1
|
Liu Z, Wang M, Hu Y, Li J, Gong W, Guo X, Song S, Zhu B. Ulva lactuca polysaccharides combined with fecal microbiota transplantation ameliorated dextran sodium sulfate-induced colitis in C57BL/6J mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:422-432. [PMID: 39212113 DOI: 10.1002/jsfa.13839] [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: 03/22/2024] [Revised: 06/26/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) of healthy donors improves ulcerative colitis (UC) patients by restoring the balance of the gut microbiota. However, donors vary in microbial diversity and composition, often resulting in weak or even ineffective FMT. Improving the efficacy of FMT through combination treatment has become a promising strategy. Ulva lactuca polysaccharides (ULP) have been found to benefit host health by regulating gut microbiota. The effect of the combination of ULP and FMT in ameliorating UC has not yet been evaluated. RESULTS The present study found that supplementation with ULP combined with FMT showed better effects in ameliorating UC than supplementation with FMT alone. Results suggested that FMT or ULP combined with FMT alleviated the symptoms of UC in mice, as evidenced by prevention of body weight loss, improvement of disease activity index and protection of the intestinal mucus. Notably, ULP in combination with FMT was more effective than FMT in reducing levels of cytokines and related inflammatory enzymes. In addition, ULP combined with FMT effectively restored the dysbiosis induced by dextran sulfate sodium (DSS) and further enriched probiotics (such as Bifidobacterium). The production of short-chain fatty acids, especially acetic acid, was also significantly enriched by ULP combined with FMT. CONCLUSION Supplementation of ULP combined with FMT could significantly ameliorate DSS-induced colitis in mice by inhibiting inflammation and restoring dysbiosis of gut microbiota. These results suggested that ULP combined with FMT has potential application in ameliorating UC. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhengqi Liu
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
- National Engineering Research Center of Seafood, National and Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, School of Food Science and Technology, Dalian Polytechnic University, Dalian, PR China
| | - Menghui Wang
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
| | - Yuanyuan Hu
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
| | - Jinjin Li
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
| | - Wei Gong
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
| | - Xiaoming Guo
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
| | - Shuang Song
- National Engineering Research Center of Seafood, National and Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, School of Food Science and Technology, Dalian Polytechnic University, Dalian, PR China
| | - Beiwei Zhu
- Shenzhen Key Laboratory of Food Nutrition and Health, College of Chemistry and Environmental Engineering, Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, PR China
- National Engineering Research Center of Seafood, National and Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, School of Food Science and Technology, Dalian Polytechnic University, Dalian, PR China
| |
Collapse
|
2
|
Biedermann L, Doulberis M, Schreiner P, Nielsen OH, The FO, Brand S, Burk S, Hruz P, Juillerat P, Krieger-Grübel C, Leu K, Leventhal GE, Misselwitz B, Scharl S, Schoepfer A, Seibold F, Herfarth H, Rogler G. Efficacy and Safety of Anthocyanin-Rich Extract in Patients with Ulcerative Colitis: A Randomized Controlled Trial. Nutrients 2024; 16:4197. [PMID: 39683589 DOI: 10.3390/nu16234197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Bilberries are effective in inducing clinical, endoscopic, and biochemical improvement in ulcerative colitis (UC) patients. The aim of this study was to investigate the efficacy of anthocyanin-rich extract (ACRE), the bioactive ingredient of bilberries, in a controlled clinical trial in moderate-to-severe UC. Methods: A multi-center, randomized, placebo-controlled, double-blind study with a parallel group was conducted. Initially, the study was planned for 100 patients; nevertheless, it prematurely ended due to COVID-19. Patients had moderate-to-severe active UC at screening (a Mayo score of 6-12, an endoscopic sub-score ≥ 2) and were randomized at baseline. The primary endpoint was a clinical response (week 8, a total Mayo score reduction ≥ 3 points). Fecal calprotectin (FC) and a centrally read endoscopic response were among the secondary endpoints. Results: Out of 48 patients (6 Swiss centers), 34 were randomized. Eighteen ACRE and eight placebo patients could be analyzed (per protocol set). Half (9/18) of ACRE patients and 3/8 of placebo patients responded clinically (p = 0.278). An improvement in the Mayo score was observed in the ACRE arm (77.8% vs. 62.5% placebo). FC dropped from 1049 ± 1139 to 557 ± 756 μg/g for ACRE but not for the placebo group (947 ± 1039 to 1040 ± 1179; p = 0.035). Serious adverse events were rare. Conclusions: ACRE treatment did not yield significant superiority to the placebo. Furthermore, the placebo response was unusually high. Moreover, there was a significant calprotectin decrease at the end of treatment, indicative of ACRE efficacy in UC.
Collapse
Affiliation(s)
- Luc Biedermann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Michael Doulberis
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
- Gastroklinik, Private Gastroenterological Practice, 8810 Horgen, Switzerland
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland
| | - Philipp Schreiner
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Ole Haagen Nielsen
- Department of Gastroenterology, Herlev and Gentofte Hospital, University of Copenhagen, 2730 Herlev, Denmark
| | - Frans Olivier The
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Stephan Brand
- Department of Gastroenterology and Hepatology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
| | - Sabine Burk
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Petr Hruz
- Department of Gastroenterology, Clarunis-University Center for Gastrointestinal and Liver Diseases, 4052 Basel, Switzerland
| | | | - Claudia Krieger-Grübel
- Department of Gastroenterology and Hepatology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
| | - Kristin Leu
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Gabriel E Leventhal
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Benjamin Misselwitz
- Department of Visceral Surgery and Medicine, Inselspital Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Sylvie Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Alain Schoepfer
- Department of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, 1011 Lausanne, Switzerland
| | - Frank Seibold
- Intesto Crohn and Colitis Center, 3012 Bern, Switzerland
| | - Hans Herfarth
- Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
3
|
Liu X, Ma Y, Guan K, Liu R, Mao K, Xu X, Li Q, Wang R. Intestinal barrier, immunity and gut microbiota-based protective effects of Lactococcus lactis HF08 and its postbiotic derivative on aging and aging colitis mice. Food Res Int 2024; 197:115164. [PMID: 39593375 DOI: 10.1016/j.foodres.2024.115164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 11/28/2024]
Abstract
The prevalence and severity of gastrointestinal diseases were increased with age. In this study, the intestinal protective effects of Lactococcus lactis HF08 (HF08) and its derived postbiotic (P-HF08) on D-gal-induced aging mice and D-gal/DSS-induced aging colitis mice were investigated. In D-gal-induced aging mice, both HF08 and P-HF08 alleviated aging-related intestinal barrier dysfunction, inflammatory status, and gut microbiota disorder. The effects of probiotic HF08 were superior to those of postbiotic P-HF08, attributed to ability of HF08 to regulate the gut microbiota. However, in D-gal/DSS-induced aging colitis mice, the effects of P-HF08 on colitis surpassed that of HF08. Specifically, both HF08 and P-HF08 could reduce symptoms of age-related colitis, including reduction of lose weight, the DAI score, colonic shortening, and colon tissue damage. The inhibitory effects of P-HF08 on intestinal inflammation surpassed those of HF08, as evidenced by the levels of colon IL-6, IL-1β, and IL-10. Western blot results demonstrated that the anti-inflammatory effects of P-HF08 were attributed to the downregulation of key proteins in the TLR4/NF-κB pathway. And four potential TLR4 inhibitors were identified from HF08 metabolites (eplerenone, genistein, indoleacrylic acid, and turanose) by molecular docking. Nevertheless, HF08 could better regulate gut microbiota and metabolite in aging-related colitis than P-HF08, which was consistent with the results on aging mice. Overall, our finding revealed that when the intestinal barrier was intact (aging), probiotics showed superior regulation of intestinal microbiota, while postbiotics offered greater safety in case of intestinal barrier damage (aging colitis). This study offered a novel perspective into the applications of probiotics and their derivatives in the aging related gastrointestinal diseases adjuvant therapy.
Collapse
Affiliation(s)
- Xiaolin Liu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Ying Ma
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Kaifang Guan
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Rongmei Liu
- Dairy Nutrition and Function, Key Laboratory of Sichuan Province, New Hope Dairy Company Limited, Chengdu 610023, China; Sichuan Engineering Laboratory for High-quality Dairy Product Preparation and Quality Control Technology, Chengdu 610000, Sichuan, China; Chengdu Molecular Power Biotechnology Co., Ltd., Chengdu 611732, Sichuan, China
| | - Kaidong Mao
- Jiangsu HOWYOU Biotechnology Company Limited, Shanghai 310000, China
| | - Xiaogang Xu
- Jiangsu HOWYOU Biotechnology Company Limited, Shanghai 310000, China
| | - Qiming Li
- Dairy Nutrition and Function, Key Laboratory of Sichuan Province, New Hope Dairy Company Limited, Chengdu 610023, China; Sichuan Engineering Laboratory for High-quality Dairy Product Preparation and Quality Control Technology, Chengdu 610000, Sichuan, China; Chengdu Molecular Power Biotechnology Co., Ltd., Chengdu 611732, Sichuan, China.
| | - Rongchun Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
4
|
Du L, Ding X, Tian Y, Chen J, Li W. Effect of anthocyanins on metabolic syndrome through interacting with gut microbiota. Pharmacol Res 2024; 210:107511. [PMID: 39577753 DOI: 10.1016/j.phrs.2024.107511] [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: 08/18/2024] [Revised: 10/22/2024] [Accepted: 11/16/2024] [Indexed: 11/24/2024]
Abstract
Metabolic syndrome, as a complex pathological condition, is caused by a series of pathogenic factors and has become a global public health challenge. Anthocyanins, a natural water-soluble flavonoid pigment, have attracted much attention due to their antioxidant, anti-inflammatory, and anticancer biological activities. After ingestion, a majority of anthocyanins is not directly absorbed but rather reaches the colon. Hence, the exertion of their biological benefits is closely intertwined with the role played by gut microbiota. In this review, we introduce the pathogenesis and intervention methods of metabolic syndrome, as well as the interaction between anthocyanins and gut microbiota. We also discuss the therapeutic potential of anthocyanins through gut microbiota in addressing a range of metabolic syndrome conditions, including obesity, type 2 diabetes mellitus, cardiovascular diseases, non-alcoholic fatty liver disease, inflammatory bowel disease, polycystic ovary syndrome, osteoporosis, and cancer.
Collapse
Affiliation(s)
- Lanlan Du
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry and Grassland, Nanjing Forestry University, Nanjing 210037, China
| | - Xiaoqin Ding
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Yuwen Tian
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China
| | - Jian Chen
- Jiangsu Key Laboratory for the Research and Utilization of Plant Resources, Institute of Botany, Jiangsu Province and Chinese Academy of Sciences, Nanjing 210014, China.
| | - Weilin Li
- State Key Laboratory of Tree Genetics and Breeding, Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry and Grassland, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
5
|
Zobrist Y, Doulberis M, Biedermann L, Leventhal GE, Rogler G. Anthocyanin-Rich Extract Mitigates the Contribution of the Pathobiont Genus Haemophilus in Mild-to-Moderate Ulcerative Colitis Patients. Microorganisms 2024; 12:2376. [PMID: 39597764 PMCID: PMC11596099 DOI: 10.3390/microorganisms12112376] [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: 10/08/2024] [Revised: 10/30/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024] Open
Abstract
Anthocyanins (ACs) have been shown to elicit anti-inflammatory and antioxidant effects in animal models of ulcerative colitis (UC). Furthermore, we previously observed in a double-blind randomized trial in UC patients that biochemical disease activity tended to be lower in patients that were exposed to AC. Here, we report on the changes in the fecal microbiome composition in these patients upon AC exposure. UC patients received a 3 g daily dose of an AC-rich bilberry extract (ACRE) for eight weeks. We determined the microbiome composition in longitudinal stool samples from 24 patients and quantified the degree of change over time. We also correlated the relative abundances of individual microbial taxa at different timepoints to fecal concentrations of calprotectin, a proxy for inflammation. Microbiome composition did not change over time as a result of the intervention, in terms of both alpha and beta diversity. However, before the intervention, the abundance of Haemophilus parainfluenzae was positively correlated with fecal calprotectin concentrations, and this correlation persisted in placebo-treated subjects throughout the study. In contrast, the correlation between H. parainfluenzae and calprotectin vanished in ACRE-treated subjects, while the relative abundance of H. parainfluenzae did not change. Our results suggest that ACRE treatment mitigates the contribution of H. parainfluenzae to inflammation. Further research is warranted to better comprehend the role of microbial composition in response to medical therapy including AC-rich extract in UC patients.
Collapse
Affiliation(s)
| | - Michael Doulberis
- Department of Gastroenterology and Hepatology, Department of Medicine, Zurich University Hospital, 8091 Zurich, Switzerland; (M.D.); (L.B.)
