1
|
Hu X, Wang R, Kille P, Maret W, Hogstrand C. Zinc amino acid chelate and the Aryl Hydrocarbon Receptor (AHR) cooperate in improving the barrier function of a Caco-2 cell intestinal epithelium. J Nutr Biochem 2025; 141:109909. [PMID: 40154643 DOI: 10.1016/j.jnutbio.2025.109909] [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: 08/12/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
Zinc and several physiologically relevant ligands of the aryl hydrocarbon receptor (AHR) are nutrients that promote intestinal barrier function. We have identified that AHR activation upregulates the expression of zinc importers in the intestinal epithelium to increase intracellular zinc concentrations, which leads to improved epithelial barrier function. Here, we investigated if an amino acid chelate of zinc, in cooperation with AHR activation, can improve the barrier function of a differentiated Caco-2 cell epithelium. Functional assays of the Caco-2 cell epithelium demonstrate that both ZnSO4 and a lysine and glutamic acid chelate of Zn, in combination with the physiological AHR agonist 6-formylindolo[3,2-b]carbazole (FICZ), increase expression of tight junction proteins at the mRNA and protein levels. FICZ increases uptake of zinc into the epithelium in the presence of ZnSO4 or the amino acid Zn chelate in the medium to equal extents. We conclude that the lysine and glutamic acid chelate of Zn is as efficacious as ZnSO4 in reducing permeability of the Caco-2 cell epithelium in the presence of FICZ. The results suggest that dietary supplementation with bioavailable forms of zinc together with nutritional AHR agonists may be beneficial in improving gut barrier function and help prevent inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Xiuchuan Hu
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, London, UK
| | - Rui Wang
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, London, UK
| | - Peter Kille
- School of Biosciences, Cardiff University, Cardiff, UK
| | - Wolfgang Maret
- Department of Nutritional Sciences, School of Life Course and Population Sciences, King's College London, London, UK
| | - Christer Hogstrand
- Department of Analytical, Environmental and Forensic Sciences, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK.
| |
Collapse
|
2
|
Sato S, Hasan AU, Obara M, Kondo Y, Taira E. Long-term consumption of moderate amounts of sucrose-sweetened drinks disrupts intestinal barrier function by impairing goblet cell differentiation. Cell Tissue Res 2025; 400:273-285. [PMID: 40072586 DOI: 10.1007/s00441-025-03961-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025]
Abstract
While the prolonged consumption of sucrose-containing beverages is known to impact many organs, their specific effects on the small intestine remain elusive. This study aimed to evaluate how regular intake of sucrose, in amounts typically consumed, affects goblet cells, which play a critical role in regulating the mucosal barrier and innate immune defenses in the small intestine. Ten-week-old male ddY mice, a model of diet-induced obesity, were given a regular diet with either plain water or 7% sucrose water. Caloric intake was monitored weekly through food and drink measurements. After 8 weeks, glucose and insulin responses were evaluated following an oral gavage of glucose or sucrose. At 14 weeks, plasma, whole small intestine, and liver samples were collected. Despite achieving an isocaloric state, mice drinking sucrose water showed approximately a 1.5-fold increase in body weight and impaired glucose tolerance. In the small intestine, genes involved in sucrose digestion and absorption (Sis, Sglt1, Glut2, and Glut5) were upregulated, while genes essential for maintaining the intestinal barrier and function (Epcam, Fabp2, Cldn1, Ocln, and Tjp1) were downregulated. Serum levels and mRNA expression of the inflammatory cytokine, interleukin-18 were elevated. Genes responsible for goblet cell differentiation and function (Hes1, Gfi1, Spdef, and Klf4) were downregulated, leading to an increase in immature goblet cells and a decrease in mucin-producing markers (Muc2, Muc4, and Muc13) in the jejunum. The findings underscore that besides obesity, long-term intake of sucrose-containing drinks provokes localized inflammation and disrupts small intestinal barrier function by impairing goblet cell differentiation and activity.
Collapse
Affiliation(s)
- Sachiko Sato
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Arif U Hasan
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan.
| | - Mami Obara
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Yukiko Kondo
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Eiichi Taira
- Department of Pharmacology, School of Medicine, Iwate Medical University, Iwate, Japan
| |
Collapse
|
3
|
Huang W, Jiang T, He J, Ruan J, Wu B, Tao R, Xu P, Wang Y, Chen R, Wang H, Yang Q, Zhang K, Jin L, Sun D, You J. Modulation of Intestinal Flora: a Novel Immunotherapeutic Approach for Enhancing Thyroid Cancer Treatment. Probiotics Antimicrob Proteins 2025; 17:1038-1063. [PMID: 39890752 DOI: 10.1007/s12602-025-10471-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Over the past 3 years, there has been a growing interest in clinical research regarding the potential involvement of intestinal flora in thyroid cancer (TC). This review delves into the intricate connection between intestinal flora and TC, focusing on the particular intestinal flora that is directly linked to the disease and identifying which may be able to predict potential microbial markers of TC. In order to shed light on the inflammatory pathways connected to the onset of TC, we investigated the impact of intestinal flora on immune modulation and the connection between chronic inflammation when investigating the role of intestinal flora in the pathogenesis of TC. Furthermore, the potential role of intestinal flora metabolites in the regulation of thyroid function was clarified by exploring the effects of short-chain fatty acids and lipopolysaccharide on thyroid hormone synthesis and metabolism. Based on these findings, we further explore the effects of probiotics, prebiotics, postbiotics, vitamins, and trace elements.
Collapse
Affiliation(s)
- Weiqiang Huang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Tao Jiang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxuan He
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Baihui Wu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Runchao Tao
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Peiye Xu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Yongpan Wang
- Department of General Surgery, The First People's Hospital of Jiashan, Jiashan Hospital Afliated of Jiaxing University, Jiaxing, 314100, China
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, SAR 999077, China
| | - Hanbing Wang
- The University of Hong Kong School of Biomedical Sciences, Hong Kong, 999077, SAR, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Jinfeng You
- Department of Obstetrics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China.
| |
Collapse
|
4
|
Zhou P, Xu HJ, Wang L. Cardiovascular protective effects of natural flavonoids on intestinal barrier injury. Mol Cell Biochem 2025; 480:3343-3362. [PMID: 39820766 DOI: 10.1007/s11010-025-05213-2] [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: 10/26/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
Natural flavonoids may be utilized as an important therapy for cardiovascular diseases (CVDs) caused by intestinal barrier damage. More research is being conducted on the protective properties of natural flavonoids against intestinal barrier injury, although the underlying processes remain unknown. Thus, the purpose of this article is to present current research on natural flavonoids to reduce the incidence of CVDs by protecting intestinal barrier injury, with a particular emphasis on intestinal epithelial barrier integrity (inhibiting oxidative stress, regulating inflammatory cytokine expression, and increasing tight junction protein expression). Furthermore, the mechanisms driving intestinal barrier injury development are briefly explored, as well as natural flavonoids having CVD-protective actions on the intestinal barrier. In addition, natural flavonoids with myocardial protective effects were docked with ZO-1 targets to find natural products with higher activity. These natural flavonoids can improve intestinal mechanical barrier function through anti-oxidant or anti-inflammatory mechanism, and then prevent the occurrence and development of CVDs.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Hui-Juan Xu
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Liang Wang
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
- Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China.
| |
Collapse
|
5
|
Côco LZ, de Souza Belisário E, Vasquez EC, Pereira TMC, Aires R, Campagnaro BP. Probiotics: a promising future in the treatment of ulcerative colitis? Pharmacol Rep 2025; 77:645-657. [PMID: 40214948 DOI: 10.1007/s43440-025-00724-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 05/13/2025]
Abstract
Ulcerative colitis is an idiopathic and chronic inflammatory bowel disease, characterized by inflammation of the mucosa of the colon and rectum. Clinical manifestations commonly include abdominal pain, diarrhea (with or without hematochezia), and weight loss. The pathogenesis of ulcerative colitis is multifactorial, involving a combination of genetic predispositions and lifestyle factors. High consumption of processed food, sedentary habits, alcohol intake, and stress are among the lifestyle factors implicated in disease onset and progression. Current treatment strategies focus on managing symptoms and inducing remission, however, the chronic nature of the disease, along with the adverse effects of conventional therapies, often compromises patient's quality of life. Therefore, exploring alternative therapies that can prolong remission and reduce symptom burden is important. Experimental evidence suggests that probiotics may extend remission duration in ulcerative colitis. Moreover, probiotics exhibit efficacy in amelioration clinical symptoms by reducing inflammation markers, preserving, and restoring intestinal epithelial. This review explores the advantages of the administration of probiotics in the treatment of ulcerative colitis, elucidating their mechanism of action.
Collapse
Affiliation(s)
- Larissa Zambom Côco
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Eduarda de Souza Belisário
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Elisardo Corral Vasquez
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Thiago Melo Costa Pereira
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Rafaela Aires
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil
| | - Bianca Prandi Campagnaro
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), R Mercurio s/n, 29102623, Vila Velha, 29102-920, ES, Brazil.
| |
Collapse
|
6
|
Zhang Y, Wang Y, Lu Y, Quan H, Wang Y, Song S, Guo H. Advanced oral drug delivery systems for gastrointestinal targeted delivery: the design principles and foundations. J Nanobiotechnology 2025; 23:400. [PMID: 40448152 DOI: 10.1186/s12951-025-03479-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Accepted: 05/20/2025] [Indexed: 06/02/2025] Open
Abstract
Oral administration has long been considered the most convenient method of drug delivery, requiring minimal expertise and invasiveness. Unlike injections, it avoids discomfort, wound infections, and complications, leading to higher patient compliance. However, the effectiveness of oral delivery is often hindered by the harsh biological barriers of the gastrointestinal tract, which limit the bioaccessibility and bioavailability of drugs. The development of oral drug delivery systems (ODDSs) represents a critical area for the advancement of pharmacotherapy. This review highlights the characteristics and precise targeting mechanisms of ODDSs. It first examines the unique properties of each gastrointestinal compartment, including the stomach, small intestine, intestinal mucus, intestinal epithelial barrier, and colon. Based on these features, it outlines the targeting strategies and design principles for ODDSs aimed at overcoming gastrointestinal barriers to enhance disease treatment. Lastly, the review discusses the challenges and potential future directions for ODDS development, emphasizing their importance for advancing drug delivery technologies and accelerating their future growth.
Collapse
Affiliation(s)
- Yafei Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yiran Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yao Lu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Heng Quan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100089, China
| | - Yuqi Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Sijia Song
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100089, China
| | - Huiyuan Guo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100089, China.
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
7
|
Wang ZX, Zhang LL, Zhao XH. Enhanced alleviation of the selenite-grafted soluble and nondigestive Chinese Yam polysaccharides on nonylphenol-induced cytotoxicity and barrier damage in intestinal epithelial cells. Sci Rep 2025; 15:17970. [PMID: 40410351 PMCID: PMC12102335 DOI: 10.1038/s41598-025-03118-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 05/19/2025] [Indexed: 05/25/2025] Open
Abstract
This study explored the in vitro alleviation of the soluble and non-digestible Chinese yam polysaccharides (YP) with covalent selenite-grafting on the nonylphenol-induced cytotoxicity and barrier damage in rat intestinal epithelial (IEC-6) cells. Two grafted products YPSe-I and particularly YPSe-II possessed much higher Se contents than YP (0.803 and 1.486 versus 0.037 g/kg), could alleviate the cytotoxicity of nonylphenol by causing higher cell viability but lower lactate dehydrogenase release and ROS production, and were capable of repairing the induced barrier damage through increasing transepithelial electrical resistance, reducing paracellular permeability, promoting the production and distribution of cytoskeleton F-actin, and up-regulating the expression levels of three tight junction proteins namely zonula occludens-1, occludin, and claudin-1. Meanwhile, the expression levels of two proteins namely p-p38 and p-JNK in the cells, which are crucial to the activation of the MAPK signaling pathway, were up-regulated by nonylphenol but down-regulated by YPSe-I and YPSe-II. The results consistently confirmed that YP and YPSe-II exhibited the respective lowest and highest activities in the cells to alleviate the nonylphenol-induced cytotoxicity and barrier damage, declaring that both YP selenization and higher selenite-grafting extent were the critical factors controlling the measured activities of YPSe-I and YPSe-II. Collectively, this selenite-grafting of YP endowed the selenized products with higher activity in the cells to reduce nonylphenol-induced cytotoxicity, especially to alleviate the induced barrier damage by inactivating the MAPK signaling pathway.
Collapse
Affiliation(s)
- Zhen-Xing Wang
- School of Biology and Food Engineering, Guangdong University of Petrochemical Technology, Maoming, 525000, China
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China
- College of Food Engineering, Harbin University of Commerce, Harbin, 150076, China
| | - Li-Li Zhang
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China
| | - Xin-Huai Zhao
- School of Biology and Food Engineering, Guangdong University of Petrochemical Technology, Maoming, 525000, China.
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
8
|
Zhang J, Jiang WD, Wu P, Liu Y, Ma YB, Shi HQ, Kuang SY, Li SW, Tang L, Zhou XQ, Feng L. Dietary addition of fraxetin improved intestinal structure and growth performance in juvenile grass carp (Ctenopharyngodon idella): as a potential novel phytogenic feed additive. J Nutr Biochem 2025:109969. [PMID: 40412568 DOI: 10.1016/j.jnutbio.2025.109969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/05/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
The well-being and development of fish are affected to varying degrees under the intensive aquaculture model, and the use of Chinese herbs for aquaculture disease control and feed additives has received increasing attention. This study examined fraxetin supplementation in juvenile grass carp to investigate its effects on growth and intestinal structure. There were 1080 grass carp (11.58 ± 0.01 g) assigned to 6 treatments, fed with fraxetin (0, 3.9, 7.9, 15.8, 31.5, and 63.1 mg/kg) for 60 days in each treatment. In our study, appropriate fraxetin significantly increased final body weight (FBW), percent weight gain (PWG), and specific growth rate (SGR) compared to the unadded group (P < 0.05), but did not affect feed efficiency (FE) (P > 0.05). The administration of 7.9 mg/kg of fraxetin significantly improved fish intestinal development and body composition. Appropriate dietary fraxetin significantly enhanced intestinal digestive enzymes and brush border enzyme activity (P < 0.05), decreased serum diamine oxidase (DAO) levels (P < 0.05), and decreased intestinal cell apoptosis (P < 0.05). Appropriate levels of fraxetin inhibited the RhoA/ROCK signaling pathway while upregulating both mRNA and protein expression of tight junction (TJ) and adherens junction (AJ) (P < 0.05). These changes significantly improved apical junction complex (AJC) integrity. In conclusion, dietary supplementation with appropriate levels of fraxetin added to the diets had a facilitating effect on digestion and absorption, improved intestinal structure, and promoted fish growth performance in juvenile grass carp. In addition, the optimal dietary fraxetin levels were evaluated to be 6.06 and 7.79 mg/kg based on linear regression analysis of PWG and DAO, respectively.
