1
|
Park SH, Won SY, Kim YC. Identification of a disease-associated germline mutation (A64T) of the ring finger protein 186 gene ( RNF186) in Korean patients with ulcerative colitis. Genes Dis 2025; 12:101438. [PMID: 40230421 PMCID: PMC11995066 DOI: 10.1016/j.gendis.2024.101438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/25/2024] [Indexed: 04/16/2025] Open
Affiliation(s)
| | | | - Yong-Chan Kim
- Department of Biological Sciences, Andong National University, Andong 36729, Republic of Korea
| |
Collapse
|
2
|
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
|
3
|
Yuan J, Su J, Zhong S, Yuan X, Zhu J, Lu S, Zhang D, Li G, Xue H, Yan M, Yue L, Zhang T. Dictamnine alleviates DSS-induced colitis mice by inhibiting ferroptosis of enterocytes via activating Nrf2-Gpx4 signaling pathway. Eur J Pharmacol 2025; 997:177464. [PMID: 40049578 DOI: 10.1016/j.ejphar.2025.177464] [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: 11/11/2024] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 03/12/2025]
Abstract
BACKGROUND The treatment of ulcerative colitis (UC) remains a huge challenge worldwide. Dictamnine is a natural product derived from Dictamnus dasycarpus Turcz. root bark and possesses multi-pharmacological properties, including anti-inflammation effects. However, its protective effect on UC and its underlying mechanisms are unknown. PURPOSE Here we explored the protective effect and underlying mechanism of dictamnine against dextran sulfate sodium (DSS)-induced colitis in mice. METHODS The experimental colitis was established by adding 3% DSS on drinking water of mice and the effects of dictamnine (10, 20, 40 mg/kg, p.o, once a day by 10 days) in colon tissues was analyzed. NCM460 cell was induced by RSL3 to detect the effect of dictamnine on ferroptosis and the underlying mechanism. Pathological damage was determined by H&E. Indicators related to intestinal permeability were detected by FITC and immunofluorescence. Cytokines levels (TNF-α、IL-1β and IL-6), antioxidant enzymes activities (MDA and GSH), the level of Fe2+ Cytokines levels and Gpx4 activity were detected by ELISA. Finally, the activation of nuclear factor erythroid 2-like 2 (Nrf2) was detected to explore the mechanism. RESULTS The results indicated that dictamnine significantly attenuated DSS-induced colon pathological damage, intestinal barrier, cytokines levels, and increased the antioxidant enzymes activities. Moreover, dictamnine attenuated ferroptosis in DSS-induced colon injury and upregulated Gpx4 expression in DSS-induced mice. Mechanistic experiments revealed that dictamnine activated Nrf2 in mice. CONCLUSION Taken together, this study evaluates that dictamnine alleviates DSS-induced colitis mice by inhibiting ferroptosis of enterocytes and its protective effects are associated with activating the Nrf2-Gpx4 signaling pathway.
Collapse
Affiliation(s)
- Jin Yuan
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China
| | - Junwei Su
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shaowen Zhong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; Adverse Drug Reaction Monitoring Center, Zhongshan Food and Drug Inspection Institute, Zhongshan, Guangdong, China
| | - Xin Yuan
- Department of Pharmacy, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianping Zhu
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China
| | - Shuangxi Lu
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China
| | - Di Zhang
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China
| | - Guiling Li
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China
| | - Hanyu Xue
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China.
| | - Min Yan
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China.
| | - Lei Yue
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China.
| | - Tianwu Zhang
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China.
| |
Collapse
|
4
|
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
|
5
|
Tanaka M, Toyonaga T, Nakagawa F, Iwamoto T, Hasegawa Y, Komatsu A, Sumiyoshi N, Shibuya N, Minemura A, Ariyoshi T, Matsumoto A, Oka K, Shimoda M, Saruta M. Dietary 3-aminobenzoic acid enhances intestinal barrier integrity and attenuates experimental colitis. Am J Physiol Gastrointest Liver Physiol 2025; 328:G801-G810. [PMID: 40338094 DOI: 10.1152/ajpgi.00406.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/21/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
Disruption of intestinal epithelial integrity and increased permeability is central to the pathogenesis of ulcerative colitis (UC). In this study, we identified 3-aminobenzoic acid (3-ABA), a dietary component abundant in azuki beans, soybeans, and chickpeas as a regulator of epithelial permeability and inflammation in the colon. Screening 119 gut microbial metabolites revealed the ability of 4-ABA, a structural isomer of 3-ABA, to enhance barrier function in Caco2 cells. Further analysis of structural isomers identified 3-ABA as the most effective, significantly increasing transepithelial electrical resistance and reducing epithelial permeability. Using liquid chromatography-mass spectrometry, 3-ABA was detected in dietary beans and human fecal samples. Fecal 3-ABA levels were significantly lower in patients with UC compared with healthy individuals. Metagenomic and functional prediction analyses revealed dysbiosis in patients with UC, characterized by an enrichment of bacterial genes involved in ABA degradation. Gene expression analysis of 3-ABA-stimulated Caco2 cells demonstrated upregulation of tight junction molecules, such as CLDN1 and TJP1, enhancing epithelial barrier integrity. In a dextran sodium sulfate-induced colitis mouse model, rectal 3-ABA administration ameliorated colitis by enhancing epithelial barrier function and reducing inflammation. These findings highlight 3-ABA's potential as a dietary therapeutic agent for UC, offering a novel strategy to enhance intestinal integrity and mitigate inflammation.NEW & NOTEWORTHY Increased intestinal epithelial permeability is central to the pathogenesis of ulcerative colitis (UC). 3-Aminobenzoic acid (3-ABA), a dietary component abundant in beans, decreased epithelial permeability and attenuated colonic inflammation in a mouse experimental colitis model. Reduced fecal 3-ABA levels in patients with UC were associated with dysbiosis-driven accelerated degradation. These findings highlight the therapeutic potential of 3-ABA in UC by targeting colonic epithelium.
Collapse
Affiliation(s)
- Miho Tanaka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takahiko Toyonaga
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Fumiyuki Nakagawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takeo Iwamoto
- Core Research Facilities, The Jikei University School of Medicine, Tokyo, Japan
| | - Yudai Hasegawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Akira Komatsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Natsuki Sumiyoshi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Naoki Shibuya
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Ayaka Minemura
- Research Department, R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Tadashi Ariyoshi
- Research Department, R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Asami Matsumoto
- Research Department, R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Kentaro Oka
- Research Department, R&D Division, Miyarisan Pharmaceutical Co., Ltd., Saitama, Japan
| | - Masayuki Shimoda
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Masayuki Saruta
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Côco LZ, de Souza Belisário E, Vasquez EC, Pereira TMC, Aires R, Campagnaro BP. Probiotics: a promising future in the treatment of ulcerative colitis? Pharmacol Rep 2025; 77:645-657. [PMID: 40214948 DOI: 10.1007/s43440-025-00724-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 05/13/2025]
Abstract
Ulcerative colitis is an idiopathic and chronic inflammatory bowel disease, characterized by inflammation of the mucosa of the colon and rectum. Clinical manifestations commonly include abdominal pain, diarrhea (with or without hematochezia), and weight loss. The pathogenesis of ulcerative colitis is multifactorial, involving a combination of genetic predispositions and lifestyle factors. High consumption of processed food, sedentary habits, alcohol intake, and stress are among the lifestyle factors implicated in disease onset and progression. Current treatment strategies focus on managing symptoms and inducing remission, however, the chronic nature of the disease, along with the adverse effects of conventional therapies, often compromises patient's quality of life. Therefore, exploring alternative therapies that can prolong remission and reduce symptom burden is important. Experimental evidence suggests that probiotics may extend remission duration in ulcerative colitis. Moreover, probiotics exhibit efficacy in amelioration clinical symptoms by reducing inflammation markers, preserving, and restoring intestinal epithelial. This review explores the advantages of the administration of probiotics in the treatment of ulcerative colitis, elucidating their mechanism of action.
Collapse
Affiliation(s)
- Larissa Zambom Côco
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Eduarda de Souza Belisário
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Elisardo Corral Vasquez
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Thiago Melo Costa Pereira
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Rafaela Aires
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Bianca Prandi Campagnaro
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil.
| |
Collapse
|
7
|
Wang H, Yang Y, Li T, Chang S, Zhu Y, Liu C. Drinking Water Temperature Impacts the Pathogenesis of DSS-Induced Ulcerative Colitis by Regulating Intestinal Barrier Function and Remodeling the Gut Microbiota Composition. FASEB J 2025; 39:e70645. [PMID: 40377203 DOI: 10.1096/fj.202500062r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/18/2025] [Accepted: 05/07/2025] [Indexed: 05/18/2025]
Abstract
Environmental factors, including poor dietary habits and unhealthy drinking patterns, contribute to ulcerative colitis (UC). While the relationship between diet-related malnutrition and UC has been extensively explored, the impact of drinking water temperature remains largely overlooked, prompting us to investigate its influence on UC pathogenesis and explore the underlying mechanisms. In the present study, we observed that, unlike external thermal and cold therapy, varying drinking water temperatures transiently altered the internal body temperature of the digestive tract. Specifically, chronic drinking of 0°C water had significant anti-inflammatory effects and preserved the integrity of the mucosal barrier in a colitis mouse model. Mechanistically, this temperature spectrum changed the composition of the gut microbiota from inflammation-prone (25°C drinking water) to a resting pattern similar to that of the negative control. Specifically, the abundances of Blautia and Parasutterella, two beneficial genera, were strongly increased in response to 0°C water, accompanied by elevated levels of short-chain fatty acids. In contrast, drinking 40°C water had opposite effects on all the examined parameters and generally aggravated the development of colitis. This study is the first to demonstrate how modifying the temperature of habitual drinking water can modulate colitis progression, providing a novel and noninvasive approach to UC management. Specifically, chronic consumption of 0°C water alleviated the severity of colitis, whereas 40°C water aggravated the disease. Therefore, by focusing on commonly consumed drinking water temperatures, our findings suggest that this simple intervention could be a safe, convenient, and effective therapeutic strategy.
Collapse
Affiliation(s)
- Huiting Wang
- Department of Endocrinology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Provincial University Key Laboratory of Drug Discovery for Metabolic Inflammatory Diseases, China Pharmaceutical University, Nanjing, China
| | - Yiheng Yang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianyu Li
- Department of Endocrinology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Provincial University Key Laboratory of Drug Discovery for Metabolic Inflammatory Diseases, China Pharmaceutical University, Nanjing, China
| | - Shengyu Chang
- Department of Endocrinology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Provincial University Key Laboratory of Drug Discovery for Metabolic Inflammatory Diseases, China Pharmaceutical University, Nanjing, China
| | - Yao Zhu
- Department of Endocrinology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Provincial University Key Laboratory of Drug Discovery for Metabolic Inflammatory Diseases, China Pharmaceutical University, Nanjing, China
| | - Chang Liu
- Department of Endocrinology, Nanjing Drum Tower Hospital, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Jiangsu Provincial University Key Laboratory of Drug Discovery for Metabolic Inflammatory Diseases, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
8
|
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
|
9
|
Takagi T, Uchiyama K, Asaeda K, Murakami E, Inoue K, Mizushima K, Hirai Y, Naito Y, Itoh Y. Association of Vascular Cell Adhesion Molecule-1 Expression in Colonic Mucosa With Mucosal Inflammation and Subsequent Relapse in Patients With Ulcerative Colitis. J Gastroenterol Hepatol 2025. [PMID: 40411296 DOI: 10.1111/jgh.17000] [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: 11/22/2024] [Revised: 04/04/2025] [Accepted: 04/26/2025] [Indexed: 05/26/2025]
Abstract
BACKGROUND AND AIM The association between vascular cell adhesion molecule-1 (VCAM-1) expression and intestinal mucosal inflammation and between VCAM-1 expression and the clinical course in patients with ulcerative colitis (UC) remains unclear. Therefore, we investigated not only the association between mucosal VCAM-1 expression and mucosal inflammation but also its association with subsequent relapse in UC patients with clinical remission. METHODS Fifty-eight patients with UC in clinical remission and 16 patients in clinical active who visited Kyoto Prefectural University of Medicine for a 2-year follow-up period were included. VCAM-1 expression was compared between patients who subsequently relapsed and those who remained in remission, and it was examined in relation to endoscopic findings, histological activity, and cytokine expression. We also investigated the expression of mucosal addressin cell adhesion molecule-1 (MAdCAM-1). RESULTS VCAM-1 was associated with clinical disease activity and endoscopic severity and was significantly elevated in histologically active mucosa compared with inactive mucosa. VCAM-1 expression co-localized with MAdCAM-1 in the mucosal and submucosal microvessels of the colon and was significantly higher in the relapse group than in the remission group. Similar results were observed for MAdCAM-1 expression. VCAM-1 expression levels were also closely correlated with those of several other cytokines. CONCLUSIONS VCAM-1 expression in the colonic mucosa of patients with UC is associated with mucosal inflammation and subsequent relapse. These results suggest that VCAM-1 may serve as a marker for relapse and therapeutic effectiveness in UC, and that treatment targeting α4 integrin is efficient and rational.