- Gastroklinik, Private Gastroenterological Practice, 8810 Horgen, Switzerland
- Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001 Aarau, Switzerland
| | - Luc Biedermann
- Department of Gastroenterology and Hepatology, Department of Medicine, Zurich University Hospital, 8091 Zurich, Switzerland; (M.D.); (L.B.)
| | | | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, Department of Medicine, Zurich University Hospital, 8091 Zurich, Switzerland; (M.D.); (L.B.)
| |
Collapse
|
6
|
Xu Q, Li Y, Qin X, Xin Y, Wang J, Zhang Y, Xu K, Yang X, Wang X. osa-miR168a, a Plant miRNA That Survives the Process of In Vivo Food Digestion, Attenuates Dextran Sulfate Sodium-Induced Colitis in Mice by Oral Administration. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25146-25160. [PMID: 39480689 DOI: 10.1021/acs.jafc.4c07283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Previous studies showed that osa-miR168a, a plant miRNA rich in fruits and vegetables, had cross-kingdom biological effects on immunocytes, silkworms, and rodents. In this study, the effects of miR168a on mouse colitis induced by dextran sulfate sodium (DSS) were investigated. The results showed that miR168a oligomers were resistant during the process of food digestion, ending up with a residual concentration of 67.8 ± 11.2 fM in mouse intestines 4 h after oral gavage. More importantly, direct oral administration of the miRNA to the colitis mice significantly ameliorated the progression of the disease, as evidenced by the reduction in DAI score, histopathological lesions, and proinflammatory cytokines. Repairing intestinal barrier function by promoting the regeneration of TJ proteins and the mucus layer, suppressing oxidative stress and colonic inflammation via modulating Nrf2 and NF-κB signaling pathways, and restoring the imbalanced gut microbiota caused by DSS are proposed mechanisms behind the anticolitis activity of miR168a. This study provided new evidence of the cross-kingdom regulatory effects of dietary miRNAs, suggesting the potential of the plant miRNA for the prevention and treatment of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Qin Xu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Ying Li
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Xinshu Qin
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yirao Xin
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Jianing Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Yi Zhang
- Department of Food Science, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Ke Xu
- Department of Joint Surgery, Hong Hui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Xingbin Yang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| | - Xingyu Wang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710062, China
| |
Collapse
|
7
|
Jeong JS, Baek GH, Kim JW, Kim JH, Chung EH, Ko JW, Kwon MJ, Kim SK, Lee SH, Kim JS, Kim TW. Korean Red Ginseng alleviates dextran sodium sulfate-induced colitis through gut microbiota modulation in mice. J Ginseng Res 2024; 48:581-591. [PMID: 39583173 PMCID: PMC11584195 DOI: 10.1016/j.jgr.2024.08.001] [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: 03/12/2024] [Revised: 07/25/2024] [Accepted: 08/08/2024] [Indexed: 11/26/2024] Open
Abstract
Background There is a growing interest in understanding the association between the gut microbiota and inflammatory bowel disease (IBD). Natural compounds, such as Korean Red Ginseng (KRG), show promise for IBD treatment because of their ability to influence gut microbiota. This study explored the effects of KRG on gut microbiota modulation and subsequent intestinal epithelial cell regeneration in an experimental colitis model. Method Using a mouse model of colitis induced by 2 % dextran sodium sulfate, the study administered 200 or 400 mg/kg/day of KRG to evaluate its biological effects. Colitis symptoms were assessed through body weight, disease activity index, colon length, and histological analysis. The microbial composition in the fecal was determined using 16S rRNA sequencing. To evaluate regeneration signals in the colon, western blotting and immunohistochemistry assays were conducted. Result Administration of KRG effectively mitigated colitis symptoms in mice, as indicated by histological examination showing alleviated epithelial damage and inflammation, along with increased mucus production. Microbiota analysis showed that KRG significantly altered microbial diversity, favoring beneficial taxa and suppressing harmful taxa. Moreover, ameliorated β-catenin/transcription factor-4 protein expression, a key signal associated with epithelial cell regeneration, was observed in the KRG treated groups, accompanied by improved intestinal linings. Conclusion These findings suggest that KRG exerts biological effects in colitis by modulating gut microbiota and creating a favorable intestinal environment, thereby reducing regenerative signals. Further research is warranted to elucidate the cellular and molecular mechanisms underlying the interaction of KRG with gut microbiota and pave the way for effective IBD therapies.
Collapse
Affiliation(s)
- Ji-Soo Jeong
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Ga-Hyeon Baek
- Department of Nano-Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Jeong-Won Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Jin-Hwa Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Eun-Hye Chung
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Je-Won Ko
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| | - Mi-Jin Kwon
- R&D Headquarters, Korea Ginseng Corporation, Gwacheon, Republic of Korea
| | - Sang-Kyu Kim
- R&D Headquarters, Korea Ginseng Corporation, Gwacheon, Republic of Korea
| | - Seung-Ho Lee
- R&D Headquarters, Korea Ginseng Corporation, Gwacheon, Republic of Korea
| | - Jun-Seob Kim
- Department of Nano-Bioengineering, Incheon National University, Incheon, Republic of Korea
- Institute for New Drug Development, College of Life Science and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Tae-Won Kim
- College of Veterinary Medicine (BK21 FOUR Program), Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
8
|
Qian S, Zhang X, Zheng X, Li R, Hao X, Tang Z, Yang Z, Sun A, Guo S, Song Y, Zhang Z, Song X, Yu L. Development of interleukin-27 recombinant Lactococcus lactis and its efficacy in treating psoriasis and colitis in mice. Int J Biol Macromol 2024; 282:137113. [PMID: 39486722 DOI: 10.1016/j.ijbiomac.2024.137113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Psoriasis and inflammatory bowel disease (IBD) are chronic immune-mediated diseases that adversely affect patients' quality of life. Interleukin (IL)-27 plays an important role in a variety of infectious diseases, autoimmune disorders, and cancers. However, its therapeutic effects in psoriasis and colitis remain underexplored. In this study, we evaluated the therapeutic potential of recombinant Lactococcus lactis (L. lactis) expressing IL-27 (pIL-27) in imiquimod-induced psoriasis and dextran sodium sulfate-induced colitis mouse models. In the psoriasis mouse model, oral administration of pIL-27 significantly reduced skin scaling, mitigated weight loss, lowered psoriasis area and severity index scores, diminished epidermal hyperplasia and inflammatory cell infiltration, and decreased inflammatory cytokine levels. In the colitis mouse model, oral administration of pIL-27 alleviated weight loss, improved disease activity index scores, prevented colon shortening, ameliorated histopathological changes, and decreased inflammatory cytokine levels. Furthermore, recombinant L. lactis expressing IL-27 could modulate the gut microbiota, increasing the amount of beneficial bacteria and reducing harmful bacteria in the intestine, thereby alleviating the progression of psoriasis and colitis. These results suggest the potential of IL-27 as a therapeutic option for treating psoriasis and IBD.
Collapse
Affiliation(s)
- Shaoju Qian
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China
| | - Xingyi Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Xiaoxiao Zheng
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Ruixue Li
- Department of Otolaryngology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Xiaoling Hao
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China; Department of Endocrine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhou Tang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Zishan Yang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China
| | - Aiping Sun
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China
| | - Sheng Guo
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China
| | - Yihang Song
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Zihan Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China
| | - Xiangfeng Song
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China
| | - Lili Yu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan 453003, China; Department of Endocrine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
9
|
Sun R, Yu P, Guo L, Huang Y, Nie Y, Yang Y. Improving the growth and intestinal colonization of Escherichia coli Nissle 1917 by strengthening its oligopeptides importation ability. Metab Eng 2024; 86:157-171. [PMID: 39389255 DOI: 10.1016/j.ymben.2024.10.002] [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: 06/12/2024] [Revised: 10/01/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
Escherichia coli Nissle 1917 (EcN), the probiotic featured with well-established safety in different host, is emerging as a favored chassis for the construction of engineered probiotics for disease treatment. However, limited by the low intestinal colonization ability of EcN, repeated administration is required to maximize the health benefits of the EcN-derived engineered probiotics. Here, using fecal metabolites as "metabolites pool", we developed a metabolomic strategy to characterize the comprehensive metabolic profile of EcN. Compared with Prevotella copri DSM 18205 (P. copri), one of the dominant microbes in gut flora, EcN exhibited minor growth advantage under the fecal metabolites-containing condition for its lower metabolic capability towards fecal metabolites. Further study indicated that EcN lacked the ability to import the oligopeptides containing more than two amino acids. The shortage of oligopeptides-derived amino acids might limit the growth of EcN by restricting its purine metabolism. Assisted with the bioinformatic and qRT-PCR analyses, we identified a tripeptides-specific importer Pc-OPT in P. copri, which was mainly distributed in genera Prevotella and Bacteroides. Overexpression of Pc-OPT improved the tripeptides importation of EcN and promoted its growth and intestinal colonization. Notably, 16S rRNA gene amplicon sequencing results indicated that strengthening the oligopeptides importation ability of EcN might promote its intestinal colonization by adjusting the gut microbial composition. Our study reveals that the growth and intestinal colonization of EcN is limited by its insufficient oligopeptides importation and paves road for promoting the efficacy of the EcN-derived synthetic probiotics by improving their intestinal colonization ability.
Collapse
Affiliation(s)
- Ruxue Sun
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Peijun Yu
- Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liying Guo
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Yufei Huang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Yanhong Nie
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 201602, China; Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yunpeng Yang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou, 225009, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 201602, China; Institute of Neuroscience, CAS Key Laboratory of Primate Neurobiology, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
10
|
Qin B, Li Z, Zhu Q, Chen T, Lan W, Cui Y, Azad MAK, Kong X. Dietary Fermented Blueberry Pomace Supplementation Improves Small Intestinal Barrier Function and Modulates Cecal Microbiota in Aged Laying Hens. Animals (Basel) 2024; 14:2786. [PMID: 39409735 PMCID: PMC11475786 DOI: 10.3390/ani14192786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
This study aimed to investigate the effects of fermented blueberry pomace (FBP) on the intestinal barrier function and cecal microbiome of aged laying hens. A total of 320 Yukou Jingfen No. 8 laying hens (345-day-old) were randomly divided into a control group, 0.25% FBP group, 0.5% FBP group, or 1.0% FBP group. The results showed that the villus height (VH) in the jejunum of the 0.25-0.5% FBP groups and villus surface area in the jejunum of the 0.25% FBP group were higher (p < 0.05), while 0.25% FBP supplementation displayed a higher (p = 0.070) VH in the ileum compared to the control group. Mucin-2 expression was upregulated (p < 0.05) in the jejunum of the 0.5% FBP group and the ileum of the 0.25-0.5% FBP groups. Compared to the control group, interleukin (IL)-4 and IL-13 expressions were upregulated (p < 0.05) in the 1.0% FBP group. Microbiota analysis revealed that Prevotella abundance in the cecum of the 0.5-1.0% FBP groups was higher (p < 0.05) than in the 0.25% FBP group. In addition, microbial function prediction analysis showed that cecal microbiota in the 0.25% FBP group were mainly enriched by alanine/aspartate/glutamate metabolism and methane metabolism. Moreover, Spearman's correlation analysis revealed the potential correlations between the abundance of the cecal microbiota and intestinal-barrier-function-related gene expressions, as well as the short-chain fatty acid content, of laying hens. In summary, dietary FBP supplementation enhanced intestinal barrier function by improving intestinal morphology, upregulating gene expressions related to barrier function, and altering the cecal microbiota of aged laying hens.
Collapse
Affiliation(s)
- Binghua Qin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang 236037, China; (W.L.); (Y.C.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhihua Li
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
| | - Qian Zhu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ting Chen
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Lan
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang 236037, China; (W.L.); (Y.C.)
| | - Yadong Cui
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang 236037, China; (W.L.); (Y.C.)
| | - Md. Abul Kalam Azad
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiangfeng Kong
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutrition Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (B.Q.); (Z.L.); (Q.Z.); (T.C.)
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang 236037, China; (W.L.); (Y.C.)