Collapse
Affiliation(s)
- Jie Zhang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Yao-Bin Ma
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - He-Qun Shi
- Guangzhou Cohoo Biotech Research & Development Centre, Guangzhou, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Shu-Wei Li
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Sichuan Animtech Feed Co. Ltd, Chengdu, 610066, Sichuan, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China.
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China; Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China.
| |
Collapse
|
9
|
Zhou Y, Song W, Wang Y, Li S, Shan C, Dong J, Xu Z, Zou H, Pan Y, Chen X, Zhang Y, Song J. Calycosin regulates gut microbiota-bile acid-FXR axis to protect rats from cerebral ischemia-reperfusion injury. Eur J Pharmacol 2025; 1000:177707. [PMID: 40348321 DOI: 10.1016/j.ejphar.2025.177707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 04/30/2025] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Recent reports have shown that metabolites derived from gut microbiota play a vital role in intestinal diseases, immune regulation, and neuroinflammation. Nowadays, calycosin has been revealed the protective mechanism from different perspectives on cerebral ischemia-reperfusion injury (CIRI), while the effect of gut microbiota-bile acid-farnesoid X receptor (FXR) axis on the inflammatory protection of CIRI has not been explored. To this end, we established a middle cerebral artery occlusion (MCAO) model firstly to assess the protection of calycosin in CIRI through neurological deficit scoring, TTC staining, and HE staining. Secondly, 16s RNA sequencing, ELISA, real-time qPCR, Western blot, and total bile acid (TBA) detection kit were utilized to detect the pharmacology of calycosin on MCAO rats. Our data indicated that calycosin could significantly improve nerve function scores, reduce cerebral infarction volume, lower serum levels of IL-10, IL-17 inflammatory factors, and TBA, increase mRNA and protein levels of ZO-1 and Occludin in brain, as well as FXR, ZO-1 and Occludin levels in colon. In summary, calycosin can exert a neuroinflammatory protective effect on CIRI in rats via regulating the gut microbiota to improve bile acid metabolism.
Collapse
Affiliation(s)
- Yujia Zhou
- the Second School of Clinical Medicine, Zhejiang Chinese Medical University, China.
| | - Wenke Song
- School of Life Sciences, Zhejiang Chinese Medical University, China.
| | - Yaru Wang
- School of Life Sciences, Zhejiang Chinese Medical University, China.
| | - Simeng Li
- School of Life Sciences, Zhejiang Chinese Medical University, China.
| | - Chuchu Shan
- School of Life Sciences, Zhejiang Chinese Medical University, China.
| | - Jingyi Dong
- School of Life Sciences, Zhejiang Chinese Medical University, China.
| | - Zhengyuan Xu
- School of Life Sciences, Zhejiang Chinese Medical University, China.
| | - Haonan Zou
- School of Life Sciences, Zhejiang Chinese Medical University, China.
| | - Yifeng Pan
- the Second Affiliated Hospital, Zhejiang University School of Medicine, China.
| | - Xingying Chen
- Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, China.
| | - Yuyan Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, China.
| | - Jingmei Song
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, China.
| |
Collapse
|
10
|
Lin Y, Gong J, Buttimer C, Pan X, Jia Y, Bai Z, Wang R, Tong H, Bao H. Effects of astaxanthin on growth performance, intestinal integrity, and microbiota in Salmonella Enteritidis-infected chickens. Poult Sci 2025; 104:105056. [PMID: 40132313 PMCID: PMC11986504 DOI: 10.1016/j.psj.2025.105056] [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: 01/17/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025] Open
Abstract
This study investigated the effects of astaxanthin (AST) supplementation in drinking water on the growth performance, intestinal barrier function, and cecal microbiota of broilers challenged with Salmonella Enteritidis. During the 20-day experiment, two hundred and forty 1-day-old male Arbor Acres birds were randomly assigned into a 2 × 2 factorial design with four groups: a non-challenged control (CON), an S. Enteritidis-challenged group (SA), a group receiving AST treatment (AST), and an S. Enteritidis-challenged group receiving AST treatment (SA+AST). Each treatment comprised six replicate groups, and challenged groups were inoculated with S. Enteritidis from day 2 to day 4. The results indicated that S. Enteritidis infection significantly reduced the average daily feed intake (ADFI) in broilers and adversely affected average daily gain (ADG) and feed conversion ratio (FCR) by day 20. AST supplementation significantly improved FCR. While S. Enteritidis infection did not significantly affect ileal mucosa antioxidation, it significantly decreased villus height and the villus height-to-crypt depth ratio (VCR), and significantly downregulated mRNA expression of ZO-1 and Occludin. However, AST supplementation significantly enhanced antioxidant capacity (T-AOC), increased villus height and VCR in the ileum, and notably upregulated ZO-1 and MUC2 expression levels, particularly mitigating the adverse effects of S. Enteritidis infection on ileal crypt depth. Furthermore, S. Enteritidis infection significantly affected both the α- and β-diversity of cecal microbiota. Infection with S. Enteritidis was associated with changes at the phylum level, including significant increases in Alistipes, unclassified_f__Lachnospiraceae, and bacteria of the Clostridia UCG-014 grouping, alongside notable decreases in Bacteroides, Akkermansia, Blautia, and Butyricicoccus. AST supplementation significantly decreased the abundance of norank_f__Ruminococcaceae and increased the abundance of Lachnoclostridium and unclassified_f__Lachnospiraceae in the challenged group. In conclusion, AST supplementation in drinking water could improve growth performance and intestinal health in broilers challenged with S. Enteritidis.
Collapse
Affiliation(s)
- Yong Lin
- Institute of Agricultural Facilities and Equipment & Key Laboratory of Protected Agriculture Engineering in the Middle and Lower Reaches of Yangtze River, Ministry of Agriculture and Rural Affair& Jiangsu Engineering Research Center for Facility Waterfowl Health Breeding Equipment, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu, PR China
| | - Jiansen Gong
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, Jiangsu, PR China
| | - Colin Buttimer
- APC Microbiome Institute, University College Cork, Cork T12 YT20, Ireland
| | - Xiaoqing Pan
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu, PR China
| | - Yimin Jia
- Key Laboratory of Animal Physiology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, PR China
| | - Zongchun Bai
- Institute of Agricultural Facilities and Equipment & Key Laboratory of Protected Agriculture Engineering in the Middle and Lower Reaches of Yangtze River, Ministry of Agriculture and Rural Affair& Jiangsu Engineering Research Center for Facility Waterfowl Health Breeding Equipment, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu, PR China
| | - Ran Wang
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu, PR China
| | - Haibing Tong
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, Jiangsu, PR China
| | - Hongduo Bao
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, Nanjing, Jiangsu, PR China.
| |
Collapse
|
11
|
Liu L, Jia R, Chen W, Chen W, Wang X, Guo Z. The lotus seed starch-EGCG complex modulates obesity in C57BL/6J mice through the regulation of the gut microbiota. Int J Biol Macromol 2025; 310:143256. [PMID: 40250649 DOI: 10.1016/j.ijbiomac.2025.143256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/23/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
The starch-polyphenol complex, identified as RS5-resistant starch, has been shown to regulate the gut environment and inhibit metabolic diseases, including obesity. In a study with C57BL/6 obese mice fed LSE, potential anti-obesity effects were demonstrated through physiological and biochemical assessments, gut microbiota analysis, and mechanistic insights. The study showed that LSE reduced mice body weight, serum total cholesterol, and triglycerides (P < 0.05). Serum inflammatory markers (TNF-α, IL-6, IL-1β) and LPS levels were significantly decreased, while glucose tolerance (AUC reduced by 29.29 %) and insulin sensitivity (AUC reduced by 31.79 %) were improved. Histological analysis indicated reduction in adipocyte size and attenuation of hepatic steatosis. Gut microbiota profiling demonstrated LSE increased beneficial bacteria genera Faecalibacterium, Bifidobacterium, and Akkermansia. This correlated with enhanced SCFA production (acetate 41.53 %, propionate 45.52 %, butyrate 57.49 % increase). These findings demonstrate that LSE exerts anti-obesity effects through modulation of the gut microbiota-SCFA-metabolic axis, supporting starch-polyphenol complexes as functional food candidates.
Collapse
Affiliation(s)
- Lu Liu
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ru Jia
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wenjing Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Wenyu Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiaoying Wang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Zebin Guo
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
12
|
Zhai S, Peng X, Liu C, Zhang R, Jin C, Jiang X, Feng P, Liang Y, Yuan X, Zhang J, Yang Y. Ginsenoside Rg1 alleviates ochratoxin A-induced liver inflammation in ducklings: Involvement of intestinal microbiota modulation and the TLR4/NF-κB pathway inhibition. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 296:118186. [PMID: 40222107 DOI: 10.1016/j.ecoenv.2025.118186] [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: 01/10/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Ochratoxin A (OTA) is a toxic fungal secondary metabolite that triggers liver inflammation in animals. OTA could disrupt intestinal microbiota balance by promoting Gram-negative bacteria growth and activating the Toll-like receptor 4 (TLR4)/Nuclear factor-kappa B (NF-κB) signaling pathway, thereby inducing liver inflammation. Ginsenoside Rg1 (Rg1) is an active component of ginseng, exhibits anti-inflammatory, antibacterial, and antioxidative properties, particularly against gram-negative bacteria. Rg1 has been shown to maintain intestinal microbiota homeostasis and inhibit the TLR4 signaling pathway to alleviate liver inflammation. Given these established mechanisms, the aim of this study was to explore the preventive effect of Rg1 in countering OTA-induced liver inflammation through modulation of intestinal microbiota and the TLR4/NF-κB signaling pathway. The results revealed that Rg1 reduced OTA residues in the cecum and enhanced intestinal barrier function. Moreover, Rg1 ameliorated the intestinal microbiota composition in OTA-treated ducklings by decreasing the relative abundance of lipopolysaccharide (LPS)-related bacteria. Rg1 also increases the abundance of short-chain fatty acid (SCFA)-producing bacteria. Additionally, Rg1 supplementation with OTA decreased the accumulation of LPS in tissues and inhibited the TLR4/NF-κB signaling pathway. Intriguingly, Rg1 maintained its beneficial effects in OTA-treated ducklings even after antibiotic treatment by inhibiting the TLR4/NF-κB pathway. These findings emphasized the importance of intestinal microbiota homeostasis and TLR4/NF-κB pathway suppression in the anti-inflammatory action of Rg1 during OTA-induced liver inflammation.
Collapse
Affiliation(s)
- Shuangshuang Zhai
- College of Animal Science and Technology, Yangtze University, Jingzhou, Hubei Province 434023, China.
| | - Xin Peng
- College of Animal Science and Technology, Yangtze University, Jingzhou, Hubei Province 434023, China
| | - Cheng Liu
- College of Animal Science and Technology, Yangtze University, Jingzhou, Hubei Province 434023, China
| | - Ran Zhang
- Shenzhen International Travel Health Care Center (Shenzhen Customs District Port Outpatient Clinics), Shenzhen Customs District, Shenzhen 518000, China
| | - Chunlong Jin
- Shenzhen International Travel Health Care Center (Shenzhen Customs District Port Outpatient Clinics), Shenzhen Customs District, Shenzhen 518000, China
| | - Xiayu Jiang
- College of Animal Science and Technology, Yangtze University, Jingzhou, Hubei Province 434023, China
| | - Peishi Feng
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang Province 310014, China
| | - Yuting Liang
- College of Animal Science and Technology, Yangtze University, Jingzhou, Hubei Province 434023, China
| | - Xi Yuan
- College of Animal Science and Technology, Yangtze University, Jingzhou, Hubei Province 434023, China
| | - Jinqiu Zhang
- College of Animal Science and Technology, Yangtze University, Jingzhou, Hubei Province 434023, China
| | - Ye Yang
- College of Animal Science and Technology, Yangtze University, Jingzhou, Hubei Province 434023, China
| |
Collapse
|
13
|
Zhao X, Ye X, Gu Y, Lou Y, Zhou Z, Ji Y, Xu D. Oxymatrine for inflammatory bowel disease in preclinical studies: a systematic review and meta-analysis. Front Med (Lausanne) 2025; 12:1542953. [PMID: 40370726 PMCID: PMC12075229 DOI: 10.3389/fmed.2025.1542953] [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: 12/10/2024] [Accepted: 04/15/2025] [Indexed: 05/16/2025] Open
Abstract
Background Inflammatory Bowel Disease (IBD) is a chronic, idiopathic inflammatory disorder of the intestines. Oxymatrine (OMT) is a naturally active substance found in the desiccated roots of Sophora flavescens. It possesses anti-tumor, antiviral, and anti-inflammatory properties. In recent years, its therapeutic role in IBD has gradually been discovered. This review aims to explore the impact of OMT on inflammatory bowel disease by animal models. Methods Conduct a systematic search in the PubMed, Embase, Web of Science, Cochrane, and Medline databases. Using SYRCLE's risk of bias tool to assess the bias risk and quality of the included studies. For some data presented as figures, Web Plot Digitizer 4.2 software was used to extract it. STATA 16.0 was selected for the final meta-analysis. Results After rigorous literature screening, 12 studies were included. The data analysis results indicated that the disease activity index (DAI), histopathological score (HS), interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), nuclear factor-κB (NF-κB), and myeloperoxidase (MPO) activity in the IBD animal models significantly decreased following intervention with oxymatrine. Furthermore, OMT also extended the colon length in the animal models and improved the expression level of zonula occludens-1(ZO-1) and occludin. These results suggested that OMT may improve the condition of IBD through anti-inflammatory, antioxidative stress and protecting the intestinal barrier. Conclusion Meta-analysis suggests oxymatrine positively affects IBD animal models. This provides new insights for the clinical treatment of inflammatory bowel disease. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/view/CRD42024570580, identifier [CRD42024570580].
