1
|
Mitra D, Armijo GK, Ober EH, Baker SM, Turner HC, Broustas CG. MIIST305 mitigates gastrointestinal acute radiation syndrome injury and ameliorates radiation-induced gut microbiome dysbiosis. Gut Microbes 2025; 17:2458189. [PMID: 39930324 PMCID: PMC11817531 DOI: 10.1080/19490976.2025.2458189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
High-dose radiation exposure results in gastrointestinal (GI) acute radiation syndrome identified by the destruction of mucosal layer, intestinal growth barrier dysfunction, and aberrant inflammatory responses. Further, radiation causes gut microbiome dysbiosis characterized by diminished microbial diversity, mostly commensal bacteria, and the spread of bacterial pathogens that trigger the recruitment of immune cells and the production of pro-inflammatory factors that lead to further GI tissue damage. Currently, there are no U.S. Food and Drug Administration (FDA) approved countermeasures that can treat radiation-induced GI injuries. To meet this critical need, Synedgen Inc. has developed a glycopolymer radiomitigator (MIIST305) that is specifically targeted to the GI tract, which acts by intercalating into the mucus layer and the glycocalyx of intestinal epithelial cells that could potentially ameliorate the deleterious effects of radiation. Male C57BL/6J adult mice were exposed to 13 Gy partial body X-irradiation with 5% bone marrow shielding and MIIST305 was administered on days 1, 3, and 5 post-irradiation. Approximately 85% of the animals survived the irradiation exposure and were apparently healthy until the end of the 30-day study period. In contrast, no control, Vehicle-treated animals survived past day 10 at this radiation dose. We show that MIIST305 improved intestinal epithelial barrier function and suppressed systemic inflammatory responses mediated by radiation-induced pro-inflammatory cytokines. Taxonomic profiling and community structure of the fecal and colonic mucosa microbiota demonstrated that MIIST305 treatment increased microbial diversity and restored abundance of beneficial commensal bacteria, including Lactobacillus and Bifidobacterium genera while suppressing potentially pathogenic bacteria Enterococcus and Staphylococcus compared with Vehicle-treated animals. In summary, MIIST305 is a novel GI-targeted therapeutic that greatly enhances survival in mice exposed to lethal radiation and protects the GI tract from injury by restoring a balanced gut microbiota and reducing pro-inflammatory responses. Further development of this drug as an FDA-approved medical countermeasure is of critical importance.
Collapse
Affiliation(s)
- Debmalya Mitra
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabriel K. Armijo
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth H. Ober
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Helen C. Turner
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Constantinos G. Broustas
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
2
|
Hu D, Zeng H, Li W, Zhang Y, Chi X, Liu X, Zhang H, Ge G, Jiao X, Xie P. Discovery of a Novel Serine-Targeting Covalent Inhibitor against HCES2A for Treating Drug-induced Diarrhea and Ulcerative Colitis. J Med Chem 2025. [PMID: 40396776 DOI: 10.1021/acs.jmedchem.5c00563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Mammalian carboxylesterases play an important role in the hydrolysis of both endogenous substrates and xenobiotics bearing ester or amide bond(s). We previously reported that bysspectin A and its derivative LC-20W were potent reversible hCES2A inhibitors. Here, a series of bysspectin A derivatives were designed and synthesized using LC-20W as the leading compound. Compound 9d was identified as a potent serine-targeting covalent inhibitor of hCES2A (IC50 = 0.12 nM), which was much more potent than that of LC-20W. Further chemoproteomics and docking simulations showed that 9d could selectively modify hCES2A at the catalytic serine (Ser228), thereby blocking its catalytic activity. Notably, 9d showed good cell-membrane permeability and was capable of inhibiting intracellular hCES2A in living cells. In vivo tests showed that 9d significantly alleviates irinotecan-induced diarrhea and dextran sulfate sodium-induced colitis. Collectively, a novel serine-targeting covalent inhibitor against hCES2A was developed, offering a promising candidate for treating drug-induced diarrhea and ulcerative colitis.
Collapse
Affiliation(s)
- Danyang Hu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Hairong Zeng
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine; Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, People's Republic of China
| | - Wenxuan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Ya Zhang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine; Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, People's Republic of China
| | - Xiaoqian Chi
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Xiaoyu Liu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Haijing Zhang
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Guangbo Ge
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine; Shanghai Frontiers Science Center of TCM Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, People's Republic of China
| | - Xiaozhen Jiao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| | - Ping Xie
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China
| |
Collapse
|
3
|
Zhang B, Tian M, Yang Y, Qiu Y, Wang L, Xiao H, Zhu X, Qin L, Yang X, Jiang Z. Dietary bile acid supplementation improves the intestinal health and growth performance of piglets partially through the FXR/AQPs pathway. Porcine Health Manag 2025; 11:28. [PMID: 40400018 DOI: 10.1186/s40813-025-00440-x] [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: 06/03/2024] [Accepted: 04/16/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Maintaining the integrity of the structure and function of piglet intestines is crucial for their growth and health. This study aims to evaluate the effects of an antibiotic free diet supplemented with bile acid on gut health and growth performance of weaned piglets, and to explore their regulatory mechanisms. METHODS Thirty-two weaned piglets were randomly divided into two groups and fed either a basal diet or a basal diet supplemented with 350 mg/kg bile acid. RESULTS Dietary supplementation with bile acid increased the average daily gain (ADG) and final weight of piglets, and reduced the diarrhea incidence (P < 0.05), which was verified to be related to the improvement of lipid absorption, amino acid transport, and intestinal barrier function. Bile acid increased the concentration of lipase and decreased the concentration of total cholesterol, total glyceride, low-density lipoprotein, and urea nitrogen in serum (P < 0.05). Meanwhile, bile acid improved the mRNA expression of amino acid transporters in the intestine. On the other hand, bile acid decreased the pH values of the stomach, jejunum, and colon, and improved intestinal morphology (P < 0.05). The real-time quantitative PCR results showed that bile acid increased the mRNA expression of Occludin and ZO-1 in the duodenum and ileum (P < 0.05). Moreover, dietary bile acid supplementation altered the composition of the ileal microbiota in piglets and increased the relative abundance of Ligilactobacillus. In vitro, bile acid improved the repair of IPEC-J2 cells after injury and was shown to be associated with the activation of farnesoid X receptors (FXR) and increased expression of tight junction proteins and aquaporins (AQPs) proteins. CONCLUSION This study found that dietary bile acid supplementation promotes the intestinal health and nutrient absorption partially through the FXR/AQPs pathway, ultimately improving growth performance of piglets.
Collapse
Affiliation(s)
- Beibei Zhang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, China
| | - Min Tian
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong, China
| | - Yahui Yang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, China
| | - Yueqin Qiu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong, China
| | - Li Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong, China
| | - Hao Xiao
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong, China
| | - Xiaoping Zhu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong, China
| | - Limei Qin
- School of Life Sciences and Engineering, Foshan University, Foshan, China.
| | - Xuefen Yang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, China.
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong, China.
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China.
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong, China.
| | - Zongyong Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangdong, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangdong, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangdong, China
| |
Collapse
|
4
|
Zhang R, Duan S, Ma X, Guo Z, Szeto IMY, Shi X, Zhao C, Yan Y, Li B. Synergistic effect of 2'-fucosyllactose and osteopontin on intestinal mucosal immunity injury. Food Funct 2025; 16:4134-4148. [PMID: 40308162 DOI: 10.1039/d4fo05287k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Previous studies have demonstrated the beneficial effects of 2'-fucosyllactose (2'-FL) and osteopontin (OPN) in modulating intestinal mucosal immunity. Nevertheless, the potential synergistic interactions and underlying mechanisms of 2'-FL and OPN have not been fully elucidated. To address this question, this study employed Sprague-Dawley (SD) rats to establish a lipopolysaccharide (LPS)-induced model simulating human microbiota-associated intestinal barrier damage. The findings indicate that the combined administration of 2'-FL and OPN exerts a considerable protective effect on the intestinal barrier, surpassing the individual efficacy of 2'-FL or OPN alone. The administration of 2'-FL and OPN mitigated body weight loss, attenuated disease activity index (DAI) scores, reduced serum myeloperoxidase (MPO) activity, and improved intestinal histopathology. In addition, 2'-FL and OPN significantly increased the mRNA expression of MUC2, ZO-1, and claudin-2, and reduced serum diamine oxidase (DAO) and D-lactate levels. 2'-FL and OPN reduced the levels of pro-inflammatory cytokines IL-6, and IL-1β, and elevated the levels of anti-inflammatory cytokines IL-10, IL-4, and secretory immunoglobulin A (sIgA) in the intestine. 2'-FL and OPN significantly improved the balance in CD4+/CD8+, Th1/Th2, and Th17/Treg cells. Moreover, 2'-FL and OPN regulated LPS-induced dysbiosis of the intestinal microbiota, increased the abundance of Lactobacillus and Romboutsia, and reduced the abundance of Escherichia-Shigella and Alloprevotella. Overall, 2'-FL and OPN synergistically protected against LPS-induced intestinal mucosal barrier damage in rats by regulating intestinal permeability, attenuating the inflammatory response, balancing intestinal immune cells, and modulating intestinal microbiota composition.
Collapse
Affiliation(s)
- Rui Zhang
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Sufang Duan
- National Center of Technology Innovation for Dairy, Hohhot 010110, China
- Inner Mongolia Yili Industrial Group, Co. Ltd, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China.
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Xinming Ma
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Zhengtao Guo
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Ignatius Man-Yau Szeto
- National Center of Technology Innovation for Dairy, Hohhot 010110, China
- Inner Mongolia Yili Industrial Group, Co. Ltd, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China.
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Xiaodan Shi
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Congrui Zhao
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Yalu Yan
- Inner Mongolia Yili Industrial Group, Co. Ltd, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China.
- Inner Mongolia Dairy Technology Research Institute Co. Ltd, Hohhot 010110, China
| | - Bailiang Li
- Food College, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
5
|
You M, Li J, Wang X, Liu Y, Chen S, Wang P. Targeting SLC7 A11 Ameliorates Ulcerative Colitis by Promoting Efferocytosis Through the ERK1/2 Pathway. Inflammation 2025:10.1007/s10753-025-02312-6. [PMID: 40360947 DOI: 10.1007/s10753-025-02312-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/28/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
OBJECTIVE AND DESIGN This study investigates the effect and underlying mechanism of targeting SLC7A11 in mitigating dextran sulfate sodium (DSS)-induced intestinal inflammation and injury in colitis. METHODS We utilized wild-type and SLC7A11-/+ mice to assess the inflammatory damage in DSS-induced colitis in vivo. In vitro, colon tissues from patients with ulcerative colitis were analyzed to compare SLC7A11 expression between inflamed and non-inflamed regions. Further mechanistic insights were obtained using Caco-2 cells and bone marrow-derived dendritic cells (BMDCs). RESULTS In human colon tissues, SLC7A11 expression was significantly elevated in inflamed regions compared to non-inflamed areas, particularly in dendritic cells. In vivo inhibition of SLC7A11 markedly alleviated DSS-induced colitis symptoms. In vitro, suppressing SLC7A11 restored the integrity of the Caco-2 monolayer intestinal epithelial model. Both knockout and inhibition of SLC7A11 enhanced ERK1/2 phosphorylation and increased efferocytosis in BMDCs. CONCLUSIONS Targeting SLC7A11 augments dendritic cell efferocytosis and preserves intestinal epithelial barrier function, potentially offering a therapeutic avenue for alleviating ulcerative colitis.
Collapse
Affiliation(s)
- Meiyi You
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Jichang Li
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Xin Wang
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Yucun Liu
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Shanwen Chen
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China.
| | - Pengyuan Wang
- Department of Gastrointestinal Surgery, Peking University First Hospital, Beijing, 100034, People's Republic of China.
| |
Collapse
|
6
|
Wang S, Yang X, Liu X, Wen Q, Xu L, Feng M, Lang J, Liu D. Iron modulates barrier integrity and stem cell function of small intestine during experimental colitis. Front Nutr 2025; 12:1545956. [PMID: 40416374 PMCID: PMC12100934 DOI: 10.3389/fnut.2025.1545956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/21/2025] [Indexed: 05/27/2025] Open
Abstract
Background Ulcerative colitis (UC) brings inconvenience to many patients with inflammatory bowel disease (IBD). Although colonic pathology is widely investigated, little attention has been paid to the disorders in small intestine of UC. In this study, we investigated the impairments of UC to small intestine and further explored how iron metabolism regulated epithelial integrity and the activity of intestinal stem cells (ISCs). Methods Mice were treated by 2.5% dextran sulfate sodium (DSS) for 7 days to established acute experimental colitis. Small intestinal tissues were collected at different time points in the process of DSS-induced colitis. Histological analysis was used to evaluate the changes of small intestine, including H&E, Alcian blue and PAS staining, immunostaining, and qRT-PCR. Iron content was modulated by the supplementation of ferric citrate or depletion by deferoxamine (DFO). The influence of iron on the barrier integrity and stem cell function was further determined by histology, IEC-6 cell, and enteroid culture. ROS content was demonstrated by DHE staining. The proliferation of intestinal stem cells (ISCs) was shown by BrdU and Olfm4 staining, and Lgr5-tdTomato mice were used for lineage tracing study. Results It was shown that during DSS-induced colitis, small intestine underwent a serious injury process, including dysregulated integrity and decreased proliferation of ISCs. Iron overload significantly exacerbated intestinal injury in tissues, epithelial cell line, and intestinal organoids. However, iron chelation by deferoxamine (DFO) would greatly suppress small intestinal injury. Mechanistically, iron overload exacerbated the generation of ROS and enhanced the infiltration of immune cells. In addition, STAT3 and ERK pathways in intestinal epithelium were impaired during experimental colitis, and iron content significantly interrupted the expression of p-STAT3 and p-ERK1/2 within small intestine. Conclusion In summary, this study proved that small intestine was also impaired in experimental colitis, and iron content could affect DSS-induced small intestinal damage and regeneration, indicating the strategy of iron supplementation in clinical practice needs to be more cautious and consider more factors.