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
11
|
Wu L, Hu Z, Luo X, Ge C, Lv Y, Zhan S, Huang W, Shen X, Yu D, Liu B. Itaconic Acid Alleviates Perfluorooctanoic Acid-Induced Oxidative Stress and Intestinal Damage by Regulating the Keap1/Nrf2/Ho-1 Pathway and Reshaping the Gut Microbiota. Int J Mol Sci 2024; 25:9826. [PMID: 39337313 PMCID: PMC11432532 DOI: 10.3390/ijms25189826] [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: 07/22/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
Itaconic acid (IA) is recognized for its potential application in treating intestinal diseases owing to the anti-inflammatory and antioxidant properties. Perfluorooctanoic acid (PFOA) can accumulate in animals and result in oxidative and inflammatory damages to multi-tissue and organ, particularly in the intestinal tract. This study aimed to explore whether IA could mitigate intestinal damage induced by PFOA exposure in laying hens and elucidate its potential underlying mechanisms. The results showed that IA improved the antioxidant capacity of laying hens and alleviated the oxidative damage induced by PFOA, as evidenced by the elevated activities of T-SOD, GSH-Px, and CAT, and the decreased MDA content in both the jejunum and serum. Furthermore, IA improved the intestinal morphological and structural integrity, notably attenuating PFOA-induced villus shedding, length reduction, and microvillus thinning. IA also upregulated the mRNA expression of ZO-1, Occludin, Claudin-1, and Mucin-2 in the jejunum, thereby restoring intestinal barrier function. Compared with the PF group, IA supplementation downregulated the gene expression of Keap1 and upregulated the HO-1, NQO1, SOD1, and GPX1 expression in the jejunum. Meanwhile, the PF + IA group exhibited lower expressions of inflammation-related genes (NF-κB, IL-1β, IFN-γ, TNF-α, and IL-6) compared to the PF group. Moreover, IA reversed the PFOA-induced imbalance in gut microbiota by reducing the harmful bacteria such as Escherichia-Shigella, Clostridium innocuum, and Ruminococcus torques, while increasing the abundance of beneficial bacteria like Lactobacillus. Correlation analysis further revealed a significant association between gut microbes, inflammatory factors, and the Keap1/Nrf2/HO-1 pathway expression. In conclusion, dietary IA supplementation could alleviate the oxidative and inflammatory damage caused by PFOA exposure in the intestinal tract by reshaping the intestinal microbiota, modulating the Keap1/Nrf2/HO-1 pathway and reducing oxidative stress and inflammatory response, thereby promoting intestinal homeostasis.
Collapse
Affiliation(s)
- Lianchi Wu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhaoying Hu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyu Luo
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chaoyue Ge
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yujie Lv
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shenao Zhan
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weichen Huang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyu Shen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dongyou Yu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- ZJU-Xinchang Joint Innovation Centre (TianMu Laboratory), Gaochuang Hi-Tech Park, Shaoxing 312500, China
| | - Bing Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- ZJU-Xinchang Joint Innovation Centre (TianMu Laboratory), Gaochuang Hi-Tech Park, Shaoxing 312500, China
| |
Collapse
|
12
|
Li W, Tang X, Liu H, Liu K, Tian Z, Zhao Y. Protective effect of 1,3-dioleoyl-2-palmitoylglycerol against DSS-induced colitis via modulating gut microbiota and maintaining intestinal epithelial barrier integrity. Food Funct 2024; 15:8700-8711. [PMID: 39076044 DOI: 10.1039/d4fo02344g] [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: 07/31/2024]
Abstract
Inflammatory bowel disease (IBD) is a challenging condition to cure that can occur at any age. The gut microbiome and intestinal epithelial barrier play a crucial role in the development of IBD. 1,3-Dioleoyl-2-palmitoylglycerol (OPO), the predominant triglyceride in breast milk, is a structural lipid with multiple physiological functions. However, the protective effect of OPO on IBD and its underlying mechanism remains unclear. This study showed that oral administration of OPO markedly ameliorated dextran sulfate sodium (DSS)-induced colitis phenotypes. OPO treatment reduced inflammation levels by suppressing the TLR4-MyD88-NF-κB signaling pathway in colitis mice. Furthermore, OPO treatment improved intestinal epithelial barrier function via promoting epithelial cell proliferation and differentiation, inhibiting cell apoptosis, and upregulating tight junction protein expression. The 16S rRNA gene sequencing revealed that OPO treatment restored microbial alpha diversity and reshaped the microbiota of colitis mice. Therefore, our study revealed that OPO exhibited a protective role in DSS-induced colitis via maintaining intestinal epithelial barrier integrity and modulating gut microbiota. Our results highlight that OPO could be used as effective supplements for individuals with IBD or intestinal dysfunctions.
Collapse
Affiliation(s)
- Wusun Li
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards & Testing Technology for Agro-Products of Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Xiaoyan Tang
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards & Testing Technology for Agro-Products of Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Hui Liu
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards & Testing Technology for Agro-Products of Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Ke Liu
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards & Testing Technology for Agro-Products of Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Zhiqing Tian
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards & Testing Technology for Agro-Products of Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Yujie Zhao
- Key Laboratory of Agro-Product Quality and Safety, Institute of Quality Standards & Testing Technology for Agro-Products of Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
13
|
Weng H, Zeng H, Wang H, Chang H, Zhai Y, Li S, Han Z. Differences in Lactation Performance, Rumen Microbiome, and Metabolome between Montbéliarde × Holstein and Holstein Cows under Heat Stress. Microorganisms 2024; 12:1729. [PMID: 39203571 PMCID: PMC11357101 DOI: 10.3390/microorganisms12081729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/03/2024] Open
Abstract
Heat stress negatively affects lactation performance and rumen microbiota of dairy cows, with different breeds showing varying levels of heat tolerance. This study aimed to compare the lactation performance of Montbéliarde × Holstein (MH, n = 13) and Holstein (H, n = 13) cows under heat stress, and 16S rRNA sequencing and liquid chromatography-mass spectrometry (LC-MS) were used to determine the rumen microbiome and metabolome in experimental cows. The results indicated that during heat stress, milk yield (p = 0.101), milk fat yield, milk protein yield, milk protein, and milk lactose (p < 0.05) in Montbéliarde × Holstein cows were higher than those in Holstein cows, whereas milk yield variation and somatic cell counts (p < 0.05) were lower than those in Holstein cows. The sequencing results indicated that the rumen of Montbéliarde × Holstein cows was significantly enriched with beneficial bacteria, such as Rikenellaceae, Allobaculum, and YRC22 (p < 0.05). In addition, correlations were observed between specific ruminal bacteria and lactation performance. Ruminal metabolites related to antioxidant and anti-inflammatory properties were significantly higher (p < 0.05) in Montbéliarde × Holstein cows than in Holstein cows. Overall, Montbéliarde × Holstein cows showed higher production efficiency under heat stress, which may be related to the different rumen mechanisms of crossbred and Holstein cows in adapting to heat stress.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhaoyu Han
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
14
|
Sun S, Zhang G, Lv S, Sun J. Potential mechanisms of traditional Chinese medicine in the treatment of liver cirrhosis: a focus on gut microbiota. Front Microbiol 2024; 15:1407991. [PMID: 39234554 PMCID: PMC11371771 DOI: 10.3389/fmicb.2024.1407991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Cirrhosis, a pathological stage that develops from various chronic liver diseases, is characterized by liver fibrosis, pseudolobular formation, and chronic inflammation. When it progresses to the decompensated phase, the mortality rate of cirrhosis can reach 80%. The role of gut microbiota in the progression of liver diseases has received significant attention. Numerous studies have shown that regulating gut microbiota has significant therapeutic effects on preventing and reversing liver cirrhosis. This article reviewed the mechanisms by which gut microbiota influence liver cirrhosis, explaining the effective therapeutic effects of traditional Chinese medicine. Through multi-directional regulation involving signaling pathways, gut microbiota diversity, and restoration of intestinal barrier function, traditional Chinese medicine has been promising in ameliorating liver cirrhosis, providing treatment options and pharmacological guidance for the occurrence and development of liver cirrhosis.
Collapse
Affiliation(s)
- Siyuan Sun
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Guangheng Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shimeng Lv
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinhui Sun
- Gastroenterology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
15
|
Li A, Liu A, Wang J, Song H, Luo P, Zhan M, Zhou X, Chen L, Zhang L. The prophylaxis functions of Lactobacillus fermentum GLF-217 and Lactobacillus plantarum FLP-215 on ulcerative colitis via modulating gut microbiota of mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:5816-5825. [PMID: 38406876 DOI: 10.1002/jsfa.13410] [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: 07/27/2023] [Revised: 12/09/2023] [Accepted: 02/18/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND The strong connection between gut microbes and human health has been confirmed by an increasing number of studies. Although probiotics have been found to relieve ulcerative colitis, the mechanism varies by the species involved. In this study, the physiological, immune and pathological factors of mice were measured and shotgun metagenomic sequencing was conducted to investigate the potential mechanisms in preventing ulcerative colitis. RESULTS The results demonstrated that ingestion of Lactobacillus fermentum GLF-217 and Lactobacillus plantarum FLP-215 significantly alleviated ulcerative colitis induced by dextran sulfate sodium (DSS), as evidenced by the increase in body weight, food intake, water intake and colon length as well as the decrease in disease activity index, histopathological score and inflammatory factor. Both strains not only improved intestinal mucosa by increasing mucin-2 and zonula occludens-1, but also improved the immune system response by elevating interleukin-10 levels and decreasing the levels of interleukin-1β, interleukin-6, tumor necrosis factor-α and interferon-γ. Moreover, L. fermentum GLF-217 and L. plantarum FLP-215 play a role in preventing DSS-induced colitis by regulating the structure of gut microbiota and promoting the formation of short-chain fatty acids. CONCLUSIONS This study may provide a reference for the prevention strategy of ulcerative colitis. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ao Li
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
| | | | - Jun Wang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
| | - Hainan Song
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
| | | | | | | | | | - Lin Zhang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
| |
Collapse
|
16
|
Wang D, Chen W, Cao J, Si L, Chen Z. Establishment and Evaluation of a Mouse Model of Experimental Ulcerative Colitis Induced by the Gavage Administration of Dextran Sulfate Sodium. Biomedicines 2024; 12:1764. [PMID: 39200228 PMCID: PMC11352140 DOI: 10.3390/biomedicines12081764] [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: 07/09/2024] [Revised: 07/22/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Given the critical role of dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) mouse models in the appraisal of associated therapeutic drugs, the optimization of the administration method and dosages is of paramount importance. Therefore, UC was induced in mice through the gavage administration of a DSS solution instead of free drinking water. The effects of varying daily dosages (2, 4, 6, and 8 g/kg) and frequencies (once or twice) of administration on the body weight and survival rate of the model mice were evaluated. Concurrently, the inflammatory indicators and tissue sections of the model mice were thoroughly evaluated. The results revealed that when the daily dosage reached 8 g/kg, the dosage exhibited a high level of toxicity, resulting in a high mortality rate among the mice. The DSS administration of 6 g/kg*2 not only elicited conspicuous symptoms, significant weight loss, substantial shortening of the colon, and significant changes in various inflammatory indicators, such as myeloperoxidase (MPO), nitric oxide (NO), reactive oxygen species (ROS), and glutathione (GSH), but it also maintained a high survival rate in the UC mice. The findings from this experiment lay a solid experimental foundation for future research on drugs intended for the treatment of UC.
Collapse
Affiliation(s)
- Dan Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (D.W.); (W.C.); (J.C.)
| | - Wei Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (D.W.); (W.C.); (J.C.)
| | - Jie Cao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (D.W.); (W.C.); (J.C.)
| | - Luqin Si
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Zehong Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (D.W.); (W.C.); (J.C.)
| |
Collapse
|
17
|
Li J, Jia J, Teng Y, Xie C, Li C, Zhu B, Xia X. Gastrodin Alleviates DSS-Induced Colitis in Mice through Strengthening Intestinal Barrier and Modulating Gut Microbiota. Foods 2024; 13:2460. [PMID: 39123651 PMCID: PMC11311408 DOI: 10.3390/foods13152460] [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: 05/07/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Inflammatory bowel diseases (IBDs) are commonly associated with dysfunctional intestinal barriers and disturbed gut microbiota. Gastrodin, a major bioactive ingredient of Gastrodia elata Blume, has been shown to exhibit anti-oxidation and anti-inflammation properties and could mitigate non-alcoholic fatty liver disease, but its role in modulating IBD remains elusive. The aim of this study was to investigate the impact of gastrodin on DSS-induced colitis in mice and explore its potential mechanisms. Gastrodin supplementation alleviated clinical symptoms such as weight loss, a shortened colon, and a high disease activity index. Meanwhile, gastrodin strengthened the intestinal barrier by increasing the 0expression of tight junction proteins and mucin. Furthermore, Gastrodin significantly reduced pro-inflammatory cytokine secretion in mice by downregulating the NF-κB and MAPK pathways. Gut microbiota analysis showed that gastrodin improved the DSS-disrupted microbiota of mice. These findings demonstrate that gastrodin could attenuate DSS-induced colitis by enhancing the intestinal barrier and modulating the gut microbiota, providing support for the development of a gastrodin-based strategy to prevent or combat IBD.
Collapse
Affiliation(s)
- Jiahui Li
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, 1 Qinggongyuan Road, Ganjingzi District, Dalian 116034, China; (J.L.); (J.J.); (Y.T.); (B.Z.)