Collapse
Affiliation(s)
- Tomohisa Takagi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department for Medical Innovation and Translational Medical Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuhiko Uchiyama
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kohei Asaeda
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Eiki Murakami
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ken Inoue
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Katsura Mizushima
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuko Hirai
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
10
|
Wahabi S, Selmi S, Gharbi K, Ayari A, Yahmdi F, Sebai H. Protective effects of Arbutus unedo extract on acetic acid-induced colitis in rats: histological, biochemical, and antioxidant assessments. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025. [PMID: 40395169 DOI: 10.1002/jsfa.14317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND This study aimed to analyze the advantageous effects of the decoction aqueous extract of Arbutus unedo fruits (AUFDE) against ulcerative colitis (UC) caused by acetic acid (AA) in rats, along with the processes involved in this protective effect. Adult male Wistar rats were divided into six groups: control (H2O), AA, AA combined with various doses of AUFDE (75, 150, and 300 mg kg-1 b.w., p.o.), and AA combined with sulfasalazine (SULF) (100 mg kg-1 b.w., p.o.) during 7 days. All rats underwent overnight fasting, followed by the induction of UC via rectal infusion of AA (300 mg kg-1 b.w.) at a concentration of 3% (v/v), administered at a volume of 5 mL kg-1 b.w. for 30 s. The colon was resected and subjected to macroscopic examination to assess ulcerated areas and the ulcer index. RESULTS The in vivo assay's findings revealed that AUFDE pretreatment attenuates histological changes and significantly reduces colonic mucosa lesions caused by AA. Also, AUFDE reduced the oxidative state that AA caused in the colonic mucosa, as indicated by elevated malondialdehyde levels and the reduction of both enzymatic antioxidants such as superoxide dismutase, catalase, and glutathione peroxidase, and non-enzymatic antioxidants such as thiol groups. Furthermore, AUFDE pretreatment controlled the dysregulation of all intracellular mediators and significantly decreased inflammatory indicators like C-reactive protein and alkaline phosphatase levels, whereas AA intoxication raised iron and calcium levels in plasma. According to our findings, AUFDE may have protected rats from AA-induced colitis. © 2025 Society of Chemical Industry.
Collapse
Affiliation(s)
- Soumaya Wahabi
- LR: Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Beja, Beja, Tunisia
| | - Slimen Selmi
- LR: Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Beja, Beja, Tunisia
| | - Khaoula Gharbi
- LR: Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Beja, Beja, Tunisia
| | - Ala Ayari
- LR: Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Beja, Beja, Tunisia
| | - Fethi Yahmdi
- Clinical Biology Laboratory, Nefza Hospital, Beja, Tunisia
| | - Hichem Sebai
- LR: Functional Physiology and Valorization of Bio-Resources, University of Jendouba, Higher Institute of Biotechnology of Beja, Beja, Tunisia
| |
Collapse
|
11
|
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
|
12
|
Kannan PR, Chen L, Lv Y, Zhao R, Hu Y, Iqbal MZ, Han Q, Kong X, Li Y. Smart Silk-Based In Situ Sol-Gel Modulates Rectal Microenvironment for Effective Ulcerative Colitis Alleviation. Adv Healthc Mater 2025:e2500984. [PMID: 40394946 DOI: 10.1002/adhm.202500984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 05/06/2025] [Indexed: 05/22/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease, with untreated cases often progressing to colorectal cancer. Current treatments aim to induce inflammatory remission but often neglect the surrounding microenvironment, which significantly impairs mucosal healing and contributes to treatment failures. This study presents a novel silk fibroin-based fucoidan (SFU) in situ rectal gel, with sol-gel transition confirmed through rheological analysis under physiological pH and temperature conditions. The SFU gel exhibits strong antioxidant activity, achieving a DPPH radical scavenging rate of 73.3 ± 1.52%. The gel efficiently reduces reactive oxygen species (ROS) and nitric oxide (NO) production, demonstrating its reliable antioxidant effects. In a DSS-induced UC mouse model, SFU effectively alleviates colitis symptoms, including weight loss and disease activity index (DAI) reduction, with improved stool consistency and reduced rectal bleeding. Moreover, SFU therapy reprograms macrophages from proinflammatory M1 to anti-inflammatory M2 phenotypes, significantly lowering IL-6 and TNF-α levels, suggesting anti-inflammatory properties. Furthermore, SFU increased tight junction proteins Occludin-1 and ZO-1, indicating gut mucosal barrier integrity. SFU treatment restores goblet cells and mucin production while preventing fibrosis, demonstrating its potential as a natural therapy for UC treatment.
Collapse
Affiliation(s)
- Perumal Ramesh Kannan
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Liuting Chen
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Yudie Lv
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Ruibo Zhao
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Yeting Hu
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, P. R. China
| | - M Zubair Iqbal
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Qianqian Han
- National Institutes for Food and Drug Control, Beijing, 100050, P. R. China
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Yao Li
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
- Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| |
Collapse
|
13
|
Wang B, Tao M, Zhu W, Li J, Hai Z. In Situ Self-Assembled Probe for Antioxidant and Anti-Inflammatory Therapy of Inflammatory Bowel Disease. ACS APPLIED BIO MATERIALS 2025; 8:4285-4293. [PMID: 40299753 DOI: 10.1021/acsabm.5c00394] [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: 05/01/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic and relapsing disease of the gastrointestinal tract. At present, antioxidant therapy is a promising strategy for IBD treatment. Since low-molecular-weight antioxidants (e.g., 2,2,6,6-tetramethylpiperidin-N-oxyl (TEMPO)) have a short in vivo half-life and inadequate cellular uptake, researchers have focused on loading antioxidants into nanostructures for improving their antioxidant and anti-inflammatory activities. As we know, in situ self-assembly with the formation of nanostructures under intracellular specific stimuli is a convenient delivery strategy to enhance the accumulation and retention of molecules at target sites in vivo. Herein, we developed an in situ self-assembled TEMPO probe TPP-FFYp-O to improve the antioxidant and anti-inflammatory effects of TEMPO in IBD. Compared to the control probe TPP-O without a self-assembly moiety, TPP-FFYp-O could successfully self-assemble into nanoparticles (NPs) under alkaline phosphatase (ALP)-guided dephosphorylation, with significantly enhanced antioxidant capacity in vitro. Cell experiments confirmed that intracellular formation of NPs by TPP-FFYp-O could improve the antioxidant and anti-inflammatory abilities of TEMPO and alleviate cellular damage. Moreover, TPP-FFYp-O exhibited good biocompatibility in vivo and significantly relieved pathological injury and inflammatory factors in the colon tissues of an IBD model compared to TPP-O. We envision that the in situ self-assembly platform can be used to load various active molecules for more applications in the future.
Collapse
Affiliation(s)
- Beibei Wang
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Menglin Tao
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Wujuan Zhu
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Jin Li
- Department of Gastroenterology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China
| | - Zijuan Hai
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| |
Collapse
|
14
|
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
|
15
|
Zhao Y, Tian X, Yan Y, Tian S, Liu D, Xu J. Lithospermic acid alleviates oxidative stress and inflammation in DSS-induced colitis through Nrf2. Eur J Pharmacol 2025; 995:177390. [PMID: 39956261 DOI: 10.1016/j.ejphar.2025.177390] [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: 11/17/2024] [Revised: 01/26/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
Lithospermic acid (LA), a plant-derived polycyclic phenolic carboxylic acid, is known for its strong anti-inflammatory and antioxidant effects. However, its effects have not yet been studied in ulcerative colitis (UC). This study aimed to assess the protective effects of LA in UC and investigate its potential mechanisms of action. Our findings indicated that LA effectively mitigated oxidative stress in mice with colitis by increasing the production of antioxidant enzymes, such as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-PX), while reducing the levels of malondialdehyde (MDA) and reactive oxygen species (ROS) (p < 0.05 for all). In NCM460 cells, LA inhibited the Lipopolysaccharide (LPS)-induced increase in ROS and preserved the mitochondrial membrane potential. In vitro and in vivo experiments confirmed that LA decreased the production of inflammatory markers (p < 0.05). Additionally, LA upregulated intestinal mucosal proteins, contributing to mucosal barrier repair. Mechanistically, LA activated the nuclear factor erythroid 2-related factor 2 (Nrf2) signalling pathway, increasing the expression of Nrf2, heme oxygenase-1 (HO-1), and NAD(P)H quinone oxidoreductase 1 (NQO1) while inhibiting nuclear factor kappa B (NF-κB) phosphorylation (p < 0.05). Notably, the inhibition of Nrf2 reversed the protective effects of LA against colitis. Molecular docking analyses support a strong interaction between Nrf2 and LA. LA mitigates colitis-related inflammation and oxidative stress mainly by activating the Nrf2 signalling pathway. These findings support the potential development of LA as a novel therapeutic agent for UC.
Collapse
Affiliation(s)
- Yulei Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China; Department of Gastroenterology, Yidu Central Hospital of Weifang, Shandong Second Medical University, Weifang, China
| | - Xiangping Tian
- Department of Gastroenterology, Yidu Central Hospital of Weifang, Shandong Second Medical University, Weifang, China
| | - Yao Yan
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Shuyue Tian
- Department of Gastroenterology, Yidu Central Hospital of Weifang, Shandong Second Medical University, Weifang, China
| | - Dandan Liu
- Department of Gastroenterology, Yidu Central Hospital of Weifang, Shandong Second Medical University, Weifang, China.
| | - Jianmin Xu
- Department of Gastroenterology, Yidu Central Hospital of Weifang, Shandong Second Medical University, Weifang, China.
| |
Collapse
|
16
|
Kanika, Kumar A, Ahmad A, Rahul, Kumar B, Mahajan S, Ali A, Kumar J, Ali N, Navik U, Parvez S, Khan R. Beta-Sitosterol-Conjugated Sinapic Acid-Engineered Nanoliposome: Biomucoadhesive and Enzyme-Responsive Targeted Oral Therapy in Ulcerative Colitis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27839-27857. [PMID: 40298241 DOI: 10.1021/acsami.5c02190] [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: 04/30/2025]
Abstract
Developing oral drug delivery systems is promising for ulcerative colitis (UC). However, the key challenges, including formulation degradation under harsh gastric conditions, poor targeting efficiency, and limited colonic residence, lead to poor therapeutic efficacy that still needs to be tackled. Effective treatment requires a safe, efficacious, enzyme- and pH-responsive, biomucoadhesive oral drug delivery system to overcome these challenges. Therefore, we have developed chitosan-armored 1,2-dimyristoyl-sn-glycero-3-phosphoglycerol (DMPG) nanoliposomes amalgamated with synthesized beta-sitosterol-sinapic acid (Be-S) conjugate, further encapsulated with 3,4-methylenedioxy-β-nitrostyrene (MNS) as NLRP3 inhibitor, termed C@MN@DMBe-S, to overcome the limitation of free MNS and sinapic acid. Formulated by the thin-film hydration method and processed through extrusion, these unilamellar liposomes demonstrated structural stability and mucoadhesive properties due to chitosan coating. This configuration protected the nanoliposomes from the gastric acidic environment and allowed retention in the inflamed colon for 48 h. The enzyme-responsive C@MN@DMBe-S nanoliposome releases sinapic acid at the inflamed colonic site via esterase activity, providing sustained and controlled release of MNS. This synergistic action delivers antioxidant and anti-inflammatory effects while influencing the gut microbiota composition by releasing short-chain fatty acids. Moreover, therapeutic investigations revealed that C@MN@DMBe-S exhibited superior efficacy compared with free MNS when administered orally. The formulation effectively downregulated NF-κB, NLRP3, Caspase-1, and IL-1β expression while upregulating MUC5AC expression, indicating enhanced anti-inflammatory and protective effects and thereby promoting mucosal healing. In addition, C@MN@DMBe-S was found to regulate immune cell expression and effectively downregulate neutrophil infiltration. This armor- and enzyme-responsive strategy elucidates the impact of oral nanomedicines on mitigating UC and is demonstrated as an effective treatment.