Collapse
Affiliation(s)
- Shubin Wang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiangjie Yang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiangjun Liu
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qin Wen
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Xu
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Mei Feng
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Medical Oncology, The Third People's Hospital of Sichuan Province, Chengdu, China
| | - Jinyi Lang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dengqun Liu
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital and Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Experimental Research, Sichuan Cancer Hospital and Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
7
|
Chaves LS, Oliveira ACP, Oliveira AP, Lopes ALF, Araujo AKS, Pacheco G, Silva KC, Martins FEC, Gomes IAB, Ramos SVS, Viana HTMC, Batista AVF, Oliveira BC, Nicolau LAD, Ribeiro FOS, Castro AV, de Araujo-Nobre AR, Silva DA, Cordeiro LMC, Góis MB, Medeiros JVR. Cashew gum fractions protect intestinal mucosa against shiga toxin-producing Escherichia coli infection: Characterization and insights into microbiota modulation. Int J Biol Macromol 2025; 311:143916. [PMID: 40324507 DOI: 10.1016/j.ijbiomac.2025.143916] [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: 02/03/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Diarrheal diseases remain a major public health concern, particularly in regions with poor sanitation. Polysaccharides extracted from natural gums have been investigated as functional agents for intestinal health, and their fractionation enables the production of oligosaccharides with potential prebiotic activity. This study aimed to produce cashew gum (CG) fractions through Smith degradation (CGD48) and partial hydrolysis (CGD24) and to evaluate their ability to modulate and protect the intestinal microbiota. Balb/c mice were administered CG (1200 mg/kg), CGD24 (800 mg/kg), or CGD48 (800 mg/kg) for 10 and 26 days, followed by infection with Shiga toxin-producing Escherichia coli (STEC) (5 × 1010 CFU/mL) for three days. Characterization assays confirmed the fragmentation of CG. Both CGD24 and CGD48 promoted the growth of beneficial bacteria with and without infection and reduced STEC colonization. Furthermore, they preserved mucin levels in the cecum and large intestine and maintained baseline levels of superoxide dismutase (SOD), suggesting protection of the intestinal mucosa. These findings indicate that CG fractions exhibit microbiota-modulating and protective effects against STEC, highlighting their therapeutic potential and the need for further studies to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Letícia S Chaves
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Antonio C P Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Ana P Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - André L F Lopes
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Andreza K S Araujo
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Gabriella Pacheco
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Katriane C Silva
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Francisco E C Martins
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Isaac A B Gomes
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Sabrine V S Ramos
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Hémilly T M C Viana
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Ana V F Batista
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Beatriz C Oliveira
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Lucas A D Nicolau
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil
| | - Fábio O S Ribeiro
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Auricélia V Castro
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Alyne Rodrigues de Araujo-Nobre
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Durcilene A Silva
- Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil
| | - Lucimara M C Cordeiro
- Department of Biochemistry and Molecular Biology, Federal University of Parana, Curitiba, PR, Brazil
| | - Marcelo B Góis
- Post-Graduation Program in Biosciences and Health, Federal University of Rondonópolis, Rondonópolis, Brazil
| | - Jand V R Medeiros
- Laboratory of Inflammation and Translational Gastroenterology (LIGAT), Postgraduate Program in Biotechnology (PPGBIOTEC), Federal University of Parnaiba Delta, Av. São Sebastião, 2819, Parnaíba, PI CEP 64202-020, Brazil; Research Center on Biodiversity and Biotechnology (BIOTEC), Federal University of Delta do Parnaíba, UFDPar, Parnaíba, PI CEP 64202-020, Brazil.
| |
Collapse
|
8
|
Tram NDT, Xu J, Chan KH, Rajamani L, Ee PLR. Bacterial clustering biomaterials as anti-infective therapies. Biomaterials 2025; 316:123017. [PMID: 39708775 DOI: 10.1016/j.biomaterials.2024.123017] [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: 08/13/2024] [Revised: 11/23/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
In Nature, bacterial clustering by host-released peptides or nucleic acids is an evolutionarily conserved immune defense strategy employed to prevent adhesion of pathogenic microbes, which is prerequisite for most infections. Synthetic anti-adhesion strategies present as non-lethal means of targeting bacteria and may potentially be used to avoid resistance against antimicrobial therapies. From bacteria-agglutinating biomolecules discovered in nature to synthetic designs involving peptides, cationic polymers and nanoparticles, the modes of actions appear broad and unconsolidated. Herein, we present a critical review and update of the state-of-the-art in synthetic bacteria-clustering designs with proposition of a more streamlined nomenclature and classification. Overall, this review aims to consolidate the conceptual framework in the field of bacterial clustering and highlight its potentials as an avenue for discovering novel antibacterial biomaterials.
Collapse
Affiliation(s)
- Nhan Dai Thien Tram
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore
| | - Jian Xu
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore
| | - Kiat Hwa Chan
- Division of Science, Yale-NUS College, 16 College Avenue West, Singapore, 138527, Singapore; NUS College, National University of Singapore, 18 College Avenue East, Singapore, 138593, Singapore
| | - Lakshminarayanan Rajamani
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore; Ocular Infections and Anti-Microbials Research Group, Singapore Eye Research Institute, Singapore, 169856, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Graduate Medical School, Singapore, 169857, Singapore
| | - Pui Lai Rachel Ee
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore.
| |
Collapse
|
9
|
Chen KH, Xu R, Ye HJ, Xu B, Cao SL, Chen HX, Chen YL, Cai YP, Xie XQ, Zhao M, Zhou L, Luo X. Evaluating the efficacy and safety of emodin, luteolin, and paeonol combination from Dahuang Mudan decoction in ameliorating ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119692. [PMID: 40157404 DOI: 10.1016/j.jep.2025.119692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 03/11/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dahuang Mudan Decoction is a classic Chinese medicine prescription for treating ulcerative colitis (UC). Previous studies have shown that Dahuang Mudan Decoction has preventive and therapeutic effects on mice with dextran sulfate sodium (DSS) induced colitis. AIM OF THE STUDY The objective of this research endeavor was to ascertain the most efficacious synergistic blend of Emodin, Luteolin, and Paeonol, the main active ingredients in Dahuang Mudan Decoction, in alleviating UC. Additionally, it sought to elucidate the underlying therapeutic mechanisms and evaluate the safety of the combined components. MATERIALS AND METHODS Employing Emodin, Luteolin, and Paeonol as starting materials, the optimal combination was selected by orthogonal design. Basic pharmacodynamics was observed in mouse model of UC induced by DSS. The pathological changes of the colon were observed using hematoxylin and eosin (H&E) staining. The changes of cytokines and proteins related to inflammation and intestinal barrier function were detected by WB, Alcian blue staining, immunofluorescence, immunohistochemistry and related kits. Subsequently, 16S rRNA sequencing was used to observe changes in the intestinal flora. To evaluate the therapeutic effect and potential mechanism of the optimal monomer composition on UC mouse model. Finally, we performed toxicity tests as part of the safety assessment of the combination of the three monomers. RESULTS The different combinations of Emodin, Luteolin, and Paeonol alleviated DSS-induced colitis to varying degrees. The ELP5 group (Emodin 5 mg/kg + Luteolin 5 mg/kg + Paeonol 15 mg/kg) and ELP9 group (Emodin 15 mg/kg + Luteolin 15 mg/kg + Paeonol 75 mg/kg) had the most significant mitigation effect on UC mice. Mechanistically, the monomeric composition provides a comprehensive treatment for UC by addressing multiple aspects, including anti-inflammatory and antioxidant effects, repairing the damaged intestinal barrier, restoring the intestinal flora structure, and regulating short-chain fatty acid levels. In addition, the combination of Emodin, Luteolin and Paeonol exhibited a more significant effect on DSS-induced colitis compared to the individual components, indicating a synergistic effect among them. In the single-dose toxicity test, no obvious abnormalities were found in the general state or major organs of the mice. In repeated toxicity tests, it was found that the combined use of three monomers had less effect on organ index, hematology and serum biochemical indexes than that of a single compound. Pathological examination showed that the three monomers had certain toxicity to mouse liver, kidney and lung when used alone and in large doses for a long time, and the toxicity was significantly reduced after combined use. CONCLUSIONS We have determined the optimal combination of three active ingredients in Dahuang Mudan Decoction to alleviate DSS induced colitis in mice by inhibiting intestinal inflammation and oxidative stress, repairing impaired intestinal barrier function, and regulating intestinal flora disturbance. The results of single administration toxicity test proved the safety of the three monomers combined, and repeated administration toxicity test clarified the safe dose range of the combined administration, and also revealed that the combined therapy exhibited superior safety compared to monotherapy.
Collapse
MESH Headings
- Animals
- Emodin/pharmacology
- Emodin/administration & dosage
- Emodin/therapeutic use
- Emodin/toxicity
- Colitis, Ulcerative/drug therapy
- Colitis, Ulcerative/chemically induced
- Colitis, Ulcerative/pathology
- Acetophenones/administration & dosage
- Acetophenones/pharmacology
- Acetophenones/toxicity
- Acetophenones/therapeutic use
- Luteolin/pharmacology
- Luteolin/administration & dosage
- Luteolin/therapeutic use
- Luteolin/toxicity
- Dextran Sulfate
- Mice
- Male
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/administration & dosage
- Drugs, Chinese Herbal/therapeutic use
- Drugs, Chinese Herbal/toxicity
- Disease Models, Animal
- Mice, Inbred C57BL
- Colon/drug effects
- Colon/pathology
- Drug Therapy, Combination
- Gastrointestinal Microbiome/drug effects
- Anti-Inflammatory Agents/pharmacology
Collapse
Affiliation(s)
- Ke-Han Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rui Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hua-Jian Ye
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shui-Ling Cao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hong-Xu Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun-Liang Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yan-Ping Cai
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xue-Qian Xie
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meng Zhao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lian Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Xia Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China; StateKey Laboratory of Traditional Chinese Medicine Syndrome, China.
| |
Collapse
|
10
|
Luo Y, Luo L, Xia M, Liu Q, Zhang G. Studies on the changes in rectal permeability and intestinal microbiota with developmental age in young rats. Front Microbiol 2025; 16:1551693. [PMID: 40336831 PMCID: PMC12058081 DOI: 10.3389/fmicb.2025.1551693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/31/2025] [Indexed: 05/09/2025] Open
Abstract
Introduction The gut contains a diverse array of commensal microorganisms, forming a vital biological barrier within the intestine that contributes to the overall intestinal mucosal barrier. However, research on the rectal barrier during early development remains limited. This study aims to investigate the relationship between intestinal microbiota and rectal barrier function in young rats. Methods We evaluated the rectal barrier structure and function in rats at 2-, 4-, and 10-week-old. Methodology included histological analysis, Muc2 expression quantification, immunofluorescence localization of tight junction proteins (ZO-1, Occludin, Claudins), blood glucose monitoring after rectal insulin administration, and 16S rDNA sequencing of rectal microbiota. Spearman correlation analysis was used to explore mechanisms linking age-dependent changes in rectal permeability to microbiota dynamics. Results Physiological rectal permeability was significantly higher in 2-week-old rats compared to 4- and 10-week-old rats (p < 0.01), although systemic biomarkers (LPS, D-LA, and LBP) showed no significant differences. The rectal microbiota exhibited marked age-dependent shifts in composition, α/β-diversity, and metabolic pathways, with increased abundance of beneficial taxa (e.g., Muribaculaceae, Akkermansia) in older rats. Correlation analysis revealed strong associations between reduced permeability, elevated Occludin expression, and microbiota maturation (R = 0.65, p < 0.001). Conclusion This study demonstrates that age-dependent maturation of the rectal barrier is closely linked to microbiota composition and tight junction protein expression, providing insights into developmental mechanisms and potential strategies for pediatric rectal drug delivery.
Collapse
Affiliation(s)
- Yunfeng Luo
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Liangming Luo
- Yudu County Hospital of Traditional Chinese Medicine, Ganzhou, China
| | - Mengle Xia
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Qian Liu
- Discipline of Chinese and Western Integrative Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Jiangxi University of Chinese Medicine, Nanchang, China
| | - Guosong Zhang
- Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
11
|
Vital KD, Pires LO, Gallotti B, Silva JL, Lima de Jesus LC, Alvarez-Leite JI, Ferreira Ê, de Carvalho Azevedo VA, Santos Martins F, Nascimento Cardoso V, Antunes Fernandes SO. Atorvastatin attenuates intestinal mucositis induced by 5-fluorouracil in mice by modulating the epithelial barrier and inflammatory response. J Chemother 2025; 37:175-192. [PMID: 38711347 DOI: 10.1080/1120009x.2024.2345027] [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: 10/25/2023] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024]
Abstract
Chemotherapy-induced intestinal mucositis is a major side effect of cancer treatment. Statins are 3-hydroxy-3-methyl glutaryl coenzyme reductase inhibitors used to treat hypercholesterolemia and atherosclerotic diseases. Recent studies have demonstrated that atorvastatin (ATV) has antioxidant, anti-inflammatory, and resulting from the regulation of different molecular pathways. In the present study, we investigated the effects of ATV on intestinal homeostasis in 5-fluorouracil (5-FU)-induced mucositis. Our results showed that ATV protected the intestinal mucosa from epithelial damage caused by 5-FU mainly due to inflammatory infiltrate and intestinal permeability reduction, downregulation of inflammatory markers, such as Tlr4, MyD88, NF-κB, Tnf-a, Il1β, and Il6 dose-dependent. ATV also improved epithelial barrier function by upregulating the mRNA transcript levels of mucin 2 (MUC2), and ZO-1 and occludin tight junction proteins. The results suggest that the ATV anti-inflammatory and protective effects on 5-FU-induced mice mucositis involve the inhibition of the TLR4/MYD88/NPRL3/NF-κB, iNos, and caspase 3.
Collapse
Affiliation(s)
- Kátia Duarte Vital
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiz Octavio Pires
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno Gallotti
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Janayne Luihan Silva
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luís Cláudio Lima de Jesus
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Ênio Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vasco Ariston de Carvalho Azevedo
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Flaviano Santos Martins
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Valbert Nascimento Cardoso
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Simone Odília Antunes Fernandes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
12
|
Zhu L, Luo Y, Liu Y, Sun S, Yuan J, Zhang L, Zhong W, Ma S, Yu Z, Zhou J, Chen X, Zhao J. Clostridium butyricum ameliorates indomethacin-induced enteropathy by promoting MUC2 secretion via suppressing the Notch pathway. Front Microbiol 2025; 16:1509876. [PMID: 40177488 PMCID: PMC11961966 DOI: 10.3389/fmicb.2025.1509876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Nonsteroidal anti-inflammatory drug (NSAID) enteropathy is a serious clinical complication with no effective treatments available. Modulating the intestinal microbiota through dietary and nutritional targets is a promising strategy for preventing NSAID enteropathy. This study aimed to investigate the protective effect and underlying mechanisms of the probiotic Clostridium butyricum (CB) on indomethacin (IND)-induced enteropathy. C57BL/6J mice received CB treatment for 14 days along with concurrent IND gavage for the final 7 days. Caco2 cells were stimulated with IND to evaluate the effect of CB supernatant (CBS) on the intestinal barrier function, and LS174T cells were used to validate the modulatory action of CBS on the Notch signaling pathway. Our findings revealed that CB treatment prevented anorexia and weight loss, reduced the severity of enteropathy, and decreased the inflammatory response of the small intestine. CB also increased the expression of tight junction proteins and reduced permeability in mice and Caco2 cells. Additionally, CB suppressed apoptosis and promoted proliferation in the small intestine. Further research found that CB increased the number of goblet cells and MUC2 secretion. Mechanistically, CB may promote MUC2 secretion by suppressing the Notch signaling pathway, consistent with the results of intervention in LS174T cells with CBS. In conclusion, CB might prevent NSAID enteropathy by increasing MUC2 secretion through the inhibition of the Notch pathway. Our study identified the potential efficacy of CB as a preventive strategy against NSAID enteropathy and showed promising prospects for CB as a food supplement.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xin Chen
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingwen Zhao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
13
|
Zhang L, Feng L, Yang L, Wan H. Noninvasive Detection and Monitoring of the Integrity of the Intestinal Barrier through NIR-II Fluorescence Imaging and Colorimetric Urinalysis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13445-13460. [PMID: 39977645 DOI: 10.1021/acsami.4c20263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
The variation of the integrity of the intestinal barrier is closely related to the occurrence and progression of various diseases, making its accurate detection and monitoring essential to provide reliable information for guiding the refinement of treatment scenarios. Herein, we developed a strategy utilizing glutathione-capped gold clusters (Au-GSH) to achieve simultaneous in vivo second near-infrared (NIR-II) fluorescence imaging and in vitro colorimetric urinalysis for noninvasive detection and monitoring of intestinal barrier integrity. After oral administration, Au-GSH can demonstrate different distribution behaviors in terms of the variation of intestinal barrier integrity. Specifically, Au-GSH could selectively permeate through compromised intestinal barrier to be distributed into the bladder in view of its ultrasmall size below the glomerular filtration cutoff, leading to the rapid excretion through urine. Such process can be visually profiled by collecting NIR-II fluorescence (>1100 nm) emitting from Au-GSH in a noninvasive real-time manner. By virtue of the peroxidase-like activity of Au-GSH, the simple colorimetric urinalysis was further established for evaluating the integrity of the intestinal barrier, advancing the detection and monitoring performance. As a result, our developed strategy successfully detected the damage of intestinal barrier integrity in ulcerative colitis (UC) mice, a common disease characterized by compromised intestinal barrier integrity. Excitingly, the monitoring of restoration of intestinal barrier integrity in both therapeutic and preventative modes for UC mice was also realized by our strategy, making it a promising diagnosis scenario for diseases related to the variation of intestinal barrier integrity.
