1
|
Shin S, Chen S, Xie K, Duhun SA, Ortiz-Cerda T. Evaluating the anti-inflammatory and antioxidant efficacy of complementary and alternative medicines (CAM) used for management of inflammatory bowel disease: a comprehensive review. Redox Rep 2025; 30:2471737. [PMID: 40056427 PMCID: PMC11892051 DOI: 10.1080/13510002.2025.2471737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic autoimmune condition whose pathogenesis has not been fully elucidated, and current treatments are not definitive and often carry several side effects. The Complementary and Alternative Medicine (CAM) offers a new approach to conventional medicine. However, their clinical application and mechanisms remain limited.Objective: The aim of this review is to evaluate the anti-inflammatory, impact on microbiota and antioxidant efficacy of currently available CAM for IBD.Methods: The literature collection was obtained from Google Scholar, MEDLINE, PubMed and Web of Science (WOS). Studies in both human and animal models, published in English language between 2018 and 2024, were selected. Sixty-seven studies were included in the current review after inclusion and exclusion screening processes.Results: Mostly, studies showed significant anti-inflammatory, gut microbiota restoring, antioxidant effects of polyphenols, polysaccharides, emodin, short-chain fatty acids (SCFA; including butyrate, propionate and acetate), and probiotics although some contrasting results were noted. Current evidence shows that polyphenols exhibit the most consistent result in alleviating IBD pathophysiology, primarily due to their significant SCFA-elevating effect.Discussion: Future studies may focus on human studies, narrowing down on individual factors which may change natural product's metabolism. Further research studies are also essential to obtain therapeutic recommendations.
Collapse
Affiliation(s)
- Sia Shin
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Siqi Chen
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Kangzhe Xie
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Suehad Abou Duhun
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Tamara Ortiz-Cerda
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Departamento de Citología e Histología Normal y Patológica, Facultad de medicina, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
2
|
Ju Y, Ma C, Huang L, Tao Y, Li T, Li H, Huycke MM, Yang Y, Wang X. Inactivation of glutathione S-transferase alpha 4 blocks Enterococcus faecalis-induced bystander effect by promoting macrophage ferroptosis. Gut Microbes 2025; 17:2451090. [PMID: 39819335 PMCID: PMC11740687 DOI: 10.1080/19490976.2025.2451090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/08/2024] [Accepted: 01/02/2025] [Indexed: 01/30/2025] Open
Abstract
Enterococcus faecalis-infected macrophages produce 4-hydroxynonenal (4-HNE) that mediates microbiota-induced bystander effect (MIBE) leading to colorectal cancer (CRC). Glutathione S-transferase alpha 4 (Gsta4), a specific detoxifying enzyme for 4-HNE, is overexpressed in human CRC and E. faecalis-induced murine CRC. However, the roles of Gsta4 in E. faecalis-induced colitis and CRC remain unclear. Herein, we demonstrate that Gsta4 is essential for MIBE by protecting macrophages from E. faecalis-induced ferroptosis. E. faecalis OG1RFSS was used to induce colitis in Gsta4-/- and Il10-/-/Gsta4-/- mice by orogastric gavage. Ferroptosis was assessed in Gsta4-deficient murine macrophages. We found that, unlike Il10-/- mice, Gsta4-/- and Il10-/-/Gsta4-/- mice colonized with E. faecalis failed to develop colitis or CRC. Immunofluorescent staining showed a reduction of macrophages in the lamina propria of E. faecalis-colonized Il10-/-/Gsta4-/- mice, as well as decreased Gpx4 expression, indicating the occurrence of ferroptosis. Ferroptosis was further confirmed in Gsta4-deficient murine macrophages infected with E. faecalis. Moreover, Gsta4 inactivation induced the upregulation of Hmox1 and phosphorylated c-Jun while blocked Nos2 expression, leading to the accumulation of intracellular ferrous iron, lipid peroxidation and, eventually, ferroptosis. Finally, Mapk8, as a ferroptosis driver, was remarkably elevated in E. faecalis-infected Gsta4-deficient macrophages. These results suggest that Gsta4 inactivation blocks MIBE by eliminating macrophages, thereby attenuates E. faecalis-induced colitis and CRC.
Collapse
Affiliation(s)
- Yuanyuan Ju
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Genetics and Reproductive Medicine, Nantong, Jiangsu, China
| | - Chunhua Ma
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Genetics and Reproductive Medicine, Nantong, Jiangsu, China
| | - Lin Huang
- Department of Gastroenterology, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yumei Tao
- Department of Pathology, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
| | - Tianqi Li
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Genetics and Reproductive Medicine, Nantong, Jiangsu, China
| | - Haibo Li
- Department of Clinical Laboratory, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
| | - Mark M. Huycke
- Stephenson Cancer Center, Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Yonghong Yang
- Department of Nephrology, Rheumatology, and Immunology, Nantong Children’s Hospital, Nantong, Jiangsu, China
- Department of Pediatrics, Nantong Maternity and Child Healthcare Hospital, Nantong, Jiangsu, China
| | - Xingmin Wang
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity and Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
- Nantong Key Laboratory of Genetics and Reproductive Medicine, Nantong, Jiangsu, China
- Stephenson Cancer Center, Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
3
|
Zhang HL, Chang J, Sun CP, Huo ZP, Feng YL, Li PY, Jia YX, Hui SW, Zhu QM, Cai JY, He Y, Qiu F, Zhang J. Andrographolide and its sulfated metabolite alleviated DSS-induced inflammatory bowel disease through regulating inflammation and immune via MAPK/NLRP3 pathways and the balance of Th17/Treg cells. Mol Immunol 2025; 183:313-320. [PMID: 40441031 DOI: 10.1016/j.molimm.2025.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 04/30/2025] [Accepted: 05/14/2025] [Indexed: 06/11/2025]
Abstract
Inflammatory Bowel Disease (IBD), is a chronic illness characterized by severe abdominal pain, diarrhea, and weight loss, seriously diminishing patients' quality of life. Andrographolide (AND), a natural diterpenoid from Andrographis paniculata, and its sulfated metabolite, andrographolide sodium bisulfite (ASB), have showed potential anti-inflammatory effects. However, their mechanism in IBD remains elusive. This study investigated the impact of AND and its sulfated derivative ASB, on inflammatory responses in IBD. Our findings revealed that AND and ASB significantly reduced disease activity index (DAI) scores and enhanced intestinal barrier function in dextran sodium sulfate (DSS)-induced mice, thereby ameliorating the course of IBD. Furthermore, AND and ASB inhibited both the mitogen-activated protein kinase (MAPK) and NLRP3 pathways to reduce the release of inflammatory cytokines IL-6 and TNF-α. This mechanism was accompanied by a restoration of immune balance through the modulation of T-helper 17 (Th17) and regulatory T (Treg) cells. The ability of AND and ASB to mitigate chronic inflammation and maintain immune equilibrium presented a promising therapeutic approach for IBD management. These findings suggested that AND and ASB might provide novel therapeutic approaches for IBD, thereby warranting further investigation into their clinical efficacy for disease treatment and maintenance of remission.
Collapse
Affiliation(s)
- Hui-Lin Zhang
- School of Chinese Materia Medica, School of Medical Technology, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China; College of Pharmacy, Dalian Medical University, Dalian 116044, People's Republic of China
| | - Jing Chang
- School of Chinese Materia Medica, School of Medical Technology, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Cheng-Peng Sun
- School of Chinese Materia Medica, School of Medical Technology, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China.
| | - Zhi-Peng Huo
- School of Chinese Materia Medica, School of Medical Technology, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China; State Key Laboratory of Chinese Medicine Modernization, Tasly Pharmaceutical Group Co. Ltd., Tianjin 300410, People's Republic of China
| | - Yan-Li Feng
- School of Chinese Materia Medica, School of Medical Technology, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Peng-Yan Li
- School of Chinese Materia Medica, School of Medical Technology, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Ya-Xue Jia
- School of Chinese Materia Medica, School of Medical Technology, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Si-Wen Hui
- School of Chinese Materia Medica, School of Medical Technology, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Qi-Meng Zhu
- School of Chinese Materia Medica, School of Medical Technology, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China
| | - Jin-Yong Cai
- State Key Laboratory of Chinese Medicine Modernization, Tasly Pharmaceutical Group Co. Ltd., Tianjin 300410, People's Republic of China
| | - Yi He
- State Key Laboratory of Chinese Medicine Modernization, Tasly Pharmaceutical Group Co. Ltd., Tianjin 300410, People's Republic of China.
| | - Feng Qiu
- School of Chinese Materia Medica, School of Medical Technology, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China.
| | - Juan Zhang
- School of Chinese Materia Medica, School of Medical Technology, Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, People's Republic of China.
| |
Collapse
|
4
|
Chen Y, Shui M, Li H, Guo M, Yuan Q, Hao W, Wang T, Zhou H, Chen Z, Wang S. Inflammation-targeted delivery of probiotics for alleviation of colitis and associated cognitive disorders through improved vitality and colonization. Biomaterials 2025; 318:123163. [PMID: 39923539 DOI: 10.1016/j.biomaterials.2025.123163] [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: 10/20/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Oral probiotic biotherapies hold significant promise for addressing intestinal inflammatory disorders. Nonetheless, due to the challenging pathological microenvironment of the gastrointestinal tract, it is difficult to achieve deliver probiotics in an inflammation-targeted manner while improving their intestinal colonization and minimizing the impact of gastrointestinal environment on their vitality. To address this, an innovative probiotics oral delivery system (EcN-Apt@HG) against ulcerative colitis (UC) was developed by conjugating IL-6 aptamer to the surface of EcN and subsequently encapsulating the probiotics in a hydrogel consisting of aldehyde-functionalized chondroitin sulfate (CS) and Poly(amidoamine) (PAMAM). As expected, the encapsulated EcN demonstrated resistance to gastrointestinal conditions, and the colonization duration of probiotics in the colon was enhanced via the preferential adhesion effect of IL-6 aptamer on the inflammatory site. The EcN-Apt@HG system restored the damaged mucosal layer, suppressed hyperactive immune responses, and reshaped the dysbiosis of intestinal microflora, thereby synergistically alleviating dextran sulfate sodium (DSS)-induced colitis. Notably, EcN-Apt@HG significantly alleviated depression-like behaviors and cognitive impairment in colitis mice through gut-brain axis interaction. This approach provides a simple and promising strategy for inflammation-targeted delivery of probiotics to the intestine and shows great potential for UC therapy and associated cognitive disorders.
Collapse
Affiliation(s)
- Yi Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Mingju Shui
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hongyi Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Miao Guo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Qin Yuan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Wei Hao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Tao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hefeng Zhou
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Zhejie Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
5
|
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
|
6
|
Chen K, Wang H, Yang Y, Tang C, Sun X, Zhou J, Liu S, Li Q, Zhao L, Gao Z. Common mechanisms of Gut microbe-based strategies for the treatment of intestine-related diseases: based on multi-target interactions with the intestinal barrier. Cell Commun Signal 2025; 23:288. [PMID: 40528179 DOI: 10.1186/s12964-025-02299-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 06/07/2025] [Indexed: 06/20/2025] Open
Abstract
The concurrent occurrence and exacerbation of multiple diseases, including geriatric diseases and chronic diseases, impose a heavy burden on human health and medical expenses. Clarifying the common mechanisms of related multifarious diseases and developing preventive and therapeutic strategies with synergistic effects for multiple diseases are of great significance in alleviating the burden on the medical system and reducing patients' burden of drug metabolism. Recent studies have revealed that gut microbiota disorders and intestinal barrier damage, which consequently cause metabolic and immunological disorders, may be a common pathological basis underlying various intestinal-related diseases. In this review, we focus on the intestinal barrier function, summarizing the multi-target interactions and common mechanisms involved in diseases related to the gut such as ulcerative colitis, colorectal cancer, and type 2 diabetes. We identified gut microbe-based strategies, including probiotics, prebiotics, synbiotics, postbiotics, as well as potential targets in faecal microbiota transplant and berberine. The common mechanisms and key targets in the treatment of these diseases mainly include increasing the abundance of beneficial genera Bifidobacterium and Lactobacillus, increasing the levels of Short Chain Fatty Acids, restoring the intestinal mechanical barrier, and suppressing gut inflammation infiltration. We aim to provide a crucial basis and direction for the development of novel drugs with therapeutic effects for multiple diseases, thereby alleviating the patients' burden of medication and enhancing the efficacy of treatment.
Collapse
Affiliation(s)
- Keyu Chen
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
- Department of Endocrinology, Academy of Chinese Medical Sciences, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Han Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yingying Yang
- China-Japan Friendship Hospital, Beijing, 100029, China
| | - Cheng Tang
- National Key Laboratory of Efficacy and Mechanism on Chinese Medicine for Metabolic Diseases, Beijing Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaowei Sun
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Jie Zhou
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Shixu Liu
- Department of Traditional Chinese Medicine, School of Basic Medicine, Qingdao University, Qingdao, 266003, China
| | - Qingwei Li
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Linhua Zhao
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Zezheng Gao
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
7
|
Cristelo C, Viegas J, Barros AS, Almeida H, das Neves J, Sarmento B, Nunes R. Oral delivery of tunable oxidation-responsive budesonide-loaded nanoparticles enhances inflammation modulation in intestinal colitis. J Control Release 2025; 384:113948. [PMID: 40513667 DOI: 10.1016/j.jconrel.2025.113948] [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: 04/02/2025] [Revised: 05/31/2025] [Accepted: 06/09/2025] [Indexed: 06/16/2025]
Abstract
The rising global prevalence and socio-economic impact of Inflammatory Bowel Disease (IBD) highlight the pressing demand of innovative solutions. Drug-targeting technologies are urgently needed to effectively deliver drugs directly to the affected areas of the gastrointestinal tract (GIT). In this work, a surface-tunable nanosystem responsive to reactive oxygen species (ROS) was developed for the focal oral delivery of budesonide to IBD affected GIT areas. Poly(lactic-co-glycolic) acid (PLGA) nanoparticles (NPs) were functionalized with a dense hydrophilic polyethylene glycol (PEG) corona linked by a ROS-sensitive moiety to obtain cleavable PEG (CleavPEG) NPs. CleavPEG NPs with nearly 100 nm and high association efficiency (∼70 %) presented an oxidation-responsive in vitro release of budesonide highly associated (> 60 %) with epithelial intestinal cells and macrophages without decreasing cell metabolic activity. In an inflamed 3D intestinal model, budesonide association to NPs allowed for minimal permeation of budesonide, when compared to its free form, with a similar reduction of IL-8, CXCL10/IP-10 and CCL20/MIP3a. Moreover, in a DSS-induced colitis mice model, CleavPEG NPs accumulated more in the colon than PEG NPs without cleavable linker, and repeated oral treatment with budesonide-loaded CleavPEG NPs decreased intestinal inflammation: confirmed by colonoscopy and quantified by a disease activity index (DAI) and levels of pro-inflammatory cytokines in colon comparable to healthy animals. CleavPEG NPs were efficiently responsive to oxidative environment, and improved budesonide efficacy in resolving inflammation, showing promise for the treatment of IBD.