Collapse
Affiliation(s)
- Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali 140306, Punjab, India
| | - Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali 140306, Punjab, India
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary AB T2N4N1, Canada
| | - Rahul
- Department of Chemistry, Malaviya National Institute of Technology, Jaipur 302017, Rajasthan, India
| | - Bhuvnesh Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali 140306, Punjab, India
| | - Shubham Mahajan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali 140306, Punjab, India
| | - Aneesh Ali
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali 140306, Punjab, India
| | - Jattin Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali 140306, Punjab, India
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda Ghudda Punjab, Bathinda 151401, Punjab, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali 140306, Punjab, India
| |
Collapse
|
17
|
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
|
18
|
Liu J, Huang H, Zhang X, Shen Y, Jiang D, Hu S, Li S, Yan Z, Hu W, Luo J, Yao H, Chen Y, Tang B. Unveiling the Cuproptosis in Colitis and Colitis-Related Carcinogenesis: A Multifaceted Player and Immune Moderator. RESEARCH (WASHINGTON, D.C.) 2025; 8:0698. [PMID: 40370501 PMCID: PMC12076167 DOI: 10.34133/research.0698] [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: 03/11/2025] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 05/16/2025]
Abstract
Cuproptosis represents a novel mechanism of cellular demise characterized by the intracellular buildup of copper ions. Unlike other cell death mechanisms, its distinct process has drawn considerable interest for its promising applications in managing inflammatory bowel disease (IBD) and colorectal cancer (CRC). Emerging evidence indicates that copper metabolism and cuproptosis may exert dual regulatory effects within pathological cellular environments, specifically modulating oxidative stress responses, metabolic reprogramming, and immunotherapeutic efficacy. An appropriate level of copper may promote disease progression and exert synergistic effects, but exceeding a certain threshold, copper can inhibit disease development by inducing cuproptosis in pathological cells. This makes abnormal copper levels a potential new therapeutic target for IBD and CRC. This review emphasizes the dual function of copper metabolism and cuproptosis in the progression of IBD and CRC, while also exploring the potential application of copper-based therapies in disease treatment. The analysis further delineates the modulatory influence of tumor immune microenvironment on cuproptosis dynamics, while establishing the therapeutic potential of cuproptosis-targeted strategies in circumventing resistance to both conventional chemotherapeutic agents and emerging immunotherapies. This provides new research directions for the development of future cuproptosis inducers. Finally, this article discusses the latest advances in potential molecular targets of cuproptosis and their related genes in the treatment of IBD and CRC, highlighting future research priorities and unresolved issues.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Gastroenterology, the Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Hairuo Huang
- China Medical University, Shenyang 110122, China
| | - Xiaojie Zhang
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Yang Shen
- Department of Radiation Oncology, Zhongshan Hospital,
Fudan University, Shanghai 200000, China
| | - DeMing Jiang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering,
Zhejiang University, Hangzhou 310007, China
| | - Shurong Hu
- Department of Gastroenterology, the Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Shuyan Li
- Department of Nursing, the Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zelin Yan
- Department of Gastroenterology, the First Affiliated Hospital of Zhejiang Chinese Medical University,
Zhejiang Provincial Key Laboratory of Gastrointestinal Diseases Pathophysiology, Hangzhou 310006, China
| | - Wen Hu
- Department of Gastroenterology, the First Affiliated Hospital of Zhejiang Chinese Medical University,
Zhejiang Provincial Key Laboratory of Gastrointestinal Diseases Pathophysiology, Hangzhou 310006, China
| | - Jinhua Luo
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Haibo Yao
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital,
Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou 310014, China
| | - Yan Chen
- Department of Gastroenterology, the Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Bufu Tang
- Department of Interventional Radiology, Zhongshan Hospital,
Fudan University, Shanghai 200000, China
| |
Collapse
|
19
|
You M, Li J, Wang X, Liu Y, Chen S, Wang P. Targeting SLC7 A11 Ameliorates Ulcerative Colitis by Promoting Efferocytosis Through the ERK1/2 Pathway. Inflammation 2025:10.1007/s10753-025-02312-6. [PMID: 40360947 DOI: 10.1007/s10753-025-02312-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/28/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
OBJECTIVE AND DESIGN This study investigates the effect and underlying mechanism of targeting SLC7A11 in mitigating dextran sulfate sodium (DSS)-induced intestinal inflammation and injury in colitis. METHODS We utilized wild-type and SLC7A11-/+ mice to assess the inflammatory damage in DSS-induced colitis in vivo. In vitro, colon tissues from patients with ulcerative colitis were analyzed to compare SLC7A11 expression between inflamed and non-inflamed regions. Further mechanistic insights were obtained using Caco-2 cells and bone marrow-derived dendritic cells (BMDCs). RESULTS In human colon tissues, SLC7A11 expression was significantly elevated in inflamed regions compared to non-inflamed areas, particularly in dendritic cells. In vivo inhibition of SLC7A11 markedly alleviated DSS-induced colitis symptoms. In vitro, suppressing SLC7A11 restored the integrity of the Caco-2 monolayer intestinal epithelial model. Both knockout and inhibition of SLC7A11 enhanced ERK1/2 phosphorylation and increased efferocytosis in BMDCs. CONCLUSIONS Targeting SLC7A11 augments dendritic cell efferocytosis and preserves intestinal epithelial barrier function, potentially offering a therapeutic avenue for alleviating ulcerative colitis.
Collapse
Affiliation(s)
- Meiyi You
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Jichang Li
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Xin Wang
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Yucun Liu
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Shanwen Chen
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China.
| | - Pengyuan Wang
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China.
| |
Collapse
|
20
|
Yang Y, Sun X, Liu B, Zhang Y, Xie T, Li J, Liu J, Zhang Q. Identifying Lactylation-related biomarkers and therapeutic drugs in ulcerative colitis: insights from machine learning and molecular docking. BMC Pharmacol Toxicol 2025; 26:103. [PMID: 40361222 PMCID: PMC12076822 DOI: 10.1186/s40360-025-00939-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Ulcerative colitis (UC), a chronic relapsing-remitting inflammatory bowel disease. Recent studies have shown that lactylation modifications may be involved in metabolic-immune interactions in intestinal inflammation through epigenetic regulation, but their specific mechanisms in UC still require in-depth validation. METHODS We conducted comparative analyses of transcriptomic profiles, immune landscapes, and functional pathways between UC and normal cohorts. Lactylation-related differentially expressed genes were subjected to enrichment analysis to delineate their mechanistic roles in UC. Through machine learning algorithms, the diagnostic model was established. Further elucidating the mechanisms and regulatory network of the model gene in UC were GSVA, immunological correlation analysis, transcription factor prediction, immunofluorescence, and single-cell analysis. Lastly, the CMap database and molecular docking technology were used to investigate possible treatment drugs for UC. RESULTS Twenty-two lactylation-related differentially expressed genes were identified, predominantly enriched in actin cytoskeleton organization and JAK-STAT signaling. By utilizing machine learning methods, 3 model genes (S100A11, IFI16, and HSDL2) were identified. ROC curves from the train and test cohorts illustrate the superior diagnostic value of our model. Further comprehensive bioinformatics analyses revealed that these three core genes may be involved in the development of UC by regulating the metabolic and immune microenvironment. Finally, regorafenib and R-428 were considered as possible agents for the treatment of UC. CONCLUSION This study offers a novel strategy to early UC diagnosis and treatment by thoroughly characterizing lactylation modifications in UC.
Collapse
Affiliation(s)
- Yao Yang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Xu Sun
- Institute of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Bin Liu
- Health Team, The 92914th Military Hospital of PLA, Lingao, Hainan, China
| | - Yunshu Zhang
- Institute of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Tong Xie
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Junchen Li
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
- Institute of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China.
| | - Jifeng Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| | - Qingkai Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
21
|
Ma J, Wu D, Xu C, He Q, Wang M, Imran M, Nazar M, Li K. Lactobacillus salivarius alleviated dextran sulfate sodium (DSS)-induced colitis in mice by mitigating oxidative stress and inflammatory responses through modulation of the intestinal flora. Microb Pathog 2025; 205:107696. [PMID: 40355053 DOI: 10.1016/j.micpath.2025.107696] [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: 03/06/2025] [Revised: 05/01/2025] [Accepted: 05/10/2025] [Indexed: 05/14/2025]
Abstract
Colitis is a multifactorial inflammatory bowel disease (IBD) involving intestinal barrier dysfunction, immune dysregulation, oxidative stress, and microbiota imbalance. Lactobacillus salivarius (L. salivarius), a probiotic with antioxidative and anti-inflammatory properties, was evaluated in a mouse model of DSS-induced colitis. Treatment with L. salivarius significantly reduced weight loss, colon shortening, and disease activity index, while improving histopathological damage. DSS-induced colitis exhibited significant oxidative stress, evidenced by decreased total antioxidant capacity, increased malondialdehyde (MDA), and reduced activity of antioxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). After the intervention of L. salivarius, MDA levels in colon tissues decreased significantly, while SOD and GSH-Px activities increased significantly. In DSS-induced colitis models, the expression of pro-inflammatory cytokines (such as TNF-α, IL-6, IL-1β) was significantly increased, while the expression of anti-inflammatory cytokine IL-10 was decreased. After the intervention of L. salivarius, the level of proinflammatory factors was significantly down-regulated, while the level of IL-10 was up-regulated. DSS treatment leads to a significant imbalance in the intestinal microbiota, characterized by an increase in the ratio of Firmicutes to Bacteroidetes (F/B ratio). The abundance of pathogenic bacteria (Alistipes, Candidatus_Soleaferrea, Frisingicoccus, Romboutsia, Streptococcus) increased, while beneficial bacteria (Anaerotruncus, Rikenella) decreased. After probiotic administration, the F/B ratio was restored significantly, and the abundance of important beneficial bacteria increased while the abundance of pathogenic bacteria decreased. These results suggest that L. salivarius alleviates DSS-induced colitis by enhancing antioxidant defense, regulating inflammatory responses, and restoring gut microbiota balance. This probiotic may offer a promising therapeutic strategy for managing ulcerative colitis (UC).
Collapse
Affiliation(s)
- Jinping Ma
- Bijie Academy of Agricultural Sciences, Bijie, 551799, China
| | - Daoyi Wu
- Bijie Academy of Agricultural Sciences, Bijie, 551799, China.
| | - Chang Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qing He
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Mingjin Wang
- Bijie Academy of Agricultural Sciences, Bijie, 551799, China
| | - Muhammad Imran
- Department of Pathology, University of Agriculture, Faisalabad, Pakistan
| | - Mudassar Nazar
- Constituent College Burewala-61010, University of Agriculture Faisalabad, Pakistan
| | - Kun Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
22
|
Li Z, Zeng L, Huang W, Zhang X, Zhang L, Xie Q. Angiogenic Factors and Inflammatory Bowel Diseases. Biomedicines 2025; 13:1154. [PMID: 40426981 DOI: 10.3390/biomedicines13051154] [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: 03/10/2025] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is characterized by chronic intestinal inflammation and impaired epithelial barrier function. Emerging evidence highlights the critical role of vascular remodeling and angiogenesis in IBD pathogenesis. This review explores the intricate relationship between blood vessels and the intestinal epithelial barrier, emphasizing how aberrant vascularization contributes to barrier dysfunction and disease progression. In IBD, excessive angiogenesis is driven by hypoxia, immune cell infiltration, and pro-inflammatory cytokines, further perpetuating inflammation and tissue damage. Key angiogenic factors, such as vascular endothelial growth factor (VEGF), angiopoietins, and platelet-derived growth factor (PDGF), are upregulated in IBD, promoting pathological vessel formation. These newly formed vessels are often immature and hyperpermeable, exacerbating leukocyte recruitment and inflammatory responses. Given the pivotal role of angiogenesis in IBD, anti-angiogenic therapies have emerged as a potential therapeutic strategy. Preclinical and clinical studies targeting VEGF and other angiogenic pathways have shown promise in reducing inflammation and promoting mucosal healing. This review summarizes current knowledge on vascular-epithelial interactions in IBD, the mechanisms driving pathological angiogenesis, and the therapeutic potential of anti-angiogenic approaches, providing insights for future research and treatment development.