Collapse
Affiliation(s)
- Li Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Lei Feng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Lin Yang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Hao Wan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| |
Collapse
|
14
|
Kilari G, Tran J, Blyth GAD, Cobo ER. Human cathelicidin LL-37 rapidly disrupted colonic epithelial integrity. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184410. [PMID: 39837472 DOI: 10.1016/j.bbamem.2025.184410] [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: 07/01/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
The intestinal barrier, held together by epithelial cells and intercellular tight junction (TJ) proteins, prevents the penetration of microbial pathogens. Concurrently, intestinal epithelial cells secrete antimicrobial peptides, including cathelicidin. Cathelicidin has direct antibacterial and immunomodulatory functions, although its role in intestinal integrity remains elusive. In this study, we demonstrate that direct stimulation of human colonic epithelial (T84) cells with human cathelicidin, LL-37, resulted in a rapid and transient increase in epithelial cell permeability. This increased permeability was associated with the TJ proteins occludin and claudin-2 degradation, mediated by these specific proteins' endocytosis and lysosomal degradation. While murine cathelicidin (CRAMP) failed to modify T84 cell permeability, LL-37 degraded TJ proteins in murine rectal epithelial (CMT-93) cells. The stimulus of (CMT-93) cells with LL-37 aggravated the cell permeability and furthered TJ degradation provoked by the intestinal pathogen, attaching/effacing (A/E) Citrobacter rodentium (C. rodentium). The number of C. rodentium that colonized CMT-93 cells was not severely impacted by the presence of LL-37. While a temporary disruption of tight junctions by LL-37 may lead to a 'leaky gut,' this study demonstrates that LL-37 increases epithelial cell permeability by degrading TJ proteins occludin and claudin-2 through endocytosis and lysosomal degradation. These immunomodulatory actions occurring at concentrations lower than those microbicidal uncover a new guise for cathelicidin modulating the epithelial barrier against A/E pathogens. Recognizing native cathelicidin's functions in a specified disease setting (e.g., colitis) will help establish it as an anti-infectious immunomodulator.
Collapse
Affiliation(s)
- Geeta Kilari
- Faculty of Veterinary Medicine, University of Calgary, Canada
| | - Jacquelyn Tran
- Faculty of Veterinary Medicine, University of Calgary, Canada
| | | | - Eduardo R Cobo
- Faculty of Veterinary Medicine, University of Calgary, Canada.
| |
Collapse
|
15
|
Yang Q, Li Y, Wang X, Ding Q, Tao Y, Li P, Lian X, Chen Y, Zhao L. A high cholesterol diet aggravates experimental colitis through SREBP2-modulated endocytosis and degradation of occludin and Zo-1 proteins. FEBS J 2025; 292:1052-1069. [PMID: 39279038 DOI: 10.1111/febs.17269] [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: 01/11/2024] [Revised: 06/01/2024] [Accepted: 09/02/2024] [Indexed: 09/18/2024]
Abstract
Disrupted cholesterol homeostasis plays a critical role in the development of multiple diseases, such as cardiovascular disease and cancer. However, the role of cholesterol in inflammatory bowel disease (IBD) remains unclear. In the present study, we investigated whether and how high levels of cholesterol in the diet affect experimental colitis in mice. A normal diet supplemented with 1.25% cholesterol (high cholesterol diet) caused more severe colitis and aggravated the disruption of intestinal tight junction structure, accompanied by higher colonic tissue total cholesterol (TC) levels in a dextran sulfate sodium (DSS)-induced experimental colitis mouse model. Cholesterol aggravated DSS-induced intestinal epithelial barrier impairment and nuclear sterol regulatory element-binding protein 2 (nSREBP2) inhibition both in vivo and in vitro. In addition, nSREBP2 overexpression ameliorated cholesterol-induced intestinal epithelial barrier disruption in Caco2 cells. Interestingly, inhibition of SREBP2 disrupted intestinal epithelial barrier in the absence of cholesterol. Furthermore, SREBP2 regulated the protein expression of tight junction proteins (occludin/Zo-1) via modulating caveolin-1-mediated endocytosis and lysosomal degradation. Analysis of UK Biobank data indicated that, in fully adjusted models, higher serum TC concentrations were an independent protective factor for IBD incidence. The sterol regulatory element-binding factor 2 (SREBF2) gene rs2228313 (G/C) genetic variant was associated with the incidence of IBD and the CC genotype of SREBF2 rs2228313 was associated with higher serum TC levels and decreased the risk of IBD. In summary, a high cholesterol diet aggravates DSS-induced colitis in mice by down-regulating nSREBP2 expression, thereby promoting the endocytic degradation of tight junction proteins. In humans, SREBF2 gene single nucleotide polymorphism rs2228313 and serum TC levels are associated with IBD incidence.
Collapse
Affiliation(s)
- Qin Yang
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Yongjia Li
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Xingxing Wang
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Qiuying Ding
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Yi Tao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, China
| | - Pan Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, China
| | - Xuemei Lian
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
- School of Public Health, Chongqing Medical University, China
| | - Yaxi Chen
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| | - Lei Zhao
- Centre for Lipid Research & Chongqing Key Laboratory of Metabolism on Lipid and Glucose, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, China
| |
Collapse
|
16
|
Li Y, Yue X, Ren X, Pang Y, Wang T, Huangfu B, Mikhailovich ZA, Vasilievich KV, Zhang M, Luan Y, Wang Q, He X. Mare milk and fermented mare milk alleviate dextran sulfate sodium salt-induced ulcerative colitis in mice by reducing inflammation and modulating intestinal flora. J Dairy Sci 2025; 108:2182-2198. [PMID: 39647629 DOI: 10.3168/jds.2024-25181] [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: 05/29/2024] [Accepted: 11/10/2024] [Indexed: 12/10/2024]
Abstract
Mare milk (MM) and fermented mare milk (FM) are specialized animal milks with high nutritional value, containing a variety of functionally active substances that are capable of resisting inflammatory responses and oxidative stress. However, little relevant research on the maintenance of intestinal homeostasis has been performed. This study aimed to investigate the effects of MM and FM on the prevention of dextran sulfate sodium salt (DSS)-induced ulcerative colitis in a mouse model and to preliminarily elucidate the underlying mechanisms. The results showed that MM and FM had different degrees of protective effects against the damage caused by DSS and alleviated ulcerative colitis by inhibiting weight loss, reducing colon length shortening, and restoring intestinal structure. Additionally, MM and FM maintained intestinal tight junction protein levels to repair barrier function, downregulated inflammatory cytokines (e.g., IL-1β, TNF-α, IL-6, and iNOS) and bolstered the body's antioxidant defense system. Moreover, MM and FM regulated dysregulation of the intestinal microenvironment by improving the diversity of the gut microbiota and reshaping its structure, including increasing the proportion of Firmicutes and Bacteroidetes and the relative abundance of beneficial bacterial genera (e.g., Akkermansia). In summary, MM and FMM can serve as dietary resources for preventing ulcerative colitis and maintaining intestinal homeostasis.
Collapse
Affiliation(s)
- Yi Li
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083
| | - Xiaoyu Yue
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China 100193
| | - Xinxin Ren
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083
| | - Yang Pang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083
| | - Teng Wang
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083
| | - Bingxin Huangfu
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083
| | | | | | - Mu Zhang
- Shenyang Agricultural University, Shenyang, China 110161
| | - Yue Luan
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China 100193
| | - Qin Wang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China 100193.
| | - Xiaoyun He
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China 100083.
| |
Collapse
|
17
|
Ding YX, Chen LL, Li KW, Zou L, Liao LM, Han XY, OuYang J, Wu YP, Zhang WD, Chu HR. Assessing the impact of moxibustion on colonic mucosal integrity and gut microbiota in a rat model of cerebral ischemic stroke: insights from the "brain-gut axis" theory. Front Neurol 2025; 16:1450868. [PMID: 40083458 PMCID: PMC11903257 DOI: 10.3389/fneur.2025.1450868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 02/08/2025] [Indexed: 03/16/2025] Open
Abstract
Objective The aim of this study is to assess the impact of moxibustion on the colonic mucosal barrier and gut microbiota in a rat model of cerebral ischemic stroke (CIS). Method The CIS rat model was established using the modified Zea Longa suture method. Successfully modeled rats were randomly allocated into a model group and a moxibustion group, with a sham surgery group serving as the control. The moxibustion group received suspended moxibustion at Dazhui (GV 14), Baihui (GV 20), Fengfu (GV 16), and bilateral Tianshu (ST 25) and Shangjuxu (ST 37) acupoints. Neurological function was assessed using the Longa score, and brain infarct size was assessed through 2,3,5-triphenyl tetrazolium chloride staining. Gut microbiota composition was analyzed using 16S rDNA amplification sequencing. Intestinal mucosal permeability was evaluated using the FITC-Dextran tracer method. The serum ET-1 levels and the expression of Occludin and ZO-1 proteins in colonic tissues were also measured. Result The model group exhibited significantly higher Longa scores, larger brain infarct size, and higher serum FITC-Dextran levels and ET-1 levels when compared with the sham surgery group (p < 0.01). The model group demonstrated decreased expression of Occludin and ZO-1 in colonic tissues (p < 0.01) and changes in gut microbiota structure. When compared to the model group, the moxibustion group demonstrated significantly lower Longa scores, smaller brain infarct size, and lower serum FITC-Dextran levels and ET-1 levels (p < 0.05). Furthermore, the moxibustion group demonstrated decreased inflammatory cell infiltration in colonic tissues, increased expression of Occludin and ZO-1 proteins in colonic tissues (p < 0.05), enhanced gut microbiota structure, and a decreased Simpson index (p < 0.05). Conclusion Moxibustion can improve the neurological dysfunction in CIS model rats. The mechanism may be associated with the improvement in gut microbiota dysbiosis, reduction in colonic mucosal permeability, and restoration of intestinal mucosal barrier damage.
Collapse
Affiliation(s)
- Yi-Xia Ding
- Department of Encephalopathy (V), The Second Affiliated Hospital of Anhui University of Chinese Medicine (Anhui Acupuncture Hospital), Hefei, Anhui, China
- Institute of Clinical Acupuncture and Moxibustion, Anhui Academy of Chinese Medicine, Hefei, Anhui, China
- Anhui Clinical Medical Research Center of Acupuncture and Moxibustion, Hefei, Anhui, China
| | - Liang-Liang Chen
- Institute of Clinical Acupuncture and Moxibustion, Anhui Academy of Chinese Medicine, Hefei, Anhui, China
- Anhui Clinical Medical Research Center of Acupuncture and Moxibustion, Hefei, Anhui, China
- Department of Spleen and Stomach Diseases, The Second Affiliated Hospital of Anhui University of Chinese Medicine (Anhui Acupuncture Hospital), Hefei, Anhui, China
| | - Kui-Wu Li
- Graduate School of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Ling Zou
- Graduate School of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Lu-Min Liao
- Graduate School of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xiao-Yu Han
- Graduate School of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jie OuYang
- Graduate School of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yue-Ping Wu
- Graduate School of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Wen-Dong Zhang
- Department of Encephalopathy (V), The Second Affiliated Hospital of Anhui University of Chinese Medicine (Anhui Acupuncture Hospital), Hefei, Anhui, China
- Institute of Clinical Acupuncture and Moxibustion, Anhui Academy of Chinese Medicine, Hefei, Anhui, China
- Anhui Clinical Medical Research Center of Acupuncture and Moxibustion, Hefei, Anhui, China
| | - Hao Ran Chu
- Institute of Clinical Acupuncture and Moxibustion, Anhui Academy of Chinese Medicine, Hefei, Anhui, China
- Anhui Clinical Medical Research Center of Acupuncture and Moxibustion, Hefei, Anhui, China
- Outpatient Department, The Second Affiliated Hospital of Anhui University of Chinese Medicine (Anhui Acupuncture Hospital), Hefei, Anhui, China
| |
Collapse
|
18
|
Zhang X, Hu R, Wang Z, Wang J, Yue Z, Wu F, Zhou W, Shah AM. Transcriptomics insights into glutamine on repairing of histamine-induced Yak rumen epithelial cells barrier damage in vitro. BMC Genomics 2025; 26:195. [PMID: 40000997 PMCID: PMC11863408 DOI: 10.1186/s12864-025-11383-6] [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: 05/21/2024] [Accepted: 02/19/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Glutamine (Gln) plays a pivotal role in maintaining the integrity of the rumen epithelial barrier in mammals. This study aimed to investigate the effects of Gln on histamine-induced barrier damage in yak rumen epithelial cells (YRECs). RESULTS RT-qPCR analysis revealed a significant decrease in the mRNA expression of tight junction proteins (ZO-1, JAM-A, Claudin-1, and Claudin-4) following 24-hour exposure to 20 µM histamine (HIS group) (P < 0.05). In the subsequent experiment, YRECs were first treated with 20 µM histamine for 24 h, followed by 8 mM glutamine for 12 h (HG group). Gln treatment reversed the histamine-induced downregulation of both mRNA and protein levels of tight junction proteins and restored the distribution of ZO-1 at the cell membrane. Transcriptome analysis revealed that co-regulated differentially expressed genes were primarily involved in the mitogen-activated protein kinase (MAPK) signaling pathway and apoptosis. These findings were further corroborated by RT-qPCR, Western blot, and flow cytometry analyses. To determine whether glutamine regulates cell barrier function through the p38 MAPK signaling pathway, 20 µM Skatole, a p38 MAPK agonist, was introduced (SK group). The results showed a significant increase in the p-p38/p38 ratio and a marked decrease in the mRNA and protein expression of tight junction proteins in the SK group compared to the HG group (P < 0.05). CONCLUSIONS Glutamine mitigates histamine-induced barrier damage in YRECs through the p38 MAPK signaling pathway and apoptosis regulation.
