1
|
Duguma T, Melaku Y, Rentsch D, Terfa A, Shenkute K. In vitro antibacterial activities, DPPH radical scavenging, and molecular simulation of isolated compounds from the leaves of Rhus ruspolii. Z NATURFORSCH C 2025; 80:183-199. [PMID: 39300914 DOI: 10.1515/znc-2024-0127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
Rhus ruspolii Engl. plant is traditionally used in Ethiopia to treat various diseases. However, the biological and phytochemical properties of the leaves are not well documented. Hence, this study aimed to isolate phytochemicals from R. ruspolii leaves and evaluate their antibacterial and DPPH radical scavenging activities. GC-MS analysis identified 16 compounds from combined fractions 6-10. Chromatographic separation and NMR analysis resulted in the isolation and characterization of palmitic acid (7), 3,4-dihydroxybenzoic acid (17), cupressuflavone (18), amentoflavone (19), shikimic acid (20), avicularin (21), and myricetin-3-O-5''-acetylarabinofuranoside (22). The inhibition zones of extracts (100 mg/mL) and isolated compounds (5 mg/mL) ranged from 8.33 ± 0.50 to 16.33 ± 0.47 mm against all evaluated bacteria. Of all isolated compounds, compounds 18 and 21 showed good activity against Gram-negative (supported by in silico molecular docking studies) and Gram-positive bacteria, respectively. The lowest (49.1 %) and the highest (91.3 %) DPPH radicals were inhibited by combined fractions 6-10 and compound 17, respectively, at 62.5 μg/mL. The SwissADME online analysis showed compounds 17 and 20 have good solubility and permeability. The Pro Tox 3.0 online analysis revealed none of the isolated compounds are fatal if swallowed. Therefore, the findings of this study support the traditional use of the plant for treating bacteria diseases.
Collapse
Affiliation(s)
- Tolessa Duguma
- Department of Applied Chemistry, 125545 Adama Science and Technology University , P.O. Box: 1888, Adama, Ethiopia
| | - Yadessa Melaku
- Department of Applied Chemistry, 125545 Adama Science and Technology University , P.O. Box: 1888, Adama, Ethiopia
| | - Daniel Rentsch
- Laboratory for Functional Polymers, Empa-Swiss Federal Laboratories for Materials Science and Technology, CH-8600 Duebendorf, Switzerland
| | - Akalu Terfa
- Department of Applied Chemistry, 125545 Adama Science and Technology University , P.O. Box: 1888, Adama, Ethiopia
| | - Kebede Shenkute
- Department of Applied Chemistry, 125545 Adama Science and Technology University , P.O. Box: 1888, Adama, Ethiopia
| |
Collapse
|
2
|
Wang Y, Li T, Dong Z, Zhang Q, Mi J, Wang Q, Lin G, Ma Q, Jia R, Huang S. Extracellular Vesicles From Lactobacillus fermentum Enhance Intestinal Barrier Integrity and Restore Gut Microbial Homeostasis in Experimental Murine Colitis. J Nutr 2025; 155:1311-1323. [PMID: 40058701 DOI: 10.1016/j.tjnut.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Lactobacillus fermentum has been shown to improve intestinal health and treat colitis; however, its precise efficacy and mechanisms in inflammatory bowel disease remain unclear. OBJECTIVES This study aimed to evaluate whether L fermentum and its metabolites, extracellular vesicles, and other components could modulate intestinal barrier function and gut microbiota to alleviate dextran sulfate sodium (DSS)-induced colitis in mice. METHODS Forty-eight mice were randomly assigned to 6 groups: control, DSS, L fermentum+DSS group (LF+DSS), heat-inactivated L fermentum+DSS group (LHF+DSS), L fermentum supernatant solution+DSS group (LSF+DSS), and L fermentum extracellular vesicles+DSS group (LEV+DSS). After a 1-wk acclimation, mice were gavaged daily for 3 wk. Fresh cultures, including live (LF+DSS), heat-inactivated (LHF+DSS), supernatant (LSF+DSS), and extracellular vesicles (LEV+DSS), were prepared daily. During the final 7 d, the control group received normal water, and the other groups received 3% DSS. Data were collected daily, followed by sample collection from the mice. RESULTS In this study, significant reductions (P < 0.05) in body weight changes, disease activity index, intestinal damage, and histology scores were observed in the treatment groups, especially LEV+DSS and LF+DSS. Additionally, compared with the DSS group, colonic mucus secretion, as well as claudin-1 and occludin expression, increased significantly (P < 0.05) in the LEV+DSS and LF+DSS groups, whereas proinflammatory cytokines IL-1β and TNF-α decreased (P < 0.05) and IL-10 increased (P < 0.05) in the LEV+DSS group. L fermentum and its components significantly regulated gut microbiota α-diversity and β-diversity, affecting overall composition. Linear discriminant analysis effect size analysis revealed an enrichment of beneficial bacteria including Prevotellaceae_UCG-001, Romboutsia, and Ruminococcus species in the LF+DSS group and Akkermansia, Odoribacter, and Marvinbryantia species in the LEV+DSS group. Both L fermentum and its extracellular vesicles significantly downregulated the gene expression of TNF-α and IL-1β, whereas the expression of IL-10 was upregulated, thereby contributing to the alleviation of colitis symptoms. CONCLUSIONS This study reveals that L fermentum alleviates colitis through modulation of the gut microbiota and reinforcement of the intestinal mucosal barrier, with its extracellular vesicles potentially playing a key role in this regulatory process.
