1
|
Liu Y, Chen N, He H, Liu L, Sun S. Sodium butyrate alleviates DSS-induced inflammatory bowel disease by inhibiting ferroptosis and modulating ERK/STAT3 signaling and intestinal flora. Ann Med 2025; 57:2470958. [PMID: 40028886 PMCID: PMC11878173 DOI: 10.1080/07853890.2025.2470958] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/14/2025] [Accepted: 02/06/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), can seriously impact patients' quality of life. Sodium butyrate (NaB), a product of dietary fiber fermentation, has been shown to alleviate IBD symptoms. Some studies have shown that it is related to ferroptosis. However, the precise mechanism linking NaB, IBD, and ferroptosis is not clear. OBJECTIVE This study aimed to demonstrate that NaB suppresses ferroptosis, thereby alleviating inflammatory bowel disease (IBD) through modulation of the extracellular regulated protein kinases/signal transducer and activator of transcription 3 (ERK/STAT3) signaling pathway and intestinal flora. METHODS An IBD model was established using 2.5% (w/v) dextran sulfate sodium (DSS). Mice were orally administered low-dose NaB, high-dose NaB , or 5-aminosalicylic acid (5-ASA). Ferroptosis-related molecules were measured using specific kits, and western blotting (WB) and real-time polymerase chain reaction (RT-qPCR) were used to determine the levels of the target molecules. RESULTS NaB alleviated symptoms in IBD mice, including reduced weight loss, prolonged colon length, reduced disease activity index (DAI), and reduced spleen index and mRNA expression of inflammatory factors. Additionally, NaB reduced the content of Fe2+ and myeloperoxidase (MPO) and increased the content of GSH and the activity of superoxide dismutase (SOD), which reflected NaB-inhibited ferroptosis. Moreover, western blotting showed that NaB enhanced STAT3 and ERK phosphorylation. In addition, NaB regulates the composition and functions of flora related to IBD. CONCLUSION NaB alleviates IBD by inhibiting ferroptosis and modulating ERK/STAT3 signaling and the intestinal flora.
Collapse
Affiliation(s)
- Yingyin Liu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Nachuan Chen
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Huaxing He
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Lulin Liu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Suxia Sun
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Rahaman MM, Wangchuk P, Sarker S. A systematic review on the role of gut microbiome in inflammatory bowel disease: Spotlight on virome and plant metabolites. Microb Pathog 2025; 205:107608. [PMID: 40250496 DOI: 10.1016/j.micpath.2025.107608] [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: 08/02/2024] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Inflammatory bowel diseases (IBD), including ulcerative colitis and Crohn's disease, arise from various factors such as dietary, genetic, immunological, and microbiological influences. The gut microbiota plays a crucial role in the development and treatment of IBD, though the exact mechanisms remain uncertain. Current research has yet to definitively establish the beneficial effects of the microbiome on IBD. Bacteria and viruses (both prokaryotic and eukaryotic) are key components of the microbiome uniquely related to IBD. Numerous studies suggest that dysbiosis of the microbiota, including bacteria, viruses, and bacteriophages, contributes to IBD pathogenesis. Conversely, some research indicates that bacteria and bacteriophages may positively impact IBD outcomes. Additionally, plant metabolites play a crucial role in alleviating IBD due to their anti-inflammatory and microbiome-modulating properties. This systematic review discusses the role of the microbiome in IBD pathogenesis and evaluates the potential connection between plant metabolites and the microbiome in the context of IBD pathophysiology.
Collapse
Affiliation(s)
- Md Mizanur Rahaman
- Biomedical Sciences and Molecular Biology, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, 4811, Australia
| | - Phurpa Wangchuk
- College of Science and Engineering, James Cook University, Nguma Bada campus, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Nguma Bada campus, McGregor Rd, Smithfield, Cairns, QLD, 4878, Australia
| | - Subir Sarker
- Biomedical Sciences and Molecular Biology, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, 4811, Australia.
| |
Collapse
|
3
|
Jori C, Ahmad A, Kumar A, Kumar B, Ali A, Ali N, Tabassum H, Khan R. Bioactive chitosan-BSA Maillard-derived chrysin-loaded nanoparticles: A gastroprotective, biomucoadhesive approach for enhanced oral therapy in ulcerative colitis. Carbohydr Polym 2025; 359:123537. [PMID: 40306769 DOI: 10.1016/j.carbpol.2025.123537] [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: 01/23/2025] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 05/02/2025]
Abstract
The current limitations of oral nanomedicines such as aminosalicylates, immunosuppressants, corticosteroids, and antibiotics include the toxic byproducts from nanocarrier synthesis, poor targeting and retention within the inflamed colon, delayed release at inflammation sites, susceptibility to gastric degradation, reduced efficacy under hypoxic conditions, MUC2 homeostasis disruption, and insufficiently addressing the disease's root causes. This research presents an innovative approach of using non-toxic, biodegradable, and biocompatible Maillard reaction-based nanoparticles (MPs) for targeted oral drug delivery in IBD therapy. Through the development of mucoadhevise chitosan-bovine serum albumin Maillard nanoparticles shielded with biocompatible, non-toxic, non-immunogenic, gastroprotective pectin (P@CMPs) encapsulating with chrysin, a flavonoid with anti-inflammatory and hyperoxia properties whose bioavailability is negatively affected by gastric degradation. P@CMPs had a spherical, uniform 300 nm hydrodynamic diameter, confirmed by TEM and FESEM. Chrysin encapsulation efficiency and loading capacity were ∼96 % and 16 %, respectively, demonstrating effective nanoparticle formulation The P@CMPs is designed to withstand the gastrointestinal environment, ensuring targeted delivery and prolonged retention in inflamed colonic regions. In a dextran sodium sulfate-induced colitis mouse model, P@CMPs markedly mitigated inflammation, suppressed proinflammatory cytokine levels, and augmented the expression of MUC2, a crucial factor for maintaining the integrity of the gut barrier. By employing non-toxic, biocompatible and biodegradable materials, our P@CMPs approach offers a promising avenue for advancing IBD treatment, addressing various challenges and precise oral delivery within the gastrointestinal system.
Collapse
Affiliation(s)
- Chandrashekhar Jori
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, Foothills Medical Centre, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Ajay Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Bhuvnesh Kumar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Aneesh Ali
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Heena Tabassum
- Division of Basic Medical Sciences, Indian Council of Medical Research, Government of India, V. Ramalingaswamy Bhawan, New Delhi 110029, India.
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge city, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India.
| |
Collapse
|
4
|
Eldesoqui M, Ali LS, Erfan OS, Dawood AF, Badawy AA, Ali SK, Mohammed ZA, Mahmoud AM, Embaby EM, El Nashar EM, Aldehri M, Zafrah H, Al-Zahrani NS, Soliman RHM. Dihydroartemisinin attenuates acetic acid-induced ulcerative colitis in rats: Suppression of inflammation and modulation of NFκβ/TNF-α/RIPK1-mediated necroptosis and apoptosis. Tissue Cell 2025; 94:102791. [PMID: 39978210 DOI: 10.1016/j.tice.2025.102791] [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: 01/11/2025] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is an inflammatory bowel disease characterized by the overproduction of reactive oxygen species (ROS) and the release of inflammatory mediators. Dihydroartemisinin (DHA) is a semi-synthetic active metabolite of artemisinin that has anti-inflammatory, antioxidant, and anti-fibrotic properties. OBJECTIVE This study aimed to assess the therapeutic benefits of DHA on acetic acid(AA) -induced UC in rats, with particular emphasis on its anti-inflammatory effects and its influence on NFκB/TNF-α/RIPK1 necroptotic pathways. METHODS Eighteen rats were allocated into control, acetic acid-induced colitis (AA), and DHA-treated (AA+DHA) groups. Colitis was caused by rectal instillation of 5 % acetic acid. DHA was supplied via intraperitoneal injection. Histological, biochemical studies of oxidative stress, inflammatory and anti-inflammatory mediators, Western blotting for TNF-α, RIPK1, and caspase 3, and immunohistochemical assessment of NFκB, TNF-α, and RIPK1, were conducted. RESULTS DHA treatment markedly diminished macroscopic damage, disease activity index, histopathology scores, and malondialdehyde (MDA) levels, enhancing glutathione (GSH) levels. Additionally, DHA decreased serum TNF-α and IL-6 and increased IL-10. Western blotting and immunohistochemistry investigations validated the reduced expression of TNF-α, RIPK1, and caspase 3 in DHA-treated rats. CONCLUSION DHA demonstrates protective properties against acetic acid-induced UC by decreasing oxidative stress and inflammation, modifying TNF-α activity to regulate apoptotic and necroptotic pathways. So, DHA may be a favorable therapeutic alternative for the management of ulcerative colitis.
Collapse
Affiliation(s)
- Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O.Box 71666, Riyadh 11597, Saudi Arabia; Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Lashin S Ali
- Department of Basic Medical Sciences, Faculty of Dentistry, Al-Ahliyya Amman University, Amman 19111, Jordan; Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Omnia S Erfan
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Amal F Dawood
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Abdelnaser A Badawy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar, Saudi Arabia.
| | - Sahar K Ali
- Department of clinical pharmacology, faculty of medicine, Zagazig university, Zagazig 44519, Egypt.
| | - Zeinab A Mohammed
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Alia Mohamed Mahmoud
- Forensic Medicine and Clinical Toxicology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Eman M Embaby
- Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Eman Mohamad El Nashar
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Postal code (62529), Saudi Arabia.
| | - Majed Aldehri
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Postal code (62529), Saudi Arabia.
| | - Hind Zafrah
- Department of Physiology, Faculty of Medicine, King Khalid University, Abha Postal code (62529), Saudi Arabia.
| | - Norah Saeed Al-Zahrani
- Department of Clinical Biochemistry, College of Medicine; King Khalid University, Abha, Postal code (62529), Saudi Arabia.
| | | |
Collapse
|
5
|
Yang QH, Zhang CN. Comparative study on the pathogenesis of Crohn’s disease and ulcerative colitis. World J Gastroenterol 2025; 31:106406. [DOI: 10.3748/wjg.v31.i19.106406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/25/2025] [Accepted: 04/25/2025] [Indexed: 05/21/2025] Open
Abstract
Inflammatory bowel disease (IBD) is an incurable disease of the digestive system; however, the therapeutic methods for IBD remain limited. The pathogenesis of IBD was systematically discussed and compared in this paper, primarily comprising Crohn’s disease and ulcerative colitis. This paper focused on six common aspects: (1) Dysregulated immune responses; (2) Gene function changes; (3) Intestinal microbes disorder and imbalance; (4) Microbial infections; (5) Associations between IBD and other inflammatory diseases; and (6) Other factors. In addition, the pathogenesis differences between these two forms of IBD were unraveled and clearly distinguished. These unique aspects of pathogenesis provide crucial insights for the precise treatment of both Crohn’s disease and ulcerative colitis. This paper illustrates the root causes and beneficial factors of resistance to IBD, which provides novel insights on early prevention, development of new therapeutic agents, and treatment options of this disease.
Collapse
Affiliation(s)
- Qi-Hang Yang
- Chinese Academy of Medical Science & Peking Union Medical College, Institute of Biomedical Engineering, Tianjin 300192, China
- University College London, Cancer Institute, London WC1E 6BT, United Kingdom
| | - Chuang-Nian Zhang
- Chinese Academy of Medical Science & Peking Union Medical College, State Key Laboratory of Advanced Medical Materials and Devices, Engineering Research Center of Pulmonary and Critical Care Medicine Technology and Device (Ministry of Education), Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Tianjin Institutes of Health Science, Tianjin 300192, China
| |
Collapse
|
6
|
Cosio D. Multimorbidity: Addressing the Elephant in the Clinic Room. Healthcare (Basel) 2025; 13:1202. [PMID: 40428038 PMCID: PMC12110903 DOI: 10.3390/healthcare13101202] [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: 03/21/2025] [Revised: 04/23/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Multimorbidity is the conjoint presence of multiple conditions in patients, which is a public health problem. Multimorbidity is like the elephant in the clinic room because it remains the unaddressed challenge we face in healthcare. Clinical health psychology has a role to play in this undertaking because it recognizes the intersection and interface of concurrent mental and/or behavioral problems and physical diseases. The current article will define multimorbidity, describe current statistics, how it differs from comorbidity, how to use the biopsychosocial model, and ways in which clinical health psychologists can manage and prevent it in their clinics. A model of how to address multimorbidity will be shared using the role of a clinical health psychologist working in a multidisciplinary pain clinic in a hospital setting serving patients who are socioeconomically disadvantaged.
Collapse
Affiliation(s)
- David Cosio
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
7
|
Qin L, Hu C, Zhao Q, Wang Y, Fan D, Lin A, Xiang L, Chen Y, Shao J. Unraveling the role of Ctla-4 in intestinal immune homeostasis through a novel Zebrafish model of inflammatory bowel disease. eLife 2025; 13:RP101932. [PMID: 40392591 PMCID: PMC12092003 DOI: 10.7554/elife.101932] [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] [Indexed: 05/22/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and relapsing immune-mediated disorder characterized by intestinal inflammation and epithelial injury. The underlying causes of IBD are not fully understood, but genetic factors have been implicated in genome-wide association studies, including CTLA-4, an essential negative regulator of T cell activation. However, establishing a direct link between CTLA-4 and IBD has been challenging due to the early lethality of CTLA-4 knockout mice. In this study, we identified zebrafish Ctla-4 homolog and investigated its role in maintaining intestinal immune homeostasis by generating a Ctla-4-deficient (ctla-4-/-) zebrafish line. These mutant zebrafish exhibited reduced weight, along with impaired epithelial barrier integrity and lymphocytic infiltration in their intestines. Transcriptomics analysis revealed upregulation of inflammation-related genes, disturbing immune system homeostasis. Moreover, single-cell RNA-sequencing analysis indicated increased Th2 cells and interleukin 13 expression, along with decreased innate lymphoid cells and upregulated proinflammatory cytokines. Additionally, Ctla-4-deficient zebrafish exhibited reduced diversity and an altered composition of the intestinal microbiota. All these phenotypes closely resemble those found in mammalian IBD. Lastly, supplementation with Ctla-4-Ig successfully alleviated intestinal inflammation in these mutants. Altogether, our findings demonstrate the pivotal role of Ctla-4 in maintaining intestinal homeostasis. Additionally, they offer substantial evidence linking CTLA-4 to IBD and establish a novel zebrafish model for investigating both the pathogenesis and potential treatments.