Collapse
Affiliation(s)
- Cecília Cristelo
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Juliana Viegas
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Andreia S Barros
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Helena Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - José das Neves
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto Universitário de Ciências da Saúde, CESPU, Avenida Central de Gandra, 1317, 4585-116 Gandra, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto Universitário de Ciências da Saúde, CESPU, Avenida Central de Gandra, 1317, 4585-116 Gandra, Portugal.
| | - Rute Nunes
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Instituto Universitário de Ciências da Saúde, CESPU, Avenida Central de Gandra, 1317, 4585-116 Gandra, Portugal
| |
Collapse
|
8
|
Luo J, Shi B, Hao C, Sun M, Xu L, Kuang H, Qu A. Chiral Zinc Sulfide Nanoparticles Scavenging Reactive Oxygen Species for Remodeling Intestinal Homeostasis. Angew Chem Int Ed Engl 2025; 64:e202503654. [PMID: 40170506 DOI: 10.1002/anie.202503654] [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: 02/13/2025] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/03/2025]
Abstract
Elevated levels of reactive oxygen species (ROS) and gut microbiota dysbiosis are crucial factors that exacerbate inflammatory bowel disease (IBD). To address this, we successfully synthesized zinc sulfide nanoparticles (ZnS NPs) with a particle size of approximately 500 nm and a maximum g-factor of 0.07, utilizing l-/d-cysteine as chiral ligands. Chirality gives NPs unique bioactivity. These chiral ZnS NPs could enter macrophages through the CD44 and clathrin pathways, which enhanced the ability to scavenge ROS, in turn significantly inhibited the NF-κB and NLRP3 signaling pathways, thereby reducing the secretion of TNF-α, IL-6, and IL-1β, while upregulating IL-10. In vivo experimental data showed that l-ZnS NPs outperformed 5-aminosalicylic acid, significantly improving body weight, reducing the IBD activity index, and attenuating tissue damage. Concurrently, l-ZnS NPs exhibited a marked prophylactic effect. The benchmark studies verified that l-ZnS NPs increased the abundance of the beneficial Lachnospiraceae NK4A136 by 10.55-fold and decreased harmful Enterobacter by 2914.00-fold, thereby reshaping the intestinal microecological balance. Pharmacokinetic and biosafety assessments confirmed the safety of l-ZnS NPs. Our findings indicate that chiral ZnS NPs hold great potential as nanodrugs for the treatment and prevention of IBD, providing an important foundation for the development of IBD therapeutic strategies.
Collapse
Affiliation(s)
- Jun Luo
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Baimei Shi
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Changlong Hao
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Maozhong Sun
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Liguang Xu
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hua Kuang
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Aihua Qu
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
9
|
Fan G, Liu Y, Tao L, Wang D, Huang Y, Yang X. Sodium butyrate alleviates colitis by inhibiting mitochondrial ROS mediated macrophage pyroptosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167756. [PMID: 40044062 DOI: 10.1016/j.bbadis.2025.167756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory bowel disease with unclear causes and limited treatment options. Sodium butyrate (NaB), a byproduct of dietary fiber in the intestine, has demonstrated efficacy in treating inflammation. However, the precise anti-inflammatory mechanisms of NaB in colon inflammation remain largely unexplored. This study aims to investigate the effects of NaB on dextran sulfate sodium (DSS)-induced colitis in rats. The findings indicate that oral administration of NaB effectively prevent colitis and reduce levels of serum or colon inflammatory factors. Additionally, NaB demonstrated in vitro inhibition of RAW264.7 inflammation cytokines induced by LPS, along with suppression of the ERK and NF-κB signaling pathway activation. Moreover, NaB mitigated LPS and Nigericin-induced RAW264.7 pyroptosis by reducing indicators of mitochondrial damage, including increased mitochondrial membrane potential (JC-1) levels and decreased Mito-ROS production. NaB increases ZO-1 and Occludin expression in CaCo2 cells by inhibiting RAW264.7 pyroptosis. These results suggest that NaB could be utilized as a therapeutic agent or dietary supplement to alleviate colitis.
Collapse
Affiliation(s)
- Guoqiang Fan
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yaxin Liu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Limei Tao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Danping Wang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yizhu Huang
- Singao Xiamen Company, Xiamen 361006, PR China
| | - Xiaojing Yang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
10
|
Qin S, Yang Z, Lei J, Xie Q, Jiang L, Fan Y, Luo Y, Wei K, Luo W, Yu B. Comparative efficacy of preventive vs. therapeutic resveratrol in modulating gut microbiota and alleviating inflammation in DSS-induced colitis. BMC Immunol 2025; 26:42. [PMID: 40437375 PMCID: PMC12121039 DOI: 10.1186/s12865-025-00718-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 05/06/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) management remains challenging due to limited preventive strategies and the low bioavailability of therapeutic agents like resveratrol (RSV). While RSV exhibits anti-inflammatory properties, its preventive potential via gut microbiome modulation remains unexplored. METHODS A murine colitis model was established using 2.5% DSS, with mice randomized into control (CON), DSS, therapeutic RSV treatment (RSV), and preventive RSV treatment (PRE) groups. Clinical outcomes, intestinal barrier integrity, inflammatory cytokines, macrophage polarization, TLR4/NF-κB signaling, and gut microbiota (16S rRNA sequencing) were systematically evaluated. RESULTS Preventive RSV (PRE) outperformed therapeutic RSV across all metrics. PRE attenuated colitis severity by 51.4% (weight loss, P < 0.001 vs. RSV) and restored mucosal architecture (P = 0.048 vs. DSS). Mechanistically, PRE normalized barrier function via transcriptional (ZO-1: 56.7% of CON; Occludin: 14-fold induction vs. DSS) and protein-level recovery (ZO-1: 96.5% of CON, P = 0.02), suppressed pro-inflammatory cytokines (TNF-α: 80.8%; IL-6: 69.9%; IL-18: >96%, P < 0.001 vs. DSS), and promoted M2 macrophage polarization (CD206: 1.7-fold vs. CON, P = 0.02) through TLR4/NF-κB inhibition (53% TLR4 reduction vs. 15% with RSV, P < 0.001). Despite comparable α-diversity between RSV and PRE, PRE uniquely enriched barrier-protective taxa (Lactococcus, Muribaculum) and restored microbial amino acid biosynthesis. Crucially, PRE's efficacy despite low systemic bioavailability implicated microbiome-mediated "luminal priming" as its primary mechanism. CONCLUSIONS This study redefines preventive RSV as a microbial ecosystem engineer that preemptively fortifies the gut against inflammation via microbiome-immune-metabolic crosstalk. By prioritizing ecological prevention over symptom suppression, our findings offer a transformative "food as medicine" strategy for IBD, highlighting RSV's potential as a chronotherapeutic agent to reshape clinical paradigms.
Collapse
Affiliation(s)
- Senmei Qin
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zongjing Yang
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jinqing Lei
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Qingli Xie
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Linsui Jiang
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yuanyuan Fan
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yonggu Luo
- Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Kecong Wei
- Department of Neurosurgery, Wuming Hospital of Guangxi Medical University, Wuming, 530199, Guangxi, China
| | - Wei Luo
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Bing Yu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China.
| |
Collapse
|
11
|
Xia Y, Zhou X, Cai P, Yan R, Ocansey DKW, Mao F. Flos lonicerae polysaccharide alleviates inflammatory bowel disease by improving intestinal microbiota and inhibiting oxidative stress and the NF-κB pathway. Int Immunopharmacol 2025; 159:114956. [PMID: 40424651 DOI: 10.1016/j.intimp.2025.114956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 05/21/2025] [Accepted: 05/22/2025] [Indexed: 05/29/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic disease that is increasingly being recognized globally. IBD is difficult to cure and is in urgent need of more treatment options. Flos lonicerae polysaccharide (FLP) is an effective component extracted from traditional Chinese medicine flos lonicerae, and previous studies have demonstrated its therapeutic potential in treating inflammatory diseases such as rhinitis and dermatitis, but there has been few in-depth research on their regulation of IBD repair mechanisms. This study focuses on the regulatory mechanisms of FLP on the structure of the gut microbiome and immune homeostasis. Results have demonstrated that treatment with FLP effectively attenuates weight loss, reduces the release of proinflammatory cytokines, and restores spleen and colon structures in mice. These effects are achieved through modifying the intestinal microbial community structure, increasing beneficial bacteria, and decreasing harmful bacteria. Furthermore, FLP significantly impacts the levels of Coenzyme Q2, a lipid closely associated with mitochondrial function. This modulation possibly regulates oxidative stress levels and the NF-κB pathway, thereby contributing to the alleviation of IBD. Therefore, FLP as a natural product with multiple biological activities, exhibits potential therapeutic effects in regulating intestinal microbiota, reducing oxidative stress, and inhibiting the NF-κB pathway, suggesting it may serve as a novel strategy for IBD treatment.
Collapse
Affiliation(s)
- Yuxuan Xia
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, Jiangsu, PR China
| | - Xiaodong Zhou
- Department of General Surgery, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, Jiangsu, PR China
| | - Peipei Cai
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou 215153, Jiangsu, PR China
| | - Ruhong Yan
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou 215153, Jiangsu, PR China
| | - Dickson Kofi Wiredu Ocansey
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, Jiangsu, PR China
| | - Fei Mao
- Department of Laboratory Medicine, the Affiliated People's Hospital, Jiangsu University, Zhenjiang 212002, Jiangsu, PR China.
| |
Collapse
|
12
|
Xiao Z, Xie J, Zhao X, Chen X, Lu Y, Xu Y, Wu M, An L, Li Q. Role of Pyroptosis in inflammatory bowel disease. Int Immunopharmacol 2025; 155:114619. [PMID: 40209313 DOI: 10.1016/j.intimp.2025.114619] [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: 01/30/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
Inflammatory bowel disease (IBD) is a serious chronic condition marked by persistent and recurrent intestinal ulcers. Although the exact cause of IBD remains unclear, it is generally accepted that a complex interaction among dietary factors, gut microbiota, and immune responses in genetically predisposed individuals contributes to its development. Pyroptosis, an inflammatory form of programmed cell death activated by inflammasomes, is marked by the rupture of cell membranes and the subsequent release of inflammatory mediators. Emerging evidence indicates that pyroptosis plays a crucial role in the pathogenesis of IBD. Moderate pyroptosis activation can enhance intestinal immune defenses, while excessive inflammasome activation can trigger an inflammatory cascade, resulting in increased damage to intestinal tissues. This article reviews the molecular mechanisms underlying pyroptosis and highlights its role in the onset and progression of IBD. Furthermore, We explore recent advancements in IBD treatment, focusing on small molecule compounds that specifically target and inhibit pyroptosis.
Collapse
Affiliation(s)
- Zhiyi Xiao
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Jiling Xie
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Xun Zhao
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Xiangjun Chen
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Yihong Lu
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Yuanzhao Xu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Manqing Wu
- Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Lingyue An
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| | - Qing Li
- Department of Gastroenterology and Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| |
Collapse
|
13
|
Pan Y, Zhao X, Chen Q, Zhao T, Ma Y, Wu H, Xiang Y, Jiang P, Li W, Yan Q, Mao S, Tao Y, Wang L, Zhu Y, Xu G. Faecalibacterium Prausnitzii extracellular vesicles regulating macrophage differentiation via homologous recombination repair in colitis model. Microbiol Res 2025; 298:128217. [PMID: 40393169 DOI: 10.1016/j.micres.2025.128217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/22/2025]
Abstract
Inflammatory Bowel Disease (IBD) is characterized by chronic inflammation influenced by the depletion of beneficial gut microbiota, a critical factor in disease onset and progression. This study investigates the therapeutic potential of extracellular vesicles (EVs) derived from Faecalibacterium prausnitzii (F.p EVs), a commensal bacterium whose reduction is linked to IBD. Our research demonstrates that F.p EVs are preferentially taken up by macrophages, where they exert their anti-inflammatory effects through the enhancement of homologous recombination (HR) repair mechanisms. Specifically, F.p EVs upregulate the expression of key proteins involved in HR repair, such as BRCA1 and BRCA2, thereby reducing DNA damage and inhibiting the cGAS-STING pathway, which is central to the inflammatory response. This modulation of macrophage function results in decreased pro-inflammatory cytokine production and enhanced intestinal barrier integrity. By elucidating these mechanisms, our study provides a clear understanding of how F.p EVs can be used to target fundamental aspects of IBD pathology, laying the groundwork for the development of more effective and targeted therapies.
Collapse
Affiliation(s)
- Yinya Pan
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Xinlu Zhao
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Qiongyun Chen
- Department of Gastroenterology, Taikang Xianlin Drum Tower Hospital, Nanjing, Jiangsu 21008, China
| | - Tao Zhao
- Department of Gastroenterology, Taikang Xianlin Drum Tower Hospital, Nanjing, Jiangsu 21008, China
| | - Yichun Ma
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 21008, China
| | - Hai Wu
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Ying Xiang
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 21008, China
| | - Ping Jiang
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Wenjun Li
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Qiong Yan
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 21008, China
| | - Shangtao Mao
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yufei Tao
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Lei Wang
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - Yun Zhu
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Nanjing Medical Center for Clinical Pharmacy, Nanjing 210008, China.
| | - Guifang Xu
- Department of Gastroenterology, Department of Pharmacy, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Gastroenterology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing Medical University, Nanjing 210008, China; Department of Gastroenterology, Taikang Xianlin Drum Tower Hospital, Nanjing, Jiangsu 21008, China; Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 21008, China.
| |
Collapse
|
14
|
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
|
15
|
Liu J, Huang H, Zhang X, Shen Y, Jiang D, Hu S, Li S, Yan Z, Hu W, Luo J, Yao H, Chen Y, Tang B. Unveiling the Cuproptosis in Colitis and Colitis-Related Carcinogenesis: A Multifaceted Player and Immune Moderator. RESEARCH (WASHINGTON, D.C.) 2025; 8:0698. [PMID: 40370501 PMCID: PMC12076167 DOI: 10.34133/research.0698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 05/16/2025]
Abstract
Cuproptosis represents a novel mechanism of cellular demise characterized by the intracellular buildup of copper ions. Unlike other cell death mechanisms, its distinct process has drawn considerable interest for its promising applications in managing inflammatory bowel disease (IBD) and colorectal cancer (CRC). Emerging evidence indicates that copper metabolism and cuproptosis may exert dual regulatory effects within pathological cellular environments, specifically modulating oxidative stress responses, metabolic reprogramming, and immunotherapeutic efficacy. An appropriate level of copper may promote disease progression and exert synergistic effects, but exceeding a certain threshold, copper can inhibit disease development by inducing cuproptosis in pathological cells. This makes abnormal copper levels a potential new therapeutic target for IBD and CRC. This review emphasizes the dual function of copper metabolism and cuproptosis in the progression of IBD and CRC, while also exploring the potential application of copper-based therapies in disease treatment. The analysis further delineates the modulatory influence of tumor immune microenvironment on cuproptosis dynamics, while establishing the therapeutic potential of cuproptosis-targeted strategies in circumventing resistance to both conventional chemotherapeutic agents and emerging immunotherapies. This provides new research directions for the development of future cuproptosis inducers. Finally, this article discusses the latest advances in potential molecular targets of cuproptosis and their related genes in the treatment of IBD and CRC, highlighting future research priorities and unresolved issues.
