mucosal damage. Results The results indicated that QCHS_JPBS formula suppressed the inflammatory response and repaired the damaged intestinal epithelial barrier in DSS-induced colitis mice. QCHS_JPBS formula promoted ISC differentiation, particularly in the direction of goblet cells. QCHS_JPBS formula restored gut dysbiosis and regulated metabolic disorders in DSS-induced colitis mice. And then, the results of fecal microbiota transplantation indicated that QCHS_JPBS formula promoted differentiation of intestinal stem cells to repair mucosal damage through gut microbiota. Finally, a total of 79 active ingredients of QCHS_JPBS formula were identified based on LC-MS analysis and EGFR, STAT3, SRC, AKT1, and HSP90AA1 were considered as potential therapeutic UC targets of QCHS_JPBS formula based on network pharmacology analysis. Conclusion The present study demonstrated that QCHS_JPBS formula promoted the differentiation of ISCs through gut microbiota to repair the damaged intestinal epithelial barrier in UC mice.
Collapse
Affiliation(s)
- Yulai Fang
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Shichen Min
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yuguang Wu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Feng Xu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Hongxin Chen
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yanan Li
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Yizhou Lu
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Jingyi Hu
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Lei Zhu
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Hong Shen
- Digestive Disease Research Institute, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| |
Collapse
|
42
|
Li S, Li H, Qi M. Exploring shared pathogenic mechanisms and biomarkers in hepatic fibrosis and inflammatory bowel disease through bioinformatics and machine learning. Front Immunol 2025; 16:1533246. [PMID: 40421012 PMCID: PMC12104268 DOI: 10.3389/fimmu.2025.1533246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/22/2025] [Indexed: 05/28/2025] Open
Abstract
Background The coexistence of hepatic fibrosis (HF) and inflammatory bowel disease (IBD) represents a significant clinical concern due to their poorly characterized shared pathogenic mechanisms. Current limitations in identifying common biomarkers for comorbid cases impede early dual diagnosis and therapeutic interventions. Methods Differentially expressed genes (DEGs) were screened, followed by Weighted Gene Co-expression Network Analysis (WGCNA) to identify disease-associated modules. The key diagnostic biomarkers were determined via a protein-protein interaction (PPI) network combined with two machine learning algorithms. The logistic regression model was subsequently developed based on these key genes. Immune cell infiltration profiling of both diseases was assessed via the CIBERSORT algorithm. The construction of genes-miRNAs and genes-TFs (Transcription Factors) regulatory networks were based on the NetworkAnalyst website. Potential drug-gene interactions were predicted utilizing the DSigDB database. The expression and distribution of these genes were validated through single-cell sequencing analysis. Results A sum of 119 up-regulated genes and 17 down-regulated genes were screened, which were enriched in categories associated with immune cell infiltration and chemotaxis, cytokine regulation, metabolic processes, enzymatic activity, and extracellular matrix deposition, based on enrichment analysis. WGCNA revealed four disease-associated gene modules. Four shared diagnostic genes for both diseases were screened, including MMP2, COL1A2, STAT1, and CXCL1. ROC curve analysis confirmed robust diagnostic performance as AUC > 0.7 for individual genes and AUC > 0.85 for combined model. M1 macrophages were significantly increased in both pathologies of diseases. A total of 462 drugs were predicted targeting these biomarkers in the DSigDB database. The four key diagnostic gene expression patterns across diverse cell subpopulations were visualized by single-cell sequencing analysis. Conclusion MMP2, COL1A2, CXCL1, and STAT1 were identified as shared biomarkers for IBD and HF, providing a molecular basis for early diagnosis and precision medicine approaches. It elucidated the similarities between HF and IBD in terms of immunity, metabolism, and fibrosis.
