1
|
Liu L, Zhao Y, Huang Z, Long Z, Qin H, Lin H, Zhou S, Kong L, Ma J, Lin Y, Li Z. Dietary supplementation of Lycium barbarum polysaccharides alleviates soybean meal-induced enteritis in spotted sea bass Lateolabrax maculatus. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:1-22. [PMID: 39949731 PMCID: PMC11815959 DOI: 10.1016/j.aninu.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/22/2024] [Accepted: 10/24/2024] [Indexed: 02/16/2025]
Abstract
The aim of this experiment was to investigate the effect of Lycium barbarum polysaccharides (LBP) on alleviating soybean meal-induced enteritis (SBMIE) in spotted sea bass Lateolabrax maculatus. The diet with 44% fishmeal (FM) content was used as a blank control, and soybean meal (SM) was used to replace 50% FM as an experimental control to induce enteritis. Then, on the basis of experimental control, 0.10%, 0.15%, and 0.20% LBP were added as experimental diets. A total of 225 spotted sea bass (44.52 ± 0.24 g) were randomly divided into 5 groups and fed the corresponding diets for 52 d. The results showed that 0.15% LBP decreased serum D-lactic acid (D-LA) content and diamine oxidase (DAO) activity (P < 0.05). In addition, in all LBP supplementation groups, the intestinal tissue morphology was significantly improved (P < 0.05); the intestinal microbial structure gradually recovered to a level close to that without adding SM; and the microbial species richness and diversity were significantly increased (P < 0.05). Through transcriptomic and metabolomic analysis, it was found that the expression of proinflammatory factors such as interleukin-1β (IL-1β), interleukin-12 (IL-12), nuclear factor kappa B subunit 2 (NF-κB2), and Toll-like receptor 2 (TLR2) were significantly down-regulated in the mitogen-activated protein kinase (MAPK) and Toll-like receptor signaling pathways (P < 0.05), and the important tight junction protein gene Occludin was up-regulated (P < 0.05). In addition, LBP down-regulated saponin metabolites and up-regulated amino acid metabolites (P < 0.05). In conclusion, LBP demonstrated a significant alleviating effect on SBMIE of spotted sea bass L. maculatus.
Collapse
Affiliation(s)
- Longhui Liu
- Fisheries College, Jimei University, Xiamen, China
- Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| | - Yanbo Zhao
- Fisheries College, Jimei University, Xiamen, China
- Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| | - Zhangfan Huang
- Fisheries College, Jimei University, Xiamen, China
- Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| | - Zhongying Long
- Fisheries College, Jimei University, Xiamen, China
- Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| | - Huihui Qin
- Fisheries College, Jimei University, Xiamen, China
- Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| | - Hao Lin
- Fisheries College, Jimei University, Xiamen, China
- Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| | - Sishun Zhou
- Fisheries College, Jimei University, Xiamen, China
- Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| | - Lumin Kong
- Fisheries College, Jimei University, Xiamen, China
- Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| | - Jianrong Ma
- Fisheries College, Jimei University, Xiamen, China
- Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| | - Yi Lin
- Fisheries College, Jimei University, Xiamen, China
- Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| | - Zhongbao Li
- Fisheries College, Jimei University, Xiamen, China
- Fujian Provincial Key Laboratory of Marine Fishery Resources and Eco-environment, Xiamen, China
| |
Collapse
|
2
|
Yu G, He J, Gao Z, Fu L, Zhang Q. Protein-bound AGEs derived from methylglyoxal induce pro-inflammatory response and barrier integrity damage in epithelial cells by disrupting the retinol metabolism. Food Funct 2024; 15:11650-11666. [PMID: 39523841 DOI: 10.1039/d4fo00364k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Advanced glycation end-products (AGEs) are complex and heterogeneous compounds widely present in processed foods. Previous studies evidenced the adverse effects of AGEs on gut homeostasis, but the precise pathological mechanisms and molecular pathways responsible for the disruption of intestinal barrier integrity by AGEs remain incompletely elucidated. In this study, protein-bound AGEs (BSA-MGO), the most common type of dietary AGE, were prepared by methylglyoxal-mediated glycation, and an in vitro human epithelial colorectal adenocarcinoma (Caco-2) cell model was employed to evaluate the impact of protein-bound AGEs on gut epithelial function. Results showed that exposure to BSA-MGO significantly increased the permeability of Caco-2 cell monolayers as evidenced by the decreased transepithelial electrical resistance value, increased paracellular transport of FITC-dextran, and down-regulated tight-junction proteins. In parallel, BSA-MGO induced pro-inflammatory responses and oxidative stress in the monolayers. Transcriptomic profiling further revealed that BSA-MGO disrupted the retinol metabolism, thereby contributing to the barrier integrity damage in epithelial cells. Overall, these results provide valuable insights into the disrupting effects of dietary AGEs on intestinal barrier function, and the perturbed pathways present potential targets for further exploration of the molecular mechanisms underlying the detrimental effect of processed foods on gut health.
Collapse
Affiliation(s)
- Gang Yu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, 18 Xue Zheng Street, Hangzhou, 310018, Zhejiang Province, China.
| | - Jianxin He
- Zhejiang Li Zi Yuan Food Co., Ltd, Jinhua, 321031, China
| | - Zhongshan Gao
- Allergy Research Center, Zhejiang University, Hangzhou, 310018, China
| | - Linglin Fu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, 18 Xue Zheng Street, Hangzhou, 310018, Zhejiang Province, China.
| | - Qiaozhi Zhang
- School of Food Science and Biotechnology, Zhejiang Gongshang University, 18 Xue Zheng Street, Hangzhou, 310018, Zhejiang Province, China.
| |
Collapse
|
3
|
Feng Y, Pan M, Li R, He W, Chen Y, Xu S, Chen H, Xu H, Lin Y. Recent developments and new directions in the use of natural products for the treatment of inflammatory bowel disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155812. [PMID: 38905845 DOI: 10.1016/j.phymed.2024.155812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/13/2024] [Accepted: 06/06/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) represents a significant global health challenge, and there is an urgent need to explore novel therapeutic interventions. Natural products have demonstrated highly promising effectiveness in the treatment of IBD. PURPOSE This study systematically reviews the latest research advancements in leveraging natural products for IBD treatment. METHODS This manuscript strictly adheres to the PRISMA guidelines. Relevant literature on the effects of natural products on IBD was retrieved from the PubMed, Web of Science and Cochrane Library databases using the search terms "natural product," "inflammatory bowel disease," "colitis," "metagenomics", "target identification", "drug delivery systems", "polyphenols," "alkaloids," "terpenoids," and so on. The retrieved data were then systematically summarized and reviewed. RESULTS This review assessed the different effects of various natural products, such as polyphenols, alkaloids, terpenoids, quinones, and others, in the treatment of IBD. While these natural products offer promising avenues for IBD management, they also face challenges in terms of clinical translation and drug discovery. The advent of metagenomics, single-cell sequencing, target identification techniques, drug delivery systems, and other cutting-edge technologies heralds a new era in overcoming these challenges. CONCLUSION This paper provides an overview of current research progress in utilizing natural products for the treatment of IBD, exploring how contemporary technological innovations can aid in discovering and harnessing bioactive natural products for the treatment of IBD.
Collapse
Affiliation(s)
- Yaqian Feng
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Mengting Pan
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Ruiqiong Li
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Weishen He
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yangyang Chen
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Shaohua Xu
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| | - Hui Chen
- Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, China.
| | - Huilong Xu
- Institute of Structural Pharmacology & TCM Chemical Biology, Fujian Key Laboratory of Chinese Materia Medica, College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| | - Yao Lin
- Fujian-Macao Science and Technology Cooperation Base of Traditional Chinese Medicine-Oriented Chronic Disease Prevention and Treatment, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| |
Collapse
|
4
|
Jia X, Huang Y, Liu G, Li Z, Tan Q, Zhong S. The Use of Polysaccharide AOP30 from the Rhizome of Alpinia officinarum Hance to Alleviate Lipopolysaccharide-Induced Intestinal Epithelial Barrier Dysfunction and Inflammation via the TLR4/NfκB Signaling Pathway in Caco-2 Cell Monolayers. Nutrients 2024; 16:2151. [PMID: 38999898 PMCID: PMC11243348 DOI: 10.3390/nu16132151] [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: 04/21/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/14/2024] Open
Abstract
Alpinia officinarum Hance is rich in carbohydrates and is flavored by natives. The polysaccharide fraction 30 is purified from the rhizome of A. officinarum Hance (AOP30) and shows excellent immunoregulatory ability when administered to regulate immunity. However, the effect of AOP30 on the intestinal epithelial barrier is not well understood. Therefore, the aim of this study is to investigate the protective effect of AOP30 on the intestinal epithelial barrier using a lipopolysaccharide (LPS)-induced intestinal epithelial barrier dysfunction model and further explore its underlying mechanisms. Cytotoxicity, transepithelial electrical resistance (TEER) values, and Fluorescein isothiocyanate (FITC)-dextran flux are measured. Simultaneously, the protein and mRNA levels of tight junction (TJ) proteins, including zonula occludens-1 (ZO-1), Occludin, and Claudin-1, are determined using Western blotting and reverse-transcription quantitative polymerase chain reaction methods, respectively. The results indicate that AOP30 restores the LPS-induced decrease in the TEER value and cell viability. Furthermore, it increases the mRNA and protein expression of ZO-1, Occludin, and Claudin-1. Notably, ZO-1 is the primary tight junction protein altered in response to LPS-induced intestinal epithelial dysfunction. Additionally, AOP30 downregulates the production of TNFα via the Toll-like receptor 4 (TLR4)/NF-κB signaling pathway. Collectively, the findings of this study indicate that AOP30 can be developed as a functional food ingredient or natural therapeutic agent for addressing intestinal epithelial barrier dysfunction. It sheds light on the role of AOP30 in improving intestinal epithelial function.
Collapse
Affiliation(s)
- Xuejing Jia
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yun Huang
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Guanghuo Liu
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zipeng Li
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Qiwei Tan
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Saiyi Zhong
- Guangdong Provincial Key Laboratory of Aquatic Products Processing and Safety, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Guangdong Provincial Engineering Technology Research Center of Seafood, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| |
Collapse
|
5
|
Li ZX, Zhuo JL, Yang N, Gao MB, Qu ZH, Han T. Effect of Lycium barbarum polysaccharide on osteoblast proliferation and differentiation in postmenopausal osteoporosis. Int J Biol Macromol 2024; 271:132415. [PMID: 38759858 DOI: 10.1016/j.ijbiomac.2024.132415] [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: 01/31/2024] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
OBJECTIVE We aimed to investigate the effect of Lycium barbarum polysaccharide (LBP) on the proliferation and differentiation of osteoblasts in postmenopausal individuals with osteoporosis using in vitro cell experiments. METHODS We assessed the effect of long-term LBP consumption on the intestinal metabolites of individuals using a simulation of the human intestinal microbiota ecosystem. We also tested the capacity of LBP in proliferating MC3T3-E1 cells using the cell counting kit-8 (CCK-8) method and analyzed the effect of intestinal metabolites on the osteogenic differentiation of MC3T3-E1 cells by testing bone metabolism viability with relevant indicators. RESULTS The level of short-chain fatty acids (SCFAs) significantly increased (p < 0.05), and the concentrations of acetic acid, propionic acid, and butyric acid all showed an upward trend after the treatment using LBP. At appropriate concentrations, the fermentation supernatant can enhance osteoblast proliferation by significantly increasing the active expression of bone-alkaline phosphatase (B-ALP) and osteocalcin (OCN) in osteoblasts (p < 0.05). CONCLUSION By modulating the metabolites of intestinal microbiota, production of SCFAs, the prebiotic properties of LBP can enhance osteoblast differentiation through in vitro simulation experiment and cell-based assay.