Collapse
Affiliation(s)
- Xuan Zhao
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaolu Ye
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yuting Gu
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yijie Lou
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhanyi Zhou
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yunxi Ji
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Daogun Xu
- Wenling Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Taizhou, Zhejiang, China
| |
Collapse
|
14
|
Huyghe P, Ceulemans M, Keita ÅV, Söderholm J, Depoortere I, Tack J, Wauters L, Vanuytsel T. The Duodenal Microenvironment in Functional Dyspepsia. J Neurogastroenterol Motil 2025; 31:186-198. [PMID: 40205896 PMCID: PMC11986653 DOI: 10.5056/jnm24176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 04/11/2025] Open
Abstract
Functional dyspepsia (FD) is a chronic gastrointestinal disorder without a readily identifiable organic cause, resulting in bothersome upper abdominal symptoms. It is a highly prevalent disorder of which the pathophysiology remains mostly elusive, despite intensive research efforts. However, recent studies have found alterations in the microenvironment of the duodenum in patients with FD. In this review we summarize the duodenal microenvironment in homeostatic conditions and the alterations found in patients with FD, highlighting the similarities and discrepancies between different studies. The most consistent findings, being an impaired duodenal barrier and duodenal immune activation, are reviewed. We discuss the potential triggers for these observed alterations, including psychological comorbidities, luminal alterations and food related triggers. In summary, this review presents the evidence of molecular and cellular changes in patients with FD, with an impaired duodenal barrier and activated mucosal eosinophils and mast cells, challenging the notion that FD is purely functional, and offering different targets for potential future treatments.
Collapse
Affiliation(s)
- Pauline Huyghe
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Matthias Ceulemans
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Åsa V Keita
- Department of Biomedical and Clinical Sciences and Department of Surgery, Linköping University, Linköping, Sweden
| | - Johan Söderholm
- Department of Biomedical and Clinical Sciences and Department of Surgery, Linköping University, Linköping, Sweden
| | - Inge Depoortere
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Lucas Wauters
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Tim Vanuytsel
- Translational Research Centre for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
15
|
Yu Z, Qiu Y, Yang Y, Wen C, Huang S, Liu T, Zhong R, Peng X. Nano Zinc Oxide Restores Gut Barrier Integrity and Modulates Microbiota to Mitigate TNBS-Induced Colitis in Mice. Biol Trace Elem Res 2025:10.1007/s12011-025-04635-9. [PMID: 40285830 DOI: 10.1007/s12011-025-04635-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/20/2025] [Indexed: 04/29/2025]
Abstract
Inflammatory bowel diseases (IBD) are chronic, relapsing gastrointestinal disorders with complex pathological mechanisms and limited treatment options, caused by various uncertain factors. Zinc plays a crucial role in wound healing, tissue regeneration, and immune responses. While zinc deficiency is common among patients with IBD, the effects of dietary zinc supplementation on the disease course remain unclear. In this study, 125 male C57BL/6 J mice were randomly divided into five groups: a control group (Con), a 2,4,6-trinitrobenzenesulfonic acid (TNBS) induction group, and three zinc supplementation groups receiving 160 ppm, 400 ppm, and 1000 ppm of zinc oxide nanoparticles ( ZnO NPs), respectively. The results showed that dietary supplementation with ZnO NPs significantly alleviated damage to the colonic mucosal epithelial cells and crypts in IBD mice, while effectively reducing the inflammatory response. Furthermore, ZnO NPs helps maintain the quantity and secretion function of goblet cells, restores normal expression levels of tight junction proteins (ZO-1, occludin), and proliferating cell nuclear antigen (PCNA), and preserves the diversity of gut microbiota. Notably, a significant protective effect was observed with dietary zinc supplementation at 160 ppm. These findings suggest that ZnO NPs significantly improves TNBS-induced intestinal inflammatory damage by modulating the gut microbiota, mucus, and mechanical barriers.
Collapse
Affiliation(s)
- Zhengqiang Yu
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, China
- Department of Technology, Sichuan Youngster Technology Co., Ltd. No. 733, Furong Avenue, Wenjiang District, Chengdu, 611130, China
| | - Yi Qiu
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, China
| | - Yingxiang Yang
- School of Life Sciences, China West Normal University, Nanchong, 637001, Sichuan, China
| | - Changlin Wen
- Department of Technology, Sichuan Youngster Technology Co., Ltd. No. 733, Furong Avenue, Wenjiang District, Chengdu, 611130, China
| | - Shiyuan Huang
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, China
| | - Tao Liu
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, China
| | - Rao Zhong
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, China
| | - Xi Peng
- Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, No. 2025, Chengluo Avenue, Chengdu, 610106, China.
| |
Collapse
|
16
|
Yang B, Xu Y, Zhang W, Zhu D, Huang B, Yang Y, Jia X, Feng L. Oral absorption mechanisms of polysaccharides and potential as carriers for the construction of nano-delivery systems: A review. Int J Biol Macromol 2025; 310:143184. [PMID: 40253019 DOI: 10.1016/j.ijbiomac.2025.143184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
Polysaccharides have garnered increasing attention in recent years for their potential in oral drug delivery within biomaterials and pharmaceuticals, owing to their excellent physicochemical properties, bioactivity, and low toxicity. However, the absorption of polysaccharides encounters multiple challenges posed by the biological, chemical, mechanical, and immune barriers of the intestinal mucosa. Therefore, elucidating the mechanisms by which polysaccharides traverse the intestinal mucosa for oral absorption is essential for their further development and application. Current studies have identified several polysaccharide absorption pathways, including transcellular transport, paracellular transport, M cell and Peyer's patches mediated transport, and intestinal flora mediated transport. Furthermore, numerous studies have demonstrated that polysaccharides can enhance the solubility, gastrointestinal stability, and permeability of small molecule components, which significantly improves their bioavailability. More importantly, nano-delivery systems utilizing polysaccharides as carriers have shown great promise in enhancing the targeting of small molecule components, thereby opening new avenues for drug delivery applications. We hope this review will provide theoretical support and inspiration for a deeper understanding of oral absorption mechanisms and the potential of polysaccharides in the development of nano-delivery systems.
Collapse
Affiliation(s)
- Bing Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; Jiangning Hospital of Chinese Medicine, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yan Xu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Weiye Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Dandan Zhu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Bin Huang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yanjun Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Liang Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; Jiangning Hospital of Chinese Medicine, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
17
|
Qin N, Liu H, Wang X, Liu Y, Chang H, Xia X. Sargassum fusiforme polysaccharides protect mice against Citrobacter rodentium infection via intestinal microbiota-driven microRNA-92a-3p-induced Muc2 production. Int J Biol Macromol 2025; 300:140271. [PMID: 39863236 DOI: 10.1016/j.ijbiomac.2025.140271] [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: 08/05/2024] [Revised: 01/07/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Sargassum fusiforme, widely consumed in Asian countries, has been proven to have various biological activities. However, the impacts and mechanisms of Sargassum fusiforme polysaccharides (SFPs) on intestinal bacterial infection are not yet fully understood. Our findings indicate that SFPs pretreatment ameliorates intestinal inflammation by reducing C. rodentium colonization, increasing colon length and levels of IL-10 and IL-22, decreasing IL-1β, IL-6, TNF-α, and IL-17 levels, inhibiting colonic crypt elongation and hyperplasia, and enhancing the intestinal mucosal barrier. The protective effects against intestinal bacterial infection are linked to enhanced clearance of C. rodentium and improvements in the intestinal mucosal barrier and C. rodentium-induced intestinal microbiota dysbiosis. Fecal microbiota transplantation experiments were conducted to evaluate the functional impact of microbiota induced by SFPs. The results suggest that intestinal microbiota modified by SFPs effectively countered C. rodentium infection. In addition, our study identified that miRNA-92a-3p is partially complementary to the 3'-UTR of the Notch1 gene, thereby repressing the Notch1-Hes1 signaling pathway and enhancing Muc2 secretion. Taken together, these findings reveal that SFPs protect mice from C. rodentium infection by activating the miR-92a-3p/Notch1-Hes1 regulatory axis driven by the intestinal microbiota, which stimulates Muc2 production to maintain intestinal barrier homeostasis.
Collapse
Affiliation(s)
- Ningbo Qin
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| | - Hongxu Liu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Xinru Wang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Yi Liu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Hong Chang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Xiaodong Xia
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
18
|
Hoseinifar SH, Arghideh M, Hosseinpour Delavar F, Paolucci M, Yazici M, Bayani M, Van Doan H. Incorporation of red macroalgae (Galaxaura oblongata) in roach (Rutilus rutilus) fingerling diet: Effects on growth, immunity, oxidative status and intestinal health. Comp Biochem Physiol B Biochem Mol Biol 2025; 277:111076. [PMID: 39894439 DOI: 10.1016/j.cbpb.2025.111076] [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: 06/23/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025]
Abstract
This study investigates the effects of the red macroalgae Galaxaura oblongata diet supplementation on roach (Rutilus rutilus) fingerlings growth and gene expression related to immunity, the intestinal barrier, and antioxidant status. Roach fingerlings (2.26 ± 0.04 g) were fed a basic diet supplemented with three different percentages of G. oblongata powder: 0.25 % (G1), 0.50 % (G2), and 1.0 % (G3) over 8 weeks, with a control group (C) receiving no supplementation. The study found that growth parameters significantly increased in the G1 and G2 groups compared to the C and G3 groups (P < 0.05). Immune responses, measured by total immunoglobulin (Ig) and lysozyme activity, showed a significant increase in the whole-body extract of the G2 group (P < 0.05) and the skin mucus of all treated groups compared to the control (P < 0.05). G. oblongata supplementation did not significantly affect catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx) activities in the whole-body extract, although GPx activity in skin mucus was significantly higher in the supplemented groups. Additionally, the gene expression of interleukin 1-beta (il-1β), occludin, gpx and sod, but not Toll-like receptor increased in G. oblongata treated groups. These results suggest that G. oblongata can serve as a beneficial feed additive in the culture of roach fingerlings, enhancing growth and immune function.
Collapse
Affiliation(s)
- Seyed Hossein Hoseinifar
- Department of Fisheries, Faculty of Fisheries and Environmental Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Mohammad Arghideh
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Institute of Aquaculture and Protection of Waters, Na Sádkách 1780, 370 05 České Budějovice, Czech Republic
| | - Fatemeh Hosseinpour Delavar
- Department of Fisheries, Faculty of Fisheries and Environmental Sciences, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Marina Paolucci
- Department of Science and Technology (DST), University of Sannio, 82100 Benevento, Italy.
| | - Metin Yazici
- Faculty of Marine Sciences and Technology, Iskenderun Technical University, Iskenderun 31200, Turkey.
| | - Mahsan Bayani
- Radin Makian Azma Mehr Ltd., Radinmehr Veterinary Laboratory, Gorgan, Iran
| | - Hien Van Doan
- Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai, Thailand; Functional Feed Innovation Center (FuncFeed), Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
19
|
Miao C, Wu Z, Wang M, Zhang B, Yu W, Li Y, Cao Z. Curcumin Alleviates DON-Induced Intestinal Epithelial Barrier Disruption by Improving Ribotoxic Stress-Associated p38 Pathway-Mediated TJ Injury, Apoptosis, and Cell Cycle Arrest. J Food Sci 2025; 90:e70217. [PMID: 40271829 DOI: 10.1111/1750-3841.70217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/19/2025] [Accepted: 04/04/2025] [Indexed: 04/25/2025]
Abstract
Deoxynivalenol (DON) is a pervasive ribotoxic stressor that induces intestinal epithelial barrier disruption by impairing tight junctions (TJs) and causing cellular damage. Curcumin (CUR), known for its enteroprotective properties and low toxicity, has been shown to attenuate DON-induced intestinal epithelial barrier injury. However, the underlying mechanisms are still unclear. In this study, we established in vivo and in vitro models using 30 male Kunming mice and IPEC-J2 cells to investigate the mechanisms by which CUR alleviates DON-induced intestinal epithelial barrier injury. The results showed that CUR markedly reduced DON-induced increases in intestinal permeability by restoring TJ protein expression (Claudin-4 and occludin) and preventing fiber-shaped actin (F-actin) contraction. CUR also attenuated DON-induced apoptosis by downregulating p53 and caspase activation and alleviated the G1 cell cycle arrest by reducing p21 expression. Mechanistically, CUR inhibited the activation of the ribosomal stress response (RSR)-associated p38 pathway, evidenced by decreased phosphorylation of p38, GSK3β, and ATF-2. The p38 activator dehydrocorydaline reversed CUR's protective effects. In conclusion, CUR alleviates DON-induced intestinal epithelial barrier disruption by improving RSR-associated p38 pathway-mediated TJ injury, apoptosis, and cell cycle arrest. These findings highlight the potential of CUR as a therapeutic agent for mitigating mycotoxin-induced intestinal dysfunction and suggest new avenues for drug target discovery.
Collapse
Affiliation(s)
- Chenjiao Miao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zuoyao Wu
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Mingyu Wang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Binwen Zhang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Wangyong Yu
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yanfei Li
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zheng Cao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
20
|
Song W, Chen J, Ai G, Xiong P, Song Q, Wei Q, Zou Z, Chen X. Mechanisms of the effects of turpiniae folium extract on growth performance, immunity, antioxidant activity and intestinal barrier function in LPS-challenged broilers. Poult Sci 2025; 104:104903. [PMID: 39985896 PMCID: PMC11904579 DOI: 10.1016/j.psj.2025.104903] [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: 11/23/2024] [Revised: 01/24/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025] Open
Abstract
Turpiniae folium extract (TFE) has shown anti-inflammatory and immunomodulatory effects in broilers. However, its mechanisms remain unclear. The aim of this study is to investigate the underlying mechanisms by which TFE influences growth performance, jejunal morphology, immune function, antioxidant capacity and barrier integrity in broilers challenged with Lipopolysaccharide (LPS). A total of 240 one-day-old female broilers were randomly divided into four groups with six replicates of ten birds each. A 2 × 2 factorial design with TFE (basal diets supplemented with 0 or 500 mg/kg TFE) and LPS challenge (intraperitoneal injection of 1 mg/kg body weight of sterile saline or LPS at 21, 23 and 25 days of age). The trial lasted for 26 days. The results showed that: Prior to the LPS challenge, dietary supplementation with TFE for 21 days increased both average daily gain (ADG) (P = 0.037) and average daily feed intake (ADFI) (P = 0.045) in broilers. During the LPS challenge period, LPS challenge led to a decline in growth performance and a negative impact on intestinal morphology, while TFE supplementation significantly reversed these adverse effects, as evidenced by increases in ADG (P = 0.004), ADFI (P = 0.046), jejunal villus height (VH) (P = 0.035), the villus height to crypt depth ratio (VH/CD) (P = 0.007) and decreases in the feed-to-gain ratio (F/G) (P = 0.025), jejunal crypt depth (CD) (P = 0.049). LPS induced inflammatory responses and oxidative stress in the jejunum, leading to a significant upregulation of pro-inflammatory factor gene and protein expression, and a marked downregulation of anti-inflammatory and antioxidant gene and protein expression. TFE supplementation mitigated these effects by yielding completely opposite results except for the expression of toll-like receptor 4 (TLR4) protein (P = 0.916). LPS negatively regulates the expression of genes and proteins involved in intestinal mucosal barrier function. In contrast, TFE supplementation significantly upregulated the expression of zonula occludens-1 (ZO-1) (P < 0.001) gene and ZO-1 (P < 0.001), occludin (OCLN) (P < 0.001), claudin (CLDN) (P < 0.001) proteins. In conclusion, dietary supplementation with TFE effectively counteracts the intestinal immune and oxidative stress induced by LPS challenge in broilers, improves intestinal mucosal barrier integrity and tissue morphology, and ultimately mitigates the negative impact of LPS on broiler growth performance. This effect may involve the modulation of the Nrf2 and nuclear factor kappa B (NF-κB) signaling pathways.