Collapse
Affiliation(s)
- Xiaohong Zhang
- Low Carbon Breeding Cattle and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agriculture University, Chengdu, 611130, China
| | - Rui Hu
- Low Carbon Breeding Cattle and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agriculture University, Chengdu, 611130, China
| | - Zhisheng Wang
- Low Carbon Breeding Cattle and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agriculture University, Chengdu, 611130, China.
| | - Junmei Wang
- Low Carbon Breeding Cattle and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agriculture University, Chengdu, 611130, China
| | - Ziqi Yue
- Low Carbon Breeding Cattle and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agriculture University, Chengdu, 611130, China
| | - Fali Wu
- Low Carbon Breeding Cattle and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agriculture University, Chengdu, 611130, China
| | - Wenjuan Zhou
- Low Carbon Breeding Cattle and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agriculture University, Chengdu, 611130, China
| | - Ali Mujtaba Shah
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| |
Collapse
|
19
|
Dai TT, Fang W, Zhu WT, Han ZL, Sun NX, Yin G, Wang DL. Atractylenolide III ameliorates DSS-induced colitis by improving intestinal epithelial barrier via suppressing the NF-κB-Mediated MLCK-pMLC signaling pathway. Food Chem Toxicol 2025; 196:115158. [PMID: 39613239 DOI: 10.1016/j.fct.2024.115158] [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: 10/06/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
This study is to demonstrate the protection of atractylenolide III (AT III) on intestinal barrier dysfunction in ulcerative colitis (UC). UC model was established by 3% dextran sulfate sodium (DSS), and TNF-α was used to induce dysfunction in the intestinal epithelial barrier. TEER, FD-4 transmembrane flux and DAI were measured. Histopathological changes was identified by H&E staining, TJ structure changes were observed by TEM, IL-1β and TNF-α contents were measured by ELISA, bacterial translocation was investigated by FISH. The expressions of ZO-1, occludin, and the proteins in the MLCK/p-MLC and NF-κB pathways were analyzed by Western blotting or immunofluorescence. The results indicated that AT III alleviate the symptoms of DSS-induced colitis, reduce the disruption of intestinal epithelial barrier, and decrease FD4. Moreover, AT III inhibited the destruction of intestinal epithelial TJ structure and bacterial translocation in UC mice. AT III reversed the high levels of IL-1β and TNF-α, the decrease of occludin, ZO-1 expressions. Furthermore, AT III showed similar effects to PDTC (pyrrolidinedithiocarbamate) in ameliorating the disruption of the TNF-α-induced TEER and FD-4 disruption, MLCK protein expression, and MLC2 phosphorylation. In conclusion, AT III mitigates the dysfunction of intestinal epithelial barrier in UC through the NF-κB-mediated MLCK/p-MLC signaling pathway.
Collapse
Affiliation(s)
| | - Wei Fang
- Anhui University of Chinese Medicine, Hefei, China
| | - Wen-Tao Zhu
- Anhui University of Chinese Medicine, Hefei, China
| | - Zhi-Li Han
- Anhui University of Chinese Medicine, Hefei, China
| | - Nian-Xia Sun
- Anhui University of Chinese Medicine, Hefei, China
| | - Gang Yin
- Anhui University of Chinese Medicine, Hefei, China
| | - Dian-Lei Wang
- Anhui University of Chinese Medicine, Hefei, China; Bozhou Vocational and Technical College, Bozhou, China.
| |
Collapse
|
20
|
Dow E, Hernandez MI, Johnston CS. Eight weeks of resistance exercise improves mood state and intestinal permeability in healthy adults: A randomized controlled trial. Physiol Rep 2025; 13:e70219. [PMID: 39924706 PMCID: PMC11807843 DOI: 10.14814/phy2.70219] [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: 11/11/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 02/11/2025] Open
Abstract
To explore a potential link between resistance exercise and the gut-brain axis, this study examined the impact of resistance exercise on intestinal permeability, as indicated by lipopolysaccharide binding protein (LBP), and mood state in healthy adults. Sedentary participants (n = 20; 39.5 ± 12.1 y; 27.4 ± 5.3 kg/m2) were randomly assigned to the resistance exercise (REX) or wait-listed control (CON) groups. REX participants strength trained 3× weekly (advancing from 45%-55% to 70%-80% 1RM for 3-4 sets over 8 weeks). Strength testing, evaluation of mood states, and collection of fasting blood occurred at baseline and weeks 4 and 8. At baseline, LBP concentrations were inversely correlated to all strength measures (r range: -0.48 to -0.57; p < 0.05). The gain in total strength [(split squat left + right)/2 + bench press] was 45% higher for REX versus CON participants (p = 0.019), and serum LBP concentrations fell 16% for REX participants and rose 9% in CON participants (p = 0.014). Mood was significantly improved by resistance training versus control (but this improvement was not related to changes in LBP; r = -0.001). These findings support a role for resistance exercise in improving mood state and intestinal barrier function, but more research is warranted to further explore the effects of resistance training on the gut-brain axis.
Collapse
Affiliation(s)
- Emily Dow
- College of Health SolutionsArizona State UniversityPhoenixArizonaUSA
| | | | - Carol S. Johnston
- College of Health SolutionsArizona State UniversityPhoenixArizonaUSA
| |
Collapse
|
21
|
Wang S, Liu X, Xu L, Lang J, Liu D. Phase-dependent iron depletion differentially regulates the niche of intestinal stem cells in experimental colitis via ERK/STAT3 signaling pathway. Front Immunol 2025; 16:1537651. [PMID: 39949764 PMCID: PMC11822217 DOI: 10.3389/fimmu.2025.1537651] [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: 12/03/2024] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Introduction Ulcerative colitis (UC) is a global gastrointestinal disease, which is mainly caused by both dysfunctional epithelial barrier and inflammation response. Iron is a critical fundamental element for both the maintenance of homeostasis and the mediation of inflammation in many tissues. However, the role and mechanism of iron in the phase of enteritis and the subsequent repairing phase of intestinal stem cells has not been elucidated. In this study, we aimed to explore whether and how iron depletion would affect the occurrence and outcome of experimental colitis. Methods Iron depletion was realized by deferoxamine (DFO) at either the early stage or late stage of dextran sulfate sodium (DSS) induced experimental colitis in mice. The gross images of colons, general health, histology, barrier integrity, and qRT-PCR were performed. Meanwhile, cell culture and colonic organoids were used to examine the influence of iron depletion in vitro. Signaling pathway and inflammatory infiltration were investigated by immunostaining. Results Iron depletion within the early stage of DSS treatment significantly inhibited the onset of the inflammatory response, maintained the integrity of the colonic epithelium, and preserved the activity of intestinal stem cells (ISCs) both in vivo and in vitro. However, both continuous iron depletion by DFO and late DFO treatment aggravated colonic injury and postponed the recovery from colitis. Early DFO-induced iron depletion was able to maintain the p-STAT3 and p-ERK1/2 signaling pathways within the colonic epithelium at the early phase of colitis, but late DFO treatment inhibited the activity of these two pathways. Discussion Our study demonstrated that the manipulation of iron depletion by DFO might greatly affect the outcomes of experimental colitis in a phase-dependent manner, which suggests that the balance of iron metabolism might be an effective therapeutic target for the clinical treatment of IBD patients.
Collapse
Affiliation(s)
- Shubin Wang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiangjun Liu
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lu Xu
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinyi Lang
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dengqun Liu
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Experimental Research, Sichuan Cancer Hospital & Institute, Sichuan Provincial Engineering Research Center for Tumor Organoids and Clinical Transformation, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
22
|
Antoine T, Béduneau A, Chrétien C, Cornu R, Bonnefoy F, Moulari B, Perruche S, Pellequer Y. Clinically relevant cell culture model of inflammatory bowel diseases for identification of new therapeutic approaches. Int J Pharm 2025; 669:125062. [PMID: 39653295 DOI: 10.1016/j.ijpharm.2024.125062] [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: 11/08/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024]
Abstract
Inflammatory Bowel Diseases (IDB) are chronic disorders characterized by gut inflammation, mucosal damage, increased epithelial permeability and altered mucus layer. No accurate in vitro model exists to simulate these characteristics. In this context, drug development for IBD or intestinal inflammation requires in vivo evaluations to verify treatments efficacy. A new model with altered mucus layer composition; altered epithelial permeability and pro-inflammatory crosstalk between immune and epithelial cells will be developed to enhance in vitro models for studying IBD treatments. The effects of dextran sulfate sodium and/or lipopolysaccharides on intestinal permeability, cytokines synthesis (IL-6, IL-8, TNF-α and IL-1β), mucins (MUC2, MUC5AC) and tight junction proteins expression (Claudin-1, ZO-1 and Occludin) were investigated in a tri-coculture model combining differentiated Caco-2/HT29-MTX cells and THP-1 cells. Two anti-inflammatory agents were evaluated to assess the model's therapeutic strategy applicability (corticoids and pro-resolving factors). Two in vitro models have been developed. The first model, characterized by increased permeability of the epithelial layer and subsequent secretion of inflammatory cytokines, can reproduce the different phases of inflammation, and enables the evaluation of preventive treatments. The second model simulates the acute phase of inflammation and allows for the assessment of curative treatments. Both models demonstrated reversibility when treated with betamethasone and pro-resolving factors. These in vitro models are valuable for selecting therapeutic agents prior to their application in in vivo models. They enable the assessment of agents' anti-inflammatory effects and their ability to permeate the inflamed epithelial layer and interact with immune cells.
Collapse
Affiliation(s)
- Thomas Antoine
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, LabEx LipSTIC (ANR-11- LABX-0021), F-25000 Besançon, France
| | - Arnaud Béduneau
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, LabEx LipSTIC (ANR-11- LABX-0021), F-25000 Besançon, France
| | - Claire Chrétien
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, LabEx LipSTIC (ANR-11- LABX-0021), F-25000 Besançon, France
| | - Raphaël Cornu
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, LabEx LipSTIC (ANR-11- LABX-0021), F-25000 Besançon, France
| | - Francis Bonnefoy
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, LabEx LipSTIC (ANR-11- LABX-0021), F-25000 Besançon, France; MED'INN'Pharma, F-25000 Besançon, France
| | - Brice Moulari
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, LabEx LipSTIC (ANR-11- LABX-0021), F-25000 Besançon, France
| | - Sylvain Perruche
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, LabEx LipSTIC (ANR-11- LABX-0021), F-25000 Besançon, France; MED'INN'Pharma, F-25000 Besançon, France
| | - Yann Pellequer
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, LabEx LipSTIC (ANR-11- LABX-0021), F-25000 Besançon, France.
| |
Collapse
|
23
|
Ran L, Lei J, Liu H, Wang D, Liu J, Yang F, Chen D. Bacillus pumilus SMU5927 protect mice from damage caused by Salmonella Enteritidis colonization. Life Sci 2025; 361:123291. [PMID: 39631534 DOI: 10.1016/j.lfs.2024.123291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 11/15/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Salmonella Enteritidis is one of the main pathogens of foodborne diseases and an important pathogen causing diarrhea in yaks. Antibiotics are the mainstay of treatment for salmonellosis, but the widespread use of antibiotics has increased Salmonella resistance. Probiotics have been shown to antagonize Salmonella and reduce Salmonella infection. Bacillus pumilus is one of the microbial feed additives approved by the Chinese Ministry of Agriculture for use in animal breeding, which has the effect of improving animal growth performance and immunity, among others. Therefore, this paper explored the anti-infective effect of Bacillus pumilus against Salmonella. RESULTS Bacillus pumilus SMU5927 significantly enhances the intestinal mechanical barrier and reduces the number of Salmonella transferred to the organs. Bacillus pumilus SMU5927 ameliorated intestinal tissue damage and attenuated intestinal inflammatory responses in mice. In addition, Bacillus pumilus increased the ratio of the Firmicutes/Bacteroidetes in the intestinal flora, increased the abundance of beneficial bacteria such as Lactobacillus, and decreased the abundance of harmful bacteria. CONCLUSION This study confirmed the role of Bacillus pumilus SMU5927 in preventing and attenuating Salmonella damage and provided ideas for the development of novel antimicrobial drugs.
Collapse
Affiliation(s)
- Longjun Ran
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Jiangying Lei
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Haifeng Liu
- Department of Veterinary Surgery, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Danni Wang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Jiahao Liu
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Falong Yang
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China
| | - Dechun Chen
- College of Animal and Veterinary Sciences, Southwest Minzu University, Chengdu 610041, China; Key Laboratory of Animal Medicine in Sichuan Province, Southwest Minzu University, Chengdu 610041, China.
| |
Collapse
|
24
|
Liu C, Zeng H, Ouyang J, Wen S, Zhou F, Jiang R, Zhang X, Wang Z, Huang J, Liu Z. Eurotium-Cristatum fermented black tea alleviates ulcerative colitis through the PPARγ-NF-κB signaling axis. Food Res Int 2025; 200:115436. [PMID: 39779091 DOI: 10.1016/j.foodres.2024.115436] [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: 07/09/2024] [Revised: 10/29/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel condition that significantly impairs patient quality of life and remains incurable. Effective dietary management is crucial for both prevention and treatment. This study investigates the effects and mechanisms of Eurotium cristatum-fermented black tea (FBT) in a dextran sulfate sodium (DSS)-induced UC mouse model using transcriptome sequencing, immunofluorescence, and flow cytometry. Our results demonstrate that FBT significantly protects intestinal integrity by suppressing NF-κB-dependent inflammatory genes through the activation of peroxisome proliferator-activated receptor gamma (PPARγ) and modulation of the NF-κB signaling pathway. FBT enhances intestinal barrier integrity by limiting microbial penetration and metabolite translocation into systemic circulation, thereby reducing systemic inflammation risk. Additionally, FBT promotes intestinal mucosa repair and maintains microecological homeostasis by regulating intestinal flora and enhancing the biosynthesis of short-chain fatty acids and amino acids. These findings suggest that FBT has promising prophylactic potential for preventing and alleviating UC by modulating multiple biological pathways, including reducing inflammation, mitigating oxidative stress, and strengthening intestinal barrier integrity. This study lays the groundwork for future research on dietary interventions for UC prevention and management.
Collapse
Affiliation(s)
- Changwei Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan 411201, China
| | - Hongzhe Zeng
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Jian Ouyang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Shuai Wen
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Fang Zhou
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Ronggang Jiang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Xinyi Zhang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Zhong Wang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China
| | - Jianan Huang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China.
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha 410128, China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China; National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
25
|
Yang Z, Khan SA, Walsh LJ, Ziora ZM, Seneviratne CJ. Classical and Modern Models for Biofilm Studies: A Comprehensive Review. Antibiotics (Basel) 2024; 13:1228. [PMID: 39766618 PMCID: PMC11726878 DOI: 10.3390/antibiotics13121228] [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: 10/21/2024] [Revised: 11/30/2024] [Accepted: 12/13/2024] [Indexed: 01/15/2025] Open
Abstract
Biofilms are structured microbial communities that adhere to various abiotic and biotic surfaces, where organisms are encased in an exo-polysaccharide matrix. Organisms within biofilms use various mechanisms that help them resist external challenges, such as antibiotics, rendering them more resistant to drugs. Therefore, researchers have attempted to develop suitable laboratory models to study the physical features of biofilms, their resistance mechanisms against antimicrobial agents, and their gene and protein expression profiles. However, current laboratory models suffer from various limitations. In this comprehensive review, we have summarized the various designs that have been used for laboratory biofilm models, presenting their strengths and limitations. Additionally, we have provided insight into improving these models to more closely simulate real-life scenarios, using newly developed techniques in additive manufacturing, synthetic biology, and bioengineering.