Collapse
Affiliation(s)
- Zhiru Li
- Clinical Medical School, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Li Zeng
- Department of Geriatric Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Wei Huang
- Department of Geriatric Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xinxing Zhang
- Department of Geriatric Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Li Zhang
- Department of Geriatric Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Qin Xie
- Department of Geriatric Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| |
Collapse
|
23
|
Jiang Y, Chen J, Du Y, Fan M, Shen L. Immune modulation for the patterns of epithelial cell death in inflammatory bowel disease. Int Immunopharmacol 2025; 154:114462. [PMID: 40186907 DOI: 10.1016/j.intimp.2025.114462] [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: 01/17/2025] [Revised: 02/23/2025] [Accepted: 03/08/2025] [Indexed: 04/07/2025]
Abstract
Inflammatory bowel disease (IBD) is an inflammatory disease of the intestine whose primary pathological presentation is the destruction of the intestinal epithelium. The intestinal epithelium, located between the lumen and lamina propria, transmits luminal microbial signals to the immune cells in the lamina propria, which also modulate the intestinal epithelium. In IBD patients, intestinal epithelial cells (IECs) die dysfunction and the mucosal barrier is disrupted, leading to the recruitment of immune cells and the release of cytokines. In this review, we describe the structure and functions of the intestinal epithelium and mucosal barrier in the physiological state and under IBD conditions, as well as the patterns of epithelial cell death and how immune cells modulate the intestinal epithelium providing a reference for clinical research and drug development of IBD. In addition, according to the targeting of epithelial apoptosis and necroptotic pathways and the regulation of immune cells, we summarized some new methods for the treatment of IBD, such as necroptosis inhibitors, microbiome regulation, which provide potential ideas for the treatment of IBD. This review also describes the potential for integrating AI-driven approaches into innovation in IBD treatments.
Collapse
Affiliation(s)
- Yuting Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaoyao Du
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 201203, China
| | - Minwei Fan
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Lan Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
24
|
Attauabi M, Madsen GR, Bendtsen F, Seidelin JB, Burisch J. Incidence, Disease Burden, and Clinical Presentation of Patients Newly Diagnosed With Inflammatory Bowel Disease in a Population-Based Inception Cohort. J Crohns Colitis 2025; 19:jjae176. [PMID: 39565294 DOI: 10.1093/ecco-jcc/jjae176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND AND AIMS Emerging data indicate a stabilizing incidence of inflammatory bowel diseases (IBD), including ulcerative colitis (UC), Crohn's disease (CD), and IBD unclassified (IBDU) in Western countries. We aimed to investigate the incidence of IBD, its initial clinical presentation, and patient-reported burden. METHODS Copenhagen IBD Inception Cohort is a prospective, population-based cohort of patients with newly diagnosed IBD according to the ECCO guidelines in the period between May 2021 and May 2023, within a catchment area covering 20% of the Danish population. RESULTS Based on 554 patients (UC: 308, CD: 201, and IBDU: 18), the incidence rates per 100 000 person-years were as follows: IBD: 23.4 (95% confidence interval, 21.5-25.4), UC: 14.0 (12.6-15.6), CD: 8.6 (7.4-9.8), and IBDU: 0.8 (0.5-1.3). The median diagnostic delay was significantly shorter for UC (2.5 months [interquartile range {IQR} 1-6]) than for CD (5 months [IQR 1.5-11], p < 0.01). Moderate-to-severe disability was reported by 34% of CD patients and 22% of UC patients (p = 0.01), severe fatigue by 30% and 26% (p = 0.43), and severely impaired health-related quality of life (HRQoL) by 43% and 30% of patients, respectively (p = 0.01). Hospitalization rates (UC: 20%, CD: 34%, p < 0.01), and need for immunomodulators, biologics, or surgery within 3 months of diagnosis, were high in both UC (3%, 7%, and 37%, respectively) and CD (31%, 18%, and 10%, respectively). CONCLUSIONS We found a high incidence of IBD in Copenhagen with a substantial disease burden characterized by early and high requirements for advanced therapies and high rates of fatigue, disability, and impaired HRQoL at diagnosis.
Collapse
Affiliation(s)
- Mohamed Attauabi
- Department of Gastroenterology and Hepatology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Gastrounit, Medical Section, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents, and Adults, Hvidovre Hospital, Hvidovre, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gorm Roager Madsen
- Gastrounit, Medical Section, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents, and Adults, Hvidovre Hospital, Hvidovre, Denmark
| | - Flemming Bendtsen
- Gastrounit, Medical Section, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents, and Adults, Hvidovre Hospital, Hvidovre, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Benedict Seidelin
- Department of Gastroenterology and Hepatology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Johan Burisch
- Gastrounit, Medical Section, Copenhagen University Hospital-Amager and Hvidovre, Hvidovre, Denmark
- Copenhagen Center for Inflammatory Bowel Disease in Children, Adolescents, and Adults, Hvidovre Hospital, Hvidovre, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
25
|
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
|
26
|
Yue S, Gong L, Tan Y, Zhang X, Liao F. IsoalloLCA-intervened regulatory T cell exosomes alleviate inflammatory bowel disease by inhibiting NF-κB-associated inflammation in intestinal epithelial cells. Int Immunopharmacol 2025; 154:114501. [PMID: 40174336 DOI: 10.1016/j.intimp.2025.114501] [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: 11/17/2024] [Revised: 02/11/2025] [Accepted: 03/14/2025] [Indexed: 04/04/2025]
Abstract
Regulatory T cells (Tregs) are the principal immune cells that exert anti-inflammatory effects within the organism. Their exosomes exhibit therapeutic efficacy across a broad spectrum of diseases owing to their high stability, low immunogenicity, and substantial penetration capacity. Recent research have indicated that isoallolithocholic acid (isoalloLCA), a metabolite associated with bile acid metabolism, may enhance Treg activity by upregulating forkhead box protein3 (Foxp3) expression. Hence, metabolite-based strategies for reinforcing Tregs may offer novel intervention options for treating related diseases. In this study, tumor necrosis factor (TNF)-α and dextran sulfate sodium (DSS) were employed to establish cellular and animal models of inflammatory bowel disease (IBD), further evaluating the therapeutic efficacy of isoalloLCA-intervened regulatory T cell exosomes (isoalloLCA-Exo) within this model. Our findings demonstrated that isoalloLCA-Exo effectively inhibit colitis progression in a murine model, as indicated by reduced inflammation, decreased apoptosis of intestinal epithelial cells, and improved intestinal barrier function. Furthermore, in vitro analyses elucidated the molecular mechanisms underlying the anti-inflammatory effects of isoalloLCA-Exo, revealing that the intervention effectively reversed TNF-α-induced inflammation and apoptosis in intestinal epithelial cells by modulating the NF-κB pathway. In conclusion, isoalloLCA-Exo can decelerate inflammatory bowel disease progression and suppress inflammatory response in intestinal epithelial cells by inhibiting NF-κB pathway. Notably, isoalloLCA-Exo exhibit superior efficacy to the traditional drug mesalazine and conventional treg exosome(NC-Exo). These findings have significant implications for optimizing Treg-derived exosome-based therapies for inflammation-related diseases.
Collapse
Affiliation(s)
- Simei Yue
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lingjiao Gong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yulin Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaodan Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Fei Liao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Wuhan University Shenzhen Research Institute, Shenzhen, Guangdong 518000, China; Central Laboratory of Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
27
|
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
|
28
|
Li X, He N, Wang H, Wu Z, Wang M, Liang H, Xiao L, Yang Z, Li C, Xu P, Dai T, Li S, Zou Y. Therapeutic effect of Faecalibacterium longum CM04-06 on DSS-induced ulcerative colitis in mice. J Appl Microbiol 2025; 136:lxaf119. [PMID: 40372371 DOI: 10.1093/jambio/lxaf119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/06/2025] [Accepted: 05/14/2025] [Indexed: 05/16/2025]
Abstract
AIMS This study explores the impact of Faecalibacterium longum CM04-06 on inflammatory bowel disease (IBD) by regulating gut microbiota in mice. METHODS AND RESULTS We reanalyzed the distribution of the CM04-06 genome in the metagenome of the IBD cohort and observed a significantly higher abundance of CM04-06 in healthy individuals compared to patients with UC or CD. The prophylactic administration of CM04-06 was evaluated for its effects on intestinal microbial diversity and community composition after a two-week trial in mice. The intestinal microbiota was characterized using metagenomic sequencing of fecal samples on the DNBSEQ platform. CM04-06 treatment resulted in a significant reduction in the Disease Activity Index (DAI) and histological scores, as well as a decrease in the levels of pro-inflammatory cytokines, including IL-1β, IL-6, and TNF-α, in both the colon and serum of DSS-induced mice. Furthermore, supplementation with CM04-06 significantly reduced the levels of pro-inflammatory cytokines in both the colon and serum. Additionally, CM04-06 enhanced the integrity of the intestinal epithelial barrier by increasing the expression of tight junction proteins and mucin. Moreover, we observed greater abundances of Faecalibaculum rodentium, Alistipes onderdonkii, Alistipes shahii, and Bifidobacterium animalis after CM04-06 treatment. CONCLUSIONS CM04-06 prevents and alleviates intestinal inflammation by modulating the composition of the microbiota community, increasing the abundance of beneficial probiotics, and suppressing pro-inflammatory cytokine levels.
Collapse
Affiliation(s)
- Xiaofang Li
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
- BGI Research, Shenzhen 518083, China
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Haoyu Wang
- BGI Research, Shenzhen 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhinan Wu
- BGI Research, Shenzhen 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengmeng Wang
- BGI Research, Shenzhen 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Liang Xiao
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen 518083, China
| | - Zizhen Yang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Cunyin Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Ping Xu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Tong Dai
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Yuanqiang Zou
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen 518083, China
| |
Collapse
|
29
|
Shen Y, Fan N, Ma S, Cheng X, Yang X, Wang G. Gut Microbiota Dysbiosis: Pathogenesis, Diseases, Prevention, and Therapy. MedComm (Beijing) 2025; 6:e70168. [PMID: 40255918 PMCID: PMC12006732 DOI: 10.1002/mco2.70168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/22/2025] Open
Abstract
Dysbiosis refers to the disruption of the gut microbiota balance and is the pathological basis of various diseases. The main pathogenic mechanisms include impaired intestinal mucosal barrier function, inflammation activation, immune dysregulation, and metabolic abnormalities. These mechanisms involve dysfunctions in the gut-brain axis, gut-liver axis, and others to cause broader effects. Although the association between diseases caused by dysbiosis has been extensively studied, many questions remain regarding the specific pathogenic mechanisms and treatment strategies. This review begins by examining the causes of gut microbiota dysbiosis and summarizes the potential mechanisms of representative diseases caused by microbiota imbalance. It integrates clinical evidence to explore preventive and therapeutic strategies targeting gut microbiota dysregulation, emphasizing the importance of understanding gut microbiota dysbiosis. Finally, we summarized the development of artificial intelligence (AI) in the gut microbiota research and suggested that it will play a critical role in future studies on gut dysbiosis. The research combining multiomics technologies and AI will further uncover the complex mechanisms of gut microbiota dysbiosis. It will drive the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Yao Shen
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
| | - Nairui Fan
- Basic Medical College of Jiamusi UniversityHeilongjiangChina
| | - Shu‐xia Ma
- Basic Medical College of Jiamusi UniversityHeilongjiangChina
| | - Xin Cheng
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
- International SchoolGuangzhou Huali College, ZengchengGuangzhouChina
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
- Guangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryGuangdong Second Provincial General HospitalSchool of MedicineJinan UniversityGuangzhouChina
| |
Collapse
|
30
|
Wei Q, Jiang H, Zeng J, Xu J, Zhang H, Xiao E, Lu Q, Huang G. Quercetin protected the gut barrier in ulcerative colitis by activating aryl hydrocarbon receptor. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156633. [PMID: 40088746 DOI: 10.1016/j.phymed.2025.156633] [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: 08/17/2024] [Revised: 02/25/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is characterized by abdominal pain and bloody diarrhoea and restoring the gut barrier is the core goal of UC treatment. Activation of aryl hydrocarbon receptor (Ahr) was reported to effectively alleviate symptoms and repair the gut barrier damage. Neutrophil extracellular traps (NETs) have been recognized as potential targets in the treatment of UC. Ahr activation has been found to be capable of upregulating Nqo1, thereby reducing the production of reactive oxygen species (ROS), which is important in the formation of NETs. Quercetin (QUE), which is derived from natural plants and herbs used in traditional Chinese medicine (TCM), is able to strengthen gut barrier function by activating Ahr. PURPOSE The aim of this study is to investigate how QUE suppresses NETs in UC and activates Ahr in neutrophils. METHODS In this study, the dextran sulfate sodium (DSS)-induced UC model was used. Histopathological assessments were performed in the paraffin slides of tissues after H&E, PAS, Masson and alcian blue staining. The concentration of cytokines was also detected using cytometric beads array kits. Based on the transcriptomic analysis of colon tissues, western blot (WB) analysis, immunohistochemistry (IHC) assays and immunofluorescence (IF) assays were conducted to validate the significantly regulated genes and pathways. In vitro, the binding of quercetin to Ahr was calculated by molecular dynamic simulations (MDS) and biolayer interferometry (BLI) analysis. Primary neutrophils isolated from mice were cocultured with LPS or PMA with or without quercetin. The regulated genes were detected using WB, real-time quantitative PCR, enzyme-linked immunosorbent assay (ELISA) and IF analysis. The agonists and antagonist of Ahr were used as the control. RESULTS After the administration of quercetin, colon inflammation and gut barrier disruption was significantly prevented through inhibiting the NF-κB pathway and upregulating the expression of Ahr/Arnt and Nqo1. The transcriptomic analysis and IHC assays showed that inflammation and NETs were greatly decreased by QUE treatment. In vitro, quercetin inhibited LPS-induced inflammatory responses through NF-κB pathway. Furthermore, MDS and BLI analysis revealed that QUE is an agonist of AHR. QUE activated Ahr translocation and reduced ROS production via regulation of Arnt and Nqo1. CONCLUSION This study proved that quercetin greatly improved gut barrier function in the DSS-induced colitis model by regulating NET formation and that quercetin was able to activate Ahr and upregulate Arnt in neutrophils to regulate NET formation.