Collapse
Affiliation(s)
- Jingwen Liu
- Department of Gastroenterology, the Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Hairuo Huang
- China Medical University, Shenyang 110122, China
| | - Xiaojie Zhang
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Yang Shen
- Department of Radiation Oncology, Zhongshan Hospital,
Fudan University, Shanghai 200000, China
| | - DeMing Jiang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering,
Zhejiang University, Hangzhou 310007, China
| | - Shurong Hu
- Department of Gastroenterology, the Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Shuyan Li
- Department of Nursing, the Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zelin Yan
- Department of Gastroenterology, the First Affiliated Hospital of Zhejiang Chinese Medical University,
Zhejiang Provincial Key Laboratory of Gastrointestinal Diseases Pathophysiology, Hangzhou 310006, China
| | - Wen Hu
- Department of Gastroenterology, the First Affiliated Hospital of Zhejiang Chinese Medical University,
Zhejiang Provincial Key Laboratory of Gastrointestinal Diseases Pathophysiology, Hangzhou 310006, China
| | - Jinhua Luo
- The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| | - Haibo Yao
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital,
Key Laboratory of Gastroenterology of Zhejiang Province, Hangzhou 310014, China
| | - Yan Chen
- Department of Gastroenterology, the Second Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Bufu Tang
- Department of Interventional Radiology, Zhongshan Hospital,
Fudan University, Shanghai 200000, China
| |
Collapse
|
16
|
Xu G, Zhou J, Liu K, Wang Y, Tsikari T, Qin F, van den Hil F, Boor PPC, Ayada I, de Vries AC, Li J, Jiang S, Offermans DM, Kainov DE, Janssen HLA, Peppelenbosch MP, Bijvelds MJC, Wang W, Orlova VV, Pan Q, Li P. Macrophage-augmented intestinal organoids model virus-host interactions in enteric viral diseases and facilitate therapeutic development. Nat Commun 2025; 16:4475. [PMID: 40368896 PMCID: PMC12078800 DOI: 10.1038/s41467-025-59639-9] [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: 11/16/2024] [Accepted: 04/29/2025] [Indexed: 05/16/2025] Open
Abstract
The pathogenesis of enteric viral infections is attributed to both viral replication and the resultant immune-inflammatory response. To recapitulate this complex pathophysiology, we engineer macrophage-augmented organoids (MaugOs) by integrating human macrophages into primary intestinal organoids. Echovirus 1, echovirus 6, rotavirus, seasonal coronavirus OC43 and SARS-CoV-2- known to directly invade the intestine- are used as disease modalities. We demonstrate that these viruses efficiently propagate in MaugOs and stimulate the host antiviral response. However, rotavirus, coronavirus OC43 and SARS-CoV-2, but not the two echoviruses, trigger inflammatory responses. Acetate, a microbial metabolite abundantly present in the intestine, potently inhibits virus-induced inflammatory responses in MaugOs, while differentially affecting viral replication in macrophages and organoids. Furthermore, we provide a proof-of-concept of combining antiviral agent with either anti-inflammatory regimen or acetate to simultaneously inhibit viral infection and inflammatory response in MaugOs. Collectively, these findings demonstrate that MaugOs are innovative tools for studying the complex virus-host interactions and advancing therapeutic development.
Collapse
Affiliation(s)
- Guige Xu
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, 271018, China
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiangrong Zhou
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Kuan Liu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Yining Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Theano Tsikari
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Fang Qin
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, China
| | - Francijna van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick P C Boor
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Ibrahim Ayada
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Annemarie C de Vries
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Jiajing Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Shijin Jiang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, 271018, China
| | - Dewy M Offermans
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Denis E Kainov
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7028, Trondheim, Norway
| | - Harry L A Janssen
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
- Toronto Centre for Liver Disease, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Marcel J C Bijvelds
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Wenshi Wang
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, China
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Pengfei Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands.
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
17
|
Li H, Zhang Y, Du S, Shen J, Liu X, Jing J. "Remodeling the intestinal immune microenvironment": immune regulation and tissue regeneration by mesenchymal stem/stromal cells in the repair microenvironment of inflammatory bowel disease. Front Immunol 2025; 16:1543702. [PMID: 40433382 PMCID: PMC12106535 DOI: 10.3389/fimmu.2025.1543702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
The global prevalence of inflammatory bowel disease (IBD) has significantly increased in recent decades. IBD is a long-term, recurring, gastrointestinal inflammatory condition that mainly comprises two primary clinical types: ulcerative colitis and Crohn's disease. The current treatment paradigm for IBD primarily focuses on symptom management. However, this approach does not support mucosal epithelial repair, maintenance of barrier homeostasis, or regulation of biological functions in the gut. Conventional therapies rely on the frequent use of high-dose medications, including antibiotics, nonsteroidal anti-inflammatory drugs, biological agents, and immunomodulators. Recently, mesenchymal stem/stromal cells (MSCs) have gained interest in tissue regeneration owing to their unique ability to differentiate and secrete regulatory factors, including extracellular vesicles (EVs), which play crucial roles in abnormal organization. Various routes of administration have been explored in preclinical and clinical studies to deliver MSCs from diverse tissue sources. The routes include intraperitoneal, intravenous, and local (intracolonic or rectal) delivery. The MSCs employed were obtained from various tissues, including bone marrow, umbilical cord, and adipose tissue. This article reviews the research framework for the application of MSCs and EVs secretion in the treatment of IBD, emphasizing key immunological effects, such as immune microenvironment regulation, intestinal barrier stabilization, and therapeutic approaches targeting intestinal barrier disorders. The discussion primarily focuses on the advantages of MSCs over other biologics, impairment of gut mucosal tissue-resident mesenchymal stem cells in IBD development, immune targets (at the cellular and molecular levels) within the framework of IBD, and the reparative effects of MSCs in the microenvironment of IBD. We aimed to present an overview of the current trends in MSC research and therapy, as well as to identify the challenges and future directions that must be addressed to advance research on MSC-mediated therapeutic strategies for IBD.
Collapse
Affiliation(s)
| | | | | | | | | | - Jie Jing
- School and Hospital of Stomatology, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
18
|
Xia L, Pan Y, Wang X, Hu R, Gao J, Chen W, He K, Cui D, Zhao Y, Liu L, Lai L, Su M. ERMAP attenuates DSS-induced colitis in mice by regulating macrophage and T cell functions. BMC Gastroenterol 2025; 25:362. [PMID: 40355813 PMCID: PMC12070682 DOI: 10.1186/s12876-025-03840-z] [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: 10/29/2024] [Accepted: 04/02/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND & AIMS Both macrophages and T cells play a critical role in inflammatory bowel disease (IBD) development. Since our previous studies have shown that a novel immune checkpoint molecule erythrocyte membrane-associated protein (ERMAP) affects macrophage polarization and negatively regulates T cell responses, we investigated the effects of ERMAP on DSS-induced colitis progression in mice. METHODS C57BL/6 mice developed a dextran sodium sulfate (DSS) colitis model, treated with control Fc protein (Control Ig) and ERMAP-Fc fusion protein (ERMAP-Ig) for 12 days to assess colitis severity by disease activity index (DAI), weight loss, colon length, histology, flow cytometry, Q-PCR, WB, ELISA, and the effect of adoptive transfer of ERMAP knockout mice (ERMAP-/-) peritoneal macrophages on DSS colitis mice. In vitro, the effects of the RAW264.7 macrophage cell line that interfered with ERMAP expression on macrophage polarization and T cells were analyzed by flow cytometry. RESULTS We show here that administration of ERMAP protein significantly increases the proportion of anti-inflammatory M2-type macrophages and inhibits T cell activation and proliferation in DSS-induced colitis mice. Knockdown of ERMAP in RAW264.7 macrophages reduces M2-type macrophage polarization and increases T cell responses. Adoptive transfer of macrophages from ERMAP-/- exacerbates DSS-induced colitis. Global gene expression analysis by RNA-seq shows that ERMAP inhibits the NOD-like receptor (NLR) protein family pathway in macrophages. CONCLUSIONS In summary, our results suggest that administration of ERMAP can protect DSS-induced colitis in mice by regulating T cell and macrophage functions. This study adds to the evidence for various mechanistic pathways associated to the pathogenesis of IBD, which could subsequently be translated to novel therapeutics.
Collapse
Affiliation(s)
- Lu Xia
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Department of Histology and Embryology, Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China
- Key Laboratory for Research on Autoimmune Diseases of Higher Education schools in Guizhou Province, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Yiwen Pan
- Department of Histology and Embryology, Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Xianbin Wang
- Department of Histology and Embryology, Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Rong Hu
- Translational Medicine Research Center of Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Jie Gao
- Translational Medicine Research Center of Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Wei Chen
- Department of Histology and Embryology, Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Keke He
- Department of Histology and Embryology, Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Dongbin Cui
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Youbo Zhao
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China
| | - Lu Liu
- The Public Health Clinical Center of Guiyang City, 6 Daying Road, Guiyang City, 550004, Guizhou, China.
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, 1390 Storrs Road, Storrs, CT, 06269, USA.
| | - Min Su
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
- Department of Histology and Embryology, Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
- Key Laboratory for Research on Autoimmune Diseases of Higher Education schools in Guizhou Province, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
- Key Laboratory of Adult Stem Cell Translational Research (Chinese Academy of Medical Sciences), Guizhou Medical University, 6 Ankang Avenue, Guian New District, Guizhou, 561113, China.
| |
Collapse
|
19
|
Zhou M, Pei B, Cai P, Yi C, Akanyibah FA, Lyu C, Mao F. Human umbilical cord mesenchymal stem cell-derived exosomes repair IBD by activating the SIRT1-FXR pathway in macrophages. Stem Cell Res Ther 2025; 16:233. [PMID: 40346712 PMCID: PMC12065267 DOI: 10.1186/s13287-025-04365-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD), a chronic immune disorder, has increasing global incidence and poor treatment outcome. Abnormal macrophage function is implicated in the pathophysiology of IBD. In this study, we investigated the mechanism by which human umbilical cord mesenchymal stem cell-derived exosomes (hucMSC-Ex) inhibit inflammation in IBD mouse and macrophage inflammation models. METHODS We established a dextran sodium sulfate (DSS)-induce BALB/c mice model of IBD and treated with hucMSC-Ex via tail vein to evaluate their repair effect on IBD mice. An in vitro macrophage inflammation model was established using lipopolysaccharide (LPS) and Nigericin (Nig) by stimulating mouse macrophage RAW264.7 and human myeloid leukemia mononuclear (THP-1) cells to assess the repair effect of hucMSC-Ex on macrophage inflammation. EX 527, an effective inhibitor of silent information regulator of transcription 1 (SIRT1), was employed in both the in vivo and in vitro models to explore the effect of hucMSC-Ex on the SIRT1-FXR (farnesoid X receptor) pathway in macrophages during the attenuation of inflammation. RESULTS HucMSC-Ex effectively inhibited inflammation in both the in vivo and in vitro models by up-regulating the expressions of SIRT1 and FXR, which reduced the acetylation level of FXR and inhibited the activation of NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome. The addition of EX 527 further proved that hucMSC-Ex can reduce the acetylation of FXR by activating the SIRT1-FXR pathway, and the decrease of FXR acetylation was directly related to the inhibition of the activity of the NLRP3 inflammasome. CONCLUSION HucMSC-Ex alleviates IBD by reducing the acetylation level of FXR through activating the SIRT1-FXR pathway in macrophages and directly negatively regulating the activation of NLRP3 inflammasomes, thus inhibiting the occurrence of the inflammatory process.
Collapse
Affiliation(s)
- Mengjiao Zhou
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212013, P. R. China
| | - Bing Pei
- Department of Clinical Laboratory, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, Jiangsu, 223800, P. R. China
| | - Peipei Cai
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang College, Zhenjiang, Jiangsu, 212028, P. R. China
| | - Francis Atim Akanyibah
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China
| | - Changkun Lyu
- School of Medical Technology, Shangqiu Medical College Shangqiu, Shangqiu, Henan, 476100, P. R. China
| | - Fei Mao
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, No.8 Dianli Road, Zhenjiang, Jiangsu, 212002, P. R. China.