Collapse
Affiliation(s)
- Zhihe Yang
- School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, QLD 4072, Australia;
- Oral Health Centre, School of Dentistry, University of Queensland, Herston, QLD 4006, Australia; (S.A.K.); (L.J.W.)
| | - Sadaf Aiman Khan
- Oral Health Centre, School of Dentistry, University of Queensland, Herston, QLD 4006, Australia; (S.A.K.); (L.J.W.)
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, QLD 4072, Australia;
- Indian Institute of Technology (IITD) Delhi, University of Queensland, New Delhi 110016, India
| | - Laurence J. Walsh
- Oral Health Centre, School of Dentistry, University of Queensland, Herston, QLD 4006, Australia; (S.A.K.); (L.J.W.)
| | - Zyta M. Ziora
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, QLD 4072, Australia;
- Indian Institute of Technology (IITD) Delhi, University of Queensland, New Delhi 110016, India
| | | |
Collapse
|
26
|
Dong D, Wang H, Bi H, Li Y, Gao T, Feng J, Li G, Guo S, Yuan H, Ni W. A pectic polysaccharide from Lycium ruthenicum Murray alleviates dextran sulfate sodium-induced colitis in mice. Curr Res Food Sci 2024; 10:100955. [PMID: 39807359 PMCID: PMC11728900 DOI: 10.1016/j.crfs.2024.100955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Inflammatory bowel disorders (IBD) can lead to severe complications like perforation, bleeding, and colon cancer, posing life-threatening risks. Lycium ruthenicum Murray (L. ruthenicum Murr.), rich in polysaccharides, has been utilized in traditional diets for thousands of years. This study explores the protective effects of the polysaccharide of L. ruthenicum on mice with dextran sulfate sodium (DSS)-induced colitis. In the present study, a pectic polysaccharide (LRWP-Ap) containing arabinogalactan (AG) and homogalacturonic acid (HG) structural domains with a Mw of 4.34 kDa was obtained from L. ruthenicum Murr. Fruit. The gavage administration of LRWP-Ap significantly alleviated symptoms of DSS-induced colitis in mice. In this process, LRWP-Ap modulated the balance of Arg-1/iNOS to regulate the metabolism of arginine, and the levels of intestinal tight junction (TJ) (ZO-1, Occludin, and Claudin 1) were increased by LRWP-Ap treatment, which promoted intestinal barrier function. In addition, LRWP-Ap alleviated the inflammatory response while increasing the anti-inflammatory response by reducing the level of proinflammatory factors, enhancing the level of anti-inflammatory factors (IL-10) and improving the balance of Treg/Th17 cells. These effects resulted in the maintenance of intestinal immune homeostasis. Moreover, LRWP-Ap modulated the gut microbiota composition and short-chain fatty acid (SCFA) content, which may maintain relatively favorable intestinal homeostasis. In general, LRWP-Ap has the potential to alleviate IBD, and the use of L. ruthenicum Murr. As a natural functional food to improve gut health in the context of DSS-induced colitis.
Collapse
Affiliation(s)
- Dai Dong
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xin min Street, Changchun, 130021, China
| | - Hailiang Wang
- The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130033, China
| | - Hongtao Bi
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, CAS, 23 Xinning Road, Xining, 810008, China
| | - Yu Li
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xin min Street, Changchun, 130021, China
| | - Tingting Gao
- School of Psychology, Chengdu Medical College, 783 Xindu Road, Chengdu, 610500, China
| | - Jingyue Feng
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xin min Street, Changchun, 130021, China
| | - Guoqiang Li
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, CAS, 23 Xinning Road, Xining, 810008, China
| | - Shiqi Guo
- The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130033, China
| | - Hongyan Yuan
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xin min Street, Changchun, 130021, China
| | - Weihua Ni
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xin min Street, Changchun, 130021, China
| |
Collapse
|
27
|
Ni W, Li Y, Feng J, Liu B, Yuan H, Tai G, Bi H. Therapeutic Efficacy and Underlying Mechanisms of a Mannoglucan from Hirsutella sinensis Mycelium on Dextran Sulfate Sodium-Induced Inflammatory Bowel Disease in Mice: Modulation of the Intestinal Barrier, Oxidative Stress and Gut Microbiota. Int J Mol Sci 2024; 25:13100. [PMID: 39684811 DOI: 10.3390/ijms252313100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Hirsutella sinensis (H. sinensis), a non-sexual form of the valuable Chinese medicinal herb, demonstrates various biological activities, such as immune modulation and antioxidative capabilities. Nonetheless, the effects of bioactive polysaccharides derived from H. sinensis on colitis have yet to be investigated. In our prior research, we extracted a mannoglucan (HSWP-1d) from H. sinensis and found that it attenuates TGF-β1-induced epithelial-mesenchymal transition. The present study investigated the protective effects of HSWP-1d against colitis induced by dextran sulfate sodium (DSS) in mice. The results demonstrate that HSWP-1d effectively ameliorates symptoms of colitis and preserves the intestinal barrier's stability by enhancing the expression of tight junction proteins. The administration of HSWP-1d results in a reduction in oxidative stress through the augmentation of antioxidative enzyme activities, concomitant with the suppression of oxidative product generation. Simultaneously, HSWP-1d reduced the levels of pro-inflammatory cytokines while elevating the levels of anti-inflammatory cytokines, effectively mitigating the inflammatory response. Furthermore, HSWP-1d influences and alters short-chain-fatty-acid (SCFA) levels, thereby enhancing the intestinal microenvironment. In conclusion, HSWP-1d contributes to intestinal well-being and holds potential as both a therapeutic choice and a supplier of essential nutrients for the amelioration of colitis.
Collapse
Affiliation(s)
- Weihua Ni
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Yu Li
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Jingyue Feng
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Boxuan Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Hongyan Yuan
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Guixiang Tai
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
| | - Hongtao Bi
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130021, China
- Qinghai Provincial Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, CAS, 23 Xinning Road, Xining 810008, China
| |
Collapse
|
28
|
Mach N. The forecasting power of the mucin-microbiome interplay in livestock respiratory diseases. Vet Q 2024; 44:1-18. [PMID: 38606662 PMCID: PMC11018052 DOI: 10.1080/01652176.2024.2340003] [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: 05/23/2023] [Accepted: 03/31/2024] [Indexed: 04/13/2024] Open
Abstract
Complex respiratory diseases are a significant challenge for the livestock industry worldwide. These diseases considerably impact animal health and welfare and cause severe economic losses. One of the first lines of pathogen defense combines the respiratory tract mucus, a highly viscous material primarily composed of mucins, and a thriving multi-kingdom microbial ecosystem. The microbiome-mucin interplay protects from unwanted substances and organisms, but its dysfunction may enable pathogenic infections and the onset of respiratory disease. Emerging evidence also shows that noncoding regulatory RNAs might modulate the structure and function of the microbiome-mucin relationship. This opinion paper unearths the current understanding of the triangular relationship between mucins, the microbiome, and noncoding RNAs in the context of respiratory infections in animals of veterinary interest. There is a need to look at these molecular underpinnings that dictate distinct health and disease outcomes to implement effective prevention, surveillance, and timely intervention strategies tailored to the different epidemiological contexts.
Collapse
Affiliation(s)
- Núria Mach
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| |
Collapse
|
29
|
Miao S, Li J, Chen Y, Zhao W, Xu M, Liu F, Zou X, Dong X. Targeting gut microbiota and metabolism profiles with coated sodium butyrate to ameliorate high-energy and low-protein diet-induced intestinal barrier dysfunction in laying hens. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 19:104-116. [PMID: 39635416 PMCID: PMC11615920 DOI: 10.1016/j.aninu.2024.06.006] [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: 03/30/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 12/07/2024]
Abstract
High energy diets are a risk factor for intestinal barrier damage. Butyrate, a major energy source for intestinal epithelial cells, has been shown to improve barrier dysfunction and modulate the gut microbiota. In this trial, we examined the preventative effect of coated sodium butyrate (CSB) on high-energy and low-protein (HELP)-induced intestinal barrier injury in laying hens, and also worked to determine the underlying mechanisms by an integrative analysis of gut microbiota and the metabolome. A total of 216 healthy 28-week-old Huafeng laying hens were randomly assigned to 3 groups with 6 replicates each: the CON group (normal diet), HELP group (HELP diet) and CH500 group (500 mg/kg CSB added to HELP diet). The duration of the trial encompassed a period of 10 weeks. The results revealed that CSB treatment improved the laying rate and mitigated the detrimental effects on intestinal barrier function and the inflammatory response induced by the HELP diet in laying hens (P < 0.05). Microbial profiling analysis revealed that the CSB treatment reshaped the HELP-perturbed gut microbiota and promoted the growth of beneficial bacteria (P < 0.05). Untargeted metabolomics analysis revealed that CSB reduced the metabolites associated with intestinal inflammation (P < 0.05). In conclusion, CSB did not merely modulate alterations in the gut microbiota composition and microbial metabolites but also yielded increased egg production, while mitigating intestinal barrier dysfunction and inflammatory responses induced by HELP in laying hens.
Collapse
Affiliation(s)
- Sasa Miao
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiankui Li
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Chen
- Hangzhou Zhejiang University Animal Hospital Co., Ltd., Hangzhou 310058, China
| | - Wenyan Zhao
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mengru Xu
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fang Liu
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoting Zou
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyang Dong
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, The Key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
30
|
Chen K, Wang H, Yang X, Tang C, Hu G, Gao Z. Targeting gut microbiota as a therapeutic target in T2DM: A review of multi-target interactions of probiotics, prebiotics, postbiotics, and synbiotics with the intestinal barrier. Pharmacol Res 2024; 210:107483. [PMID: 39521027 DOI: 10.1016/j.phrs.2024.107483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
The global epidemic of type 2 diabetes mellitus (T2DM) imposes a substantial burden on public health and healthcare expenditures, thereby driving the pursuit of cost-effective preventive and therapeutic strategies. Emerging evidence suggests a potential association between dysbiosis of gut microbiota and its metabolites with T2DM, indicating that targeted interventions aimed at modulating gut microbiota may represent a promising therapeutic approach for the management of T2DM. In this review, we concentrated on the multifaceted interactions between the gut microbiota and the intestinal barrier in the context of T2DM. We systematically summarized that the imbalance of beneficial gut microbiota and its metabolites may constitute a viable therapeutic approach for the management of T2DM. Meanwhile, the mechanisms by which gut microbiota interventions, such as probiotics, prebiotics, postbiotics, and synbiotics, synergistically improve insulin resistance in T2DM are summarized. These mechanisms include the restoration of gut microbiota structure, upregulation of intestinal epithelial cell proliferation and differentiation, enhancement of tight junction protein expression, promotion of mucin secretion by goblet cells, and the immunosuppressive functions of regulatory T cells (Treg) and M2 macrophages. Collectively, these actions contribute to the amelioration of the body's metabolic inflammatory status. Our objective is to furnish evidence that supports the clinical application of probiotics, prebiotics, and postbiotics in the management of T2DM.
Collapse
Affiliation(s)
- Keyu Chen
- Institute of Metabolic Diseases, Guang' anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Department of Endocrinology, 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
| | - Xiaofei Yang
- Beijing University of Chinese Medicine, 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.
| | - Guojie Hu
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Zezheng Gao
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
31
|
Arrari F, Ortiz-Flores RM, Lhamyani S, Garcia-Fuentes E, Jabri MA, Sebai H, Bermudez-Silva FJ. Protective Effects of Spirulina Against Lipid Micelles and Lipopolysaccharide-Induced Intestinal Epithelium Disruption in Caco-2 Cells: In Silico Molecular Docking Analysis of Phycocyanobilin. Nutrients 2024; 16:4074. [PMID: 39683467 DOI: 10.3390/nu16234074] [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: 10/25/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Damage to intestinal epithelial cells is present in obesity and other diseases because of inflammatory and oxidative processes. This damage compromises the gastrointestinal barrier, killing enterocytes, altering intestinal permeability, and eliciting abnormal immune responses that promote chronic inflammation. Recent evidence shows that spirulina is a potent natural agent with antioxidant and anti-inflammatory properties. OBJECTIVES This study was conducted to evaluate the effect of spirulina aqueous extract (SPAE) on the alterations of the intestinal epithelium induced by lipid micelles (LMs) and/or inflammation induced by lipopolysaccharides (LPSs) in the Caco-2 cell line. METHODS In the current research, we assessed the protective actions of SPAE against cytotoxicity, oxidative stress, inflammation, and epithelial barrier perturbation by using an in vitro model, the intestinal Caco-2 cells, treated with LPSs and/or LMs. We also performed an in silico molecular docking analysis with spirulina's bioactive compound, phycocyanobilin. RESULTS Our results showed that SPAE has no cytotoxic effect on Caco-2 cells. On the contrary, it improved cell viability and exhibited anti-inflammatory and antioxidant actions. SPAE also protected against endoplasmic reticulum stress and tight junction proteins, thus improving the epithelial barrier. The in silico study revealed a strong binding affinity of the phycocyanobilin compound with human SOD and NADPH oxidase and a good binding affinity towards COX-2 and iNOS. CONCLUSIONS Taken together, these findings demonstrate the beneficial actions of SPAE on Caco-2 cells, suggesting it may be useful in preserving the epithelial intestinal barrier in human conditions involving oxidative stress and inflammation such as obesity.