Collapse
Affiliation(s)
- Qiuzhu Wei
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Haixu Jiang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China; School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jia Zeng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Jie Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Honglin Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Enfan Xiao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Qingyi Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Guangrui Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 102488, China.
| |
Collapse
|
31
|
Mei T, Wang Y, Zhao C, Wu J, Ye X. Observation of the efficacy of upadacitinib as salvage therapy for refractory pediatric inflammatory bowel disease. J Pediatr Gastroenterol Nutr 2025; 80:812-815. [PMID: 40012236 DOI: 10.1002/jpn3.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/01/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025]
Affiliation(s)
- Tianlu Mei
- Department of Gastroenterology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yi Wang
- Department of Pediatrics, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Chunna Zhao
- Department of Gastroenterology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Jie Wu
- Department of Gastroenterology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xiaolin Ye
- Department of Gastroenterology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
32
|
Manoria P, Noor MT. Correlation of serum vitamin D levels with serum interleukin-23 levels in patients of ulcerative colitis. Hum Immunol 2025; 86:111305. [PMID: 40199019 DOI: 10.1016/j.humimm.2025.111305] [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: 07/24/2024] [Revised: 03/21/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025]
Abstract
Ulcerative Colitis (UC) is a chronic inflammatory condition resulting from an abnormal immune response to gut microbiota, leading to cytokine dysregulation, including elevated interleukin-23 (IL-23) levels. Emerging evidence suggests that vitamin D (VD) plays a crucial role in immune modulation. However, its correlation with IL-23 in UC is not well addressed. This study aims to elucidate the relationship between serum VD and IL-23 levels in UC patients. We included forty-four UC patients and forty-four healthy controls. VD insufficiency was more common in UC patients (n = 14) compared to controls (n = 5). Significant increases in IL-23 levels were observed from remission (46.6 ± 4.3 pg/mL) to severe stages (218.5 ± 62.41 pg/mL), while VD levels did not show a similar trend. IL-23 levels also rose significantly with disease extent, from proctitis to pancolitis. A significant negative correlation was found between VD and IL-23 levels (r = -0.3175; P = 0.035). IL-23 and pulse rate were significant predictors of UC in our cohort. Our findings highlight VD insufficiency to be prevalent in UC patients, with VD levels negatively correlating with IL-23 levels, which increase with disease severity and extent. Further, understanding the interplay between VD and IL-23 will help design therapeutic interventions to modulate immune response and disease progression.
Collapse
Affiliation(s)
- Piyush Manoria
- Department of Gastroenterology and Hepatology, Manoria Hospital, Bhopal, Madhya Pradesh, India.
| | - Mohd T Noor
- Department of Gastroenterology, Sri Aurobindo Medical College and PG Institute, Indore, Madhya Pradesh, India.
| |
Collapse
|
33
|
Belvončíková P, Gromová B, Gardlík R, Tamášová B. Challenges and insights in detecting early inflammation in DSS-induced colitis using chemiluminescence. Photochem Photobiol Sci 2025; 24:765-777. [PMID: 40312581 DOI: 10.1007/s43630-025-00724-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/14/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is a complex inflammatory condition with limited non-invasive tools to monitor early-stage inflammation. This study aimed to investigate the early stages of inflammation in acute and chronic murine models of dextran sulfate sodium (DSS)-induced colitis using in vivo and ex vivo chemiluminescence imaging. METHODS Two DSS-induced colitis models were used: an acute model over 7 days and a chronic model over 6 weeks. Body weight, stool consistency, and fecal occult blood (FOB) tests were monitored. Chemiluminescence imaging was used to assess inflammation in vivo and ex vivo, complemented by colonoscopy in the chronic model. RESULTS In the acute model, DSS-treated mice exhibited weight loss, colon shortening, and positive FOB tests by day 7. Ex vivo chemiluminescence signals exhibited a significant increase as early as day 5 (p < 0.001), while in vivo imaging showed minimal changes. In the chronic model, periodic DSS exposure resulted in recurrent inflammation, with positive FOB tests and significantly elevated ex vivo and in vivo chemiluminescence signals during the final DSS cycle (p < 0.05). Colonoscopy confirmed inflammation progression. DISCUSSION This study demonstrates the progression of inflammation in acute and chronic colitis models. However, in vivo chemiluminescence imaging did not reliably detect the onset of inflammation, limiting its application for early-stage disease detection. Ex vivo chemiluminescence and FOB tests provided more consistent insights into inflammation dynamics, addressing the need for improved non-invasive monitoring tools in UC research.
Collapse
Affiliation(s)
- Paulína Belvončíková
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08, Bratislava, Slovakia
| | - Barbora Gromová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08, Bratislava, Slovakia
| | - Roman Gardlík
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08, Bratislava, Slovakia
| | - Barbora Tamášová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08, Bratislava, Slovakia.
| |
Collapse
|
34
|
Varricchi G, Poto R, Criscuolo G, Strisciuglio C, Nair P, Marone G. TL1A, a novel alarmin in airway, intestinal, and autoimmune disorders. J Allergy Clin Immunol 2025; 155:1420-1434. [PMID: 40010414 DOI: 10.1016/j.jaci.2025.02.018] [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: 12/11/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
The term alarmin denotes a broad class of molecules rapidly released to alert the immune system through the engagement of specific receptors on immune cells. Three alarmin cytokines-thymic stromal lymphopoietin, IL-33, and IL-25-are released from epithelial and certain stromal cells. TNF-like cytokine 1A (TL1A) is a member of the TNF cytokine superfamily, first identified in human endothelial cells. TL1A is now considered a novel alarmin expressed by human and mouse bronchial and intestinal epithelial cells. TL1A exerts its biological activities by binding to a trimeric receptor DR3 (death receptor 3), expressed on a wide spectrum of immune and structural cells, including lung fibroblasts, endothelial cells, and bronchial epithelial cells. TL1A has been implicated in experimental and human inflammatory bowel diseases as well as in airway inflammation and remodeling in severe asthma. A monoclonal antibody anti-TL1A (tulisokibart) is effective in inducing clinical remission in ulcerative colitis patients. Increasing evidence suggests that TL1A is also involved in certain autoimmune disorders, such as rheumatoid arthritis and psoriasis. These emerging findings broaden the role of TL1A in various human inflammatory conditions. Several clinical trials are currently evaluating the safety and efficacy of monoclonal antibodies targeting TL1A in asthma or inflammatory bowel disease patients.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy; Istituto Endotipi in Oncologia, Metabolismo e Immunologia "G. Salvatore" (IEOMI), National Research Council (CNR), Naples, Italy.
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy; Istituti Clinici Scientifici Maugeri-IRCCS Scientific Institute of Telese Terme, Benevento, Italy
| | - Gjada Criscuolo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy
| | - Caterina Strisciuglio
- Department of Woman, Child and General and Specialistic Surgery, University of Campania "L. Vanvitelli," Naples, Italy
| | - Parameswaran Nair
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Research Institute of St Joe's Hamilton, St Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy
| |
Collapse
|
35
|
Ercan G, Aygün H, Akbaş A, Çınaroğlu OS, Erbas O. Suramin Exerts an Ameliorative Effect on Acetic Acid-Induced Acute Colitis in Rats by Demonstrating Potent Antioxidant and Anti-Inflammatory Properties. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:829. [PMID: 40428786 DOI: 10.3390/medicina61050829] [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: 04/09/2025] [Revised: 04/26/2025] [Accepted: 04/27/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: The purpose of this study was to evaluate potential protective effects of suramin on inflammation, oxidative stress, and histopathological damage a rat model of acute colitis created with acetic acid. Materials and Methods: Wistar albino (male) rats were randomly assigned to three groups: control (n = 10), colitis + saline (n = 10), and colitis + suramin (n = 10). Rectal instillation of 4% acetic acid was used to induce acute colitis. Suramin (10 mg/kg/day) or saline was administered intraperitoneally for 15 days. Plasma concentrations of pentraxin 3 (PTX3), tumor necrosis factor-alpha (TNF-α), neutrophil extracellular traps (NETs), and malondialdehyde (MDA) were determined using enzyme-linked immunosorbent assay (ELISA) and spectrophotometric methods. In addition, vascular endothelial growth factor (VEGF) and TNF-α levels in colonic tissue were also measured. Histopathological evaluations were conducted using hematoxylin and eosin staining. Results: Significant increases in plasma and tissue inflammatory markers, oxidative stress parameters, and histopathological scores were observed when compared to control group; values were higher in colitis group. Suramin treatment significantly reduced plasma PTX3, TNF-α, NETs, and MDA levels, and colonic TNF-α and VEGF concentrations compared to the untreated colitis group. Histological analysis showed reduced epithelial injury and leukocyte presence in rats receiving suramin. Conclusions: Our findings demonstrate that suramin significantly attenuates inflammatory and oxidative damage in an experimental model of acute colitis. These results suggest that suramin may possess therapeutic potential in intestinal inflammation; however, this effect requires further support through advanced experimental and clinical studies.
Collapse
Affiliation(s)
- Gulcin Ercan
- Department of General Surgery, Sultan 2. Abdulhamid Han Educational and Research Hospital, Istanbul Provincial Directorate of Health, Istanbul 34865, Turkey
| | - Hatice Aygün
- Faculty Medicine, Department of Physiology, Tokat Gaziosmanpaşa University, Tokat 60250, Turkey
| | - Ahmet Akbaş
- Department of General Surgery, Faculty of Medicine, Karadeniz Technical University, Trabzon 61080, Turkey
| | - Osman Sezer Çınaroğlu
- Department of Emergency Medicine, Faculty of Medicine, Izmir Katip Çelebi University, Izmir 35620, Turkey
| | - Oytun Erbas
- Faculty of Medicine, BAMER, Biruni University, Istanbul 34015, Türkiye
| |
Collapse
|
36
|
Lyu JQ, Xiao FJ, Wang KY, Liu YJ, Hui JM, Lin J. Research trends and hotspots of metabolites and inflammatory bowel disease: a bibliometric analysis. Front Microbiol 2025; 16:1548233. [PMID: 40365072 PMCID: PMC12069343 DOI: 10.3389/fmicb.2025.1548233] [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: 12/19/2024] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Purpose The purpose of this study is to analyze the current research status and explore the relationship between metabolites and inflammatory bowel disease (IBD), providing insights for future research. Methods In this study, we retrieved publications on metabolites and IBD from the Web of Science Core Collection (WOSCC), covering the period from 1994 to 2024. We conducted descriptive and visual analyses of the topics, journals, countries/regions, institutions, authors, and citation counts of these publications. Results From January 1994 to June 2024, a total of 509 relevant publications were retrieved from the WOSCC, with the number of publications steadily increasing each year. These articles were published in 222 journals, with the top three most productive journals being inflammatory bowel diseases (36 publications), Alimentary Pharmacology & Therapeutics (16 publications), and Digestive Diseases and Sciences (13 publications). The leading countries in publication output were China (154 publications, 30.3%), the USA (101 publications, 19.8%), and the UK (32 publications, 6.3%), with total citation counts of 3,175, 7,439, and 1,444, respectively. The most recent trending keywords in this field include "gut microbiota," "inflammation," and "pathogenesis." Conclusion Recent research on the relationship between metabolites and inflammatory bowel disease (IBD) has grown significantly, deepening our understanding of their connection. Further exploration of this relationship could not only enhance the quality of life for IBD patients but also offer new insights into potential cures for the disease.