Collapse
Affiliation(s)
- Shangkun Li
- Clinical Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Haoyu Li
- Clinical Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Mingran Qi
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|
43
|
Zhang C, Shu Y, Li Y, Wang F, Gan J, Wang Y, Feng X, Guo M. Chinese yam (Dioscorea) polysaccharide ameliorates ulcerative colitis in mice via modulating disorders of intestinal microecology and metabolism. Int J Biol Macromol 2025; 315:144110. [PMID: 40360104 DOI: 10.1016/j.ijbiomac.2025.144110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/25/2025] [Accepted: 05/09/2025] [Indexed: 05/15/2025]
Abstract
Clinical research has demonstrated that non-starch polysaccharides from natural sources exhibit protective and therapeutic effects on ulcerative colitis (UC). In this study, a non-starch polysaccharide (CYP-A) with a molecular weight of 1.54 × 103 kDa was isolated from a speciality Chinese yam (Dioscorea) variety, the Ma yam, consisting mainly of mannose and glucose. The results indicated that CYP-A alleviated colitis symptoms induced by dextran sulfate sodium (DSS), repaired the mucus barrier, and protected the integrity of the intestinal mechanical barrier. Furthermore, CYP-A suppressed pro-inflammatory cytokine production, reduced oxidative stress, and modulated the biological barriers by facilitating the colonization of norank_f-_Muribaculaceae, Dubosella, Faecalibaculum and Enterorhabdus, while reducing Escherichia-Shigella and Bacteroides. Notably, CYP-A reshaped the metabolic pathways related to steroid hormone biosynthesis as well as phenylalanine, tyrosine, and histidine metabolism. These findings highlight the potential of CYP-A as a nutraceutical for targeting UC and improving gut health.
Collapse
Affiliation(s)
- Caixuan Zhang
- Laboratory of Dietary Component Interactions and Precision Nutrition, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Ying Shu
- Laboratory of Dietary Component Interactions and Precision Nutrition, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Yang Li
- Laboratory of Dietary Component Interactions and Precision Nutrition, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Fanyu Wang
- Laboratory of Dietary Component Interactions and Precision Nutrition, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Juntian Gan
- Laboratory of Dietary Component Interactions and Precision Nutrition, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Yiqian Wang
- Laboratory of Dietary Component Interactions and Precision Nutrition, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Xiaojuan Feng
- College of Ecology and Environment, Baotou Teacher's College, Baotou 014000, China
| | - Mingzhu Guo
- Laboratory of Dietary Component Interactions and Precision Nutrition, College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China.
| |
Collapse
|
44
|
Ma Y, Jing J, Gao Y, Yu Y, Mao J, Zhang Y, Li T. MLIF inhibits inflammation and maintains intestinal flora homeostasis in a dextran sulfate sodium (DSS)-induced colitis mouse model. Food Chem Toxicol 2025; 202:115545. [PMID: 40354872 DOI: 10.1016/j.fct.2025.115545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/14/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease primarily affecting the colon, characterized by mucosal inflammation and ulceration. Monocyte locomotion inhibitory factor (MLIF), a heat-stable pentapeptide derived from Entamoeba histolytica, has demonstrated the anti-inflammatory capacity. The aim of the current work was to test the protective effects of MLIF in a dextran sulfate sodium (DSS)-induced colitis mouse model. Our findings indicated that MLIF significantly inhibition of colitis development, including body weight, DAI score, colon length, and spleen index. MLIF slowing the progression of inflammation in the colon of mice exposed to DSS, evidenced by HE staining and mRNA expression levels of Il1b, Il6, Il18 and Il10. MLIF significantly alleviated intestinal barrier dysfunction in mice exposed to DSS, evidenced by AB-PAS staining and mRNA expression levels of Tjp1, Ocln and Muc2. Importantly, the administration of MLIF in colitis mice exerted beneficial effects on the gut microbiota, enhancing microbial diversity and abundance, and promoting the restoration of gut microbiota homeostasis. Non-targeted metabolomics results suggest that the benefits of MLIF may arise from its modulation of tryptophan metabolism pathways. In conclusion, MLIF prevention inflammation induction and preserves intestinal homeostasis against colitis induced by DSS.