- Institute of Hematology, Jiangsu University, Zhenjiang, Jiangsu, 212013, P. R. China.
| |
Collapse
|
20
|
Chen CH, Yu KC, Hsu LJ, Chiu WT, Hsu KS. Pro-inflammatory macrophages contribute to developing comorbid anxiety-like behaviors through gastrointestinal vagal afferent signaling in experimental colitis mice. Brain Behav Immun 2025; 128:620-633. [PMID: 40348137 DOI: 10.1016/j.bbi.2025.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 04/09/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025] Open
Abstract
Anxiety symptoms are commonly observed in individuals with inflammatory bowel disease (IBD), but the mechanistic link between IBD and comorbid anxiety remains incompletely understood. Our previous study revealed that vagal gut-brain signaling contributes to driving comorbid anxiety-like behaviors in dextran sulfate sodium (DSS)-induced colitis mice, but how vagus nerve senses and transmits information to the brain in response to changes in the colonic microenvironment following DSS treatment remain elusive. Here, we identify a critical contribution of pro-inflammatory CD86+ macrophages to activate gut-innervating vagal afferents and ultimately drive anxiety-like behaviors in DSS-treated mice. An increased number of F4/80+ macrophages accumulated closely with gut-innervating vagal afferent fibers following DSS treatment. Depletion of macrophages alleviated DSS-induced anxiety-like behaviors, whereas peripheral delivery of lipopolysaccharide-activated M1 macrophages promoted anxiety-like behaviors, which were prevented by bilateral vagal afferent ablation. Moreover, differential expression levels of anxiety-like behaviors were positively correlated with neuronal activity changes in the nucleus tractus solitarius, locus coeruleus, and basolateral amygdala. Finally, treatment with either anti-α4β7 integrin antagonist vedolizumab or neutralizing anti-interleukin-1β monoclonal antibody effectively alleviated DSS-induced anxiety-like behaviors. Collectively, these findings unravel a mechanism of macrophage-to-vagus nerve communication via cytokine signaling responsible for comorbid anxiety associated with experimental colitis and suggest that pro-inflammatory CD86+ macrophages may represent a potential therapeutic target for psychological comorbidities in patients with IBD.
Collapse
Affiliation(s)
- Chin-Hao Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Kuan-Chieh Yu
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Li-Jin Hsu
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - Kuei-Sen Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
21
|
Liu K, Tian X, Fei S, Song Y, Abd El-Aty AM, Tan M. Macrophage and mitochondrion dual-targeting astaxanthin nanoparticles prepared by Maillard reaction for colonic inflammation alleviation. MARINE LIFE SCIENCE & TECHNOLOGY 2025; 7:352-365. [PMID: 40417255 PMCID: PMC12102426 DOI: 10.1007/s42995-024-00255-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 08/21/2024] [Indexed: 05/27/2025]
Abstract
This study demonstrated the design of whey protein isolate (WPI)-mannose (Man) conjugates with triphenylphosphonium bromide (TPP) through self-assembly to prepare macrophage and mitochondrion dual-targeting astaxanthin (AXT) nanoparticles (AXT@TPP-WPI-Man). The nanoparticles displayed spherical structures with a well-dispersed size of approximately 206.1 ± 39.2 nm, with good biocompatibility, stability, and targeting capabilities. In vitro experiments demonstrated the specific accumulation of AXT@TPP-WPI-Man in mitochondria and exhibited good targeting ability toward macrophages. The AXT@TPP-WPI-Man effectively reduced reactive oxygen species and preserved the normal mitochondrial membrane potential. The AXT@TPP-WPI-Man treated ulcerative colitis mice exhibited a 52.32% increase in colon length with significant improvement in weight loss, disease activity index scores, and reduced release of inflammatory cytokines. Immunofluorescence staining indicated AXT@TPP-WPI-Man alleviated ulcerative colitis by reducing M1 polarization in colonic macrophages while promoting M2 polarization. The dual-targeting AXT@TPP-WPI-Man has the potential to improve astaxanthin bioavailability, presenting a promising delivery method for the treatment of ulcerative colitis. Supplementary Information The online version contains supplementary material available at 10.1007/s42995-024-00255-9.
Collapse
Affiliation(s)
- Kangjing Liu
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034 China
- School of Food Science and Technology, Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian, 116034 China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 China
| | - Xueying Tian
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034 China
- School of Food Science and Technology, Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian, 116034 China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 China
| | - Siyuan Fei
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034 China
- School of Food Science and Technology, Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian, 116034 China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 China
| | - Yukun Song
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034 China
- School of Food Science and Technology, Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian, 116034 China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 China
| | - A. M. Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211 Egypt
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, 25240 Erzurum, Turkey
| | - Mingqian Tan
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034 China
- School of Food Science and Technology, Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian, 116034 China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034 China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034 China
| |
Collapse
|
22
|
Zhu Y, Fang Z, Bai J, Wang L, Chen J, Zhang Z, Wang Q, Sheng W, Pan X, Gao Z, Xu D, Wu P, Sun B. Orally Administered Functional Polyphenol-Nanozyme-Armored Probiotics for Enhanced Amelioration of Intestinal Inflammation and Microbiota Dysbiosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411939. [PMID: 40067175 PMCID: PMC12061243 DOI: 10.1002/advs.202411939] [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] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/23/2025] [Indexed: 05/04/2025]
Abstract
Maintaining microbiota balance and enhancing the antioxidant performance of nanozyme-based probiotic systems are crucial for effective inflammatory bowel disease (IBD) therapy. Despite significant advancements, developing a green and safe coating technology that functionalizes probiotics with nanozymes while preserving the activity of both components remains a challenge. To address this, chitosan-modified epigallocatechin gallate (EGCG-CS, EC)is synthesized, leveraging the intrinsic adhesive and coordination properties of polyphenols to capture gold nanozymes (AuNPs), forming ECA complexes that enhance nanozyme activity. When coated onto Escherichia coli Nissle 1917 (EcN), the resulting ECA@EcN system effectively scavenged reactive oxygen species (ROS), improving probiotic viability and promoting colon accumulation. Mechanistically, ECA protected EcN by suppressing the activation of the Flagellar Assembly and Branched-Chain Amino Acid Synthesis pathways, ultimately alleviating inflammation and modulating intestinal microbial communities to relieve IBD symptoms. Given the biocompatibility of its components and the environmentally friendly assembly approach, this polyphenol-nanozyme-armored probiotic system represents a promising platform for IBD treatment.
Collapse
Affiliation(s)
- Yong Zhu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Ziqu Fang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Jie Bai
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Longhui Wang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Jiaqing Chen
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Zehua Zhang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Qiang Wang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Weiwei Sheng
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Xueyin Pan
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Zhenyuan Gao
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Dengqiu Xu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Pengkai Wu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Beicheng Sun
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| |
Collapse
|
23
|
Tan T, Li J, Fan W, Shang K, Yang C, Liu X, Zhu S, Liu T, Wang J, Li Y, Lin Y. Tetrahedral Framework Nucleic Acid Relieves Sepsis-Induced Intestinal Injury by Regulating M2 Macrophages. Cell Prolif 2025; 58:e13803. [PMID: 39844345 PMCID: PMC12099223 DOI: 10.1111/cpr.13803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/22/2024] [Accepted: 12/30/2024] [Indexed: 01/24/2025] Open
Abstract
This study aimed to clarify the role and mechanism of tetrahedral framework nucleic acids (tFNAs) in regulating M2 macrophages to reduce intestinal injury. An intestinal injury model was established by intraperitoneal injection of lipopolysaccharides (LPS) in mice to explore the alleviating effects of tFNAs on intestinal injury. Inflammatory factors were detected by quantitative polymerase chain reaction (qPCR) and enzyme-linked immunosorbent assay (ELISA). The intestinal barrier and permeability were assessed using western blotting and immunohistochemistry. Macrophages in the gut were localised and quantified using immunofluorescence. Western blotting was used to investigate the role and mechanism of tFNAs in regulating macrophages and alleviating inflammation in the injured intestines. These results show that tFNAs attenuated sepsis-induced intestinal injury. tFNAs can also promote the intestinal barrier reconstruction and reduce intestinal permeability. In vivo, tFNAs accelerated the aggregation of M2 macrophages at an early stage of injury and reduced the number of M1 macrophages in the intestine. In addition, tFNAs enhanced the clearance ability of intestinal macrophages. They activated the signalling and transcription activating factor 1(STAT1) and cytokine signalling inhibitory factor 1/3 (SOCS1/3) pathways by increasing the expression of the phagocytic receptor Mertk. These findings indicated that tFNAs can alleviate sepsis-induced intestinal injury by regulating M2 macrophages, providing a new option for treating intestinal injury.
Collapse
Affiliation(s)
- Tingting Tan
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
- Key Laboratory of Pathogen‐Host InteractionMinistry of EducationBeijingPeople's Republic of China
- School of Basic Medicine and Clinical PharmacyChina Pharmaceutical UniversityNanjingPeople's Republic of China
| | - Jiajie Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan University, Chengdu, Department of Burns and Plastic SurgeryChengduPeople's Republic of China
| | - Wensi Fan
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
| | - Kangni Shang
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
- School of Basic Medicine and Clinical PharmacyChina Pharmaceutical UniversityNanjingPeople's Republic of China
| | - Chujun Yang
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
| | - Xiaohao Liu
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
| | - Shihui Zhu
- Shanghai Children's Medical CenterShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Tong Liu
- Department of Critical Care Medicine, Zhongshan HospitalFudan UniversityShanghaiPeople's Republic of China
| | - Junjie Wang
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
- Key Laboratory of Pathogen‐Host InteractionMinistry of EducationBeijingPeople's Republic of China
| | - Yingchuan Li
- Department of Critical Care Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiPeople's Republic of China
- Key Laboratory of Pathogen‐Host InteractionMinistry of EducationBeijingPeople's Republic of China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan University, Chengdu, Department of Burns and Plastic SurgeryChengduPeople's Republic of China
| |
Collapse
|
24
|
Lu H, Zhou Q, Li J, Xu S, Yu L, Zhu Y, Zhang H, Shi C, Zuo T, Xu M, Su M, Zhang Y, Hu R, Shubhra QTH, Deng H, Hu X, Cai X. CO-Releasing Polyoxometalates Nanozyme with Gut Mucosal Immunity and Microbiota Homeostasis Remodeling Effects for Restoring Intestinal Barrier Integrity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500116. [PMID: 40079617 PMCID: PMC12061238 DOI: 10.1002/advs.202500116] [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] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Disruption of the intestinal epithelial barrier, driven by imbalances in gut mucosal immunity and microbial homeostasis, is central to the onset and progression of inflammatory bowel disease (IBD). This study introduces a CO-releasing polyoxometalates (POMs) nanozyme (PMC), synthesized by coordinating pentacarbonyl manganese bromide with molybdenum-based POM nanoclusters. PMC demonstrates targeted accumulation at IBD-affected sites, efficient scavenging of reactive oxygen species (ROS), and responsive CO release, resulting in multiple therapeutic effects. Extensive in vitro and in vivo studies have validated the exceptional capacity of PMC to repair intestinal barrier, attributed to their potent antioxidant and anti-inflammatory properties, thereby achieving significant therapeutic efficacy in ulcerative colitis treatment. 16S rRNA sequencing indicated that PMC efficiently remodeled the gut microbiota composition. Single-cell RNA sequencing indicates a reduction in pro-inflammatory M1 macrophages, alongside suppressed ROS and inflammatory signaling pathways. Concurrently, an increase in reparative M2 macrophages and intestinal stem cells is observed, in addition to significant activation of the VEGF signaling pathway in macrophages and the NOTCH pathway in stem cells, underscoring the potential of PMC to restore immune balance and promote tissue repair. This study positions PMC as a promising, multifunctional therapeutic agent for IBD treatment owing to its robust intestinal barrier-restoring capability.
Collapse
Affiliation(s)
- Hongyang Lu
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Qiang Zhou
- Ruian People's HospitalThe Third Affiliated Hospital of Wenzhou Medical UniversityWenzhou325016China
| | - Jiayu Li
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Shengming Xu
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Li Yu
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Yinci Zhu
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - He Zhang
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Chengge Shi
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Tianci Zuo
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Mengzhu Xu
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Mingli Su
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Yanmei Zhang
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Rongdang Hu
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Quazi T. H. Shubhra
- Institute of ChemistryUniversity of Silesia in KatowiceSzkolna 9Katowice40‐006Poland
| | - Hui Deng
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Xiaowen Hu
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Xiaojun Cai
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| |
Collapse
|
25
|
Li C, Liu W, Fu A, Yang H, Yi G. Potential therapeutic strategies targeting efferocytosis for inflammation resolution and tissue repair in inflammatory bowel disease. Cell Immunol 2025; 411-412:104957. [PMID: 40315792 DOI: 10.1016/j.cellimm.2025.104957] [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: 08/21/2024] [Revised: 04/11/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025]
Abstract
Efferocytosis, the process by which apoptotic cells (ACs) are recognized and cleared by phagocytes, is a critical mechanism in maintaining intestinal immune homeostasis and promoting the resolution of inflammation. Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), is characterized by chronic intestinal inflammation, wherein defective efferocytosis contributes to the accumulation of ACs, secondary necrosis, and sustained mucosal damage. This review delineates the molecular mechanisms underlying efferocytosis and systematically examines its functional roles across five key intestinal phagocytic cell types: macrophages, dendritic cells (DCs), neutrophils, intestinal epithelial cells (IECs), and Paneth cells (PCs). Particular emphasis is placed on the dysregulation of efferocytosis capacity in IBD pathogenesis and the consequences of impaired apoptotic cell clearance in both professional and non-professional phagocytes. Furthermore, we evaluate emerging therapeutic strategies designed to restore or enhance efferocytosis, including modulation of macrophage polarization, LC3-associated phagocytosis pathways, nanotechnology-enabled delivery systems, and stem cell-based interventions. A comprehensive understanding of cell-type-specific efferocytosis in the intestinal microenvironment offers promising directions for the development of targeted, inflammation-resolving therapies for IBD.
Collapse
Affiliation(s)
- Chaoquan Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Wanting Liu
- Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China
| | - Aoni Fu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Haotian Yang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Guanghui Yi
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Institute of Pharmacy and Pharmacology, Hunan province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
26
|
Gao Z, Shao S, Xu Z, Nie J, Li C, Du C. IDO1 induced macrophage M1 polarization via ER stress-associated GRP78-XBP1 pathway to promote ulcerative colitis progression. Front Med (Lausanne) 2025; 12:1524952. [PMID: 40370742 PMCID: PMC12075526 DOI: 10.3389/fmed.2025.1524952] [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: 11/08/2024] [Accepted: 04/17/2025] [Indexed: 05/16/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disorder distinguished by alternating phases of remission and exacerbation. Restoring immune balance through the modulation of M1 macrophage polarization represents a potentially valuable therapeutic strategy for UC. Indoleamine 2,3-dioxygenase-1 (IDO1) has been shown to contribute to macrophage plasticity, but its role in the pathogenesis of UC via regulating M1 macrophage polarization has not been studied yet. For the clinical component, we analyzed IDO1 expression in UC using bioinformatics analysis of Gene Expression Omnibus (GEO) datasets and validated the result using western blotting of colonic tissues from new recruited UC patients. Colitis was induced in mice via dextran sulfate sodium (DSS) treatment and subsequently treated with oral administration of 1-methyl-DL-tryptophan (1-MT), an inhibitor of IDO1 pathway. The results indicated that IDO1 expression was significantly elevated in UC patients and correlated with M1 macrophage polarization observed in both human data and colitis mice. Furthermore, 1-MT markedly ameliorated DSS-induced weight loss, colonic shortening and disease severity via inhibiting IDO1 expression level, downregulating GRP78-XBP1 pathway and reducing M1 proportion. Notably, in vitro study revealed that overexpressing IDO1 in RAW264.7 cells induced macrophage M1 polarization with increased expression levels of GRP78 and XBP1, which was attenuated by 1-MT treatment. Additionally, the catalytic effect exerted by IDO1 overexpression on M1 polarization was neutralized by employing an inhibitor targeting the endoplasmic reticulum (ER) stress pathway. Thus, our findings suggest that IDO1 may promote UC progression by skewing macrophages towards M1 polarization through ER stress-associated GRP78-XBP1 pathway.