Collapse
Affiliation(s)
- Lulu Qin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Chongbin Hu
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Qiong Zhao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Yong Wang
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Dongdong Fan
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Aifu Lin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Lixin Xiang
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Ye Chen
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
- Department of Genetic and Metabolic Disease, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouChina
| | - Jianzhong Shao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Shi L, Chen L, Jin G, Yang Y, Zhu F, Zhou G. Si-Ni Decoction as a Potential Treatment for Ulcerative Colitis: Modulation of Gut Microbiota and AKT1 Inhibition Through Network Pharmacology and in vivo Validation. J Inflamm Res 2025; 18:6263-6280. [PMID: 40391232 PMCID: PMC12087465 DOI: 10.2147/jir.s516556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/26/2025] [Indexed: 05/21/2025] Open
Abstract
Background Sini Decoction (SND), a time-honored formulation in traditional Chinese medicine, consists of three key ingredients: aconite, licorice, and ginger rhizome. It has been used for more than a thousand years to relieve symptoms associated with acute gastroenteritis, dyspepsia, and abdominal discomfort, but its therapeutic efficacy in ulcerative colitis (UC) and the mechanisms involved have not been validated. In this study, a comprehensive approach integrating network pharmacology, molecular docking, molecular dynamics simulation and experimentation was used to assess the efficacy of SND in the treatment of UC and to explore its molecular mechanisms. Methods The bioactive compounds associated with ulcerative colitis (UC) were identified using the TCMSP database, with potential targets predicted via the Swiss Target Prediction database. Protein-protein interaction networks were constructed using the STRING database and Cytoscape and the most important genes were identified. Subsequently, molecular docking was combined with molecular dynamics simulations using molecular docking to assess the binding affinity of the main active ingredient of SND to AKT1. To evaluate the therapeutic effects of SND, we utilized a dextran sodium sulfate-induced UC mouse model. Additionally, fecal samples were collected for analysis of the intestinal microbiota to explore the influence of SND on gut flora composition. Results Fifteen bioactive components from SND were identified, and their activities were validated. The results indicated that AKT serine/threonine kinase 1 is a core target of SND for the treatment of UC. The anti-inflammatory, intestinal barrier-protective, and microbiota-regulating effects of SND were confirmed in animal models, alongside evidence of its inhibitory effect on AKT1. Conclusion The active ingredients of SND were screened, with a focus on AKT1 inhibition, to reduce inflammation in UC, protect the intestinal barrier, and regulate the intestinal microbiota, demonstrating significant therapeutic potential.
Collapse
Affiliation(s)
- Lihao Shi
- Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Leilei Chen
- Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Guiyuan Jin
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, People’s Republic of China
| | - Yonghong Yang
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, People’s Republic of China
| | - Fengqin Zhu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining, People’s Republic of China
| | - Guangxi Zhou
- Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining, People’s Republic of China
| |
Collapse
|
11
|
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
|
12
|
Zhai J, Li Y, Liu J, Dai C. Neuroimmune interactions: The bridge between inflammatory bowel disease and the gut microbiota. Clin Transl Med 2025; 15:e70329. [PMID: 40400119 PMCID: PMC12095209 DOI: 10.1002/ctm2.70329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND The multidimensional regulatory mechanism of the gut-brain-immune axis in the context of inflammatory bowel disease (IBD) has garnered significant attention, particularly regarding how intestinal microbiota finely regulates immune responses through immune cells and sensory neurons. MAIN BODY Metabolites produced by intestinal microbiota influence the phenotype switching of immune cells via complex signalling pathways, thereby modulating their anti-inflammatory and pro-inflammatory functions during intestinal inflammation. Furthermore, sensory neurons exhibit heightened sensitivity to microbial-derived signals, which is essential for preserving intestinal balance and controlling pathological inflammation by integrating peripheral environmental signals with local immune responses. The dynamic equilibrium between immune cells and the neuroimmunoregulation mediated by sensory neurons collectively sustains immune homeostasis within the intestine. However, this coordination mechanism is markedly disrupted under the pathological conditions associated with IBD. CONCLUSION An in-depth exploration of the interactions among immune cells, gut microbiota and sensory neurons may yield significant insights into the pathological mechanisms underlying IBD and guide the creation of new treatment approaches. KEY POINTS The gut microbiota regulates the gut-brain-immune axis, modulating neuroimmune interactions in IBD. Microbiota-derived metabolites influence immune cells, thereby affecting neurons. Neurons secrete mediators, enabling bidirectional neuroimmune communication essential for intestinal homeostasis. Disruptions contribute to IBD, offering therapeutic targets.
Collapse
Affiliation(s)
- Jinxia Zhai
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| | - Yingjie Li
- Department of GastroenterologyFirst Affiliated Hospital, Jinzhou Medical UniversityJinzhou CityLiaoning ProvinceChina
| | - Jiameng Liu
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| | - Cong Dai
- Department of GastroenterologyFirst Affiliated Hospital, China Medical UniversityShenyang CityLiaoning ProvinceChina
| |
Collapse
|
13
|
Malik S, Naqvi SAA, Shadali AH, Khan H, Christof M, Niu C, Schwartz DA, Adler DG. Fecal Microbiota Transplantation (FMT) and Clinical Outcomes Among Inflammatory Bowel Disease (IBD) Patients: An Umbrella Review. Dig Dis Sci 2025; 70:1873-1896. [PMID: 40038211 DOI: 10.1007/s10620-025-08946-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 02/19/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND AND AIMS Recent systematic reviews and meta-analyses (SRMAs) have shown inconsistent effectiveness of FMT among patients with IBD. This study aimed to appraise the evidence for clinically relevant outcomes with FMT in patients with IBD using published SRMAs. METHODS We searched major databases from inception through Nov 2023 to identify SRMAs assessing the effectiveness of FMT in patients with IBD. Primary outcomes included clinical remission, clinical response, endoscopic remission/response, a composite endpoint, and adverse effects. We included SRMAs investigating FMT's effect in patients with IBD using RCTs and observational studies data. Methodological quality and evidence certainty were assessed using AMSTAR 2 and GRADE. RESULTS Out of 106 citations, 16 SRMAs were included with varying study sizes (2 to 60 primary studies) and participants (112 to 1169 per SRMA). Five SRMAs assessed FMT in IBD, while 11 focused on Ulcerative Colitis (UC). Seven SRMAs included RCTs only, and nine included both RCTs and observational studies. Methodological quality was critically low in 9 SRMAs (56%) and low in 7 studies (44%). FMT showed clinical remission benefit in all 16 SRMAs, with varying certainty: 3 high, 4 moderate, 4 low, and 5 very low. Endoscopic remission/response was reported in 5 meta-analyses on UC, with 1 high, 3 moderate, and 1 very low certainty. Combined clinical remission and endoscopic response were reported in 3 SRMAs on UC, with 1 low and 2 moderate certainty. Adverse events were reported in 6 SRMAs, with 1 high, 3 moderate, 1 low, and 1 very low certainty. CONCLUSION Current evidence shows potential benefits of FMT in IBD, particularly UC, supported by significant associations in 16 meta-analyses. However, poor methodological quality and variability in evidence certainty call for high-quality RCTs to strengthen the evidence.
Collapse
Affiliation(s)
- Sheza Malik
- Internal Medicine, Rochester General Hospital, Rochester, NY, USA
| | | | | | - Hajra Khan
- Rawalpindi Medical College, Rawalpindi, Pakistan
| | | | - Chengu Niu
- Internal Medicine, Rochester General Hospital, Rochester, NY, USA
| | - David A Schwartz
- Gastroenterology and Hepatology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas G Adler
- Gastroenterology and Hepatology, Porter Adventist Hospital in Denver, Denver, CO, USA.
| |
Collapse
|
14
|
Hua S, Zhang Z, Zhang Z, Liu L, Yu S, Xiao Y, Liu Y, Wei S, Xu Y, Chen YG. Genetic disruption of the circadian gene Bmal1 in the intestinal epithelium reduces colonic inflammation. EMBO Rep 2025:10.1038/s44319-025-00464-y. [PMID: 40307620 DOI: 10.1038/s44319-025-00464-y] [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: 09/07/2024] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 05/02/2025] Open
Abstract
Disruption of the circadian clock is associated with the development of inflammatory bowel disease (IBD), but the underlying mechanisms remain unclear. Here, we observe that mice in the early active phase (Zeitgeber time 12, ZT12) of the circadian clock are more tolerant to dextran sodium sulfate (DSS)-induced colitis, compared to those in the early resting phase (ZT0). The expression of the circadian gene Bmal1 peaks in the early resting phase and declines in the early active phase. Bmal1 knockout in the intestinal epithelium reduces DSS-induced inflammatory symptoms. Mechanistically, BMAL1 promotes apoptosis by binding to apoptosis-related genes, including Bax, p53, and Bak1, and promotes their expression. Intriguingly, we observe circadian apoptotic rhythms in the homeostatic intestinal epithelium, while Bmal1 deletion reduces cell apoptosis. Consistently, reducing Bmal1 expression by the REV-ERBα agonist SR9009 has the best therapeutic efficacy against DSS-induced colitis at ZT0. Collectively, our data demonstrate that the Bmal1-centered circadian clock is involved in intestinal injury repair.
Collapse
Affiliation(s)
- Shan Hua
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Ze Zhang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Zhe Zhang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Liansheng Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Shicheng Yu
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Yanhui Xiao
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Yuan Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Siting Wei
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Ying Xu
- Cambridge-Su Genomic Resource Center, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ye-Guang Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
15
|
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
|
16
|
Li J, Geng Z, Yin L, Huang J, Niu M, Zhang K, Song X, Wang Y, Zuo L, Hu J. Engeletin Targets Mitochondrial Dysfunction to Attenuate Oxidative Stress and Experimental Colitis in Intestinal Epithelial Cells Through AMPK/SIRT1/PGC-1α Signaling. Antioxidants (Basel) 2025; 14:524. [PMID: 40427406 PMCID: PMC12108241 DOI: 10.3390/antiox14050524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease and ulcerative colitis, is characterized by chronic intestinal inflammation and epithelial barrier disruption. Emerging evidence highlights mitochondrial dysfunction as a pivotal contributor to IBD pathogenesis, where impaired mitochondrial homeostasis in intestinal epithelial cells (IECs) disrupts redox balance, exacerbates oxidative stress, and triggers apoptosis, further compromising barrier integrity. This study investigated the therapeutic effects of Engeletin (Eng), a dihydroflavonoid from Smilax glabra Roxb., in dextran sulfate sodium (DSS)-induced colitis mice and colonic organoid models. Eng administration (10, 20, 40 mg/kg) significantly alleviated colitis symptoms, including weight loss, disease activity index (DAI) scores, and colon shortening, while restoring intestinal barrier integrity through the upregulation of tight junction proteins (ZO-1, claudin-1) and goblet cell preservation. Eng suppressed NF-κB-mediated inflammation and activated the Nrf2 antioxidant pathway, as well as reduced oxidative stress markers (MDA, CAT, GSH, and SOD). It attenuated epithelial apoptosis by balancing pro- and anti-apoptotic proteins (Bax/Bcl2, c-caspase3) and ameliorated mitochondrial dysfunction via enhanced ATP production, mtDNA levels, and complex I/IV activity. Mechanistically, Eng activated the AMPK/SIRT1/PGC-1α axis, and pharmacological inhibition of PGC-1α abolished its mitochondrial protective and anti-apoptotic effects. These findings demonstrate that Eng alleviates colitis by targeting mitochondrial homeostasis and oxidative stress through AMPK/SIRT1/PGC-1α signaling, offering a multitargeted strategy for IBD therapy.
Collapse
Affiliation(s)
- Jing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China; (J.L.); (L.Y.); (K.Z.); (Y.W.)
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, Bengbu 233004, China; (Z.G.); (J.H.); (M.N.); (X.S.); (L.Z.)
| | - Zhijun Geng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, Bengbu 233004, China; (Z.G.); (J.H.); (M.N.); (X.S.); (L.Z.)
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - Lixia Yin
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China; (J.L.); (L.Y.); (K.Z.); (Y.W.)
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, Bengbu 233004, China; (Z.G.); (J.H.); (M.N.); (X.S.); (L.Z.)
| | - Ju Huang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, Bengbu 233004, China; (Z.G.); (J.H.); (M.N.); (X.S.); (L.Z.)
| | - Minzhu Niu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, Bengbu 233004, China; (Z.G.); (J.H.); (M.N.); (X.S.); (L.Z.)
| | - Keni Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China; (J.L.); (L.Y.); (K.Z.); (Y.W.)
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, Bengbu 233004, China; (Z.G.); (J.H.); (M.N.); (X.S.); (L.Z.)
| | - Xue Song
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, Bengbu 233004, China; (Z.G.); (J.H.); (M.N.); (X.S.); (L.Z.)
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - Yueyue Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China; (J.L.); (L.Y.); (K.Z.); (Y.W.)
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, Bengbu 233004, China; (Z.G.); (J.H.); (M.N.); (X.S.); (L.Z.)
| | - Lugen Zuo
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, Bengbu 233004, China; (Z.G.); (J.H.); (M.N.); (X.S.); (L.Z.)
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China
| | - Jianguo Hu
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu 233004, China; (J.L.); (L.Y.); (K.Z.); (Y.W.)