Collapse
Affiliation(s)
- Yulin Ma
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, 200125, China; School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Jing Jing
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, 200125, China
| | - Yuan Gao
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yongsheng Yu
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Junqin Mao
- Department of Clinical Pharmacy, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201803, China
| | - Yuefan Zhang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Tiejun Li
- Department of Pharmacy, Punan Hospital, Pudong New District, Shanghai, 200125, China.
| |
Collapse
|
45
|
Li L, Dai L, Lin M, He S, Du H, Lin D, Wang Y, Zhang F, Tao S, Sun X, Huang X, Liu H, Wang Q, He L, Wu K, You J, Zhang M, Fu C, Tu H, Ye N, Liu J, Gao F. Colonic Submucosa Targeted Delivery of Probiotic and Rhein for Ulcerative Colitis Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2409711. [PMID: 40344311 DOI: 10.1002/advs.202409711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 04/01/2025] [Indexed: 05/11/2025]
Abstract
Ulcerative colitis (UC) is a chronic disease. A significant challenge for the management of UC is to achieve delivery of drugs to the multi-layer colonic barriers, as existing drugs are difficult to penetrate these depths. In this study, a novel drug delivery system using yeast cell wall microparticles (YPs) are developed to co-encapsulate Bacillus subtilis (BS) and Rhein (Rh) termed Rh-YBS. This system specifically targets colonic microfold cells, enabling direct delivery of BS to the colonic submucosa. Additionally, Rh enhances BS colonization in the submucosa through floral regulation. Studies indicate that Rh-YBS can effectively reach and proliferate within the submucosa in vivo. In a DSS-induced UC mouse model, Rh-YBS stimulates the CGRP-related neural pathway; BS activation in the submucosa leads to increased CGRP secretion, prompting goblet cells to secrete mucus and thereby repairing the mucosa. Furthermore, the Rh-YBS also provide a preventive benefit against UC. In summary, Rh-YBS represents an innovative drug delivery system for mucosal repair in UC treatment, activating a unique mechanism involving the CGRP-related neural pathway.
Collapse
Affiliation(s)
- Lingqiang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Linxin Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Meisi Lin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Shuang He
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Hongye Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Dasheng Lin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
- Chengdu Huashen Technology Group Co., Ltd., Chengdu, 611137, China
| | - Yanbin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Fenglian Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Sian Tao
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Xiaoluo Sun
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Xinggui Huang
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Haihui Liu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Qian Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Lingling He
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Kunhe Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Jieshu You
- College of Pharmacy, Shenzhen Technology University, Shenzhen, Guangdong, 518118, China
| | - Minyue Zhang
- Division of Hematology of Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - He Tu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Naijing Ye
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Jibin Liu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| | - Fei Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy / School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611130, China
| |
Collapse
|
46
|
Dolinger MT, Aronskyy I, Spencer EA, Pittman N, Dubinsky MC. Early intestinal ultrasound response to biologic and small molecule therapy is accurate to predict treat-to-target endoscopic outcomes in children with ulcerative colitis: results from the prospective super sonic-UC study. J Crohns Colitis 2025; 19:jjaf075. [PMID: 40312920 DOI: 10.1093/ecco-jcc/jjaf075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND AND AIMS Stride-II recommends monitoring early biomarker targets to achieve treat-to-target (T2T) endoscopic remission (ER) in ulcerative colitis (UC). Predictive capabilities of intestinal ultrasound (IUS) for ER remain unknown. We evaluated IUS response to predict ER in children with UC. METHODS Prospective longitudinal cohort study of children with UC (Mayo endoscopic score [MES ≥2) starting advanced therapy undergoing IUS (including Milan Ultrasound Criteria [MUC], Civitelli Ulcerative Colitis Index, and International Bowel Ultrasound Group Segmental Activity Score), fecal calprotectin (FC), C-reactive protein (CRP), and Pediatric Ulcerative Colitis Activity Index at baseline, week 8, and T2T. Primary outcome was accuracy to predict T2T ER (MES = 0) for change in bowel wall thickness (BWT) from baseline to week 8, and absolute BWT at week 8. Logistic regression with forward selection determined an optimal prediction model for endoscopic outcomes. RESULTS Of 42 children, 21 (50%) achieved ER. Week 8 BWT ≤ 2.7 mm (OR 6.4 [95% CI, 1.8-27.0], P = .007), MUC < 6.0 (OR 5.7 [95% CI, 1.5-25.3], P = .015), and FC ≤ 177 (OR 4.5 [95% CI, 1.1-23.6], P = .049) were associated with ER. CONCLUSIONS Combining noninvasive biomarkers of BWT and the MUC on IUS, and FC, is a feasible tight control monitoring strategy in children with UC that is predictive of endoscopic outcomes. Larger, multicenter validation studies are needed to understand how an IUS and FC monitoring strategy may improve outcomes in children with UC.