Collapse
Affiliation(s)
- Wenjing Song
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Jiang Chen
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Gaoxiang Ai
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Pingwen Xiong
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Qiongli Song
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Qipeng Wei
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Zhiheng Zou
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China
| | - Xiaolian Chen
- Institute of Animal Husbandry and Veterinary Science, Jiangxi Academy of Agricultural Sciences, Nanchang 330200, PR China; Jiangxi Province Key Laboratory of Animal Green and Healthy Breeding, Nanchang 330200, PR China.
| |
Collapse
|
21
|
Chen YP, Shi LL, Li YY, Zhang YM, Zhang SZ, Hou HD, Chen Y, Zhuo QH, Liu YQ, Wei BJ, Zhang LY. Isoliquiritigenin alleviates radiation-induced intestinal injury in lung cancer by inhibiting TNF-α/caspase3 signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04007-z. [PMID: 40137968 DOI: 10.1007/s00210-025-04007-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/02/2025] [Indexed: 03/29/2025]
Abstract
To construct a model of intestinal injury induced by radiotherapy for lung cancer and to study the protective effect and mechanism of isoliquiritigenin. The lungs of mice were irradiated with 0, 2, 4, 6, 8 Gy X-rays to screen the optimal radiation dose. A mouse model of lung cancer was established, and the tumor was irradiated once. At 3, 7, and 10 days after irradiation, H&E was used to detect the pathological manifestations of colon tissue in mice, and WB was used to detect the expression level of tight junction protein in colon tissue, so as to screen the best time point and study its possible mechanism. Molecular docking was used to study the tightness of isoliquiritigenin binding to TNF-α. Isoliquiritigenin (40 mg/kg) was given on the next day after 4 Gy X-ray irradiation. The levels of TNF-α and apoptosis, intestinal mucosal barrier function, MUC2 protein expression, and colon stem cell proliferation were detected. 4Gy X-ray local irradiation induced obvious colon injury in mice, and the injury was obvious on the 7th day. Isoliquiritigenin significantly improved the general condition, colonic histopathological changes, intestinal stem cell proliferation, and colonic tight junction function of lung cancer-bearing mice after radiotherapy. Further studies have found that isoliquiritigenin can downregulate the activation of TNF-α/Caspase-3 signaling pathway by inhibiting the expression of pro-inflammatory factor TNF-α, alleviate the apoptosis of colonic epithelial cells, improve the upregulation of colonic tight junction function, regulate the expression of MUC2, and promote the proliferation of intestinal stem cells, which may be related to the stable binding of isoliquiritigenin to TNF-α. Radiotherapy-induced bystander effect of lung cancer may be related to the abnormal expression of TNF-α. Isoliquiritigenin may downregulate the expression of TNF-α by binding to TNF-α, inhibit the apoptosis of colon cells, promote the proliferation of intestinal stem cells, and maintain the intestinal mucosal barrier to alleviate the colon injury induced by radiation bystander effect.
Collapse
Affiliation(s)
- Ya-Ping Chen
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities Gansu University of Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Liang-Liang Shi
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities Gansu University of Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yang-Yang Li
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities Gansu University of Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yi-Ming Zhang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Shang-Zu Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities Gansu University of Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Hong-Dou Hou
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities Gansu University of Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yan Chen
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities Gansu University of Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Qi-Hong Zhuo
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities Gansu University of Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yong-Qi Liu
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities Gansu University of Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Gansu university of Traditional Chinese Medicine, Lanzhou, China
| | - Ben-Jun Wei
- Key Laboratory of Dunhuang Medicine and Transformation at Provincial and Ministerial Level, Gansu university of Traditional Chinese Medicine, Lanzhou, China.
| | - Li-Ying Zhang
- Provincial-Level Key Laboratory for Molecular Medicine of Major Diseases and the Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and Universities Gansu University of Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China.
| |
Collapse
|
22
|
Zhang B, Tian M, Qiu Y, Wu J, Cui C, Liu S, Hou J, Tian C, Wang L, Gao K, Jiang Z, Yang X. Glucuronolactone Restores the Intestinal Barrier and Redox Balance Partly Through the Nrf2/Akt/FOXO1 Pathway to Alleviate Weaning Stress-Induced Intestinal Dysfunction in Piglets. Antioxidants (Basel) 2025; 14:352. [PMID: 40227425 PMCID: PMC11939252 DOI: 10.3390/antiox14030352] [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: 02/06/2025] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
(1) Background: Glucuronolactone (GLU) is a glucose metabolite with antioxidant activity. At present, the exact role of it in regulating the intestinal health of piglets under weaning stress is not clear. The purpose of this study is to investigate the effects of GLU on the growth performance and intestinal health of piglets under weaning stress and to explore potential mechanisms. (2) Methods: Twenty-four weaned piglets were randomly assigned into two groups, with one group receiving a basal diet and the other group receiving an experimental diet supplemented with 200 mg/kg of GLU. (3) Results: GLU increased the ADG, ADFI, and final body weight of piglets, while reducing the diarrhea rate. Mechanistically, GLU alleviates weaning stress-induced intestinal oxidative stress and inflammatory responses in piglets partly through activating the Nrf2-Akt signaling pathway to suppress the transcriptional activity of FOXO1, while also inhibiting the activation of the TLR4-MAPK signaling pathway to reduce the secretion of pro-inflammatory cytokines. Moreover, GLU increased the relative abundance of Lactobacillus reuteri in the ileum of piglets and improved the composition of the gut microbiota. (4) Conclusions: GLU reduced inflammation and oxidative stress through the Nrf2/Akt/FOXO1 signaling pathway and improved intestinal health, resulting in improved growth performance of the piglets.
Collapse
Affiliation(s)
- Beibei Zhang
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Min Tian
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Yueqin Qiu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Jing Wu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Chenbin Cui
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Shilong Liu
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Jing Hou
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Chaoyang Tian
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Li Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Kaiguo Gao
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Zongyong Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| | - Xuefen Yang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- State Key Laboratory of Swine and Poultry Breeding Industry, Guangzhou 510640, China
- Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, China
- Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou 510640, China
| |
Collapse
|
23
|
Muscia Saez V, Perdicaro DJ, Cremonini E, Costantino VV, Fontana AR, Oteiza PI, Vazquez Prieto MA. Grape pomace extract attenuates high fat diet-induced endotoxemia and liver steatosis in mice. Food Funct 2025; 16:2515-2529. [PMID: 40029158 DOI: 10.1039/d4fo06332e] [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: 03/05/2025]
Abstract
Obesity is a prominent global health concern associated with chronic inflammation and metabolic disorders, such as insulin resistance, type 2 diabetes, and non-alcoholic fatty liver disease (NAFLD). Excessive consumption of saturated fats exacerbates these conditions by increasing intestinal barrier permeability and circulating endotoxins. This study aims to investigate, in a murine model of high-fat diet (HFD)-induced obesity, the potential beneficial effects of a grape pomace extract (GPE), rich in phenolic compounds, at mitigating endotoxemia, and liver steatosis. Underlying mechanisms were characterized in an in vitro model of intestinal inflammation and permeabilization, as induced by tumor necrosis factor alpha (TNFα) in Caco-2 cell monolayers. Consumption of a HFD (60% calories from fat) for 13 weeks induced obesity, insulin resistance, and liver damage, evidenced by higher levels of plasma alanine aminotransferase (ALT), hepatic triglycerides content, and steatosis. In addition, HFD caused metabolic endotoxemia, hepatic toll-like receptor 4 (TLR4) upregulation and inflammation. GPE supplementation significantly reduced body weight and subcutaneous and visceral adipose tissue weight, and attenuated metabolic dysregulation. Furthermore, GPE decreased circulating LPS levels and mitigated HFD-mediated hepatic TLR4 upregulation, nuclear factor kappa B (NF-κB) activation, and downstream expression of proteins involved in oxidative stress and inflammation (NOX4, TNFα, and F4/80). In Caco-2 cells, GPE mitigated TNFα-induced monolayer permeabilization, decreased tight junction (TJ) protein levels, enhanced cellular oxidant production, activated redox-sensitive signaling, i.e., NF-κB and ERK1/2, and increased NOX1 and MLCK mRNA levels, the latter being a key regulator of monolayer permeability. The above findings suggest that GPE may protect against HFD-induced obesity and associated metabolic dysfunction (insulin resistance and NAFLD) by modulating intestinal barrier integrity and related endotoxemia.
Collapse
Affiliation(s)
- V Muscia Saez
- Laboratorio de Nutrición y Fisiopatología de la Obesidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo e Instituto de Medicina y Biología Experimental de Cuyo (IMBECU)-CONICET, M5502JMA, Mendoza, Argentina.
| | - D J Perdicaro
- Laboratorio de Nutrición y Fisiopatología de la Obesidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo e Instituto de Medicina y Biología Experimental de Cuyo (IMBECU)-CONICET, M5502JMA, Mendoza, Argentina.
| | - E Cremonini
- Departments of Nutrition and Environmental Toxicology, University of California, Davis, USA
| | - V V Costantino
- Laboratorio de Fisiopatología Renal, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo e Instituto de Medicina y Biología Experimental de Cuyo (IMBECU)-CONICET, Argentina
| | - A R Fontana
- Laboratorio de Bioquímica Vegetal, Instituto de Biología Agrícola de Mendoza (IBAM), Facultad de Ciencias Agrarias, CONICET-Universidad Nacional de Cuyo, M5528AHB, Chacras de Coria, Argentina
| | - P I Oteiza
- Departments of Nutrition and Environmental Toxicology, University of California, Davis, USA
| | - M A Vazquez Prieto
- Laboratorio de Nutrición y Fisiopatología de la Obesidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo e Instituto de Medicina y Biología Experimental de Cuyo (IMBECU)-CONICET, M5502JMA, Mendoza, Argentina.
| |
Collapse
|
24
|
Liu Y, Liu Q, Ma M, Zhang J, Liu M, Mosenthin R, Zhao L, Huang S, Ma Q. Dietary arabinogalactan modulates immunity and improves gut barrier in broilers via regulating metabolome and gut microbiome. Carbohydr Polym 2025; 352:123223. [PMID: 39843118 DOI: 10.1016/j.carbpol.2025.123223] [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: 09/28/2024] [Revised: 12/10/2024] [Accepted: 01/02/2025] [Indexed: 01/24/2025]
Abstract
The extraction of polysaccharides from wood by-products is recognized as a green re-utilization approach to shape a recycling-oriented society. In this research, we identified the structural properties of arabinogalactan (AG) extracted from Larix sibirica Ledeb wood chips and verified its efficacy as an additive in broiler framing. Results showed that the molecular weight of AG is 19.805 KDa. Methylation analysis and NMR spectra indicate that AG has a 1,6-linked Galp backbone, side residues mainly branched at C-1,3,6 on β-D-Galp. The Ara residues were substituted at C-3 of 1,6-linked Galp consisting of α-L-Araf-(1→3)-α-L-Araf-(1 → 3)-α-L-Araf (1→ and α-L-Araf-(1 → 4) β-D-Galp-(1 → 3)-β-D-Galp-(1→. As a dietary supplement in broiler model, AG treatment improved the body weight of broilers especially breast and leg muscle weight. Furthermore, AG could regulate host immune response, gut microbiota composition, and metabolic activity, especially promoting lipid metabolism. By means of serum non-targeted metabolomics analysis, enrichment of pantothenate and CoA biosynthesis and beta-alanine metabolism pathways could be determined. AG treatment led to a rise in bacteria that produce SCFAs, with elevated concentrations of acetic and butyric acids. In conclusion, AG can be considered as a potential dietary supplement to beneficially affect host's health status.
Collapse
Affiliation(s)
- Yafei Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Qingxiu Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Miaolin Ma
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jiatu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Meiling Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Rainer Mosenthin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; Institute of Animal Science, University of Hohenheim, 70593 Stuttgart, Germany
| | - Lihong Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Qiugang Ma
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
25
|
Supjaroen P, Niamsi W, Thummarati P, Laiwattanapaisal W. An In Vitro Cell Model of Intestinal Barrier Function Using a Low-Cost 3D-Printed Transwell Device and Paper-Based Cell Membrane. Int J Mol Sci 2025; 26:2524. [PMID: 40141167 PMCID: PMC11941856 DOI: 10.3390/ijms26062524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Current in vitro methods for intestinal barrier assessment predominantly utilize two-dimensional (2D) membrane inserts in standard culture plates, which are widely recognized for their inability to replicate the microenvironment critical to intestinal barrier functionality. Our study focuses on creating an alternative method for intestinal barrier function by integrating a 3D-printed transwell device with a paper-based membrane. Caco-2 cells were grown on a Matrigel-modified paper membrane, in which the tight junction formation was evaluated using TEER measurements. Neutrophil-like dHL-60 cells were employed for neutrophil extracellular trap (NET) formation experiments. Furthermore, intestinal barrier dysfunction was demonstrated using NET-isolated and Staurosporine interventions. Intestinal barrier characteristics were investigated through immunofluorescence staining of specific proteins and scanning electron microscopy (SEM). Our paper-based intestinal barrier exhibited an increased resistance in a time-dependent manner, consistent with immunofluorescence images of Zonulin Occludens-1 (ZO-1) expression. Interestingly, immunofluorescence analysis revealed changes in the morphology of the intestinal barrier and the formation of surface villi. These disruptions were found to alter the localization of tight junctions, impacting epithelial polarization and surface functionality. Moreover, we successfully demonstrated the permeability of a paper-based intestinal barrier using FITC-dextran assay. Hence, the 3D-printed transwell device integrated with a paper membrane insert presents a straightforward, cost-effective, and sustainable platform for an in vitro cell model to evaluate intestinal barrier function.