Collapse
Affiliation(s)
- Zi-Xiang Li
- Department of Clinical Nutrition, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jia-Lu Zhuo
- Department of Clinical Nutrition, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ning Yang
- Department of Clinical Nutrition, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ming-Bo Gao
- Department of Clinical Nutrition, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Zhi-Hua Qu
- Department of Clinical Nutrition, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ting Han
- Department of Clinical Nutrition, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| |
Collapse
|
6
|
Wang L, Song X, Zhou Y, Xia Y, Yang Z, Chen X, Shi R, Geng Z, Zhang X, Wang Y, Li J, Hu J, Zuo L. Sclareol protected against intestinal barrier dysfunction ameliorating Crohn's disease-like colitis via Nrf2/NF-B/MLCK signalling. Int Immunopharmacol 2024; 133:112140. [PMID: 38669952 DOI: 10.1016/j.intimp.2024.112140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Inflammation-induced intestinal barrier dysfunction is not only a pathological feature of Crohn's disease (CD) but also an important therapeutic target. Sclareol (SCL) is a nontoxic natural plant compound with anti-inflammatory effect, but its role in CD has not been established. METHODS In vivo studies of mice with TNBS-induced colitis were carried out to evaluate the effects of SCL on CD-like colitis and intestinal barrier function. In vitro, a TNF-α-induced colonic organoid model was established to test the direct effect of SCL on inflammation-induced intestinal barrier injure and inflammatory response. The Nrf2/NF-κB/MLCK signalling was analysed to explore the mechanism of SCL. RESULTS In vivo, SCL largely alleviated the colitis in TNBS mice, as evidenced by improvements in the weight loss, colitis symptoms, endoscopic score, macroscopic histological score, and histological inflammation score. Moreover, SCL significantly improved intestinal barrier dysfunction, manifested as reduced intestinal permeability and decreased intestinal bacterial translocation in TNBS mice. Importantly, SCL antagonised the intestinal mucosal inflammation while protecting tight junctions in TNBS mice. In vitro, SCL largely depressed pro-inflammatory cytokines levels and improved intestinal epithelial permeability in a TNF-α-induced colonic organoid model. In the context of CD, the protective effects of SCL against inflammation and intestinal barrier damage are at least partially results from the Nrf2 signalling activation and the NF-κB/MLCK signalling inhibition. CONCLUSIONS SCL improved intestinal barrier dysfunction and alleviated CD-like colitis, possibly through modulation of Nrf2/NF-κB/MLCK signalling. In view of SCL's safety profile, there is hope that it will be useful in the clinic.
Collapse
Affiliation(s)
- Lian Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Xue Song
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Yueqing Zhou
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Yongsheng Xia
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Zi Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Xiaohua Chen
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Ruohan Shi
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Zhijun Geng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Xiaofeng Zhang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Yueyue Wang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Jing Li
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China
| | - Jianguo Hu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China.
| | - Lugen Zuo
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, Anhui, China.
| |
Collapse
|
7
|
Li R, Yang P, Liu B, Ye Z, Zhang P, Li M, Gong Y, Huang Y, Yang L, Li M. Lycium barbarum polysaccharide remodels colon inflammatory microenvironment and improves gut health. Heliyon 2024; 10:e30594. [PMID: 38774318 PMCID: PMC11107222 DOI: 10.1016/j.heliyon.2024.e30594] [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: 07/17/2023] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/24/2024] Open
Abstract
Aim Disturbed intestinal microbiota has been implicated in the inflammatory microenvironment of the colon, which usually results in ulcerative colitis (UC). Given the limitations of these drugs, it is important to explore alternative means of protecting the gut health from UC. This study aimed to investigate the potential of polysaccharides as beneficial nutrients in the regulation of the gut microbiota, which determines the inflammatory microenvironment of the colon. Materials and methods Mice were treated with dextran sulfate sodium (DSS) to evaluate the effects and mechanisms of Lycium barbarum polysaccharide (LBP) in remodeling the inflammatory microenvironment and improving gut health. Body weight and disease activity indices were monitored daily. Hematoxylin and eosin staining was used to analyze colon dynamics. The levels of inflammatory indicators and expression of MUC-2, claudin-1, ZO-1, and G-protein-coupled receptor 5 (TGR5) were determined using assay kits and immunohistochemistry, respectively. 16S rRNA high-throughput sequencing of the intestinal microbiota and liquid chromatography-tandem mass spectrometry for related bile acids were used. Results LBP significantly improved the colonic tissue structure by upregulating MUC-2, claudin-1, and ZO-1 protein expression. The bacterial genus Dubosiella was dominant in healthy mice, but significantly decreased in mice treated with DSS. LBP rehabilitated Dubosiella in the sick guts of DSS mice to a level close to that of healthy mice. The levels of other beneficial bacterial genera Akkermansia and Bifidobacterium were also increased, whereas those of the harmful bacterial genera Turicibacter, Clostridium_sensu_stricto_1, Escherichia-Shigella, and Faecalibaculum decreased. The activity of beneficial bacteria promoted the bile acids lithocholic and deoxycholic acids in mice with UC, which improved the gut barrier function through the upregulation of TGR5. Conclusion The inflammatory microenvironment in the gut is determined by the balance of the gut microbiota. LBP showed great potential as a beneficial nutrient for rehabilitating Dubosiella which is dominant in the gut of healthy mice. Nutrient-related LBP may play an important role in gut health management.
Collapse
Affiliation(s)
- Rong Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ping Yang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Bowen Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ziru Ye
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Puyue Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Mingjian Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yanju Gong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yong Huang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Lan Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Min Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| |
Collapse
|
8
|
Xu X, Wang Q, Tong P, Li X, Meng X, Wu Y, Yuan J, Chen H, Gao J. Effects of medicine food homologous materials on food allergy-associated factors: intestinal oxidative stress, intestinal inflammation and Th2 immune response. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:3936-3946. [PMID: 38268027 DOI: 10.1002/jsfa.13276] [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: 05/04/2023] [Revised: 12/17/2023] [Accepted: 01/04/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Food allergies could be regulated via Th1/Th2 balance, intestinal oxidative stress and inflammation, which were considered as food allergy-associated factors. Medicine-food homologous materials (MFHM) were considered as a significant factor with respect to preventing human diseases. To evaluate the associations between MFHM and food allergy-associated factors, two types of MFHM with the remarkable function of anti-oxidation and anti-inflammation, Gardeniae fructus (Gar) and Sophorae glos (Sop), were chosen. RESULTS By constructing an H2O2-induced oxidative stress model of Caco-2 cells and an intestinal inflammatory cell model of Caco-2 cells with tumor necrosis factor-α and interleukin (IL)-13, the contents of anti-oxidative enzymes (SOD and GSH), inflammatory factor (IL-8) and tight junction proteins (zonula occludens-1, occludin and claudin-1) in Caco-2 cells were determined. Moreover, the anti-allergic effects of digestive Sop and Gar were evaluated by measuring the levels of Th1/Th2/Treg cytokines in the spleen cells of sensitized mice. The results showed that the SOD and GSH were obviously increased and the gene and protein expression of IL-8 and claudin-1 were improved with the incubation of digested Sop. Th2 cytokine was reduced and Th1/Th2 balance was promoted on coincubation with ovalbumin (OVA) and digested Sop in the splenocytes. However, the digested Gar had no effect. CONCLUSION The digested Sop not only had suppressive effects on intestinal oxidative stress and inflammation, but also had regulative effects on Th1/Th2 balance. This finding demonstrated that not all of the MFHM with anti-oxidant and anti-inflammatory effects have anti-allergic activities. The present study may be contributing toward establishing a screening model to identify the anti-allergic MFHM. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaoqian Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science& Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, China
| | - Qian Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science& Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, China
| | - Ping Tong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- College of Food Science& Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, China
| | - Xuanyi Meng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Yong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Juanli Yuan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, China
- College of Pharmaceutical Science, Nanchang University, Nanchang, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, China
| | - Jinyan Gao
- College of Food Science& Technology, Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, China
| |
Collapse
|
9
|
Yu X, Nie W, Chen X, Zou LF, Xu BC, Chen CG. Anionic polysaccharides benefit the bioavailability of pork myofibrillar protein gels: Evidence from a perspective of protein absorption and metabolism. Int J Biol Macromol 2024; 263:130246. [PMID: 38378115 DOI: 10.1016/j.ijbiomac.2024.130246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/14/2024] [Accepted: 02/14/2024] [Indexed: 02/22/2024]
Abstract
This study aimed to probe the bioavailability of myofibrillar protein (MP) gels in mice as affected by incorporating anionic xanthan (XMP) and sodium alginate (SMP)/cationic chitosan (CSMP)/neutral curdlan (CMP) and konjac (KMP), respectively. The results showed that the numbers of peptides absorbed were obviously higher in anionic XMP and SMP groups (88 and 126, respectively) than in the cationic CSMP (51) group. The contents of free amino acids absorbed in SMP and XMP were significantly greater than that in CSMP and CMP groups (P < 0.05). Furthermore, the antioxidant capacity of bioactive compounds absorbed in the SMP group was higher than those in the other groups (P < 0.05), and the expression of tight junction protein (Occludin and ZO-1) was up-regulated in SMP group. The low contents of free ammonia, indole and p-cresol were observed in the anionic XMP, SMP and neutral KMP groups, compared to CSMP group. This work highlights the benefits of anionic polysaccharides (sodium alginate and xanthan) in developing low-fat meat products with high MP bioavailability.