Collapse
Affiliation(s)
- Michael Todd Dolinger
- Division of Pediatric Gastroenterology, Susan and Leonard Feinstein Inflammatory Bowel Disease Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Illya Aronskyy
- Division of Pediatric Gastroenterology, Susan and Leonard Feinstein Inflammatory Bowel Disease Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Elizabeth A Spencer
- Division of Pediatric Gastroenterology, Susan and Leonard Feinstein Inflammatory Bowel Disease Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nanci Pittman
- Division of Pediatric Gastroenterology, Susan and Leonard Feinstein Inflammatory Bowel Disease Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Marla C Dubinsky
- Division of Pediatric Gastroenterology, Susan and Leonard Feinstein Inflammatory Bowel Disease Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
47
|
Salomon B, Sudhakar P, Bergemalm D, Andersson E, Grännö O, Carlson M, Hedin CRH, Söderholm JD, Öhman L, Lindqvist CM, Kruse R, Repsilber D, Verstockt B, Vermeire S, Halfvarson J. Characterization of Inflammatory Bowel Disease Heterogeneity Using Serum Proteomics: A Multicenter Study. J Crohns Colitis 2025; 19:jjae169. [PMID: 39495605 PMCID: PMC12063088 DOI: 10.1093/ecco-jcc/jjae169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/08/2024] [Accepted: 11/03/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Recent genetic and transcriptomic data highlight the need for improved molecular characterization of inflammatory bowel disease (IBD). Proteomics may advance the delineation of IBD phenotypes since it accounts for post-transcriptional modifications. AIMS We aimed to assess the IBD spectrum based on inflammatory serum proteins and identify discriminative patterns of underlying biological subtypes across multiple European cohorts. METHODS Using proximity extension methodology, we measured 86 inflammation-related serum proteins in 1551 IBD patients and 312 healthy controls (HC). We screened for proteins exhibiting significantly different levels among IBD subtypes and between IBD and HC. Classification models for differentiating between Crohn's disease (CD) and ulcerative colitis (UC) were employed to explore the IBD spectrum based on estimated probability scores. RESULTS Levels of multiple proteins, such as interleukin-17A, matrix metalloproteinase-10, and fibroblast growth factor-19, differed (fold-change >1.2; false discovery rate <0.05) between ileal versus colonic IBD. Using multivariable models, a protein signature reflecting the IBD spectrum was identified, positioning colonic CD between UC and ileal CD, which were at opposite ends of the spectrum. Based on area under the curve (AUC) estimates, classification models more accurately differentiated UC from ileal CD (median AUCs > 0.73) than colonic CD (median AUCs < 0.62). Models differentiating colonic CD from ileal CD demonstrated intermediate performance (median AUCs: 0.67-0.69). CONCLUSIONS Our findings in serum proteins support the presence of a continuous IBD spectrum rather than a clear separation of CD and UC. Within the spectrum, disease location may reflect a more similar disease than CD versus UC, as colonic CD resembled UC more closely than ileal CD.