Collapse
Affiliation(s)
- Yanwei Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; School of Life Science, Shanxi University, Taiyuan, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Tiantian Li
- Academy of National Food and Strategic Reserves Administration, Beijing, China
| | - Zhuo Dong
- Hubei International Travel Healthcare Center, Hubei, China
| | - Qiyue Zhang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jingqiu Mi
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Qingfeng Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Gang Lin
- Institute of Quality Standards and Testing Technology for Agricultural Products, Chinese Academy of Agricultural Science, Beijing, China
| | - Qiugang Ma
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Ru Jia
- School of Life Science, Shanxi University, Taiyuan, China.
| | - Shimeng Huang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China.
| |
Collapse
|
3
|
Huang M, Zhang Y, Xu X, Duan R, Yang H. Chronic chlorothalonil exposure inhibits locomotion and interferes with the gut-liver axis in Pelophylax nigromaculatus tadpoles. Sci Rep 2025; 15:14573. [PMID: 40280937 PMCID: PMC12032272 DOI: 10.1038/s41598-025-98081-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Chlorothalonil is a widely used fungicide that has a negative effect on individual movement, but its impact pathway needs further refinement. Here, the effects of exposure to chlorothalonil on the locomotion behavior of Pelophylax nigromaculatus tadpoles (GS23) were measured at three different levels (0 µg/L, 10 µg/L, and 50 µg/L), and the possible pathways of its effects were analyzed from the gut-liver axis. Chlorothalonil exposure levels of 10 µg/L and 50 µg/L significantly reduced the average speed of P. nigromaculatus tadpoles by 26% and 32.7%, respectively, and significantly decreased the locomotor frequency by 27.1% and 58.6%, respectively. Gut microbiota analysis revealed chlorothalonil exposure significantly increased the abundance of Firmicutes, while significantly decreased the abundance of Actinobacteriota, Pseudomonas, and Rhodococcus. Metabolomics analysis identified that chlorothalonil treatment changed amino acid-related metabolism pathways in the gut and liver and altered the glycerophospholipid metabolism pathway in the liver. This study indicated that chlorothalonil can affect individual locomotor abilities and interfering with the gut-liver axis of aquatic animals. These findings establish that chlorothalonil compromises aquatic organism motility through a multi-target mechanism involving gut microbiota modulation, amino acid metabolic interference, and hepatic lipid pathway disruption.
Collapse
Affiliation(s)
- Minyi Huang
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, Hunan, China
- Key Laboratory of Development, Utilization, Quality and Safety Control of Characteristic Agricultural Resources in Central Hunan Province, Loudi, 417000, Hunan, China
| | - Yuhao Zhang
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, Hunan, China
| | - Xiang Xu
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, Hunan, China
| | - Renyan Duan
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, Hunan, China.
- Key Laboratory of Development, Utilization, Quality and Safety Control of Characteristic Agricultural Resources in Central Hunan Province, Loudi, 417000, Hunan, China.
| | - Hui Yang
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, 417000, Hunan, China
| |
Collapse
|
4
|
Lao J, Chen M, Yan S, Gong H, Wen Z, Yong Y, Jia D, Lv S, Zou W, Li J, Tan H, Yin H, Kong X, Liu Z, Guo F, Ju X, Li Y. Lacticaseibacillus rhamnosus G7 alleviates DSS-induced ulcerative colitis by regulating the intestinal microbiota. BMC Microbiol 2025; 25:168. [PMID: 40133818 PMCID: PMC11938729 DOI: 10.1186/s12866-025-03904-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Ulcerative colitis (UC) is an intestinal disease caused by many factors that seriously harms the health of humans and animals. Probiotics are currently widely used to treat intestinal inflammation; however, different strains are specific, and the functions and effects of different strains are still unclear. In this study, Lacticaseibacillus rhamnosus G7 isolated from herdsmen yogurt was used. The results of the in vitro evaluation revealed that it had good tolerance and safety. In mice with colitis, G7 alleviated weight loss and colon shortening and reduced the DAI score. After G7 treatment, the levels of proinflammatory factors (IL-1β, IL-6 and TNF-α) and histopathological scores decreased, whereas the level of IL-10 increased. In addition, G7 rebalanced the intestinal microbial composition of colitis model mice by increasing the abundance of Faecalibaculum and decreasing the abundance of Bacteroides and Escherichia_Shigella. In summary, G7 has great potential in the prevention of colitis.
Collapse
Affiliation(s)
- Jianlong Lao
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
- Marine Medical Research and Development Centre, Shenzheng Institute of Guangdong Ocean University, Shenzheng, 518120, China
| | - Man Chen
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Shuping Yan
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Han Gong
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Zhaohai Wen
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Yanhong Yong
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Dan Jia
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shuting Lv
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Wenli Zou
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Junmei Li
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Huiming Tan
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Hong Yin
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xiangying Kong
- Haibei Integrated Service Center for Agriculture and Animal Husbandry, Haibei, Qinghai, 810299, China
| | - Zengyuan Liu
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Fucheng Guo
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Xianghong Ju
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China.
- Marine Medical Research and Development Centre, Shenzheng Institute of Guangdong Ocean University, Shenzheng, 518120, China.
| | - Youquan Li
- College of Coastal Agricultural Sciences, Department of Veterinary Medicine, Guangdong Ocean University, Zhanjiang, 524088, China.
- Marine Medical Research and Development Centre, Shenzheng Institute of Guangdong Ocean University, Shenzheng, 518120, China.
| |
Collapse
|
5
|
Zhao Y, Cui D, Xiao Y, Han X, Jiang M, Gong Y. Clinical Analysis and Network Pharmacology in Revealing the Mechanism of Daifu Decoction on the Relapse of UC. Drug Des Devel Ther 2025; 19:1629-1653. [PMID: 40070532 PMCID: PMC11895689 DOI: 10.2147/dddt.s497944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
Background Daifu Decoction (DFD), a patented herbal prescription used to prevent and treat ulcerative colitis (UC). This study aimed to reveal the effect of DFD on the relapse of UC and its mechanism via integrated retrospective clinical analysis, network pharmacology and in vivo and in vitro experimental validation. Methods First, the clinical data of UC patients treated with DFD were reviewed from a real-world study (RWS), and the relapse at 24 weeks after drug withdrawal was recorded to evaluate the relapse rate. Next, the chemical components of DFD were identified via ultra performance liquid chromatography‒mass spectrometry (UPLC‒MS), and the differentially expressed genes (DEGs) between UC patients in the active and remission stages were screened as disease targets related to the relapse of UC from the Gene Expression Omnibus (GEO) database. The core components, targets and key signalling pathways of DFD for preventing the relapse of UC were discussed via network pharmacology. Finally, the above results were verified via molecular docking and in vivo and in vitro experiments. Results A total of 475 UC patients were included, and the relapse rate of UC treated with DFD was 23.9%. Additionally, the 221 components identified by UPLC-MS and 398 DEGs related to the relapse of UC enriched the main pathway of the relapse of UC was IL-17 signaling pathway and the inflammatory-related targets, such as IL6, PTGS2, MMP7, MMP3, MMP1. Moreover, molecular docking revealed that the core components of DFD were able to bind to inflammation-related targets, and in vivo and in vitro experiments demonstrated that DFD could inhibit the IL-17 pathway, increase the level of claudin-1, and control inflammation to prevent UC relapse. Conclusion DFD can effectively prevent the relapse of UC which may be related to inhibiting the activation of IL-17 signalling pathway.