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China;
| | - Jinhui Jia
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, 1 Qinggongyuan Road, Ganjingzi District, Dalian 116034, China; (J.L.); (J.J.); (Y.T.); (B.Z.)
| | - Yue Teng
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, 1 Qinggongyuan Road, Ganjingzi District, Dalian 116034, China; (J.L.); (J.J.); (Y.T.); (B.Z.)
| | - Chunyuan Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China;
| | - Chunwei Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China;
| | - Beiwei Zhu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, 1 Qinggongyuan Road, Ganjingzi District, Dalian 116034, China; (J.L.); (J.J.); (Y.T.); (B.Z.)
| | - Xiaodong Xia
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, 1 Qinggongyuan Road, Ganjingzi District, Dalian 116034, China; (J.L.); (J.J.); (Y.T.); (B.Z.)
| |
Collapse
|
18
|
Wang J, Yuan ZY, Wang XY, Zhu JX, Huang WF, Xu GH, Yi LT. Anthocyanins-rich cranberry extract attenuates DSS-induced IBD in an intestinal flora independent manner. Curr Res Food Sci 2024; 9:100815. [PMID: 39161885 PMCID: PMC11332073 DOI: 10.1016/j.crfs.2024.100815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/25/2024] [Accepted: 07/20/2024] [Indexed: 08/21/2024] Open
Abstract
Cranberry is abundantly rich in anthocyanins, a type of flavonoid with potent antioxidant properties and the resistance against certain diseases. In this study, anthocyanin-rich cranberry extract was extracted, purified, and its components were analyzed. 92.18 % of anthocyanins was obtained and the total content of anthocyanins was 302.62 mg/g after AB-8 resin purification. Quantification analysis showed that the extract mainly contained cyanidin-3-galactoside, procyanidin B2 and procyanidin B4. Then we explored its effects on dextran sulfate sodium (DSS)-induced inflammatory bowel disease (IBD) in mice. The supplementation of cranberry extract resulted in an alleviation of IBD symptoms, evidenced by improvements in the disease activity index (DAI), restoration of colon length and colonic morphology. Cranberry extract reversed the elevated iron and malondialdehyde (MDA) levels and restored glutathione (GSH) levels in IBD mice. Further analysis revealed that cranberry modulated ferroptosis-associated genes and reduced expression of pro-inflammatory cytokines. Although cranberry influenced the intestinal flora balance by reducing Proteobacteria and Escherichia-Shigella, and increasing Lactobacillus, as well as enhancing SCFAs content, these effects were not entirely dependent on intestinal flora modulation, as indicated by antibiotic intervention and fecal microbiota transplantation (FMT) experiments. In conclusion, our findings suggest that the beneficial impact of cranberry extract on IBD may primarily involve the regulation of colonic ferroptosis, independent of significant alterations in intestinal flora.
Collapse
Affiliation(s)
- Jun Wang
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian province, PR China
| | - Zhong-Yu Yuan
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian province, PR China
| | - Xin-Yu Wang
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian province, PR China
| | - Ji-Xiao Zhu
- Research Center of Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Wei-Feng Huang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361003, Fujian province, PR China
| | - Guang-Hui Xu
- Xiamen Medicine Research Institute, Xiamen, 361008, Fujian province, PR China
| | - Li-Tao Yi
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian province, PR China
- Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen, 361021, Fujian province, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, Fujian province, PR China
| |
Collapse
|
19
|
Zhang D, Wan H, Zhao R, Zhang Y, Chen H. Eudragit S100 coated iron oxide-chitosan nanocomposites for colon targeting of 5-aminosalicylic acid ameliorate ulcerative colitis by improving intestinal barrier function and inhibiting NLRP3 inflammasome. Int Immunopharmacol 2024; 139:112661. [PMID: 39008936 DOI: 10.1016/j.intimp.2024.112661] [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: 04/15/2024] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
The therapeutic effect of 5-amino salicylic acid (5-ASA), a first-line therapeutic agent for the treatment of ulcerative colitis (UC), is limited by the modest bioavailability afforded by its oral administration. In this study, a 5-ASA oral delivery system was developed using Eudragit S100-coated iron oxide-chitosan nanocomposites (ES-IOCS/5-ASA) to address this issue. According to drug release studies in vitro, ES-IOCS/5-ASA only released a small amount of drug in simulated gastric fluid with a pH of 1.2. However, in a medium with a pH of 7.5, a relatively rapid and complete release was noted. 5-ASA-loaded iron oxide-chitosan nanocomposites (IOCS/5-ASA) could be effectively taken up by NCM460 cells and performed better anti-inflammatory effects than free 5-ASA. At the same time, IOCS/5-ASA improved barrier damage in DSS-induced NCM460 cells. In vivo models of dextran sulphate sodium (DSS)-induced colitis were used to assess the therapeutic efficacy of oral administration of ES-IOCS/5-ASA. ES-IOCS/5-ASA significantly relieved DSS-induced colitis and enhanced the integrity of the intestinal epithelial barrier. ES-IOCS/5-ASA also reduced the expression of NLRP3, ASC and IL-1β. Additionally, iron oxide nanoparticles used as nanozymes could alleviate inflammation. In summary, this study indicates that ES-IOCS/5-ASA exert anti-inflammatory effects on DSS-induced colitis by improving intestinal barrier function and inhibiting NLRP3 inflammasome expression, presenting a viable therapeutic choice for the treatment of UC.
Collapse
Affiliation(s)
- Dandan Zhang
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Hao Wan
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, Jiangsu, People's Republic of China
| | - Ran Zhao
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yu Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, Jiangsu, People's Republic of China.
| | - Hong Chen
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China.
| |
Collapse
|
20
|
Lu S, Xu Y, Zhang H, Liu Z, Xu J, Zheng B, Shi D, Qiu F. Glycyrol Relieves Ulcerative Colitis by Promoting the Fusion of ZO-1 with the Cell Membrane through the Enteric Glial Cells GDNF/RET Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:14653-14662. [PMID: 38860840 DOI: 10.1021/acs.jafc.4c00239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
The damage to the mechanical barrier of the intestinal mucosa is the initiating factor and the core link of the progression of ulcerative colitis (UC). Protecting the mechanical barrier of the intestinal mucosa is of great significance for improving the health status of UC patients. ZO-1 is a key scaffold protein of the mechanical barrier of the intestinal mucosa, and its fusion with the membrane of the intestinal epithelium is a necessary condition to maintain the integrity of the mechanical barrier of the intestinal mucosa. Enteric glial cells (EGCs) play an important role in the maintenance of intestinal homeostasis and have become a new target for regulating intestinal health in recent years. In this study, we found that glycyrol (GC), a representative coumarin compound isolated from Licorice (Glycyrrhiza uralensis Fisch, used for medicine and food), can alleviate UC by promoting the production of neurotrophic factor GDNF in mice EGCs. Specifically, we demonstrated that GC promotes the production of GDNF, then activates its receptor RET, promotes ZO-1 fusion with cell membranes, and protects the intestinal mucosal mechanical barrier. The results of this study can provide new ideas for the prevention and treatment of UC.
Collapse
Affiliation(s)
- Shangyun Lu
- Nutritional and Food Science Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan 030001, China
- Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, Taiyuan 030001, China
| | - Yang Xu
- Nutritional and Food Science Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Huixia Zhang
- Nutritional and Food Science Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Ziling Liu
- Nutritional and Food Science Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Jiali Xu
- Nutritional and Food Science Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Bowen Zheng
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Dongxing Shi
- Nutritional and Food Science Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan 030001, China
- Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, Taiyuan 030001, China
| | - Fubin Qiu
- Nutritional and Food Science Research Institute, Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Taiyuan 030001, China
- Center for Ecological Public Health Security of Yellow River Basin, Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
21
|
Zhu J, Wang H, Aisikaer M, Yisimayili Z, Yang T, Zhou W, Zhao J, Yunusi K, Aximujiang K. L.acidophilus HSCC LA042 and HKL suspension ameliorate DSS-induced ulcerative colitis in mice by improving the intestinal barrier inhibiting the NLRP3 inflammasome and pathogenic bacteria. Heliyon 2024; 10:e33053. [PMID: 39027449 PMCID: PMC11254534 DOI: 10.1016/j.heliyon.2024.e33053] [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: 01/20/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024] Open
Abstract
Ulcerative Colitis(UC) is a chronic intestinal inflammation affecting the intestines, yet its underlying causes remain unclear. In recent decades, the global prevalence of UC has been on the rise, leading to an increasing demand for therapeutic drugs with minimal side effects. Huan Kui Le (HKL), a traditional Chinese medicine compound, has demonstrated promising efficacy when combined with Lactobacillus acidophilus (Lac.) for UC intervention. However, the precise therapeutic mechanism of this combination remains unknown. The study focused on understanding the mechanisms of UC by examining the effects of Lac. and HKL (LH) treatment. The outcomes discovered that the disruption of gut microbiota, triggered by the activation of the NLRP3 inflammasome, plays a crucial role in UC development. This disruption exacerbates UC symptoms by causing disturbances in inflammatory cytokines and mucosal permeability. We investigated the dynamic changes following the application of this treatment using 16S rRNA sequencing, HE, WB, IHC, and ELISA. Compared with the UC group, LH treatment reduced colon pathological injury, improved colon length, and decreased IL-1 β serum levels. Furthermore, it restored the expression of TJs and preserved mucosal barrier integrity. LH treatment also mitigated colon injury by attenuating the expression of pyroptosis-related genes and proteins, such as NLRP3 and Caspase-1. Additionally, LH treatment altered the gut microbiota's microecology, characterized by a reduction in pathogenic bacteria abundance like Escherichia-shigella and an increase in beneficial bacteria abundance like Akkermansia and Erysipelatoclostridium. Overall, our findings indicate that LH therapy may be associated with intestinal barrier repair, inflammasome inhibition, and gut microbiota regulation, suggesting its potential as a UC treatment.
Collapse
Affiliation(s)
- Jiwei Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Hanming Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Muaitaer Aisikaer
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | | | - Tongtong Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Wenjun Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Jianfeng Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Kurexi Yunusi
- Uygur Medical College, Xinjiang Medical University, Urumqi, 830017, China
| | - Kasimujiang Aximujiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Molecular Biology for Endemic Disease, Urumqi, Xinjiang, 830017, China
| |
Collapse
|
22
|
Zhang P, Xue Y, Cao Z, Guo Y, Pang X, Chen C, Zhang W. Raffinose Ameliorates DSS-Induced Colitis in Mice by Modulating Gut Microbiota and Targeting the Inflammatory TLR4-MyD88-NF-κB Signaling Pathway. Foods 2024; 13:1849. [PMID: 38928791 PMCID: PMC11203344 DOI: 10.3390/foods13121849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/02/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
This study aimed to explore the protective effects of raffinose (Raf) against inflammatory bowel disease in mice with colitis. Mice were administered 100, 200, or 400 mg/kg Raf for 21 d, followed by drinking-water containing 3% dextran sulfate sodium salt (DSS) for 3 d. Thereafter, the phenotype, pathological lesions in the colon, cytokines levels, and gut microbiota were evaluated. Treatment with Raf reduced the severity of the pathological changes in the colon, mitigating the reduction in colon length. Following Raf intervention, serum levels of inflammatory cytokines (IL-2, IL-6, IL-1β, and TNF-α) tended to return to normal. These results suggest that the anti-inflammatory effects of Raf are associated with a reduction in TLR4-MyD88-NF-κB pathway expression in mouse colonic tissues. Analysis of gut microbiota abundance and its correlation with colitis parameters revealed that DSS-induced dysbiosis was partially mitigated by Raf. In conclusion, Raf exerts a protective effect in colitis by modulating the gut microbiota and TLR4-MyD88-NF-κB pathway.
Collapse
|
23
|
Xiang T, Wang J, Li H. Current applications of intestinal organoids: a review. Stem Cell Res Ther 2024; 15:155. [PMID: 38816841 PMCID: PMC11140936 DOI: 10.1186/s13287-024-03768-3] [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: 02/13/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
In the past decade, intestinal organoid technology has paved the way for reproducing tissue or organ morphogenesis during intestinal physiological processes in vitro and studying the pathogenesis of various intestinal diseases. Intestinal organoids are favored in drug screening due to their ability for high-throughput in vitro cultivation and their closer resemblance to patient genetic characteristics. Furthermore, as disease models, intestinal organoids find wide applications in screening diagnostic markers, identifying therapeutic targets, and exploring epigenetic mechanisms of diseases. Additionally, as a transplantable cellular system, organoids have played a significant role in the reconstruction of damaged epithelium in conditions such as ulcerative colitis and short bowel syndrome, as well as in intestinal material exchange and metabolic function restoration. The rise of interdisciplinary approaches, including organoid-on-chip technology, genome editing techniques, and microfluidics, has greatly accelerated the development of organoids. In this review, VOSviewer software is used to visualize hot co-cited journal and keywords trends of intestinal organoid firstly. Subsequently, we have summarized the current applications of intestinal organoid technology in disease modeling, drug screening, and regenerative medicine. This will deepen our understanding of intestinal organoids and further explore the physiological mechanisms of the intestine and drug development for intestinal diseases.