Collapse
Affiliation(s)
- Benita Salomon
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Padhmanand Sudhakar
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, India
| | - Daniel Bergemalm
- Faculty of Medicine and Health, Department of Gastroenterology, Örebro University, Örebro, Sweden
| | - Erik Andersson
- Faculty of Medicine and Health, Department of Gastroenterology, Örebro University, Örebro, Sweden
| | - Olle Grännö
- Faculty of Medicine and Health, Department of Laboratory Medicine, Clinical Microbiology, Örebro University, Örebro, Sweden
| | - Marie Carlson
- Department of Medical Sciences, Gastroenterology Research Group, Uppsala University, Uppsala, Sweden
| | - Charlotte R H Hedin
- Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
- Department of Gastroenterology, Dermatovenereology, and Rheumatology, Centre for Digestive Health, Karolinska University Hospital, Stockholm, Sweden
| | - Johan D Söderholm
- Department of Surgery, Linköping University, Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lena Öhman
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Carl Mårten Lindqvist
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Robert Kruse
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
- Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
- Faculty of Medicine and Health, Department of Clinical Research Laboratory, Örebro University, Örebro, Sweden
| | - Dirk Repsilber
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Bram Verstockt
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, KU Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic Diseases and Metabolism, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Jonas Halfvarson
- Faculty of Medicine and Health, Department of Gastroenterology, Örebro University, Örebro, Sweden
| |
Collapse
|
48
|
Pertsinidou E, Salomon B, Bergemalm D, Salihovic S, Hedin CRH, Ling Lundström M, Keita ÅV, Magnusson MK, Eriksson C, Bengtson MB, Grännö O, Aabrekk TB, Movérare R, Rydell N, Ekoff H, Rönnelid J, D’Amato M, Detlie TE, Huppertz-Hauss G, Opheim R, Ricanek P, Kristensen VA, Öhman L, Söderholm JD, Kruse R, Lindqvist CM, Carlson M, Repsilber D, Høivik ML, Halfvarson J. Anti-integrin αvβ6 IgG antibody as a diagnostic and prognostic marker in ulcerative colitis: A cross-sectional and longitudinal study defining a specific disease phenotype. J Crohns Colitis 2025; 19:jjaf062. [PMID: 40251889 PMCID: PMC12086997 DOI: 10.1093/ecco-jcc/jjaf062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Indexed: 04/21/2025]
Abstract
BACKGROUND AND AIMS The diagnostic and prognostic properties of anti-integrin αvβ6 immunoglobulin G (IgG) autoantibodies in ulcerative colitis (UC) are poorly understood. We aimed to assess the diagnostic performance of anti-integrin αvβ6 autoantibodies and examine their association with disease outcomes. METHODS Serum samples from a Swedish inception cohort of patients with suspected inflammatory bowel disease (IBD, n = 473) were analyzed using an in-house fluorescence enzyme immunoassay based on EliA technology. Findings were validated in a Norwegian population-based inception cohort (n = 570). Diagnostic performance was assessed by calculating the area under the curve (AUC) with 95% confidence intervals and determining sensitivity and specificity. Reclassification was evaluated using the net reclassification index. RESULTS In the discovery cohort, patients with UC, IBD-unclassified, or colonic Crohn's disease exhibited higher median autoantibody levels compared to symptomatic and healthy controls. In the validation cohort, the autoantibody demonstrated 79% sensitivity and 94% specificity for UC vs symptomatic controls at a cut-off of 400 UA/l. Its diagnostic performance (AUC = 0.92, 95% CI, 0.89-0.95) was superior to hs-CRP (AUC = 0.65, 95% CI, 0.60-0.70, P < .001) and faecal calprotectin (fcalpro) (AUC = 0.88, 95% CI, 0.84-0.92, P = .09). Combining the autoantibody with fcalpro further improved diagnostic accuracy (AUC = 0.97, 95% CI, 0.95-0.98) and patient reclassification (P < .001). Autoantibody positivity was associated with a severe phenotype of UC, characterised by increased inflammatory activity and higher IL-17A and granzyme B levels. Higher autoantibody levels were linked to an aggressive disease course, remaining stable in aggressive UC but decreasing in indolent disease (P = .003). CONCLUSIONS Anti-integrin αvβ6 is a reliable diagnostic and prognostic marker for UC, with potential clinical implementation.
