1
|
Wang L, Zheng W, Men Q, Ren X, Song S, Ai C. Curcumin-loaded polysaccharide microparticles alleviated DSS-induced ulcerative colitis by improving intestinal microecology and regulating MAPK/NF-κB/Nrf2/NLRP3 pathways. Int J Biol Macromol 2024; 281:136687. [PMID: 39427805 DOI: 10.1016/j.ijbiomac.2024.136687] [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: 05/13/2024] [Revised: 09/19/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Curcumin (Cur) exerts many benefits on the host, but its application is limited by its poor bioavailability. In this study, composite polysaccharide microparticles loading Cur (Cur-CPM) was prepared by food-grade materials and gel technology. Its properties were analyzed via the in vitro and in vivo models, and then its benefit on gut health was assessed in DSS-treated mice. Compared to free Cur, CPM extended the residence time and absorption efficiency of Cur in the intestine, effectively ameliorating the symptoms of colitis. Cur-CPM alleviated colonic inflammation by inhibiting the activation of the MAPK and NF-κB pathways and suppressing NLRP3 inflammasome activity, affecting the expression of inflammation-related cytokines and mediators. In addition, Cur-CPM regulated the levels of antioxidants and oxidants in the colon tissues via Nrf2 activation, alleviating oxidative stress. Cur-CPM protected gut barrier function by maintaining the integrity of colonic mucosal layer and tight junction. The underlying mechanism can be attributed not only to the anti-inflammatory and antioxidant activities of Cur but also to modulation of Cur and CPM on the gut microbiota and metabolites. It suggests that Cur-CPM holds the potential to be developed as a functional component to enhance gut health.
Collapse
Affiliation(s)
- Lu Wang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Weiyun Zheng
- School of Agronomy and Life Science, Shanxi Datong University, Datong 037009, PR China
| | - Qiuyue Men
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Xiaomeng Ren
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
2
|
Mio K, Goto Y, Matsuoka T, Komatsu M, Ishii C, Yang J, Kobayashi T, Aoe S, Fukuda S. Barley β-glucan consumption improves glucose tolerance by increasing intestinal succinate concentrations. NPJ Sci Food 2024; 8:69. [PMID: 39349520 PMCID: PMC11444033 DOI: 10.1038/s41538-024-00311-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/15/2024] [Indexed: 10/02/2024] Open
Abstract
Barley is rich in β-glucan, which can alter gut microbiota and metabolome profiles, potentially affecting host metabolism. However, the microbiota and metabolites increased by barley β-glucan remain unclear. In this study, we focused on the gut-microbiota-derived metabolite succinate and investigated the microbiome and metabolome profiles altered by barley β-glucan intake. C57BL/6 J mice were fed a standard or middle-fat diet containing barley flour rich in β-glucan or barley flour without β-glucan, and their gut microbiota and metabolome profiles were analyzed. The results showed increased Bacteroides, Parasutterella, and succinate due to barley β-glucan intake independent of diet differences. Next, we used mice lacking slc13a2, a gene that is involved in the cellular uptake of succinate. Wild-type mice showed improved glucose tolerance after the intake of barley β-glucan, but this effect was attenuated in the slc13a2-deficient mice. These results suggest that barley β-glucan intake increases succinate and succinate-producing bacteria and affects glucose metabolism.
Collapse
Affiliation(s)
- Kento Mio
- Research and Development Department, Hakubaku co., Ltd., Yamanashi, Japan.
| | - Yuka Goto
- Research and Development Department, Hakubaku co., Ltd., Yamanashi, Japan
| | - Tsubasa Matsuoka
- Research and Development Department, Hakubaku co., Ltd., Yamanashi, Japan
| | - Mitsuko Komatsu
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Chiharu Ishii
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Jiayue Yang
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Toshiki Kobayashi
- Research and Development Department, Hakubaku co., Ltd., Yamanashi, Japan
| | - Seiichiro Aoe
- Graduate School of Studies in Human Culture, Otsuma Women's University, Tokyo, Japan.
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan.
- Laboratory for Regenerative Microbiology, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Gut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan.
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.