Collapse
Affiliation(s)
- Yangyang Zhao
- Department of Traditional Chinese Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, People’s Republic of China
| | - Danyang Cui
- Department of Traditional Chinese Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, People’s Republic of China
| | - Yanan Xiao
- Department of Traditional Chinese Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, People’s Republic of China
| | - Xu Han
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing, People’s Republic of China
| | - Miao Jiang
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing, People’s Republic of China
| | - Yang Gong
- Department of Traditional Chinese Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, People’s Republic of China
| |
Collapse
|
6
|
Huang Y, Ru Q, Ruan H, Zhang J, Wang Y, Wang C, Chen C, Yu D, Luo J, Yang M. Changyanning tablet alleviates Crohn's disease by inhibiting GPX4-mediated ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119415. [PMID: 39870334 DOI: 10.1016/j.jep.2025.119415] [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: 12/17/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Changyanning tablets (CYN) are a marketed traditional Chinese medicine composed of Diijincao (Euphorbia humifusa Willd.), Jinmaoercao (Hedyotis chrysotricha (Palib.) Merr.), Zhangshugen (root of Cinnamomum camphora (L.) J.Presl), Xiangru (Elsholtzia ciliate (Thunb.) Hyl.), and Fengxiangshuye (leaf of Liquidambar formosana Hance). They possess the functions of clearing heat, removing dampness, and regulating qi. CYN is used for the treatment of diarrhea and dysentery caused by damp heat in the large intestine, with symptoms such as diarrhea, or stools with pus and blood, tenesmus, abdominal pain and distension, acute and chronic gastroenteritis, diarrhea, bacterial diarrhea, and indigestion in children. AIM OF THE STUDY This study aims to explore the intervention effects of CYN on Crohn's disease (CD) and its potential mechanisms. MATERIALS AND METHODS The therapeutic effect and potential mechanism of CYN on CD were investigated based on the 2,4,6-Trinitrobenzenesulfonic acid solution (TNBS)-induced rat model. In vivo and in vitro experiments confirmed that CYN can alleviate CD by inhibiting GPX4-mediated ferroptosis. siRNA was used to knock down GPX4 for reverse validation. Finally, active components of CYN inhibiting ferroptosis were identified using UPLC-MS and the RSL3-induced HCoEpiC ferroptosis cell model. RESULTS CYN significantly improved ferroptosis-related indicators (GSH, MDA, GPX4, and SLC7A11) in the colons of TNBS-induced CD rats. Screening with three ferroptosis inducers (RSL3, FINO2, and erastin) revealed that CYN was most effective against RSL3 (a ferroptosis inducer targeting GPX4)-induced apoptosis. Subsequently, the resistance effect of CYN on RSL3-induced ferroptosis was confirmed in vitro. Further in vivo experiments showed that CYN alleviated local CD-like intestinal injury induced by RSL3 enema. siRNA knockdown of GPX4 in HCoEpiC cells further validated GPX4 as major target of CYN in inhibiting ferroptosis. Finally, UPLC-MS and in vitro experiments identified rutin, rosmarinic acid, and kaempferol-3-O-sophoroside as key active components of CYN for inhibiting ferroptosis. CONCLUSIONS CYN alleviates CD by inhibiting GPX4-mediated ferroptosis, highlighting its clinical potential for treating CD and enhancing the understanding of the pathogenic and therapeutic mechanisms associated with CD.
Collapse
Affiliation(s)
- Ying Huang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Qing Ru
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Haonan Ruan
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Jing Zhang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Yunyun Wang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Chuang Wang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Changyong Chen
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Defa Yu
- Jiangxi Kangenbei Traditional Chinese Medicine Co., Ltd., Shangrao, 334400, China.
| | - Jiaoyang Luo
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Meihua Yang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
7
|
Moutsoglou D, Syal A, Lopez S, Nelson EC, Chen L, Kabage AJ, Fischer M, Khoruts A, Vaughn BP, Staley C. Novel Microbial Engraftment Trajectories Following Microbiota Transplant Therapy in Ulcerative Colitis. J Crohns Colitis 2025; 19:jjae142. [PMID: 39240145 DOI: 10.1093/ecco-jcc/jjae142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND AND AIMS Microbiota transplant therapy (MTT) is an emerging treatment for ulcerative colitis (UC). One proposed mechanism for the benefit of MTT is through engraftment of donor microbiota; however, engraftment kinetics are unknown. We identified SourceTracker as an efficient method both to determine engraftment and for the kinetic study of engrafting donor taxa to aid in determining the mechanism of how this therapy may treat UC. METHODS Ulcerative colitis patients received either encapsulated (drug name MTP-101C) or placebo capsules daily for 8 weeks followed by a 4-week washout period. Amplicon sequence data from donors and patients were analyzed using the Bayesian algorithm SourceTracker. RESULTS Twenty-seven patients were enrolled, 14 to placebo and 13 to MTT. Baseline Shannon and Chao1 indices negatively correlated with week 12 donor engraftment for patients treated with active drug capsules but not for placebo patients. SourceTracker engraftment positively correlated with the week 12 distance from donors measured using the Bray-Curtis similarity metric in treated patients but not with placebo. Engraftment at week 12 was significantly higher in the MTT group than in the placebo group. We identified engrafting taxa from donors in our patients and quantified the proportion of donor similarity or engraftment during weeks 1 through 8 (active treatment) and week 12, 4 weeks after the last dose. CONCLUSION SourceTracker can be used as a simple and reliable method to quantify donor microbial community engraftment and donor taxa contribution in patients with UC and other inflammatory conditions treated with MTT.