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, Bengbu 233004, China; (Z.G.); (J.H.); (M.N.); (X.S.); (L.Z.)
| |
Collapse
|
17
|
Weinhaeusel A, Huber J, Schoenthaler S, Beigel F, Noehammer C, Vierlinger K, Siebeck M, Gropp R. Autoantibody Profiling in Ulcerative Colitis: Identification of Early Immune Signatures and Disease-Associated Antigens for Improved Diagnosis and Monitoring. Int J Mol Sci 2025; 26:4086. [PMID: 40362323 PMCID: PMC12072058 DOI: 10.3390/ijms26094086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/17/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Ulcerative colitis (UC) is a major form of inflammatory bowel disease (IBD) characterised by chronic immune-mediated inflammation. While serological biomarkers for IBD diagnosis and differentiation have been explored, autoantibody-based profiling remains underdeveloped. This study aimed to elucidate antibody signatures in manifested and pre-diagnostic UC patients compared to controls using a high-content protein microarray. Serum and plasma samples from manifested and pre-diagnostic UC cohorts were analysed using AIT's 16k protein microarray, presenting 6369 human proteins. The pre-diagnostic cohort, consisting of 33 UC cases and 33 controls, included longitudinal samples collected before diagnosis, while the severe UC cohort, comprising 49 severe UC patients and 23 controls, included individuals undergoing treatment. Immunoglobulin G (IgG) autoantibody reactivity was assessed to identify differentially reactive antigens (DIRAGs) linked to UC onset, disease progression, and activity. In manifested UC, 691 DIRAGs showed higher reactivity in cases. In the pre-diagnostic cohort, 966 DIRAGs were identified, with 803 antigens exhibiting increased reactivity in cases. Longitudinal analysis revealed 1371 DIRAGs, with 1185 showing increased reactivity closer to diagnosis when comparing samples collected 4-11 months before UC diagnosis to earlier time points 9-24 months prior, highlighting potential early biomarkers. A significant overlap of 286 antigens, corresponding to 41 percent of identified DIRAGs, was observed between severe and pre-diagnostic UC datasets, with an odds ratio of 3.8 and a p-value below 2.2 × 10-16, confirming reliability and biological relevance. Additionally, 21 antigens correlated with simple clinical colitis activity index (SCCAI) scores. Reactome pathway analysis identified 49 pathways associated with DIRAGs in pre-diagnostic UC, distinct from 24 pathways in manifested UC, with an overlap of five key pathways related to protein folding, immune regulation, and viral infection, reflecting differences in disease onset and manifestation. Autoantibody profiling reveals early immune signatures in UC, offering novel biomarkers for preclinical diagnosis and disease monitoring. The overlap between pre-diagnostic and manifested UC antigenic profiles reinforces their biological relevance, linking them to molecular pathology. These findings highlight antibody profiling as an additional omics layer, paving the way for new diagnostic and therapeutic strategies in UC management.
Collapse
Affiliation(s)
- Andreas Weinhaeusel
- Austrian Institute of Technology GmbH (AIT), Giefinggasse, 1210 Vienna, Austria; (J.H.); (S.S.); (C.N.); (K.V.)
| | - Jasmin Huber
- Austrian Institute of Technology GmbH (AIT), Giefinggasse, 1210 Vienna, Austria; (J.H.); (S.S.); (C.N.); (K.V.)
| | - Silvia Schoenthaler
- Austrian Institute of Technology GmbH (AIT), Giefinggasse, 1210 Vienna, Austria; (J.H.); (S.S.); (C.N.); (K.V.)
| | - Florian Beigel
- Department of Medicine II, Hospital of the Ludwig-Maximilian University Munich, 81377 Munich, Germany;
| | - Christa Noehammer
- Austrian Institute of Technology GmbH (AIT), Giefinggasse, 1210 Vienna, Austria; (J.H.); (S.S.); (C.N.); (K.V.)
| | - Klemens Vierlinger
- Austrian Institute of Technology GmbH (AIT), Giefinggasse, 1210 Vienna, Austria; (J.H.); (S.S.); (C.N.); (K.V.)
| | - Matthias Siebeck
- Department of General, Visceral und Transplantation Surgery, Hospital of the Ludwig-Maximilian University Munich, 80336 Munich, Germany; (M.S.); (R.G.)
| | - Roswitha Gropp
- Department of General, Visceral und Transplantation Surgery, Hospital of the Ludwig-Maximilian University Munich, 80336 Munich, Germany; (M.S.); (R.G.)
| |
Collapse
|
18
|
Costa M, Pottier M, Jacob M, Zarnitzky P, Segain B, Figeac M, Sebda S, Leprêtre F, Meresse B, Demaret J, Foligné B, Standaert A, Bertin B. Relevance of mouse and human IBD patient-derived colon organoids to investigate intestinal macrophage differentiation. J Leukoc Biol 2025; 117:qiaf004. [PMID: 39832522 DOI: 10.1093/jleuko/qiaf004] [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: 07/04/2024] [Revised: 10/09/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
The gastrointestinal tract is a remarkable example of complex biology, with a constant dialogue between the intestinal epithelium, in close contact with the microbiota, and the immune cells that protect the gut from infection. Organoids have revolutionized our approach to modeling the intestinal cellular compartment and have opened new avenues for unraveling the mechanisms involved in intestinal homeostasis and chronic pathogenesis, such as inflammatory bowel disease. To date, few models have been established to explore the role of the colon, which is, however, the main site of inflammation in ulcerative colitis. Here, we used conditioned media produced by colon organoids from mice or humans (control patients and patients with ulcerative colitis) to investigate the relationship between macrophages and the colon epithelium. We addressed transcriptomic profiles of organoid conditioned media-stimulated bone marrow-derived macrophages and found that these cells exhibited a unique anti-inflammatory signature distinct from that of conventional in vitro IL-4/IL-13 M2-differentiated macrophages. In addition, organoid conditioned media induced a clear CD5 antigen-like-mediated immunoregulatory effect characterized by a significant reduction in lipopolysaccharide-induced inducible nitric oxide synthase expression. In line, organoid conditioned media from human colons inhibited lipopolysaccharide-dependent inflammatory cytokine expression in human monocyte-derived macrophages. Interestingly, the inflammatory marker CD68 was reduced by organoid conditioned media from control patients but not from patients with ulcerative colitis, suggesting epithelial dysfunction in patients with ulcerative colitis. Our results report new regulatory mechanisms in the colon and highlight the importance of developing new in vitro models to better characterize the relationship between the intestinal epithelium and immune mucosal cells.
Collapse
Affiliation(s)
- Maxime Costa
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Muriel Pottier
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Marie Jacob
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Pauline Zarnitzky
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Benjamin Segain
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Martin Figeac
- Plateau de Génomique Fonctionnelle et Structurale, CHU Lille, Univ. Lille, Lille France
| | - Shéhérazade Sebda
- Plateau de Génomique Fonctionnelle et Structurale, CHU Lille, Univ. Lille, Lille France
| | - Frédéric Leprêtre
- Plateau de Génomique Fonctionnelle et Structurale, CHU Lille, Univ. Lille, Lille France
| | - Bertrand Meresse
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Julie Demaret
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Benoit Foligné
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Annie Standaert
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Benjamin Bertin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| |
Collapse
|
19
|
Zhang W, Jing X, Li B, Wu X. Clearance of Cell-Free DNA: A Novel Target for Therapeutic Utilization in Multiple Systemic Disorders. ACS Biomater Sci Eng 2025; 11:2069-2079. [PMID: 40178087 DOI: 10.1021/acsbiomaterials.5c00049] [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: 04/05/2025]
Abstract
Cell-free DNA (cfDNA) holds significant promise for diagnostic and therapeutic advancements in medicine. This review delineates the utility of cfDNA in diagnostics and its therapeutic potential through clearance mechanisms for an array of diseases. Damage-associated molecular patterns (DAMPs) are endogenous molecules released by host cells during stress, or injury. As a trigger for inflammatory responses via damage-associated molecular patterns (DAMPs), cfDNA's removal via nanotechnological approaches can attenuate inflammation and promote tissue repair. While the application of cfDNA clearance is particularly auspicious in cancer, sepsis, and inflammatory conditions, it is confronted with challenges including toxicity, specificity, and the rigors of clinical trial validation. Collectively, this review delineates novel therapeutic targets to inform the development of innovative treatment strategies.
Collapse
Affiliation(s)
- Wenjun Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi030001, China
| | - Xuan Jing
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi030001, China
| | - Bing Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi030001, China
| | - Xiuping Wu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, Shanxi030001, China
| |
Collapse
|
20
|
Ma Z, McAninch S, Liu Z, Zhang C, Chen H, He J, Yang W, Panganiban RP, Cong Y, Yochum G, Brasier AR, Pinchuk IV, Tian B, Zhou J. Orally Bioavailable BRD4 BD1 Inhibitor ZL0516 Effectively Suppresses Colonic Inflammation in Animal Models of Inflammatory Bowel Disease. ACS Pharmacol Transl Sci 2025; 8:1152-1167. [PMID: 40242579 PMCID: PMC11997885 DOI: 10.1021/acsptsci.5c00068] [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: 01/22/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025]
Abstract
Inflammatory bowel disease (IBD), a chronic, progressive, and recurrent gastrointestinal inflammatory disorder, poses a significant threat to global health and exerts an adverse effect on the quality of life. Currently, there is a lack of effective therapies for IBD. Developing novel targeted therapies for IBD, particularly orally effective therapeutics, is a vital need for IBD patients. Herein, we first demonstrate that BRD4/NF-κB signaling is aberrantly activated in the colons of human IBD biopsy samples compared to that of normal healthy controls. ZL0516, a potent, selective, and orally bioavailable BRD4 BD1 inhibitor, significantly inhibits the TNFα- and LPS-induced expression of inflammatory cytokines in human colonic epithelial cells (HCECs) and peripheral blood mononuclear cells (PBMCs) with low cytotoxicity. Intriguingly, when administered in a preventive mode, ZL0516 significantly blocks dextran sulfate sodium (DSS)-induced murine colitis. When used in a therapeutic mode, ZL0516 effectively suppresses colonic inflammation in several IBD-relevant animal models: DSS-, oxazolone (OXA)-, and flagellin (Cbir1) T cell-induced chronic murine colitis models of IBD. ZL0516 suppresses IBD inflammatory responses in vitro and in vivo by blocking the activation of the BRD4/NF-κB signaling pathway. Also, we found that RVX208, a selective BRD4 BD2 inhibitor in Phase III clinical development, only displayed marginal effects in these IBD animal models. Collectively, our results demonstrate that specific BRD4 BD1 inhibition is a novel therapeutic strategy for IBD-associated colonic inflammation, and orally effective inhibitor ZL0516 is a promising candidate for the development of a novel therapeutic regimen against IBD.
Collapse
Affiliation(s)
- Zonghui Ma
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Steven McAninch
- Department
of Medicine, Penn State Health Milton S.
Hershey Medical Center, Hershey, Pennsylvania 17033, United States
| | - Zhiqing Liu
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Cun Zhang
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Haiying Chen
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Jing He
- Department
of Pathology, University of Texas Medical
Branch (UTMB), Galveston, Texas 77555, United States
| | - Wenjing Yang
- Division
of Gastroenterology and Hepatology, Department of Medicine, Feinberg
School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Ronaldo P. Panganiban
- Department
of Medicine, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Yingzi Cong
- Division
of Gastroenterology and Hepatology, Department of Medicine, Feinberg
School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Gregory Yochum
- Division
of Colon and Rectal Surgery, Department of Surgery, and Department
of Biochemistry and Molecular Biology, Penn
State Milton S. Hershey Medical Center, Hershey, Pennsylvania 17033, United States
| | - Allan R. Brasier
- Institute
for Clinical and Translational Research (ICTR) School of Medicine
and Public Health, 4248 Health Sciences Learning Center, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Irina V. Pinchuk
- Department
of Medicine, Penn State Health Milton S.
Hershey Medical Center, Hershey, Pennsylvania 17033, United States
| | - Bing Tian
- Department
of Internal Medicine, University of Texas
Medical Branch (UTMB), Galveston, Texas 77555, United States
| | - Jia Zhou
- Chemical
Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch (UTMB), Galveston, Texas 77555, United States
| |
Collapse
|
21
|
Cao Y, Xiao S, Fang Y, Yang J, Hu Z, Zhang H, Liu X, Liu D, Zhou Z, Wang P. Fluxapyroxad induces chronic colonic inflammation via inhibiting intestinal aryl hydrocarbon receptors in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 973:179134. [PMID: 40112552 DOI: 10.1016/j.scitotenv.2025.179134] [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: 02/13/2025] [Revised: 03/11/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Fluxapyroxad, the most extensively utilized succinate dehydrogenase inhibitor (SDHI) fungicide, lacks comprehensive research on potential risks associated with chronic toxicity. To investigate its effects on chronic colonic inflammation and elucidate the underlying mechanisms, a mouse model was employed to assess oral exposure to fluxapyroxad at no observed adverse effect level (NOEL) for 13 weeks, in vitro and in silico models were utilized as well. The results revealed reduced body weight gain, colon length reduction, crypt damage, goblet cell loss in the colon, impaired intestinal barrier integrity, and an elevation of proinflammatory cytokines, including IL-6, IL-1β, and TNF-α following fluxapyroxad exposure in mice. These findings suggested that fluxapyroxad induced chronic colonic inflammation. Furthermore, fluxapyroxad decreased interleukin 22 levels and antibacterial peptide secretion by inhibiting Aryl hydrocarbon receptors (AhR) activation, which was confirmed in vitro experiments. Molecular docking analysis indicated that fluxapyroxad spontaneously formed halogen bonds and bound hydrophobic interactions with AhR, which might act as an AhR inhibitor. These results indicated that AhR inhibition may represent one of the primary mechanisms for chronic colonic inflammation induced by fluxapyroxad exposure. This study shed light on the association between low acute pesticide exposure to fluxapyroxad and chronic colonic inflammation development while contributing to pesticide safety assessment.