Collapse
Affiliation(s)
- Fatma Arrari
- Laboratory of Functional Physiology and Valorization of Bio-Resources, Higher Institute of Biotechnology of Beja, University of Jendouba, Beja 9000, Tunisia
| | - Rodolfo-Matias Ortiz-Flores
- Centro de Investigacion Biomedica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERDEM), Instituto de Investigacion Biomedica de Malaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Regional Universitario de Malaga, UGC Endocrinología y Nutricion, 29009 Malaga, Spain
| | - Said Lhamyani
- Centro de Investigacion Biomedica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERDEM), Instituto de Investigacion Biomedica de Malaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Regional Universitario de Malaga, UGC Endocrinología y Nutricion, 29009 Malaga, Spain
| | - Eduardo Garcia-Fuentes
- Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas (CIBERehd), Instituto de Investigacion Biomedica de Malaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Universitario Virgen de la Victoria, UGC de Aparato Digestivo, 29010 Malaga, Spain
| | - Mohamed-Amine Jabri
- Laboratory of Functional Physiology and Valorization of Bio-Resources, Higher Institute of Biotechnology of Beja, University of Jendouba, Beja 9000, Tunisia
| | - Hichem Sebai
- Laboratory of Functional Physiology and Valorization of Bio-Resources, Higher Institute of Biotechnology of Beja, University of Jendouba, Beja 9000, Tunisia
| | - Francisco-Javier Bermudez-Silva
- Centro de Investigacion Biomedica en Red de Diabetes y Enfermedades Metabolicas Asociadas (CIBERDEM), Instituto de Investigacion Biomedica de Malaga y Plataforma en Nanomedicina-IBIMA Plataforma BIONAND, Hospital Regional Universitario de Malaga, UGC Endocrinología y Nutricion, 29009 Malaga, Spain
| |
Collapse
|
32
|
Sivri D, Şeref B, Şare Bulut M, Gezmen Karadağ M. Evaluation of the Effect of Probiotic Supplementation on Intestinal Barrier Integrity and Epithelial Damage in Colitis Disease: A Systematic Review. Nutr Rev 2024:nuae180. [PMID: 39602817 DOI: 10.1093/nutrit/nuae180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
CONTEXT Previous reviews have focused on the effects of probiotics on colitis, but there is a need to understand their impact on barrier integrity and tight junction protein improvement in colitis. OBJECTIVE This study aimed to systematically examine the effects of probiotic use on barrier integrity in colitis disease. This study was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. DATA SOURCES A systematic search in PubMed, Web of Science, Scopus, and Cochrane databases identified 2537 articles. DATA EXTRACTION As a result of the search, 2537 articles were accessed. Study results were summarized descriptively through discussions by intervention conditions, study population, measurement methods, and key findings. The included studies were independently reviewed and all authors reached consensus on the quality and major findings from the included articles. Forty-six studies that met the inclusion criteria were analyzed within the scope of the systematic review. RESULTS Although the study primarily utilized probiotics from the Lactobacillaceae family (notably, L casei, L reuteri, L rhamnosus, L plantarum, and L pentosus) and the Bifidobacteriaceae family (notably, B breve, B animalis, and B dentium), other probiotics also demonstrated positive effects on tight junction proteins. These effects are attributed to the production of bioactive and metabolic compounds, as well as short-chain fatty acids, which combat pathogens and reduce anti-inflammatory agents. However, it was observed that the effects of these probiotics on tight junction proteins varied depending on the strain and dose. CONCLUSION The beneficial effects of probiotics on remission in inflammatory bowel disease are well documented. Studies show that probiotics generally improve intestinal barrier function, but factors such as dose, duration, and bacterial species combinations need further clarification. Additionally, comprehensive studies are needed to understand how improved barrier function affects absorption in individuals. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42023452774.
Collapse
Affiliation(s)
- Dilek Sivri
- Department of Nutrition and Dietetics, Anadolu University, Eskişehir, Türkiye
| | - Betül Şeref
- Department of Nutrition and Dietetics, Karamanoğlu Mehmetbey University, Karaman, Türkiye
| | - Melike Şare Bulut
- Department of Nutrition and Dietetics, Biruni University, Istanbul, Türkiye
| | | |
Collapse
|
33
|
Miao J, Cui L, Zeng H, Hou M, Wang J, Hang S. Lactiplantibacillus plantarum L47 and inulin affect colon and liver inflammation in piglets challenged by enterotoxigenic Escherichia coli through regulating gut microbiota. Front Vet Sci 2024; 11:1496893. [PMID: 39664894 PMCID: PMC11631943 DOI: 10.3389/fvets.2024.1496893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 11/07/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction Infection by pathogenic bacteria during weaning is a common cause of diarrhea and intestinal inflammation in piglets. Supplementing the diet with synbiotics is beneficial for animal health. The strain of Lactiplantibacillus plantarum L47 (L47) isolated in our lab exhibited good probiotic properties when combined with inulin. Here, the effectiveness of combining L47 and inulin (CLN) in protecting against enterotoxigenic Escherichia coli (ETEC) induced colon and liver inflammation in weaned piglets was evaluated. Methods Twenty-eight piglets aged 21 days were randomly assigned into 4 groups: CON (control), LI47 (oral CLN culture fluid, 1010 CFU/d of L47 and 1 g/d of inulin), ECON (oral ETEC culture fluid, 1010 CFU/d), and ELI47 (oral CLN and ETEC culture fluid). After 24 days, the colon and liver samples were collected for further analysis. Results and discussion CLN alleviated colon damage caused by ETEC challenge, as evidenced by an increase of colonic crypt depth, mRNA expression of tight junction Claudin-1 and Occludin, GPX activity, the concentration of IL-10 and sIgA (p < 0.05). Moreover, there was a decrease in MDA activity, the load of E. coli, the concentration of LPS, gene expression of TLR4, and the concentration of TNF-α and IL-6 (p < 0.05) in colonic mucosa. Additionally, CLN counteracted liver damage caused by ETEC challenge by modulating pathways associated with immunity and disease occurrence (p < 0.05). Conclusion Supplementing with CLN alleviated colon inflammation induced by ETEC challenge by decreasing the E. coli/LPS/TLR4 pathway and regulating hepatic immune response and disease-related pathways, suggesting that CLN could protect intestinal and liver health in animals.
Collapse
Affiliation(s)
| | | | | | | | | | - Suqin Hang
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
34
|
Blais A, Takakura N, Grauso M, Puel-Artero C, Blachier F, Lan A. Dietary Bovine Lactoferrin Reduces the Deleterious Effects of Lipopolysaccharide Injection on Mice Intestine. Nutrients 2024; 16:4040. [PMID: 39683434 DOI: 10.3390/nu16234040] [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: 10/11/2024] [Revised: 10/31/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Injection of lipopolysaccharides (LPS) in experimental models induces a systemic inflammatory response that is associated with deleterious effects on intestinal morphology and physiology. In this study, we have studied in female mice the effects of dietary supplementation with bovine lactoferrin (bLF) given before intraperitoneal injection of LPS on jejunum and colon. METHODS The first study evaluated the efficiency of different bLF and LPS concentrations to determine the optimal experimental conditions. For the second study mice were fed with 1% bLF before the LPS challenge (3 mg/kg body weight). Plasmatic markers of inflammation, intestinal morphology, permeability, and expression of genes related to epithelial differentiation, epithelial barrier function and intestinal inflammation in both small intestine and colon were evaluated. RESULTS bLF ingestion before the LPS challenge reduced the TNF-α circulating concentration, compared to control animals. This decrease in plasma TNF-α was correlated with improved intestinal permeability. The morphology of jejunal epithelium, which was affected by LPS challenge, was partly maintained by bLF. Measurement of the expression of genes encoding proteins involved in epithelial differentiation, intestinal inflammation, and epithelial barrier function suggests an overall protective effect of bLF against the adverse effects of LPS in the jejunum. In the colon, the effects of bLF ingestion on the subsequent LPS challenge, although protective, remain different when compared with those observed on jejunum. CONCLUSIONS Taken together, our data indicate that bLF dietary supplementation does have a protective effect on the deleterious intestinal alterations induced by LPS systemic inflammation.
Collapse
Affiliation(s)
- Anne Blais
- UMR-PNCA, Université Paris-Saclay, AgroParisTech, INRAE, 91120 Palaiseau, France
| | - Natsuko Takakura
- UMR-PNCA, Université Paris-Saclay, AgroParisTech, INRAE, 91120 Palaiseau, France
| | - Marta Grauso
- UMR-PNCA, Université Paris-Saclay, AgroParisTech, INRAE, 91120 Palaiseau, France
| | | | - François Blachier
- UMR-PNCA, Université Paris-Saclay, AgroParisTech, INRAE, 91120 Palaiseau, France
| | - Annaïg Lan
- UMR-PNCA, Université Paris-Saclay, AgroParisTech, INRAE, 91120 Palaiseau, France
| |
Collapse
|
35
|
Wang X, Yang Z, Zhang W, Xing L, Luo R, Cao S. Obstacles, research progress, and prospects of oral delivery of bioactive peptides: a comprehensive review. Front Nutr 2024; 11:1496706. [PMID: 39610876 PMCID: PMC11602335 DOI: 10.3389/fnut.2024.1496706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024] Open
Abstract
Bioactive peptides hold significant potential for enhancing human health, however, their limited oral bioavailability poses a substantial barrier to their widespread use in the food and pharmaceutical industries. This article reviews the key factors influencing the absorption efficiency of oral bioactive peptides, including issues related to bitter taste perception, challenges in gastrointestinal environmental stability, and limitations in transmembrane transport. Furthermore, it highlights the latest technologies, such as osmotic technology, chemical modification, and advanced delivery systems, and discusses their advantages in enhancing the stability of bioactive peptides and facilitating intestinal absorption. In addition, the application and challenges of common delivery systems such as liposomes, emulsions, polymer nanoparticles, and hydrogels in oral bioactive peptide delivery are also discussed. This paper aims to provide a theoretical foundation for scientific research and practical applications of oral delivery of bioactive peptides, thereby promoting the further development of bioactive peptides in the context of human health.
Collapse
Affiliation(s)
- Xinyu Wang
- School of Food Science and Engineering, Ningxia University, Yinchuan, China
| | - Zeyao Yang
- School of Food Science and Engineering, Ningxia University, Yinchuan, China
| | - Wangang Zhang
- Key Lab of Meat Processing and Quality Control, MOE, School of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Lujuan Xing
- Key Lab of Meat Processing and Quality Control, MOE, School of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Ruiming Luo
- School of Food Science and Engineering, Ningxia University, Yinchuan, China
| | - Songmin Cao
- School of Food Science and Engineering, Ningxia University, Yinchuan, China
| |
Collapse
|
36
|
Zhang Y, Xun L, Qiao R, Jin S, Zhang B, Luo M, Wan P, Zuo Z, Song Z, Qi J. Advances in research on the role of high carbohydrate diet in the process of inflammatory bowel disease (IBD). Front Immunol 2024; 15:1478374. [PMID: 39588368 PMCID: PMC11586370 DOI: 10.3389/fimmu.2024.1478374] [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: 08/12/2024] [Accepted: 10/25/2024] [Indexed: 11/27/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, systemic gastrointestinal disorder characterized by episodic inflammation that requires life-long management. Although the etiology of IBD is not fully understood, it is hypothesized to involve a multifaceted interplay among genetic susceptibility, the host immune response, and environmental factors. Previous studies have largely concluded that IBD is associated with this complex interplay; however, more recent evidence underscores the significant role of dietary habits as risk factors for the development of IBD. In this review, we review the molecular mechanisms of high-sugar and high-fat diets in the progression of IBD and specifically address the impacts of these diets on the gut microbiome, immune system regulation, and integrity of the intestinal barrier, thereby highlighting their roles in the pathogenesis and exacerbation of IBD.
Collapse
Affiliation(s)
- Ying Zhang
- School of Medicine, Kunming University of Science and Technology, Kunming, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Linting Xun
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Ran Qiao
- Colleges of Letters and Science, University of Wisconsin–Madison, Madison, WI, United States
| | - Shumei Jin
- Yunnan Institute of Food and Drug Supervision and Control, Medical Products Administration of Yunnan Province, Kunming, China
| | - Bing Zhang
- Yunnan Provincial Key Laboratory of Modern Information Optics, Kunming University of Science and Technology, Kunming, China
| | - Mei Luo
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Ping Wan
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Kunming, China
- Yunnan Clinical Research Center for Geriatric Disorders, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Zan Zuo
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Zhengji Song
- School of Medicine, Kunming University of Science and Technology, Kunming, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Kunming, China
| | - Jialong Qi
- School of Medicine, Kunming University of Science and Technology, Kunming, China
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People’s Hospital of Yunnan Province, Kunming, China
- Yunnan Clinical Research Center for Geriatric Disorders, The First People’s Hospital of Yunnan Province, Kunming, China
- Yunnan Provincial Key Laboratory of Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
37
|
Jung N, Schreiner J, Baur F, Vogel-Kindgen S, Windbergs M. Predicting nanocarrier permeation across the human intestine in vitro: model matters. Biomater Sci 2024; 12:5775-5788. [PMID: 39402906 DOI: 10.1039/d4bm01092b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
For clinical translation of oral nanocarriers, simulation of the intestinal microenvironment during in vitro testing is crucial to evaluate interactions with the intestinal mucosa. However, studies are often conducted using simplistic cell culture models, overlooking key physiological factors, and potentially leading to an overestimation of nanocarrier permeation. In this study, we systematically investigate different tissue models of the human intestine under static cultivation and dynamic flow conditions and analyze the impact of altered tissue characteristics on nanocarrier permeation. Our results reveal that the selection of cell types as well as the respective culture condition have a notable impact on the physiological characteristics of the resulting tissues. Tissue layer thickness, mucus secretion, and barrier impairment, all increase with increasing amounts of goblet cells and the application of dynamic flow conditions. Permeation studies with poly(lactic-co-glycolic acid) (PLGA) nanocarriers with and without polyethylene glycol (PEG) coating elucidate that the amount of mucus present in the respective model is the limiting factor for the permeation of PLGA nanocarriers, while tissue topography presents the key factor influencing PEG-PLGA nanocarrier permeation. Furthermore, both nanocarriers exhibit diametrically opposite permeation kinetics compared to soluble compounds. In summary, these findings reveal the critical role of the implemented test systems on permeation assessment and emphasize that, in the context of preclinical nanocarrier testing, the choice of in vitro model matters.
Collapse
Affiliation(s)
- Nathalie Jung
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Jonas Schreiner
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Florentin Baur
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Sarah Vogel-Kindgen
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| | - Maike Windbergs
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
38
|
Mitra D, Armijo GK, Ober EH, Baker SM, Turner HC, Broustas CG. MIIST305 mitigates gastrointestinal acute radiation syndrome injury and ameliorates radiation-induced gut microbiome dysbiosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619652. [PMID: 39484519 PMCID: PMC11526895 DOI: 10.1101/2024.10.22.619652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
High-dose radiation exposure results in gastrointestinal (GI) acute radiation syndrome identified by the destruction of mucosal layer, intestinal epithelial barrier dysfunction, and aberrant inflammatory responses. In addition, radiation causes gut microbiome dysbiosis characterized by diminished microbial diversity, reduction in the abundance of beneficial commensal bacteria, and the spread of bacterial pathogens that trigger the recruitment of immune cells and the production of pro-inflammatory factors that lead to further GI tissue damage. Currently, there are no FDA-approved countermeasures that can treat radiation-induced GI injury. To meet this critical need, Synedgen Inc., has developed a glycopolymer radiomitigator (MIIST305) that is specifically targeted to the GI tract which acts by intercalating into the mucus layer and the glycocalyx of intestinal epithelial cells that could potentially ameliorate the deleterious effects of radiation. Male C57BL/6J adult mice were exposed to 13 Gy total body X-irradiation with 5% bone marrow shielding and MIIST305 was administered on days 1, 3, and 5 post-irradiation. Approximately 85% of the animals survived the irradiation exposure and were apparently healthy until the end of the 30-day study period. In contrast, no control, vehicle-treated animals survived past day 10 at this radiation dose. We show that MIIST305 improved intestinal epithelial barrier function and suppressed systemic inflammatory response mediated by radiation-induced pro-inflammatory cytokines. Taxonomic profiling and community structure of the fecal and colonic mucosa microbiota demonstrated that MIIST305 treatment increased microbial diversity and restored abundance of beneficial commensal bacteria, including Lactobacillus and Bifidobacterium genera, while suppressing potentially pathogenic bacteria compared with vehicle-treated animals. In summary, MIIST305 is a novel GI-targeted therapeutic that greatly enhances survival in mice exposed to lethal radiation and protects the GI tract from injury by restoring a balanced gut microbiota and effectively reducing proinflammatory responses. Further development of this drug as an FDA-approved medical countermeasure will be of critical importance in the event of a radiation public health emergency.