Collapse
Affiliation(s)
- Daphne Moutsoglou
- Department of Gastroenterology, Minneapolis VA Health Care System, MN 55417, USA
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Aneesh Syal
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Sharon Lopez
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Elizabeth C Nelson
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Lulu Chen
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Amanda J Kabage
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Monika Fischer
- Division of Gastroenterology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Byron P Vaughn
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Christopher Staley
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
8
|
Zhang Z, Fang Y, He Y, Farag MA, Zeng M, Sun Y, Peng S, Jiang S, Zhang X, Chen K, Xu M, Han Z, Zhang J. Bifidobacterium animalis Probio-M8 improves sarcopenia physical performance by mitigating creatine restrictions imposed by microbial metabolites. NPJ Biofilms Microbiomes 2024; 10:144. [PMID: 39632843 PMCID: PMC11618631 DOI: 10.1038/s41522-024-00618-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
Sarcopenia is a major health challenge due to an aging population. Probiotics may improve muscle function through gut-muscle axis, but their efficacy and mechanisms in treating sarcopenia remain unclear. This study investigated the impact of Bifidobacterium animalis subsp. lactis Probio-M8 (Probio-M8) on old mice and sarcopenia patients. We analyzed 43 subjects, including gut microbiome, fecal metabolome, and serum metabolome, using a multi-omics approach to assess whether Probio-M8 can improve sarcopenia by modulating gut microbial metabolites. Probio-M8 significantly improved muscle function in aged mice and enhanced physical performance in sarcopenia patients. It reduced pathogenic gut species and increased beneficial metabolites such as indole-3-lactic acid, acetoacetic acid, and creatine. Mediating effect analyses revealed that Probio-M8 effectively reduced n-dodecanoyl-L-homoserine lactone level in gut concurrent with increased creatine circulation, to significantly enhance host physical properties. These findings provide new insights into probiotics as a potential treatment for sarcopenia by modulating gut microbiota metabolism.
Collapse
Affiliation(s)
- Zeng Zhang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, Hainan, China
| | - Yajing Fang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, Hainan, China
| | - Yangli He
- Department of Health Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Cairo, Egypt
| | - Min Zeng
- Department of Health Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yukai Sun
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, Hainan, China
| | - Siqi Peng
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, Hainan, China
| | - Shuaiming Jiang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, Hainan, China
| | - Xian Zhang
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Kaining Chen
- Department of Endocrinology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Meng Xu
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, Hainan, China
| | - Zhe Han
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, Hainan, China
| | - Jiachao Zhang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, Hainan, China.
- One Health Institute, Hainan University, Haikou, Hainan, China.
| |
Collapse
|
9
|
Tian H, Ling N, Guo C, Gao M, Wang Z, Liu B, Sun Y, Chen Y, Ji C, Li W. Immunostimulatory activity of sea buckthorn polysaccharides via TLR2/4-mediated MAPK and NF-κB signaling pathways in vitro and in vivo. Int J Biol Macromol 2024; 283:137678. [PMID: 39566757 DOI: 10.1016/j.ijbiomac.2024.137678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/14/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
SP0.1-1, derived from Sea buckthorn (Hippophae rhamnoides L.), has been discovered to exhibit unique antioxidant activity. In this study, we investigated the immunomodulatory activity and mechanisms of SP0.1-1 on macrophage RAW 264.7 cells in vitro and immunosuppressive mice induced by cyclophosphamide in vivo. The results indicated SP0.1-1 strengthened the immune functions via promoting the proliferation of RAW264.7 cells and phagocytic activity, along with stimulating the release of NO, ROS and cytokines including TNF-α, IL-6, IL-1β and IFN-γ. Western blot and molecular docking analysis demonstrated that SP0.1-1 attached to the prime receptors TLR2 and TLR4 in RAW264.7 cells, and triggered the activation of MyD88-mediated MAPK and NF-κB signaling pathways, thereby exerting the immune response in RAW264.7 cells. However, the intervention of specific inhibitors against TLR2, TLR4, JNK, ERK, p38 and NF-κB blocked the TLR-mediated MAPK and NF-κB signaling pathways and downregulated the levels of NO and the aforementioned cytokines, thus suppressing the activation of macrophages. Therefore, it can be speculated that SP0.1-1 activated the macrophages principally via the TLR2/4-MyD88-mediated MAPK and NF-κB signaling pathways. Additionally, SP0.1-1 could protect against the cyclophosphamide-induced immunosuppression in mice, manifested by the improvement of body weight, immune organ indices, phagocytic index, and the relievement of spleen damage, along with the enhancement of cytokines TNF-α, IL-6, IFN-γ and immunoglobulin IgG and IgM. These findings will shed light on the molecular mechanism of SP0.1-1 on the immunoregulatory effect, and lay the foundation for exploiting a potential immunostimulatory agent of SP0.1-1.
Collapse
Affiliation(s)
- Haiyan Tian
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Na Ling
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China.
| | - Chunqiu Guo
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Mingze Gao
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Zihao Wang
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Bing Liu
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Yuan Sun
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Yin Chen
- School of Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China
| | - Chenfeng Ji
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China.
| | - Wenlan Li
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China.
| |
Collapse
|
10
|
Mo K, Shen Y, Su D, Lv L, Du J, Ding H, Huang X. Pharmacokinetic-Pharmacodynamic Modeling of the Immune-Enhancing Effect of Shikimic Acid in Growing Pigs. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:26224-26235. [PMID: 39542831 PMCID: PMC11613447 DOI: 10.1021/acs.jafc.4c09250] [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: 09/28/2024] [Revised: 11/03/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Shikimic acid (SA), extracted from the fruit of shikimi-no-ki, is used both as a preservative in the food industry and as an intermediate for a variety of active ingredients with a wide range of pharmacological functions. A deeper understanding of the pharmacokinetic process of SA in pigs and its impact on humoral immunity could prove invaluable in facilitating its clinical application in veterinary and human medicine. The pharmacokinetic study employed a two-period, two-sequence, crossover design to animal experiments and developed a novel method of pig plasma preparation using water as an extractant and ionization promoter, followed by purification and enrichment on a MAX solid phase extraction (SPE) column. The results showed that SA is rapidly absorbed after intragastric administration (50 mg/kg BW), reaching a plasma Cmax of 10,823.44 ng/mL at 1.78 h, followed by rapid elimination, with a t1/2 of 1.81 h, consistent with a one-compartment model. The results for intravenous administration (2 mg/kg BW) were consistent with a two-compartment open model with a t1/2 of 3.66 h, with concentrations below the limit of quantification (LOQ) observed beyond 12 h postdose. The absolute bioavailability of SA in pigs was calculated to be 21.68%. Furthermore, the Pearson's correlation analysis demonstrated a strong positive correlation between SA concentration in pig plasma and the changes of C3, C4 and IgG, IgA, and IgM (0.6 < R < 1, P < 0.0001). A more detailed pharmacokinetic-pharmacodynamic (PK-PD) modeling analysis of the intravenous group revealed the EC50/Cmax values of approximately 10%, with all γ values exceeding 3. This study was the inaugural investigation into the pharmacokinetics of SA in growing pigs, and it also revealed that SA has the potential to act as an immunopotentiator.