Collapse
Affiliation(s)
- Zijian Gao
- Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Shuai Shao
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Zhen Xu
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Jiao Nie
- Department of Gastroenterology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Chenglin Li
- Department of Oncology, Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
| | - Chao Du
- Linyi People’s Hospital, Shandong Second Medical University, Linyi, China
- Department of Gastroenterology, Weihai Municipal Hospital, Shandong University, Weihai, China
| |
Collapse
|
27
|
Yang J, des Rieux A, Malfanti A. Stimuli-Responsive Nanomedicines for the Treatment of Non-cancer Related Inflammatory Diseases. ACS NANO 2025; 19:15189-15219. [PMID: 40249331 PMCID: PMC12045021 DOI: 10.1021/acsnano.5c00700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Nanomedicines offer a means to overcome the limitations associated with traditional drug dosage formulations by affording drug protection, enhanced drug bioavailability, and targeted drug delivery to affected sites. Inflamed tissues possess unique microenvironmental characteristics (including excessive reactive oxygen species, low pH levels, and hypoxia) that stimuli-responsive nanoparticles can employ as triggers to support on-demand delivery, enhanced accumulation, controlled release, and activation of anti-inflammatory drugs. Stimuli-responsive nanomedicines respond to physicochemical and pathological factors associated with diseased tissues to improve the specificity of drug delivery, overcome multidrug resistance, ensure accurate diagnosis and precision therapy, and control drug release to improve efficacy and safety. Current stimuli-responsive nanoparticles react to intracellular/microenvironmental stimuli such as pH, redox, hypoxia, or specific enzymes and exogenous stimuli such as temperature, magnetic fields, light, and ultrasound via bioresponsive moieties. This review summarizes the general strategies employed to produce stimuli-responsive nanoparticles tailored for inflammatory diseases and all recent advances, reports their applications in drug delivery, and illustrates the progress made toward clinical translation.
Collapse
Affiliation(s)
- Jingjing Yang
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Anne des Rieux
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
- Department
of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
28
|
Zhu D, Ma X, Wang J, Chen T, Yang J, Liu Y, Lin Z, Wu M, Hu TY, Zhang Y. A Sequential Release Micro-nano System for Colitis Therapy via Gut Microbiota and Immune Regulation. Angew Chem Int Ed Engl 2025; 64:e202424409. [PMID: 39980315 DOI: 10.1002/anie.202424409] [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: 12/31/2024] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 02/22/2025]
Abstract
Commencing with the breakdown of the intestinal barrier, various pathogenic factors, such as dysbiosis of the gut microbiota, harmful inflammatory cytokines, and immune system imbalance, collectively contribute to the development of colitis. Numerous interventions focusing on single factors have been developed to provide short-term therapeutic benefits, but the continued existence of unresolved pathogenic factors can lead to disease exacerbation. Here we have designed a multicomponent system-inulin microspheres encapsulating selenium-containing nanomicelles, aiming to tackle the multiple factors associated with colitis. This micro-nano drug delivery platform achieves sequential release of drugs in the inflamed colon, with each component of the system functioning independently and jointly. The outer layer of inulin supplies nutrients for probiotics. The inner core comprises selenocystamine and 3-oxolithocholic acid, which polarize macrophages towards an anti-inflammatory phenotype and regulate adaptive immunity by inhibiting TH17-cell differentiation, respectively. In an acute colitis mouse model, this therapeutic system ameliorates colonic inflammation, enhances the abundance of gut microbiota, and modulates the mucosal immune system, showing potential in preventing colitis.
Collapse
Affiliation(s)
- Dongdong Zhu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xiaocao Ma
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Jingguo Wang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Tiantian Chen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Jiahui Yang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yan Liu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhun Lin
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Minhao Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Tony Y Hu
- Center of Cellular and Molecular Diagnosis, Tulane University School of Medicine, New Orleans, Louisiana, 70112, United States
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| |
Collapse
|
29
|
Tao M, Wang L, Chen C, Tang M, Wang Y, Zhang J, Zhao X, Feng Q, Chen J, Yan W, Lin R, Fu Y. Developmentally endothelial locus-1 facilitates intestinal inflammation resolution by suppressing the Cmpk2-cGAS-STING pathway and promoting reparatory macrophage transition. J Adv Res 2025:S2090-1232(25)00274-7. [PMID: 40288675 DOI: 10.1016/j.jare.2025.04.030] [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: 10/17/2024] [Revised: 03/25/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
INTRODUCTION Abnormalities in inflammation resolution function are intimately linked to chronic inflammation, and proresolution therapies may offer novel opportunities for IBD treatment. Developmental endothelial locus 1 (DEL-1), a natural modulator of tissue immunity and inflammation resolution, has not been studied in IBD. OBJECTIVES We aimed to investigate the expression and functions of DEL-1 in IBD. METHODS Assessment of DEL-1 expression in patients, murine models, and cellular levels. To explore the effects of DEL-1 in the acute and recovery phases of inflammation, overexpression plasmids, adeno-associated viruses for DEL-1 knockdown, and DEL-1-Fc fusion proteins were administered to cells and mice. Additionally, the potential mechanism of DEL-1 in IBD was demonstrated using flow cytometry, RNA-Seq, ChIP, dual-luciferase reporter assays and 16S rRNA. RESULTS DEL-1 levels were significantly reduced in IBD patients, colitis mice and macrophages, while the levels increased with inflammation to resolve. Transfection with DEL-1 overexpression plasmid or DEL-1-Fc intervention reduces levels of inflammatory cytokines in both phases and upregulates reparative gene levels in the recovery phase. DEL-1 knockdown inhibits inflammation resolution of colitis. Mechanistically, we demonstrated that DEL-1 inhibits Cmpk2-dependent mtDNA synthesis, thereby inhibiting the cGAS-STING pathway to ameliorate intestinal inflammation. Moreover, DEL-1 promotes reparative macrophage transition in the repair model of colitis. Spi1 was identified as a transcription factor that regulates Cmpk2 and the reparative gene Il10. Intervention with overexpression plasmid of Spi1 or Cmpk2 or the STING agonist DMXAA reverses the effects of DEL-1. In parallel, DEL-1 also inhibits neutrophil recruitment, repairs the intestinal barrier, and improves intestinal microbiota dysbiosis. CONCLUSION We report the first demonstration that DEL-1 significantly ameliorates colonic inflammation in colitis mice. Our findings elucidate a novel mechanism wherein DEL-1 exerts its protective effects by suppressing the Cmpk2-cGAS-STING pathway and promoting reparative macrophage transition. These results collectively position DEL-1 as a promising therapeutic avenue for IBD.
Collapse
Affiliation(s)
- Meihui Tao
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Wang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoyue Chen
- Department of Gastroenterology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mengfan Tang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanping Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyue Zhang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhao
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinyu Feng
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junfa Chen
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yan
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yu Fu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
30
|
Cicia D, Biscu F, Iannotti FA, Miraglia M, Ferrante C, Iaccarino N, Cadenas de Miguel S, Chiavaroli A, Schiano Moriello A, De Cicco P, Nanì MF, Zanoletti L, Ke BJ, van Baarle L, Talavera K, Randazzo A, Elia I, Capasso R, Matteoli G, Pagano E, Izzo AA. Dietary targeting of TRPM8 rewires macrophage immunometabolism reducing colitis severity. Cell Death Dis 2025; 16:343. [PMID: 40280909 PMCID: PMC12032354 DOI: 10.1038/s41419-025-07553-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 04/29/2025]
Abstract
The interplay between diet, host genetics, microbiota, and immune system has a key role in the pathogenesis of inflammatory bowel disease (IBD). Although the causal pathophysiological mechanisms remain unknown, numerous dietary nutrients have been shown to regulate gut mucosal immune function, being effective in influencing innate or adaptive immunity. Here, we proved that transient receptor potential melastatin 8 (TRPM8), a non-selective cation channel, mediates LPS- evoked Ca2+ influx in macrophages leading to their activation. Additionally, we showed that TRPM8 is selectively blocked by the dietary flavonoid luteolin, which induced a pro-tolerogenic phenotype in pro-inflammatory macrophages. Accordingly, genetic deletion of Trpm8 in macrophages caused a deficit in the activation of pro-inflammatory metabolic and transcriptional reprogramming, leading to reduced production of key pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. The TRPM8 anti-inflammatory effect was found to be dependent on lactate which in turn induces IL-10 gene expression. Adoptive transfer of TRPM8-deficient bone marrow in wild-type mice improved intestinal inflammation in a model of colitis. Accordingly, oral administration of luteolin protected mice against colitis through an impairment in the innate immune response. Our study reveals the potential of targeting TRPM8 through specific nutrient interventions to regulate immune function in sub-clinical scenarios or to treat inflammatory diseases, primarily driven by chronic immune responses, such as IBD.
Collapse
Affiliation(s)
- D Cicia
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
- Laboratory of Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - F Biscu
- Laboratory of Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - F A Iannotti
- Institute of Biomolecular Chemistry ICB, CNR, Pozzuoli, Naples, Italy
| | - M Miraglia
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - C Ferrante
- Department of Pharmacy, Gabriele d'Annunzio University, Chieti, Italy
| | - N Iaccarino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - S Cadenas de Miguel
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - A Chiavaroli
- Department of Pharmacy, Gabriele d'Annunzio University, Chieti, Italy
| | | | - P De Cicco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - M F Nanì
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - L Zanoletti
- Laboratory of Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - B-J Ke
- Laboratory of Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - L van Baarle
- Laboratory of Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - K Talavera
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven and VIB Center for Brain and Disease Research, Leuven, Belgium
| | - A Randazzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - I Elia
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute, Leuven, Belgium
| | - R Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
| | - G Matteoli
- Laboratory of Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium.
| | - E Pagano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| | - A A Izzo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
31
|
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
|
32
|
Ye R, Guo J, Yang Z, Wang Z, Chen Y, Huang J, Dong Y. Somatostatin and Mannooligosaccharide Modified Selenium Nanoparticles with Dual-Targeting for Ulcerative Colitis Treatment. ACS NANO 2025; 19:14914-14930. [PMID: 40214514 DOI: 10.1021/acsnano.5c00355] [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: 04/23/2025]
Abstract
Inflammatory bowel disease (IBD) is a prevalent condition worldwide, characterized by complex etiologies, limited efficacy of clinical drug treatments, and potential adverse effects. In this study, we designed 269 nm selenium nanoparticles with double-cell targeting for ulcerative colitis treatment. Somatostatin (SST) and mannooligosaccharide (MOS) were employed to functionalize an Eucommia ulmoides polysaccharide selenium nanoparticle (EUP-SeNP), resulting in the formulation of SST/MOS@EUP-SeNP. Nanoparticles were engineered to target intestinal epithelial cells and macrophages through specific cell surface receptors, enabling dual-targeted treatment. In addition, sodium alginate (SA) microspheres incorporating SST/MOS@EUP-SeNP were prepared for oral administration, protecting the nanoparticles from gastric fluid. The results showed that SA/SST/MOS@EUP-SeNP could preferentially target the inflamed colon tissue and adhere to the colon, enhance the intestinal barrier function, regulate the level of colon inflammation, enhance antioxidant capacity, and regulate the composition of intestinal microbes to effectively relieve the colitis induced by sodium glucan sulfate (DSS). Meanwhile, SA/SST/MOS@EUP-SeNP had excellent biocompatibility both in vivo and in vitro. To some extent, this study can provide a reference for the treatment of IBD.
Collapse
Affiliation(s)
- Ruihua Ye
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jianying Guo
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Zhongjin Yang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100070, China
| | - Zixu Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yaoxing Chen
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jiaqiang Huang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, Ministry of Education, China Agricultural University, Beijing 100193, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yulan Dong
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
33
|
Vámos E, Vántus VB, Deák P, Kálmán N, Sturm EM, Nayak BB, Makszin L, Loránd T, Gallyas FJ, Radnai B. MIF tautomerase inhibitor TE-11 prevents inflammatory macrophage activation and glycolytic reprogramming while reducing leukocyte migration and improving Crohn's disease-like colitis in male mice. Front Immunol 2025; 16:1558079. [PMID: 40330457 PMCID: PMC12053165 DOI: 10.3389/fimmu.2025.1558079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/28/2025] [Indexed: 05/08/2025] Open
Abstract
Background & aims Crohn's disease (CD) is a chronic inflammatory disorder primarily affecting the gastrointestinal tract. Leukocyte recruitment, M1 macrophage polarization and associated metabolic reprogramming are hallmarks of its pathomechanism. Here, we tested TE-11, a potent MIF tautomerase inhibitor (IC50 = 5.63 μmol/dm3) in experimental Crohn's disease in male mice, in leukocyte recruitment and in inflammatory M1 macrophage activation. Methods 2,4,6-trinitrobenzenesulfonic acid-(TNBS)-induced colitis was utilized as a CD-model in male mice. We performed macroscopic scoring and cytokine measurements. We also analyzed MIF-induced leukocyte migration and evaluated apoptosis. LPS+IFN-γ-induced RAW264.7 cells were applied as a M1 macrophage model. We performed qPCR, ROS and nitrite determinations, ELISA measurements, mitochondrial oxygen consumption rate and extracellular acidification rate determinations. Results TE-11 improved mucosal damage, reduced inflammation score and concentration of IL-1β and IL-6 in the colon. It inhibited MIF-induced human blood eosinophil and neutrophil migration and counteracted the anti-apoptotic effect of MIF. In macrophages, MIF inhibition prevented M1 polarization by downregulating HIF-1α gene expression in LPS+IFN-γ-activated cells. Additionally, the molecule reduced mRNA transcription and protein expression of chemokine CCL-2 and cytokine IL-6 while further increasing SOD2 gene transcription and decreased ROS and nitrite production in macrophages. During inflammatory metabolic reprogramming, TE-11 prevented LPS+IFN-γ-induced metabolic shift from OXPHOS to glycolysis. Similarly to anti-inflammatory M2 cells, TE-11 improved mitochondrial energy production by increasing basal respiration, ATP production, coupling efficiency, maximal respiration and spare respiratory capacity. Conclusion Comprehensively, TE-11, a MIF tautomerase inhibitor ameliorates CD-like colitis, reduces MIF-induced eosinophil and neutrophil migration and prevents M1 polarization and associated metabolic reprogramming; therefore, it may prove beneficial as a potential drug candidate regarding CD therapy.