Collapse
Affiliation(s)
- Pitaksit Supjaroen
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (P.S.)
| | - Wisanu Niamsi
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (P.S.)
| | - Parichut Thummarati
- Centre of Excellence for Biosensors and Bioengineering (CEBB), Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wanida Laiwattanapaisal
- Centre of Excellence for Biosensors and Bioengineering (CEBB), Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
26
|
Čuljak N, Bendelja K, Leboš Pavunc A, Butorac K, Banić M, Savić Mlakar A, Cvetić Ž, Hrsan J, Novak J, Šušković J, Kos B. In Vitro Analysis of Probiotic Properties Related to the Adaptation of Levilactobacillus brevis to Intestinal Microenvironment and Involvement of S-Layer Proteins. Int J Mol Sci 2025; 26:2425. [PMID: 40141069 PMCID: PMC11942123 DOI: 10.3390/ijms26062425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Although rare, the ability to produce surface S-layer proteins is beneficially associated with particular Lactobacillus strains being investigated as probiotics. Therefore, this work aimed to study specific probiotic functionalities of selected Levilactobacillus brevis strains MB1, MB2, MB13 and MB20, isolated from human milk microbiota, and to assess the contribution of S-proteins. Firstly, Rapid Annotation using Subsystem Technology revealed that cell wall-related genes were abundant in analysed L. brevis genomes. Furthermore, the results demonstrated that S-proteins mediate aggregation capacity and competitive exclusion of selected pathogens by L. brevis strains. The improvement of Caco-2 epithelial monolayer barrier function was demonstrated by the increase in JAM-A and occludin expressions when L. brevis strains or S-proteins were added, with the effect being most pronounced after treatment with MB2 and S-proteins of MB1. L. brevis strains, especially MB20, exerted the potential to adhere to recombinant human ZG16. Strain MB2 and MB20-S-proteins improved the barrier function of HT29 epithelial monolayer, as evidenced by increased ZG16 expression. Analysed L. brevis strains and S-proteins differentially affected the protein expression of IL-1β, IL-6 and IL-8, and IL-10 cytokines. The most prominent effect was observed by S-proteins of MB20, since IL-1β production was decreased while IL-10 production was significantly increased.
Collapse
Affiliation(s)
- Nina Čuljak
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Krešo Bendelja
- Center for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, 10000 Zagreb, Croatia; (K.B.); (A.S.M.); (Ž.C.)
| | - Andreja Leboš Pavunc
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Katarina Butorac
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Martina Banić
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Ana Savić Mlakar
- Center for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, 10000 Zagreb, Croatia; (K.B.); (A.S.M.); (Ž.C.)
| | - Željko Cvetić
- Center for Research and Knowledge Transfer in Biotechnology, University of Zagreb, Rockefellerova 10, 10000 Zagreb, Croatia; (K.B.); (A.S.M.); (Ž.C.)
| | - Jana Hrsan
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Jasna Novak
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Jagoda Šušković
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| | - Blaženka Kos
- Department of Biochemical Engineering, Faculty of Food Technology and Biotechnology, University of Zagreb, Pierottijeva 6, 10000 Zagreb, Croatia; (N.Č.); (A.L.P.); (K.B.); (M.B.); (J.H.); (J.Š.); (B.K.)
| |
Collapse
|
27
|
Zhang L, Huang D, Gu J, Liang H, Ren M. The Significant Enhancing Effect of Vitamin B 6-Fortified Feed on the Intestinal Digestive Efficiency, Immunity, and Antioxidant Defense Mechanisms of Juvenile Largemouth Bass ( Micropterus salmoides). Antioxidants (Basel) 2025; 14:313. [PMID: 40227288 PMCID: PMC11939574 DOI: 10.3390/antiox14030313] [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: 02/15/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 04/15/2025] Open
Abstract
A 12-week aquaculture trial was conducted to evaluate the effects of vitamin B6 on the intestinal health of largemouth bass (Micropterus salmoides). Six feeds with a vitamin B6 content of 2.03 (control group), 2.91, 3.30, 6.03, 9.53, and 21.79 mg/kg were prepared. The results were as follows. Regarding digestive efficiency, the 9.53 mg/kg group showed significantly higher activities of AMY, LPS, and TRY compared to the control group; the 6.03 mg/kg group exhibited increased AKP and Na+/K+ ATPase activities. Regarding immunity, the 6.03 mg/kg group had markedly higher relative expressions of zo-1 and occ than the control group; the 9.53 mg/kg group showed significantly higher relative expressions of il-10, tgf-β, igm, and cd83, while il-8 and tnf-α were notably lower, and nf-κb was noticeably decreased in 21.79 mg/kg group. For antioxidant capacity, the 6.03 mg/kg group had markedly higher activities of CAT, SOD, GSH-Px, and T-AOC levels, compared to the control group; the MDA level in the control group was markedly higher than in the other groups. The relative expressions of nrf2, cat, Cu-Zn sod, and gpx were highest in 9.53 mg/kg group and significantly higher than in the control group. In conclusion, an appropriate level of vitamin B6 in the feed is vital for protecting the intestinal health of largemouth bass.
Collapse
Affiliation(s)
- Leimin Zhang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Dongyu Huang
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Jiaze Gu
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Hualiang Liang
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| | - Mingchun Ren
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China
| |
Collapse
|
28
|
Yang S, Liu H, Liu Y. Advances in intestinal epithelium and gut microbiota interaction. Front Microbiol 2025; 16:1499202. [PMID: 40104591 PMCID: PMC11914147 DOI: 10.3389/fmicb.2025.1499202] [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: 09/20/2024] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
The intestinal epithelium represents a critical interface between the host and external environment, serving as the second largest surface area in the human body after the lungs. This dynamic barrier is sustained by specialized epithelial cell types and their complex interactions with the gut microbiota. This review comprehensively examines the recent advances in understanding the bidirectional communication between intestinal epithelial cells and the microbiome. We briefly highlight the role of various intestinal epithelial cell types, such as Paneth cells, goblet cells, and enteroendocrine cells, in maintaining intestinal homeostasis and barrier function. Gut microbiota-derived metabolites, particularly short-chain fatty acids and bile acids, influence epithelial cell function and intestinal barrier integrity. Additionally, we highlight emerging evidence of the sophisticated cooperation between different epithelial cell types, with special emphasis on the interaction between tuft cells and Paneth cells in maintaining microbial balance. Understanding these complex interactions has important implications for developing targeted therapeutic strategies for various gastrointestinal disorders, including inflammatory bowel disease, metabolic disorders, and colorectal cancer.
Collapse
Affiliation(s)
- Sen Yang
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Pediatrics, The Fifth Peoples Hospital of Chengdu, Chengdu, China
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Lu W, Wen J. Anti-Inflammatory Effects of Hydrogen Sulfide in Axes Between Gut and Other Organs. Antioxid Redox Signal 2025; 42:341-360. [PMID: 39655451 DOI: 10.1089/ars.2023.0531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Significance: Hydrogen sulfide (H2S), a ubiquitous small gaseous signaling molecule, plays a critical role in various diseases, such as inflammatory bowel disease (IBD), rheumatoid arthritis (RA), ischemic stroke, and myocardial infarction (MI) via reducing inflammation, inhibiting oxidative stress, and cell apoptosis. Recent Advances: Uncontrolled inflammation is closely related to pathological process of ischemic stroke, RA, MI, and IBD. Solid evidence has revealed the axes between gut and other organs like joint, brain, and heart, and indicated that H2S-mediated anti-inflammatory effect against IBD, RA, MI, and ischemic stroke might be related to regulating the functions of axes between gut and other organs. Critical Issues: We reviewed endogenous H2S biogenesis and the H2S-releasing donors, and revealed the anti-inflammatory effects of H2S in IBD, ischemic stroke, RA, and MI. Importantly, this review outlined the potential role of H2S in the gut-joint axis, gut-brain axis, and gut-heart axis as a gasotransmitter. Future Direction: The rate, location, and timing of H2S release from its donors determine its potential success or failure as a useful therapeutic agent and should be focused on in the future research. Therefore, there is still a need to explore internal and external sources monitoring and controlling H2S concentration. Moreover, more efficient H2S-releasing compounds are needed; a better understanding of their chemistry and properties should be further developed. Antioxid. Redox Signal. 42, 341-360.
Collapse
Affiliation(s)
- Weizhuo Lu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
30
|
Qiu C, Chen Y, Xia H, Duan J, Zhang L, Zhang Y, Chen Z, Zhang L. Hsa_circ_0004662 Accelerates the Progression of Ulcerative Colitis via the microRNA-532/HMGB3 Signalling Axis. J Cell Mol Med 2025; 29:e70430. [PMID: 40099942 PMCID: PMC11916553 DOI: 10.1111/jcmm.70430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/18/2024] [Accepted: 02/04/2025] [Indexed: 03/20/2025] Open
Abstract
Increasing research has indicated that circular RNAs (circRNAs) are crucial for the development of ulcerative colitis (UC). Thus, we attempted to identify the role of hsa_circ_0004662 in UC progression. Hsa_circ_0004662 expression was determined via qRT-PCR. Lipopolysaccharide (LPS)-induced inflammation in normal colonic epithelial cells (ECs). The hsa_circ_0004662 content was then assessed in a mucosal inflammatory bowel disease (IBD) model. Cell proliferation was examined via CCK-8 and EdU uptake assays. Apoptotic rates were analysed via flow cytometry. The protein content was quantified via Western blotting. Enzyme-linked immunosorbent assay kits were used to detect IL-1β, TNF-α and IL-6, and dual-luciferase reporter (DLR) assays were used to identify interactions between miR-532 and circ_0004662 or HMGB3. An animal model of UC was also developed for confirmation. In this study, we identified the function of hsa_circ_0004662 in promoting UC progression. Hsa_circ_0004662 was upregulated in clinical UC tissues and LPS-induced colonic ECs, and its knockdown inhibited apoptosis, reduced inflammatory cytokine release and promoted cell proliferation in vitro. Mechanistically, hsa_circ_0004662 acted as a molecular sponge for miR-532, which targets HMGB3. The hsa_circ_0004662/miR-532/HMGB3 axis was further validated in a DSS-induced colitis mouse model, where hsa_circ_0004662 knockdown attenuated inflammation and tissue damage. These findings suggested that hsa_circ_0004662 contributes to UC progression through the miR-532/HMGB3 signalling pathway, offering potential targets for UC therapy.
Collapse
Affiliation(s)
- Chunhua Qiu
- Department of Gastroenterology, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Yun Chen
- Department of Geriatric Gastroenterology, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Huan Xia
- Geriatrics Research Institute, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Jun Duan
- Department of Geriatric Gastroenterology, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Lu Zhang
- Department of Geriatric Gastroenterology, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - You Zhang
- Department of Geriatric Gastroenterology, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Ziyang Chen
- Department of Gastroenterology, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Li Zhang
- Department of Geriatric Gastroenterology, Sichuan Provincial People's Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
31
|
Tadese DA, Mwangi J, Luo L, Zhang H, Huang X, Michira BB, Zhou S, Kamau PM, Lu Q, Lai R. The microbiome's influence on obesity: mechanisms and therapeutic potential. SCIENCE CHINA. LIFE SCIENCES 2025; 68:657-672. [PMID: 39617855 DOI: 10.1007/s11427-024-2759-3] [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: 06/21/2024] [Accepted: 10/16/2024] [Indexed: 01/03/2025]
Abstract
In 2023, the World Obesity Atlas Federation concluded that more than 50% of the world's population would be overweight or obese within the next 12 years. At the heart of this epidemic lies the gut microbiota, a complex ecosystem that profoundly influences obesity-related metabolic health. Its multifaced role encompasses energy harvesting, inflammation, satiety signaling, gut barrier function, gut-brain communication, and adipose tissue homeostasis. Recognizing the complexities of the cross-talk between host physiology and gut microbiota is crucial for developing cutting-edge, microbiome-targeted therapies to address the global obesity crisis and its alarming health and economic repercussions. This narrative review analyzed the current state of knowledge, illuminating emerging research areas and their implications for leveraging gut microbial manipulations as therapeutic strategies to prevent and treat obesity and related disorders in humans. By elucidating the complex relationship between gut microflora and obesity, we aim to contribute to the growing body of knowledge underpinning this critical field, potentially paving the way for novel interventions to combat the worldwide obesity epidemic.
Collapse
Affiliation(s)
- Dawit Adisu Tadese
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - James Mwangi
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lei Luo
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Zhang
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Xiaoshan Huang
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Brenda B Michira
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shengwen Zhou
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peter Muiruri Kamau
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiumin Lu
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Ren Lai
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
32
|
Zhang M, Zhao F, Guo M, Duan M, Xie Y, Qiu L. Vitamin E alleviates zebrafish intestinal damage and microbial disturbances caused by pyraclostrobin. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2025; 208:106221. [PMID: 40015832 DOI: 10.1016/j.pestbp.2024.106221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/26/2024] [Accepted: 11/23/2024] [Indexed: 03/01/2025]
Abstract
Pyraclostrobin (PY) is highly toxic to aquatic organisms, and its increased residues in aquatic environments may have harmful effects on the intestine of aquatic creatures. Previous research demonstrated that vitamin E (VE) alleviated the acute toxicity of PY to zebrafish. This study further explored the mitigation effect of VE on PY-induced intestinal toxicity in fish and the underlying mechanisms by exposing adult zebrafish to PY (10, 20 μg/L) with or without 4 μM VE supplementation for 21 days. The results showed that VE alleviated the gut histopathological lesions caused by PY. VE co-exposure also improved PY-induced intestinal inflammation and restored the expression level of genes encoding intestinal tight junction protein. Furthermore, VE restored the anti-oxidation level inhibited by PY and reduced pro-apoptotic cytokine level and apoptotic enzyme activity increased by PY. 16S rRNA high-throughput sequencing showed that VE improved the zebrafish intestinal flora imbalance caused by 20 μg/L PY, increased the relative abundance of beneficial bacterium Cetobacterium, and reduced the relative abundance of pathogenic bacteria. In conclusion, VE alleviated PY-induced intestinal toxicity via repairing the damaged intestinal mucosal barrier, inhibiting inflammation, reducing oxidative stress and apoptosis, and improving the intestinal microbial disorder in zebrafish.