Collapse
Affiliation(s)
| | | | - Dehai Su
- Guangdong Key Laboratory
for Veterinary Drug Development and Safety Evaluation, College of
Veterinary Medicine, South China Agricultural
University, Guangzhou 510642, China
| | - Linyi Lv
- Guangdong Key Laboratory
for Veterinary Drug Development and Safety Evaluation, College of
Veterinary Medicine, South China Agricultural
University, Guangzhou 510642, China
| | - Juan Du
- Guangdong Key Laboratory
for Veterinary Drug Development and Safety Evaluation, College of
Veterinary Medicine, South China Agricultural
University, Guangzhou 510642, China
| | - Huanzhong Ding
- Guangdong Key Laboratory
for Veterinary Drug Development and Safety Evaluation, College of
Veterinary Medicine, South China Agricultural
University, Guangzhou 510642, China
| | - Xianhui Huang
- Guangdong Key Laboratory
for Veterinary Drug Development and Safety Evaluation, College of
Veterinary Medicine, South China Agricultural
University, Guangzhou 510642, China
| |
Collapse
|
11
|
Gu Y, Zhang J, Zheng H, Qin Y, Zheng M, Hu Y, Xin J. Therapeutic Effect of Shikimic Acid on Heat Stress-Induced Myocardial Damage: Assessment via Network Pharmacology, Molecular Docking, Molecular Dynamics Simulation, and In Vitro Experiments. Pharmaceuticals (Basel) 2024; 17:1485. [PMID: 39598396 PMCID: PMC11597562 DOI: 10.3390/ph17111485] [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: 09/30/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Rising global temperatures have been linked to an increased incidence of heat stress (HS)-induced myocardial damage. METHODS This study aimed to investigate the therapeutic potential of shikimic acid (SA) on HS-induced myocardial damage using network pharmacology, molecular docking, molecular dynamics (MD) simulations, and in vitro experiments. RESULTS Network pharmacology analysis indicated that SA significantly attenuates the inflammatory response to HS by modulating 60 targets, including TNF, IL-6, and STAT3, which are enriched in the PI3K/AKT signaling pathway. Molecular docking and MD simulation analyses demonstrated that SA forms stable complexes with TNF (-6.642 kcal/mol) and IL-6 (-7.261 kcal/mol), with no significant conformational changes over a 100 ns simulation period. In vitro experiments demonstrated that SA, within the concentration range of 250 μM to 31.25 μM, significantly promoted the proliferation of normal HL-1 cells by an average of 31.0%. Moreover, it enhanced the survival rate of HL-1 cells exposed to 43 °C for 3 h by approximately 59.9% and downregulated the expression of Hsp90 and Hsp70. Additionally, this concentration range of SA reduced the expression of TNF-α, IL-6, TLR2, and COL1A1. CONCLUSIONS These findings offer evidence for the therapeutic potential of SA in HS-induced myocardial damage.
Collapse
Affiliation(s)
- Yan Gu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.G.); (J.Z.); (H.Z.); (Y.Q.)
| | - Jingyi Zhang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.G.); (J.Z.); (H.Z.); (Y.Q.)
| | - Haohong Zheng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.G.); (J.Z.); (H.Z.); (Y.Q.)
| | - Yuyang Qin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.G.); (J.Z.); (H.Z.); (Y.Q.)
| | - Min Zheng
- Guangxi Center for Animal Disease Control and Prevention, Nanning 530001, China;
| | - Yanchun Hu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.G.); (J.Z.); (H.Z.); (Y.Q.)
| | - Jialiang Xin
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China; (Y.G.); (J.Z.); (H.Z.); (Y.Q.)
| |
Collapse
|
12
|
Peng Y, Zhu J, Li Y, Yue X, Peng Y. Almond polysaccharides inhibit DSS-induced inflammatory response in ulcerative colitis mice through NF-κB pathway. Int J Biol Macromol 2024; 281:136206. [PMID: 39362427 DOI: 10.1016/j.ijbiomac.2024.136206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Ulcerative colitis (UC), a type of inflammatory bowel disease (IBD), is a chronic recurrent inflammatory disease of the colon. Our previous findings demonstrated that almond polysaccharide (AP-1) exhibits significant anti-inflammatory activity in vitro. Therefore, this study aimed to explore the ameliorative effect of AP-1 on dextrose sodium sulfate (DSS)-induced UC mice and to elucidate its possible mechanism of action. By observing changes in body weight, fecal viscosity, stool blood, disease activity index, and colon length, we found that AP-1 attenuated inflammation. It inhibited TNF-α, IL-1β, and IL-6 while boosting anti-inflammatory IL-10 levels. Histomorphologically, AP-1 protected against DSS-induced colonic tissue damage by reducing inflammatory cell infiltration and mucosal injury. It also lowered myeloperoxidase (MPO) and NO while increasing total superoxide dismutase (T-SOD) and glutathione peroxidase (GSH-Px) in colonic tissues. Moreover, using the Western blot technique, AP-1 was shown to inhibit the phosphorylation of p65 and IκB-α proteins in the NF-κB/iNOS/COX2 signaling pathway and down-regulate the expression of inflammation-associated proteins COX2 and iNOS, thus slowing down and ameliorating inflammatory processes. Therefore, the safe and effective beneficial effects of AP-1 make it a promising therapeutic strategy for relieving IBD, especially UC.
Collapse
Affiliation(s)
- Yanqi Peng
- Department of Food Science, College of Public Health, Shenyang Medical College, Shenyang 110034, China; College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Jiayi Zhu
- College of Public Health, Shenyang Medical College, Shenyang 110034, China
| | - Yingshuo Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Xiqing Yue
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China.
| | - Yanyu Peng
- Department of Histology and Embryology, Shenyang Medical College, Shenyang 110034, China; Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, China.