- Metagen Inc., Tsuruoka, Yamagata, Japan.
| |
Collapse
|
3
|
Huang LS, Yeh YM, Chiu SF, Huang PJ, Chu LJ, Huang CY, Cheng FW, Chen LC, Lin HC, Shih YW, Lin WN, Huang KY. Intestinal microbiota analysis of different Blastocystis subtypes and Blastocystis-negative individuals in Taiwan. Biomed J 2024; 47:100661. [PMID: 37774792 PMCID: PMC11341923 DOI: 10.1016/j.bj.2023.100661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/13/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Intestinal parasitic infections are the most common infectious diseases among Southeast Asian migrant workers in Taiwan, especially for infections with Blastocystis hominis. However, little is known about the impact of Blastocystis subtypes (STs) on the gut microbiota. METHODS We retrospectively evaluated the prevalence of intestinal parasites in a teaching hospital in Northern Taiwan in the period of 2015-2019. Blastocystis-positive stool specimens were collected for ST analysis by polymerase chain reaction in 2020. Intestinal microbiota analyses of different Blastocystis STs and Blastocystis-free individuals were conducted by 16S rRNA sequencing. RESULTS A total of 13,859 subjects were analyzed, of which 1802 cases (13%) were diagnosed with intestinal parasitic infections. B. hominis infections were the most prevalent (n = 1546, 85.7%). ST analysis of Blastocystis-positive samples (n = 150) indicated that ST1 was the most common type, followed by ST3, ST4, ST2, ST7, and ST5. Different Blastocystis STs (ST1, ST3, and ST4) were associated with distinct richness and diversity of the microbiota. Taxonomic profiles revealed that Akkermansia muciniphila was significantly enriched for all analyzed Blastocystis STs, whereas Holdemanella biformis was more abundant in the Blastocystis-free group. Additionally, Succinivibrio dextrinosolvens and Coprococcus eutactus were specifically more abundant in ST3 carriers than in non-infected individuals. CONCLUSION This study demonstrates that A. muciniphila is positively associated with all Blastocystis STs, while H. biformis was negatively associated with them. Several bacteria were enriched in specific STs, highlighting the need for further microbiota analysis at the ST level to elucidate the pathogenicity of Blastocystis.
Collapse
Affiliation(s)
- Li-San Huang
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei, Taiwan; Department of Inspection, Taipei City Hospital, Renai Branch, Taipei, Taiwan; Host-Parasite Interactions Laboratory, National Defense Medical Center, Taipei, Taiwan
| | - Yuan-Ming Yeh
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Shu-Fang Chiu
- Department of Inspection, Taipei City Hospital, Renai Branch, Taipei, Taiwan; Host-Parasite Interactions Laboratory, National Defense Medical Center, Taipei, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Po-Jung Huang
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Lichieh Julie Chu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan; Department of Otolaryngology - Head & Neck Surgery, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Ching-Yun Huang
- Host-Parasite Interactions Laboratory, National Defense Medical Center, Taipei, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Fang-Wen Cheng
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei, Taiwan; Host-Parasite Interactions Laboratory, National Defense Medical Center, Taipei, Taiwan
| | - Lih-Chyang Chen
- Department of Medicine, Mackay Medical College, New Taipei, Taiwan
| | - Hsin-Chung Lin
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei, Taiwan; Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ya-Wen Shih
- School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Wei-Ning Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu Jen Catholic University, New Taipei, Taiwan
| | - Kuo-Yang Huang
- Graduate Institute of Pathology and Parasitology, National Defense Medical Center, Taipei, Taiwan; Host-Parasite Interactions Laboratory, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
4
|
Padhi S, Sarkar P, Sahoo D, Rai AK. Potential of fermented foods and their metabolites in improving gut microbiota function and lowering gastrointestinal inflammation. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024. [PMID: 38299734 DOI: 10.1002/jsfa.13313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/12/2024] [Accepted: 01/13/2024] [Indexed: 02/02/2024]
Abstract