Collapse
Affiliation(s)
- Xia Yu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, Anhui province, People's Republic of China.
| | - Wen Nie
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, Anhui province, People's Republic of China; School of Biological Science and Food Engineering, Chuzhou University, Chuzhou 239000, Anhui province, People's Republic of China
| | - Xing Chen
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.
| | - Li-Fang Zou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, Anhui province, People's Republic of China.
| | - Bao-Cai Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, Anhui province, People's Republic of China; Key Laboratory for Animal Food Green Manufacturing and Resource Mining of Anhui Province, Hefei University of Technology, Hefei, 230601, Anhui province, People's Republic of China
| | - Cong-Gui Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, Anhui province, People's Republic of China; Key Laboratory for Animal Food Green Manufacturing and Resource Mining of Anhui Province, Hefei University of Technology, Hefei, 230601, Anhui province, People's Republic of China.
| |
Collapse
|
10
|
Fu S, Tian X, Peng C, Zhang D, Zhou L, Yuan Y, He J, Guo L, Qiu Y, Ye C, Liu Y, Zong B. Baicalin inhibited PANX-1/P2Y6 signaling pathway activation in porcine aortic vascular endothelial cells infected by Glaesserella parasuis. Heliyon 2024; 10:e23632. [PMID: 38187335 PMCID: PMC10770501 DOI: 10.1016/j.heliyon.2023.e23632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
Glaesserella parasuis can induce endothelial barrier damage in piglets, although the mechanism by which this pathogen triggers inflammatory damage remains unclear. Baicalin possesses anti-inflammatory and anti-oxidant activities. However, whether baicalin can relieve endothelial barrier damage caused by Glaesserella parasuis infection has not yet been studied. Hence, we evaluated the ability of baicalin to counteract the changes induced by Glaesserella parasuis in porcine aortic vascular endothelial cells. The results showed that Glaesserella parasuis could upregulate the expression of pannexin 1 channel protein and promote the release of adenosine triphosphate, adenosine diphosphate, adenosine 3'-monophosphate, uridine triphosphate, uridine diphosphate, and uridine monophosphate in porcine aortic vascular endothelial cells. The expression level of purinergic receptor P2Y6 was upregulated in porcine aortic vascular endothelial cells triggered by Glaesserella parasuis. In addition, Glaesserella parasuis could activate phospholipase C-protein kinase C and myosin light chain kinase-myosin light chain signaling pathways in porcine aortic vascular endothelial cells. Baicalin could inhibit pannexin 1 channel protein expression, reduce adenosine triphosphate, adenosine diphosphate, adenosine 3'-monophosphate, uridine triphosphate, uridine diphosphate, and uridine monophosphate release, and attenuate the expression level of P2Y6 in porcine aortic vascular endothelial cells induced by Glaesserella parasuis. Baicalin could also reduce the activation of phospholipase C-protein kinase C and myosin light chain kinase-myosin light chain signaling pathways in porcine aortic vascular endothelial cells triggered by Glaesserella parasuis. Our study report that Glaesserella parasuis could promote pannexin 1 channel protein expression, induce nucleosides substance release, and P2Y6 expression in porcine aortic vascular endothelial cells and baicalin could inhibit the expression levels of pannexin 1, nucleosides substance, and P2Y6 in the porcine aortic vascular endothelial cells induced by Glaesserella parasuis, which might be served as some targets for treatment of inflammation disease caused by Glaesserella parasuis.
Collapse
Affiliation(s)
- Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| | - Xinyue Tian
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| | - Chun Peng
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| | - Dan Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| | - Linglu Zhou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| | - Yuzhen Yuan
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| | - Jing He
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| | - Ling Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| | - Chun Ye
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| | - Yu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| | - Bingbing Zong
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, PR China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan 430023, PR China
| |
Collapse
|
11
|
Fan S, Zhou Y, Zhao Y, Daglia M, Zhang J, Zhu Y, Bai J, Zhu L, Xiao X. Metabolomics reveals the effects of Lactiplantibacillus plantarum dy-1 fermentation on the lipid-lowering capacity of barley β-glucans in an in vitro model of gut-liver axis. Int J Biol Macromol 2023; 253:126861. [PMID: 37714241 DOI: 10.1016/j.ijbiomac.2023.126861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/11/2023] [Accepted: 09/09/2023] [Indexed: 09/17/2023]
Abstract
Bioactive polysaccharides known as the biological response modifiers, can directly interact with intestinal epithelium cells (IEC) and regulate key metabolic processes such as lipid metabolism. Here, the coculture of Caco-2/HT29 monolayer (>400 Ω × cm2) and HepG2 cells was developed to mimic the gut-liver interactions. This system was used to investigate the effects of raw and fermented barley β-glucans (RBG and FBG) on lipid metabolism by directly interacting with IEC. Both RBG and FBG significantly and consistently reduced the lipid droplets and triacylglycerol levels in monoculture and coculture of HepG2 overloaded with oleic acid. Notably, FBG significantly and distinctly elevated PPARα (p < 0.05) and PPARα-responsive ACOX-1 (p < 0.01) gene expressions, promoting lipid degradation in cocultured HepG2. Moreover, the metabolomics analyses revealed that FBG had a unique impact on extracellular metabolites, among them, the differential metabolite thiomorpholine 3-carboxylate was significantly and strongly correlated with PPARα (r = -0.68, p < 0.01) and ACOX-1 (r = -0.76, p < 0.01) expression levels. Taken together, our findings suggest that FBG-mediated gut-liver interactions play a key role in its lipid-lowering effects that are superior to those of RBG. These results support the application of Lactiplantibacillus fermentation for improving hypolipidemic outcomes.
Collapse
Affiliation(s)
- Songtao Fan
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Yurong Zhou
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Yansheng Zhao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Maria Daglia
- Department of Pharmacy, University of Naples Federico II, Naples, Italy; International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, China
| | - Jiayan Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Ying Zhu
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Juan Bai
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Lin Zhu
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Xiang Xiao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
12
|
Jia J, Zheng W, Tang S, Song S, Ai C. Scytosiphon lomentaria fucoidan ameliorates DSS-induced colitis in dietary fiber-deficient mice via modulating the gut microbiota and inhibiting the TLR4/NF-κB/MLCK pathway. Int J Biol Macromol 2023; 253:127337. [PMID: 37820918 DOI: 10.1016/j.ijbiomac.2023.127337] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/28/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
The prevalence of ulcerative colitis (UC) poses a serious threat to human health. This study showed that fiber-deficient diet (FD) increased the susceptibility of mice to low dosage of DSS-induced UC, and a UC model was established by feeding mice with DSS and FD to evaluate the effect of Scytosiphon lomentaria fucoidan (SLF) on UC. SLF ameliorated the symptoms of UC, as evidenced by increases in colon length, goblet cells and glycoprotein and reduction in inflammatory cell infiltration and intestinal epithelial injury. SLF alleviated oxidative stress and inhibited colonic inflammation by reducing the levels of lipopolysaccharides and pro-inflammatory cytokines and suppressing the activation of nuclear factor kappa B pathway. SLF protected tight junction integrity by reducing the level of myosin light chain kinase and increasing the levels of claudin, zonula occludens-1 and occludin. SLF improved serum metabolites profile and affected multiple metabolic pathways that are crucial to human health, e.g. butanoate metabolism. The underlying mechanism can be associated with modulation of the gut microbiota and metabolites, including increases in short chain fatty acids and reduction in Proteobacteria, Bacteroides and Romboutsia. It suggests that SLF could be developed as a prebiotic polysaccharide to benefit human health by improving intestinal microecology.
Collapse
Affiliation(s)
- Jinhui Jia
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Weiyun Zheng
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuangru Tang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
13
|
Tao Q, Liu XW, Zhang ZD, Ma N, Lu XR, Ge WB, Li JY, Yang YJ. Protective Effect and Mechanism of Aspirin Eugenol Ester on Lipopolysaccharide-Induced Intestinal Barrier Injury. Int J Mol Sci 2023; 24:17434. [PMID: 38139262 PMCID: PMC10743450 DOI: 10.3390/ijms242417434] [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: 10/31/2023] [Revised: 12/11/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Intestinal inflammation is a complex and recurrent inflammatory disease. Pharmacological and pharmacodynamic experiments showed that aspirin eugenol ester (AEE) has good anti-inflammatory, antipyretic, and analgesic effects. However, the role of AEE in regulating intestinal inflammation has not been explored. This study aimed to investigate whether AEE could have a protective effect on LPS-induced intestinal inflammation and thus help to alleviate the damage to the intestinal barrier. This was assessed with an inflammation model in Caco-2 cells and in rats induced with LPS. The expression of inflammatory mediators, intestinal epithelial barrier-related proteins, and redox-related signals was analyzed using an enzyme-linked immunosorbent assay (ELISA), Western blotting, immunofluorescence staining, and RT-qPCR. Intestinal damage was assessed by histopathological examination. Changes in rat gut microbiota and their functions were detected by the gut microbial metagenome. AEE significantly reduced LPS-induced pro-inflammatory cytokine levels (p < 0.05) and oxidative stress levels in Caco-2 cells and rats. Compared with the LPS group, AEE could increase the relative expression of Occludin, Claudin-1, and zonula occludens-1 (ZO-1) and decrease the relative expression of kappa-B (NF-κB) and matrix metalloproteinase-9. AEE could significantly improve weight loss, diarrhea, reduced intestinal muscle thickness, and intestinal villi damage in rats. Metagenome results showed that AEE could regulate the homeostasis of the gut flora and alter the relative abundance of Firmicutes and Bacteroidetes. Flora enrichment analysis indicated that the regulation of gut flora with AEE may be related to the regulation of glucose metabolism and energy metabolism. AEE could have positive effects on intestinal inflammation-related diseases.
Collapse
Affiliation(s)
- Qi Tao
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Xi-Wang Liu
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Zhen-Dong Zhang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Ning Ma
- College of Veterinary Medicine, Hebei Agricultural University, Baoding 071001, China;
| | - Xiao-Rong Lu
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Wen-Bo Ge
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Jian-Yong Li
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| | - Ya-Jun Yang
- Key Lab of New Animal Drug Project of Gansu Province, Key Lab of Veterinary Pharmaceutical Development of Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou 730050, China; (Q.T.); (X.-W.L.); (Z.-D.Z.); (X.-R.L.); (W.-B.G.)
| |
Collapse
|
14
|
Li ZY, Lin LH, Liang HJ, Li YQ, Zhao FQ, Sun TY, Liu ZY, Zhu JY, Gu F, Xu JN, Hao QY, Zhou DS, Zhai HH. Lycium barbarum polysaccharide alleviates DSS-induced chronic ulcerative colitis by restoring intestinal barrier function and modulating gut microbiota. Ann Med 2023; 55:2290213. [PMID: 38061697 PMCID: PMC10836275 DOI: 10.1080/07853890.2023.2290213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/26/2023] [Indexed: 12/18/2023] Open
Abstract
PURPOSE This study examined the protective effects and mechanism of Lycium barbarum polysaccharides (LBP) in the context of intestinal barrier function and intestinal microbiota in mice with dextran sulfate sodium (DSS)-induced chronic ulcerative colitis (UC). METHODS C57BL/6J male mice were assigned to a standard normal diet without DSS (control group), a normal diet with DSS (DSS group, 2% DSS given discontinuously for 3 weeks) or a normal diet supplemented with LBP (1% dry feed weight, LBP group, 2% DSS given discontinuously for 3 weeks) for a total of 8 weeks, at which point colonic tissues and caecal contents were collected. RESULTS LBP exerted a significant effect against colitis by increasing body weight, colon length, DAI and histopathological scores. LBP inhibited proinflammatory cytokines (IL-1β, IL-6, iNOS and TNF-α) expression, improved anti-inflammatory cytokine (IL-10) expression, promoted the expression of tight junction proteins (Occludin and ZO-1) via nuclear factor erythroid 2-related factor 2 (Nrf2) activation and decreased Claudin-2 expression to maintain the intestinal mucosal barrier. In addition, the abundances of some probiotics (Ruminococcaceae, Lactobacillus, Butyricicoccus, and Akkermansia) were decreased with DSS treatment but increased obviously with LBP treatment. And LBP reduced the abundance of conditional pathogens associated with UC (Mucispirillum and Sutterella). Furthermore, LBP improved the production of short-chain fatty acids (SCFAs), including acetic acid, propionic acid, butyric acid and isobutyric acid. CONCLUSION LBP can alleviate DSS-induced UC by regulating inflammatory cytokines and tight junction proteins. Moreover, LBP promotes probiotics, suppresses conditional pathogens and increases SCFAs production, showing a strong prebiotic effect.