| |
Collapse
|
13
|
Lao J, Yan S, Yong Y, Li Y, Wen Z, Zhang X, Ju X, Li Y. Lacticaseibacillus casei IB1 Alleviates DSS-Induced Inflammatory Bowel Disease by Regulating the Microbiota and Restoring the Intestinal Epithelial Barrier. Microorganisms 2024; 12:1379. [PMID: 39065147 PMCID: PMC11278699 DOI: 10.3390/microorganisms12071379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/18/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Inflammatory bowel disease (IBD) is becoming an increasingly serious health problem in humans and animals. Probiotics can inhibit the development of IBD. Due to the specificity of the strains, the function and mechanism of action of different strains are still unclear. Here, a DSS-induced colitis mouse model was utilized to investigate the ability and mechanism by which Lacticaseibacillus casei IB1 alleviates colitis. Treatment with L. casei IB1 improved DSS-induced colitis in mice, as indicated by increased body weight, colon length, and goblet cell numbers and decreased disease activity index (DAI), proinflammatory factor (TNF-α, IL-1β, and IL-6) levels, and histopathological scores after intake of IB1. IB1 supplementation also improved the expression of tight junction proteins and inhibited the activation of the MAPK and NF-κB signaling pathways to alleviate intestinal inflammation. In addition, IB1 rebalanced the intestinal microbial composition of colitis mice by increasing the abundance of Faecalibaculum and Alistipes and decreasing the abundance of Bacteroides and Escherichia_Shigella. In summary, L. casei IB1 showed great potential for relieving colitis by regulating the microbiota and restoring the epithelial barrier. It can be used as a potential probiotic for the prevention and treatment of UC in the future.
Collapse
Affiliation(s)
- Jianlong Lao
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (S.Y.); (Y.Y.); (Y.L.); (Z.W.); (X.Z.)
- Marine Medical Research and Development Centre, Shenzhen Institute of Guangdong Ocean University, Shenzhen 518120, China
| | - Shuping Yan
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (S.Y.); (Y.Y.); (Y.L.); (Z.W.); (X.Z.)
| | - Yanhong Yong
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (S.Y.); (Y.Y.); (Y.L.); (Z.W.); (X.Z.)
| | - Yin Li
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (S.Y.); (Y.Y.); (Y.L.); (Z.W.); (X.Z.)
| | - Zhaohai Wen
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (S.Y.); (Y.Y.); (Y.L.); (Z.W.); (X.Z.)
| | - Xiaoyong Zhang
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (S.Y.); (Y.Y.); (Y.L.); (Z.W.); (X.Z.)
| | - Xianghong Ju
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (S.Y.); (Y.Y.); (Y.L.); (Z.W.); (X.Z.)
- Marine Medical Research and Development Centre, Shenzhen Institute of Guangdong Ocean University, Shenzhen 518120, China
| | - Youquan Li
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China; (J.L.); (S.Y.); (Y.Y.); (Y.L.); (Z.W.); (X.Z.)
- Marine Medical Research and Development Centre, Shenzhen Institute of Guangdong Ocean University, Shenzhen 518120, China
| |
Collapse
|
14
|
Li J, Luo X, Shiu PHT, Cheng Y, Nie X, Rangsinth P, Lau BWM, Zheng C, Li X, Li R, Lee SMY, Fu C, Seto SW, Zhang J, Leung GPH. Protective effects of Amauroderma rugosum on dextran sulfate sodium-induced ulcerative colitis through the regulation of macrophage polarization and suppression of oxidative stress. Biomed Pharmacother 2024; 176:116901. [PMID: 38878683 DOI: 10.1016/j.biopha.2024.116901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/30/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Amauroderma rugosum (AR) is a medicinal mushroom commonly used to treat inflammation, gastric disorders, epilepsy, and cancers due to its remarkable anti-inflammatory and anti-oxidative properties. This study was designed to evaluate the pharmacological effects of AR and its underlying mechanism of action against ulcerative colitis (UC) in vitro and in vivo. METHODS A UC mouse model was established by administration of dextran sulfate sodium (DSS). AR extract was administered intragastrically to mice for 7 days. At the end of the experiment, histopathology, macrophage phenotype, oxidative stress, and inflammatory status were examined in vivo. Furthermore, RAW 264.7, THP-1, and Caco-2 cells were used to elucidate the mechanism of action of AR in vitro. RESULTS AR extract (0.5-2 mg/mL) significantly suppressed lipopolysaccharide (LPS) and interferon-gamma (IFN-γ)-induced M1 macrophage (pro-inflammatory) polarization in both RAW 264.7 and THP-1 cells. LPS-induced pro-inflammatory mediators (nitric oxide, TNF-α, IL-1β, MCP-1, and IL-6) were reduced by AR extract in a concentration-dependent manner. Similarly, AR extract downregulated MAPK signaling activity in LPS-stimulated RAW 264.7 cells. AR extract elicited a concentration-dependent increase in the mRNA expression of M2 (anti-inflammatory) phenotype markers (CD206, Arg-1, Fizz-1, and Ym-1) in RAW 264.7 cells. Moreover, AR extract suppressed DSS-induced ROS generation and mitochondrial dysfunction in Caco-2 cells. The in vivo experiment revealed that AR extract (200 mg/kg) increased colon length compared to the DSS-treated group. In addition, disease activity index, spleen ratio, body weight, oxidative stress, and colonic inflammation were markedly improved by AR treatment in DSS-induced UC mice. Finally, AR suppressed M1 and promoted M2 macrophage polarization in UC mice. CONCLUSION The AR extract protected against DSS-induced UC by regulating macrophage polarization and suppressing oxidative stress. These valuable findings suggest that adequate intake of AR can prevent and/or treat UC.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China; The Research Centre for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Xi Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Polly Ho-Ting Shiu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yanfen Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Nie
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Panthakarn Rangsinth
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Benson Wui Man Lau
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Chengwen Zheng
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xuebo Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Renkai Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Simon Ming-Yuen Lee
- Department of Food Science and Nutrition, Faculty of Science, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sai-Wang Seto
- Department of Food Science and Nutrition, Faculty of Science, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China; The Research Centre for Chinese Medicine Innovation, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China.