Collapse
Affiliation(s)
- Eszter Vámos
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary
| | - Viola Bagóné Vántus
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Deák
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary
| | - Nikoletta Kálmán
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary
| | - Eva Maria Sturm
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Barsha Baisakhi Nayak
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Lilla Makszin
- Institute of Bioanalysis, Medical School, Szentágothai Research Center, University of Pécs, Pécs, Hungary
| | - Tamás Loránd
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary
| | - Ferenc Jr Gallyas
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary
| | - Balázs Radnai
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
34
|
Cui C, Tang J, Chen J, Zhang B, Li R, Zhang Q, Qiu C, Chen R, Min G, Sun Z, Weng H. Lactobacillus acidophilus extracellular vesicles-coated UiO-66-NH 2@siRNA nanoparticles for ulcerative colitis targeted gene therapy and gut microbiota modulation. J Nanobiotechnology 2025; 23:301. [PMID: 40247297 PMCID: PMC12007195 DOI: 10.1186/s12951-025-03376-0] [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: 12/26/2024] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
Ulcerative colitis (UC) is a complex and chronic inflammatory bowel disease whose pathogenesis involves genetic and environmental factors, which poses a challenge for treatment. Here, we have designed an innovative integrated therapeutic strategy using Lactobacillus acidophilus extracellular vesicles (EVs) to encapsulate UiO-66-NH2 nanoparticles bounded with TNF-α siRNA (EVs@UiO-66-NH2@siRNA) for UC treatment. This system shows superior affinity to inflammation-related cells due to the Lactobacillus acidophilus EVs can maintain immune homeostasis by regulating the secretion of cytokines in vitro. siRNA can specifically target the key inflammatory TNF-α in UC and silence its gene expression, thereby regulating the process of inflammatory response. After oral administration, EVs@UiO-66-NH2@siRNA demonstrates an accurate delivery of TNF-α siRNA to colonize the colon site and exerts a siRNA therapeutic effect by inhibiting the expression of TNF-α, which alleviates the intestinal inflammation in DSS-induced UC model. Moreover, this system can modulate the types and compositional structures of gut microbiota and metabolites to achieve an anti-inflammatory phenotype, which is helpful for the repair of intestinal homeostasis. We also have proved that UiO-66-NH2 nanoparticles exhibit a high loading capacity for TNF-α siRNA and good pH responsiveness, improving the potent release of siRNA in colon tissue. Collectively, the EVs@UiO-66-NH2@siRNA nano-delivery system demonstrate a feasible combination therapeutic strategy for UC through gut microecology modulation, immune regulation and TNF-α siRNA silence, which may provide a potential targeted treatment approach for inflammatory bowel disease.
Collapse
Affiliation(s)
- Chenyang Cui
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Jiaze Tang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Jie Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Beining Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Ruonan Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Qiang Zhang
- Department of Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Chunjing Qiu
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Rongchen Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Geng Min
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zhaowei Sun
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Haibo Weng
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
35
|
Yang H, Zhang X, Wu J, Xiao Y, Dai L, Wang G, Zhang X, Hu C, He S, Yuan Z. Probiotic Membrane-Modified Nanocomposite Alleviates Inflammation and Microbiota Dysbiosis in Colitis by Scavenging Oxidative Stress and Restoring Immune Homeostasis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22245-22265. [PMID: 40184333 DOI: 10.1021/acsami.4c22004] [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: 04/06/2025]
Abstract
Inflammatory bowel disease (IBD) is a complex chronic intestinal disorder in which excessive oxidative stress, dysregulated immune response, and microbiota dysbiosis contribute to recurrent episodes of inflammation in the colonic mucosa. Current clinical treatments focusing solely on inflammation resolution often exhibit limited efficacy due to the inability to fundamentally improve the pathological microenvironment. Herein, a probiotic membrane-modified drug delivery nanocomposite, namely, MPDA@Cur@EM, is developed for the comprehensive treatment of IBD. It contains two components: the curcumin-loaded mesoporous polydopamine nanoparticle (MPDA@Cur) as the core and the Escherichia coli Nissle 1917 outer membrane (EM) as the surface. For MPDA@Cur, the pathological microenvironment triggers the responsive release of curcumin. Importantly, MPDA@Cur can effectively alleviate the inflammatory response of LPS-activated macrophages through MPDA-mediated ROS scavenging and curcumin-induced M2 polarization. In the dextran sulfate sodium (DSS)-induced colitis model, the EM coating not only allows for the targeting enrichment of orally administered MPDA@Cur@EM to the inflamed colonic mucosa, but also participates in the regulation of intestinal flora. Consequently, MPDA@Cur@EM efficiently attenuates the inflammatory reaction and restores the intestinal barrier functions, demonstrated by the multipronged manner of restoring redox balance, remodeling immune homeostasis, and reshaping the gut microecology. Collectively, this work provides a safe and promising codelivery strategy of probiotic product, antioxidative nanoenzyme, and therapeutic drug for comprehensive management of IBD.
Collapse
Affiliation(s)
- Huan Yang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Xu Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Jianshuang Wu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Yao Xiao
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Liangliang Dai
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Gaoyang Wang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Xiaohong Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Chenghu Hu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Shuixiang He
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Zhang Yuan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| |
Collapse
|
36
|
Creoli M, Di Paola A, Tarallo A, Aziz S, Miele E, Martinelli M, Casertano M, Colucci A, Cenni S, Marrapodi MM, Staiano A, Rossi F, Strisciuglio C. Effects of CB2 Receptor Modulation on Macrophage Polarization in Pediatric Inflammatory Bowel Disease. Int J Mol Sci 2025; 26:3720. [PMID: 40332343 PMCID: PMC12027514 DOI: 10.3390/ijms26083720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Macrophages play a crucial role in maintaining intestinal homeostasis and can exhibit either pro-inflammatory M1 or anti-inflammatory M2 phenotypes. The cannabinoid receptor type 2 (CB2) is involved in immune regulation and may represent a therapeutic target in inflammatory bowel disease (IBD). Our study investigates the phenotype of circulating macrophages and CB2 expression in children with IBD, assessing the role of CB2 stimulation in macrophage polarization, iron metabolism, and intestinal barrier function. Macrophages were isolated from 17 children with ulcerative colitis (UC), 21 with Crohn's disease (CD), and 12 healthy controls (CTR). Cells were treated with a CB2 agonist (JWH-133) and an inverse agonist (AM630). CB2 expression and macrophage polarization were assessed by Western blot. Iron metabolism was evaluated through IL-6, hepcidin levels, FPN-1 expression, and iron concentration. Inflammation was assessed by cytokine release. An in vitro "immunocompetent gut" model was used to study the effects of CB2 stimulation on macrophage polarization and intestinal barrier function. CB2 expression was reduced in IBD macrophages. Compared to controls, IBD patients showed increased M1 markers and pro-inflammatory cytokines, with a reduction in M2 markers and IL-13. Altered iron metabolism was observed, with increased [Fe3+], hepcidin release, and DMT1 expression, and reduced FPN-1. CB2 stimulation restored iron metabolism, induced M2 polarization, and improved intestinal barrier function. CB2 could represent a novel therapeutic target for IBD by modulating macrophage function, iron metabolism, and mucosal barrier restoration.
Collapse
Affiliation(s)
- Mara Creoli
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 2, 80138 Naples, Italy; (M.C.); (A.D.P.); (A.C.); (M.M.M.); (C.S.)
| | - Alessandra Di Paola
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 2, 80138 Naples, Italy; (M.C.); (A.D.P.); (A.C.); (M.M.M.); (C.S.)
- Department of Life Sciences, Health and Health Professions, Link Campus University, Via del Casale di San Pio V, 00165 Rome, Italy
| | - Antonietta Tarallo
- Department of Translational Medical Science, Section of Pediatrics, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (A.T.); (E.M.); (M.M.); (M.C.); (A.S.)
| | - Sohail Aziz
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Santa Maria di Costantinopoli 16, 80138 Naples, Italy; (S.A.); (S.C.)
| | - Erasmo Miele
- Department of Translational Medical Science, Section of Pediatrics, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (A.T.); (E.M.); (M.M.); (M.C.); (A.S.)
| | - Massimo Martinelli
- Department of Translational Medical Science, Section of Pediatrics, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (A.T.); (E.M.); (M.M.); (M.C.); (A.S.)
| | - Marianna Casertano
- Department of Translational Medical Science, Section of Pediatrics, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (A.T.); (E.M.); (M.M.); (M.C.); (A.S.)
| | - Antonio Colucci
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 2, 80138 Naples, Italy; (M.C.); (A.D.P.); (A.C.); (M.M.M.); (C.S.)
| | - Sabrina Cenni
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Santa Maria di Costantinopoli 16, 80138 Naples, Italy; (S.A.); (S.C.)
| | - Maria Maddalena Marrapodi
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 2, 80138 Naples, Italy; (M.C.); (A.D.P.); (A.C.); (M.M.M.); (C.S.)
| | - Annamaria Staiano
- Department of Translational Medical Science, Section of Pediatrics, University of Naples “Federico II”, Via S. Pansini 5, 80131 Naples, Italy; (A.T.); (E.M.); (M.M.); (M.C.); (A.S.)
| | - Francesca Rossi
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 2, 80138 Naples, Italy; (M.C.); (A.D.P.); (A.C.); (M.M.M.); (C.S.)
| | - Caterina Strisciuglio
- Department of Woman, Child and General and Specialist Surgery, University of Campania “Luigi Vanvitelli”, Via Luigi de Crecchio 2, 80138 Naples, Italy; (M.C.); (A.D.P.); (A.C.); (M.M.M.); (C.S.)
| |
Collapse
|
37
|
Jeong MR, Hwang JW, Choi M, Seok SH. MHC class II + macrophage differentiation is impaired in metastasized lungs via PGE 2 receptor EP2. Cell Rep 2025; 44:115574. [PMID: 40232933 DOI: 10.1016/j.celrep.2025.115574] [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: 10/08/2024] [Revised: 01/17/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Monocytes differentiate into macrophages (Mφs) to facilitate lung metastasis, but the monocyte-to-Mφ transition during this process is not well understood. To investigate, we performed bulk RNA sequencing on Mφs isolated from the lungs of mice bearing Lewis lung carcinoma tumors and from naive lungs. Our results showed impaired differentiation of monocytes into major histocompatibility complex (MHC) class II+ Mφs, with an upregulation of PGE2-inducible genes, including Arg1, in tumor-associated Mφs (TAMs). In vitro experiments confirmed that prostaglandin E2 (PGE2) inhibits the differentiation of MHC class II+ Mφs while promoting Arg1+ Mφs via the E prostanoid 2 (EP2) receptor, accompanied by DNA methylation. Whole-genome bisulfite sequencing revealed that PGE2-EP2 signaling drives the hypermethylation and downregulation of gene sets related to myeloid cells in non-neoplastic tissues. Our study highlights PGE2-EP2-driven DNA methylation in the monocyte-to-TAM transition, suggesting potential therapeutic avenues for lung metastasis.
Collapse
Affiliation(s)
- Mi Reu Jeong
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jung Woo Hwang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Seung Hyeok Seok
- Macrophage Lab, Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Cancer Research Institute, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
38
|
Kim YI, Ko I, Yi EJ, Kim J, Hong YR, Lee W, Chang SY. NAD + modulation of intestinal macrophages renders anti-inflammatory functionality and ameliorates gut inflammation. Biomed Pharmacother 2025; 185:117938. [PMID: 40022994 DOI: 10.1016/j.biopha.2025.117938] [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: 01/16/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Macrophages can maintain gut immune homeostasis by driving clearance of infection, but also can prevent chronic inflammation and induce tissue repair. Reduced nicotinamide adenine dinucleotide (NAD+) levels in macrophages have been reported to be associated with the onset of severe colitis. Given that dysregulation of gut macrophages plays a significant role in inflammatory bowel disease (IBD), they represent a potential target for novel therapies. Here we show an IBD therapeutic candidate LMT503, a substrate that modulates NADH quinone oxidoreductase (NQO1), which induces anti-inflammatory macrophage polarization by NAD+ enhancement. To determine the anti-inflammatory effect of LMT503, a dextran sulfate sodium (DSS)-induced colitis mouse model was used in this study. Treatment of bone marrow-derived macrophages (BMDMs) with LMT503 increased IL-10 and Arg1 levels but decreased levels of TNF-α, iNOS, and IL-6. LMT503 also increased levels of SIRT1, SIRT3, and SIRT6, suggesting that macrophages were driven to an anti-inflammatory character. In a murine DSS-induced colitis model, oral treatment with LMT503 ameliorated colonic inflammation and decreased infiltrating monocytes and neutrophils. Although NAD+ enhancement did not alter CX3CR1intCD206- or CX3CR1hiCD206+ colon macrophage population, it decreased levels of TNF-α and iNOS and increased IL-10 level, with colonic macrophages showing an anti-inflammatory character shift. Depletion of CX3CR1 expressing gut resident macrophages abrogated the immune regulatory effect of LMT503 in the colon. These data suggest that LMT503 is a therapeutic candidate that can target macrophages to drive polarization with an immunosuppressive character and ameliorate IBD.