Collapse
Affiliation(s)
- Zhi-Yu Li
- Department of Gastroenterology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Lan-Hui Lin
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Department of Gastroenterology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - He-Jun Liang
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Department of Gastroenterology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Ya-Qi Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fu-Qian Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ting-Yi Sun
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zi-Yu Liu
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Department of Gastroenterology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Jing-Yi Zhu
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Department of Gastroenterology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Feng Gu
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Department of Gastroenterology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Jia-Ning Xu
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Department of Gastroenterology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Qi-Yuan Hao
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Department of Gastroenterology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - De-Shan Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hui-Hong Zhai
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Department of Gastroenterology, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Gao F, He Q, Wu S, Zhang K, Xu Z, Kang J, Quan F. Catalpol ameliorates LPS-induced inflammatory response by activating AMPK/mTOR signaling pathway in rat intestinal epithelial cells. Eur J Pharmacol 2023; 960:176125. [PMID: 37890606 DOI: 10.1016/j.ejphar.2023.176125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 10/01/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023]
Abstract
Intestinal inflammation is a common clinical intestinal disease. Catalpol, a natural iridoid compound, has been shown to have anti-inflammatory, anti-oxidant and anti-apoptotic functions, but the mechanism of its protection against intestinal inflammation is still unclear. This study investigated the protective effect and potential mechanism of catalpol on the lipopolysaccharide (LPS)-induced inflammatory response of intestinal epithelial cell-6 (IEC-6). The results showed that catalpol could inhibit LPS-induced inflammatory response by dose-dependently reducing the release of inflammatory factors, such as tumor necrosis (TNF)-α, interleukin (IL)-1β and IL-6, and inhibiting the nuclear factor kappa-B (NF-κB) signaling pathway. Catalpol ameliorated cellular oxidative stress by reducing reactive oxygen species (ROS) and malondialdehyde (MDA) levels and increasing superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) expression. Meanwhile, catalpol also inhibited cell apoptosis, decreased the expression of B-cell lymphoma 2 (Bcl-2) - associated X (Bax), caspase 3 and caspase 9, and increased the expression of Bcl-2. This study found that catalpol activates AMP-activated protein kinase (AMPK) signaling pathway and inhibit mammalian target of rapamycin (mTOR) phosphorylationthe. In a further study, after inhibiting AMPK with dorsomorphin, the anti-inflammatory effects of catalpol were significantly reduced. Therefore, catalpol ameliorates LPS-induced inflammatory response by activating AMPK/mTOR signaling pathway in IEC-6 cells.
Collapse
Affiliation(s)
- Feng Gao
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Qifu He
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Shenghui Wu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Kang Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Zhiming Xu
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Jian Kang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Fusheng Quan
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
16
|
Jiang F, Wu M, Li R. The significance of long non-coding RNAs in the pathogenesis, diagnosis and treatment of inflammatory bowel disease. PRECISION CLINICAL MEDICINE 2023; 6:pbad031. [PMID: 38163004 PMCID: PMC10757071 DOI: 10.1093/pcmedi/pbad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024] Open
Abstract
Inflammatory bowel diseases (IBD) are a group of chronic relapsing gastrointestinal inflammatory diseases with significant global incidence. Although the pathomechanism of IBD has been extensively investigated, several aspects of its pathogenesis remain unclear. Long non-coding RNAs (lncRNAs) are transcripts with more than 200 nucleotides in length that have potential protein-coding functions. LncRNAs play important roles in biological processes such as epigenetic modification, transcriptional regulation and post-transcriptional regulation. In this review, we summarize recent advances in research on IBD-related lncRNAs from the perspective of the overall intestinal microenvironment, as well as their potential roles as immune regulators, diagnostic biomarkers and therapeutic targets or agents for IBD.
Collapse
Affiliation(s)
- Fei Jiang
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou 221000, China
- Department of Laboratory Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, China
| | - Min Wu
- Drug Discovery Section, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Rongpeng Li
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou 221000, China
| |
Collapse
|
17
|
Lian YZ, Liu YC, Chang CC, Nochi T, Chao JCJ. Combined Lycium barbarum Polysaccharides with Plasmon-Activated Water Affect IFN-γ/TNF-α Induced Inflammation in Caco-2 Cells. Pharmaceuticals (Basel) 2023; 16:1455. [PMID: 37895926 PMCID: PMC10610401 DOI: 10.3390/ph16101455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
The effects of Lycium barbarum polysaccharides (LBP) and plasmon-activated water (PAW) against IFN-γ/TNF-α induced inflammation in human colon Caco-2 cells were investigated. Cells were divided into the control, induction, LBP treatment (100-500 μg/mL), and combination groups with PAW. Inflammation was induced 24 h with 10 ng/mL IFN-γ when cell confluency reached >90%, and various doses of LBP with or without PAW were treated for 3 h, and subsequently 50 ng/mL TNF-α was added for another 24 h to provoke inflammation. Combination of LBP with PAW significantly decreased the secretion of IL-6 and IL-8. Cyclooxygenase-2 and inducible NO synthase expression was attenuated in all LBP-treated groups with or without PAW. NLRP3 inflammasome and related protein PYCARD expression were inhibited by LBP at the highest dose (500 μg/mL). All doses of LBP alone significantly decreased p-ERK expression, but combination with PAW increased p-ERK expression compared to those without PAW. Additionally, 250 and 500 μg/mL of LBP with or without PAW inhibited procaspase-3/caspase-3 expression. Therefore, LBP possesses anti-inflammation and anti-apoptosis by inhibiting the secretion of inflammatory cytokines and the expression of NLRP3 inflammasome-related protein. The combination with PAW exerts additive or synergistic effect on anti-inflammation.
Collapse
Affiliation(s)
- Yu Zhi Lian
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan;
| | - Yu-Chuan Liu
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, Taipei Medical University, Taipei 110301, Taiwan;
- Cell Physiology and Molecular Image Research Center, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei 110301, Taiwan
| | - Chun-Chao Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110301, Taiwan;
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei 110301, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Tomonori Nochi
- Laboratory of Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8577, Japan;
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8577, Japan
| | - Jane C.-J. Chao
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110301, Taiwan;
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 110301, Taiwan
- Master Program in Global Health and Health Security, Taipei Medical University, Taipei 110301, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, Taipei 110301, Taiwan
| |
Collapse
|
18
|
Liu W, Liu H, Wang Y, Zhao Z, Balasubramanian B, Jha R. Effects of Enteromorpha prolifera polysaccharides on growth performance, intestinal barrier function and cecal microbiota in yellow-feathered broilers under heat stress. J Anim Sci Biotechnol 2023; 14:132. [PMID: 37814279 PMCID: PMC10563363 DOI: 10.1186/s40104-023-00932-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/21/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Global warming leading to heat stress (HS) is becoming a major challenge for broiler production. This study aimed to explore the protective effects of seaweed (Enteromorpha prolifera) polysaccharides (EPS) on the intestinal barrier function, microbial ecology, and performance of broilers under HS. A total of 144 yellow-feathered broilers (male, 56 days old) with 682.59 ± 7.38 g were randomly assigned to 3 groups: 1) TN (thermal neutral zone, 23.6 ± 1.8 °C), 2) HS (heat stress, 33.2 ± 1.5 °C for 10 h/d), and 3) HSE (HS + 0.1% EPS). Each group contained 6 replicates with 8 broilers per replicate. The study was conducted for 4 weeks; feed intake and body weights were measured at the end of weeks 2 and 4. At the end of the feeding trial, small intestine samples were collected for histomorphology, antioxidant, secretory immunoglobulin A (sIgA) content, apoptosis, gene and protein expression analysis; cecal contents were also collected for microbiota analysis based on 16S rDNA sequencing. RESULTS Dietary EPS promoted the average daily gain (ADG) of broilers during 3-4 weeks of HS (P < 0.05). At the end of HS on broilers, the activity of total superoxide dismutase (T-SOD), glutathione S-transferase (GST), and the content of sIgA in jejunum were improved by EPS supplementation (P < 0.05). Besides, dietary EPS reduced the epithelial cell apoptosis of jejunum and ileum in heat-stressed broilers (P < 0.05). Addition of EPS in HS group broilers' diet upregulated the relative mRNA expression of Occludin, ZO-1, γ-GCLc and IL-10 of the jejunum (P < 0.05), whereas downregulated the relative mRNA expression of NF-κB p65, TNF-α and IL-1β of the jejunum (P < 0.05). Dietary EPS increased the protein expression of Occludin and ZO-1, whereas it reduced the protein expression of NF-κB p65 and MLCK (P < 0.01) and tended to decrease the protein expression of TNF-α (P = 0.094) in heat-stressed broilers. Furthermore, the proportions of Bacteroides and Oscillospira among the three groups were positively associated with jejunal apoptosis and pro-inflammatory cytokine expression (P < 0.05) and negatively correlated with jejunal Occludin level (P < 0.05). However, the proportions of Lactobacillus, Barnesiella, Subdoligranulum, Megasphaera, Collinsella, and Blautia among the three groups were positively related to ADG (P < 0.05). CONCLUSIONS EPS can be used as a feed additive in yellow-feathered broilers. It effectively improves growth performance and alleviates HS-induced intestinal injury by relieving inflammatory damage and improving the tight junction proteins expression. These beneficial effects may be related to inhibiting NF-κB/MLCK signaling pathway activation and regulation of cecal microbiota.
Collapse
Affiliation(s)
- Wenchao Liu
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, People's Republic of China
| | - Huimei Liu
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, People's Republic of China
| | - Yaoyao Wang
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, People's Republic of China
| | - Zhongxiang Zhao
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, Guangdong, 524088, People's Republic of China
| | | | - Rajesh Jha
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
19
|
Yu Y, Xia Y, Sun N, Tian Y, Chen X, Fan L, Zhao C, Xia C, Yang A, Liu H. Extraction and Purification, Structural Characterization and Biological Activity of a Polysaccharide, PRP-1, from Plumeria rubra. Chem Biodivers 2023; 20:e202300866. [PMID: 37537695 DOI: 10.1002/cbdv.202300866] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023]
Abstract
Polysaccharides derived from the flowers of Plumeria rubra (PRP) have shown a variety of beneficial effects on improving human health. However, the structural features and bioactivities of PRP remain unclear. A novel neutral polysaccharide (named PRP-1) with a molecular weight of 23 kDa was extracted and purified from the flowers of P. rubra. PRP-1 was consisted of arabinose, galactose, glucose, xylose and mannose, with a molar ratio of 1.49: 27.89: 50.24: 13.02: 7.36. The structural characterization based on the methylation and 1D/2D nuclear magnetic resonance analyses indicated that PRP-1 was composed of →4)-Glcp-(1→, →4,6)-Glcp-(1→, →4)-Galp-(1→, →2)-Galp-(1→, t-Gal(p), →4)-Manp-(1→, →4,6)-Manp-(1→, t-Man(p), →2)-Xylp-(1→, and t-Xyl(p). Scanning electron microscopy revealed that PRP-1 possess a compact three-dimensional curling network structure in the terms of morphology. PRP-1 exhibited anti-inflammatory activity, which have moderate inhibitory effects on TNF-α and IL-6 production in lipopolysaccharide (LPS)-induced RAW 264.7 cells. In addition, PRP-1 showed ABTS, OH radicals scavenging and the Fe2+ chelating effects in a concentration dependent manner. In α-glucosidase inhibition assay, PRP-1 did not exhibit inhibitory activity. Overall, these results provide a scientific basis for the utilization of the flowers of P. rubra as a potential functional food ingredient.