Collapse
Affiliation(s)
- Jia-Qi Lyu
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Fang-Jun Xiao
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, Guangdong, China
| | - Ke-Ying Wang
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ying-Jie Liu
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jing-Mei Hui
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, Guangdong, China
| | - Jie Lin
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, Guangdong, China
| |
Collapse
|
37
|
Lu H, Zhang G, Wang K, Liu K, Gao Y, Chen J, Li Y, Yan J. The Role of Lactiplantibacillus plantarum CGMCC9513 in Alleviating Colitis by Synergistic Enhancement of the Intestinal Barrier Through Modulating Gut Microbiota and Activating the Aryl Hydrocarbon Receptor. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10551-0. [PMID: 40301232 DOI: 10.1007/s12602-025-10551-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2025] [Indexed: 05/01/2025]
Abstract
Ulcerative colitis (UC) has become a global health issue. This study evaluated whether administering Lactiplantibacillus plantarum CGMCC9513 to dextran sulfate sodium (DSS)-induced colitis mice could alleviate colitis by modulating gut microbiota imbalance and activating the aryl hydrocarbon receptor (AhR) to enhance intestinal barrier function. The anti-inflammatory effect and AhR activation ability of L. plantarum CGMCC9513 were evaluated with lipopolysaccharide (LPS)-induced cell inflammation model; 25 male BALB/c mice were divided into blank group (CNG), model group (DSS), L. plantarum CGMCC9513-treated group (LPDT), and L. plantarum CGMCC9513 control group (LP). The mice were pre-administered L. plantarum CGMCC9513 for 14 days and continued to receive it during DSS induction. Symptoms during induction, goblet cell count, expression of MUC2 and Occludin proteins, and changes in gut microbiota were observed. Subsequently, the expression of cytokines interleukin-10 (IL-10), tumor necrosis factor (TNF-α), interleukin-1β (IL-1β) and AhR activation status was determined. The study found that L. plantarum CGMCC9513 could alleviate cell inflammation induced by LPS and activate AhR in vitro. For colitis mice, it could reduce colonic mucosal damage and enhance intestinal barrier function. Regarding gut microbiota changes, L. plantarum CGMCC9513 mainly downregulated Bacteroides, Blautia, Escherichia-Shigella, and Lachnospiraceae_ NK4A136_group and upregulated Firmicutes, Lactobacillus. It reduces the risk of bacterial translocation and increases beneficial gut bacteria. L. plantarum CGMCC9513 reduced the expression of pro-inflammatory cytokines TNF-α and IL-1β while increasing the expression of anti-inflammatory cytokine IL-10. Meanwhile, increased expression of AhR and Cytochrome P450 1A1 (CYP1A1) proteins indicated AhR activation by L. plantarum CGMCC9513. In conclusion, L. plantarum CGMCC9513 can synergistically enhance intestinal barrier alleviation in colitis mice by modulating gut microbiota imbalance and activating AhR.
Collapse
Affiliation(s)
- Hongyu Lu
- Medical School of Guangxi University, Nanning, 530004, China
| | - Guoqing Zhang
- Medical School of Guangxi University, Nanning, 530004, China
| | - Kaidi Wang
- Medical School of Guangxi University, Nanning, 530004, China
| | - Kefei Liu
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Yingrui Gao
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Jinyan Chen
- Tianjin Shengji Group., Co., Ltd, Huayuan Industrial Zone, No. 2, Hai Tai Development 2 Road, Tianjin, 300384, China
| | - Yixiang Li
- Medical School of Guangxi University, Nanning, 530004, China.
| | - Jianhua Yan
- Medical School of Guangxi University, Nanning, 530004, China.
| |
Collapse
|
38
|
Thapa HB, Passegger CA, Fleischhacker D, Kohl P, Chen YC, Kalawong R, Tam-Amersdorfer C, Gerstorfer MR, Strahlhofer J, Schild-Prüfert K, Zechner EL, Blesl A, Binder L, Busslinger GA, Eberl L, Gorkiewicz G, Strobl H, Högenauer C, Schild S. Enrichment of human IgA-coated bacterial vesicles in ulcerative colitis as a driver of inflammation. Nat Commun 2025; 16:3995. [PMID: 40301356 PMCID: PMC12041585 DOI: 10.1038/s41467-025-59354-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/21/2025] [Indexed: 05/01/2025] Open
Abstract
The gut microbiome contributes to chronic inflammatory responses in ulcerative colitis (UC), but molecular mechanisms and disease-relevant effectors remain unclear. Here we analyze the pro-inflammatory properties of colonic fluid obtained during colonoscopy from UC and control patients. In patients with UC, we find that the pelletable effector fraction is composed mostly of bacterial extracellular vesicles (BEVs) that exhibit high IgA-levels and incite strong pro-inflammatory responses in IgA receptor-positive (CD89+) immune cells. Biopsy analyses reveal higher infiltration of CD89+ immune cells in the colonic mucosa from patients with UC than control individuals. Further studies show that IgA-coated BEVs, but not host-derived vesicles nor soluble IgA, are potent activators of pro-inflammatory responses in CD89+ cells. IgA-coated BEVs also exacerbate intestinal inflammation in a dextran sodium sulfate colitis model using transgenic mice expressing human CD89. Our data thus implicate a link between IgA-coated BEVs and intestinal inflammation via CD89+ immune cells, and also hint a potential new therapeutic target for UC.
Collapse
Affiliation(s)
- Himadri B Thapa
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Christina A Passegger
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | | | - Paul Kohl
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Yi-Chi Chen
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Ratchara Kalawong
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Carmen Tam-Amersdorfer
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Michael R Gerstorfer
- Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
| | - Jana Strahlhofer
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | - Ellen L Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed, Graz, Austria
- Field of Excellence Biohealth - University of Graz, Graz, Austria
| | - Andreas Blesl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Lukas Binder
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Georg A Busslinger
- Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
| | - Leo Eberl
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Gregor Gorkiewicz
- BioTechMed, Graz, Austria
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Herbert Strobl
- Division of Immunology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
- BioTechMed, Graz, Austria
| | - Christoph Högenauer
- BioTechMed, Graz, Austria.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
| | - Stefan Schild
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- BioTechMed, Graz, Austria.
- Field of Excellence Biohealth - University of Graz, Graz, Austria.
- Austrian Agency for Health and Food Safety (AGES), Institute for Medical Microbiology and Hygiene, Graz, Austria.
| |
Collapse
|
39
|
Kuo SN, Wu PX, Huang SL, Hsu YC, Huang JH. Thermo-responsive methylcellulose/hyaluronic acid-mesalamine hydrogel in targeted drug delivery for ulcerative colitis. RSC Adv 2025; 15:14126-14135. [PMID: 40313324 PMCID: PMC12044526 DOI: 10.1039/d5ra00216h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 04/17/2025] [Indexed: 05/03/2025] Open
Abstract
Current treatments for ulcerative colitis (UC), including mesalamine (Me) enemas, face limitations such as poor colonic retention, systemic side effects, and suboptimal patient compliance. To address these challenges, this study developed a thermo-responsive hydrogel combining hyaluronic acid-mesalamine (HA-Me) conjugates with methylcellulose (MC), providing a targeted and sustained drug delivery platform for UC treatment. HA-Me conjugates were synthesized via a nucleophilic addition-elimination reaction, with FT-IR and 1H-NMR confirming successful conjugation and a grafting ratio of 12.45%. Rheological analysis revealed a lower critical solution temperature (LCST) of 36.7-37.7 °C, ensuring gelation at body temperature when the MC concentration was 5-7 wt%. The optimized hydrogel exhibits intestinal retention properties, thereby improving drug bioavailability. The results confirmed that this hydrogel not only improved drug release time but also provided a protective barrier for inflamed wounds, facilitating wound healing, reducing the risk of reinfection, and improving medical compliance. Its mucoadhesive properties further supported effective drug delivery and localized therapeutic effects. This study highlights the potential of the MC/HA-Me hydrogel as a platform for overcoming the limitations of conventional UC treatments, offering opportunities for tailored therapeutic applications and future clinical development.
Collapse
Affiliation(s)
- Sheng-Nan Kuo
- Department of Chemical Engineering, National Tsing Hua University Hsinchu 30013 Taiwan +886-3-5743051
| | - Pei-Xhan Wu
- Department of Chemical Engineering, National United University Miaoli 36003 Taiwan +886-37-382-209
| | - Shu-Ling Huang
- Department of Chemical Engineering, National United University Miaoli 36003 Taiwan +886-37-382-209
- Science/International Master Program of Translation Medicine, National United University Miaoli 36003 Taiwan
| | - Yu-Ci Hsu
- Department of Chemical Engineering, National United University Miaoli 36003 Taiwan +886-37-382-209
| | - Jen-Huang Huang
- Department of Chemical Engineering, National Tsing Hua University Hsinchu 30013 Taiwan +886-3-5743051
| |
Collapse
|
40
|
Ahmadi A, Shokoohizadeh L, Sheikhesmaili F, Mirzaei MK, Mohammadi A, Nikkhoo B, Khodaei H, Alikhani MY, Yousefimashouf R. Gut microbiomes and treatment-resistant ulcerative colitis: a case-control study using qPCR. BMC Microbiol 2025; 25:254. [PMID: 40295906 PMCID: PMC12036124 DOI: 10.1186/s12866-025-03963-z] [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: 02/10/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The gut microbiome has been identified as a pivotal factor in ulcerative colitis (UC), given its role as the main reservoir of microbes in the body. This community of microorganisms, present in variable concentrations in the digestive tract, makes a wide range of beneficial roles for the host. However, the role of the gut microbiome in patients with refractory UC is still significant, so this study aimed to further investigate the role of these bacteria in patients with refractory UC. METHODS This case-control study was conducted on stool samples from four distinct groups: the first group comprised new patients diagnosed with ulcerative colitis (all of them had responded to treatment after follow-up) (N = 24); the second group consisted of patients with treatment-resistant ulcerative colitis (N = 23); the third group included first-degree relatives of group 1 patients (N = 24); and the fourth group consisted of first-degree relatives of group 2 patients (N = 23). The research tools employed in this study included a questionnaire, quantitative real-time PCR (qPCR) test, and culture on stool samples. RESULT The mean age of patients in groups 1 and 2 was 45.88 ± 18.51 and 41.30 ± 13.01 years, while the mean age of controls in groups 3 and 4 was 37.29 ± 9.62 and 40.96 ± 13.01 years, respectively. Stool culture results for pathogenic bacteria were negative in all four groups. The of history of consuming dairy products containing probiotics was highest in Group 1, with 22 (91.67%) subjects, while the lowest was observed in Group 3, with 16 (66.67%). The highest history of self-administered antibiotic use was observed in Group 2, with 13 cases (56.52%), while the lowest was noted in Group 3, with 4 cases (16.67%). The findings indicated a statistically significant relationship (P < 0.05) between Groups 2 and 4 with respect to the E. coli and Bifidobacterium ssp. microbial population. Additionally, a significant relationship was identified between the Lactobacillus ssp., Bifidobacterium ssp., and Bacteroides ssp. microbial community between groups 1 and 2 (P < 0.05). CONCLUSION The findings of this study demonstrated that several intestinal microbiomes have a substantial impact on the management of ulcerative colitis. The results of this study suggest that by comparing the gut microbiome of treatment-resistant and individuals newly diagnosed with ulcerative colitis, we can gain a better understanding of microbiome differences that may influence treatment outcomes. The results of this study may also lead to the identification of new therapeutic strategies that are based on regulating the gut microbiome. These strategies could include the use of fecal microbiome transplantation (FMT), probiotics, prebiotics, or specific bacteria-based therapies.