Foods prepared using microbial conversion of major and minor food components, which are otherwise known as fermented foods continue to impact human health. The live microorganisms and transformed metabolites can also have a deep influence on the gut microbiota, the multifaceted population of microorganisms dwelling inside the gut play a key role in wellbeing of an individual. The probiotic strains delivered through the consumption of fermented food and other bioactive components such as polyphenolic metabolites, bioactive peptides, short-chain fatty acids and others including those produced via gut microbiota mediated transformations have been proposed to balance the gut microbiota diversity and activity, and also to regulate the inflammation in the gut. However, little is known about such effects and only a handful of fermented foods have been explored to date. We herein review the recent knowledge on the dysbiotic gut microbiota linking to major gut inflammatory diseases. Also, evidences that fermented food consumption modulates the gut microbiota, and its impact on the gut inflammation and inflammatory diseases have been discussed. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Srichandan Padhi
- Nutrition Biotechnlogy Division, National Agri-Food Biotechnology Institute, Mohali, India
| | - Puja Sarkar
- Nutrition Biotechnlogy Division, National Agri-Food Biotechnology Institute, Mohali, India
| | | | - Amit Kumar Rai
- Nutrition Biotechnlogy Division, National Agri-Food Biotechnology Institute, Mohali, India
| |
Collapse
|
5
|
Zhou X, Zhang H, Li S, Jiang Y, Deng J, Yang C, Chen X, Jiang L. Effects of different levels of Citri Sarcodactylis Fructus by-products fermented feed on growth performance, serum biochemical, and intestinal health of cyan-shank partridge birds. Sci Rep 2023; 13:20130. [PMID: 37978234 PMCID: PMC10656579 DOI: 10.1038/s41598-023-47303-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 11/11/2023] [Indexed: 11/19/2023] Open
Abstract
This research aimed to investigate the effects of supplements containing fermented feed made from Citri Sarcodactylis Fructus by-products (CSFBP-Fermented feed) on the growth performance, immunological function, and gut health of broilers. 1080 cyan-shank partridge birds aged 47 days were chosen and casually distributed to four groups, each with 6 replicates and 45 birds per replicate. The experimental groups were provided with 1% (group T2), 3% (group T3) and 5% (group T4) of CSFBP-fermented feed in the basic diet, while the control group (group T1) received the basic diet. The findings revealed that supplementation with CSFBP-Fermented feed reduced ADFI and FCR and improved ADG in birds (P < 0.05). MDA levels in the serum of birds fed CSFBP-fermented feed were lower than in the control group (P < 0.05). The CAT activity in the serum of broilers increased after supplementation with 3% CSFBP-Fermented feed (P < 0.05). Supplementing broilers with CSFBP-fermented feed enhanced VH in the ileum, jejunum, and duodenum (P < 0.05). The addition of 3% CSFBP-Fermented feed decreased CD in the jejunum (P < 0.05). The addition of 3% and 5% CSFBP-Fermented feed increased the mRNA expression of ZO-1 and Occludin in the jejunum of broiler chickens and reduced the mRNA expression of IL-6 (P < 0.05). The addition of 3% CSFBP-Fermented feed increased the mRNA expression of Claudin in the jejunum of broiler chickens and reduced IL-1β mRNA expression (P < 0.05). Compared to the control group, all experimental groups exhibited decreased mRNA expression of TNF-α and INF-γ in the jejunal mucosa of the birds (P < 0.05). According to research using high-throughput sequencing of microorganisms' 16S rDNA, and an analysis of α-diversity found that supplementing broilers with 3% CSFBP-Fermented feed decreased the number of bacteria in their cecum (P < 0.05). Bacteroidota was higher in all groups after supplementation with CSFBP-Fermented feed. At the genus level, after addition with 3% CSFBP-Fermented feed, the abundance of Bacteroide and Prevotellaceae_Ga6A1_group were higher than the control group (33.36% vs 29.95%, 4.35% vs 2.94%). The abundance of Rikenellaceae_RC9_gut_group and Fusobacterium were lower than the control group (5.52% vs. 7.17%,0.38% vs. 1.33%). In summary, supplementing the diet with CSFBP-Fermented feed can promote the growth of performance by enhancing intestinal morphology, and barrier function, as well as modulating intestinal inflammatory factors and microbial composition in broilers.
Collapse
Affiliation(s)
- Xinhong Zhou
- Leshan Academy of Agriculture Science, Leshan, 614001, Sichuan, China
- College of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China
| | - Huaidan Zhang
- Leshan Academy of Agriculture Science, Leshan, 614001, Sichuan, China
| | - Shiyi Li
- Leshan Academy of Agriculture Science, Leshan, 614001, Sichuan, China
| | - Yilong Jiang
- Leshan Academy of Agriculture Science, Leshan, 614001, Sichuan, China
| | - Jicheng Deng
- Leshan Academy of Agriculture Science, Leshan, 614001, Sichuan, China
| | - Chuanpeng Yang
- Leshan Academy of Agriculture Science, Leshan, 614001, Sichuan, China
| | - Xianxin Chen
- Leshan Academy of Agriculture Science, Leshan, 614001, Sichuan, China.
| | - Li Jiang
- College of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, 621010, Sichuan, China.