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
15
|
Chang YQ, Moon SK, Wang YQ, Xie LM, Cho HS, Kim SK. Supplemental effects of different production methods of pine needle additives on growth performance, intestinal environment, meat quality and serum of broiler chickens. Anim Biosci 2024; 37:1263-1276. [PMID: 38754853 PMCID: PMC11222840 DOI: 10.5713/ab.24.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 05/18/2024] Open
Abstract
OBJECTIVE Pine needles are rich in many nutrients and exhibit antibacterial and antioxidant biological activities; however, the effects of different production methods of pine needle additives on the growth performance and intestinal flora of broiler chickens are not known. METHODS Normal diets were supplemented with pine needle fermentation juice (PNF), pine needle soaking juice (PNS), or pine needle powder (PNP), and the associated effects on growth performance, relative organ weights, intestinal development, intestinal histological morphology, intestinal flora, meat quality, and serum indicators in broiler chickens were observed. RESULTS The results showed that PNF, PNS, and PNP all significantly improved feed utilisation and promoted the growth and development of broilers. All three additives also significantly improved the structure of the intestinal flora, specifically increasing the diversity of bacteria; increasing the abundance of beneficial bacteria, such as Faecalibacterium, Rikenella, and Blautia; and decreasing the abundance of harmful bacteria, such as Staphylococcus. The antioxidant properties of pine needles were also found to intensify lipid metabolic reactions in the blood, thus leading to lower triglycerides and total cholesterol. Meanwhile, high doses of PNF reduced jejunum and ileum weights and also increased meat yellowness. Lastly, none of PNF, PNS, or PNP had an effect on relative organ weights or intestinal histological morphology. CONCLUSION The addition of pine needles to the diet of broiler chickens can effectively promote their growth performance as well as improve their intestinal flora and serum status without side effects; in particular, the dose of 0.2% of either PNF and PNS is expected to have the capacity to replace growth-promoting antibiotics as diet additives.
Collapse
Affiliation(s)
- Yi-Qiang Chang
- Department of Animal Science and Technology, Konkuk University, Seoul 05029,
Korea
| | - Seung-Kyu Moon
- Department of Animal Science and Technology, Konkuk University, Seoul 05029,
Korea
| | - Yan-Qing Wang
- Department of Animal Science and Technology, Konkuk University, Seoul 05029,
Korea
| | - Liu-Ming Xie
- Department of Animal Science and Technology, Konkuk University, Seoul 05029,
Korea
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047,
China
| | - Hang-sul Cho
- Department of Animal Science and Technology, Konkuk University, Seoul 05029,
Korea
| | - Soo-Ki Kim
- Department of Animal Science and Technology, Konkuk University, Seoul 05029,
Korea
| |
Collapse
|
16
|
Ali Agha AS, Alshaer W, Aburjai T. Advancements and Challenges in Aptamer-Based Therapeutics and Diagnostics Across Diverse Medical Domains: A Comprehensive Review. JORDAN JOURNAL OF PHARMACEUTICAL SCIENCES 2024; 17:344-361. [DOI: 10.35516/jjps.v17i2.1895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Aptamers, which are single-stranded DNA or RNA molecules, are increasingly recognized as important tools in diagnostics and therapeutics across various medical disciplines such as oncology, respiratory diseases, and neurological disorders. This review provides a comprehensive evaluation of the recent progress and obstacles encountered in the field of aptamer-based applications. Aptamers have shown promise in oncology for early cancer detection and targeted drug delivery, effectively reducing off-target effects. They also hold potential for significantly impacting the management of respiratory conditions such as asthma and Chronic Obstructive Pulmonary Disease (COPD) by selectively targeting cytokines and regulating the inflammatory response. In the realm of neurological disorders, aptamers offer novel methods by influencing the gut-brain axis and proposing potential approaches for early detection and specific therapy. Despite these notable benefits, persistent challenges remain in areas such as molecular stability, delivery mechanisms, and economic viability. This review offers a comprehensive overview of aptamer-based diagnostics and therapeutics while exploring potential avenues for future research.
Collapse
|
17
|
Bai J, Wang Y, Li F, Wu Y, Chen J, Li M, Wang X, Lv B. Research advancements and perspectives of inflammatory bowel disease: A comprehensive review. Sci Prog 2024; 107:368504241253709. [PMID: 38778725 PMCID: PMC11113063 DOI: 10.1177/00368504241253709] [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] [Indexed: 05/25/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease with increasing incidence, such as Crohn's disease and ulcerative colitis. The accurate etiology and pathogenesis of IBD remain unclear, and it is generally believed that it is related to genetic susceptibility, gut microbiota, environmental factors, immunological abnormalities, and potentially other factors. Currently, the mainstream therapeutic drugs are amino salicylic acid agents, corticosteroids, immunomodulators, and biological agents, but the remission rates do not surpass 30-60% of patients in a real-life setting. As a consequence, there are many studies focusing on emerging drugs and bioactive ingredients that have higher efficacy and long-term safety for achieving complete deep healing. This article begins with a review of the latest, systematic, and credible summaries of the pathogenesis of IBD. In addition, we provide a summary of the current treatments and drugs for IBD. Finally, we focus on the therapeutic effects of emerging drugs such as microRNAs and lncRNAs, nanoparticles-mediated drugs and natural products on IBD and their mechanisms of action.
Collapse
Affiliation(s)
- Junyi Bai
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Ying Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Fuhao Li
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yueyao Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Meng Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xi Wang
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
18
|
Wen X, Peng H, Zhang H, He Y, Guo F, Bi X, Liu J, Sun Y. Wheat Bran Polyphenols Ameliorate DSS-Induced Ulcerative Colitis in Mice by Suppressing MAPK/NF-κB Inflammasome Pathways and Regulating Intestinal Microbiota. Foods 2024; 13:225. [PMID: 38254526 PMCID: PMC10814686 DOI: 10.3390/foods13020225] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/23/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Wheat bran (WB) is the primary by-product of wheat processing and contains a high concentration of bioactive substances such as polyphenols. This study analyzed the qualitative and quantitative components of polyphenols in wheat bran and their effects on ulcerative colitis (UC) using the dextran sulfate sodium (DSS)-induced colitis model in mice. The potential mechanism of wheat bran polyphenols (WBP) was also examined. Our findings indicate that the main polyphenol constituents of WBP were phenolic acids, including vanillic acid, ferulic acid, caffeic acid, gallic acid, and protocatechuic acid. Furthermore, WBP exerted remarkable protective effects against experimental colitis. This was achieved by reducing the severity of colitis and improving colon morphology. Additionally, WBP suppressed colonic inflammation via upregulation of the anti-inflammatory cytokine IL-10 and downregulation of pro-inflammatory cytokines (TNF-α, IL-6, IL-1β) in colon tissues. Mechanistically, WBP ameliorated DSS-induced colitis in mice by inhibiting activation of the MAPK/NF-κB pathway. In addition, microbiome analysis results suggested that WBP modulated the alteration of gut microbiota caused by DSS, with an enhancement in the ratio of Firmicutes/Bacteroidetes and adjustments in the number of Helicobacter, Escherichia-Shigella, Akkermansia, Lactobacillus, Lachnospiraceae_NK4A136_group at the genus level. To conclude, the findings showed that WBP has excellent prospects in reducing colonic inflammation in UC mice.