Collapse
Affiliation(s)
- Amjad Ahmadi
- Infectious Disease Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Leili Shokoohizadeh
- Infectious Disease Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Farshad Sheikhesmaili
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammadali Khan Mirzaei
- Institute of Virology, Helmholtz Munich, German Research Centre for Environmental Health, Neuherberg, Germany
- Chair of Prevention of Microbial Infectious Diseases, Central Institute of Disease Prevention, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Asadollah Mohammadi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Bahram Nikkhoo
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Hakim Khodaei
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Yousef Alikhani
- Infectious Disease Research Center, Avicenna Institute of Clinical Sciences, Avicenna Health Research Institute, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Rasoul Yousefimashouf
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
41
|
Miraghaee DS, Khalili A, Bayat G, Mousavi Z, Nazari M, Hosseini M, Goudarzvand M, Mazloom R. A single dose of nicotine modulates heart rate variability in rats with induced-ulcerative colitis. Auton Neurosci 2025; 260:103282. [PMID: 40306144 DOI: 10.1016/j.autneu.2025.103282] [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: 08/26/2023] [Revised: 12/02/2024] [Accepted: 04/18/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND AND AIMS Nicotine, a widely used toxic substance, has various receptors scattered throughout the body that have shown opposite effects on inflammatory disorders. However, the effects of nicotine on heart rate variability in ulcerative colitis are unclear. Therefore, the present study aimed to determine the effect of acute nicotine injection on heart rate variability in a rat model of ulcerative colitis. METHODS Six male Wistar rat groups, containing vehicle, UC (induction of ulcerative colitis without treatment), and nicotine (0.5, 1, 1.5, and 2 mg/kg), were assessed. First, the rats were anesthetized and the initial electrocardiogram was recorded. Twenty-four hours after the induction of ulcerative colitis with 4 % acetic acid by rectal injection, a second electrocardiogram was recorded. Finally, 15 min after nicotine injection in each group, the last electrocardiogram was recorded. Linear and nonlinear indices of heart rate variability were extracted from the recorded R-R intervals. RESULTS A single injection of nicotine at high doses increased the standard deviation of R-R intervals, root mean square of successive differences between normal heartbeats, ratio of the short-term deviation to the long-term deviation of R-R intervals, and entropy of R-R intervals in ulcerative colitis animals (at least P < 0.05). CONCLUSIONS Acute injection of nicotine at doses 1.5 and 2 mg/kg can improve R-R interval linear indices, balance the ratio of short-term deviation to long-term deviation, and modify the entropy in the induced ulcerative colitis rats. However, further research is needed for the clinical use of acute nicotine injection in ulcerative colitis.
Collapse
Affiliation(s)
- Diba Sadat Miraghaee
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Azadeh Khalili
- Department of Physiology-Pharmacology-Medical Physics, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Bayat
- Department of Physiology-Pharmacology-Medical Physics, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Zahra Mousavi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Milad Nazari
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark; DANDRITE, the Danish Research Institute of Translational Neuroscience, Aarhus, Denmark.
| | - Marjan Hosseini
- Department of Physiology-Pharmacology-Medical Physics, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mahdi Goudarzvand
- Department of Physiology-Pharmacology-Medical Physics, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| | - Roham Mazloom
- Department of Physiology-Pharmacology-Medical Physics, Faculty of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
42
|
Sailer FK, Palmer MA, Aliraj B, Heering J, Brockmann A, Elewa MAF, Röhrig A, Proschak E, Stepniak DT, Ramsey S, Brüne B, Weigert A. S1PR4-dependent effects of Etrasimod on primary human myeloid immune cell activation. Front Pharmacol 2025; 16:1590816. [PMID: 40342995 PMCID: PMC12058506 DOI: 10.3389/fphar.2025.1590816] [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: 03/10/2025] [Accepted: 04/07/2025] [Indexed: 05/11/2025] Open
Abstract
Background Sphingosine-1-phosphate (S1P) and its five receptors S1PR1-5 play an essential role in the migration, differentiation and activation of various immune cells. Several S1PR modulators with distinct selectivity have been recently approved for the treatment of various inflammatory diseases. Among those are Ozanimod, an S1PR1/5 modulator approved for the treatment of ulcerative colitis and multiple sclerosis, and Etrasimod, an S1PR1/4/5 modulator approved for the treatment of ulcerative colitis. Chronic autoinflammatory diseases such as the inflammatory bowel diseases (IBDs) Crohn's disease and ulcerative colitis are characterized by an abundance of disease-propagating immune cells in the gastrointestinal tract. Since currently available treatment options such as biologics provide a sometimes inadequate treatment response, one alternative strategy to treat IBDs is the use of S1P receptor modulators. Objective We aimed to investigate if targeting S1PR4 affects the impact of Etrasimod on the activation of primary human immune cells, and to elucidate the mode of action of Etrasimod on S1PR4. Methods Primary human macrophages, plasmacytoid dendritic cells and neutrophils were pretreated with S1P, Etrasimod (S1PR1/4/5), Ozanimod (S1PR1/5), Siponimod (S1PR1/5), CYM 50308 (S1PR4 agonist) and CYM 50358 (S1PR4 antagonist), and then stimulated with Zymosan A, ODN 2336 and PMA, respectively. We measured cytokine and chemokine production by macrophages and plasmacytoid dendritic cells via CBA/Legendplex, and survival and activation markers for neutrophils via flow cytometry. Confocal microscopy of S1PR-expressing CHO-K1 cell lines was used to study receptor internalization. Results We found that signaling induced by S1P, Etrasimod and the S1PR4 agonist attenuates CCL20 and CXCL5 production by Zymosan-stimulated macrophages, and these findings were confirmed by S1PR4 knockdown. Additionally, S1PR4 was involved in the regulation of IFN-α production by ODN2336-stimulated plasmacytoid dendritic cells. Lastly, both Etrasimod and the S1PR4 agonist reduced the activation level of PMA-stimulated neutrophils. Regarding receptor dynamics, we show that Etrasimod induces internalization of S1PR4. Conclusion Taken together, our data show that S1PR4 takes on an essential role in the regulation of various immunological functions, and that Etrasimod can act as a superagonist/functional antagonist of S1PR4.
Collapse
Affiliation(s)
- Fiona K. Sailer
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Megan A. Palmer
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Blerina Aliraj
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jan Heering
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Frankfurt am Main, Germany
| | - Andreas Brockmann
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Mohammed A. F. Elewa
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
- Biochemistry Department, Faculty of Pharmacy, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | - Aissa Röhrig
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
- Department of Internal Medicine 1, Goethe University Hospital, Frankfurt am Main, Germany
| | - Ewgenij Proschak
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Frankfurt am Main, Germany
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Simeon Ramsey
- Inflammation and Immunology, Pfizer Inc., Cambridge, MA, United States
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
43
|
Wang M, Jiang Y, Chen Z, Jiang D, Jiang X, Ye J, Wang H, Liu Y. Colon-Targeted Mucoadhesive PLGA Microspheres Loaded with Ramulus Mori Alkaloids for Enhanced Water-Soluble Drug Delivery in Ulcerative Colitis Treatment. Molecules 2025; 30:1878. [PMID: 40363686 PMCID: PMC12073386 DOI: 10.3390/molecules30091878] [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: 03/16/2025] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammation disease with severe impact on quality of life, with limited treatment options. Ramulus Mori alkaloids (SZ-A) from Morus alba show promise for UC treatment due to their safety and pharmacological effects, including anti-inflammation and barrier repair. However, their clinical use has been limited by gastrointestinal flatulence as a side effect due to their pharmacological action as an α-glucosidase inhibitor targeting the small intestine following oral administration. Therefore, constructing a colon-targeted formulation to deliver SZ-A is an advantageous strategy to improve UC therapy. In this study, we used the complex formed by thiolated hyaluronic acid, which has mucosal adhesion and inflammation-targeting properties, and SZ-A as an intermediate carrier and prepared sodium alginate-modified PLGA microspheres (SZ-A@MSs) with the double emulsion method to achieve efficient encapsulation of SZ-A. Specifically, sodium alginate serves as a gastric acid protectant and microbiota-responsive material, enabling the precise and responsive release of microspheres in the colonic region. SZ-A@MSs have a particle size of about 30 µm, a drug loading of about 12.0%, and an encapsulation efficiency of about 31.7% and function through intestinal adhesion to and targeting of inflammatory sites. SZ-A@MSs showed antioxidant and anti-inflammatory abilities in Raw264.7 cells. In vivo imaging results suggest that SZ-A@MSs have good colon site retention and sustained-release effect. Pharmacodynamic results show that SZ-A@MSs display good efficacy, including the ability to inhibit weight loss, inhibit colonic atrophy, and inhibit the secretion of inflammatory factors. In conclusion, SZ-A@MSs have good colon-targeting properties, can improve therapeutic effects, and provide a potential treatment method for UC.
Collapse
Affiliation(s)
- Mo 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, China; (M.W.); (Y.J.); (Z.C.); (D.J.); (X.J.); (J.Y.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yu Jiang
- 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, China; (M.W.); (Y.J.); (Z.C.); (D.J.); (X.J.); (J.Y.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Zhiyang Chen
- 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, China; (M.W.); (Y.J.); (Z.C.); (D.J.); (X.J.); (J.Y.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Dengbao Jiang
- 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, China; (M.W.); (Y.J.); (Z.C.); (D.J.); (X.J.); (J.Y.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xuan Jiang
- 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, China; (M.W.); (Y.J.); (Z.C.); (D.J.); (X.J.); (J.Y.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jun Ye
- 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, China; (M.W.); (Y.J.); (Z.C.); (D.J.); (X.J.); (J.Y.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Hongliang 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, China; (M.W.); (Y.J.); (Z.C.); (D.J.); (X.J.); (J.Y.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Yuling 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, China; (M.W.); (Y.J.); (Z.C.); (D.J.); (X.J.); (J.Y.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
44
|
Li X, Yan C, Li S, Shen L, Huo L. Mechanism of LncRNA CBR3-AS1 in regulating pyroptosis of intestinal epithelial cells in ulcerative colitis. J Bioenerg Biomembr 2025:10.1007/s10863-025-10060-3. [PMID: 40257734 DOI: 10.1007/s10863-025-10060-3] [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: 11/27/2024] [Accepted: 04/02/2025] [Indexed: 04/22/2025]
Abstract
Ulcerative colitis (UC) is a common chronic relapsing inflammatory disease that threatens human life. This study aims to explore the mechanism of LncRNA CBR3-AS1 in pyroptosis of intestinal epithelial cells in UC. The levels of CBR3-AS1, KLF2, and SUGT1 in UC cells were detected. After downregulating CBR3-AS1 expression, cell viability and pyroptosis were measured, followed by the detection of SOD and MDA levels. The binding of CBR3-AS1 to EZH2, enrichment of EZH2 and H3K27me3 on the KLF2 promoter, and binding of KLF2 to the SUGT1 promoter were assayed. The role of CBR3-AS1 in pyroptosis was validated in animal models. We found that CBR3-AS1 and SUGT1 were increased in UC cells, and KLF2 was decreased. After downregulation of CBR3-AS1, cell viability was increased and pyroptosis was alleviated. CBR3-AS1 recruited EZH2 to occupy the KLF2 promoter, leading to increased H3K27me3 levels and suppressed KLF2 expression, reducing the enrichment of KLF2 on the SUGT1 promoter, finally promoting SUGT1 expression. SUGT1 overexpression or KLF2 downregulation alleviated the protective effect of silencing CBR3-AS1 on pyroptosis in UC cells. CBR3-AS1 downregulation alleviates cell pyroptosis in colonic tissues. In conclusion, CBR3-AS1 exacerbated pyroptosis of intestinal epithelial cells in UC via the KLF2/SUGT1 pathway.