Collapse
Affiliation(s)
- Mengna Zhang
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Feng Zhao
- College of Agriculture, Guangxi University, Nanning, Guangxi 530004, China
| | - Mengyu Guo
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Manman Duan
- Institute of Rural Revitalization, Dezhou University, Dezhou, Shandong 253023, China
| | - Yao Xie
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Lihong Qiu
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
33
|
Gao A, Lv J, Su Y. The Inflammatory Mechanism of Parkinson's Disease: Gut Microbiota Metabolites Affect the Development of the Disease Through the Gut-Brain Axis. Brain Sci 2025; 15:159. [PMID: 40002492 PMCID: PMC11853208 DOI: 10.3390/brainsci15020159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Parkinson's disease is recognized as the second most prevalent neurodegenerative disorder globally, with its incidence rate projected to increase alongside ongoing population growth. However, the precise etiology of Parkinson's disease remains elusive. This article explores the inflammatory mechanisms linking gut microbiota to Parkinson's disease, emphasizing alterations in gut microbiota and their metabolites that influence the disease's progression through the bidirectional transmission of inflammatory signals along the gut-brain axis. Building on this mechanistic framework, this article further discusses research methodologies and treatment strategies focused on gut microbiota metabolites, including metabolomics detection techniques, animal model investigations, and therapeutic approaches such as dietary interventions, probiotic treatments, and fecal transplantation. Ultimately, this article aims to elucidate the relationship between gut microbiota metabolites and the inflammatory mechanisms underlying Parkinson's disease, thereby paving the way for novel avenues in the research and treatment of this condition.
Collapse
Affiliation(s)
| | | | - Yanwei Su
- Department of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (A.G.); (J.L.)
| |
Collapse
|
34
|
Cheng Y, Lin S, Cao Z, Yu R, Fan Y, Chen J. The role of chronic low-grade inflammation in the development of sarcopenia: Advances in molecular mechanisms. Int Immunopharmacol 2025; 147:114056. [PMID: 39799736 DOI: 10.1016/j.intimp.2025.114056] [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: 06/24/2024] [Revised: 12/16/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
With the exacerbation of global population aging, sarcopenia has become an increasingly recognized public health issue. Sarcopenia, characterized by a progressive decline in skeletal muscle mass, strength, and function, significantly impacts the quality of life in the elderly. Herein, we explore the role of chroniclow-gradeinflammation in the development of sarcopenia and its underlying molecular mechanisms, including chronic inflammation-associated signaling pathways, immunosenescence, obesity and lipid infiltration, gut microbiota dysbiosis and intestinal barrier disruption, and the decline of satellite cells. The interplay and interaction of these molecular mechanisms provide new perspectives on the complexity of the pathogenesis of sarcopenia and offer a theoretical foundation for the development of future therapeutic strategies.
Collapse
Affiliation(s)
- Ying Cheng
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Shangjin Lin
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Ziyi Cao
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Runzhi Yu
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040 China
| | - Yongqian Fan
- Department of Orthopedics, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China.
| | - Jie Chen
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040 China.
| |
Collapse
|
35
|
Qin J, Liu Y, Cao M, Zhang Y, Bai G, Shi B. Bacillus subtilis MZ-01 alleviates diarrhea caused by ETEC K88 by reducing inflammation and promoting intestinal health. J Appl Microbiol 2025; 136:lxaf018. [PMID: 39821304 DOI: 10.1093/jambio/lxaf018] [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: 01/05/2024] [Revised: 08/19/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025]
Abstract
AIMS The purpose of this study was to investigate the effects of Bacillus subtilis supplementation on the health of weaned piglets and whether B. subtilis supplementation can reduce the damage of piglets induced by ETEC K88. METHODS AND RESULTS The experiment was designed with a 2 × 2 factorial arrangement, comprising the control group, B. subtilis (PRO) group, Escherichia coli K88 (ETEC) group, and B. subtilis + ETEC (PRO + ETEC) group. Regardless of the presence of ETEC, the addition of PRO increased the piglets' final body weight, average daily gain, and daily feed intake. Additionally, PRO primarily achieves a reduction in heat-stable enterotoxin (ST) levels, suppresses the expression of NF-κB, TLR4, and MyD88 mRNA in the jejunum and ileum, lowers pro-inflammatory factors in the blood and small intestine, enhances the expression of tight junction proteins in the small intestine, improves the composition of the colonic microbiota, increases colonic short-chain fatty acid contents, thereby alleviating diarrhea and mitigating bodily damage caused by ETEC K88 infection. CONCLUSION The addition of B. subtilis MZ-01 alleviated ETEC K88-induced piglet diarrhea by reducing ST levels, decreasing pro-inflammatory factors in the blood and intestine, and enhancing the intestinal barrier and tight junction proteins.
Collapse
Affiliation(s)
- Jianwei Qin
- College of Animal Science and Technology, Northeast Agricultural University, Changjiang Road, Harbin, 150030, PR China
| | - Yang Liu
- College of Animal Science and Technology, Northeast Agricultural University, Changjiang Road, Harbin, 150030, PR China
| | - Mingming Cao
- College of Animal Science and Technology, Northeast Agricultural University, Changjiang Road, Harbin, 150030, PR China
| | - Yue Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Changjiang Road, Harbin, 150030, PR China
| | - Guangdong Bai
- College of Animal Science and Technology, Northeast Agricultural University, Changjiang Road, Harbin, 150030, PR China
| | - Baoming Shi
- College of Animal Science and Technology, Northeast Agricultural University, Changjiang Road, Harbin, 150030, PR China
| |
Collapse
|
36
|
Xin J, He L, Li Y, Pu Q, Du X, Ban F, Han D. Sanguinarine chloride hydrate mitigates colitis symptoms in mice through the regulation of the intestinal microbiome and metabolism of short-chain fatty acids. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167579. [PMID: 39561858 DOI: 10.1016/j.bbadis.2024.167579] [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: 09/13/2024] [Revised: 10/21/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
Sanguinarine constitutes the main components of Macleaya cordata, and exhibits diverse biological and pharmacological activities. This study investigated the effects of sanguinarine chloride hydrate (SGCH) on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) mice. Five groups were designed to investigate the effects of SGCH on the pathological symptoms, the mRNA expression levels of inflammatory cytokines, colonic mucosal barrier damage, microbiota composition, and SCFAs metabolism in UC mice. The administration of SGCH in DSS-induced UC mice resulted in the amelioration of pathological symptoms, as evidenced by an increase in body weight, a decrease in disease activity index score, elongation of colon length, reduction in spleen index, and improvement in colon injury. SGCH can regulate the expression of inflammatory cytokines (IL-6, TNF-α, IL-1β and IL-10) and tight junction proteins (ZO-1 and Occludin) associated with UC. SGCH exhibited a significant decrease in NF-κB P65 mRNA expression levels, accompanied by a significantly reduced protein level of NF-κB P-P65/P65. Further studies revealed SGCH effectively reversed the decrease in intestinal microbiota diversity induced by UC, thereby promoting the growth of beneficial bacteria such as Akkermansia, Alistipes, and norank_o_Clostridia_UCG-014. Correlation analysis demonstrated a positive association between butanoic acid, propanoic acid, isobutyric acid, isovaleric acid, valeric acid, hexanoic acid with Colidextribacter, while Coriobacteriaceae_UCG-002 exhibited a negative correlation with butanoic acid, acetic acid and propanoic acid. In conclusion, the administration of SGCH can ameliorate clinical symptoms in UC mice, regulate the expression of inflammatory cytokines and tight junction proteins, modulate intestinal microbiota metabolism and SCFAs production.
Collapse
Affiliation(s)
- Jige Xin
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Lin He
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Yanlin Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Qiqi Pu
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Xuan Du
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Fuze Ban
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Diangang Han
- Technology Center of Kunming Customs, Kunming 650200, China.
| |
Collapse
|
37
|
Feng BY, Chen PL, Yan L, Huang WF, Li CF, Yi LT, Xu GH. Long-term Pu-erh tea alleviates inflammatory bowel disease via the regulation of intestinal microbiota and maintaining the intestinal mucosal barrier. Food Sci Biotechnol 2025; 34:743-755. [PMID: 39958166 PMCID: PMC11822139 DOI: 10.1007/s10068-024-01696-9] [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: 04/19/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 02/18/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal condition with increasing global prevalence. Current therapies are limited, leading to exploration of novel treatments like Pu-erh tea, a fermented tea recognized for its health benefits. This study shows that long-term consumption of Pu-erh tea significantly reduces IBD symptoms in DSS-induced mice by moderating inflammation and enhancing oxidative responses in the colon. Pu-erh tea notably increases the abundance of specific gut microbiota, particularly enhancing Firmicutes, Bacteroidota, and Proteobacteria phyla, and raising levels of Lactobacillus and Muribaculaceae genera. Key species such as Lactobacillus johnsonii, Lactobacillus reuteri, and Lactobacillus murinus also showed increased abundance. Additionally, Pu-erh tea helps restore the integrity of the intestinal barrier. These findings highlight the potential of Pu-erh tea as a complementary dietary strategy for IBD, potentially improving disease management and patient outcomes through its effects on the intestinal microbiota and mucosal barrier. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-024-01696-9.
Collapse
Affiliation(s)
- Bi-Yun Feng
- Fujian University of Traditional Chinese Medicine College of Pharmacy, Fuzhou, 350108 Fujian People’s Republic of China
- Xiamen Medicine Research Institute, Xiamen, 361008 Fujian People’s Republic of China
| | - Pei-Lu Chen
- Fujian University of Traditional Chinese Medicine College of Pharmacy, Fuzhou, 350108 Fujian People’s Republic of China
- Xiamen Medicine Research Institute, Xiamen, 361008 Fujian People’s Republic of China
| | - Ling Yan
- Fujian University of Traditional Chinese Medicine College of Pharmacy, Fuzhou, 350108 Fujian People’s Republic of China
- Xiamen Medicine Research Institute, Xiamen, 361008 Fujian People’s Republic of China
| | - Wei-Feng Huang
- Department of Gastroenterology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003 Fujian People’s Republic of China
| | - Cheng-Fu Li
- Xiamen Hospital of Traditional Chinese Medicine, Xiamen, 361009 Fujian People’s Republic of China
| | - Li-Tao Yi
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021 Fujian People’s Republic of China
- Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen, 361021 Fujian People’s Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021 Fujian People’s Republic of China
| | - Guang-Hui Xu
- Fujian University of Traditional Chinese Medicine College of Pharmacy, Fuzhou, 350108 Fujian People’s Republic of China
- Xiamen Medicine Research Institute, Xiamen, 361008 Fujian People’s Republic of China
- Xiamen Key Laboratory of Natural Medicine Research and Development, Xiamen, 361021 Fujian People’s Republic of China
| |
Collapse
|
38
|
Sun S, Li Y, Li Y, Niu Y, Hu Z, Deng C, Chen Y, Hu B, Huang Y, Deng X. Delayed Administration of IGFBP7 Improved Bone Defect Healing via ZO-1 Dependent Vessel Stabilization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406965. [PMID: 39698844 PMCID: PMC11809352 DOI: 10.1002/advs.202406965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/25/2024] [Indexed: 12/20/2024]
Abstract
The vascular response following injury is pivotal for successful bone-defect repair but constitutes a major hurdle in the field of regenerative medicine. Throughout this process, vessel stabilization is crucial to provide an adequate nutrient supply and facilitate efficient waste removal. Therefore, this study investigated whether promoting vascular stabilization improves bone defect repair outcomes. The findings show that insulin-like growth factor-binding protein (IGFBP) 7 exhibits a novel biological function in attenuating vascular permeability and enhancing vascular wall integrity. The potential underlying mechanism involves the up-regulation of insulin-like growth factor 1 receptor (IGF1R) expression by IGFBP7 on endothelial cell membrane, followed by activation of the downstream PI3K/AKT signaling pathway and upregulated expression of the tight junction protein zonula occludens-1 (ZO-1). IGFBP7 delayed administration in mice with cranial defects significantly improved bone defect healing by increasing ZO-1 and CD31 co-localization within vessel walls and optimizing the perfusion function of the final vascular network. Furthermore, the application of the typical tight junction regulator AT1001 effectively promoted ZO-1-dependent vascular stabilization and facilitated bone defect repair. This study presents a new approach to enhance bone defect healing via vascular stabilization-targeted interventions and significantly advances the understanding of the complex interplay between osteogenesis and angiogenesis in bone defect healing.
Collapse
Affiliation(s)
- Shiyu Sun
- Department of General DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yao Li
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yuman Li
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Yuting Niu
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Zhewen Hu
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Chenyu Deng
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Department of OrthodonticsPeking University School and Hospital of StomatologyBeijing100081China
| | - Yiming Chen
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Bo Hu
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Ying Huang
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
| | - Xuliang Deng
- Department of Geriatric DentistryPeking University School and Hospital of StomatologyBeijing100081P. R. China
- National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental MaterialsBeijing100081P. R. China
| |
Collapse
|
39
|
Jeong YE, Shea K, Ford KA. Unraveling Caco-2 cells through functional and transcriptomic assessments. Regul Toxicol Pharmacol 2025; 156:105771. [PMID: 39761805 DOI: 10.1016/j.yrtph.2025.105771] [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: 06/06/2024] [Revised: 11/20/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
The static Caco-2 monolayer is an extensively utilized model for predicting the permeability of small molecules during the drug development process. While these cells can differentiate and develop key functional and morphological features that emulate human enterocytes, they do not fully replicate the complexity of human intestinal physiology. In this study, we investigated functional and morphological aspects of Caco-2 cells, alongside their transcriptomic profiles, with a particular emphasis on genes encoding drug-metabolizing enzymes and drug transporters. We found that Caco-2 cells not only established a robust and bio-relevant permeable intestinal barrier but also demonstrated functional maturity and differentiation in the intestinal epithelium, substantiated by the activities of important enzymes and an efflux transporter. However, our targeted gene expression analyses revealed that substantial disparities were found in mRNA transcript levels among Caco-2 cells and human biopsy samples. These findings highlight that, although Caco-2 cells are valuable for assessing the passive transport of drugs, their accuracy for predicting active transport or small intestinal drug metabolism is constrained by their transcriptomic divergence from human intestinal tissues. This study highlights the importance of understanding the Caco-2 model's inherent limitations and provides insights that could inform its appropriate application in drug development and regulatory decision-making.