Collapse
Affiliation(s)
- Tao Xiang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Hui Li
- Surgical Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
24
|
Kumkum R, Aston-Mourney K, McNeill BA, Hernández D, Rivera LR. Bioavailability of Anthocyanins: Whole Foods versus Extracts. Nutrients 2024; 16:1403. [PMID: 38794640 PMCID: PMC11123854 DOI: 10.3390/nu16101403] [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: 04/18/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
Anthocyanins have gained significant popularity in recent years for their diverse health benefits, yet their limited bioavailability poses a challenge. To address this concern, technologies have emerged to enhance anthocyanin concentration, often isolating these compounds from other food constituents. However, the extent to which isolated anthocyanins confer health benefits compared to their whole-food counterparts remains unclear. This review explores the current literature on anthocyanin bioavailability and metabolism in the body, with a focus on comparing bioavailability when consumed as extracts versus whole foods rich in anthocyanins, drawing from in vitro, in vivo, and human clinical studies. While direct comparisons between anthocyanin bioavailability in whole foods versus isolates are scarce, prevailing evidence favours whole-food consumption over anthocyanin extracts. Further clinical investigations, preferably with direct comparisons, are needed to validate these findings and elucidate the nuanced interplay between anthocyanins and food matrices, informing future research directions and practical recommendations.
Collapse
Affiliation(s)
| | | | | | | | - Leni R. Rivera
- Institute for Innovation in Physical and Mental Health and Clinical Translation (IMPACT), Deakin University, Geelong 3220, Australia; (R.K.); (K.A.-M.); (B.A.M.); (D.H.)
| |
Collapse
|
25
|
Chu L, Zhang S, Wu W, Gong Y, Chen Z, Wen Y, Wang Y, Wang L. Grape seed proanthocyanidin extract alleviates inflammation in experimental colitis mice by inhibiting NF-κB signaling pathway. ENVIRONMENTAL TOXICOLOGY 2024; 39:2572-2582. [PMID: 38205677 DOI: 10.1002/tox.24129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/07/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024]
Abstract
Ulcerative colitis (UC) is a complex inflammatory disease of colorectum that induces abnormal immune responses and severely affects the quality of life of the patients. Grape seed proanthocyanidin extract (GSPE) exerts anti-inflammatory and antioxidant functions in many inflammatory diseases. The objective of this study was to investigate the potential therapeutic effects and underlying mechanisms of GSPE in UC using a dextran sodium sulfate (DSS)-induced mouse UC model and a lipopolysaccharide (LPS)-stimulated RAW264.7 macrophage model. In this study, we found that the GSPE markedly prevented DSS-induced weight loss and colon length shortening in UC mice. Further investigations showed that GSPE significantly attenuated the expression of pro-inflammatory cytokines TNF-α, IL-6, and IL-1β, and elevated the expression of anti-inflammatory cytokine IL-10 in the colon tissues and serum of DSS-induced colitis mice by suppressing NF-κB signaling pathway. Furthermore, LPS-induced inflammation in RAW264.7 cells was also reversed by GSPE. Taken together, our results confirm that GSPE can ameliorate inflammatory response in experimental colitis via inhibiting NF-κB signaling pathway. This study advances the research progress on a potentially effective therapeutic strategy for inflammatory bowel diseases.
Collapse
Affiliation(s)
- Lei Chu
- Clinical Laboratory, The People's Hospital of Danyang & Affiliated Danyang Hospital of Nantong University, Danyang, China
| | - Shaoru Zhang
- Clinical Laboratory, The People's Hospital of Danyang & Affiliated Danyang Hospital of Nantong University, Danyang, China
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Weidong Wu
- Clinical Laboratory, The People's Hospital of Danyang & Affiliated Danyang Hospital of Nantong University, Danyang, China
| | - Yuqing Gong
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Zhenshi Chen
- Clinical Laboratory, The People's Hospital of Danyang & Affiliated Danyang Hospital of Nantong University, Danyang, China
| | - Yanting Wen
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Yong Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Lihui Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
26
|
Li S, Zhuge A, Chen H, Han S, Shen J, Wang K, Xia J, Xia H, Jiang S, Wu Y, Li L. Sedanolide alleviates DSS-induced colitis by modulating the intestinal FXR-SMPD3 pathway in mice. J Adv Res 2024:S2090-1232(24)00128-0. [PMID: 38582300 DOI: 10.1016/j.jare.2024.03.026] [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: 11/29/2023] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/08/2024] Open
Abstract
INTRODUCTION Inflammatory bowel disease (IBD) is a global disease with limited therapy. It is reported that sedanolide exerts anti-oxidative and anti-inflammatory effects as a natural phthalide, but its effects on IBD remain unclear. OBJECTIVES In this study, we investigated the impacts of sedanolide on dextran sodium sulfate (DSS)-induced colitis in mice. METHODS The mice were administered sedanolide or vehicle followed by DSS administration, after which colitis symptoms, inflammation levels, and intestinal barrier function were evaluated. Transcriptome analysis, 16S rRNA sequencing, and targeted metabolomics analysis of bile acids and lipids were performed. RESULTS Sedanolide protected mice from DSS-induced colitis, suppressed the inflammation, restored the weakened epithelial barrier, and modified the gut microbiota by decreasing bile salt hydrolase (BSH)-expressing bacteria. The downregulation of BSH activity by sedanolide increased the ratio of conjugated/unconjugated bile acids (BAs), thereby inhibiting the intestinal farnesoid X receptor (FXR) pathway. The roles of the FXR pathway and gut microbiota were verified using an intestinal FXR-specific agonist (fexaramine) and germ-free mice, respectively. Furthermore, we identified the key effector ceramide, which is regulated by sphingomyelin phosphodiesterase 3 (SMPD3). The protective effects of ceramide (d18:1/16:0) against inflammation and the gut barrier were demonstrated in vitro using the human cell line Caco-2. CONCLUSION Sedanolide could reshape the intestinal flora and influence BA composition, thus inhibiting the FXR-SMPD3 pathway to stimulate the synthesis of ceramide, which ultimately alleviated DSS-induced colitis in mice. Overall, our research revealed the protective effects of sedanolide against DSS-induced colitis in mice, which indicated that sedanolide may be a clinical treatment for colitis. Additionally, the key lipid ceramide (d18:1/16:0) was shown to mediate the protective effects of sedanolide, providing new insight into the associations between colitis and lipid metabolites.
Collapse
Affiliation(s)
- Shengjie Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Aoxiang Zhuge
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hui Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jian Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Kaicen Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiafeng Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - He Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shiman Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Youhe Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250000, China.
| |
Collapse
|
27
|
Xie L, Chen T, Li H, Xiao J, Wang L, Kim SK, Huang Z, Xie J. An Exopolysaccharide from Genistein-Stimulated Monascus Purpureus: Structural Characterization and Protective Effects against DSS-Induced Intestinal Barrier Injury Associated with the Gut Microbiota-Modulated Short-Chain Fatty Acid-TLR4/MAPK/NF-κB Cascade Response. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7476-7496. [PMID: 38511260 DOI: 10.1021/acs.jafc.3c09290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Inflammatory bowel disease is a major health problem that can lead to prolonged damage to the digestive system. This study investigated the effects of an exopolysaccharide from genistein-stimulated Monascus purpureus (G-EMP) in a mouse model of colitis to clarify its molecular mechanisms and identified its structures. G-EMP (Mw = 56.4 kDa) was primarily consisted of → 4)-α-D-Galp-(1 →, → 2,6)-α-D-Glcp-(1→ and →2)-β-D-Manp-(1 → , with one of the branches being α-D-Manp-(1 →. G-EMP intervention reduced the loss of body weight, degree of colonic damage and shortening, disease activity index scores, and histopathology scores, while restoring goblet cell production and oxidative homeostasis, repairing colonic functions, and regulating inflammatory cytokines. RNA sequencing and Western blot analysis indicated that G-EMP exerts anti-inflammatory properties by suppressing the TLR4/MAPK/NF-κB inflammatory signaling pathway. G-EMP modulated the gut microbiota by improving its diversities, elevating the relative abundances of beneficial bacteria, declining the Firmicutes/Bacteroidota value, and regulating the level of short-chain fatty acids (SCFAs). Correlation analysis demonstrated strong links between SCFAs, gut microbiota, and the inflammatory response, indicating the potential of G-EMP to prevent colitis.
Collapse
Affiliation(s)
- Liuming Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
- Sino-German Joint Research Institute, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Republic of Korea
| | - Ting Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Hong Li
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
- Sino-German Joint Research Institute, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Jindan Xiao
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Linchun Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
- Sino-German Joint Research Institute, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Soo-Ki Kim
- Department of Animal Science and Technology, Konkuk University, Seoul 05029, Republic of Korea
| | - Zhibing Huang
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
- Sino-German Joint Research Institute, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| | - Jianhua Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, No. 235 Nanjing East Road, Nanchang 330047, China
| |
Collapse
|
28
|
Liu Y, Fernandes I, Mateus N, Oliveira H, Han F. The Role of Anthocyanins in Alleviating Intestinal Diseases: A Mini Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:5491-5502. [PMID: 38446808 DOI: 10.1021/acs.jafc.3c07741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Anthocyanins are phytonutrients with physiological activity belonging to the flavonoid family whose transport and absorption in the human body follow specific pathways. In the upper gastrointestinal tract, anthocyanins are rarely absorbed intact by active transporters, with most reaching the colon, where bacteria convert them into metabolites. There is mounting evidence that anthocyanins can be used for prevention and treatment of intestinal diseases, including inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), and colorectal cancer (CRC), through the protective function on the intestinal epithelial barrier, immunomodulation, antioxidants, and gut microbiota metabolism. Dietary anthocyanins are summarized in this comprehensive review with respect to their classification and structure as well as their absorption and transport mechanisms within the gastrointestinal tract. Additionally, the review delves into the role and mechanism of anthocyanins in treating common intestinal diseases. These insights will deepen our understanding of the potential benefits of natural anthocyanins for intestinal disorders.
Collapse
Affiliation(s)
- Yang Liu
- College of Enology, Northwest A&F University, Yangling 712100, China
| | - Iva Fernandes
- LAQV, REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, Porto 4169-007 Porto, Portugal
| | - Nuno Mateus
- LAQV, REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, Porto 4169-007 Porto, Portugal
| | - Hélder Oliveira
- LAQV, REQUIMTE, Department of Chemistry and Biochemistry, Faculty of Sciences, Porto 4169-007 Porto, Portugal
| | - Fuliang Han
- College of Enology, Northwest A&F University, Yangling 712100, China
- Shaanxi Engineering Research Center for Viti-Viniculture, Northwest A&F University, Yangling 712100, China
- Heyang Experimental Demonstration Station, Northwest A&F University, Weinan 715300, China
- Ningxia Helan Mountain's East Foothill Wine Experiment and Demonstration Station, Northwest A&F University, Yongning 750104, China
| |
Collapse
|
29
|
Li F, Sun Q, Chen L, Zhang R, Zhang Z. Unlocking the health potential of anthocyanins: a structural insight into their varied biological effects. Crit Rev Food Sci Nutr 2024:1-21. [PMID: 38494796 DOI: 10.1080/10408398.2024.2328176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Anthocyanins have become increasingly important to the food industry due to their colorant features and many health-promoting activities. Numerous studies have linked anthocyanins to antioxidant, anti-inflammatory, anticarcinogenic properties, as well as protection against heart disease, certain types of cancer, and a reduced risk of diabetes and cognitive disorders. Anthocyanins from various foods may exhibit distinct biological and health-promoting activities owing to their structural diversity. In this review, we have collected and tabulated the key information from various recent published studies focusing on investigating the chemical structure effect of anthocyanins on their stability, antioxidant activities, in vivo fate, and changes in the gut microbiome. This information should be valuable in comprehending the connection between the molecular structure and biological function of anthocyanins, with the potential to enhance their application as both colorants and functional compounds in the food industry.