Collapse
Affiliation(s)
- Xi Li
- Department of Gastroenterology, The First Affiliated Hospital of Shanxi Medical University, 85 Jiefang South Road, Yingze District, Taiyuan, Shanxi Province, 030000, China
- Department of Gastroenterology, Changzhi People's Hospital, 502 Changxing Middle Road, Changzhi, 046000, China
| | - Caiwen Yan
- Department of Gastroenterology, Changzhi People's Hospital, 502 Changxing Middle Road, Changzhi, 046000, China
| | - Suxia Li
- Department of Gastroenterology, The First Affiliated Hospital of Shanxi Medical University, 85 Jiefang South Road, Yingze District, Taiyuan, Shanxi Province, 030000, China
| | - Lujun Shen
- Department of Gastroenterology, Changzhi People's Hospital, 502 Changxing Middle Road, Changzhi, 046000, China
| | - Lijuan Huo
- Department of Gastroenterology, The First Affiliated Hospital of Shanxi Medical University, 85 Jiefang South Road, Yingze District, Taiyuan, Shanxi Province, 030000, China.
| |
Collapse
|
45
|
Adam KM, Ali EW, Elangeeb ME, Abuagla HA, Elamin BK, Ahmed EM, Edris AM, Ahmed AAEM, Eltieb EI. Intelligent Care: A Scientometric Analysis of Artificial Intelligence in Precision Medicine. Med Sci (Basel) 2025; 13:44. [PMID: 40265391 PMCID: PMC12015873 DOI: 10.3390/medsci13020044] [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: 02/17/2025] [Revised: 03/24/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025] Open
Abstract
The integration of advanced computational methods into precision medicine represents a transformative advancement in healthcare, enabling highly personalized treatment strategies based on individual genetic, environmental, and lifestyle factors. These methodologies have significantly enhanced disease diagnostics, genomic analysis, and drug discovery. However, rapid expansion in this field has resulted in fragmented understandings of its evolution and persistent knowledge gaps. This study employs a scientometric approach to systematically map the research landscape, identify key contributors, and highlight emerging trends in precision medicine. Methods: A scientometric analysis was conducted using data retrieved from the Scopus database, covering publications from 2019 to 2024. Tools such as VOSviewer and R-bibliometrix package (version 4.3.0) were used to perform co-authorship analysis, co-citation mapping, and keyword evolution tracking. The study examined annual publication growth, citation impact, research productivity by country and institution, and thematic clustering to identify core research areas. Results: The analysis identified 4574 relevant publications, collectively amassing 70,474 citations. A rapid growth trajectory was observed, with a 34.3% increase in publications in 2024 alone. The United States, China, and Germany emerged as the top contributors, with Harvard Medical School, the Mayo Clinic, and Sichuan University leading in institutional productivity. Co-citation and keyword analysis revealed three primary research themes: diagnostics and medical imaging, genomic and multi-omics data integration, and personalized treatment strategies. Recent trends indicate a shift toward enhanced clinical decision support systems and precision drug discovery. Conclusions: Advanced computational methods are revolutionizing precision medicine, spurring increased global research collaboration and rapidly evolving methodologies. This study provides a comprehensive knowledge framework, highlighting key developments and future directions. The insights derived can inform policy decisions, funding allocations, and interdisciplinary collaborations, driving further advancements in healthcare solutions.
Collapse
Affiliation(s)
- Khalid M. Adam
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia; (E.W.A.); (M.E.E.); (H.A.A.); (E.M.A.); (A.M.E.); (A.A.E.M.A.); (E.I.E.)
| | - Elshazali W. Ali
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia; (E.W.A.); (M.E.E.); (H.A.A.); (E.M.A.); (A.M.E.); (A.A.E.M.A.); (E.I.E.)
| | - Mohamed E. Elangeeb
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia; (E.W.A.); (M.E.E.); (H.A.A.); (E.M.A.); (A.M.E.); (A.A.E.M.A.); (E.I.E.)
| | - Hytham A. Abuagla
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia; (E.W.A.); (M.E.E.); (H.A.A.); (E.M.A.); (A.M.E.); (A.A.E.M.A.); (E.I.E.)
| | - Bahaeldin K. Elamin
- Department of Microbiology and Clinical Parasitology, College of Medicine, University of Bisha, P.O. Box 1290, Bisha 67714, Saudi Arabia;
| | - Elsadig M. Ahmed
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia; (E.W.A.); (M.E.E.); (H.A.A.); (E.M.A.); (A.M.E.); (A.A.E.M.A.); (E.I.E.)
| | - Ali M. Edris
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia; (E.W.A.); (M.E.E.); (H.A.A.); (E.M.A.); (A.M.E.); (A.A.E.M.A.); (E.I.E.)
| | - Abubakr A. Elamin Mohamed Ahmed
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia; (E.W.A.); (M.E.E.); (H.A.A.); (E.M.A.); (A.M.E.); (A.A.E.M.A.); (E.I.E.)
| | - Elmoiz I. Eltieb
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, P.O. Box 255, Bisha 67714, Saudi Arabia; (E.W.A.); (M.E.E.); (H.A.A.); (E.M.A.); (A.M.E.); (A.A.E.M.A.); (E.I.E.)
| |
Collapse
|
46
|
Braga-Neto MB, Qazi T, Fulmer C, Holubar SD, Fiocchi C, Ivanov AI, Rieder F. Cellular and molecular mechanisms in the pathogenesis of pouchitis: more than just the microbiota. Gut 2025:gutjnl-2024-334445. [PMID: 40240062 DOI: 10.1136/gutjnl-2024-334445] [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: 11/25/2024] [Accepted: 03/28/2025] [Indexed: 04/18/2025]
Abstract
Pouchitis, defined as inflammation of the ileal pouch, is the most common complication following restorative proctocolectomy for refractory ulcerative colitis. Antibiotics remain the first line of therapy for pouchitis, but the majority of patients develop subsequent episodes and some are refractory to antibiotic therapy. This highlights the need for more effective treatment options and points to a more complex pathophysiology beyond the role of th pouch microbiome, similar to what is seen in inflammatory bowel disease. In this review, we outline the putative mechanisms of pouchitis, including genetic predisposition, microbiome alterations, dysfunction of the intestinal barrier and the immune system and review the available animal models of pouchitis. In addition, we introduce the concept of pouchitis as a possible transmural disease and discuss the potential role of non-immune cells, including stromal cells, in perpetuating inflammation and intestinal barrier dysfunction. We discuss future directions, implications for novel therapies and propose novel multicellular disease models that can better capture the complexity of pouchitis pathogenesis.
Collapse
Affiliation(s)
- Manuel B Braga-Neto
- Department of Gastroenterology, Hepatology & Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Taha Qazi
- Department of Gastroenterology, Hepatology & Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Clifton Fulmer
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Stefan D Holubar
- Department of Colon and Rectal Surgery, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Claudio Fiocchi
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Andrei I Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Florian Rieder
- Department of Gastroenterology, Hepatology & Nutrition, Digestive Disease Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Program for Global Translational Inflammatory Bowel Disease, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
47
|
Zhao Z, Lu H, Wang J, Wu T, Xu S, Ge Y, You Q, Jiang Z, Lu M. Discovery of β-amino acid substituted naphthalene sulfonamide derivatives as potent Kelch-like ECH-associated protein 1-nuclear factor erythroid 2-related factor 2 (Keap1-Nrf2) protein-protein interaction inhibitors for ulcerative colitis management. Eur J Med Chem 2025; 288:117384. [PMID: 39965408 DOI: 10.1016/j.ejmech.2025.117384] [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: 12/20/2024] [Revised: 01/24/2025] [Accepted: 02/02/2025] [Indexed: 02/20/2025]
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a key regulator of cellular defense system against oxidative insults. Directly inhibiting the Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 protein-protein interaction (PPI) has emerged as a promising approach to activate Nrf2 for the treatment of diseases associated with oxidative stress. Herein, we identified β-amino acids as privileged structural fragments for designing novel naphthalene sulfonamide-based Keap1-Nrf2 PPI inhibitors. Comprehensive structure-activity relationship (SAR) exploration identified compound 19 as the optimal inhibitor with an IC50 of 0.55 μM for disrupting the Keap1-Nrf2 interaction and a Kd of 0.50 μM for binding to Keap1. Further studies demonstrated that 19 effectively activated the Nrf2-regulated cytoprotective system and provided protective effects against dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in both in vitro and in vivo models. These findings highlight the potential of β-amino acid substituted naphthalene sulfonamide Keap1-Nrf2 inhibitor 19 as a prospective therapeutic agent for UC via Keap1 targeting.
Collapse
Affiliation(s)
- Ziquan Zhao
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hongjin Lu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Junjie Wang
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Tingting Wu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Shicheng Xu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuxin Ge
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University Medical College, Suzhou, 215123, China.
| | - Zhengyu Jiang
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Mengchen Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University Medical College, Suzhou, 215123, China.
| |
Collapse
|
48
|
Pan X, Xian P, Li Y, Zhao X, Zhang J, Song Y, Nan Y, Ni S, Hu K. Chemotaxis-driven hybrid liposomes recover intestinal homeostasis for targeted colitis therapy. J Control Release 2025; 380:829-845. [PMID: 39961435 DOI: 10.1016/j.jconrel.2025.02.036] [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: 10/13/2024] [Revised: 02/05/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
Inflammatory bowel disease (IBD) is closely linked to the dysregulation of intestinal homeostasis, accompanied by intestinal epithelial barrier destruction, dysbiosis of gut microbiota, subsequent inflammatory factor infiltration, and excessive oxidative stress. Conventional therapeutics focus on suppressing inflammation and often suffer from metabolic instability as well as limited targeting, thereby leading to suboptimal remission rates and severe side effects. Here, we designed bacterial outer membrane vesicle (OMV, from Stenotrophomonas maltophilia)-fused and borneol-modified liposomes (BO/OMV-lipo@LU) for targeted delivery of luteolin to recover intestinal homeostasis by alleviating inflammation and modulating dysregulated intestinal epithelial barrier, redox balance, and gut microbiota in IBD. In a Caco-2/HT29-MTX monolayer model, the OMV and borneol-bifunctionalized liposomes enhanced the uptake efficiency of unfunctionalized liposomes with a 2-fold increase. Owing to the chemotaxis-driven colon-targeting ability of OMVs and the ability of borneol to promote intestinal epithelial uptake, the hybrid liposomes successfully targeted the inflamed colon. In a colitis mouse model, BO/OMV-lipo@LU exhibited enhanced efficacy following oral administration. The BO/OMV-lipo@LU treatment increased the colon length and body weights of mice suffering colitis by 40 % and 15 %, respectively, with values comparable to the healthy control group. Notably, BO/OMV-lipo@LU alleviated proinflammatory markers, modulated redox balance, and restored the intestinal epithelial barrier. In addition, the formulation increased the abundance of beneficial microbiota while decreasing the abundance of harmful microbiota. These results demonstrated that this biomimetic nanoplatform could be exploited as a safe and effective gut-targeted delivery system in IBD treatment.
Collapse
Affiliation(s)
- Xier Pan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Peng Xian
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yushu Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao Zhao
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiaxin Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yangjie Song
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yunrong Nan
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shuting Ni
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
49
|
Bu F, Chen K, Chen S, Jiang Y. Gut microbiota and intestinal immunity interaction in ulcerative colitis and its application in treatment. Front Cell Infect Microbiol 2025; 15:1565082. [PMID: 40292216 PMCID: PMC12031664 DOI: 10.3389/fcimb.2025.1565082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic, non-specific inflammatory bowel disease characterized by inflammation and injury of the colonic mucosa, exhibiting an increasing global incidence. Although research into UC pathogenesis is ongoing, the precise mechanisms remain to be fully elucidated. Studies indicate that UC development results from a complex interplay of factors, including genetic predisposition, environmental exposures, gut microbial dysbiosis, and immune dysregulation. Specifically, UC pathogenesis involves aberrant immune responses triggered by interactions between the host and gut microbiota. A complex, dynamic relationship exists between the microbial community and the host immune system throughout UC pathogenesis. Accumulating evidence suggests that changes in microbiota composition significantly impact gut immunity. This review will examine the intricate balance between the gut microbiota and mucosal immunity in UC progression and discuss potential therapeutic applications, providing a reference for further clinical treatment of this patient population.
Collapse
Affiliation(s)
| | | | - Siche Chen
- Department of Colorectal Surgery, Zhejiang Provincial People’s Hospital,
Affiliated People’s Hospital of Hangzhou Medical College, HangZhou, China
| | - Yi Jiang
- Department of Colorectal Surgery, Zhejiang Provincial People’s Hospital,
Affiliated People’s Hospital of Hangzhou Medical College, HangZhou, China
| |
Collapse
|
50
|
Park J, Wu Y, Le QV, Kim JS, Xu E, Lee J, Oh YK. Self-disassembling nanoparticles as oral nanotherapeutics targeting intestinal microenvironment. Nat Commun 2025; 16:3365. [PMID: 40204740 PMCID: PMC11982569 DOI: 10.1038/s41467-025-58513-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
Inspired by the survival strategies of pyomelanin-producing microbes, we synthesize pyomelanin nanoparticles (PMNPs) from homogentisic acid- γ-lactone via auto-oxidation and investigate their biomedical potential. PMNPs possess distinct physicochemical properties, including reactive oxygen species scavenging and microenvironment-responsive self-disassembly. Under intestinal conditions, PMNPs self-disassemble and penetrate the nanoscale pores of the mucin layer. In an inflammatory bowel disease model, orally administered PMNPs withstand gastric acidity and, in their solubilized form, interact with macrophages and epithelial cells. They significantly reduce reactive oxygen species levels, exert anti-inflammatory effects, and restore gut microbiota composition. Compared to conventional nanoparticles and 5-aminosalicylic acid, PMNPs exhibit greater therapeutic efficacy. Clinical symptoms and intestinal inflammation are alleviated, and the gut microbiota is restored to near-normal levels. These findings underscore the therapeutic potential of PMNPs for inflammatory bowel disease treatment and suggest broader biomedical applications.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Jung Suk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Enzhen Xu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy, Korea University, Sejong, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|