Collapse
Affiliation(s)
- Ye Eun Jeong
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA.
| | - Katherine Shea
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA.
| | - Kevin A Ford
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The U.S. Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA.
| |
Collapse
|
40
|
Nie X, Zhao F, Yin Y, Lu Q, Dai Y, Wang R, Ji Y, Zhang H, Zhu C. The potential of supplementing compound organic trace elements at lower levels in Chinese yellow- feathered broiler diets, part II: Impacts on growth performance, gut health, intestinal microbiota, and fecal mineral excretion. Poult Sci 2025; 104:104797. [PMID: 39827692 PMCID: PMC11787591 DOI: 10.1016/j.psj.2025.104797] [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: 07/23/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
This study aimed to investigate the effects of reducing inorganic trace minerals (ITM) by supplementing compound organic trace minerals (OTM) chelates on growth performance, fecal mineral excretion, intestinal health, and cecal microbiota of yellow-feathered broilers. A total of 960 one day old male broilers were randomly assigned to 6 treatments, among which birds were fed with the basal diets (negative control, NC), or supplemented with 1,000 mg/kg (positive control, PC), 300, and 500 mg/kg ITM or OTM, respectively. Dietary supplementation of OTM significantly increased the average daily gain (ADG) during 22-53 d and 1-53 d, and reduced the fecal emissions of Fe, Cu, Zn, and Mn of Chinese yellow-feathered broilers (P < 0.05). Furthermore, the OTM300 group significantly reduced the crypt depth in the duodenum, and increased the ratio of villus height to crypt depth (V/C) in the duodenum and jejunum (P < 0.05). The mRNA expression of TGF-β, Bcl-2, CAT, and GPX4 as well as tight junction proteins (occludin, ZO-1, claudin-1, and claudin-5) in jejunum mucosa were significantly increased by compound OTM when comparing with ITM300 group (P < 0.05). Moreover, dietary compound OTM significantly changed the Chao1 index and β diversity index of cecal microbiota of Chinese yellow-feathered broilers. The abundances of Firmicutes (phylum), Eubacterium_coprostanoligenes_group (family) and Oscillibacter (genus) were increased, while the abundances of Bacteroidetes (phylum) and Rikenellaceae RC9 group (genus) were decreased by OTM treatment. Spearman correlation analysis showed that the mRNA of occludin and jejunal V/C ratio were positively correlated with the abundance of Firmicutes (phylum), but negatively correlated with the abundance of Bacteroidota (phylum). In addition, the abundance of Eubacterium_coprostanoligenes_group (family) was positively correlated with the mRNA of claudin-1, Bcl-2, and TGF-β. PICRUST prediction of microbial function revealed that OTM treatment enriched the pathways related to amino acid metabolism and DNA replication. In conclusion, dietary supplementation at lower levels of compound OTM to replace ITM could improve growth performance and intestinal health, and reduce the fecal excretion of trace elements by modulation of cecal microbiota community and diversity in Chinese yellow-feathered broilers.
Collapse
Affiliation(s)
- Xiaoyan Nie
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Fei Zhao
- Hunan DeBon Bio-Tech Co., Ltd., Hengyang 421500, China
| | - Yucheng Yin
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Qi Lu
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Yang Dai
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Rui Wang
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Yiwen Ji
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Huihua Zhang
- School of Animal Science and Technology, Foshan University, Foshan 528225, China
| | - Cui Zhu
- School of Animal Science and Technology, Foshan University, Foshan 528225, China.
| |
Collapse
|
41
|
Wang J, Zhang Z, Wang J, Shi L, Wang S, Niu B, Tian X, Lv Q, Wei L, Li M, Liu Y. Bacillus coagulans alleviates intestinal barrier injury induced by Klebsiella pneumoniae in rabbits by regulating the TLR4/MyD88/NF-κB signalling pathway. Vet Microbiol 2025; 301:110364. [PMID: 39755051 DOI: 10.1016/j.vetmic.2024.110364] [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: 09/22/2024] [Revised: 12/16/2024] [Accepted: 12/29/2024] [Indexed: 01/06/2025]
Abstract
Probiotics effectively alleviate host diarrhoea, but the specific mechanism is not clear. Therefore, we explored the protective mechanism of Bacillus coagulans (BC) on intestinal barrier injury induced by Klebsiella pneumoniae (K. pneumoniae) in rabbits by HE, immunofluorescence and 16S rRNA. The results showed that BC pretreatment alleviated the changes in average daily gain, average daily feed intake and FCR caused by K. pneumoniae in rabbits. Moreover, BC alleviated the inflammatory cell infiltration, intestinal villus reduction, crypt deepening and goblet cell reduction caused by K. pneumoniae in rabbits. Further research revealed that BC improved the intestinal barrier by improving the mechanical barrier, chemical barrier, immune barrier and microbial barrier. Specifically, BC improved the intestinal mechanical barrier by improving the intestinal structure, increasing the protein expression of PCNA, increasing the number of goblet cells, and altering the expression of occludin, claudin-1 and ZO-1. BC improved the intestinal chemical barrier by regulating the expression of MUC1 and MUC2 and inhibited the TLR4/MyD88/NF-κB signalling pathway by altering the expression levels of the inflammatory factors IL-1β, IL-6 and TNF-α, thus optimizing the intestinal immune barrier. In addition, adding BC to the diet improved the intestinal microbial barrier of rabbits by reducing the abundance of harmful bacteria and increasing the abundance of beneficial bacteria. In summary, BC protects against K. pneumoniae-induced intestinal barrier damage by improving intestinal morphology, mitigating the inflammatory response and regulating the microbial composition. Among the pretreatments, the pretreatment effect of 1 × 106 CFU/g was the best. This study provides a theoretical basis for the use of BC to prevent and treat diarrhoea caused by K. pneumoniae in rabbits.
Collapse
Affiliation(s)
- Jianing Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Ziqiang Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Jiajia Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Lihui Shi
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Shuaishuai Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Bingyu Niu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Xiaonuo Tian
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Qiongxia Lv
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Lan Wei
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Mengyun Li
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China
| | - Yumei Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan 471000, China.
| |
Collapse
|
42
|
Hu D, Li X, Duan X, Yang L, Luo B, Wang L, Hu Z, Zhou Y, Qian P. Recombinant Saccharomyces cerevisiae EBY100/pYD1-FaeG: a candidate for an oral subunit vaccine against F4+ ETEC infection. Appl Environ Microbiol 2025; 91:e0181724. [PMID: 39601541 PMCID: PMC11784076 DOI: 10.1128/aem.01817-24] [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: 09/11/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Diarrheal diseases attributable to multidrug-resistant F4+ enterotoxigenic Escherichia coli (ETEC) are escalating in severity, posing significant risks to the health and safety of both humans and animals. This study used Saccharomyces cerevisiae EBY100 to display the FaeG subunit of F4 colonizing factor as an oral vaccine against F4+ ETEC infection. Mice were orally immunized twice with 108 CFU of EBY100/pYD1-FaeG, followed by a challenge with F4+ ETEC EC6 on day 7 post-immunization. The results showed that the recombinant strain EBY100/pYD1-FaeG orally enhanced the growth of the small intestine villi, significantly boosted the expression of tight junction proteins (ZO-1, Occludin, MUC2, and Claudin) (P < 0.05), and modulated the gut microbiota composition. Additionally, immunization with EBY100/pYD1-FaeG also upregulated the levels of IL-2, IL-4, and IFN-γ in the intestines of mice (P < 0.01), while serum IgG and fecal sIgA titer significantly increased (P < 0.05). These immune responses enhanced the capacity to fight against ETEC, leading to an increased survival rate of mice and relieved damage to tissues and organs of mice infection. In summary, the study suggested that the recombinant Saccharomyces cerevisiae EBY100/pYD1-FaeG could effectively stimulate the immune response and generate specific antibodies against F4+ ETEC, showing its potential to serve as a subunit oral vaccine candidate for preventing F4+ ETEC infection.IMPORTANCEThe multidrug-resistant F4+ enterotoxigenic Escherichia coli (ETEC) strains are the primary clinical pathogens responsible for post-weaning diarrhea in piglets, resulting in substantial economic losses in the pig farming industry. In the study, we developed an oral vaccine candidate, Saccharomyces cerevisiae EBY100/pYD1-FaeG, to prevent diarrhea caused by multidrug-resistant F4+ ETEC. Oral administration of EBY100/pYD1-FaeG significantly enhanced immune responses, improved intestinal health, and provided protection against F4+ ETEC infection in mice. This approach offers a potential application prospect for preventing F4+ ETEC infections that lead to post-weaning diarrhea in clinical settings and provides a promising solution for addressing the growing threat of antibiotic resistance in bacterial pathogens.
Collapse
Affiliation(s)
- Dayue Hu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiangmin Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaochao Duan
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liuyue Yang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Baizhi Luo
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Linkang Wang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zihui Hu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yang Zhou
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ping Qian
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Centre for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
43
|
Chen Y, Sun W, Mei H, Zhu S. Partially hydrolyzed guar gum alleviates neurological deficits and gastrointestinal dysfunction in mice with traumatic brain injury. Neurosurg Rev 2025; 48:103. [PMID: 39883194 DOI: 10.1007/s10143-024-03161-x] [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: 10/31/2024] [Revised: 12/06/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025]
Abstract
Traumatic brain injury (TBI)-associated neuroinflammation and neurotoxicity can induce gastrointestinal dysfunction through the brain-gut axis. Partially hydrolyzed guar gum (PHGG) was demonstrated to exert beneficial health effects by altering gut microbiota and short-chain fatty acids (SCFAs) production. Our study aimed to explore the effects of PHGG on gastrointestinal dysfunction in TBI mouse models. Controlled cortical impact (CCI)-induced TBI mouse models were administrated with PHGG (600 mg/kg/d) for 21 consecutive days. Behavioral tests (modified neurological severity score and beam walk test) and Y‑maze assay were performed to evaluate neurological functions and cognitive impairment. Enzyme-linked immunosorbent assay, reverse transcription-quantitative polymerase chain reaction, and western blotting examined the levels of inflammatory cytokines, intestinal mucosal damage markers, intestinal tight junction proteins, and NLRP3 inflammasome-related molecules in the serum, cerebral cortex, and colon tissues. The histological changes in the cerebral cortex and colon tissues were observed through hematoxylin and eosin and Nissl staining. Liquid chromatography/mass spectrometry analyzed SCFA amounts in the cecum contents and bile acid levels in the serum. PHGG administration alleviated neurological deficits and cognitive perturbations, reduced neuroinflammation, and attenuated cortical tissue damage and neuron loss in TBI mice. PHGG ameliorated intestinal barrier impairment, upregulated intestinal production of SCFAs, and elevated serum bile acid levels in TBI mice. Besides, PHGG treatment repressed NLRP3 inflammasome activation in TBI mice. Overexpressing NLRP3 reversed the beneficial effects of PHGG against TBI in mice. PHGG ameliorates neuroinflammation and gastrointestinal dysfunction in TBI murine models by inhibiting NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Yao Chen
- Department of Infection Control, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu Province, 225300, China
| | - Wenbin Sun
- Department of Critical Care Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Zhou shan hui shui Community,199 Hailing South Road, Taizhou, Jiangsu Province, 225300, China
| | - Haifeng Mei
- Department of Critical Care Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Zhou shan hui shui Community,199 Hailing South Road, Taizhou, Jiangsu Province, 225300, China
| | - Shang Zhu
- Department of Critical Care Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Zhou shan hui shui Community,199 Hailing South Road, Taizhou, Jiangsu Province, 225300, China.
| |
Collapse
|
44
|
Aitkenhead R, Waldron M, Conway GE, Horner K, Heffernan SM. The Influence of Dietary Supplements on Exercise-Induced Gut Damage and Gastrointestinal Symptoms: A Systematic Review and Meta-Analysis. Nutrients 2025; 17:443. [PMID: 39940302 PMCID: PMC11820470 DOI: 10.3390/nu17030443] [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: 01/08/2025] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/14/2025] Open
Abstract
Endurance exercise, especially under heat stress, temporarily compromises the integrity of the intestinal barrier in healthy individuals. Consequently, there is growing interest in developing effective dietary strategies to alleviate exercise-induced gastrointestinal symptoms and gut damage. This meta-analysis investigated the effects of dietary supplements on mitigating these challenges. The search was performed in November 2024 following PRISMA guidelines, and 26 peer-reviewed studies were included across three meta-analyses: (1) gastrointestinal symptoms, (2) circulating intestinal fatty acid-binding protein (i-FABP), and (3) exercise performance. The moderating effect of variables was assessed via sub-group analysis and meta-regression. Overall, there was no pooled effect of supplement interventions on gastrointestinal symptoms (Hedges' g = 0.42, 95% CI -0.17: 1.02, p = 0.15), and probiotics had a moderate significant effect for gastrointestinal symptoms (Hedges' g = -0.62, 95% CI -1.01; 1.01, p = 0.05). There was a significant increase in i-FABP concentrations pre- to post exercise (∆ 106%; Hedges' g = 1.01, 95% CI 0.63; 1.38, p = 0.01). There were no pooled or sub-group differences for exercise performance for any supplements (p = 0.53). Moderate-to-large heterogeneity was observed across studies (I2 ≥ 58.6%), and candidate moderators (exercise duration, modality, and environmental temperature) had no significant effect on any outcomes (p > 0.05). A significant increase in circulating i-FABP during exercise was observed. However, when examining the effects of different supplement categories, although significance was observed for a select few supplements, the changes in i-FABP, gastrointestinal symptoms, and exercise performance were outside of clinical relevance. Although probiotics showed a moderate significant effect for gastrointestinal symptoms, the conflicting findings across studies may have been due to inadequate control of confounding variables across studies. Further research is required to assess the alternative dietary supplements' effects on gastrointestinal health and exercise performance, particularly under varied environmental conditions, where more rigorous control for cofounding factors is implemented.
Collapse
Affiliation(s)
- Robyn Aitkenhead
- A-STEM Centre, Faculty of Science and Engineering, Swansea University, Swansea SA1 8EN, UK; (R.A.); (S.M.H.)
| | - Mark Waldron
- A-STEM Centre, Faculty of Science and Engineering, Swansea University, Swansea SA1 8EN, UK; (R.A.); (S.M.H.)