Collapse
Affiliation(s)
- Fangfang Li
- Food Science Program, Division of Food, Nutrition & Exercise Sciences, University of Missouri, Columbia, Missouri, USA
| | - Quancai Sun
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Florida, USA
| | - Long Chen
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ruojie Zhang
- Food Science Program, Division of Food, Nutrition & Exercise Sciences, University of Missouri, Columbia, Missouri, USA
| | - Zipei Zhang
- Food Science Program, Division of Food, Nutrition & Exercise Sciences, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
30
|
Liu M, Ma J, Xu J, Huangfu W, Zhang Y, Ali Q, Liu B, Li D, Cui Y, Wang Z, Sun H, Zhu X, Ma S, Shi Y. Fecal microbiota transplantation alleviates intestinal inflammatory diarrhea caused by oxidative stress and pyroptosis via reducing gut microbiota-derived lipopolysaccharides. Int J Biol Macromol 2024; 261:129696. [PMID: 38280701 DOI: 10.1016/j.ijbiomac.2024.129696] [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: 07/07/2023] [Revised: 01/07/2024] [Accepted: 01/21/2024] [Indexed: 01/29/2024]
Abstract
Infancy is a critical period in the maturation of the gut microbiota and a phase of susceptibility to gut microbiota dysbiosis. Early disturbances in the gut microbiota can have long-lasting effects on host physiology, including intestinal injury and diarrhea. Fecal microbiota transplantation (FMT) can remodel gut microbiota and may be an effective way to treat infant diarrhea. However, limited research has been conducted on the mechanisms of infant diarrhea and the regulation of gut microbiota balance through FMT, primarily due to ethical challenges in testing on human infants. Our study demonstrated that elevated Lipopolysaccharides (LPS) levels in piglets with diarrhea were associated with colon microbiota dysbiosis induced by early weaning. Additionally, LPS upregulated NLRP3 levels by activating TLR4 and inducing ROS production, resulting in pyroptosis, disruption of the intestinal barrier, bacterial translocation, and subsequent inflammation, ultimately leading to diarrhea in piglets. Through microbiota regulation, FMT modulated β-PBD-2 secretion in the colon by increasing butyric acid levels. This modulation alleviated gut microbiota dysbiosis, reduced LPS levels, attenuated oxidative stress and pyroptosis, inhibited the inflammatory response, maintained the integrity of the intestinal barrier, and ultimately reduced diarrhea in piglets caused by colitis. These findings present a novel perspective on the pathogenesis, pathophysiology, prevention, and treatment of diarrhea diseases, underscoring the significance of the interaction between FMT and the gut microbiota as a critical strategy for treating diarrhea and intestinal diseases in infants and farm animals.
Collapse
Affiliation(s)
- Mengqi Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Jixiang Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Junying Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Weikang Huangfu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Yan Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Qasim Ali
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Boshuai Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China; Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China; Henan Forage Engineering Technology Research Center, Zhengzhou, Henan 450002, China
| | - Defeng Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China; Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China; Henan Forage Engineering Technology Research Center, Zhengzhou, Henan 450002, China
| | - Yalei Cui
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China; Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China; Henan Forage Engineering Technology Research Center, Zhengzhou, Henan 450002, China
| | - Zhichang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China; Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China; Henan Forage Engineering Technology Research Center, Zhengzhou, Henan 450002, China
| | - Hao Sun
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China; Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China; Henan Forage Engineering Technology Research Center, Zhengzhou, Henan 450002, China
| | - Xiaoyan Zhu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China; Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China; Henan Forage Engineering Technology Research Center, Zhengzhou, Henan 450002, China
| | - Sen Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China; Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China; Henan Forage Engineering Technology Research Center, Zhengzhou, Henan 450002, China
| | - Yinghua Shi
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China; Henan Key Laboratory of Innovation and Utilization of Grassland Resources, Zhengzhou, China; Henan Forage Engineering Technology Research Center, Zhengzhou, Henan 450002, China.
| |
Collapse
|
31
|
Nishiyama K, Kato Y, Nishimura A, Mi X, Nagata R, Mori Y, Azuma YT, Nishida M. Pharmacological Activation of TRPC6 Channel Prevents Colitis Progression. Int J Mol Sci 2024; 25:2401. [PMID: 38397074 PMCID: PMC10889536 DOI: 10.3390/ijms25042401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
We recently reported that transient receptor potential canonical (TRPC) 6 channel activity contributes to intracellular Zn2+ homeostasis in the heart. Zn2+ has also been implicated in the regulation of intestinal redox and microbial homeostasis. This study aims to investigate the role of TRPC6-mediated Zn2+ influx in the stress resistance of the intestine. The expression profile of TRPC1-C7 mRNAs in the actively inflamed mucosa from inflammatory bowel disease (IBD) patients was analyzed using the GEO database. Systemic TRPC3 knockout (KO) and TRPC6 KO mice were treated with dextran sulfate sodium (DSS) to induce colitis. The Zn2+ concentration and the mRNA expression levels of oxidative/inflammatory markers in colon tissues were quantitatively analyzed, and gut microbiota profiles were compared. TRPC6 mRNA expression level was increased in IBD patients and DSS-treated mouse colon tissues. DSS-treated TRPC6 KO mice, but not TRPC3 KO mice, showed severe weight loss and increased disease activity index compared with DSS-treated WT mice. The mRNA abundances of antioxidant proteins were basically increased in the TRPC6 KO colon, with changes in gut microbiota profiles. Treatment with TRPC6 activator prevented the DSS-induced colitis progression accompanied by increasing Zn2+ concentration. We suggest that TRPC6-mediated Zn2+ influx activity plays a key role in stress resistance against IBD, providing a new strategy for treating colitis.
Collapse
Affiliation(s)
- Kazuhiro Nishiyama
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.N.); (Y.K.); (X.M.)
- Laboratory of Prophylactic Pharmacology, Osaka Metropolitan University Graduate School of Veterinary Science, Osaka 598-8531, Japan;
| | - Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.N.); (Y.K.); (X.M.)
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan;
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- SOKENDAI (Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
| | - Xinya Mi
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.N.); (Y.K.); (X.M.)
| | - Ryu Nagata
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan;
| | - Yasuo Mori
- Graduate School of Engineering, Kyoto University, Kyoto 615-8530, Japan;
| | - Yasu-Taka Azuma
- Laboratory of Prophylactic Pharmacology, Osaka Metropolitan University Graduate School of Veterinary Science, Osaka 598-8531, Japan;
| | - Motohiro Nishida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.N.); (Y.K.); (X.M.)
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan;
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- SOKENDAI (Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
32
|
Zheng L, Han Z, Zhang J, Kang J, Li C, Pang Q, Liu S. Lactiplantibacillus plantarum and Saccharomyces cerevisiae-Fermented Coconut Water Alleviates Dextran Sodium Sulfate-Induced Enteritis in Wenchang Chicken: A Gut Microbiota and Metabolomic Approach. Animals (Basel) 2024; 14:575. [PMID: 38396543 PMCID: PMC10886277 DOI: 10.3390/ani14040575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/19/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
In order to investigate the potential mechanisms of probiotic-fermented coconut water in treating enteritis, this study conducted a comprehensive analysis of the effects of probiotic intervention on the recovery from Dextran Sodium Sulfate-induced acute enteritis in Wenchang chicks. The analysis encompassed the assessment of growth performance, serum indicators, intestinal tissue structure, and metagenomic and metabolomic profiles of cecal contents in 60 Wenchang chicks subjected to intervention. This approach aimed to elucidate the impact of probiotic intervention on the recovery process from acute enteritis at both the genetic and metabolic levels in the avian model. The results revealed that intervention with Saccharomyces cerevisiae Y301 improved the growth rate of chicks. and intervention with Lactiplantibacillus plantarum MS2c regulated the glycerophospholipid metabolism pathway and reshaped the gut microbiota structure in modeling chicks with acute enteritis, reducing the abundance of potentially pathogenic bacteria from the Alistipes and increasing the abundance of potentially beneficial species from the Christensenellaceae. This intervention resulted in the production of specific gut metabolites, including Gentamicin C and polymyxin B2, recognized for their therapeutic effects on acute enteritis. The combined intervention of S. cerevisiae Y301 and L. plantarum MS2c not only enhanced growth performance but also mitigated intestinal wall damage and increased the abundance of gut metabolites such as gentamicin C and polymyxin B2, thereby mitigating symptoms of enteritis. Furthermore, this combined intervention reduced the levels of serum immune markers, including IL-10, IL-6, TNF-α, IFN-γ, and D-lactic acid, thus mitigating intestinal epithelial cell damage and promoting acute enteritis recovery. This study provides crucial insights into the mechanisms of action of probiotics and probiotic-fermented coconut water in acute enteritis recovery, offering new perspectives for sustainable farming practices for Wenchang chicken.
Collapse
Affiliation(s)
- Leijie Zheng
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (L.Z.)
| | - Zhe Han
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (L.Z.)
| | - Jiachao Zhang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (L.Z.)
| | - Jiamu Kang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (L.Z.)
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China
| | - Congfa Li
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (L.Z.)
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China
- Key Laboratory of Tropical Agricultural Products Processing Technology of Haikou City, Haikou 570228, China
| | - Qing Pang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (L.Z.)
| | - Sixin Liu
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (L.Z.)
- Key Laboratory of Tropical Agricultural Products Processing Technology of Haikou City, Haikou 570228, China
| |
Collapse
|
33
|
Mai H, Yang X, Xie Y, Zhou J, Wang Q, Wei Y, Yang Y, Lu D, Ye L, Cui P, Liang H, Huang J. The role of gut microbiota in the occurrence and progression of non-alcoholic fatty liver disease. Front Microbiol 2024; 14:1257903. [PMID: 38249477 PMCID: PMC10797006 DOI: 10.3389/fmicb.2023.1257903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is the most prevalent cause of chronic liver disease worldwide, and gut microbes are associated with the development and progression of NAFLD. Despite numerous studies exploring the changes in gut microbes associated with NAFLD, there was no consistent pattern of changes. Method We retrieved studies on the human fecal microbiota sequenced by 16S rRNA gene amplification associated with NAFLD from the NCBI database up to April 2023, and re-analyzed them using bioinformatic methods. Results We finally screened 12 relevant studies related to NAFLD, which included a total of 1,189 study subjects (NAFLD, n = 654; healthy control, n = 398; obesity, n = 137). Our results revealed a significant decrease in gut microbial diversity with the occurrence and progression of NAFLD (SMD = -0.32; 95% CI -0.42 to -0.21; p < 0.001). Alpha diversity and the increased abundance of several crucial genera, including Desulfovibrio, Negativibacillus, and Prevotella, can serve as an indication of their predictive risk ability for the occurrence and progression of NAFLD (all AUC > 0.7). The occurrence and progression of NAFLD are significantly associated with higher levels of LPS biosynthesis, tryptophan metabolism, glutathione metabolism, and lipid metabolism. Conclusion This study elucidated gut microbes relevance to disease development and identified potential risk-associated microbes and functional pathways associated with NAFLD occurrence and progression.
Collapse
Affiliation(s)
- Huanzhuo Mai
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Xing Yang
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Yulan Xie
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Jie Zhou
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Qing Wang
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Yiru Wei
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Yuecong Yang
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Dongjia Lu
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
| | - Li Ye
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Nanning, China
| | - Ping Cui
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Nanning, China
- Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Hao Liang
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
- Joint Laboratory for Emerging Infectious Diseases in China (Guangxi)-ASEAN, Nanning, China
- Life Sciences Institute, Guangxi Medical University, Nanning, China
| | - Jiegang Huang
- School of Public Health, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| |
Collapse
|
34
|
Li Q, Lin L, Zhang C, Zhang H, Ma Y, Qian H, Chen XL, Wang X. The progression of inorganic nanoparticles and natural products for inflammatory bowel disease. J Nanobiotechnology 2024; 22:17. [PMID: 38172992 PMCID: PMC10763270 DOI: 10.1186/s12951-023-02246-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024] Open
Abstract
There is a growing body of evidence indicating a close association between inflammatory bowel disease (IBD) and disrupted intestinal homeostasis. Excessive production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), along with an increase in M1 proinflammatory macrophage infiltration during the activation of intestinal inflammation, plays a pivotal role in disrupting intestinal homeostasis in IBD. The overabundance of ROS/RNS can cause intestinal tissue damage and the disruption of crucial gut proteins, which ultimately compromises the integrity of the intestinal barrier. The proliferation of M1 macrophages contributes to an exaggerated immune response, further compromising the intestinal immune barrier. Currently, intestinal nanomaterials have gained widespread attention in the context of IBD due to their notable characteristics, including the ability to specifically target regions of interest, clear excess ROS/RNS, and mimic biological enzymes. In this review, we initially elucidated the gut microenvironment in IBD. Subsequently, we delineate therapeutic strategies involving two distinct types of nanomedicine, namely inorganic nanoparticles and natural product nanomaterials. Finally, we present a comprehensive overview of the promising prospects associated with the application of nanomedicine in future clinical settings for the treatment of IBD (graphic abstract). Different classes of nanomedicine are used to treat IBD. This review primarily elucidates the current etiology of inflammatory bowel disease and explores two prominent nanomaterial-based therapeutic approaches. First, it aims to eliminate excessive reactive oxygen species and reactive nitrogen species. Second, they focus on modulating the polarization of inflammatory macrophages and reducing the proportion of pro-inflammatory macrophages. Additionally, this article delves into the treatment of inflammatory bowel disease using inorganic metal nanomaterials and natural product nanomaterials.
Collapse
Affiliation(s)
- Qingrong Li
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Liting Lin
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Cong Zhang
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China
| | - Hengguo Zhang
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Yan Ma
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China
| | - Haisheng Qian
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China.