Collapse
Affiliation(s)
- Young-In Kim
- Laboratory of Microbiology, Department of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon 16499, Republic of Korea; Korea Initiative for fostering University of Research and Innovation (KIURI) Program, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Inseok Ko
- Lmito Therapeutics, Yongin-si 16827, Republic of Korea; Department of Chemistry Education, Graduate Department of Chemical Materials, Pusan National University, Busan, Republic of Korea
| | - Eun-Je Yi
- Laboratory of Microbiology, Department of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon 16499, Republic of Korea
| | - Jusik Kim
- Lmito Therapeutics, Yongin-si 16827, Republic of Korea
| | - Yong Rae Hong
- Lmito Therapeutics, Yongin-si 16827, Republic of Korea
| | - Wheeseong Lee
- Lmito Therapeutics, Yongin-si 16827, Republic of Korea
| | - Sun-Young Chang
- Laboratory of Microbiology, Department of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon 16499, Republic of Korea.
| |
Collapse
|
39
|
Mandal M, Mamun MAA, Rakib A, Kumar S, Park F, Hwang DJ, Li W, Miller DD, Singh UP. Modulation of occludin, NF-κB, p-STAT3, and Th17 response by DJ-X-025 decreases inflammation and ameliorates experimental colitis. Biomed Pharmacother 2025; 185:117939. [PMID: 40036995 DOI: 10.1016/j.biopha.2025.117939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/01/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
SCOPE Inflammatory bowel disease (IBD) involves a range of immune-mediated disorders marked by systemic and local intestinal inflammation. We synthesized a novel compound DJ-X-025 and uncovered its anti-inflammatory properties using lipopolysaccharide (LPS)-induced RAW 264.7 macrophages in vitro and a dextran sodium sulfate (DSS)-induced model of colitis. METHODS AND RESULTS We evaluated the alteration in cell morphology, cytoskeletal proteins, and inflammatory markers of DJ-X-025 treated LPS-stimulated RAW 264.7 macrophages. We administered DJ-X-025 by oral gavage in DSS-induced colitis, examined colon histology, and alterations of immune cells by flow cytometry, and performed molecular studies using RT-qPCR and western blot analysis. DJ-X-025 treatment markedly altered the morphology of LPS-treated RAW 264.7 macrophages from elongated to round shapes, modulated actin and tubulin, and reduced the level of inflammatory markers like TNF-α, IL-1β, IL-6, and iNOS. Further, we observed that DJ-X-025 steered to improve colon length, muscularis mucosa thickness, and colon inflammatory score compared to the DSS group alone. DJ-X-025 effectively inverted the increased population of activated T cells, Th17, and macrophages in lamina propria by DSS treatment, leading to a substantial reduction in the inflammatory response in the colon. Strikingly, DJ-X-025 treatment enhanced the expression of occludin and diminished the expression of NF-κB and phosphorylation of STAT3 in the colon of DSS-treated mice compared to DSS-alone. Additionally, DJ-X-025 induced the expression of Foxp3 in the colon and, reduced systemic inflammatory cytokine/chemokine levels further supporting its immunomodulatory effects. These results suggest that DJ-X-025 is linked to the induction of occludin expression and decreased expression of p-STAT3/NF-κB and Th17 response in the colon, which together suppresses systemic and colon inflammatory cytokines for effective amelioration of experimental colitis. CONCLUSION These findings suggest that DJ-X-025 might be a promising therapeutic agent for the treatment of IBD.
Collapse
Affiliation(s)
- Mousumi Mandal
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Md Abdullah Al Mamun
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Ahmed Rakib
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Udai P Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
40
|
Liu Y, Zhang JT, Sun M, Song J, Sun HM, Wang MY, Wang CM, Liu W. Targeting ferroptosis in the treatment of ulcerative colitis by traditional Chinese medicine: A novel therapeutic strategies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156539. [PMID: 39987602 DOI: 10.1016/j.phymed.2025.156539] [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: 11/09/2024] [Revised: 02/04/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND The incidence of ulcerative colitis (UC) has been rising rapidly in recent years, and there is currently no effective method to prevent its recurrence. Owing to its long treatment duration, difficulty in treatment, prolonged remission, and high costs, it has attracted global attention. Exploring safe, effective, and sustainable treatment regimens has become an urgent global issue. Traditional Chinese medicine (TCM) has unique advantages such as low cost, low drug resistance, and fewer side effects, and has accumulated rich experience in the treatment of UC. PURPOSE Ferroptosis, as a new form of non-apoptotic cell death, is characterized by iron homeostatic imbalance and lipid peroxidation in the redox system. Studies have shown that inhibited ferroptosis in intestinal epithelial cells can protect the intestinal mucosa. Targeted intervention in ferroptosis may be a new direction for the treatment of UC. METHODS We conducted a systematic literature search with Google Scholar, PubMed, Web of Science, ScienceDirect and X-mol databases have been utilized to retrieve relevant literature up to October 2024, using keywords included ferroptosis, Inflammatory bowel disease (IBD), UC, Crohn's disease and TCM, Chinese traditional prescription, Chinese medicine extract and active ingredients. The existing literature was comprehensively studied and sorted out. RESULTS Currently, UC is mainly treated with drugs, including corticosteroids, amino salicylates, biologics, and immunomodulators, but drug resistance and adverse reactions are common. Increasing evidence suggests that TCM may treat UC by interfering with ferroptosis. Scholars have confirmed that TCM can inhibit ferroptosis, and recent studies have shown that TCM can not only inhibit iron dependent lipid peroxidation in intestinal cells but also enhance the antioxidant and anti-inflammatory abilities of intestinal mucosa, thus playing a role in the treatment of UC. This review explores the relevance of TCM intervention in ferroptosis and the treatment of UC, discusses the possible mechanisms of ferroptosis in UC, and aims to provide a basis for the diagnosis and treatment of UC. CONCLUSION It is revealed that TCM targeted ferroptosis has a good application prospect in the treatment of UC, providing a theoretical basis for elucidating the pathogenesis of UC and the study of TCM targeting ferroptosis regulating lipid metabolism in the treatment of UC, and providing a new perspective for the treatment of IBD in the future.
Collapse
Affiliation(s)
- Ying Liu
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Jing-Tian Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Meng Sun
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Jian Song
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Hai-Ming Sun
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Meng-Yang Wang
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Chun-Mei Wang
- College of Pharmacy, Beihua University, jilin 132013, China
| | - Wei Liu
- College of Pharmacy, Beihua University, jilin 132013, China.
| |
Collapse
|
41
|
Liu J, Niu D, Tang Y, Zheng R, Qin Y, Cheng X, Pan S, Yuan J, Shi X, Yang J. Beta-hydroxy-beta-methylbutyrate (HMB) ameliorates DSS-induced colitis by inhibiting ERK/NF-κB activation in macrophages. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156492. [PMID: 39978274 DOI: 10.1016/j.phymed.2025.156492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 01/20/2025] [Accepted: 02/09/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND β-Hydroxy β-Methylbutyrate (HMB), derived from leucine, is known for its role in anti-oxidation and anti-inflammation. But, the application of HMB in IBD treatment is not fully understood, highlighting the requirement for further research. PURPOSE We aimed to examine the effects of HMB treatment on DSS-induced chronic colitis in mice and explore its underlying mechanisms. METHODS To simulate colonic inflammation, a murine colitis model was generated by using DSS induction. Critical indicators such as body weight, colon length, disease activity index (DAI), and gross pathology were thoroughly monitored. Immunohistochemistry assay was conducted to assess the expression of Occludin and F4/80. Flow cytometry was employed to evaluate the expression levels of CD80 and CD86. qPCR was performed to measure cytokine expression (IL-6, IL-1β, TNF-α, IL-22, CXCL2, iNOS). RNA sequencing was carried out using bone-marrow derived dendritic macrophage cells (BMDMs). RESULTS Our study indicates that HMB treatment substantially mitigated colonic damage in murine models of DSS-induced colitis, highlighting its anti-inflammatory potential. Notably, HMB significantly enhanced the expression of Occludin in these mice. Furthermore, HMB downregulated proinflammatory markers such as IL-6, IL-1β, and TNF-α as well as CXCL2 in the colon tissue. In vitro experiments also revealed that HMB reduced production of proinflammatory cytokines induced by DSS and suppressed the expression levels of CD80 and CD86 in macrophage cells. On a mechanistic level, we demonstrated the anti-inflammatory effects of HMB by reducing the phosphorylation of p-ERK and p-p65, thereby limiting cytokine production in both in vivo and in vitro settings. CONCLUSION These findings indicate that HMB possesses anti-inflammation against intestinal inflammation and may hold promise as a potential therapeutic candidate for IBD treatment. There's growing interest in combining traditional anti-inflammatory agents with supplements like HMB to improve outcomes in complex IBD cases. HMB's role in established muscle preservation and reduction of systemic inflammation as described in this study could make it a valuable adjunct in IBD therapy.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Clinical Nutrition, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215000, PR China
| | - Danye Niu
- Department of Clinical Nutrition, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215000, PR China
| | - Yu Tang
- Department of Clinical Nutrition, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215000, PR China
| | - Ruoheng Zheng
- Institute of Clinical Medicine Research, Suzhou Hospital, The Affiliated Hospital of Medical School, Nanjing University, Lijiang Road No. 1, Suzhou 215153, PR China
| | - Yinyin Qin
- Institute of Clinical Medicine Research, Suzhou Hospital, The Affiliated Hospital of Medical School, Nanjing University, Lijiang Road No. 1, Suzhou 215153, PR China
| | - Xiuqin Cheng
- Department of Gastroenterology, Suzhou Hospital, The Affiliated Hospital of Medical School, Nanjing University, Lijiang Road No. 1, Suzhou 215153, PR China
| | - Shubo Pan
- Department of Gastroenterology, Suzhou Hospital, The Affiliated Hospital of Medical School, Nanjing University, Lijiang Road No. 1, Suzhou 215153, PR China
| | - Jinfei Yuan
- Institute of Clinical Medicine Research, Suzhou Hospital, The Affiliated Hospital of Medical School, Nanjing University, Lijiang Road No. 1, Suzhou 215153, PR China
| | - Xiaohua Shi
- Department of Gastroenterology, Suzhou Hospital, The Affiliated Hospital of Medical School, Nanjing University, Lijiang Road No. 1, Suzhou 215153, PR China
| | - Jiao Yang
- Institute of Clinical Medicine Research, Suzhou Hospital, The Affiliated Hospital of Medical School, Nanjing University, Lijiang Road No. 1, Suzhou 215153, PR China; Department of Gastroenterology, Suzhou Hospital, The Affiliated Hospital of Medical School, Nanjing University, Lijiang Road No. 1, Suzhou 215153, PR China.
| |
Collapse
|
42
|
Zhuang J, Hai Y, Lu X, Sun B, Fan R, Zhang B, Wang W, Han B, Luo L, Yang L, Zhang C, Zhao M, Wei G. A Self-Assembled Metabolic Regulator Reprograms Macrophages to Combat Cytokine Storm and Boost Sepsis Immunotherapy. RESEARCH (WASHINGTON, D.C.) 2025; 8:0663. [PMID: 40171016 PMCID: PMC11959697 DOI: 10.34133/research.0663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 04/03/2025]
Abstract
Sepsis, a life-threatening inflammatory disorder characterized by multiorgan failure, arises from a dysregulated immune response to infection. Modulating macrophage polarization has emerged as a promising strategy to control sepsis-associated inflammation. The endogenous metabolite itaconate has shown anti-inflammatory potential by suppressing the stimulator of interferon genes (STING) pathway, but its efficacy is inhibited by hyperactive glycolysis, which sustains macrophage overactivation. Here, we revealed a critical crosstalk between the itaconate-STING axis and glycolysis in macrophage-mediated inflammation. Building on this interplay, we developed a novel nanoparticle LDO (lonidamine disulfide 4-octyl-itaconate), a self-assembled metabolic regulator integrating an itaconate derivative with the glycolysis inhibitor Lonidamine. By concurrently targeting glycolysis and STING pathways, LDO reprograms macrophages to restore balanced polarization. In sepsis models, LDO effectively attenuates CCL2-driven cytokine storms, alleviates acute lung injury, and significantly enhances survival via metabolic reprogramming. This study offers a cytokine-regulatory strategy rooted in immunometabolism, providing a foundation for the translational development of immune metabolite-based sepsis therapies.
Collapse
Affiliation(s)
- Junyan Zhuang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Yongrui Hai
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Xintong Lu
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Borui Sun
- Department of Anesthesiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Renming Fan
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Bingjie Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Wenhui Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Bingxue Han
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| | - Li Luo
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi’an 710038, Shaanxi, China
| | - Le Yang
- Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi’an 710038, Shaanxi, China
| | - Chun Zhang
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Minggao Zhao
- Department of Pharmacy, Tangdu Hospital, Air Force Military Medical University, Xi’an 710038, Shaanxi, China
| | - Gaofei Wei
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
| |
Collapse
|
43
|
Gao C, Yang Z, Song R, Sheng H, Zhu L. Nanotechnology-based drug delivery system for targeted therapy of ulcerative colitis from traditional Chinese medicine: A review. Int J Pharm 2025; 673:125375. [PMID: 39965734 DOI: 10.1016/j.ijpharm.2025.125375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/27/2025] [Accepted: 02/15/2025] [Indexed: 02/20/2025]
Abstract
Ulcerative colitis (UC) is a chronic autoimmune disease and seriously affects the normal life of patients. Conventional therapeutic drugs are difficult to meet clinical needs. Traditional Chinese medicine (TCM) ingredients could effectively alleviate the symptoms of UC by anti-inflammatory, anti-oxidative, regulating the gut microbiota, and repairing the colonic epithelial barrier, but their low solubility and bioavailability severely limit their clinical application. Nano-drug delivery systems (NDDS) combined with TCM ingredients is a promising option for treating UC, and they could significantly enhance the stability, solubility, and bioavailability of TCM ingredients. The review describes the anti-UC mechanisms of TCM ingredients, systematically summarizes various kinds of NDDS for TCM ingredients according to different routes of administration, and highlights the advantages of NDDS for TCM ingredients in the treatmentof UC. In addition, we discuss the limitations of existing NDDS for TCM ingredients and the development direction in the future. This review will provide a basis for the future development of anti-UC NDDS for TCM ingredients.