Collapse
Affiliation(s)
- Xusheng Wen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.W.); (Y.H.); (F.G.); (X.B.); (J.L.)
| | - Han Peng
- Department of Food Science and Technology, University of California, Davis, 1 Shields Ave., Davis, CA 95616, USA;
| | - Hua Zhang
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China;
| | - Yangzheng He
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.W.); (Y.H.); (F.G.); (X.B.); (J.L.)
| | - Fanghua Guo
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.W.); (Y.H.); (F.G.); (X.B.); (J.L.)
| | - Xin Bi
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.W.); (Y.H.); (F.G.); (X.B.); (J.L.)
| | - Jiahua Liu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.W.); (Y.H.); (F.G.); (X.B.); (J.L.)
| | - Yong Sun
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China; (X.W.); (Y.H.); (F.G.); (X.B.); (J.L.)
| |
Collapse
|
19
|
Alhameed RA, Semreen MH, Hamad M, Giddey AD, Sulaiman A, Al Bataineh MT, Al-Hroub HM, Bustanji Y, Alzoubi KH, Soares NC. Multi-Omics Profiling of Candida albicans Grown on Solid Versus Liquid Media. Microorganisms 2023; 11:2831. [PMID: 38137975 PMCID: PMC10745582 DOI: 10.3390/microorganisms11122831] [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: 10/21/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023] Open
Abstract
Candida albicans is a common pathogenic fungus that presents a challenge to healthcare facilities. It can switch between a yeast cell form that diffuses through the bloodstream to colonize internal organs and a filamentous form that penetrates host mucosa. Understanding the pathogen's strategies for environmental adaptation and, ultimately, survival, is crucial. As a complementary study, herein, a multi-omics analysis was performed using high-resolution timsTOF MS to compare the proteomes and metabolomes of Wild Type (WT) Candida albicans (strain DK318) grown on agar plates versus liquid media. Proteomic analysis revealed a total of 1793 proteins and 15,013 peptides. Out of the 1403 identified proteins, 313 proteins were significantly differentially abundant with a p-value < 0.05. Of these, 156 and 157 proteins were significantly increased in liquid and solid media, respectively. Metabolomics analysis identified 192 metabolites in total. The majority (42/48) of the significantly altered metabolites (p-value 0.05 FDR, FC 1.5), mainly amino acids, were significantly higher in solid media, while only 2 metabolites were significantly higher in liquid media. The combined multi-omics analysis provides insight into adaptative morphological changes supporting Candida albicans' life cycle and identifies crucial virulence factors during biofilm formation and bloodstream infection.
Collapse
Affiliation(s)
- Rouba Abdulsalam Alhameed
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates; (R.A.A.); (M.H.); (A.S.); (H.M.A.-H.); (Y.B.); (K.H.A.)
| | - Mohammad H. Semreen
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates; (R.A.A.); (M.H.); (A.S.); (H.M.A.-H.); (Y.B.); (K.H.A.)
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates
| | - Mohamad Hamad
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates; (R.A.A.); (M.H.); (A.S.); (H.M.A.-H.); (Y.B.); (K.H.A.)
- College of Health Sciences, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates
| | - Alexander D. Giddey
- Center for Applied and Translational Genomics, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates;
| | - Ashna Sulaiman
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates; (R.A.A.); (M.H.); (A.S.); (H.M.A.-H.); (Y.B.); (K.H.A.)
| | - Mohammad T. Al Bataineh
- Center for Biotechnology, Department of Molecular Biology and Genetics, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates;
| | - Hamza M. Al-Hroub
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates; (R.A.A.); (M.H.); (A.S.); (H.M.A.-H.); (Y.B.); (K.H.A.)
| | - Yasser Bustanji
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates; (R.A.A.); (M.H.); (A.S.); (H.M.A.-H.); (Y.B.); (K.H.A.)
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Karem H. Alzoubi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates; (R.A.A.); (M.H.); (A.S.); (H.M.A.-H.); (Y.B.); (K.H.A.)
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates
| | - Nelson C. Soares
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates; (R.A.A.); (M.H.); (A.S.); (H.M.A.-H.); (Y.B.); (K.H.A.)
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27227, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), 1649-016 Lisbon, Portugal
- Centre for Toxicogenomics and Human Health (ToxOmics), Faculdade de Lisboa, NOVA School, 1169-056 Lisbon, Portugal
| |
Collapse
|
20
|
Chu H, Liu W, Zhao C, Yin T, Shi J, Zhang W. Glycated Casein by TGase-Type Exerts Protection Potential against DSS-Induced Colitis via Inhibiting TLR4/NF-κB Signaling Pathways in C57BL/6J Mice. Foods 2023; 12:3431. [PMID: 37761139 PMCID: PMC10528845 DOI: 10.3390/foods12183431] [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: 08/26/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Glycation by transglutaminase (TGase)-type could effectively improve the structure and functional properties of proteins. However, the influence on intestinal inflammation or the underlying mechanisms has not been investigated. The goal of this research was to compare the bioactivities between glycated casein generated from the TGase-catalyzed reaction and oligochitosan as well as casein using a mouse model of dextran sulfate sodium (DSS)-induced intestinal inflammation to examine the protective effects and the underlying mechanism of glycated casein on intestinal inflammation. Eight groups of C57BL/6 mice were randomly assigned in this study: Control group: standard diet for 35 days; Model group: standard diet for 28 days and then colitis induction; Pretreated groups: different levels (200, 400, 800 mg/kg BW) of casein or glycated casein for 28 days before colitis induction. The mice were drinking water containing a 3% DSS solution for seven days of mice to cause colitis. The results indicated that glycated casein and casein at 200-800 mg/kg BW all relieved DSS-induced weight loss, reduced disease activity index (DAI) score, alleviated colon length shortening, weakened the destruction of colonic mucosal structure, decreased serum LPS, and MPO, IL-1β, IL-6 and TNF-α levels in serum and colon, as well as regulated the expression of proteins involved in the TLR4/NF-κB signaling pathway in a concentration-dependent manner. Glycated caseinate showed a better protective effect against DSS-induced colitis than casein, highlighting that the TGase-type glycation of proteins as a potential functional food ingredient might be a helpful method for gut health.
Collapse
Affiliation(s)
- Hui Chu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Weiling Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Cong Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Tong Yin
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Jia Shi
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
- Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Wei Zhang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
- Department of Food Science, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|