Collapse
Affiliation(s)
- Debmalya Mitra
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabriel K. Armijo
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth H. Ober
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Helen C. Turner
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Constantinos G. Broustas
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
39
|
Kellermann L, Hansen SL, Maciag G, Granau AM, Johansen JV, Teves JM, Bressan RB, Pedersen MT, Soendergaard C, Baattrup AM, Hammerhøj A, Riis LB, Gubatan J, Jensen KB, Nielsen OH. Influence of Vitamin D Receptor Signalling and Vitamin D on Colonic Epithelial Cell Fate Decisions in Ulcerative Colitis. J Crohns Colitis 2024; 18:1672-1689. [PMID: 38747639 PMCID: PMC11479711 DOI: 10.1093/ecco-jcc/jjae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 04/18/2024] [Accepted: 05/14/2024] [Indexed: 10/17/2024]
Abstract
BACKGROUND AND AIMS Epidemiological studies have shown that subnormal levels of vitamin D (25[OH]D) are associated with a more aggravated clinical course of ulcerative colitis [UC]. Despite an increased focus on the therapeutic importance of vitamin D and vitamin D receptor [VDR] signalling, the mechanisms underlying the effects of the vitamin D-VDR axis on UC remain elusive. Therefore, we aimed to investigate whether exposure to active vitamin D (1,25[OH]2D3/VDR) signalling in human organoids could influence the maintenance of the colonic epithelium. METHODS Intestinal VDR expression was studied by immunohistochemistry, RNA expression arrays, and single-cell RNA sequencing of colonic biopsy specimens obtained from patients with UC and healthy individuals. To characterise the functional and transcriptional effects of 1,25[OH]2D3, we used patient-derived colonic organoids. The dependency of VDR was assessed by knocking out the receptor with CRISPR/Cas9. RESULTS Our results suggest that 1,25[OH]2D3/VDR stimulation supports differentiation of the colonic epithelium and that impaired 1,25[OH]2D3/VDR signalling thereby may compromise the structure of the intestinal epithelial barrier, leading to flares of UC. Furthermore, a transcriptional response to VDR activity was observed primarily in fully differentiated cells at the top of the colonic crypt, and this response was reduced during flares of UC. CONCLUSIONS We identified an important role of vitamin D signalling in supporting differentiated cell states in the human colonic epithelium, and thereby maintenance of the intestinal barrier integrity. This makes the vitamin D-VDR signalling axis an interesting target for therapeutic efforts to achieve and maintain remission in patients with UC.
Collapse
Affiliation(s)
- Lauge Kellermann
- Department of Gastroenterology, Herlev Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Herlev, Denmark
| | - Stine Lind Hansen
- Novo Nordisk Foundation Center for Stem Cell Medicine [reNEW], Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Grzegorz Maciag
- Novo Nordisk Foundation Center for Stem Cell Medicine [reNEW], Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Agnete Marie Granau
- Department of Gastroenterology, Herlev Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Herlev, Denmark
| | | | - Joji Marie Teves
- Novo Nordisk Foundation Center for Stem Cell Medicine [reNEW], Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen N, Denmark
| | - Raul Bardini Bressan
- Novo Nordisk Foundation Center for Stem Cell Medicine [reNEW], Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | | | - Christoffer Soendergaard
- Department of Gastroenterology, Herlev Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Herlev, Denmark
| | - Astrid Moeller Baattrup
- Novo Nordisk Foundation Center for Stem Cell Medicine [reNEW], Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Alexander Hammerhøj
- Department of Gastroenterology, Herlev Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Herlev, Denmark
| | - Lene Buhl Riis
- Department of Pathology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - John Gubatan
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kim Bak Jensen
- Novo Nordisk Foundation Center for Stem Cell Medicine [reNEW], Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Ole Haagen Nielsen
- Department of Gastroenterology, Herlev Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, Herlev, Denmark
| |
Collapse
|
40
|
Lacy BE, Cangemi DJ. Opioids and the Gastrointestinal Tract: The Role of Peripherally Active µ-Opioid Receptor Antagonists in Modulating Intestinal Permeability. Am J Gastroenterol 2024; 119:1970-1978. [PMID: 38870087 PMCID: PMC11446513 DOI: 10.14309/ajg.0000000000002887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
Opioid receptors are found throughout the gastrointestinal tract, including the large intestine. Many patients treated with opioids experience opioid-induced constipation (OIC). Laxatives are not effective in most patients, and in those who do initially respond, the efficacy of laxatives generally diminishes over time. In addition, OIC does not spontaneously resolve for most patients. However, complications of opioids extend far beyond simply slowing gastrointestinal transit. Opioid use can affect intestinal permeability through a variety of mechanisms. Toll-like receptors are a crucial component of innate immunity and are tightly regulated within the gut epithelium. Pathologic µ-opioid receptor (MOR) and toll-like receptor signaling, resulting from chronic opioid exposure, disrupts intestinal permeability leading to potentially harmful bacterial translocation, elevated levels of bacterial toxins, immune activation, and increased cytokine production. Peripherally active MOR antagonists, including methylnaltrexone, are effective at treating OIC. Benefits extend beyond simply blocking the MOR; these agents also act to ameliorate opioid-induced disrupted intestinal permeability. In this review, we briefly describe the physiology of the gastrointestinal epithelial border and discuss the impact of opioids on gastrointestinal function. Finally, we consider the use of peripherally active MOR antagonists to treat disrupted intestinal permeability resulting from opioid use and discuss the potential for improved morbidity and mortality in patients treated with methylnaltrexone for opioid-induced bowel disorders.
Collapse
|
41
|
Moya AMTM, Alexandrino TD, Morari J, Reguengo LM, Velloso LA, Leal RF, Junior SB, Pereira APA, Pastore GM, Bicas JL, Cazarin CBB. The Consumption of the Fibrous Fraction of Solanum lycocarpum St. Hil. Does Not Preserve the Intestinal Mucosa in TNBS-Induced Rats. Foods 2024; 13:2949. [PMID: 39335878 PMCID: PMC11431493 DOI: 10.3390/foods13182949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Solanum lycocarpum St. Hil. is considered a natural anti-inflammatory. In traditional medicine, it is used to reduce cholesterol levels in the treatment of obesity. Foods capable of conferring a protective and nutritious effect have been used to prevent or attenuate the clinical symptoms of inflammatory bowel diseases. Ulcerative colitis is a multifactorial inflammatory bowel disease. This study investigated the impact of the consumption of the fibrous fraction (FF) and resistant starch (RS) of fruta-do-lobo in an experimental model of colitis induced with the use 2,4,6-trinitrobenzene sulphonic acid (TNBS) in rats. The different colitis groups all experienced decreased weight gain, which could be linked to the inflammatory process (p = 0.603). Additionally, the experimental model led to increased oxidative stress, higher levels of pro-inflammatory cytokines, and the elevated gene expression of these cytokines. Despite this, consuming the fibrous fraction of fruta-do-lobo (RS and FF) did not appear to protect the animals against the inflammatory process. Regarding the expression of TNF-α, only the group treated with the drug mesalamine had a reduced serum level of this inflammatory marker (p = 0.03). Our results showed that the diet containing RS and FF did not protect the intestinal mucosa against TNBS inflammation. New studies on the variation in the time of consumption or the supplemented dose of fruta-do-lobo fibers could help to elucidate their effects in protecting the mucosa.
Collapse
Affiliation(s)
- Amanda Maria Tomazini Munhoz Moya
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| | - Thaís Dolfini Alexandrino
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| | - Joseane Morari
- School of Medical Sciences, Universidade Estadual de Campinas, Rua Tessália Vieira de Camargo, 126, Campinas 13083-887, São Paulo, Brazil; (J.M.); (L.A.V.); (R.F.L.)
| | - Livia Mateus Reguengo
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| | - Licio Augusto Velloso
- School of Medical Sciences, Universidade Estadual de Campinas, Rua Tessália Vieira de Camargo, 126, Campinas 13083-887, São Paulo, Brazil; (J.M.); (L.A.V.); (R.F.L.)
| | - Raquel Franco Leal
- School of Medical Sciences, Universidade Estadual de Campinas, Rua Tessália Vieira de Camargo, 126, Campinas 13083-887, São Paulo, Brazil; (J.M.); (L.A.V.); (R.F.L.)
| | - Stanislau Bogusz Junior
- São Carlos Institute of Chemistry (IQSC), University of São Paulo (USP), São Carlos 13566-590, São Paulo, Brazil;
| | - Ana Paula Aparecida Pereira
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
- Faculty of Nutrition, Federal University of Mato Grosso, Avenida Fernando Correa da Costa, 2367, Boa Esperança, Cuiabá 78068-600, Mato Grosso, Brazil
| | - Glaucia Maria Pastore
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| | - Juliano Lemos Bicas
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| | - Cinthia Baú Betim Cazarin
- School of Food Engineering, Universidade Estadual de Campinas, Rua Monteiro Lobato, 80, Campinas 13083-862, São Paulo, Brazil; (A.M.T.M.M.); (T.D.A.); (L.M.R.); (A.P.A.P.); (G.M.P.); (J.L.B.)
| |
Collapse
|
42
|
Grancieri M, de São José VPB, Toledo RCL, Verediano TA, Sant'Ana C, Lúcio HG, Gonzalez de Mejia E, Martino HSD. Effect of digested chia seed protein on the gut microbiota and colon morphology of mice fed a high-saturated-fat diet. Food Funct 2024; 15:9284-9297. [PMID: 39162475 DOI: 10.1039/d4fo02199a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
The present study aimed to investigate the effect of digested total protein (DTP) from chia seed on the gut microbiota and morphology of mice fed with a high-fat diet. Forty-four male C57BL/6 mice were divided into 4 groups: AIN (standard diet), HF (high-fat diet), AIN + DTP (standard diet supplemented with 400 mg of digested chia seed protein), and HF + DTP (high-fat diet supplemented with 400 mg of digested chia seed protein) during 8 weeks. Colon morphology, tight junction's gene expression, and gut microbiota composition were evaluated. The consumption of digested chia seed protein (DTP) increased the crypts width, longitudinal and circular muscular layer. Furthermore, the AIN + DTP group enhanced the expression of tight junction proteins, including occludin and claudin, while the AIN + DTP and HF + DTP groups increase the zonula occludens expression. The α-diversity analysis showed a reduction in bacterial dominance in the HF + DTP group. All the experimental groups were grouped in different cluster, showing differences in the microbiota community in the β-diversity analyzes. The Firmicutes/Bacteroidetes ratio did not differ among the groups. The genera Olsenella and Dubosiella were increased in the AIN + DTP group, but the Oscillospiraceae_unclassified was increased in the HF + DTP group. The Alistipes was increased, while the Roseburia and Akkermansia were decreased in the AIN + DTP and HF + DTP groups. Then, the consumption of DTP from chia seed improved the gut microbiota composition and mucosal integrity, counteracting the adverse effects of high-fat diet.
Collapse
Affiliation(s)
- Mariana Grancieri
- Department of Pharmacy and Nutrition, Federal University of Espírito Santo, Alegre, ES, 29500-000, Brazil
| | | | - Renata Celi Lopes Toledo
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, 36571-000, Brazil.
| | | | - Cintia Sant'Ana
- Department of Pharmacy and Nutrition, Federal University of Espírito Santo, Alegre, ES, 29500-000, Brazil
| | - Haira Guedes Lúcio
- Department of Nutrition and Health, Universidade Federal de Viçosa, Viçosa, Minas Gerais, 36571-000, Brazil.
| | - Elvira Gonzalez de Mejia
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, Illinois, 61801, USA
| | | |
Collapse
|
43
|
Feng H, Ping K, Yang Y, Liu Z, Song Q, Chen S, Meng Y, He Q, Hu Y, Dong J. Quercetin alleviates difenoconazole-induced growth inhibition in carp through intestinal-brain axis. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 204:106066. [PMID: 39277382 DOI: 10.1016/j.pestbp.2024.106066] [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: 05/27/2024] [Revised: 07/01/2024] [Accepted: 07/29/2024] [Indexed: 09/17/2024]
Abstract
Difenoconazole (DIF) is frequently used for the management of fungal infections in fruit and vegetables and excessive residues in the aquatic environment can have adverse effects on fish such as growth inhibition. A treatment based on the dietary additive quercetin (QUE) is a promising approach to positively regulate the state of fish growth. This study focused on whether and how QUE alleviated DIF-induced growth inhibition in fish. In this study, carp were exposed to DIF (0.3906 mg/L) for consecutive 30 d, which showed growth inhibition. Disruption of the intestinal barrier led to elevated levels of intestinal lipopolysaccharide (LPS) and an inflammatory response. Through the intestinal-brain axis, LPS entered the brain where it disrupted the blood-brain barrier, triggered neuroinflammation, caused brain cell apoptosis, and damaged nerves in addition to other things. The dietary supplementation of QUE (400 mg/kg) reduced the levels of LPS in the intestinal and brain, while reducing inflammation and increasing the expression of appetite factors, thereby reducing growth inhibition in carp. This work provided evidence for QUE from the intestinal-brain axis perspective as a potential candidate for alleviating growth inhibition in fish.
Collapse
Affiliation(s)
- Huimiao Feng
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Kaixin Ping
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yue Yang
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Zhijun Liu
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Qimei Song
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Si Chen
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yu Meng
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Qian He
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yuxuan Hu
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jingquan Dong
- Jiangsu Marine Pharmaceutical Resources Development Engineering Research Center, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| |
Collapse
|
44
|
Li Y, Guo Y, Geng C, Song S, Yang W, Li X, Wang C. Vitamin D/vitamin D receptor protects intestinal barrier against colitis by positively regulating Notch pathway. Front Pharmacol 2024; 15:1421577. [PMID: 39130644 PMCID: PMC11310051 DOI: 10.3389/fphar.2024.1421577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024] Open
Abstract
Objective Vitamin D/Vitamin D receptor (VD/VDR) signaling and the Notch pathway are involved in intestinal barrier restoration in colitis; however, their relationship and underlying mechanism are largely unknown. Therefore, this study aimed to investigate the role and mechanism of VD/VDR and the Notch pathways in intestinal barrier protection. Methods Genetic Vdr knockout (VDR KO) and VD deficient (VDd) mice were established, and colitis was induced by feeding 2.5% dextran sodium sulfate (DSS) water. Mechanistic studies, including real-time PCR, immunofluorescence, Western blotting and dual-luciferase reporter assays, were performed on cultured Caco-2 cells and intestinal organoids. Results VD deficiency and VDR genetical KO increased the severity of DSS-induced colitis in mice, which presented a higher disease activity index score, increased intestinal permeability, and more severe intestinal histological damage than controls, accompanied by decreased and disrupted claudin-1 and claudin-3. Moreover, inhibition of Notch pathway by LY411,575 aggravated the severity of DSS-induced colitis and intestinal injury. In Caco-2 cells and intestinal organoids, the expression of Notch-1, N1ICD and Hes1 decreased upon downregulation or KO of VDR but increased upon paricalcitol (PAR, a VDR agonist) treatment. Meanwhile, PAR rescued claudin-1 and claudin-3 impairments that resulted from TNF-α exposure but failed to restore claudin-3 upon Notch inhibition. The dual-luciferase reporter assay further suggested that VD/VDR positively regulated the Notch signaling pathway by modulating Notch-1 transcription. Conclusion VD/VDR positively modulates Notch activation by promoting Notch-1 transcription to maintain intestinal tight junction integrity and barrier function. This highlights the VD/VDR-Notch pathway as a potential new therapeutic target for protecting the intestinal barrier against ulcerative colitis.