Collapse
Affiliation(s)
- Yue Cao
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Shouchun Xiao
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Yaofeng Fang
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China
| | - Jiaxing Yang
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Zeyu Hu
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China
| | - Hongjun Zhang
- Institute for the Control of Agrochemicals, Ministry of Agriculture and Rural Affairs, China, No. 22 Maizidian Street, Chaoyang, Beijing 100125, PR China.
| | - Xueke Liu
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Donghui Liu
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Zhiqiang Zhou
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Peng Wang
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| |
Collapse
|
22
|
Campisi D, Hawkins N, Bonjour K, Wollert T. The Role of WIPI2, ATG16L1 and ATG12-ATG5 in Selective and Nonselective Autophagy. J Mol Biol 2025:169138. [PMID: 40221132 DOI: 10.1016/j.jmb.2025.169138] [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: 12/05/2024] [Revised: 02/24/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Autophagy is a conserved cellular recycling pathway that delivers damaged or superfluous cytoplasmic material to lysosomes for degradation. In response to cytotoxic stress or starvation, autophagy can also sequester bulk cytoplasm and deliver it to lysosomes to regenerate building blocks. In macroautophagy, a membrane cisterna termed phagophore that encloses autophagic cargo is generated. The formation of the phagophore depends on a conserved machinery of autophagy related proteins. The phosphatidylinositol(3)-phosphate binding protein WIPI2 facilitates the transition from phagophore initiation to phagophore expansion by recruiting the ATG12-ATG5-ATG16L1 complex to phagophores. This complex functions as an E3-ligase to conjugate ubiquitin-like ATG8 proteins to phagophore membranes, which promotes tethering of cargo to phagophore membranes, phagophore expansion, maturation and the fusion of autophagosomes with lysosomes. ATG16L1 also has important functions independently of ATG12-ATG5 in autophagy and beyond. In this review, we will summarize the functions of WIPI2 and ATG16L1 in selective and nonselective autophagy.
Collapse
Affiliation(s)
- Daniele Campisi
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - N'Toia Hawkins
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - Kennedy Bonjour
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France
| | - Thomas Wollert
- Membrane Biochemistry and Transport, Institut Pasteur, Université de Paris, UMR3691 CNRS, 75015 Paris, France.
| |
Collapse
|
23
|
Zhang Y, Zhao Q, Wu Z, Chen N, Li N, Liu J, Zhang M, Li S, Chi Y, Liu Y. Mesenteric Lymphatic B Cells Migrate to the Gut and Aggravate TNBS-Induced Rat Colitis via Regulating Intestinal T Cells. Int J Mol Sci 2025; 26:3519. [PMID: 40331987 PMCID: PMC12026553 DOI: 10.3390/ijms26083519] [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: 02/06/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Inflammatory bowel disease (IBD), comprising Crohn's disease (CD) and ulcerative colitis (UC), is affecting a growing global population. Unlike UC, which is characterized by inflammation confined to the intestinal mucosa and submucosa, CD involves transmural inflammation of the intestine. Although the lymphatic system is believed to play a role in the pathogenesis of CD, its exact contribution remains poorly understood. Mesenteric lymphatics (MLs), which drain interstitial fluid and immune cells into mesenteric lymph nodes, have been implicated in this process. In the present study, we aimed to investigate the role of ML immune cells in TNBS-induced colitis in rats. Flow cytometry analysis revealed an increased ratio of B cells and altered B cell function in the MLs of colitis rats compared to controls. The adoptive transfer of mesenteric lymphatic B (MLB) cells isolated from colitis rats to recipient rats exacerbated colitis and was associated with the enhanced migration of MLB cells to the gut. RNA sequencing analysis demonstrated a significant upregulation of genes associated with inflammation and immune responses in MLB cells from colitis rats, particularly key molecules involved in T cell activation, such as cluster of differentiation 27 (Cd27) and cluster of differentiation 40 (Cd40), and the chemotactic receptor C-C motif chemokine receptor 8 (Ccr8), which mediates B cell migration in response to T cells. Mechanistically, MLB cells from colitis rats were recruited to the colon by intra-intestinal T cells through the Ccr8-C-C motif chemokine ligand 1 (Ccl1) axis, where they subsequently exacerbated inflammatory responses via enhanced differentiation. These observations indicate that the migration of MLB cells to the gut exacerbates TNBS-induced colitis in rats by modulating intestinal T cells.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Q.Z.); (Z.W.); (N.C.)
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing 100044, China
| | - Qinghe Zhao
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Q.Z.); (Z.W.); (N.C.)
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing 100044, China;
| | - Zhe Wu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Q.Z.); (Z.W.); (N.C.)
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing 100044, China
| | - Ning Chen
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Q.Z.); (Z.W.); (N.C.)
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing 100044, China
| | - Na Li
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing 100044, China;
| | - Jiao Liu
- Center of Medical and Health Analysis, Peking University Health Science Center, Beijing 100191, China;
| | - Menglei Zhang
- Department of Animal Laboratory, Peking University People’s Hospital, Beijing 100044, China; (M.Z.); (S.L.)
| | - Shuolei Li
- Department of Animal Laboratory, Peking University People’s Hospital, Beijing 100044, China; (M.Z.); (S.L.)
| | - Yujing Chi
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Q.Z.); (Z.W.); (N.C.)
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People’s Hospital, Beijing 100044, China;
| | - Yulan Liu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing 100044, China; (Y.Z.); (Q.Z.); (Z.W.); (N.C.)
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing 100044, China
| |
Collapse
|
24
|
Xue C, Chu Q, Shi Q, Zeng Y, Lu J, Li L. Wnt signaling pathways in biology and disease: mechanisms and therapeutic advances. Signal Transduct Target Ther 2025; 10:106. [PMID: 40180907 PMCID: PMC11968978 DOI: 10.1038/s41392-025-02142-w] [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: 08/19/2024] [Revised: 11/13/2024] [Accepted: 12/29/2024] [Indexed: 04/05/2025] Open
Abstract
The Wnt signaling pathway is critically involved in orchestrating cellular functions such as proliferation, migration, survival, and cell fate determination during development. Given its pivotal role in cellular communication, aberrant Wnt signaling has been extensively linked to the pathogenesis of various diseases. This review offers an in-depth analysis of the Wnt pathway, detailing its signal transduction mechanisms and principal components. Furthermore, the complex network of interactions between Wnt cascades and other key signaling pathways, such as Notch, Hedgehog, TGF-β, FGF, and NF-κB, is explored. Genetic mutations affecting the Wnt pathway play a pivotal role in disease progression, with particular emphasis on Wnt signaling's involvement in cancer stem cell biology and the tumor microenvironment. Additionally, this review underscores the diverse mechanisms through which Wnt signaling contributes to diseases such as cardiovascular conditions, neurodegenerative disorders, metabolic syndromes, autoimmune diseases, and cancer. Finally, a comprehensive overview of the therapeutic progress targeting Wnt signaling was given, and the latest progress in disease treatment targeting key components of the Wnt signaling pathway was summarized in detail, including Wnt ligands/receptors, β-catenin destruction complexes, and β-catenin/TCF transcription complexes. The development of small molecule inhibitors, monoclonal antibodies, and combination therapy strategies was emphasized, while the current potential therapeutic challenges were summarized. This aims to enhance the current understanding of this key pathway.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
25
|
Gong W, Liu Z, Wang Y, Huang W, Yang K, Gao Z, Guo K, Xiao Z, Zhao W. Reprogramming of Treg cell-derived small extracellular vesicles effectively prevents intestinal inflammation from PANoptosis by blocking mitochondrial oxidative stress. Trends Biotechnol 2025; 43:893-917. [PMID: 39689981 DOI: 10.1016/j.tibtech.2024.11.017] [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: 07/22/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic relapsing immune-mediated inflammatory disorder of the alimentary tract without exact etiology. Mitochondrial reactive oxygen species (mtROS) derived from mitochondrial dysfunction impair intestinal barrier function, increase gut permeability, and facilitate immune cell invasion, and, therefore, are considered to have a pivotal role in the pathogenesis of IBD. Here, we reprogrammed regulatory T cell (Treg)-derived exosomes loaded with the antioxidant trace element selenium (Se) and decorated them with the synthetic mitochondria-targeting SS-31 tetrapeptide via a peptide linker. This linker can be cleaved by matrix metalloproteinases (MMPs) in inflammatory lesions. This actively targetable exosome-derived delivery system is protected from intestinal inflammation by scavenging excessive mtROS and preventing immunologically programmed cell death pyroptosis, necroptosis, and apoptosis, known as PANoptosis. Our results suggest that this engineered exosome delivery platform represents a promising targeted therapeutic strategy for the treatment of IBDs.
Collapse
Affiliation(s)
- Wenbin Gong
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhenni Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Yuqiu Wang
- Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Wenbo Huang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Kui Yang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhenhai Gao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Kun Guo
- Department of General Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Zhengtao Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China.
| | - Wei Zhao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
26
|
Rasmussen MS, Hansen LØ, Deding U, Ellebæk MB, Kjeldsen J, Bjørsum-Meyer T. Applicability of colon capsule endoscopy for monitoring ulcerative colitis: a systematic review. Scand J Gastroenterol 2025; 60:336-342. [PMID: 40084907 DOI: 10.1080/00365521.2025.2475081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/18/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND As the target of therapy in Ulcerative colitis (UC) has changed from symptomatic relief to mucosal healing, endoscopic visualization is mandatory. Colon capsule endoscopy (CCE) may serve as a less invasive and more tolerable alternative to standard colonoscopy (SC) for the monitoring of UC. OBJECTIVES To evaluate the diagnostic accuracy, adverse events and tolerability for CCE compared to SC. DESIGN Systematic review. DATA SOURCES A systematic literature search was conducted in PubMed, Embase and Web of Science. METHODS Search results were imported into Covidence and screened. Included studies underwent risk of bias assessment using Methodological Index for Non-Randomized Studies (MINORS), and relevant data, including completeness of the procedure, type of bowel preparation and adverse events, was extracted. Pooled estimates of diagnostic accuracy were calculated from the studies providing the necessary data. RESULTS Out of 2804 articles, six studies were eligible for inclusion. Three provided the necessary data to calculate pooled estimates of diagnostic accuracy in recognizing mucosal inflammation: pooled sensitivity of 93%, specificity of 68.8%, positive predictive value of 89.4%, and negative predictive value of 78.6%. The adverse events, such as nausea and abdominal distension, were predominantly related to bowel preparation regimens. CONCLUSION CCE has the potential for monitoring UC. However, the specificity and NPV must be improved. Bowel preparation regimens must be optimized to improve patient experience and the effectiveness of CCE. REGISTRATION Prospero ID CRD42023450210.
Collapse
Affiliation(s)
- Mathilde Simone Rasmussen
- Department of Surgery, Odense University Hospital, Svendborg, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Lea Østergaard Hansen
- Department of Surgery, Odense University Hospital, Svendborg, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Ulrik Deding
- Department of Surgery, Odense University Hospital, Svendborg, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mark Bremholm Ellebæk
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, Surgical Research Unit, Odense University Hospital, Odense, Denmark
| | - Jens Kjeldsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Research, Research unit of Medical Gastroenterology, Odense University Hospital, Odense, Denmark
| | - Thomas Bjørsum-Meyer
- Department of Surgery, Odense University Hospital, Svendborg, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
27
|
Nakajima M, Iwao Y, Okabayashi K, Kanai Y, Shimoda M. Pathological characteristics of inflammatory bowel diseases. J Med Ultrason (2001) 2025; 52:187-196. [PMID: 40025407 DOI: 10.1007/s10396-025-01520-9] [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: 11/28/2024] [Accepted: 12/24/2024] [Indexed: 03/04/2025]
Abstract
Inflammatory bowel disease (IBD) is an inflammatory disorder in which intestinal homeostasis is disrupted for some reason. Among them, ulcerative colitis (UC) and Crohn's disease (CD) are frequently referred to as IBD in the narrow sense, characterized by relapse episodes and remission periods. The differential diagnosis of IBD involves a broad spectrum of inflammatory or infectious diseases that mimic UC and/or CD, as well as others that may complicate existing IBD. Accordingly, these differential diseases and modifying factors should be considered in their pathological diagnosis, and a careful diagnosis should be made in close collaboration with clinicians. Here, we provide a pathological overview of UC, CD, and their differential diseases, as well as IBD-associated cancers, demonstrating their typical gross and histological features. Further, we introduce a pathological scoring system for biopsy specimens to diagnose IBD that may potentially be integrated into clinical practice.
Collapse
Affiliation(s)
- Makoto Nakajima
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yasushi Iwao
- Center for Preventive Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Koji Okabayashi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Department of Pathology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan.
| |
Collapse
|
28
|
George AT, Rubin DT. Artificial Intelligence in Inflammatory Bowel Disease. Gastrointest Endosc Clin N Am 2025; 35:367-387. [PMID: 40021234 DOI: 10.1016/j.giec.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
Artificial intelligence (AI) is being increasingly studied and implemented in gastroenterology. In inflammatory bowel disease (IBD), numerous AI models are being developed to assist with IBD diagnosis, standardization of endoscopic and radiologic disease activity, and predicting outcomes. Further prospective, multicenter studies representing diverse populations and novel applications are needed prior to routine implementation in clinical practice and expected improved outcomes for clinicians and patients.