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| | - Xianwen Wang
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China.
| |
Collapse
|
35
|
Zhao R, Zhang Q, Huang T, Tian Y, Guan G, Lin Y. Effect of the Combination of Clostridium butyricum and Mycelium of Phellinus igniarius on Intestinal Microbiota and Serum Metabolites in DSS-Induced Colitis. Nutrients 2024; 16:153. [PMID: 38201981 PMCID: PMC10780906 DOI: 10.3390/nu16010153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/26/2023] [Accepted: 12/30/2023] [Indexed: 01/12/2024] Open
Abstract
Clostridium butyricum (CB) and Phellinus igniarius (PI) have anti-inflammatory, immune regulation, anti-tumor, and other functions. This study aimed to explore the therapeutic effect of CB and mycelium of PI (MPI) alone and in combination on colitis mice induced by dextran sodium sulfate (DSS). Mice were randomly assigned to five groups: (1) control (CTRL), (2) DSS, (3) CB, (4) MPI, and (5) CB + MPI (CON). The weight of the mice was recorded daily during the experiment, and the length of the colon was measured on the last day of the experiment. The colons were collected for hematoxylin and eosin staining, colon contents were collected for intestinal flora analysis, and serum was collected for metabolite analysis. The results showed that compared with the DSS group, CB, MPI, and CON treatments inhibited the weight loss and colon length shortening caused by DSS, significantly increased the concentrations of interleukin (IL)-4, IL-10, and superoxide dismutase, and significantly decreased the concentrations of IL-6, tumor necrosis factor-α, and myeloperoxidase. Gene sequence analysis of 16S rRNA showed that CB, MPI, and CON treatments changed the composition and structure of intestinal microorganisms. Metabolome results showed that CB, MPI, and CON treatments changed serum metabolites in DSS-treated mice, including dodecenoylcarnitine, L-urobilinogen, and citric acid. In conclusion, CB, MPI, and CON treatments alleviated DSS-induced colitis in mice by regulating intestinal flora and metabolites, with the CON group having the best effect.
Collapse
Affiliation(s)
- Rou Zhao
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China;
| | - Qiaoyi Zhang
- Agricultural Bioengineering Institute, Changsha 410128, China; (Q.Z.); (T.H.); (Y.T.); (G.G.)
| | - Ting Huang
- Agricultural Bioengineering Institute, Changsha 410128, China; (Q.Z.); (T.H.); (Y.T.); (G.G.)
| | - Yun Tian
- Agricultural Bioengineering Institute, Changsha 410128, China; (Q.Z.); (T.H.); (Y.T.); (G.G.)
| | - Guiping Guan
- Agricultural Bioengineering Institute, Changsha 410128, China; (Q.Z.); (T.H.); (Y.T.); (G.G.)
| | - Yuanshan Lin
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China;
| |
Collapse
|
36
|
Lin WS, Cheng WC, Pan MH. Virofree Associates with the Modulation of Gut Microbiomes and Alleviation of DSS-Induced IBD Symptoms in Mice. ACS OMEGA 2023; 8:41427-41437. [PMID: 37969979 PMCID: PMC10633932 DOI: 10.1021/acsomega.3c05159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/29/2023] [Indexed: 11/17/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic, nonspecific inflammation of the intestines that primarily comprises Crohn's disease and ulcerative colitis. The incidence and prevalence of IBD have been increasing globally, highlighting the significance of research and prophylactic interventions. Virofree, a mixture of various botanical extracts (including grapes, cherries, olive leaves, marigolds, green tea, and others), has shown significant potential in disease prevention. This study examined the effects of Virofree on intestinal inflammation and the gut microbiota in mice using a dextran sulfate sodium (DSS)-induced model. The mice showed no adverse reactions when administered Virofree. Virofree administration reduced the disease activity index as indicated by amelioration of DSS-induced symptoms in the mice, including weight loss, diarrhea, and rectal bleeding. Regarding the gut microbiota, Virofree intervention modulated the DSS-induced decrease in gut microbial diversity; the Virofree group showed no increase in the phyla Proteobacteria or Verrucomicrobia while displaying an increase in the genus Duncaniella, bacteria that may have protective properties. These findings suggest that Virofree may have a direct or indirect impact on the composition of the gut microbiota and that it can alleviate the imbalance of the microbiome and intestinal inflammation caused by DSS treatment.
Collapse
Affiliation(s)
- Wei-Sheng Lin
- Institute
of Food Science and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Wan-Chen Cheng
- Institute
of Food Science and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Min-Hsiung Pan
- Institute
of Food Science and Technology, National
Taiwan University, Taipei 10617, Taiwan
- Department
of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- Department
of Health and Nutrition Biotechnology, Asia
University, Taichung 41354, Taiwan
| |
Collapse
|
37
|
Jin Y, Chen L, Yu Y, Hussain M, Zhong H. Bioactive Components in Fruit Interact with Gut Microbes. BIOLOGY 2023; 12:1333. [PMID: 37887043 PMCID: PMC10604038 DOI: 10.3390/biology12101333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/30/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023]
Abstract
Fruits contain many bioactive compounds, including polysaccharides, oligosaccharides, polyphenols, anthocyanins, and flavonoids. All of these bioactives in fruit have potentially beneficial effects on gut microbiota and host health. On the one hand, fruit rich in active ingredients can act as substrates to interact with microorganisms and produce metabolites to regulate the gut microbiota. On the other hand, gut microbes could promote health effects in the host by balancing dysbiosis of gut microbiota. We have extensively analyzed significant information on bioactive components in fruits based on Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA). Although the deep mechanism of action of bioactive components in fruits on gut microbiota needs further study, these results also provide supportive information on fruits as a source of dietary active ingredients to provide support for the adjunctive role of fruits in disease prevention and treatment.
Collapse
Affiliation(s)
- Yuanyuan Jin
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China; (Y.J.); (Y.Y.)
| | - Ling Chen
- Sanya Branch of Hainan Food and Drug Inspection Institute, Sanya 572011, China;
| | - Yufen Yu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China; (Y.J.); (Y.Y.)
| | - Muhammad Hussain
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China; (Y.J.); (Y.Y.)
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China; (Y.J.); (Y.Y.)
| |
Collapse
|
38
|
Do HJ, Kim YS, Oh TW. Effect of Polycan, a β-Glucan from Aureobasidium pullulans SM-2001, on Inflammatory Response and Intestinal Barrier Function in DSS-Induced Ulcerative Colitis. Int J Mol Sci 2023; 24:14773. [PMID: 37834221 PMCID: PMC10572787 DOI: 10.3390/ijms241914773] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Ulcerative colitis (UC), a subtype of inflammatory bowel disease, is a chronic gastrointestinal inflammatory disease with unclear etiology and pathophysiology. Herein, we determined the effects of extracellular polysaccharides purified from Aureobasidium pullulans SM-2001 (Polycan) on tight junction protein expression, inflammation, and apoptosis in a dextran sodium sulfate (DSS)-induced acute colitis model. Fifty mice were divided into normal, DSS, DSS + Polycan 250 mg/kg (Polycan 250), DSS + Polycan 500 mg/kg (Polycan 500), and DSS + 5-aminosalicylic acid 100 mg/kg (5-ASA) groups. Their body weights, colon lengths, histological changes in colon tissue, and tight junction function were observed. Results showed that Polycan 250, Polycan 500, and 5-ASA significantly inhibited body weight loss compared with DSS. Similar to 5-ASA, Polycan 500 exhibited preventive effects on colon length shortening and histological changes in colon tissues. Polycan inhibited the DSS-induced decrease in fluorescein isothiocyanate-dextran permeability and myeloperoxidase activity. Moreover, Polycan significantly recovered serum cytokine (e.g., tumor necrosis factor-α, interleukin (IL)-6, and IL-1β) or mRNA expression in colon tissue compared with DSS. Polycan also inhibited apoptosis by reducing caspase-3 activity and the Bcl-2 associated X/B-cell lymphoma 2 (Bcl-2) ratio. Additionally, DSS treatment significantly reduced microbial abundance and diversity, but the administration of Polycan reversed this effect. Collectively, Polycan protected intestinal barrier function and inhibited inflammation and apoptosis in DSS-induced colitis.
Collapse
Affiliation(s)
- Hyun Ju Do
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea;
| | - Young-Suk Kim
- Glucan Co., Ltd., 25-15, Worasan-ro 950 beon-gil, Munsan-eup, Jinju-si 52840, Gyeongsangnam-do, Republic of Korea;
| | - Tae Woo Oh
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 41062, Republic of Korea
- Department of Korean Convergence Medical Science, University of Science & Technology (UST), 1672 Yuseongdae-ro, Daejeon 34054, Yuseong-gu, Republic of Korea
| |
Collapse
|
39
|
Song X, Huang Q, Yang Y, Ma L, Liu W, Ou C, Chen Q, Zhao T, Xiao Z, Wang M, Jiang Y, Yang Y, Zhang J, Nan Y, Wu W, Ai K. Efficient Therapy of Inflammatory Bowel Disease (IBD) with Highly Specific and Durable Targeted Ta 2 C Modified with Chondroitin Sulfate (TACS). ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301585. [PMID: 37224059 DOI: 10.1002/adma.202301585] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/06/2023] [Indexed: 05/26/2023]
Abstract
Non-invasive localization of lesions and specific targeted therapy are still the main challenges for inflammatory bowel disease (IBD). Ta, as a medical metal element, has been widely used in the treatment of different diseases because of its excellent physicochemical properties but is still far from being explored in IBD. Here, Ta2 C modified with chondroitin sulfate (CS) (TACS) is evaluated as a highly targeted therapy nanomedicine for IBD. Specifically, TACS is modified with dual targeting CS functions due to IBD lesion-specific positive charges and high expression of CD44 receptors. Thanks to the acid stability, sensitive CT imaging function, and strong reactive oxygen species (ROS) elimination ability, oral TACS can accurately locate and delineate IBD lesions through non-invasive CT imaging, and specifically targeted treat IBD effectively because high levels of ROS are a central factor in the progression of IBD. As expected, TACS has much better imaging and therapeutic effects than clinical CT contrast agent and first-line drug 5-aminosalicylic acid, respectively. The mechanism of TACS treatment mainly involves protection of mitochondria, elimination of oxidative stress, inhibiting macrophage M1 polarization, protection of intestinal barrier, and restoration of intestinal flora balance. Collectively, this work provides unprecedented opportunities for oral nanomedicines to targeted therapy of IBD.
Collapse
Affiliation(s)
- Xiangping Song
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiong Huang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Yuqi Yang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Liang Ma
- Hubei Key Laboratory of Optical Information and Pattern Recognition, Wuhan Institute of Technology, Wuhan, 430205, China
| | - Wenguang Liu
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Tianjiao Zhao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Mingyuan Wang
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yitian Jiang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Yunrong Yang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Jinping Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, P. R. China
| | - Yayun Nan
- Geriatric Medical Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China
| | - Wei Wu
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment., Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410008, China
| |
Collapse
|
40
|
Li YY, Cui Y, Dong WR, Liu TT, Zhou G, Chen YX. Terminalia bellirica Fruit Extract Alleviates DSS-Induced Ulcerative Colitis by Regulating Gut Microbiota, Inflammatory Mediators, and Cytokines. Molecules 2023; 28:5783. [PMID: 37570753 PMCID: PMC10421151 DOI: 10.3390/molecules28155783] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/26/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease significantly impacting patients' lives. This study aimed to elucidate the alleviating effect of ethyl acetate extract (TBEA) from Terminalia bellirica fruit on UC and to explore its mechanism. TBEA was the fraction with the best anti-inflammatory activity screened using in vitro anti-inflammatory assays, and HPLC initially characterized its composition. The mice model of ulcerative colitis was established after free drinking of 2.5% dextran sulfate sodium for six days, and the experimental group was treated with 50 mg/kg and 100 mg/kg TBEA for seven days. We found that TBEA significantly alleviated symptoms in UC mice, including a physiologically significant reduction in disease activity index and pathological damage to colonic tissue. TBEA dramatically slowed down oxidative stress and inflammatory process in UC mice, as evidenced by decreasing myeloperoxidase and malondialdehyde activities and increasing glutathione and catalase levels by reducing the concentrations of IL-6, IL-1β, TNF-α, and NO in UC mice, as well as by regulating key proteins in the IL-6/JAK2/STAT3 pathway. Meanwhile, TBEA maintained intestinal homeostasis by regulating intestinal flora structure. Our study provides new ideas for developing TBEA into a new drug to treat UC.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu-Xin Chen
- Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), School of Biological Engineering and Food, Hubei University of Technology, Wuhan 430068, China
| |
Collapse
|
41
|
Chen H, Sun R, Wang J, Yao S, Batool SS, Yu Z, Huang S, Huang J. Bacillus amyloliquefaciens alleviates the pathological injuries in mice infected with Schistosoma japonicum by modulating intestinal microbiome. Front Cell Infect Microbiol 2023; 13:1172298. [PMID: 37265494 PMCID: PMC10230073 DOI: 10.3389/fcimb.2023.1172298] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 04/24/2023] [Indexed: 06/03/2023] Open
Abstract
Schistosoma japonicum causes serious pathological organ damage and alteration of the intestinal microbiome in the mammalian host, threatening the health of millions of people in China. Bacillus amyloliquefaciens has been reported to be able to alleviate the damage to the gut and liver and maintain the homeostasis of the intestinal microenvironment. However, it was unclear whether B. amyloliquefaciens could alleviate the hepatic and intestinal symptoms caused by S. japonicum. In this study, the intragastric administration of B. amyloliquefaciens was performed to treat S. japonicum-infected mice during the acute phase. Histopathological analysis and 16S rRNA gene sequencing were used to evaluate the pathological damage and changes in the intestinal microbiome. The results of the study showed that B. amyloliquefaciens treatment significantly reduced the degree of granuloma and fibrosis in infected mice. Additionally, recovery of diversity in the intestinal microbiome, decrease in the relative abundance of potential pathogenic bacteria such as Escherichia-Shigella, and reshaping of the interactive network between genera in the intestine were also observed after treatment with B. amyloliquefaciens. Our findings indicated that treatment with B. amyloliquefaciens effectively alleviated the pathological injuries of the liver and intestine in mice infected with S. japonicum by modulating the intestinal microbiome, implying that this probiotic can function as an effective therapeutic agent against schistosomiasis. We hope our study will provide auxiliary strategies and methods for the early prevention of schistosomiasis japonica.