Collapse
Affiliation(s)
- Yanni Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Yaoyu Guo
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Chong Geng
- Laboratory of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, China
| | - Shuailing Song
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Wenjuan Yang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Xiao Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Chunhui Wang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
45
|
Chen P, Ding WL, Xu BW, Rehman MU, Liu KL, He YF, Li SY, Jian FC, Huang 黄 SC淑. Aflatoxin B1 as a complicit in intestinal damage caused by Eimeria ovinoidalis in lambs: Novel insights to reveal parasite-gut battle. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 947:174539. [PMID: 38977103 DOI: 10.1016/j.scitotenv.2024.174539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/25/2024] [Accepted: 07/04/2024] [Indexed: 07/10/2024]
Abstract
Mycotoxins, unavoidable contaminants in feed and feed ingredients, have the potential to influence the incidence and severity of various diseases upon ingestion. Sheep coccidiosis is an enteric disease caused by protozoa of Eimeria spp. However, the extent to which the presence of aflatoxin b1 (AFB1) synergistically exacerbates damage to intestinal health in lambs with Eimeria remains unclear. 50-day-old female lambs were randomly assigned to a 2 × 2 factorial arrangement of treatments for 15 days to assess the impact of AFB1 exposure on lambs with or without Eimeria (E.) ovinoidalis infection. Our findings reveal that AFB1 synergistically intensifies damage to intestinal health in lambs challenged by E. ovinoidalis. This is evidenced by disruptions to the intestinal microbiota and reductions in the production of short-chain fatty acids. AFB1 further aggravates damage to the cecal mechanical barrier. Additionally, AFB1 contributes to the entry of lipopolysaccharide into the bloodstream, activating the inflammatory response. Interestingly, AFB1 exposure history results in an early peak of oocyst excretion and a decreased number of oocyst excretion in E. ovinoidalis infected lambs. This may be closely linked to the destruction of the intestinal epithelial cell structure and its apoptosis, as indicated by a decreased ratio of Bcl-2 to Bax and increased caspase-3 levels. Mechanistically, proteomics analysis identified mitochondrial dysfunction (inhibition of the oxidative phosphorylation pathway) as the primary factor intensifying intestinal epithelial cell destruction caused by coccidia, exacerbated by AFB1 through the inhibiting the conversion of NADH to NAD+ in the cecum of lambs via down-regulation of the PGC-1α/NRF1/TFAM pathway. Overall, these results offer novel insights into the AFB1 complicity in accelerating intestinal damage caused by E. ovinoidalis in lambs. Targeting the mitochondrial oxidative phosphorylation pathway of the intestine may represent a new therapeutic strategy against the detrimental effects of mycotoxin and coccidia.
Collapse
Affiliation(s)
- Pan Chen
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Wen-Li Ding
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Bo-Wen Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Mujeeb Ur Rehman
- Directorate Planning & Development, Livestock and Dairy Development Department, Balochistan, Pakistan
| | - Kai-Li Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Yan-Feng He
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Sen-Yang Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Fu-Chun Jian
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China.
| | - Shu-Cheng 淑成 Huang 黄
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China.
| |
Collapse
|
46
|
Zhang W, Wang Y, Zhu M, Liu K, Zhang HL. Gut flora in multiple sclerosis: implications for pathogenesis and treatment. Neural Regen Res 2024; 19:1480-1488. [PMID: 38051890 PMCID: PMC10883522 DOI: 10.4103/1673-5374.387974] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/25/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT Multiple sclerosis is an inflammatory disorder characterized by inflammation, demyelination, and neurodegeneration in the central nervous system. Although current first-line therapies can help manage symptoms and slow down disease progression, there is no cure for multiple sclerosis. The gut-brain axis refers to complex communications between the gut flora and the immune, nervous, and endocrine systems, which bridges the functions of the gut and the brain. Disruptions in the gut flora, termed dysbiosis, can lead to systemic inflammation, leaky gut syndrome, and increased susceptibility to infections. The pathogenesis of multiple sclerosis involves a combination of genetic and environmental factors, and gut flora may play a pivotal role in regulating immune responses related to multiple sclerosis. To develop more effective therapies for multiple sclerosis, we should further uncover the disease processes involved in multiple sclerosis and gain a better understanding of the gut-brain axis. This review provides an overview of the role of the gut flora in multiple sclerosis.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Neurology, the First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Ying Wang
- Department of Neurology, the First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Mingqin Zhu
- Department of Neurology, the First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Kangding Liu
- Department of Neurology, the First Hospital of Jilin University, Jilin University, Changchun, Jilin Province, China
| | - Hong-Liang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, Beijing, China
| |
Collapse
|
47
|
Han Y, Spicer J, Huang Y, Bunt C, Liu M, Wen J. Advancements in oral insulin: A century of research and the emergence of targeted nanoparticle strategies. EUR J LIPID SCI TECH 2024; 126. [DOI: 10.1002/ejlt.202300271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Indexed: 01/03/2025]
Abstract
AbstractWith the growing prevalence of diabetes, there is an urgent demand for a user‐friendly treatment option that minimizes side effects related to the use of subcutaneous injections. Scientists have dedicated over a century to developing an oral dosage form of insulin that can be administrated orally. The oral route of administration is the most desirable route for regularly dosed drugs in terms of safety and patient compliance. However, oral delivery of insulin remains a formidable challenge due to its intrinsically limited ability to cross the intestinal epithelium membrane and susceptibility to enzymatic degradation. This article reviews oral insulin research over the past decade, with a particular focus on surface modifications of nanoparticles (NPs). Various strategies involving controlling surface charges, utilizing protective proteins, and targeting specific receptors with ligands have been explored. Notably, surface modifications of the NPs for targeting specific intestinal receptors have shown promise in enhancing insulin oral absorption and bioavailability. Advanced technologies such as oral microneedles and gene therapy have also been developed, but their safety requires further assessment. Despite encouraging preclinical results across numerous strategies, the current clinical evidence is less optimistic. In summary, the present findings highlight the substantial journey that still lies ahead before achieving successful oral delivery of insulin.Practical Applications: This review provides a summary of recent progress in oral insulin delivery, particularly highlighting surface‐modified functional nanoparticles serving as an effective drug delivery system, which offers valuable information to the researchers. Due to the limited effectiveness of oral protein drugs caused by biological barriers, innovative technologies and drug delivery systems have been developed to overcome these obstacles and achieve therapeutic goals. This review concluded that surface modifications to nanoparticles can improve insulin stability and permeability, thereby enhancing oral bioavailability. It could assist researchers in developing more effective and patient‐friendly oral drug delivery systems.
Collapse
Affiliation(s)
- Yue Han
- School of Pharmacy Faculty of Medical and Health Sciences The University of Auckland Auckland New Zealand
| | - Julie Spicer
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland Auckland New Zealand
| | - Yuan Huang
- Key Laboratory of Drug Targeting and Drug Delivery, West China School of Pharmacy, Sichuan University Chengdu China
| | - Craig Bunt
- The Department of Food Science University of Otago Dunedin New Zealand
| | - Mengyang Liu
- School of Pharmacy Faculty of Medical and Health Sciences The University of Auckland Auckland New Zealand
| | - Jingyuan Wen
- School of Pharmacy Faculty of Medical and Health Sciences The University of Auckland Auckland New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland Auckland New Zealand
| |
Collapse
|
48
|
Peng Y, Huang Y, Li H, Li C, Wu Y, Chen ZS, Wang X, Liao F, Miao C. Huangqin Qingre Chubi Capsule inhibits rheumatoid arthritis by regulating intestinal flora and improving intestinal barrier. Front Pharmacol 2024; 15:1422245. [PMID: 38989143 PMCID: PMC11233690 DOI: 10.3389/fphar.2024.1422245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/04/2024] [Indexed: 07/12/2024] Open
Abstract
Background Changes in intestinal flora and intestinal barrier in patients with preclinical and diagnosed rheumatoid arthritis (RA) suggest that intestinal flora and intestinal barrier play an important role in the induction and persistence of RA. Huangqin Qingre Chubi Capsule (HQC) is a clinically effective herbal formula for the treatment of RA, but its therapeutic mechanism has not been fully clarified. Materials and methods In this study, real-time qPCR (RT-qPCR), 16SrRNA sequencing, Western blot (WB), immunofluorescence and other methods were used to investigate whether HQC inhibited RA. Results Based on research in collages-induced arthritis (CIA) model in mice, human colon cancer cell line (Caco-2), and fibroblast-like synoviocytes (FLS) from RA patients, we found that intestinal flora was disturbed in CIA model group, intestinal barrier was damaged, and lipolyaccharide (LPS) level was increased, and HQC could regulate intestinal flora and intestinal barrier and reduce LPS translocation into blood. Antibiotic depletion weakened the anti-RA effect of HQC, and HQC fecal microbiota transplantation alleviated RA pathology. In addition, LPS increased the expression of RA pathologic factors MMP3, Fibronectin and inflammatory factors IL-6, TNF-α, IL-1β and IL-8, indicating that elevated peripheral blood level of LPS was related to RA pathology. Conclusion The dysregulation of intestinal flora and the disruption of intestinal barrier are significant factors in the development of RA. HQC improves RA by regulating intestinal flora, intestinal barrier and inhibiting LPS translocation into blood. The study unveiles RA's new pathogenesis and laid a scientific groundwork for advancing HQC therapy for RA.
Collapse
Affiliation(s)
- Yanhui Peng
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yurong Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hui Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Chen Li
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Yajie Wu
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, United States
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Faxue Liao
- Department of Orthopaedics, The First Affiliated Hospital, Anhui Public Health Clinical Center, Anhui Medical University, Hefei, China
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
- Institute of Prevention and Treatment of Rheumatoid Arthritis, Anhui University of Chinese Medicine, Hefei, Anhui, China
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hefei, China
| |
Collapse
|
49
|
Wang Z, Guo Z, Liu L, Ren D, Zu H, Li B, Liu F. Potential Probiotic Weizmannia coagulans WC10 Improved Antibiotic-Associated Diarrhea in Mice by Regulating the Gut Microbiota and Metabolic Homeostasis. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10308-1. [PMID: 38900235 DOI: 10.1007/s12602-024-10308-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
Antibiotic-associated diarrhea (AAD) is a common side effect of long-term and heavy antibiotic therapy. Weizmannia coagulans (W. coagulans) is an ideal probiotic because of its high viability, stability, and numerous health benefits to the host. In this study, the strains were first screened for W. coagulans WC10 (WC10) with a high combined ability based on their biological properties of gastrointestinal tolerance, adhesion, and short-chain fatty acid production ability. The effect of WC10 on mice with AAD was further evaluated. The results showed that WC10 was effective in improving the symptoms of AAD, effectively restoring antibiotic-induced weight loss, and reducing diarrhea status score and fecal water content. In addition, WC10 decreased the expression of pro-inflammatory cytokines and increased the expression of anti-inflammatory cytokines, alleviated intestinal tissue damage and inflammation, and improved intestinal epithelial barrier function by decreasing serum levels of enterotoxin, DAO, and D-lactic acid, and by increasing the expression of the intestinal mucosal immune factors sIgA and occludin. Importantly, the composition and function of the gut microbiota gradually recovered after WC10 treatment, increasing the number of SCFAs-producing Bifidobacterium and Roseburia. Subsequently, the short-chain fatty acid (SCFA) content was examined and WC10 significantly increased acetate, propionate, and butyrate production. Additionally, metabolomic analysis also showed that WC10 reversed the antibiotic interference with major metabolic pathways. These findings provide a solid scientific basis for the future application of W. coagulans WC10 in the treatment of AAD.
Collapse
Affiliation(s)
- Zengbo Wang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China
- Food College, Northeast Agricultural University, Harbin, 150030, China
| | - Zengtao Guo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China
- Food College, Northeast Agricultural University, Harbin, 150030, China
| | - Libo Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China
- Food College, Northeast Agricultural University, Harbin, 150030, China
| | - Daxi Ren
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hang Zu
- Heilongjiang Ubert Dairy Co., Heilongjiang, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China.
- Food College, Northeast Agricultural University, Harbin, 150030, China.
| | - Fei Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, China.
- Food College, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
50
|
Xu Y, Xu J, Zhu Y, Mao H, Li J, Kong X, Zhu X, Zhang J. Investigating gut microbiota-blood and urine metabolite correlations in early sepsis-induced acute kidney injury: insights from targeted KEGG analyses. Front Cell Infect Microbiol 2024; 14:1375874. [PMID: 38887493 PMCID: PMC11180806 DOI: 10.3389/fcimb.2024.1375874] [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: 01/24/2024] [Accepted: 05/13/2024] [Indexed: 06/20/2024] Open
Abstract
Background The interplay between gut microbiota and metabolites in the early stages of sepsis-induced acute kidney injury (SA-AKI) is not yet clearly understood. This study explores the characteristics and interactions of gut microbiota, and blood and urinary metabolites in patients with SA-AKI. Methods Utilizing a prospective observational approach, we conducted comparative analyses of gut microbiota and metabolites via metabolomics and metagenomics in individuals diagnosed with SA-AKI compared to those without AKI (NCT06197828). Pearson correlations were used to identify associations between microbiota, metabolites, and clinical indicators. The Comprehensive Antibiotic Resistance Database was employed to detect antibiotic resistance genes (ARGs), while Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways informed on metabolic processes and microbial resistance patterns. Results Our study included analysis of four patients with SA-AKI and five without AKI. Significant disparities in bacterial composition were observed, illustrated by diversity indices (Shannon index: 2.0 ± 0.4 vs. 1.4 ± 0.6, P = 0.230; Simpson index: 0.8 ± 0.1 vs. 0.6 ± 0.2, P = 0.494) between the SA-AKI group and the non-AKI group. N6, N6, N6-Trimethyl-L-lysine was detected in both blood and urine metabolites, and also showed significant correlations with specific gut microbiota (Campylobacter hominis and Bacteroides caccae, R > 0, P < 0.05). Both blood and urine metabolites were enriched in the lysine degradation pathway. We also identified the citrate cycle (TCA cycle) as a KEGG pathway enriched in sets of differentially expressed ARGs in the gut microbiota, which exhibits an association with lysine degradation. Conclusions Significant differences in gut microbiota and metabolites were observed between the SA-AKI and non-AKI groups, uncovering potential biomarkers and metabolic changes linked to SA-AKI. The lysine degradation pathway may serve as a crucial link connecting gut microbiota and metabolites.
Collapse
Affiliation(s)
- Yaya Xu
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiaotong University, Shanghai, China
| | - Jiayue Xu
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiaotong University, Shanghai, China
| | - Yueniu Zhu
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiaotong University, Shanghai, China
| | - Haoyun Mao
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiaotong University, Shanghai, China
| | - Jiru Li
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiaotong University, Shanghai, China
| | - Xiangmei Kong
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiaotong University, Shanghai, China
| | - Xiaodong Zhu
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiaotong University, Shanghai, China
| | - Jianhua Zhang
- Department of Pediatric Respiratory, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|