Collapse
Affiliation(s)
- Alvin T George
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - David T Rubin
- Department of Medicine, Inflammatory Bowel Disease Center, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
29
|
Liang C, Wang Z, Mai Y, Li J, Dai Q, Yuan Y, Wang M, Liu Y, Zhang W, Li Y, Lu X, Lin Z, Mao T. Mendelian randomization study of circulating leukocytes counts reveals causal associations with inflammatory bowel disease. Medicine (Baltimore) 2025; 104:e41969. [PMID: 40153772 PMCID: PMC11957634 DOI: 10.1097/md.0000000000041969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 03/07/2025] [Indexed: 03/30/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic recurrent IBD, whose cause involves the interaction between genetic and environmental factors. Although there is a recognized link between immune response and IBD, the causal relationship between circulating immune cell counts and IBD remains controversial. This study aimed to elucidate the causal relationship between genetically predicted circulating immune cell counts and IBD. We conducted a bidirectional 2-sample Mendelian randomization (MR) study using aggregated statistics from genome-wide association studies. The causal relationship between 5 circulating leukocytes cells (monocytes, lymphocytes, eosinophils, basophils and neutrophils) counts and IBD, including ulcerative colitis (UC) and Crohn disease (CD) was analyzed. Horizontal pleiotropy test and heterogeneity test were used to ensure the stability of the results. Our findings indicated that monocytes, lymphocytes, eosinophils, and basophils count were not significantly associated with IBD, however, elevated circulating neutrophils count was significantly associated with higher risk of IBD [odds ratio (OR) = 1.0017; 95% confidence interval (CI) = 1.0004-1.003; P = .009] and UC [OR = 2.465; 95% CI = 1.236-4.916; P = .01]. In addition, we also found that IBD [OR: 12.07; 95% CI = 1.909-76.316; P = .008] and CD [OR = 1.014; 95% CI = 1.004-1.023; P = .005] were significantly associated with higher circulating neutrophils count in reverse MR. This MR study provides genetic evidence for the causal relationship between the genetically predicted increase in circulating neutrophils count and the risk of IBD (UC and CD). This finding stresses the need for further exploring physiological functions of neutrophils in order to develop effective strategies against IBD.
Collapse
Affiliation(s)
- Chengtao Liang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Zhibin Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yuhe Mai
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Junxiang Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Qiuhong Dai
- Qinhuangdao Hospital of Traditional Chinese Medicine, Qinhuangdao, PR China
| | - Yali Yuan
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Muyuan Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yuyue Liu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Wenji Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Yitong Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Xinyu Lu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Zhengdao Lin
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| | - Tangyou Mao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, PR China
| |
Collapse
|
30
|
Humeidi R, Oshiro-Rapley N, Gu X, An JS, Ananthakrishnan AN, Creasey EA, Daly MJ, Schreiber SL, Graham DB, Seyedsayamdost MR, Xavier RJ. The Ulcerative Colitis-Associated Gene NXPE1 Catalyzes Glycan Modifications on Colonic Mucin. J Am Chem Soc 2025; 147:10618-10628. [PMID: 40067145 PMCID: PMC12076216 DOI: 10.1021/jacs.5c00769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Colonic mucus forms a first line of defense against bacterial invasion while providing nutrition to support coinhabiting microbes in the gut. Mucus is composed of polymeric networks of mucin proteins, which are heavily modified post-translationally. The full compendium of enzymes responsible for these modifications and their roles in health and disease remain incompletely understood. Herein, we determine the biochemical function of NXPE1, a gene implicated in ulcerative colitis (UC), and demonstrate that it encodes an acetyltransferase that modifies mucin glycans. Specifically, NXPE1 utilizes acetyl-CoA to regioselectively modify the mucus sialic acid, 5-N-acetylneuraminic acid (Neu5Ac), at the 9-OH group to generate 9-O-acetylated Neu5Ac (Neu5,9Ac2). We further demonstrate that colonic organoids derived from donors harboring the missense variant NXPE1 G353R, which is protective against UC, exhibit severely impaired acetylation of Neu5Ac on mucins. Together, our findings support a model in which NXPE1 masks the alcohols of mucus sialoglycans via acetylation, which is important for modulating mucus barrier properties that limit interactions with commensal microbes.
Collapse
Affiliation(s)
- Ranad Humeidi
- Program for Chemistry & Chemical Biology, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Noriko Oshiro-Rapley
- Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Xiebin Gu
- Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Joon Soo An
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Ashwin N. Ananthakrishnan
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Elizabeth A. Creasey
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Mark J. Daly
- Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Stuart L. Schreiber
- Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
- Department of Chemistry & Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Daniel B. Graham
- Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Mohammad R. Seyedsayamdost
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Ramnik J. Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02141, USA
- Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
31
|
Jeong HS, Choi Y, Kim DW, Je JY, Lee SJ, Choi CH. Multicompartmental Hydrogel Microspheres with a Concentric Thin Oil Layer: Protecting and Targeting Therapeutic Agents for Inflammatory Bowel Disease. ACS APPLIED BIO MATERIALS 2025; 8:2251-2262. [PMID: 40030207 DOI: 10.1021/acsabm.4c01763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Although oral delivery of therapeutic agents offers numerous benefits, its application is limited due to the digestive tract's harsh conditions (e.g., strong acidity and high osmolarity), which impair activity and create challenges in achieving targeted release into the intestine. Here, we present multicompartmental hydrogel microspheres equipped with a concentric oil layer to significantly enhance the oral drug delivery efficiency for treating inflammatory bowel disease (IBD). These microspheres are created through the utilization of triple-emulsion droplets, featuring intermediate oil layers that distinctively separate two prepolymer phases, allowing us to fine-tune the composition of each compartment through a tailored polymerization strategy. We demonstrate that the oil layer can protect the encapsulated material by preventing exposure to the acidic environment of the stomach during the digestive process. Unlike aqueous core capsules, the core is composed of hydrogel, which provides high stability even under high osmolarity conditions in the stomach. By fine-tuning the shell's composition, we can develop capsules that release selectively in response to the gut's pH conditions. We demonstrate the system's efficacy by preserving the anti-inflammatory activities of 5-aminosalicylic acid (5-ASA) and Lys-Pro-Val (KPV) under stomach conditions and maintaining their therapeutic effects on colonic epithelial cell migration and proliferation.
Collapse
Affiliation(s)
- Hye-Seon Jeong
- School of Chemical Engineering, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Yoon Choi
- Division of Cosmetic Science and Technology, Daegu Haany University, 1 Haanydaero, Gyeongsan, Gyeongbuk 38610, Republic of Korea
| | - Do-Wan Kim
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Jae-Young Je
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Sei-Jung Lee
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Chang-Hyung Choi
- School of Chemical Engineering, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| |
Collapse
|
32
|
Li Y, Zhang Q, Liu X, Wang Y, Yang C, Wu Y, Xiao B, Feng Y, Wu A, Yi J, Wu J, Liang Z, Yuan Z. Citrinin-Induced Intestinal Onset of Pyroptosis via the IP3R1-GRP75-VDAC1 Complex-Mediated Mitochondrial Oxidative Stress. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5803-5815. [PMID: 40000072 DOI: 10.1021/acs.jafc.4c11218] [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: 02/27/2025]
Abstract
Citrinin (CTN) is commonly found in animal feed and stored grains and poses a serious threat to human and animal health. Formation of the IP3R1-GRP75-VDAC1 complex has been shown to play a key role in intestinal defense against harmful stimuli, but the mechanism of its action in CTN-exposure-induced enterotoxicity is not clear. Therefore, the aim of this study was to investigate the role of the IP3R1-GRP75-VDAC1 complex in CTN-exposure-induced intestinal and IPEC-J2 monolayer cell damage in mice. It was shown that CTN exposure triggered intestinal cell pyroptosis and increased IP3R1-GRP75-VDAC1 complex formation as well as mitochondrial levels of calcium ions and mitochondrial reactive oxygen species (mtROS). And mtROS is considered to be a key factor in cellular pyroptosis. Therefore, the removal of mtROS by using Mito-Tempo was found to attenuate CTN-exposure-induced cellular pyroptosis but failed to attenuate mitochondrial calcium ion overload. However, silencing of GRP75 alleviated CTN-exposure-induced increases in the level of mtROS, mitochondrial calcium ions, and subsequent cellular pyroptosis. Therefore, this study confirms that CTN exposure induces cellular juxtaposition in intestinal tissues and points out that mitochondrial oxidative stress mediated by the IP3R1-GRP75-VDAC1 complex is a key mechanism by which CTN exposure triggers intestinal cellular pyroptosis.
Collapse
Affiliation(s)
- Yuanyuan Li
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Qike Zhang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Xiaofang Liu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Yongkang Wang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Chenglin Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - You Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Bo Xiao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Yiya Feng
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Aoao Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Jine Yi
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
| | - Jing Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
- Institute of Yunnan Circular Agricultural Industry, Puer 665000, P. R. China
| | - Zengenni Liang
- Dongting Laboratory, Hunan Provincial Key Laboratory for Fruits and Vegetables Storage Processing and Quality Safety, Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha 410125, P. R. China
- Yulushan Laboratory, Changsha 410128, P. R. China
| | - Zhihang Yuan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, P. R. China
- Institute of Yunnan Circular Agricultural Industry, Puer 665000, P. R. China
| |
Collapse
|
33
|
Zubair M, Abouelnazar FA, Iqbal MA, Pan J, Zheng X, Chen T, Shen W, Yin J, Yan Y, Liu P, Mao F, Chu Y. Mesenchymal stem cell-derived exosomes as a plausible immunomodulatory therapeutic tool for inflammatory diseases. Front Cell Dev Biol 2025; 13:1563427. [PMID: 40129569 PMCID: PMC11931156 DOI: 10.3389/fcell.2025.1563427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/21/2025] [Indexed: 03/26/2025] Open
Abstract
Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs), especially, exosomes are considered to have diverse therapeutic effects for various significant diseases. MSC-derived exosomes (MSCex) offer substantial advantages over MSCs due to their long-term preservation, stability, absence of nuclei and fewer adverse effects such as infusion toxicity, thereby paving the way towards regenerative medicine and cell-free therapeutics. These exosomes harbor several cellular contents such as DNA, RNA, lipids, metabolites, and proteins, facilitating drug delivery and intercellular communication. MSCex have the ability to immunomodulate and trigger the anti-inflammatory process hence, playing a key role in alleviating inflammation and enhancing tissue regeneration. In this review, we addressed the anti-inflammatory effects of MSCex and the underlying immunomodulatory pathways. Moreover, we discussed the recent updates on MSCex in treating specific inflammatory diseases, including arthritis, inflammatory bowel disease, inflammatory eye diseases, and respiratory diseases such as asthma and acute respiratory distress syndrome (ARDS), as well as neurodegenerative and cardiac diseases. Finally, we highlighted the challenges in using MSCex as the successful therapeutic tool and discussed future perspectives.
Collapse
Affiliation(s)
- Muhammad Zubair
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Fatma A. Abouelnazar
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
- Faculty of Applied Health Sciences Technology, Pharos University, Alexandria, Egypt
| | | | - Jingyun Pan
- Department of Traditional Chinese Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Xuwen Zheng
- Department of Emergency, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Tao Chen
- Department of Gastroenterology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Wenming Shen
- Department of Emergency, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Jinnan Yin
- Department of Emergency, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Pengjun Liu
- Department of Gastroenterology, Wujin Hospital Affiliated With Jiangsu University, Changzhou, China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ying Chu
- Wujin Clinical College, Xuzhou Medical University, Changzhou, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
34
|
Yan Y, Tian L, Zhao Y, Xuan B, Xu X, Ding J, Li W, Zhou YL, Zhang Y, Ma Y, Ning L, Wang Z, Jiang Y, Zhu X, Huang X, Hu M, Shen N, Gao X, Fang JY, Cui Z, Cao Z, Chen H, Wang X, Hong J. Bacteroides fragilis Toxin Suppresses METTL3-Mediated m6A Modification in Macrophage to Promote Inflammatory Bowel Disease. J Crohns Colitis 2025; 19:jjae179. [PMID: 40065724 DOI: 10.1093/ecco-jcc/jjae179] [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] [Indexed: 05/13/2025]
Abstract
BACKGROUND AND AIMS Bacteroides fragilis toxin (BFT), produced by enterotoxigenic B. fragilis (ETBF), is crucial for ETBF-induced colitis. This study aims to investigate the impact of BFT-host interactions on N6-methyladenosine (m6A) modification of host mRNA and its underlying mechanisms. METHODS Single-cell sequencing was employed to identify the cell types involved in ETBF-induced colitis in inflammatory bowel disease patients and dextran sodium sulfate-induced colitis mice. An ETBF strain with the bft gene deleted (ETBF[Δbft]) was utilized to investigate the role of ETBF components. The biological functions and mechanisms of BFT-induced m6A modifications, as well as the target genes, were explored in vitro and in vivo. RESULTS Inflammatory macrophages are enriched in the intestinal mucosal tissue of both inflammatory bowel disease patients and mice with high levels of ETBF. Additionally, ETBF triggers the activation of inflammatory macrophages, subsequently inducing downstream inflammatory responses. Remarkably, BFT secreted by ETBF reduced METTL3 transcription by inhibiting FOXD3 expression and induced a dramatic reduction of m6A modifications in inflammatory macrophages. Moreover, BFT promotes the expression of its target ITGA5 expression by diminishing YTHDF2-dependent mRNA degradation. Targeting integrin subunit alpha 5 using Cilengitide significantly alleviated ETBF-induced colitis by decreasing the level of inflammatory factors in macrophages. CONCLUSIONS Our study reveals that BFT produced by ETBF leads to a reduction of m6A modifications by reducing METTL3 transcription and promotes ITGA5 expression in inflammatory macrophages. These findings provide new insights into the modulation of human m6A epitranscriptome in macrophages by gut microbiota and its significance in inflammatory bowel disease progression.