Collapse
Affiliation(s)
- Hao Chen
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Ruizheng Sun
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jingyan Wang
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Siqi Yao
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Syeda Sundas Batool
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Zheng Yu
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Shuaiqin Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
| | - Jing Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- Human Microbiome and Health Group, Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| |
Collapse
|
42
|
Sung TS, Ryoo SB, Lee CH, Choi SM, Nam JW, Kim HB, Lee JY, Lim JD, Park KJ, Lee HT. Prokinetic Activity of Mulberry Fruit, Morus alba L. Nutrients 2023; 15:nu15081889. [PMID: 37111108 PMCID: PMC10143206 DOI: 10.3390/nu15081889] [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: 03/16/2023] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
The fruit of Morus alba L. (MAF) has been consumed as a food worldwide. MAF has also been widely used in traditional medicine for thousands of years in East Asia, and its diverse bioactivities have been reported in numerous publications. However, no prokinetic activity has been reported for MAF or its components. In the present study, therefore, we investigated the effects of MAF on gastrointestinal motor function by measuring the intestinal transit rate (ITR) of Evans blue in mice in vivo. The ITR values accelerated by MAF were significantly higher than those accelerated by cisapride or metoclopramide, suggesting that MAF has potential as a new prokinetic agent to replace cisapride and metoclopramide. We also investigated the effects of MAF on myogenic and neurogenic contractions in human intestinal smooth muscles by measuring spontaneous contractions of smooth muscle strips, smooth muscle contractions induced by neural stimulation, and migrating motor complexes from intestinal segments in the human ileum and sigmoid colon in situ. MAF increased both myogenic and neurogenic contractions to enhance ileal and colonic motility in the human intestine. Taken together, these results indicate that MAF enhanced intestinal motility by increasing both myogenic and neurogenic contractions, thereby accelerating the ITR.
Collapse
Affiliation(s)
- Tae Sik Sung
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Seung-Bum Ryoo
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Chang-Hyun Lee
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, College of Engineering, Dong-Eui University, Busan 47340, Republic of Korea
| | - Seon-Min Choi
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, College of Engineering, Dong-Eui University, Busan 47340, Republic of Korea
| | - Joo-Won Nam
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Hyun-Bok Kim
- National Institute of Agricultural Sciences, Rural Development Administration, Wanju 55365, Republic of Korea
| | - Ji Young Lee
- Department of Ophthalmology and Visual Science, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jung-Dae Lim
- Department of Herbal Medicine Resource, Kangwon National University, Samcheok 25949, Republic of Korea
| | - Kyu Joo Park
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hyun-Tai Lee
- Biopharmaceutical Engineering Major, Division of Applied Bioengineering, College of Engineering, Dong-Eui University, Busan 47340, Republic of Korea
- Core-Facility Center for Tissue Regeneration, Dong-Eui University, Busan 47340, Republic of Korea
| |
Collapse
|
43
|
Blackcurrant Alleviates Dextran Sulfate Sodium (DSS)-Induced Colitis in Mice. Foods 2023; 12:foods12051073. [PMID: 36900589 PMCID: PMC10000425 DOI: 10.3390/foods12051073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Previous studies have reported that anthocyanin (ACN)-rich materials have beneficial effects on ulcerative colitis (UC). Blackcurrant (BC) has been known as one of the foods rich in ACN, while studies demonstrating its effect on UC are rare. This study attempted to investigate the protective effects of whole BC in mice with colitis using dextran sulfate sodium (DSS). Mice were orally given whole BC powder at a dose of 150 mg daily for four weeks, and colitis was induced by drinking 3% DSS for six days. Whole BC relieved symptoms of colitis and pathological changes in the colon. The overproduction of pro-inflammatory cytokines such as IL-1β, TNF-α, and IL-6 in serum and colon tissues was also reduced by whole BC. In addition, whole BC significantly lowered the levels of mRNA and protein of downstream targets in the NF-κB signaling pathway. Furthermore, BC administration increased the expression of genes related to barrier function: ZO-1, occludin, and mucin. Moreover, the whole BC modulated the relative abundance of gut microbiota altered with DSS. Therefore, the whole BC has demonstrated the potential to prevent colitis through attenuation of the inflammatory response and regulation of the gut microbial composition.
Collapse
|
44
|
Li H, Xiao C, Wang F, Guo X, Zhou Z, Jiang Y. Blueberry-Mulberry Extract Alleviates Cognitive Impairment, Regulates Gut Metabolites, and Inhibits Inflammation in Aged Mice. Foods 2023; 12:foods12040860. [PMID: 36832936 PMCID: PMC9956669 DOI: 10.3390/foods12040860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/22/2023] Open
Abstract
Cognitive impairment is associated with aging; however, the underlying mechanism remains unclear. Our previous study found that polyphenol-rich blueberry-mulberry extract (BME) had an antioxidant capability and effectively alleviated cognitive impairment in a mouse model of Alzheimer's disease. Thus, we hypothesized that BME would improve cognitive performance in naturally aging mice and assessed its effects on related signaling pathways. Eighteen-month-old C57BL/6J mice were gavaged with 300 mg/kg/d of BME for 6 weeks. Behavioral phenotypes, cytokine levels, tight junction protein levels, and the histopathology of the brain were assessed, and 16S ribosomal RNA sequencing and targeted metabolome analyses were used for gut microbiota and metabolite measurements. Our results showed that the cognitive performance of aged mice in the Morris water maze test was improved after BME treatment, neuronal loss was reduced, IL-6 and TNF-α levels in the brain and intestine were decreased, and the levels of intestinal tight junction proteins (ZO-1 and occludin) were increased. Further, 16S sequencing showed that BME significantly increased the relative abundance of Lactobacillus, Streptococcus, and Lactococcus and decreased the relative abundance of Blautia, Lachnoclostridium, and Roseburia in the gut. A targeted metabolomic analysis showed that BME significantly increased the levels of 21 metabolites, including α-linolenic acid, vanillic acid, and N-acetylserotonin. In conclusion, BME alters the gut microbiota and regulates gut metabolites in aged mice, which may contribute to the alleviation of cognitive impairment and to inflammation inhibition in both the brain and the gut. Our results provide a basis for future research on natural antioxidant intervention as a treatment strategy for aging-related cognitive impairment.
Collapse
Affiliation(s)
- Hui Li
- Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Changhao Xiao
- Institute of Environmental and Operational Medicine, Tianjin 300050, China
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Feng Wang
- Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Xuqi Guo
- Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Zhongkai Zhou
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yugang Jiang
- Institute of Environmental and Operational Medicine, Tianjin 300050, China
- Correspondence: ; Tel.: +86-139-2077-0716
| |
Collapse
|
45
|
Effect of anthocyanins on gut health markers, Firmicutes-Bacteroidetes ratio and short-chain fatty acids: a systematic review via meta-analysis. Sci Rep 2023; 13:1729. [PMID: 36720989 PMCID: PMC9889808 DOI: 10.1038/s41598-023-28764-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/24/2023] [Indexed: 02/02/2023] Open
Abstract
Researchers discovered that diets rich in anthocyanin-rich fruits and vegetables significantly impacted gut flora. To conclude, large-scale randomized controlled clinical trials are challenging to conduct; therefore, merging data from multiple small studies may aid. A systematic review collects and analyses all research on a particular subject and design. This comprehensive review and meta-analysis examined the influence of dietary anthocyanins on Firmicutes/Bacteroide (Fir/Bac) and short-chain fatty acids (SCFAs) content. The current meta-analysis followed the guidelines of PRISMA-the preferred reporting items for systematic reviews and meta-analyses. Diets high in anthocyanins substantially reduced the Fir/Bac ratio in the assessed trials. Among three SCFAs, the highest impact was observed on acetic acid, followed by propionic acid, and then butanoic acid. The meta-analysis results also obtained sufficient heterogeneity, as indicated by I2 values. There is strong evidence that anthocyanin supplementation improves rodent gut health biomarkers (Fir/Bac and SCFAs), reducing obesity-induced gut dysbiosis, as revealed in this systematic review/meta-analysis. Anthocyanin intervention duration and dosage significantly influenced the Fir/Bac ratio and SCFA. Anthocyanin-rich diets were more effective when consumed over an extended period and at a high dosage.
Collapse
|
46
|
Wang N, Chen W, Cui C, Zheng Y, Yu Q, Ren H, Liu Z, Xu C, Zhang G. The Peanut Skin Procyanidins Attenuate DSS-Induced Ulcerative Colitis in C57BL/6 Mice. Antioxidants (Basel) 2022; 11:2098. [PMID: 36358470 PMCID: PMC9686776 DOI: 10.3390/antiox11112098] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/12/2022] [Accepted: 10/21/2022] [Indexed: 08/10/2023] Open
Abstract
Polyphenols from peanut skin have been reported to possess many beneficial functions for human health, including anti-oxidative, antibacterial, anticancer, and other activities. To date, however, its anti-inflammatory effect and the underlying mechanism remain unclear. In this study, the anti-inflammatory effect of peanut skin procyanidins extract (PSPE) and peanut skin procyanidins (PSPc) were investigated by a dextran sodium sulfate (DSS)-induced colitis mouse model. The results showed that both PSPE and PSPc supplementation reversed the DSS-induced body weight loss and reduced disease activity index (DAI) values, accompanied by enhanced goblet cell numbers and tight junction protein claudin-1 expression in the colon. PSPE and PSPc treatment also suppressed the inflammatory responses and oxidative stress in the colon by down-regulating IL-1β, TNF-α, and MDA expressions. Meanwhile, PSPE and PSPc significantly altered the gut microbiota composition by increasing the relative abundance of Clostridium XlVb and Anaerotruncus, and inhibiting the relative abundance of Alistipes at the genus level. PSPE and PSPc also significantly elevated the production of short-chain fatty acids (SCFAs) in mice with colitis. The correlation analysis suggested that the protective effects of PSPE and PSPc on colitis might be related to the alteration of gut microbiota composition and the formation of SCFAs. In conclusion, the current research indicates that supplementation of PSPE and PSPc could be a promising nutritional strategy for colitis prevention and treatment.
Collapse
Affiliation(s)
- Na Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
- College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
- International Joint Research Center for Animal Immunology, Zhengzhou 450002, China
- Key Laboratory of Nutrition and Healthy Food of Zhengzhou, Zhengzhou 450002, China
| | - Weixuan Chen
- Key Laboratory of Nutrition and Healthy Food of Zhengzhou, Zhengzhou 450002, China
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Chenxu Cui
- College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
- Key Laboratory of Nutrition and Healthy Food of Zhengzhou, Zhengzhou 450002, China
| | - Yuru Zheng
- College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
- Key Laboratory of Nutrition and Healthy Food of Zhengzhou, Zhengzhou 450002, China
| | - Qiuying Yu
- College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
- International Joint Research Center for Animal Immunology, Zhengzhou 450002, China
- Key Laboratory of Nutrition and Healthy Food of Zhengzhou, Zhengzhou 450002, China
| | - Hongtao Ren
- College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
- Key Laboratory of Nutrition and Healthy Food of Zhengzhou, Zhengzhou 450002, China
| | - Zhigang Liu
- Key Laboratory of Nutrition and Healthy Food of Zhengzhou, Zhengzhou 450002, China
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Chao Xu
- College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
- Key Laboratory of Nutrition and Healthy Food of Zhengzhou, Zhengzhou 450002, China
| | - Gaiping Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
- International Joint Research Center for Animal Immunology, Zhengzhou 450002, China
| |
Collapse
|