Collapse
Affiliation(s)
- Yongshi Yu
- School of Food Science and Engineering, Foshan University, No.33 Guangyun Road, Nanhai District, Foshan, China
| | - Yingchi Xia
- School of Medicine, Foshan University, No.33 Guangyun Road, Nanhai District, Foshan, China
| | - Ningyun Sun
- School of Food Science and Engineering, Foshan University, No.33 Guangyun Road, Nanhai District, Foshan, China
| | - Yamei Tian
- School of Food Science and Engineering, Foshan University, No.33 Guangyun Road, Nanhai District, Foshan, China
| | - Xin Chen
- School of Medicine, Foshan University, No.33 Guangyun Road, Nanhai District, Foshan, China
| | - Lixia Fan
- School of Medicine, Foshan University, No.33 Guangyun Road, Nanhai District, Foshan, China
| | - Chaochao Zhao
- School of Medicine, Foshan University, No.33 Guangyun Road, Nanhai District, Foshan, China
| | - Chenglai Xia
- Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, No.11 Renmin West Road, Chancheng District, Foshan, China
| | - Anping Yang
- School of Medicine, Foshan University, No.33 Guangyun Road, Nanhai District, Foshan, China
| | - Hui Liu
- School of Food Science and Engineering, Foshan University, No.33 Guangyun Road, Nanhai District, Foshan, China
- School of Medicine, Foshan University, No.33 Guangyun Road, Nanhai District, Foshan, China
| |
Collapse
|
20
|
Wang Y, Ke W, Gan J, Zhu H, Xie X, He G, Liu S, Huang Y, Tang H. MicroRNA-29b-3p promotes intestinal permeability in IBS-D via targeting TRAF3 to regulate the NF-κB-MLCK signaling pathway. PLoS One 2023; 18:e0287597. [PMID: 37428806 PMCID: PMC10332595 DOI: 10.1371/journal.pone.0287597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 06/08/2023] [Indexed: 07/12/2023] Open
Abstract
Irritable bowel syndrome with predominant diarrhea (IBS-D) is characterized by increased intestinal permeability. Previous studies have shown that the microRNA-29 gene is involved in the regulation of intestinal permeability in patients with IBS-D. NF-κB was proved to play a key role in inflammatory response of intestine and resultant disruption of tight junction integrity, whose activity could be inhibited by TNF Receptor-Associated Factor 3 (TRAF3). However, the exact mechanism that induces increased intestinal permeability in IBS-D patients has not been clarified. In this study, we found that microRNA-29b‑3p (miR-29b-3p) was significantly upregulated, while TRAF3 was decreased and the NF-κB-MLCK pathway was activated within the colonic tissue of IBS-D patients. Subsequently, we confirmed the targeting relationship between miR-29b-3p and TRAF3 through a double-luciferase reporter assay. Lentivirus transfection of NCM460 cells with miR-29b-3p-overexpressing and -silencing vectors demonstrated that the expression of TRAF3 was negatively correlated with the level of miR-29b-3p. The NF-κB/MLCK pathway was activated in the miR-29b-3p-overexpressing group and inhibited to some extent in the miR-29b-3p-silencing group. Results in WT and miR-29 knockout mice showed that miR-29b-3p levels were increased, TRAF3 levels were decreased, and the NF-κB/MLCK signaling was activated in the WT IBS-D group as compared with the WT control group. The protein levels of TRAF3 and TJs in the miR-29b-/- IBS-D group were partially recovered and NF-κB/MLCK pathway indicators were, to a certain extent, decreased as compared with the WT IBS-D group. These results suggested that miR-29b-3p deletion enhances the TRAF3 level in IBS-D mice and alleviates the high intestinal permeability. In brief, through the analysis of intestinal tissue samples from IBS-D patients and miR-29b-/- IBS-D mice, we showed that miR-29b-3p is involved in the pathogenesis of intestinal hyperpermeability in IBS-D via targeting TRAF3 to regulate the NF-κB-MLCK signaling pathway.
Collapse
Affiliation(s)
- Yongfu Wang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wei Ke
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianfeng Gan
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - He Zhu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiangyu Xie
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guodong He
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shan Liu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yusheng Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hongmei Tang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
21
|
Gong S, Zheng J, Zhang J, Han J. Arabinogalactan ameliorates benzo[a]pyrene-induced intestinal epithelial barrier dysfunction via AhR/MAPK signaling pathway. Int J Biol Macromol 2023:124866. [PMID: 37196716 DOI: 10.1016/j.ijbiomac.2023.124866] [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: 11/12/2022] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
Benzo[a]pyrene (B[a]P), a kind of pollutant, can disrupt the gut microbiota, but its effects on the function of intestinal epithelial barrier (IEB) is still unclear. Arabinogalactan (AG), a natural polysaccharide, can protect intestinal tract. Thus, the purpose of this study was to evaluate the effect of B[a]P on IEB function and the mitigation effect of AG on the IEB dysfunction induced by B[a]P using a Caco-2 cell monolayer model. We found B[a]P could damage the IEB integrity by inducing cell cytotoxicity, increasing lactate dehydrogenase leakage, decreasing the transepithelial electrical resistance, and increasing fluorescein isothiocyanate-dextran flux. The mechanism of B[a]P-induced IEB damage may through induction of oxidative stress, including increasing reactive oxygen species levels, decreasing glutathione levels, reducing the activity of superoxide dismutase, and increasing malonaldehyde levels. Moreover, it can be due to increasing secretion of pro-inflammatory cytokines (interleukin [IL]-1β, IL-6, and tumor necrosis factor [TNF]-α), down-regulated expression of tight junction (TJ) proteins (claudin-1, zonula occludens [ZO]-1, and occludin), and induced activation of aryl hydrocarbon receptor (AhR)/mitogen activated protein kinase (MAPK) signaling pathway. Remarkably, AG ameliorated B[a]P-induced IEB dysfunction through inhibited oxidative stress and pro-inflammatory factor secretion. Our study demonstrated B[a]P could damage the IEB and AG could alleviate this damage.
Collapse
Affiliation(s)
- Shaoying Gong
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jiachen Zheng
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Junjie Zhang
- College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Jianchun Han
- College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
22
|
Wang Y, Zhang J, Zhang B, Lu M, Ma J, Liu Z, Huang J, Ma J, Yang X, Wang F, Tang X. Modified Gegen Qinlian decoction ameliorated ulcerative colitis by attenuating inflammation and oxidative stress and enhancing intestinal barrier function in vivo and in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2023; 313:116538. [PMID: 37086872 DOI: 10.1016/j.jep.2023.116538] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 04/12/2023] [Accepted: 04/20/2023] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Modified Gegen Qinlian decoction (MGQD), which was first documented in Treatise on Febrile Disease, is recognized as a classic prescription to treat ulcerative colitis (UC). However, its protective mechanism against UC remains to be fully elucidated. AIM OF THE STUDY To explore the impact and the potential molecular mechanism of MGQD on dextran sodium sulfate (DSS)-induced UC mice and tumor necrosis factor alpha (TNF-α)-induced Caco-2 cell monolayer model of intestinal barrier. MATERIALS AND METHODS The chemical components of MGQD and MGQD drug containing serum (MGQD-DS) were characterized by LC-MS/MS. The therapeutic effect of MGQD on DSS-induced UC was evaluated based on body weight, disease activity index (DAI), colon length, colonic histopathological injury, inflammatory cytokines, oxidative stress response and intestinal barrier function. Cell Counting Kit (CCK)-8 assay was applied to detect the effect of MGQD-DS on the viability of Caco-2 cells. Additionally, TNF-α-induced Caco-2 cell monolayer model of intestinal barrier was established in vitro. The Caco-2 cell monolayers were administered blank serum or MGQD-DS to observe the effects of MGQD-DS on transepithelial electrical resistance (TEER), permeability of fluorescein isothiocyanate (FITC)-dextran, inflammatory cytokines, oxidative stress indicators and intestinal epithelial barrier (IEB). RESULTS MGQD significantly improved symptoms and pathological damage in UC mice by downregulating the expression of interleukin (IL)-1β and malondialdehyde (MDA), attenuating the loss of goblet cells and the destruction of intestinal epithelial ultrastructure, and upregulating the expression of superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), zonula occludens-1 (ZO-1), Occludin, Claudin-1 and E-cadherin. In vitro, MGQD-DS significantly reduced the flux of FITC-dextran, increased the TEER, inhibited the expression of IL-21, IL-17A and MDA, and promoted the expression of IL-4, IL-10, transforming growth factor-β (TGF-β), SOD, CAT, GSH, Occludin and E-cadherin in TNF-α-induced Caco-2 cell monolayer model of intestinal barrier. CONCLUSION MGQD can ameliorate DSS-induced UC mice and TNF-α-induced Caco-2 cell monolayer model of intestinal barrier, and the protective effect is related to its inhibition of inflammation, alleviation of oxidative stress, and repair of intestinal barrier damage.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Department of Gastroenterology, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, 100191, China; Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Jiaqi Zhang
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Beihua Zhang
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Mengxiong Lu
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Department of Gastroenterology, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, 100191, China; Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Jing Ma
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Zhihong Liu
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Department of Gastroenterology, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, 100191, China; Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Jinke Huang
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Jinxin Ma
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Department of Gastroenterology, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, 100191, China; Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Xuefei Yang
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Department of Gastroenterology, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, 100191, China; Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, 100191, China
| | - Fengyun Wang
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
| | - Xudong Tang
- Department of Gastroenterology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China; Department of Gastroenterology, Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, 100191, China; Academy of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
23
|
Huo J, Pei W, Liu G, Sun W, Wu J, Huang M, Lu W, Sun J, Sun B. Huangshui Polysaccharide Exerts Intestinal Barrier Protective Effects through the TLR4/MyD88/NF- κB and MAPK Signaling Pathways in Caco-2 Cells. Foods 2023; 12:foods12030450. [PMID: 36765977 PMCID: PMC9914309 DOI: 10.3390/foods12030450] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 01/20/2023] Open
Abstract
Several reports have demonstrated that natural polysaccharides exert protective effects on intestinal barrier function. In our previous study, we isolated a polysaccharide named HSP-W from Huangshui (HS). In the present study, the protective role of HSP-W in LPS-induced intestinal barrier dysfunction was determined by several molecular biological techniques. The results showed that HSP-W treatment alleviated the deduced TEER and increased the permeability of intestinal epithelial cells induced by LPS through inhibiting the release of inflammatory cytokines and enhancing the expression of tight junction (TJ) proteins. The underlying molecular mechanisms were elucidated by RNA-seq technique, which indicated that the differentially expressed genes (DEGs) between the LPS-treated and LPS+HSP-W-treated groups were enriched in the "MAPK" signaling pathway. Notably, the overlapping DEGs reversed by HSP-W intervention highlighted the pathways of the "Toll-like receptor" and "NF-κB" signaling pathways. The suppression of p38 and NF-κB were mediated by the inhibition of MyD88. Furthermore, HSP-W treatment prevented the translocation of NF-κB to nucleus, thus inhibiting the release of TNF-α, IL-6, and IL-1β. Overall, HSP-W has beneficial effects on LPS-induced inflammation; it protects the intestinal barrier from injury in Caco-2 cells through inhibiting the TLR4/MyD88/NF-κB and p38 MAPK signaling pathways.