Collapse
Affiliation(s)
- Chengcheng Gao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zerun Yang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ruirui Song
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
44
|
Pang L, Huang Y, Li R, Guo L, Man C, Yang X, Jiang Y. Effects of postbiotics produced by Lactobacillus plantarum JM015 isolated from traditional fermented dairy products on Salmonella-induced intestinal inflammation: A preventive strategy. Food Chem 2025; 469:142549. [PMID: 39708644 DOI: 10.1016/j.foodchem.2024.142549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/03/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Affiliation(s)
- Lidong Pang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yan Huang
- Sanmenxia Polytechnic, Sanmenxia, 472000, China
| | - Runze Li
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Ling Guo
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Chaoxin Man
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xinyan Yang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Yujun Jiang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
45
|
Fang X, Zhang Y, Ke Z, Zhang Y, Lin Y, Huang Y, Zhou J, Su H, Xu J, Liu Y. The m6A reader HNRNPC is a key regulator in DSS-induced colitis by modulating macrophage phenotype. iScience 2025; 28:111812. [PMID: 40124522 PMCID: PMC11927749 DOI: 10.1016/j.isci.2025.111812] [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: 04/06/2024] [Revised: 07/25/2024] [Accepted: 01/10/2025] [Indexed: 03/25/2025] Open
Abstract
m6A regulators were demonstrated to modulate the functions of intestinal epithelial and immune cells in the ulcerative colitis. This study aimed to elucidate whether and how the m6A reader heterogeneous nuclear ribonucleoprotein C (HNRNPC) regulates macrophage function in the colitis. We observed elevated HNRNPC in the inflammatory Raw264.7 cells and macrophages in the dextran sodium sulfate (DSS)-induced colitis. Knocking down HNRNPC can mitigate LPS-induced activation of macrophages in vitro. Furthermore, adoptive transfer of macrophages with HNRNPC knockdown significantly alleviated colitis compared to those transfected with negative control siRNA. Additionally, RNA sequencing illuminated that HNRNPC regulated functions of macrophages by inhibiting alternative mRNA slicing, involving adjusting acute inflammatory response, and promoting cell chemotaxis and migration. Besides, HNRNPC can govern the stability of Itgb7, and Itgb7 might be an effective target for HNRNPC in macrophages. Our findings highlight the crucial role and therapeutic potential of HNRNPC inhibition in macrophages in alleviating colitis.
Collapse
Affiliation(s)
- Xiaohui Fang
- Department of Gastroenterology, Peking University Peoples Hospital, No.11, Xizhimen South Street, Xicheng District, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Yu Zhang
- Department of Gastroenterology, Peking University Peoples Hospital, No.11, Xizhimen South Street, Xicheng District, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Ziliang Ke
- Department of Gastroenterology, Peking University Peoples Hospital, No.11, Xizhimen South Street, Xicheng District, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Yang Zhang
- Department of Gastroenterology, Peking University Peoples Hospital, No.11, Xizhimen South Street, Xicheng District, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Yiken Lin
- Department of Gastroenterology, Peking University Peoples Hospital, No.11, Xizhimen South Street, Xicheng District, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Yibo Huang
- Department of Gastroenterology, Peking University Peoples Hospital, No.11, Xizhimen South Street, Xicheng District, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Jianhua Zhou
- Institute of Clinical Molecular Biology & Central Laboratory, Peking University People’s Hospital, Beijing 100044, China
| | - Huiting Su
- Institute of Clinical Molecular Biology & Central Laboratory, Peking University People’s Hospital, Beijing 100044, China
| | - Jun Xu
- Department of Gastroenterology, Peking University Peoples Hospital, No.11, Xizhimen South Street, Xicheng District, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| | - Yulan Liu
- Department of Gastroenterology, Peking University Peoples Hospital, No.11, Xizhimen South Street, Xicheng District, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, No. 11, Xizhimen South Street, Xicheng District, Beijing 100044, China
| |
Collapse
|
46
|
Quan T, Li R, Gao T. The Intestinal Macrophage-Intestinal Stem Cell Axis in Inflammatory Bowel Diseases: From Pathogenesis to Therapy. Int J Mol Sci 2025; 26:2855. [PMID: 40243444 PMCID: PMC11988290 DOI: 10.3390/ijms26072855] [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: 02/19/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
The gut plays a crucial role in digestion and immunity, so its balance is essential to overall health. This balance relies on dynamic interactions between intestinal epithelial cells, immune cells, and crypt stem cells. Inflammatory bowel disease (IBD), which consists of ulcerative colitis and Crohn's disease, is a chronic relapsing inflammatory disease of the gastrointestinal tract closely related to immune dysfunction. Stem cells, known for their ability to self-renew and differentiate, play an important role in repairing damaged intestinal epithelium and maintaining homeostasis in vivo. Macrophages are key gatekeepers of intestinal immune homeostasis and have a significant impact on IBD. Current research has focused on the link between epithelial cells and stem cells, but interactions with macrophages, which have been recognized as attractive targets for the development of new therapeutic approaches to disease, have been less explored. Recently, the developing field of immunometabolism has reinforced that metabolic reprogramming is a key determinant of macrophage function and subsequent disease progression. The aim of this review is to explore the role of the macrophage-stem cell axis in the maintenance of intestinal homeostasis and to summarize potential approaches to treating IBD by manipulating the cellular metabolism of macrophages, as well as the main opportunities and challenges faced. In summary, our overview provides a framework for understanding the critical role of macrophage immunometabolism in maintaining gut health and potential therapeutic targets.
Collapse
Affiliation(s)
| | | | - Ting Gao
- College of Veterinary Medicine, China Agricultural University, Beijing 100083, China; (T.Q.); (R.L.)
| |
Collapse
|
47
|
Luo T, Hou L, Cao Y, Li M, Sheng X, Cheng W, Yan L, Zheng L. Tea Extracellular Vesicle-Derived MicroRNAs Contribute to Alleviate Intestinal Inflammation by Reprogramming Macrophages. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6745-6757. [PMID: 40047388 DOI: 10.1021/acs.jafc.5c01990] [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: 03/20/2025]
Abstract
The clinical use of conventional medications for inflammatory bowel disease (IBD) is often limited by significant side effects. The extracellular vesicles derived from plant-based diets have shown promise in mitigating disease. Here, we discovered that natural extracellular vesicles from tea (TEVs) can achieve an appropriate transition from proinflammatory (M1) to anti-inflammatory (M2) macrophages and inhibit inflammation response both in vitro and in vivo. More importantly, the therapeutic effects of TEVs were at least partially attributed to RNA in a DSS-induced colitis model. Small RNA sequencing revealed a distinct enrichment of miRNAs in TEVs, with target genes primarily linked to IBD. TEVs were absorbed by macrophages in a time-dependent manner, carrying miRNAs that modulate gene expression within host cells. Notably, TEV-derived osa-miR166d-5p and gma-miR396a-3p were shown to enhance M2 macrophage polarization and reduce inflammation in vitro. Mechanistically, the osa-miR166d-5p- and gma-miR396a-3p-mediated targeting of the 3'-UTRs of AKT1 and IKBKB decreased NF-κB levels. Overall, we demonstrated that TEVs can ameliorate mouse colitis by reprogramming macrophage polarization and contain a unique miRNA repertoire, including osa-miR166d-5p and gma-miR396a-3p, with a novel function of alleviating intestinal inflammation.
Collapse
Affiliation(s)
- Tianyu Luo
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Linhai Hou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Yaqi Cao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Meiqi Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Xinyue Sheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Wenqi Cheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Ling Yan
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
- Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230009, China
- Anhui Province Key Laboratory of Agricultural Products Modern Processing, Hefei 230009, China
| | - Lei Zheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
- Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230009, China
- Research Laboratory of Agricultural Environment and Food Safety, Anhui Modern Agricultural Industry Technology System, Hefei 230009, China
| |
Collapse
|
48
|
Cao R, Zhou J, Liu J, Wang Y, Dai Y, Jiang Y, Yamauchi A, Atlas D, Jin T, Zhou J, Wang C, Tan Q, Chen Y, Yodoi J, Tian H. TXM-CB13 Improves the Intestinal Mucosal Barrier and Alleviates Colitis by Inhibiting the ROS/TXNIP/TRX/NLRP3 and TLR4/MyD88/NF-κB/NLRP3 Pathways. Inflammation 2025:10.1007/s10753-025-02282-9. [PMID: 40085192 DOI: 10.1007/s10753-025-02282-9] [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: 05/31/2024] [Revised: 02/11/2025] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
The activation of inflammasomes (NLRP3 and NLRP1) is central to the pathogenesis of inflammatory bowel disease (IBD). Here we examined the protective effects of a thioredoxin-mimetic peptide CB13 (TXM-CB13), known for its antioxidative stress and anti-inflammatory properties. We examined the effects of TXM-CB13 on dextran sulfate sodium (DSS)-induced colitis and lipopolysaccharide (LPS)-induced NLRP3 inflammasome activation in RAW264.7 macrophages. TXM-CB13 appeared to alleviate symptoms of DSS-induced colitis and to significantly suppress the protein and mRNA levels of NLRP3, Mlck, and IL-1β in colonic tissues. Additionally, TXM-CB13 treatment increased the levels of the intestinal barrier proteins Occludin, ZO-1, and NLRP1, as shown through immunohistochemistry and Western blot analysis. In vitro, TXM-CB13 inhibited LPS-induced TLR4 signaling, reducing MyD88 levels and consequently attenuating the activation of the NF-κB pathways, including p-IκB-α/IκB-α and p-NF-κB-p65/NF-κB-p65. This inhibition further reduced the activation of the NLRP3 inflammasome components, NLRP3, ASC, Caspase-1, GSDMD, and IL-1β. In addition, TXM-CB13 prevented the ROS-mediated dissociation of TXNIP from TRX, inhibiting NLRP3 activation. These findings suggest that TXM-CB13 is a potential therapeutic candidate for IBD through its modulation of the TLR4/MyD88/NF-κB/NLRP3 and ROS/TXNIP/TRX/NLRP3 pathways.
Collapse
Affiliation(s)
- Ruijie Cao
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Jinhui Zhou
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Jiale Liu
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Yaxuan Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Yandong Dai
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Yun Jiang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Akira Yamauchi
- Department of Breast Surgery, Misugi-kai Sato Hospital Breast Center, HIrakata, Osaka, Japan
| | - Daphne Atlas
- Dept. Of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Tiancheng Jin
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Jiedong Zhou
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Cuixue Wang
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Qihuan Tan
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Yifei Chen
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China
| | - Junji Yodoi
- Laboratory of Infection and Prevention, Department of Biological Response, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Hai Tian
- Department of Basic Medicine, Medical College, Shaoxing University, Shaoxing, China.
- Jiaozhimei Biotechnology (Shaoxing) Co., Ltd., Shaoxing, China.
| |
Collapse
|
49
|
Wang Y, Chu T, Meng C, Bian Y, Li J. Piezo1-specific Deletion in Macrophage Protects the Progression of Chronic Inflammatory Bowel Disease in Mice. Cell Mol Gastroenterol Hepatol 2025; 19:101495. [PMID: 40081571 DOI: 10.1016/j.jcmgh.2025.101495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND & AIMS Piezo1, a recently identified mechanically activated nonselective cation channel protein, demonstrates sensitivity to various mechanical stimuli, such as matrix stiffness and shear stress. Although accumulating evidence implicates Piezo1 channels in numerous physiologic and pathophysiologic processes, its involvement in dextran sulfate sodium (DSS)-induced acute and chronic inflammatory bowel disease (IBD) remains incompletely understood. This study aimed to investigate the effect of Piezo1 channels in macrophage polarization and its associated functions in IBD. METHODS DSS-induced inflammatory bowel disease model was established in Piezo1td/Tdt or Piezo1fl/fl and Piezo1△LysM male mice. Additionally, bone marrow-derived macrophages from Piezo1fl/fl and Piezo1△LysM male mice were isolated to elucidate the downstream targets of Piezo1 and the associated underlying molecular mechanisms. RESULTS Our findings revealed that Piezo1 deficiency in macrophages could protect mice from DSS-induced chronic IBD, as evidenced by improved colon length and the preservation of colon structure. The mitigation of inflammation during chronic IBD progression was observed with Piezo1 deficiency in macrophages, characterized by reduced macrophage accumulation, M1 macrophage polarization, T helper 1 infiltration, and decreased inflammatory cytokine secretion. Further investigations unveiled that Piezo1-deficient macrophages inhibit the expression and activity of Nod-like receptor protein 3 and nuclear factor kappa B in colon tissues and bone marrow-derived macrophages while regulating the nuclear translocation of p65. Conversely, macrophage Piezo1 activation enhanced inflammatory cytokine secretion by activating Nod-like receptor protein 3/nuclear factor kappa B pathways. CONCLUSIONS Myeloid Piezo1 mediates colonic immune response, and disrupting Piezo1 inhibits the progression of chronic IBD. This study provides hitherto undocumented evidence of the pivotal role of macrophage Piezo1 channels in regulating the progression of chronic IBD. Targeting macrophage Piezo1 may offer a promising therapeutic strategy against chronic IBD.
Collapse
Affiliation(s)
- Yuman Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong Province, China; College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Tianjiao Chu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong Province, China
| | - Chengzhen Meng
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong Province, China
| | - Yifei Bian
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong Province, China.
| | - Jing Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Ji'nan, Shandong Province, China.
| |
Collapse
|
50
|
Yang S, Fan L, Yin L, Zhao Y, Li W, Zhao R, Jia X, Dong F, Zheng Z, Zhao D, Wang J. Ginseng exosomes modulate M1/M2 polarisation by activating autophagy and target IKK/IкB/NF-кB to alleviate inflammatory bowel disease. J Nanobiotechnology 2025; 23:198. [PMID: 40065319 PMCID: PMC11895377 DOI: 10.1186/s12951-025-03292-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Exosomes are involved in intercellular communication and regulation of the inflammatory microenvironment. In a previous study, we demonstrated that fresh ginseng exosomes (GEs) alleviated inflammatory bowel disease. However, the precise mechanism by which GEs activate the immune system and subsequently inhibit the formation of intestinal inflammatory microenvironment remains unknown. METHODS Herein, we investigated the effects of GEs on autophagy, macrophage polarisation, intestinal inflammation, and the epithelial barrier by means of transcriptome sequencing, network pharmacology, transmission electron microscopy, immunoblotting, flow cytometry and small molecule inhibitors. RESULTS GEs significantly activated autophagy and M2-like macrophage polarisation, which could be blocked by the autophagy inhibitor 3-methyladenine. In the co-culture system of macrophages and intestinal epithelial cells, macrophages treated with GEs secreted more interleukin-10 (IL-10) and significantly reduced Nitric oxide (NO) levels in intestinal epithelial cells in vitro. Furthermore, GEs acted directly on intestinal epithelial cells through the IKK/IкB/NF-кB signalling pathway to reduce inflammation and restore the intestinal barrier. Orally administered GEs could restore disrupted colonic barriers, alleviate inflammatory bowel responses, and regulate the polarisation of intestinal macrophages in vivo. CONCLUSION In summary, GEs may be a potential treatment for inflammatory bowel disease, and targeting autophagy and macrophage polarisation may help alleviate intestinal inflammation.
Collapse
Affiliation(s)
- Song Yang
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Liangliang Fan
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Lijia Yin
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Yueming Zhao
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Wenjing Li
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Ronghua Zhao
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Xuxia Jia
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Fusong Dong
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Ze Zheng
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Daqing Zhao
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Jiawen Wang
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China.
- Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, 17176, Stockholm, Sweden.
| |
Collapse
|