Collapse
Affiliation(s)
- Eleftheria Pertsinidou
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
- Thermo Fisher Scientific, Uppsala, Sweden
| | - Benita Salomon
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Daniel Bergemalm
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Samira Salihovic
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Charlotte R H Hedin
- Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Gastroenterology, Dermatovenereology and Rheumatology, Centre for Digestive Health, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Ling Lundström
- Department of Medical Sciences: Gastroenterology and Hepatology, Uppsala University, Uppsala, Sweden
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Maria K Magnusson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carl Eriksson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - May-Bente Bengtson
- Department of Gastroenterology, Vestfold Hospital Trust, Tønsberg, Norway
| | - Olle Grännö
- Department of Laboratory Medicine, Clinical Microbiology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Tone B Aabrekk
- Department of Gastroenterology, Vestfold Hospital Trust, Tønsberg, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Robert Movérare
- Thermo Fisher Scientific, Uppsala, Sweden
- Department of Medical Sciences: Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | | | - Helena Ekoff
- Thermo Fisher Scientific, Uppsala, Sweden
- Department of Medical Sciences: Gastroenterology and Hepatology, Uppsala University, Uppsala, Sweden
| | - Johan Rönnelid
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Mauro D’Amato
- Gastrointestinal Genetics Lab, CIC bioGUNE—BRTA, Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Department of Medicine and Surgery, LUM University, Casamassima, Italy
| | - Trond E Detlie
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | | | - Randi Opheim
- Institute of Health and Society, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Oslo University Hospital, Oslo, Norway
| | - Petr Ricanek
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
- Department of Gastroenterology, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Vendel A Kristensen
- Department of Gastroenterology, Oslo University Hospital, Oslo, Norway
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Lena Öhman
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johan D Söderholm
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Robert Kruse
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Carl M Lindqvist
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Marie Carlson
- Department of Medical Sciences: Gastroenterology and Hepatology, Uppsala University, Uppsala, Sweden
| | - Dirk Repsilber
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Marte L Høivik
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Oslo University Hospital, Oslo, Norway
| | - Jonas Halfvarson
- Department of Gastroenterology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
Collaborators
Sven Almer, Hans Strid, Henrik Hjortswang, Francesca Bresso, Johann Hreinsson, André Blomberg, Adam Carstens,
Collapse
|
49
|
Vermeire S, Nitcheu J, Gineste P, Flatres A, Santo J, Scherrer D, Peyrin-Biroulet L, Dulai PS, Danese S, Dubinsky M, Tilg H, Siegmund B, Hisamatsu T, Shan K, Rabbat CJ, Sands BE. Obefazimod in patients with moderate-to-severely active ulcerative colitis: efficacy and safety analysis from the 96-week open-label maintenance phase 2b study. J Crohns Colitis 2025; 19:jjaf074. [PMID: 40417999 PMCID: PMC12124117 DOI: 10.1093/ecco-jcc/jjaf074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND AND AIMS Obefazimod is an oral small molecule that selectively enhances the expression of a single micro-RNA (miRNA), miR-124. Obefazimod has demonstrated safety and efficacy in patients with moderate-to-severely active ulcerative colitis (UC) in a phase 2b induction trial. This analysis presents the 2-year outcome data of the open-label maintenance (OLM) study. METHODS Patients received placebo or obefazimod 25, 50, or 100 mg once-daily (od) during the induction trial and, irrespective of their clinical response, could enter the 96-week OLM study with obefazimod 50 mg od. Safety was monitored through monthly visits in the first year and quarterly visits in the second year. Efficacy was assessed at weeks 48 and 96 using nonresponder imputation (NRI) for missing data. RESULTS Of 222 eligible patients, 217 were enrolled and 164 (75.6%) completed week 96 of the OLM study. Clinical response was achieved at weeks 48 and 96 in 177 (81.6%) and 158 (72.8%) patients and clinical remission in 119 (54.8%) and 114 (52.5%) of patients. A total of 133 (61.3%) and 128 (59.0%) patients showed endoscopic improvement, and 72 (33.2%) and 78 (35.9%) endoscopic remission. In total, 148/217 patients (68.2%) reported at least 1 treatment-emergent adverse event (TEAE). The most frequent TEAEs were COVID-19 (14.3%), headache (11.5%), UC (7.8%), and nasopharyngitis (6.9%). No new safety risks emerged over 96 weeks. CONCLUSIONS The 96-week OLM study supports the long-term efficacy and favorable safety profile of obefazimod 50 mg od. A phase 3 program with obefazimod in patients with moderate-to-severe UC is ongoing. TRIAL REGISTRATION NAME/NUMBER A phase 2b, open-label, efficacy and safety study of ABX464 as maintenance therapy in patients with moderate-to-severe UC. NCT04023396.