Collapse
Affiliation(s)
- Jiaying Huo
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, China
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Wenhao Pei
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, China
| | - Guoying Liu
- Anhui Gujing Distillery Co. Ltd., Bozhou 236820, China
| | - Weizheng Sun
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Jihong Wu
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, China
- Correspondence: ; Tel.: +86-156-5271-2036
| | - Mingquan Huang
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, China
| | - Wei Lu
- Anhui Gujing Distillery Co. Ltd., Bozhou 236820, China
| | - Jinyuan Sun
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, China
| | - Baoguo Sun
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
24
|
Structure elucidation and intestinal barrier protection of an α-D-glucan in Huangshui. Int J Biol Macromol 2022; 223:595-605. [PMID: 36370853 DOI: 10.1016/j.ijbiomac.2022.11.059] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/29/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
WLY-0, as an α-D-glucan with a molecular weight (Mw) of 11.12 kDa, was successfully isolated and purified from Huangshui (HS). The results of methylation and NMR indicated that the mainchain of WLY-0 was (1 → 4)-α-D-glucan, with side chains linking at O-6. Meanwhile, the surface morphology characterization showed that WLY-0 had an irregular flake-like morphology with a rough and uneven surface and varies in sizes from nanometers to microns. Furthermore, WLY-0 relieved the increased paracellular permeability of FD4 and decreased TEER challenged by LPS, meanwhile inhibited the production of pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) and up-regulated the expression of TJ protein (Occludin, Claudin-1, ZO-1, and JAM-A) in Caco-2 cells, so to improve the intestinal barrier function. Our findings about the structural characteristics and biological activities of WLY-0 provided a scientific foundation for the utilization of HS as a potent source of an effective adjuvant in intestinal barrier injury treatment.
Collapse
|
25
|
Corrie L, Gulati M, Awasthi A, Vishwas S, Kaur J, Khursheed R, Porwal O, Alam A, Parveen SR, Singh H, Chellappan DK, Gupta G, Kumbhar P, Disouza J, Patravale V, Adams J, Dua K, Singh SK. Harnessing the dual role of polysaccharides in treating gastrointestinal diseases: As therapeutics and polymers for drug delivery. Chem Biol Interact 2022; 368:110238. [DOI: 10.1016/j.cbi.2022.110238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/27/2022] [Accepted: 10/21/2022] [Indexed: 12/01/2022]
|
26
|
The polysaccharides from the fruits of Lycium barbarum L. modify the gut community profile and alleviate dextran sulfate sodium-induced colitis in mice. Int J Biol Macromol 2022; 222:2244-2257. [DOI: 10.1016/j.ijbiomac.2022.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/11/2022] [Accepted: 10/03/2022] [Indexed: 11/05/2022]
|
27
|
The Barrier-Enhancing Function of Soluble Yam (Dioscorea opposita Thunb.) Polysaccharides in Rat Intestinal Epithelial Cells, as Affected by the Covalent Se Conjugation. Nutrients 2022; 14:nu14193950. [PMID: 36235602 PMCID: PMC9571917 DOI: 10.3390/nu14193950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/18/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
The non-starch yam polysaccharides (YP) are the bioactive substances of edible yam, while Se is an essential nutrient for the human body. Whether a covalent conjugation of Se to YP might cause bioactivity change for the resultant selenylated YP in the intestine is still insufficiently studied, including the critical intestinal barrier function. In this study, two selenylated YP products, namely, YPSe-I and YPSe-II, with corresponding Se contents of 795 and 1480 mg/kg, were obtained by the reaction of YP and Na2SeO3 in the presence of HNO3 and then assessed for their bioactivities to a cell model (i.e., rat intestinal epithelial IEC-6 cells). The results showed that YP, YPSe-I, and YPSe-II at 5–80 μg/mL dosages could promote cell growth with treatment times of 12–24 h. The three samples also could improve barrier integrity via increasing cell monolayer resistance and anti-bacterial activity against E. coli or by reducing paracellular permeability and bacterial translocation. Additionally, the three samples enhanced F-actin distribution and promoted the expression of the three tight junction proteins, namely, zonula occluden-1, occludin, and claudin-1. Meanwhile, the expression levels of ROCK and RhoA, two critical proteins in the ROCK/RhoA singling pathway, were down-regulated by these samples. Collectively, YPSe-I and, especially, YPSe-II were more potent than YP in enhancing the assessed bioactivities. It is thus concluded that this chemical selenylation of YP brought about enhanced activity in the cells to promote barrier integrity, while a higher selenylation extent of the selenylated YP induced much activity enhancement. Collectively, the results highlighted the important role of the non-metal nutrient Se in the modified polysaccharides.
Collapse
|
28
|
Han B, Ma Y, Liu Y. Fucoxanthin Prevents the Ovalbumin-Induced Food Allergic Response by Enhancing the Intestinal Epithelial Barrier and Regulating the Intestinal Flora. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:10229-10238. [PMID: 35947424 DOI: 10.1021/acs.jafc.2c04685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
This study aimed to determine whether fucoxanthin alleviated ovalbumin (OVA)-induced food allergy (FA) and explored the possible mechanisms. The results indicated that supplementation with fucoxanthin at 10.0-20.0 mg/kg per day for 7 weeks inhibited food anaphylaxis and the production of immunoglobulin (Ig) E, IgG, histamine, and related cytokines while alleviating allergic symptoms in sensitized mice. Fucoxanthin enhanced the intestinal epithelial barrier by up-regulating tight junction (TJ) protein expression and promoting regenerating islet-derived protein III-gamma (RegIIIγ) and secretory IgA (sIgA) secretion. In addition, fucoxanthin induced the secretion of anti-inflammatory factors (interleukin (IL)-10 and transforming growth factor β (TGF-β)) by regulatory T (Treg) cells and decreased the pro-inflammatory factor levels (IL-4, tumor necrosis factor-α (TNF-α), IL-17, and IL-1β), ameliorating intestinal inflammation. Compared with the model group, beneficial bacteria, such as Lactobacillaceae, increased in the intestinal flora, while pathogenic bacteria like Helicobacteraceae, Desulfovibrionaceae, and Streptococcaceae decreased. Therefore, fucoxanthin may effectively prevent FA by enhancing the intestinal epithelial barrier and reshaping the intestinal flora.
Collapse
Affiliation(s)
- Bing Han
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Yu Ma
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
| | - Yixiang Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, China
- Collaborative Innovation Center of Provincial and Ministerial Co-construction for Marine Food Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
29
|
Lycium Genus Polysaccharide: An Overview of its Extraction, Structures, Pharmacological Activities and Biological Applications. SEPARATIONS 2022. [DOI: 10.3390/separations9080197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Polysaccharide is considered to be the main active ingredient of the genus Lycium L., which is taken from the dried fruit of the famous Chinese herbal medicine and precious tonic known as wolfberry. Traditional uses include nourishing the liver and kidney and improving eyesight, with widespread use in the clinical practice of traditional Chinese medicine. Many studies have focused on the isolation and identification of the genus Lycium L. polysaccharide and its biological activities. However, the variety of raw materials and the mechanisms of polysaccharides differ. After extraction, the structure and biological activity of the obtained polysaccharides also differ. To date, approximately 58 kinds of polysaccharides have been isolated and purified from the Lycium genus, including water-soluble polysaccharides; homogeneous polysaccharides; pectin polysaccharides; acidic heteropolysaccharides; and arabinogalactans, which are composed of arabinose, glucosamine, galactose, glucose, xylose, mannose, fructose, ribose, galacturonic acid, and glucuronic acid. Pharmacological studies have shown that LBPs exhibit a variety of important biological activities, such as protection of nerves; promotion of reproduction; and anti-inflammatory, hepatoprotective, hypoglycemic, and eyesight-improving activities. The aim this paper is to summarize previous and current references to the isolation process, structural characteristics, and biological activities of the genus Lycium L. polysaccharide. This review will provide a useful reference for further research and application of the genus Lycium L. polysaccharide in the field of functional food and medicine.
Collapse
|
30
|
Lycium barbarum polysaccharide modulates gut microbiota to alleviate rheumatoid arthritis in a rat model. NPJ Sci Food 2022; 6:34. [PMID: 35864275 PMCID: PMC9304368 DOI: 10.1038/s41538-022-00149-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 07/01/2022] [Indexed: 11/08/2022] Open
Abstract
Rheumatoid arthritis (RA) seriously impairs the quality of life of sufferers. It has been shown that Lycium barbarum polysaccharide (LBP), a natural active indigestible ingredient with medicinal and edible functions, can effectively relieve RA, however, whether this effect is related to gut microbiota is not known. This study aimed to explore the RA alleviating mechanism of LBP mediated by gut microbiota using a collagen-induced arthritis rat model. The results showed that LBP significantly changed the gut microflora structure accompanied with the RA alleviation. Specifically, a LBP intervention reduced the relative abundance of Lachnospiraceae_NK4A136_group and uncultured_bacterium_f_Ruminococcaceae and significantly increased the abundance of Romboutsia, Lactobacillus, Dubosiella and Faecalibaculum. The mRNA contents of several colonic epithelial genes including Dpep3, Gstm6, Slc27a2, Col11a2, Sycp2, SNORA22, Tnni1, Gpnmb, Mypn and Acsl6, which are potentially associated to RA, were down-regulated due to the DNA hypermethylation, possibly caused by the elevating content of a bacterial metabolite S-adenosyl methionine (SAM). In conclusion, our current study suggests that LBP alleviated RA by reshaping the composition of intestinal microflora which may generate SAM, inducing DNA hypermethylation of RA-related genes in the host intestinal epithelium and subsequently reducing their expression.