Collapse
Affiliation(s)
- Yuqing Yan
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Li Tian
- Department of Gastroenterology, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Hunan, China
| | - Ying Zhao
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Baoqin Xuan
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Xitao Xu
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Jinmei Ding
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Weixun Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Yi-Lu Zhou
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Yue Zhang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Yanru Ma
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Lijun Ning
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Zhenyu Wang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Yi Jiang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Xiaoqiang Zhu
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Xiaowen Huang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Muni Hu
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Nan Shen
- Department of Infectious Disease, Shanghai Children's Medical Center, National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiang Gao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Jing-Yuan Fang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Zhe Cui
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhijun Cao
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Haoyan Chen
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| | - Xiaoyan Wang
- Department of Gastroenterology, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Hunan, China
| | - Jie Hong
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai, China
| |
Collapse
|
35
|
Gao S, Ge Y, Huang H, Wang L, Zhang W. Adipose-Derived Mesenchymal Stem Cell Exosomes Encapsulating siIL1R2 Facilitate the Repair of DSS-Induced Intestinal Mucosal Injury. Immunol Invest 2025:1-17. [PMID: 40035289 DOI: 10.1080/08820139.2025.2468959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
BACKGROUND Interleukin-1 receptor 2 (IL1R2) and C-C motif chemokine receptor 2 (CCR2) as critical mediators of immune modulation and inflammation. This study aims to evaluate their functions in dextran sulfate sodium (DSS)-induced intestinal injury. METHODS A DSS-induced intestinal injury model was established in C57BL/6 mice. Pharmacological inhibitors targeting IL1R2 or CCR2 were administered. Adipose-derived mesenchymal stem cell (ADMSC)-derived exosomes were isolated and loaded with IL1R2-siRNA, which were then administered to intestinal epithelial cells (IEC-6) or DSS-challenged mice. RESULTS IL1R2 and CCR2 were upregulated in DSS-treated colon tissues. Pharmacological inhibition of IL1R2 or CCR2 improved body weight, restored colon length, reduced serum TNF-α and IL-6 levels, and preserved epithelial integrity in mice. miR-128-3p enriched in ADMSC-derived exosomes significantly reduced CCR2 expression in IEC-6 cells. Further loading of an IL1R2 siRNA in these exosomes led to a simultaneous inhibition of IL1R2. These exosomes reduced lipopolysaccharide-induced apoptosis and inflammation in IEC-6 cells and improved histological outcomes in DSS-challenged mice. CONCLUSION IL1R2 and CCR2 are key mediators of inflammation in DSS-induced intestinal injury. Dual inhibition of IL1R2 and CCR2 holds great promise for alleviating inflammatory responses and improving histological presentations in inflammatory bowel disease.
Collapse
Affiliation(s)
- Song Gao
- Department of Gastrointestinal Surgery, the Fifth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Yajuan Ge
- Department of Abdominal Ultrasound, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - He Huang
- Department of Gastrointestinal Surgery, the Fifth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Lei Wang
- Department of Gastrointestinal Surgery, the Fifth Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Wenbin Zhang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| |
Collapse
|
36
|
Wu W, Zhang J, Qu X, Chen T, Li J, Yang Y, Chen L, Hoover A, Guo F, Kong C, Bao B, Lin Q, Zhou M, Zhu L, Wu X, Ma Y. Enabling Targeted Drug Delivery for Treatment of Ulcerative Colitis with Mucosal-Adhesive Photoreactive Hydrogel. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404836. [PMID: 39900372 PMCID: PMC11948015 DOI: 10.1002/advs.202404836] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 11/01/2024] [Indexed: 02/05/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease. UC treatments are limited by significant adverse effects associated with non-specific drug delivery, such as systematic inhibition of the host immune system. Endoscopic delivery of a synthetic hydrogel material with biocompatible gelation that can efficiently cover irregular tissue surfaces provides an effective approach for targeted drug delivery at the gastrointestinal (GI) tract. An ideal integration of synthetic material with intestinal epithelium entails an integrated and preferable chemically bonded interface between the hydrogel and mucosal surface. In this study, a photo-triggered coupling reaction is leveraged as the crosslinking platform to develop a mucosal-adhesive hydrogel, which is compatible with endoscope-directed drug delivery for UC treatment. The results demonstrated superior spatiotemporal specificity and drug pharmacokinetics with this delivery system in vivo. Delivery of different drugs with the hydrogel leads to greatly enhanced therapeutic efficacy and significantly reduced systemic drug exposure with rat colitis models. The study presents a strategy for targeted and persistent drug delivery for UC treatment.
Collapse
Affiliation(s)
- Wen Wu
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityNo. 270 Dongan RoadShanghai200032China
| | - Jian Zhang
- Ben May Department for Cancer ResearchUniversity of ChicagoGCIS W408B, 929 E 57th StreetChicagoIL60637USA
| | - Xiao Qu
- Department of EndoscopyFudan University Shanghai Cancer CenterNo. 270 Dongan RoadShanghai200032China
| | - Ting Chen
- School of Biomedical EngineeringShanghai Jiao Tong UniversityNo. 800 Dongchuan RoadShanghai200240China
| | - Jinming Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityNo. 270 Dongan RoadShanghai200032China
| | - Yongzhi Yang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityNo. 270 Dongan RoadShanghai200032China
| | - Lifeng Chen
- Ben May Department for Cancer ResearchUniversity of ChicagoGCIS W408B, 929 E 57th StreetChicagoIL60637USA
| | - Alex Hoover
- Ben May Department for Cancer ResearchUniversity of ChicagoGCIS W408B, 929 E 57th StreetChicagoIL60637USA
| | - Fanying Guo
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityNo. 270 Dongan RoadShanghai200032China
| | - Cheng Kong
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityNo. 270 Dongan RoadShanghai200032China
| | - Bingkun Bao
- School of Biomedical EngineeringShanghai Jiao Tong UniversityNo. 800 Dongchuan RoadShanghai200240China
| | - Qiuning Lin
- School of Biomedical EngineeringShanghai Jiao Tong UniversityNo. 800 Dongchuan RoadShanghai200240China
| | - Mengxin Zhou
- School of Chemistry and Molecular EngineeringEast China University of Science and TechnologyNo.130 Meilong RoadShanghai200237China
| | - Linyong Zhu
- School of Biomedical EngineeringShanghai Jiao Tong UniversityNo. 800 Dongchuan RoadShanghai200240China
| | - Xiaoyang Wu
- Ben May Department for Cancer ResearchUniversity of ChicagoGCIS W408B, 929 E 57th StreetChicagoIL60637USA
| | - Yanlei Ma
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityNo. 270 Dongan RoadShanghai200032China
| |
Collapse
|
37
|
Yang YJ, Jeon SR. Metabolic musculoskeletal disorders in patients with inflammatory bowel disease. Korean J Intern Med 2025; 40:181-195. [PMID: 40102707 PMCID: PMC11938716 DOI: 10.3904/kjim.2024.359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 03/20/2025] Open
Abstract
Inflammatory bowel disease (IBD), which includes Crohn's disease and ulcerative colitis, is a chronic inflammatory disorder that affects not only the gastrointestinal tract but also extraintestinal organs, leading to various extraintestinal manifestations and complications. Among these, musculoskeletal disorders such as osteoporosis, sarcopenia, and axial and peripheral spondyloarthritis are the most commonly observed. These conditions arise from complex mechanisms, including chronic inflammation, malnutrition, gut dysbiosis, and glucocorticoid use, all of which contribute to reduced bone density, muscle loss, and joint inflammation. Osteoporosis and sarcopenia may co-occur as osteosarcopenia, a condition that heightens the risk of fractures, impairs physical performance, and diminishes quality of life, particularly in elderly patients with IBD. Holistic management strategies, including lifestyle modifications, calcium, and vitamin D supplementation, resistance training, and pharmacological interventions, are essential for mitigating the impact of these conditions. Spondyloarthritis, which affects both axial and peripheral joints, further complicates disease management and significantly compromises joint health. Timely diagnosis and appropriate medical interventions, such as administration of nonsteroidal anti-inflammatory drugs and biologics, are critical for preventing chronic joint damage and disability. Moreover, a multidisciplinary approach that addresses both metabolic and inflammatory aspects is essential for optimizing physical function and improving treatment outcomes in patients who have IBD with musculoskeletal involvement.
Collapse
Affiliation(s)
- Young Joo Yang
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon,
Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon,
Korea
| | - Seong Ran Jeon
- Digestive Disease Center, Institute for Digestive Research, Soonchunhyang University College of Medicine, Seoul,
Korea
| |
Collapse
|
38
|
Fokam Tagne MA, Noubissi PA, Foyet Fondjo A, Nono Njomguep L, Ngakou Mukam J, Sokeng Dongmo S, Kamgang R. Effects of aqueous extract of Waltheria indica (Sterculiaceae) leafy stems on acetic acid-induced ulcerative colitis in rats. Inflammopharmacology 2025; 33:1505-1516. [PMID: 39934536 DOI: 10.1007/s10787-025-01651-y] [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: 10/24/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025]
Abstract
Ulcerative colitis is one of the inflammatory bowel diseases that manifest itself by uncontrolled inflammation of colon. The objective of this work was to evaluate the effects of aqueous extract of Waltheria indica on acetic acid-induced ulcerative colitis in rats. Six (6) groups of five (5) rats each, were anesthetized with a ketamine (50 mg/kg)/valium (10 mg/kg) mixture after eighteen (18) fasting hours. Colitis was induced by intrarectal administration of 1 mL of acetic acid (5%) in animals. Five (5) hours later, the normal control (NC) and the colitis control (CC) received distilled water (10 mL/kg bw), the positive control (Pre5) received prednisolone (5 mg/kg) and the other three test groups received the W. indica extract at 50 (Wi50), 100 (Wi100) and 200 (Wi200) mg/kg bw, orally for 7 days. At the end of the treatment, the animals were sacrificed and the blood was collected from the carotid artery, part in the ethylenediaminetetraacetate (EDTA) tube for hematological analyzes and part in dry tubes for biochemical assays. The abdomen was then opened, the colon, liver, spleen, lungs and heart were removed, drained, weighed and the indexes of each organ were determined. The extract at 200 mg/kg reduced myeloperoxidase (MPO) and inhibited the production of interleukin-1 beta (IL-1β) and interleukin-6(IL-6) in the colon and serum. The extract significantly increased the blood platelet level of the colitis rats. Thus, these results suggest that Walthera indica extract may have therapeutic potential for the treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Michel Archange Fokam Tagne
- Department of Biological Sciences, Faculty of Science, University of Ngaoundere, P.O. Box 454, Ngaoundere, Cameroon.
| | - Paul Aimé Noubissi
- Department of Zoology and Animal Physiology, Faculty of Science, University of Buea, Buea, Cameroon
| | - Angèle Foyet Fondjo
- Department of Applied Sciences for Health, Higher Institute of Applied Sciences, University Institute of Gulf of Guinea, 237, Douala, Cameroon
| | - Laurelle Nono Njomguep
- Department of Biological Sciences, Faculty of Science, University of Ngaoundere, P.O. Box 454, Ngaoundere, Cameroon
| | - Joseph Ngakou Mukam
- Animal Physiology Laboratory, Faculty of Science, University of Yaoundé I, Yaoundé, Cameroon
| | - Sélestin Sokeng Dongmo
- Department of Biological Sciences, Faculty of Science, University of Ngaoundere, P.O. Box 454, Ngaoundere, Cameroon
| | - René Kamgang
- Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon
| |
Collapse
|
39
|
Hu CH, Chen Y, Jin TY, Wang Z, Jin B, Liao J, Ding CY, Zhang A, Tang WY, Zhang LX, Xu LY, Ning FM, Liang G, Wei XH, Wang Y. A derivative of tanshinone IIA and salviadione, 15a, inhibits inflammation and alleviates DSS-induced colitis in mice by direct binding and inhibition of RIPK2. Acta Pharmacol Sin 2025; 46:672-686. [PMID: 39443729 PMCID: PMC11845706 DOI: 10.1038/s41401-024-01399-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory conditions primarily affecting the gastrointestinal tract. Previous studies established the role of the NF-κB signaling pathway in the development of IBDs, suggesting that anti-inflammatory therapies might offer a viable treatment strategy. Tanshinone IIA and salviadione, both derived from Salviae Miltiorrhizae Radix et Rhizoma, possess anti-inflammatory and anti-oxidative activities. A series of new compounds were synthesized by hybridizing salviadione with tanshinone. Among these compounds, 15a showed beneficial effects in LPS-induced acute lung injury and diabetes-induced renal injury mouse models. The current study explored the therapeutic efficacy of 15a using both acute and chronic colitis models and elucidated the underlying mechanisms. DSS-induced colitis models were established in mice, where acute colitis was treated with compound 15a (5 or 10 mg·kg-1·d-1) for 8 days, while chronic colitis mice received compound 15a (5 or 10 mg·kg-1·d-1, i.g.) during 2.5% DSS administration. The 15a treatment significantly alleviated DSS-induced pathological and inflammatory damages in both acute and chronic colitis mouse models. In mouse intestinal epithelial cell line MODE-K, pretreatment with compound 15a (5 or 10 μM) significantly suppressed LPS + L18-MDP-induced inflammatory responses. The receptor-interacting serine/threonine kinase 2 (RIPK2) was identified as a direct binding target of compound 15a using microarrays and recombinant human proteins. Moreover, 15a could directly bind to and inhibit the phosphorylation of RIPK2, leading to the suppression of the NF-κB and MAPK signaling pathways. Furthermore, LEU153 and VAL32 were identified within the KD domain of RIPK2 as critical amino residues for the binding of 15a. Briefly, the current findings demonstrate that compound 15a holds promise as a therapeutic agent for managing acute and chronic colitis.