Collapse
Affiliation(s)
- Severine Vermeire
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | | | | | | | | | | | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, INFINY Institute, INSERM NGERE, CHRU Nancy, Vandœuvre-lès-Nancy, France
| | - Parambir S Dulai
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Marla Dubinsky
- Pediatric GI and Nutrition, Mount Sinai Kravis Children’s Hospital, New York, NY, United States
| | - Herbert Tilg
- Department of Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Britta Siegmund
- Department of Gastroenterology, Infectiology and Rheumatology, Charité-Universitaetsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tadakazu Hisamatsu
- Department of Gastroenterology and Hepatology, Kyorin University Hospital, Tokyo, Japan
| | | | | | - Bruce E Sands
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
50
|
Ji ZH, Xie WY, Wu HY, Yuan B. Coix Seed Polysaccharide Mitigates Ulcerative Colitis in Mice through the Modulation of Gut Microbiota and Improvement of Intestinal Metabolism Balance. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11067-11079. [PMID: 40274530 DOI: 10.1021/acs.jafc.5c02458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease with a rising incidence globally, whereas existing treatments exhibit significant limitations. Coix seed polysaccharide (CSP), a component of traditional Chinese medicine known for its immunomodulatory and antioxidant properties, has not been thoroughly investigated for its role in UC. In this study, CSP was prepared via water extraction and ethanol precipitation, and its protective effects and mechanisms were evaluated using a dextran sulfate sodium salt (DSS)-induced UC mouse model. The results demonstrated that CSP significantly ameliorated DSS-induced UC symptoms, including weight loss, an elevated Disease Activity Index, colon shortening, increased levels of inflammatory cytokines, and intestinal barrier damage. Moreover, CSP reshaped the DSS-induced gut microbiota dysbiosis by increasing gut microbial diversity and regulating the abundance of specific genera, such as increasing Anaerotruncus. Metabolomic analysis revealed that CSP significantly modulated the levels of 116 metabolites, particularly enhancing the beneficial metabolite 3-hydroxybutyrate. Importantly, the preventive effect of CSP on UC was dependent on the gut microbiota and could be transferred via fecal microbiota transplantation. This study demonstrates that CSP, a microecology-regulating polysaccharide, effectively modulates gut microbiota and alleviates symptoms of UC. These findings support the potential of CSP as a dietary supplement for UC prevention and underscore its value in the development of medicinal foods and functional food applications.
Collapse
Affiliation(s)
- Zhong-Hao Ji
- Laboratory of Brain Diseases and Cognitive Behavior, Department of Basic Medicine, Changzhi Medical College, Changzhi 046000, Shanxi, China
| | - Wen-Yin Xie
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, Jilin, China
| | - Hong-Yu Wu
- Beihua University School of Basic Medical Science, Beihua University, Jilin 132011, Jilin, China
| | - Bao Yuan
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, Jilin, China
| |
Collapse
|