Collapse
|
31
|
Gao J, Cao S, Xiao H, Hu S, Yao K, Huang K, Jiang Z, Wang L. Lactobacillus reuteri 1 Enhances Intestinal Epithelial Barrier Function and Alleviates the Inflammatory Response Induced by Enterotoxigenic Escherichia coli K88 via Suppressing the MLCK Signaling Pathway in IPEC-J2 Cells. Front Immunol 2022; 13:897395. [PMID: 35911699 PMCID: PMC9331657 DOI: 10.3389/fimmu.2022.897395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Intestinal epithelial barrier injury disrupts immune homeostasis and leads to many intestinal disorders. Lactobacillus reuteri (L. reuteri) strains can influence immune system development and intestinal function. However, the underlying mechanisms of L. reuteri LR1 that regulate inflammatory response and intestinal integrity are still unknown. The present study aimed to determine the effects of LR1 on the ETEC K88-induced intestinal epithelial injury on the inflammatory response, intestinal epithelial barrier function, and the MLCK signal pathway and its underlying mechanism. Here, we showed that the 1 × 109 cfu/ml LR1 treatment for 4 h dramatically decreased interleukin-8 (IL-8) and IL-6 expression. Then, the data indicated that the 1 × 108 cfu/ml ETEC K88 treatment for 4 h dramatically enhanced IL-8, IL-6, and tumor necrosis factor-α (TNF-α) expression. Furthermore, scanning electron microscope (SEM) data indicated that pretreatment with LR1 inhibited the ETEC K88 that adhered on IPEC-J2 and alleviated the scratch injury of IPEC J2 cells. Moreover, LR1 pretreatment significantly reversed the declined transepithelial electrical resistance (TER) and tight junction protein level, and enhanced the induction by ETEC K88 treatment. Additionally, LR1 pretreatment dramatically declined IL-8, IL-17A, IL-6, and TNF-α levels compared with the ETEC K88 group. Then, ETEC K88-treated IPEC-J2 cells had a higher level of myosin light-chain kinase (MLCK), higher MLC levels, and a lower Rho-associated kinase (ROCK) level than the control group, while LR1 pretreatment significantly declined the MLCK and MLC expression and enhanced ROCK level in the ETEC K88-challenged IPEC-J2 cells. Mechanistically, depletion of MLCK significantly declined MLC expression in IPEC-J2 challenged with ETEC K88 compared to the si NC+ETEC K88 group. On the other hand, the TER of the si MLCK+ETEC K88 group was higher and the FD4 flux in the si MLCK+ETEC K88 group was lower compared with the si NC+ETEC K88 group. In addition, depletion of MLCK significantly enhanced Claudin-1 level and declined IL-8 and TNF-α levels in IPEC-J2 pretreated with LR1 followed by challenging with ETEC K88. In conclusion, our work indicated that L. reuteri LR1 can decline inflammatory response and improve intestinal epithelial barrier function through suppressing the MLCK signal pathway in the ETEC K88-challenged IPEC-J2.
Collapse
Affiliation(s)
- Jingchun Gao
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- College of Animal Sciences and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Shuting Cao
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Hao Xiao
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Shenglan Hu
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Kang Yao
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Kaiyong Huang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Zongyong Jiang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Li Wang
- State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- *Correspondence: Li Wang,
| |
Collapse
|
32
|
Tissue distribution of Lycium barbarum polysaccharides in rat tissue by fluorescein isothiocyanate labeling. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
33
|
Research progress of Lycium barbarum L. as functional food: phytochemical composition and health benefits. Curr Opin Food Sci 2022. [DOI: 10.1016/j.cofs.2022.100871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
34
|
Chen YS, Lian YZ, Chen WC, Chang CC, Tinkov AA, Skalny AV, Chao JCJ. Lycium barbarum Polysaccharides and Capsaicin Inhibit Oxidative Stress, Inflammatory Responses, and Pain Signaling in Rats with Dextran Sulfate Sodium-Induced Colitis. Int J Mol Sci 2022; 23:ijms23052423. [PMID: 35269566 PMCID: PMC8910612 DOI: 10.3390/ijms23052423] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/14/2022] [Accepted: 02/18/2022] [Indexed: 12/22/2022] Open
Abstract
Ulcerative colitis (UC) is an inflammatory disease with chronic relapsing symptoms. This study investigated the effects of Lycium barbarum polysaccharides (LBP) and capsaicin (CAP) in dextran sulfate sodium (DSS)-induced UC rats. Rats were divided into normal, DSS-induced UC, and UC treated with 100 mg LBP/kg bw, 12 mg CAP/kg bw, or 50 mg LBP/kg bw and 6 mg CAP/kg bw. Rats were fed LBP or CAP orally by gavage for 4 weeks, and UC model was established by feeding 5% DSS in drinking water for 6 days during week 3. Oral CAP and mixture significantly reduced disease activity index. Oral LBP significantly decreased serum malondialdehyde, interleukin (IL)-6, colonic tumor necrosis factor (TNF)-α levels, and protein expression of transient receptor potential cation channel V1 (TRPV1) and transient receptor potential ankyrin 1 (TRPA1), but increased serum catalase activity. Oral CAP significantly suppressed serum IL-6, colonic TRPV1 and TRPA1 protein expression, but elevated IL-10 levels, serum superoxide dismutase and catalase activities. The mixture of LBP and CAP significantly reduced serum IL-6, colonic TNF-α and TRPA1 protein. In conclusion, administration of LBP and/or CAP attenuate DSS-induced UC symptoms through inhibiting oxidative stress, proinflammatory cytokines, and protein expression of TRPV1 and TRPA1.
Collapse
Affiliation(s)
- Yu-Shan Chen
- School of Nutrition and Health Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei 110301, Taiwan; (Y.-S.C.); (Y.Z.L.)
- Department of Dietetics, Taipei Medical University Hospital, 252 Wu-Hsing Street, Taipei 110301, Taiwan
| | - Yu Zhi Lian
- School of Nutrition and Health Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei 110301, Taiwan; (Y.-S.C.); (Y.Z.L.)
| | - Wen-Chao Chen
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, 252 Wu-Hsing Street, Taipei 110301, Taiwan; (W.-C.C.); (C.-C.C.)
| | - Chun-Chao Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, 252 Wu-Hsing Street, Taipei 110301, Taiwan; (W.-C.C.); (C.-C.C.)
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei 110301, Taiwan
| | - Alexey A. Tinkov
- Laboratory of Molecular Dietetics, I.M. Sechenov First Moscow State Medical University, 2–4 Bolshaya Pirogovskaya Street, 119435 Moscow, Russia; (A.A.T.); (A.V.S.)
- Institute of Bioelementology, Orenburg State University, Pobedy Avenue, 13, 460018 Orenburg, Russia
| | - Anatoly V. Skalny
- Laboratory of Molecular Dietetics, I.M. Sechenov First Moscow State Medical University, 2–4 Bolshaya Pirogovskaya Street, 119435 Moscow, Russia; (A.A.T.); (A.V.S.)
- Institute of Bioelementology, Orenburg State University, Pobedy Avenue, 13, 460018 Orenburg, Russia
- Federal Research Centre of Biological Systems and Agrotechnologies of the Russian Academy of Sciences, 9 Yanvarya Street, 29, 460000 Orenburg, Russia
| | - Jane C.-J. Chao
- School of Nutrition and Health Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei 110301, Taiwan; (Y.-S.C.); (Y.Z.L.)
- Master Program in Global Health and Development, Taipei Medical University, 250 Wu-Hsing Street, Taipei 110301, Taiwan
- Nutrition Research Center, Taipei Medical University Hospital, 252 Wu-Hsing Street, Taipei 110301, Taiwan
- Correspondence: ; Tel.: +886-2-2736-1661 (ext. 6548); Fax: +886-2-2737-3112
| |
Collapse
|
35
|
Huo J, Wu Z, Sun W, Wang Z, Wu J, Huang M, Wang B, Sun B. Protective Effects of Natural Polysaccharides on Intestinal Barrier Injury: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:711-735. [PMID: 35078319 DOI: 10.1021/acs.jafc.1c05966] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Owing to their minimal side effects and effective protection from oxidative stress, inflammation, and malignant growth, natural polysaccharides (NPs) are a potential adjuvant therapy for several diseases caused by intestinal barrier injury (IBI). More studies are accumulating on the protective effects of NPs with respect to IBI, but the underlying mechanisms remain unclear. Thus, this review aims to represent current studies that investigate the protective effects of NPs on IBI by directly maintaining intestinal epithelial barrier integrity (inhibiting oxidative stress, regulating inflammatory cytokine expression, and increasing tight junction protein expression) and indirectly regulating intestinal immunity and microbiota. Furthermore, the mechanisms underlying IBI development are briefly introduced, and the structure-activity relationships of polysaccharides with intestinal barrier protection effects are discussed. Potential developments and challenges associated with NPs exhibiting protective effects against IBI have also been highlighted to guide the application of NPs in the treatment of intestinal diseases caused by IBI.
Collapse
Affiliation(s)
- Jiaying Huo
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510640, People's Republic of China
| | - Ziyan Wu
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, People's Republic of China
| | - Weizheng Sun
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510640, People's Republic of China
| | - Zhenhua Wang
- Center for Mitochondria and Healthy Aging, College of Life Science, Yantai University, Yantai, Shandong 264005, People's Republic of China
| | - Jihong Wu
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- College of Chemistry and Materials Engineering, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, People's Republic of China
| | - Mingquan Huang
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, People's Republic of China
| | - Bowen Wang
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, People's Republic of China
| | - Baoguo Sun
- Key Laboratory of Brewing Molecular Engineering of China Light Industry, Beijing Technology and Business University, Beijing 100048, People's Republic of China
- Beijing Laboratory of Food Quality and Safety, Beijing Technology and Business University, Beijing 100048, People's Republic of China
| |
Collapse
|
36
|
Sun Q, Du M, Kang Y, Zhu MJ. Prebiotic effects of goji berry in protection against inflammatory bowel disease. Crit Rev Food Sci Nutr 2022:1-25. [PMID: 34991393 DOI: 10.1080/10408398.2021.2015680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The prevalence of inflammatory bowel disease (IBD) is increasing, which is concerning because IBD is a known risk factor for the development of colorectal cancer. Emerging evidence highlights environmental factors, particularly dietary factors and gut microbiota dysbiosis, as pivotal inducers of IBD onset. Goji berry, an ancient tonic food and a nutraceutical supplement, contains a range of phytochemicals such as polysaccharides, carotenoids, and polyphenols. Among these phytochemicals, L. barbarum polysaccharides (LBPs) are the most important functional constituents, which have protective effects against oxidative stress, inflammation, and neurodegeneration. Recently, the beneficial effects of goji berry and associated LBPs consumption were linked to prebiotic effects, which can prevent dysbiosis associated with IBD. This review assessed pertinent literature on the protective effects of goji berry against IBD focusing on the gut microbiota and their metabolites in mediating the observed beneficial effects.
Collapse
Affiliation(s)
- Qi Sun
- School of Food Science, Washington State University, Pullman, Washington, USA
| | - Min Du
- Department of Animal Science, Washington State University, Pullman, Washington, USA
| | - Yifei Kang
- School of Food Science, Washington State University, Pullman, Washington, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, Washington, USA
| |
Collapse
|
37
|
Chen W, Zhu X, Wang L, Xin X, Zhang M. Effects of Two Polysaccharides from Lepidium meyenii (Maca) on Intestinal Immunity and Inflammation in vitro. Food Funct 2022; 13:3441-3452. [DOI: 10.1039/d1fo02659c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In our previous studies, two polysaccharides (MC-1 and MC-2) were identified in the roots of maca (Lepidium meyenii). In this study, the effects of these two polysaccharides on intestinal immunity...
Collapse
|
38
|
ZHANG X, Li Y, Zhang C, Chi H, Liu C, Li A, Yu W. Postbiotics derived from Lactobacillus plantarum 1.0386 Ameliorates lipopolysaccharide-induced tight junction injury via MicroRNA-200c-3p Mediated Activation of MLCK-MLC Pathway in Caco-2 Cells. Food Funct 2022; 13:11008-11020. [DOI: 10.1039/d2fo00001f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
L. plantarum 1.0386 could repair the intestinal epithelial tight junction injury, and the present study was designed to further explore the role of its postbiotics, including surface protein (1.0386-Slp), peptidase...