Collapse
Affiliation(s)
- Cheng-Hong Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Yue Chen
- Chemical Biology Research Center, Wenzhou Medical University, School of Pharmaceutical Sciences, Wenzhou, 325035, China
| | - Tian-Yang Jin
- Chemical Biology Research Center, Wenzhou Medical University, School of Pharmaceutical Sciences, Wenzhou, 325035, China
| | - Zhe Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Bo Jin
- Chemical Biology Research Center, Wenzhou Medical University, School of Pharmaceutical Sciences, Wenzhou, 325035, China
| | - Jing Liao
- Chemical Biology Research Center, Wenzhou Medical University, School of Pharmaceutical Sciences, Wenzhou, 325035, China
| | - Chun-Yong Ding
- Pharm-X Center, College of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ao Zhang
- Pharm-X Center, College of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei-Yang Tang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Ling-Xi Zhang
- Chemical Biology Research Center, Wenzhou Medical University, School of Pharmaceutical Sciences, Wenzhou, 325035, China
| | - Lei-Yu Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Fang-Min Ning
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, 310051, China
| | - Xiao-Hong Wei
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Yi Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, China.
| |
Collapse
|
40
|
Wu J, Ye W, Yu J, Zhou T, Zhou N, K P Ng D, Li Z. Engineered bacteria and bacterial derivatives as advanced therapeutics for inflammatory bowel disease. Essays Biochem 2025; 69:EBC20253003. [PMID: 40014418 DOI: 10.1042/ebc20253003] [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: 01/02/2025] [Accepted: 01/29/2025] [Indexed: 03/01/2025]
Abstract
Inflammatory bowel disease (IBD), a chronic and relapsing-remitting condition, is inadequately managed by conventional therapies that often lack targeting specificity and carry significant side effects, particularly failing to address intestinal barrier repair and microbial balance. Probiotics, with their strong colonization capabilities, present a novel approach to drug delivery. Various engineering strategies have been developed to enhance the targeting ability of probiotics to inflammation sites, enabling precise delivery or in situ synthesis of therapeutic molecules to expand their multifunctional potential. This review discusses the recent advancements in bacterial modifications, including surface physico-chemical and biological coating, genetic engineering, outer membrane vesicles, minicells, and bacterial ghosts, all of which can enhance therapeutic localization. We also outline critical preclinical considerations, such as delivery frequency, systemic distribution, immune evasion, and gene contamination risks, for clinical translation. These engineered bacteria and bacterial derivatives hold great promise for personalized and sustained IBD treatments, providing a new frontier for therapy tailored to the complex inflammatory environment of IBD.
Collapse
Affiliation(s)
- Jingyuan Wu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, P. R. China
| | - Wanlin Ye
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, P. R. China
| | - Jie Yu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, P. R. China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Tuoyu Zhou
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, P. R. China
- The Second Affiliated Hospital, School of Medicine, The Chinese University of Hong Kong, Shenzhen & Longgang District People's Hospital of Shenzhen, Guangdong, 518172, P. R. China
| | - Nuo Zhou
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, P. R. China
| | - Dennis K P Ng
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N. T., Hong Kong, P. R. China
| | - Zhaoting Li
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, P. R. China
| |
Collapse
|
41
|
Ullah H, Arbab S, Chang C, Bibi S, Muhammad N, Rehman SU, Suleman, Ullah I, Hassan IU, Tian Y, Li K. Gut microbiota therapy in gastrointestinal diseases. Front Cell Dev Biol 2025; 13:1514636. [PMID: 40078367 PMCID: PMC11897527 DOI: 10.3389/fcell.2025.1514636] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
The human gut microbiota, consisting of trillions of microorganisms, plays a crucial role in gastrointestinal (GI) health and disease. Dysbiosis, an imbalance in microbial composition, has been linked to a range of GI disorders, including inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), celiac disease, and colorectal cancer. These conditions are influenced by the interactions between the gut microbiota, the host immune system, and the gut-brain axis. Recent research has highlighted the potential for microbiome-based therapeutic strategies, such as probiotics, prebiotics, fecal microbiota transplantation (FMT), and dietary modifications, to restore microbial balance and alleviate disease symptoms. This review examines the role of gut microbiota in the pathogenesis of common gastrointestinal diseases and explores emerging therapeutic approaches aimed at modulating the microbiome. We discuss the scientific foundations of these interventions, their clinical effectiveness, and the challenges in their implementation. The review underscores the therapeutic potential of microbiome-targeted treatments as a novel approach to managing GI disorders, offering personalized and alternative options to conventional therapies. As research in this field continues to evolve, microbiome-based interventions hold promise for improving the treatment and prevention of gastrointestinal diseases.
Collapse
Affiliation(s)
- Hanif Ullah
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, Nursing Key Laboratory of Sichuan Province, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Safia Arbab
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Chengting Chang
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, Nursing Key Laboratory of Sichuan Province, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Saira Bibi
- Department of Zoology Hazara University Manshera, Dhodial, Pakistan
| | - Nehaz Muhammad
- Hebei Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology, Hebei Collaborative Innovation Center for Eco-Environment, College of Life Sciences, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Sajid Ur Rehman
- School of Public Health and Emergency Management, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Suleman
- Department of Zoology, Government Post Graduate Collage, Swabi, Pakistan
- Higher Education Department, Civil Secretariat Peshawar, Peshawar, Pakistan
| | - Irfan Ullah
- Department of Biotechnology and Genetics Engineering, Hazara University, Manshera, Pakistan
| | - Inam Ul Hassan
- Department of Microbiology, Hazara University Manshera, Manshera, Pakistan
| | - Yali Tian
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, Nursing Key Laboratory of Sichuan Province, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| | - Ka Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, Nursing Key Laboratory of Sichuan Province, West China Hospital, West China School of Nursing, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
42
|
He E, Shi B, Jia M, Sun W, Chang K, Jiang H, Zhao W, Zhao H, Dong L, Die X, Feng W, Cui H. Hirschsprung's disease may increase the incidence of inflammatory bowel disease through alterations in CA1. Pediatr Res 2025:10.1038/s41390-025-03938-w. [PMID: 39988713 DOI: 10.1038/s41390-025-03938-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/17/2024] [Accepted: 01/26/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND The role of Hirschsprung's disease (HSCR) for the development of inflammatory bowel disease (IBD) and the common pathogenesis of the diseases remains unclear. The objective is to investigate the relationship between HSCR and IBD. METHODS In our study, the Mendelian randomization approach was employed to analyze the causal relationships. A further search was conducted for differentially expressed genes (DEGs) between disease and control tissues in HSCR and IBD. Subsequently, the potential pathway mechanisms were subjected to an enrichment analysis. Furthermore, the molecular docking was employed to investigate the binding relationship between potential therapeutic targets and drugs. RESULTS The results show HSCR have an increased risk of developing IBD (IVW: OR = 1.048, P < 0.05; weighted median: OR = 1.065, P < 0.05). A total of 111 DEGs were identified in IBD, while 471 DEGs were observed in HSCR. CA1 was identified as core gene and exhibited lower expression levels in IBD (P < 0.05). Concomitantly, CA1 exhibited reduced expression levels in inflamed tissues. And the TNF and IL17 signaling pathway were found closely related to CA1 expression. CONCLUSION In total, our study shows HSCR promote the occurrence of IBD and reveals pathogenesis. Our results suggest CA1 may provide novel insight for the treatment of HSCR complicated with IBD. IMPACT Individuals with HSCR are at a higher risk of developing IBD (IVW: OR = 1.048, P < 0.05; Weighted median: OR = 1.065, P < 0.05). Patients with IBD exhibited lower expression levels of CA1 (P < 0.05). Furthermore, CA1 expression was found to be lower in inflamed tissues (P < 0.05). CA1 may provide novel insight for the treatment of HSCR complicated with IBD.
Collapse
Affiliation(s)
- Enyang He
- Tianjin Medical University, Tianjin, China
- Graduate School of Tianjin Medical University, Tianjin, China
| | - Bowen Shi
- Department of General Surgery, Tianjin Children's Hospital, Tianjin, China
| | - Miao Jia
- Tianjin Medical University, Tianjin, China
- Graduate School of Tianjin Medical University, Tianjin, China
| | - Wenjing Sun
- Tianjin Medical University, Tianjin, China
- Graduate School of Tianjin Medical University, Tianjin, China
| | - Kaili Chang
- Tianjin Medical University, Tianjin, China
- Graduate School of Tianjin Medical University, Tianjin, China
| | - Hongyv Jiang
- Tianjin Medical University, Tianjin, China
- Graduate School of Tianjin Medical University, Tianjin, China
| | - Wei Zhao
- Tianjin Medical University, Tianjin, China
- Graduate School of Tianjin Medical University, Tianjin, China
| | - Hailan Zhao
- Tianjin Medical University, Tianjin, China
- Graduate School of Tianjin Medical University, Tianjin, China
| | - Liang Dong
- Department of General Surgery, Tianjin Children's Hospital, Tianjin, China
| | - Xiaohong Die
- Department of General & Neonatal Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Feng
- Department of General & Neonatal Surgery, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Hualei Cui
- Tianjin Medical University, Tianjin, China.
- Department of General Surgery, Tianjin Children's Hospital, Tianjin, China.
| |
Collapse
|
43
|
Li Q, Song X, Su P, Lv X, Liu X, Chen X, Tang J, Gao X, Chao K. Risk factors and clinical characteristics of Clostridium difficile colonization and infection in patients with inflammatory bowel disease exposed to Vedolizumab: a multicenter retrospective study. Therap Adv Gastroenterol 2025; 18:17562848251321707. [PMID: 39975482 PMCID: PMC11837063 DOI: 10.1177/17562848251321707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 02/03/2025] [Indexed: 02/21/2025] Open
Abstract
Background Vedolizumab (VDZ), a humanized monoclonal antibody that selectively inhibits the binding of the α4β7 integrin, has been approved for treating inflammatory bowel disease (IBD). Long-term safety studies of VDZ in clinical trials identified Clostridium difficile infection (CDI) as the major opportunistic infection. Objectives We aimed to address the incidence and risk factors of C. difficile colonization (CDC) and CDI in a real-world setting among IBD patients treated with VDZ. Design Retrospective multicenter study. Methods We retrospectively included IBD patients who tested negative for C. difficile before initiating standard VDZ therapy at four tertiary hospitals from November 1, 2021, to November 31, 2023. The primary outcome was the occurrence of CDC after VDZ initiation, and the secondary outcome was the occurrence of CDI and severe CDI. Results A total of 454 patients were included in the final analysis. The median follow-up time was 12.9 (8.2-16.3) months, and the study was followed for 2488.6 person-months. The CDC occurred in 28 patients (6.2%), including 23 (11.4%) patients with ulcerative colitis (UC; 18 asymptomatic carriers and 5 with symptomatic CDI) and 5 (2.0%) patients with Crohn's disease (asymptomatic carriers). Multivariate analysis showed that age >40 years old and UC were independent risk factors for the occurrence of the CDC after VDZ initiation. The incidence of CDI was 1.1%, and all patients were able to continue VDZ therapy after receiving antibiotic treatment. No risk factors were found to be significantly associated with CDI. There were no cases of severe CDI or deaths within 30 days. Conclusion The incidence of CDC after VDZ treatment was 6.2% and the majority of patients identified as asymptomatic carriers and were able to continue VDZ treatment. Age (>40 years old) and UC were the risk factors for CDC.
Collapse
Affiliation(s)
- Qing Li
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xiaomei Song
- Department of Gastroenterology, Chongqing General Hospital, Chongqing, P.R. China
| | - Peizhu Su
- Department of Gastroenterology, The First People’s Hospital of Foshan, Foshan, P.R. China
| | - Xiaoping Lv
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Xinyu Liu
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xuemin Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, P.R. China
| | - Jian Tang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xiang Gao
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Kang Chao
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Road II, Tianhe District, Guangzhou 510000, P.R. China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| |
Collapse
|
44
|
Chen L, Xu Y, Ai F, Shen S, Luo Y, Li X. Dissecting the rising tide of inflammatory bowel disease among youth in a changing world: insights from GBD 2021. Int J Colorectal Dis 2025; 40:44. [PMID: 39964411 PMCID: PMC11836149 DOI: 10.1007/s00384-025-04821-0] [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] [Accepted: 02/03/2025] [Indexed: 02/21/2025]
Abstract
OBJECTIVES This study investigates the alarming epidemiological trends of inflammatory bowel disease (IBD) among children and young adults, highlighting the associated disease burden on global health. MATERIALS AND METHODS Utilizing data from the Global Burden of Disease (GBD) study 2021, we conducted a comprehensive analysis of age-standardized incidence rates (ASIR), age-standardized mortality rates (ASMR), disability-adjusted life years (DALYs), and estimated annual percentage changes (EAPC). Future trends were forecasted using the Bayesian age-period-cohort model. RESULTS From 1990 to 2021, IBD incidence and DALY rates remained persistently high, with a concerning upward trend noted among children and young adults. While men experienced a decline in DALY rates, women faced increasing burdens. In 2021, high-income regions, particularly North America, reported the highest incidence and DALY rates, contrasting sharply with Central Latin America, which exhibited the lowest ASIR. Southeast Asia presented the most favorable DALY rates. A notable negative correlation was identified between DALY rates and socio-demographic index (SDI) at the national level, with high and high-middle SDI countries continuing to bear a substantial burden, while low and middle SDI nations faced rising challenges. CONCLUSIONS The persistent high burden of IBD in children and young adults signifies a critical public health concern. The marked geographical and gender disparities underscore the urgent need for tailored regional and population-based strategies aimed at primary prevention and effective management. This study illuminates the pressing necessity for policy interventions to address the growing epidemic of IBD among vulnerable populations.
Collapse
Affiliation(s)
- Libin Chen
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, Hunan, China
| | - Yifu Xu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Feiyan Ai
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, Hunan, China
| | - Shourong Shen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, Hunan, China
| | - Yanwei Luo
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Xiayu Li
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, Hunan, China.
| |
Collapse
|
45
|
Li X, Cao L, Li J, Li Z, Ma H, Cheng S, Xu H, Zhao Y. Orally Administrated Inulin-Modified Nanozymes for CT-Guided IBD Theranostics. Int J Nanomedicine 2025; 20:2119-2131. [PMID: 39990289 PMCID: PMC11846537 DOI: 10.2147/ijn.s497558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/06/2025] [Indexed: 02/25/2025] Open
Abstract
Background Inflammatory bowel disease (IBD) is a chronic inflammatory bowel disease with no clinical cure. Excessive production of reactive oxygen species (ROS) at the inflammatory sites leads to the onset and progression of IBD. And the current non-invasive imaging methods are not ideal for the diagnosis and monitoring of IBD. Methods Herein, we developed inulin (IN)-coated cerium oxide nanoparticles (CeO2@IN NPs) for treatment and monitoring of IBD guided by computed tomography (CT). The physicochemical properties, ROS scavenging ability and CT imaging capabilities of CeO2@IN were investigated in vitro. Moreover, the therapeutic and targeted inflammation imaging effects of CeO2@IN were validated in dextran sulfate sodium (DSS)-induced colitis model. Results CeO2@IN with catalase (CAT) and superoxide dismutase (SOD) capabilities effectively scavenged ROS, thus protecting the cells against oxidative stress. In colitis model mice, orally administered CeO2@IN successfully traversed the gastrointestinal tract to reach the colon under the protection of IN, and effectively reduced intestinal inflammation, thereby maintaining the intestinal epithelial integrity. Notably, CeO2@IN performed better than conventional CT contrast agents for gastrointestinal tract imaging, particularly in detecting the inflamed areas in the colon. In addition, CeO2@IN exhibited excellent biocompatibility in vitro and in vivo. Conclusion The study provided a novel integrated diagnostic and therapeutic tool for the treatment and monitoring of IBD, presenting great potential as a clinical application for IBD.