- Welsh Institute of Performance Science, Swansea University, Swansea SA1 8EN, UK
- School of Health and Behavioral Sciences, University of the Sunshine Coast, Sippy Downs, QLD 4556, Australia
| | - Gillian E. Conway
- In Vitro Toxicology Group, Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK;
| | - Katy Horner
- School of Public Health, Physiotherapy and Sport Science, University College, Belfield, D04 V1W8 Dublin, Ireland;
| | - Shane M. Heffernan
- A-STEM Centre, Faculty of Science and Engineering, Swansea University, Swansea SA1 8EN, UK; (R.A.); (S.M.H.)
| |
Collapse
|
45
|
Ali M, Xu C, Wang M, Hina Q, Ji Y, Anwar S, Lu S, He Q, Qiu Y, Li K. Gut Barrier Dysfunction and Microbiota Variations in Cryptosporidiosis: A Comprehensive Review. Vet Sci 2025; 12:85. [PMID: 40005845 PMCID: PMC11861801 DOI: 10.3390/vetsci12020085] [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: 12/26/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Cryptosporidiosis is a zoonotic protozoan parasite-born disease, equally significant in both animals and humans, especially affecting immunocompromised individuals (e.g., AIDS patients) and neonates. The prime concerns of this review article are to demonstrate the disruption of the intestinal barrier and variations in the gut microbiome during cryptosporidiosis, and to explore host gut-parasite interactions that can lead to the development of novel therapeutics. The review concluded that the enteric barrier is particularly maintained by tight junction proteins (e.g., occludin, claudin, and ZO-1, etc.) and mucosal immunity, both of which are severely compromised during Cryptosporidium spp. infections, resulting in increased intestinal barrier permeability, inflammatory responses, diarrhea, and ultimately death in severe cases. Cryptosporidium-induced dysbiosis is characterized by reduced microbial diversity and richness, a shift from commensal to pathogenic bacteria, as evidenced by increased pro-inflammatory taxa like Proteobacteria, and reduced proportions of beneficial SCFAs producing bacteria, e.g., Firmicutes. Recent investigations have highlighted the interrelations between gut microbiota and epithelial barrier integrity, especially during cryptosporidiosis, demonstrating the modulations regarding tight junctions (TJs), immune reactions, and SCFA production, all of which are main players in alleviating this protozoal parasitic infection. This review comprehensively describes the fine details underlying these impairments, including autophagy-mediated TJs' degradation, inflammasome activation, and gut microbiome-driven alterations in metabolic pathways, providing the latest relevant, and well-organized piece of knowledge regarding intestinal barrier alterations and microbial shifts during cryptosporidiosis. This work emphasizes the future need for longitudinal studies and advanced sequencing techniques to understand host gut microbiota-parasite interactions, aiming to formulate innovative strategies to mitigate cryptosporidiosis.
Collapse
Affiliation(s)
- Munwar Ali
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Chang Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Mingyue Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Qazal Hina
- Department of Animal Nutrition, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Yaru Ji
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Subiha Anwar
- Department of Animal Husbandry, University of Agriculture, Faisalabad 38000, Pakistan
| | - Sijia Lu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Qing He
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yawei Qiu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Kun Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
46
|
Li B, Wang H, Wang M, Liang H, Hu T, Yang J, Li S, You X, Xia B, Yuan Y, Zou Y, Miao Y, Sun Y. Genome analysis of Bifidobacterium adolescentis and investigation of its effects on inflammation and intestinal barrier function. Front Microbiol 2025; 15:1496280. [PMID: 39911710 PMCID: PMC11794259 DOI: 10.3389/fmicb.2024.1496280] [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: 09/14/2024] [Accepted: 12/23/2024] [Indexed: 02/07/2025] Open
Abstract
Numerous studies have confirmed that gut microbiota is a key driver in the occurrence and progression of inflammatory bowel disease (IBD). Based on the bacterial collection constructed in our previous studies, we founded that Bifidobacterium adolescentis AF91-08b2A has the potential beneficial function. We designed cohort studies, genomic studies and animal experiments to further explore the probiotic function of Bifidobacterium adolescentis AF91-08b2A and its therapeutic effect on IBD. The depletion of B. adolescentis in individuals with IBD suggested its significance for intestinal health. Genomic analysis highlighted the probiotic attributes of B. adolescentis AF91-08b2A, including resistance to antibiotics and stress, and metabolic pathways related to energy and carbohydrate metabolism, which are likely to enhance its therapeutic efficacy. In DSS-induced mice colitis model, the strain significantly enhanced the disease activity index (DAI), curbed weight loss, and attenuated colonic damage. It effectively modulated the immune response by reducing the levels of pro-inflammatory cytokines such as IL-6, IL-1β, IL-17A, IFN-γ, and TNF-α, while promoting the secretion of anti-inflammatory cytokines like IL-4, IL-10, and TGF-β1. The restoration of tight junction proteins ZO-1, occludin, and claudin-2 by B. adolescentis AF91-08b2A demonstrated its capacity to safeguard the intestinal epithelial barrier. Collectively, our findings indicate B. adolescentis AF91-08b2A as a valuable therapeutic option for UC, with its multifaceted approach to reducing inflammation and fortifying the intestinal barrier.
Collapse
Affiliation(s)
- Bo Li
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
| | - Haoyu Wang
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Mengmeng Wang
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hewei Liang
- BGI Research, Shenzhen, China
- BGI Research, Wuhan, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, China
| | - Tongyuan Hu
- BGI Research, Shenzhen, China
- BGI Research, Wuhan, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, China
| | - Jinlong Yang
- BGI Research, Shenzhen, China
- BGI Research, Kunming, China
- College of Forensic Science, Xi’an Jiaotong University, Xi’an, China
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xinbi You
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
- Yunnan Geriatric Medical Center, Kunming, China
| | - Binbin Xia
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
- Yunnan Geriatric Medical Center, Kunming, China
| | - Yue Yuan
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
| | - Yuanqiang Zou
- BGI Research, Shenzhen, China
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, China
| | - Yinglei Miao
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
| | - Yang Sun
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Clinical Research Center for Digestive Diseases, Kunming, China
- Yunnan Geriatric Medical Center, Kunming, China
| |
Collapse
|
47
|
Nakamura A, Matsumoto M. Role of polyamines in intestinal mucosal barrier function. Semin Immunopathol 2025; 47:9. [PMID: 39836273 PMCID: PMC11750915 DOI: 10.1007/s00281-024-01035-4] [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: 04/08/2024] [Accepted: 12/27/2024] [Indexed: 01/22/2025]
Abstract
The intestinal epithelium is a rapidly self-renewing tissue; the rapid turnover prevents the invasion of pathogens and harmful components from the intestinal lumen, preventing inflammation and infectious diseases. Intestinal epithelial barrier function depends on the epithelial cell proliferation and junctions, as well as the state of the immune system in the lamina propria. Polyamines, particularly putrescine, spermidine, and spermine, are essential for many cell functions and play a crucial role in mammalian cellular homeostasis, such as that of cell growth, proliferation, differentiation, and maintenance, through multiple biological processes, including translation, transcription, and autophagy. Although the vital role of polyamines in normal intestinal epithelial cell growth and barrier function has been known since the 1980s, recent studies have provided new insights into this topic at the molecular level, such as eukaryotic initiation factor-5A hypusination and autophagy, with rapid advances in polyamine biology in normal cells using biological technologies. This review summarizes recent advances in our understanding of the role of polyamines in regulating normal, non-cancerous, intestinal epithelial barrier function, with a particular focus on intestinal epithelial renewal, cell junctions, and immune cell differentiation in the lamina propria.
Collapse
Affiliation(s)
- Atsuo Nakamura
- Dairy Science and Technology Institute, Kyodo Milk Industry Co. Ltd, 20-1 Hirai, Hinode-Machi, Nishitama-Gun, Tokyo, 190-0182, Japan
| | - Mitsuharu Matsumoto
- Dairy Science and Technology Institute, Kyodo Milk Industry Co. Ltd, 20-1 Hirai, Hinode-Machi, Nishitama-Gun, Tokyo, 190-0182, Japan.
| |
Collapse
|
48
|
Perruchot MH, Boudry G, Mayeur-Nickel F, Grondin M, Wiart-Letort S, Giblin L, Grundy MML. In Vitro Evaluation of Intestinal Barrier Function after Exposure to Digested Pea Ingredients─Food Matrix Effect. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:584-594. [PMID: 39681414 PMCID: PMC11726683 DOI: 10.1021/acs.jafc.4c09963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 12/18/2024]
Abstract
Dietary fibers (DF) are important components of human and animal diets. However, they can decrease protein digestibility and absorption and thus the nutritional value of a food. The aim of this study was to investigate how the form of delivery of pea DF impacted the integrity of the intestinal barrier and, thereby, the potential absorption of molecules. To this end, two pea flours, with either intact or ruptured cell walls, and two controls, pea fibers and pea protein, were digested in vitro and the digesta obtained applied onto a jejunum porcine cell line (IPEC-J2 cells). Cell viability and integrity were evaluated by transepithelial electrical resistance measurement, colorimetric assay (MTS), and immunohistochemistry for tight junction proteins. Additionally, the diffusion of FITC-dextran (FD4) and lucifer yellow (LY) through the epithelial cell monolayers was monitored. The digested pea samples did not alter the IPEC-J2 viability and permeability. For instance, no difference in the diffusion of molecules either FD4 or LY across the monolayers was observed between the different digesta and the control. Similarly, no effect was observed in ZO-1 labeling intensity compared to the control. This study demonstrated that intestinal integrity was maintained whether pea cell walls were intact or ruptured.
Collapse
Affiliation(s)
| | - Gaëlle Boudry
- NUMECAN,
INSERM, INRAE, Université de Rennes, Saint Gilles 35590, France
| | | | | | | | - Linda Giblin
- Teagasc
Food Research Centre, Moorepark, Fermoy, Co Cork P61 C996, Ireland
| | | |
Collapse
|
49
|
Sun X, Yun L, Xie K, Liu R, Ren X, Zeng B, Cao X, Li Z, Zhou G, Liu B, Peng L, Yuan L. Probiotic Bacillus pumilus LV149 enhances gut repair, modulates microbiota, and alters transcriptome in DSS-induced colitis mice. Front Microbiol 2025; 15:1507979. [PMID: 39845056 PMCID: PMC11753000 DOI: 10.3389/fmicb.2024.1507979] [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: 10/08/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
Purpose Gut microbiota dysbiosis significantly impacts ulcerative colitis (UC) progression and exacerbation. Probiotics show promise in UC management. This study evaluated the effects of different doses of Bacillus pumilus LV149, an aquatic-derived probiotic, on gut injury repair in male C57BL/6 mice with dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) and investigated the underlying mechanisms. Methods UC was induced by allowing mice free access to a 3% DSS solution for 7 days, with concurrent daily oral gavage of either a low (LV149-L, 1 × 108 CFU/day/mouse) or high (LV149-H, 1 × 109 CFU/day/mouse) dose of LV149. The effects were assessed through physiological parameters, intestinal barrier integrity, inflammation, gut microbiota composition, and transcriptomic changes. Results LV149 significantly improved pathological symptoms, including weight loss and disease activity index (DAI), and reduced colon shortening in a dose-dependent manner and inflammatory damage. The intervention also restored gut barrier function by upregulating mucins, goblet cell counts, and tight junction proteins (ZO-1, occludin, and claudin-1) in colonic tissue, along with reducing serum lipopolysaccharide (LPS) levels. Notably, only the LV149-H significantly decreased the expression of pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, while both doses increased the expression of the anti-inflammatory cytokine IL-10 in a dose-dependent in colonic tissue. LV149 further modulated the gut microbiota, increasing beneficial bacteria and reducing pathogenic populations. Transcriptomic analysis indicated that LV149-L may exert gut repair effects via the IL-17 signaling pathway, whereas LV149-H appears to act through the JAK-STAT signaling pathway. Conclusion This study demonstrated that LV149, particularly at a higher dose, effectively mitigated DSS-induced colonic injury by modulating gut microbiota, enhancing gut barrier integrity, and reducing inflammation. The dose-dependent effects underscored LV149-H's potential as a therapeutic agent for UC due to its stronger anti-inflammatory properties and gut-protective effects.
Collapse
Affiliation(s)
- Xinyu Sun
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Long Yun
- Huzhou Key Laboratory of Translational Medicine, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Keming Xie
- Medical College of Jiaying University, Jiaying University, Meizhou, China
| | - Renhui Liu
- School of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Xinyue Ren
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bokun Zeng
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xudong Cao
- Department of Chemical and Biological Engineering, University of Ottawa, Ottawa, ON, Canada
| | - Zhi Li
- Key Laboratory of Aquacultural Biotechnology Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, China
| | - Guihao Zhou
- Division of Medicine, University College London, London, United Kingdom
| | - Bang Liu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Luo Peng
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Lihong Yuan
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
50
|
Khaskheli AA, Niknafs S, Meijer MMY, Tan X, Ferket PR, Roura E. The in ovo screening of 27 single essential oils showed selective effects on hatchability, performance and gene expression relevant to gut functions in broilers at hatch. Poult Sci 2025; 104:104670. [PMID: 39693964 PMCID: PMC11720607 DOI: 10.1016/j.psj.2024.104670] [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: 09/14/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024] Open
Abstract
The early post-hatching phase remains to be one of the most vulnerable phases in broiler production. Some essential oils have been reported to improve gut health and growth in broiler chickens when applied to post-hatching diets. However, in-feed applications are unable to prevent the health challenges observed immediately after hatching. Thus, pre-hatch interventions need to be considered. A research project was developed with the aim of investigating the impact of in ovo application of 27 selected essential oils (EOs) on foetal development with emphasis on gut integrity in broiler hatchlings. The eggs were incubated under standard conditions until day 17.5, when 1 mL of each EO preparation (5 µL EO + 5 µL polysorbate-80 + 990 µL saline) was injected into the amnion. Hatchability, body weight and organ weights (residual yolk, gizzard-proventriculus, intestines, liver, and heart) were measured at hatch. Five essential oils eugenol, clove, tea tree, lemongrass, and thyme, significantly (P < 0.05) reduced hatchability (66.67 %, 58.33, 83.30 and 83.30 %) compared to the saline (96.80 %), were discarded from the rest of the study. The other 22 essential oils were investigated in a second phase to assess their impact on expression of gut biomarkers including: a) jejunum integrity; b) digestive enzymes and nutrient transporters; and c) immune system. The results indicated that lemon myrtle significantly increased and oregano EO decreased body weight at hatch (BW0) compared to the saline (P < 0.05). Ylang ylang, clary sage, bergamot, lemon myrtle, and black pepper upregulated the expressions of biomarkers regulating gut integrity and barrier functions (ZO-1, ZO-2, CLDN1, MARVELD2, EGFR and EGF), nutrients transporters (EAAT3, PEPT1, I-FABP1, SGLT1), and digestive enzymes (APN, SI). Ylang ylang, turmeric acid, star anise, clary sage, and black pepper upregulated the expression of gut immunity biomarkers IL1B, IL10, IGMH, CD3D, and BU1 compared to the saline. In conclusion, in ovo delivery of selected EOs has the potential to improve embryonic development relevant to nutrient digestion and absorption, gut integrity and immunity in broilers.
Collapse
Affiliation(s)
- Asad A Khaskheli
- Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Australia
| | - Shahram Niknafs
- Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Australia
| | - Mila M Y Meijer
- Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Australia
| | - Xinle Tan
- Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Australia
| | - Peter R Ferket
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, USA
| | - Eugeni Roura
- Queensland Alliance for Agriculture and Food Innovation (QAAFI), The University of Queensland, Australia.
| |
Collapse
|