Collapse
|
39
|
Liu ZC, Yu WW, Zhou HC, Lan ZC, Wu T, Xiong SM, Yan L, Liu HB. Lycium barbarum polysaccharides ameliorate LPS-induced inflammation of RAW264.7 cells and modify the behavioral score of peritonitis mice. J Food Biochem 2021; 45:e13889. [PMID: 34426988 DOI: 10.1111/jfbc.13889] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/26/2021] [Accepted: 07/19/2021] [Indexed: 01/17/2023]
Abstract
In the present study, the anti-inflammatory effect of Lycium barbarum polysaccharide (LBP) and the possible molecular mechanism thereof were examined, so as to perceive the pharmacological action of LBP. With acute peritonitis in mice as the inflammatory model, the protective effect of LBP on peritonitis mice was evaluated by recording the effect of behavioral scores, studying the pathological damage of intestine and liver, and detecting the levels of inflammatory cytokines. Additionally, by establishing an lipopolysaccharide (LPS)-induced RAW264.7 macrophage model, the effect of LBP on RAW264.7 cell phenotype and culture supernatant inflammatory markers was observed. Finally, the activation of inflammation-related target genes, such as iNOS, Toll-like receptor 4 (TLR4), nuclear factor-κB (NF-κB) p65, and IκBα, were further detected. The results reveal that pretreatment with LBP could decrease the behavioral score of inflammatory mice, inhibit the secretion of pro-inflammatory factors, and reduce liver and intestine injury. LBP can regulate the effect of lipopolysaccharide on the polarization of RAW264.7 cells, and reduce the production of NO and cytokines (TNF-α, IL-1β, IL-6). Further, LBP pretreatment was found to be able to significantly reduce the expression of iNOS, TLR4, NF-κB p65, and IκBα in macrophages. The present research provides evidence that LBP exerts potential anti-inflammatory activity in LPS-induced RAW264.7 macrophages via inhibiting TLR4 and NF-κB inflammatory sites and improving the behavior score of peritonitis mice. PRACTICAL APPLICATIONS: In recent years, the number of deaths worldwide has continued to rise as a result of inflammation. Despite said rise in deaths, many synthetic drugs with anti-inflammatory properties are significantly expensive and also have a host of side effects. Thus, the development of new anti-inflammatory drugs derived from medicinal plants has broad application potential. As such, in the present study, lipopolysaccharide (LPS)-induced macrophages were used to establish inflammatory cell models to verify the anti-inflammatory effect of Lycium barbarum polysaccharides (LBP). Findings were made that LBP could reduce the expression levels of inflammatory cytokines and NO by regulating macrophage polarization and NF-κB translocation, and thus, could exert anti-inflammatory activity. In addition, by intraperitoneal injection of LPS to establish peritonitis mice models, LBP pretreatment was found to have significantly modified the behavioral score of mice, while decreasing the secretion of inflammatory factors and the damage to several organs. The present study provides a basis for further understanding the effects of LBP in acute inflammation.
Collapse
Affiliation(s)
- Zhi-Chang Liu
- Second Clinical Medical College, Lanzhou University, Lanzhou, P.R. China.,Gansu Provincial Key Laboratory of Stem Cells and Gene Drugs, Lanzhou, P.R. China
| | - Wen-Wen Yu
- Second Clinical Medical College, Lanzhou University, Lanzhou, P.R. China
| | - Hai-Cun Zhou
- Second Clinical Medical College, Lanzhou University, Lanzhou, P.R. China.,Department of General Surgery, Gansu Maternal and Child Health Care Hospital, Lanzhou, P.R. China
| | - Zheng-Cang Lan
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, P.R. China
| | - Tong Wu
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, P.R. China
| | - Shi-Meng Xiong
- Second Clinical Medical College, Lanzhou University, Lanzhou, P.R. China.,Gansu Provincial Key Laboratory of Stem Cells and Gene Drugs, Lanzhou, P.R. China
| | - Long Yan
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, P.R. China.,Clinical Medical College, Northwest Minzu University, Lanzhou, P.R. China
| | - Hong-Bin Liu
- Second Clinical Medical College, Lanzhou University, Lanzhou, P.R. China.,Clinical Medical College, Northwest Minzu University, Lanzhou, P.R. China
| |
Collapse
|
40
|
Li M, Ge Q, Du H, Jiang P, Bao Z, Chen D, Lin S. Potential Mechanisms Mediating the Protective Effects of Tricholoma matsutake-Derived Peptides in Mitigating DSS-Induced Colitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:5536-5546. [PMID: 33955220 DOI: 10.1021/acs.jafc.1c01908] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Intestinal barrier dysfunction and inflammatory cytokine secretion play crucial roles in inflammatory bowel disease (IBD). Herein, we investigated the protective effects of Tricholoma matsutake-derived peptides SDIKHFPF and SDLKHFPF against dextran sulfate sodium-induced colitis. Both peptides alleviated colitis signs, including diarrhea, weight loss, bloody stools, colon shortening, and histopathological changes, while reducing mucus destruction, goblet cell exhaustion, and intestinal permeability. SDIKHFPF and SDLKHFPF protected the barrier function by promoting the expression of tight junction (TJ) zonula occludens-1 and occludin within the colon, as well as attenuating colonic inflammation through myeloperoxidase and pro-inflammatory cytokine suppression. Western blotting indicated that the peptides suppressed myosin light chain kinase (MLCK) and nuclear factor kappa B (NF-κB) levels, inhibiting MLC phosphorylation. SDLKHFPF was more potent than SDIKHFPF. These findings suggest that peptide SDLKHFPF mitigates colitis by regulating TJ protein expression and pro-inflammatory cytokine production via NF-κB/MLCK/p-MLC signaling, improving the barrier function.
Collapse
Affiliation(s)
- Mengqi Li
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Qi Ge
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Hanting Du
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Pengfei Jiang
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Zhijie Bao
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Dong Chen
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Songyi Lin
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| |
Collapse
|
41
|
Pehlİvan KarakaŞ F, CoŞkun H, SoytÜrk H, Bozat BG. Anxiolytic, antioxidant, and neuroprotective effects of goji berry polysaccharides in ovariectomized rats: experimental evidence from behavioral, biochemical, and immunohistochemical analyses. ACTA ACUST UNITED AC 2020; 44:238-251. [PMID: 33110362 PMCID: PMC7585160 DOI: 10.3906/biy-2003-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/08/2020] [Indexed: 01/10/2023]
Abstract
Recent studies have indicated that polysaccharides, the main component of the Lycium barbarum L. fruit, have beneficial effects (e.g., anxiolytic, antioxidant, and neuroprotective) on humans and rodents. However, the effects of different dosages of such polysaccharides on ovariectomized rats and their underlying mechanisms in the brain have not been evaluated in the literature. Here, we aimed to evaluate the effects of the high and low doses of polysaccharides obtained from Lycium barbarum fruits (HD-LBP and LD-LBP, respectively) on anxious behaviors via behavioral (using the OFT and EPM), biochemical (using ELISA), and immunohistochemical (using immunohistochemical staining) measures in detail. Two weeks after ovariectomy, the rats were randomly assigned to either the treatment conditions [control (DW, 3 mL/kg, p.o., per day), LD-LBP (20 mg/kg, 3 mL/kg, p.o., per day), HD-LBP (200 mg/kg, 3 mL/kg, p.o., per day), 17 β-ES (1 mg/kg, 3 mL/kg, p.o., per day), DZ(1 mg/kg, 3 mL/kg, p.o., per day)] or operation type [SHAM (pseudo-ovariectomized) and OVX (ovariectomized)]. The treatments were applied for 30 consecutive days, and then serum and brain tissue samples of all rats were collected. Biochemical (SOD, CAT, GPX, MDA, and 17 β-ES) and immunohistochemical (BDNF, SER, and apoptosis) analyses of the samples were performed as well. The rats administered HD-LBP and LD-LBP were less anxious than the control groups. The HD-LBP-treated rats had high levels of SOD and low levels of MDA in their serum samples. Moreover, HD-LBP and drug-treated groups had a high number of SER receptors and BDNF-positive cells and a low number of TUNEL-positive cells in their hippocampal brain tissues. The HD-LBP treatments decrease anxious behavior by increasing antioxidant enzyme activities, hippocampal SER and BDNF neurotransmitter levels and decreasing the TUNEL-positive cell count of ovariectomized rats. Given these findings, we suggest that menopause-induced symptoms of anxiety can be reduced by polysaccharides obtained from goji berry fruits, and that these findings will be beneficial for the production studies of natural herbal-origin antianxiety (anxiolytic) drugs in the future.
Collapse
Affiliation(s)
- Fatma Pehlİvan KarakaŞ
- Department of Biology, Faculty of Science and Art, Bolu Abant İzzet Baysal University, Bolu Turkey
| | - Hamit CoŞkun
- Department of Psychology, Faculty of Science and Art, Bolu Abant İzzet Baysal University, Bolu Turkey
| | - Hayriye SoytÜrk
- Department of Poultry Science andTechnology, Faculty of Agriculture and Natural Science, Bolu Abant İzzet Baysal University, Bolu Turkey
| | - Bihter Gökçe Bozat
- Disciplinary Neuroscience, Health Sciences Institute, Bolu Abant İzzet Baysal University, Bolu Turkey
| |
Collapse
|
42
|
Lu Y, Sun J, Hu M, Kong X, Zhong W, Li C. Network Pharmacology Analysis to Uncover the Potential Mechanisms of Lycium barbarum on Colorectal Cancer. Interdiscip Sci 2020; 12:515-525. [PMID: 33048277 DOI: 10.1007/s12539-020-00397-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 09/01/2020] [Accepted: 09/14/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Studies have shown that extracts from Lycium barbarum exerted protective effects against colorectal cancer (CRC) cells. We used the network pharmacology method to determine the effects of L. barbarum on CRC and to predict core targets, biological functions, pathways, and mechanisms of action. METHOD We obtained the active compounds and their targets in L. barbarum via use of the Traditional Chinese Medicine System Pharmacology Database (TCMSP), gathered the CRC targets from Malacards, TTD, GeneCards, and DisGeNET, and chosen the overlapped targets as the candidate targets. After protein-protein interaction (PPI) network analysis, 20 with the highest node degree were selected as the core targets, and their enrichment and pathways were analyzed. Furthermore, we employed iGEMDOCK to validate the compound-target relation. RESULT Eventually, 103 overlapped targets were chosen as the candidate targets. Targets with the top 20 highest node degree were selected as the core targets. Gene Ontology (GO) enrichment analysis indicated that the core targets were enriched in cell proliferation regulation, extracellular space, cytokine receptor binding, and so on. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis proved that the core targets were significantly enriched in bladder cancer, pathways in cancer. The docking results demonstrated that beta-sitosterol, glycitein, and quercetin had good binding activity to CRC putative targets. CONCLUSION Our work successfully predicted the functioning ingredients and potential targets of L. barbarum in CRC and illustrated the potential pathways and mechanisms comprehensively. Nevertheless, these results still call for in vitro and in vivo experiments to validate.
Collapse
Affiliation(s)
- Yi Lu
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jiachen Sun
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Minhui Hu
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xianhe Kong
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Weijie Zhong
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China.,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Chujun Li
- Department of Gastrointestinal Endoscopy, The Sixth Affiliated Hospital, Sun Yat-sen University, 26 Yuancun Erheng Road, Guangzhou, 510655, People's Republic of China. .,Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|