Collapse
Affiliation(s)
- Xinwen Li
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Lin Cao
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Jianmin Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Zhengyang Li
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Hongyu Ma
- Image Center, Cangzhou Integrated Traditional and Western Medicine Hospital, Cangzhou, 061000, People’s Republic of China
| | - Shifeng Cheng
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Hongyi Xu
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Yang Zhao
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| |
Collapse
|
46
|
Kim EJ, Jeong HS, Park JY, Je JY, Choi CH, Lee SJ. The inflammatory bowel disease and gut microbiome are restored by employing metformin-loaded alginate-shelled microcapsules. J Control Release 2025; 378:490-502. [PMID: 39710207 DOI: 10.1016/j.jconrel.2024.12.041] [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: 03/18/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic or recurrent inflammatory disorder affecting various parts of the gastrointestinal tract. Metformin, a widely prescribed hypoglycemic drug for type 2 diabetes, has shown potential in reducing IBD risk. However, its oral administration faces significant challenges due to the harsh gastrointestinal environment, requiring higher or more frequent doses to achieve therapeutic effects, which increases the risk of side effects. To address this, we developed alginate-shelled hydrogel microcapsules with a thin oil layer for targeted intestinal delivery of metformin. The oil layer protects metformin from gastric acid and ensures its release specifically in the intestines. In a dextran sulfate sodium-induced colitis mouse model, metformin-loaded hydrogel microcapsules (MHM) significantly reduced disease activity, intestinal epithelial damage, and macrophage infiltration linked to pro-inflammatory cytokine factors. Additionally, MHM improved dysbiosis of specific bacterial genera, including Bacteroides vulgatus, Lactobacillus (L.) gasseri, L. reuteri, and L. intestinalis, optimizing the abundance and composition of microorganisms. These findings indicate that encapsulating metformin within alginate shelled-microcapsules with a thin oil layer presents a potential delivery system for IBD treatment.
Collapse
Affiliation(s)
- Eun-Ju Kim
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, Republic of Korea; Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Hye-Seon Jeong
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Ji-Yeon Park
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Jae-Young Je
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Chang-Hyung Choi
- School of Chemical Engineering, Yeungnam University, 280 Daehak-ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Sei-Jung Lee
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea.
| |
Collapse
|
47
|
Pinto S, Šajbenová D, Benincà E, Nooij S, Terveer EM, Keller JJ, van der Meulen–de Jong AE, Bogaards JA, Steyerberg EW. Dynamics of Gut Microbiota After Fecal Microbiota Transplantation in Ulcerative Colitis: Success Linked to Control of Prevotellaceae. J Crohns Colitis 2025; 19:jjae137. [PMID: 39225490 PMCID: PMC11836888 DOI: 10.1093/ecco-jcc/jjae137] [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: 05/19/2024] [Revised: 07/21/2024] [Accepted: 09/02/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) is an experimental treatment for ulcerative colitis (UC). We aimed to study microbial families associated with FMT treatment success. METHODS We analyzed stools from 24 UC patients treated with 4 FMTs weekly after randomization for pretreatment during 3 weeks with budesonide (n = 12) or placebo (n = 12). Stool samples were collected 9 times pre-, during, and post-FMT. Clinical and endoscopic response was assessed 14 weeks after initiation of the study using the full Mayo score. Early withdrawal due to worsening of UC symptoms was classified as non-response. RESULTS Nine patients (38%) reached remission at week 14, and 15 patients had a partial response or non-response at or before week 14. With a Dirichlet multinomial mixture model, we identified 5 distinct clusters based on the microbiota composition of 180 longitudinally collected patient samples and 27 donor samples. A Prevotellaceae-dominant cluster was associated with poor response to FMT treatment. Conversely, the families Ruminococcaceae and Lachnospiraceae were associated with a successful clinical response. These associations were already visible at the start of the treatment for a subgroup of patients and were retained in repeated measures analyses of family-specific abundance over time. Responders were also characterized by a significantly lower Simpson dominance compared to non-responders. CONCLUSIONS The success of FMT treatment of UC patients appears to be associated with specific gut microbiota families, such as control of Prevotellaceae. Monitoring the dynamics of these microbial families could potentially be used to inform treatment success early during FMT. CLINICAL TRIAL REGISTRATION NUMBER The study was registered in the Netherlands Trial Register, with reference number NL9858.
Collapse
Affiliation(s)
- Susanne Pinto
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Dominika Šajbenová
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Elisa Benincà
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Sam Nooij
- Leiden University Center for Infectious Diseases (LUCID) Research, Leiden University Medical Center, Leiden, The Netherlands
| | - Elisabeth M Terveer
- Leiden University Center for Infectious Diseases (LUCID) Research, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Donor Feces Bank, LUCID Medical Microbiology & Infection Control, Leiden University Medical Center, Leiden, The Netherlands
| | - Josbert J Keller
- Netherlands Donor Feces Bank, LUCID Medical Microbiology & Infection Control, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gastroenterology, Haaglanden Medisch Centrum, The Hague, The Netherlands
| | | | - Johannes A Bogaards
- Department of Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity (AI&I), Amsterdam UMC, Amsterdam, The Netherlands
| | - Ewout W Steyerberg
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
48
|
Chong J, Chen Z, Ma J, He L, Zhu Y, Lu Z, Qiu Z, Chen C, Chen Y, Jiang F. Mechanistic investigation and the optimal dose based on baicalin in the treatment of ulcerative colitis-A preclinical systematic review and meta-analysis. BMC Gastroenterol 2025; 25:50. [PMID: 39901089 PMCID: PMC11792396 DOI: 10.1186/s12876-025-03629-0] [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: 09/21/2024] [Accepted: 01/20/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a type of inflammatory bowel disease, and current treatments often fall short, necessitating new therapeutic options. Baicalin shows therapeutic promise in UC animal models, but a systematic review is needed. METHODS A systematic search was conducted across databases including PubMed, EBSCO, Web of Science, and Science Direct, up to March 2024, identifying randomized controlled trials (RCTs) examining baicalin's impact on UC in animal models. Seventeen studies were selected through manual screening. Meta-analyses and subgroup analyses utilized Rev Man 5.3 and Stata 15.0 software to assess symptom improvement. RESULTS From 1304 citations, 17 were analyzed. Baicalin significantly modulated various biomarkers: HCS (SMD = -3.91), DAI (MD = -2.75), spleen index (MD = -12.76), MDA (SMD = -3.88), IL-6 (SMD = -10.59), IL-1β (SMD = -3.98), TNF-α (SMD = -8.05), NF-κB (SMD = -5.46), TLR4 (MD = -0.38), RORγ (MD = -0.89), MCP-1 (MD = -153.25), MPO (SMD = -7.34), Caspase-9 (MD = -0.93), Caspase-3 (MD = -0.45), FasL (MD = -1.20)) and enhanced BWC (MD = 0.06), CL (MD = 1.39), ZO-1 (MD = 0.44), SOD (SMD = 3.04), IL-10 mRNA (MD = 3.14), and FOXP3 (MD = 0.45) levels. Baicalin's actions may involve the PI3K/AKT, TLR4/NF-κB, IKK/IKB, Bcl-2/Bax, Th17/Treg, and TLRs/MyD88 pathways. Optimal therapeutic outcomes were predicted at dosages of 60-150 mg/kg over 10-14 weeks. CONCLUSION Baicalin demonstrates a multifaceted therapeutic potential in UC, attributed to its anti-inflammatory, antioxidant, anti-apoptotic, and intestinal barrier repair properties. While higher doses and longer treatments appear beneficial, further research, particularly human clinical trials, is necessary to verify its effectiveness and safety in people.
Collapse
Affiliation(s)
- Jinchen Chong
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Zepeng Chen
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, PR China
| | - Jiaze Ma
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Linhai He
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Yijia Zhu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Zhihua Lu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Zhengxi Qiu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
- Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Chen Chen
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China
| | - Yugen Chen
- Department of Colorectal Surgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, PR China.
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China.
- Jiangsu Collaborative Innovation Center of Chinese Medicine in Prevention and Treatment of Tumor, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China.
| | - Feng Jiang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, PR China.
- Department of Colorectal Surgery, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, PR China.
| |
Collapse
|
49
|
Wang X, Zhang G, Bian Z, Chow V, Grimaldi M, Carivenc C, Sirounian S, Li H, Sladekova L, Motta S, Luperi Y, Gong Y, Costello C, Li L, Jachimowicz M, Guo M, Hu S, Wilson D, Balaguer P, Bourguet W, Mani S, Bonati L, Peng H, March J, Wang H, Wang S, Krause HM, Liu J. An abundant ginger compound furanodienone alleviates gut inflammation via the xenobiotic nuclear receptor PXR in mice. Nat Commun 2025; 16:1280. [PMID: 39900639 PMCID: PMC11791082 DOI: 10.1038/s41467-025-56624-0] [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: 08/01/2024] [Accepted: 01/22/2025] [Indexed: 02/05/2025] Open
Abstract
The literature documenting the value of drug-like molecules found in natural products is vast. Although many dietary and herbal remedies have been found to be effective for treating intestinal inflammation, the identification of their active components has lagged behind. In this study, we find that a major ginger component, furanodienone (FDN), is a selective pregnane X receptor (PXR) ligand with agonistic transcriptional outcomes. We show that FDN binds within a sub-pocket of the PXR ligand binding domain (LBD), with subsequent alterations in LBD structure. Using male mice, we show that orally provided FDN has potent PXR-dependant anti-inflammatory outcomes that are colon-specific. Increased affinity and target gene activation in the presence of synergistically acting agonists indicates further opportunities for augmenting FDN activity, efficacy and safety. Collectively, these results support the translational potential of FDN as a therapeutic agent for the treatment and prevention of colonic diseases.
Collapse
Affiliation(s)
- Xiaojuan Wang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Guohui Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, People's Republic of China
| | - Zhiwei Bian
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Vimanda Chow
- Department of Chemistry, York University, Toronto, ON, Canada
| | - Marina Grimaldi
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Coralie Carivenc
- Centre de Biologie Structurale, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - Savannah Sirounian
- Centre de Biologie Structurale, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - Hao Li
- Department of Molecular Pharmacology; Department of Genetics; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lucia Sladekova
- Department of Molecular Pharmacology; Department of Genetics; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Stefano Motta
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy
| | - Yulia Luperi
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy
| | - Yufeng Gong
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
- College of Marine Life Sciences, Ocean University of China, Qingdao, Shandong, China
| | - Cait Costello
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Matthew Jachimowicz
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Miao Guo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, People's Republic of China
| | - Shian Hu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Derek Wilson
- Department of Chemistry, York University, Toronto, ON, Canada
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Université Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - William Bourguet
- Centre de Biologie Structurale, INSERM, CNRS, Université de Montpellier, Montpellier, France
| | - Sridhar Mani
- Department of Molecular Pharmacology; Department of Genetics; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Laura Bonati
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Milan, Italy
| | - Hui Peng
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
- School of the Environment, University of Toronto, Toronto, ON, Canada
| | - John March
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, USA
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, People's Republic of China.
| | - Henry M Krause
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| | - Jiabao Liu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
50
|
Wen Z, Zhu L, He W, Liang T, Zhong Q, Long J, Su L. Exploring the causal inference of inflammatory bowel disease and ischemic stroke: a bidirectional two-sample Mendelian randomization study. J Thromb Thrombolysis 2025; 58:340-348. [PMID: 39720960 DOI: 10.1007/s11239-024-03065-z] [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] [Accepted: 12/05/2024] [Indexed: 12/26/2024]
Abstract
Growing evidence suggests inflammatory bowel disease (IBD) is linked to ischemic stroke (IS); however, the results are inconclusive. Therefore, it remains uncertain whether the association between IBD and IS is causal. Herein, we performed a bidirectional Mendelian randomization (MR) study to examine the causal association of IBD with IS. We obtained summary-level data for IBD and IS from several publicly released genome-wide association studies to conduct a two-sample bidirectional Mendelian randomization (MR) analysis. Herein, the inverse-variance weighted method was utilized as the primary approach. Then, we applied the weighted median and MR-Egger estimators for the follow-up sensitivity analyses. In addition, the MR-Egger intercept test was performed to detect the potential directional pleiotropy. Genetically predicted IBD was not causally associated with IS and IS subtypes (IS: OR = 0.99, 95% CI 0.98-1.01, p = 0.49; large artery atherosclerosis stroke: OR = 1.00, 95% CI: 0.96-1.05, p = 0.88; cardioembolic stroke: OR = 0.99, 95% CI 0.96-1.03, p = 0.75; small-vessel occlusion stroke: OR = 1.02, 95% CI 0.99-1.05, p = 0.16). Moreover, we did not find a significant causal effect of UC or CD on IS and IS subtypes. Furthermore, there was no significant association observed between IS and IBD in the reverse MR analysis. The estimates were consistent across sensitivity analyses. Our MR analysis does not support a bidirectional causal association between IBD and IS, despite observational studies reporting an association of IBD with IS.
Collapse
Affiliation(s)
- Zheng Wen
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Lulu Zhu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China
| | - Wanting He
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China
| | - Tian Liang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China
| | - Qingqing Zhong
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China
| | - Jianxiong Long
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China.
| | - Li Su
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning, Guangxi, 530021, China.
| |
Collapse
|