| |
Collapse
|
6
|
Wu L, Hu J, Yi X, Lv J, Yao J, Tang W, Zhang S, Wan M. Gut microbiota interacts with inflammatory responses in acute pancreatitis. Therap Adv Gastroenterol 2023; 16:17562848231202133. [PMID: 37829561 PMCID: PMC10566291 DOI: 10.1177/17562848231202133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/01/2023] [Indexed: 10/14/2023] Open
Abstract
Acute pancreatitis (AP) is one of the most common acute abdominal conditions, and its incidence has been increasing for years. Approximately 15-20% of patients develop severe AP (SAP), which is complicated by critical inflammatory injury and intestinal dysfunction. AP-associated inflammation can lead to the gut barrier and function damage, causing dysbacteriosis and facilitating intestinal microbiota migration. Pancreatic exocrine deficiency and decreased levels of antimicrobial peptides in AP can also lead to abnormal growth of intestinal bacteria. Meanwhile, intestinal microbiota migration influences the pancreatic microenvironment and affects the severity of AP, which, in turn, exacerbates the systemic inflammatory response. Thus, the interaction between the gut microbiota (GM) and the inflammatory response may be a key pathogenic feature of SAP. Treating either of these factors or breaking their interaction may offer some benefits for SAP treatment. In this review, we discuss the mechanisms of interaction of the GM and inflammation in AP and factors that can deteriorate or even cure both, including some traditional Chinese medicine treatments, to provide new methods for studying AP pathogenesis and developing therapies.
Collapse
Affiliation(s)
- Linjun Wu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
- Hospital of Chinese Traditional Medicine of Leshan, Leshan, China
| | - Jing Hu
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
- Hospital of Chinese Traditional Medicine of Leshan, Leshan, China
| | - Xiaolin Yi
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
- Intensive Care Unit, Suining Municipal Hospital of TCM, Suining, China
| | - Jianqin Lv
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Wenfu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Chengdu, China
| | - Shu Zhang
- Department of Emergency Medicine, Emergency Medical Laboratory, West China
- Hospital, Sichuan University, Guo Xue Road 37, Chengdu 610041, Sichuan, China
| | - Meihua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China
- Hospital, Sichuan University, Guo Xue Road 37, Chengdu 610041, China
| |
Collapse
|
7
|
Yang Y, Cui B, Lv Y, Lu X, Shen W, Feng M, Ding X, Dong P, Wang Y. Plateau pika fecal microbiota transplantation ameliorates inflammatory bowel disease manifestations in a mouse model of colitis. Front Microbiol 2023; 14:1228778. [PMID: 37795305 PMCID: PMC10546031 DOI: 10.3389/fmicb.2023.1228778] [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: 05/26/2023] [Accepted: 08/21/2023] [Indexed: 10/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a serious global public health concern. Although the pathogenesis of the disease is currently unknown, it has been reported to be associated with both intestinal microbiota and inflammatory mediators. There is evidence suggesting that the feces of the Plateau pika is useful for treating gastrointestinal injuries and pain. Although fecal microbiota transplantation is highly efficacious intervention for IBD prevention, however, potential the transfer of pathogenic microbes or toxic substances is potentially hazardous. Fortunately, micropore filtering of the donor feces can minimize the risk of bacterial infection allowing retention of the therapeutic effects of the residual bacteriophages. Here, we demonstrated that Plateau pika feces not only alleviated the IBD symptoms but also promoted optimal structure and composition of the intestinal microbiota. Additionally, Plateau pika feces transfer also enhanced phenotypic features, such as, body-weight, disease activity index, and histological scores. In conclusion, Plateau pika feces was found to protect mice against colitis induced by dextran sodium sulfate by reducing inflammation and regulating microbial dysbiosis. These findings suggest the potential of Plateau pika feces as an alternative therapy for IBD.
Collapse
Affiliation(s)
- Yayuan Yang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Baiqiang Cui
- Department of Thoracic Surgery, Gansu Province Hospital, Lanzhou, China
| | - Yanan Lv
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangnan Lu
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Wenxiang Shen
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Min Feng
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xuezhi Ding
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Pengcheng Dong
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yu Wang
- Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
8
|
Mao B, Xiang Q, Tang X, Zhang Q, Liu X, Zhao J, Cui S, Zhang H. Lactobacillus reuteri CCFM1175 and Lactobacillus paracasei CCFM1176 Could Prevent Capsaicin-Induced Ileal and Colonic Injuries. Probiotics Antimicrob Proteins 2023:10.1007/s12602-023-10106-1. [PMID: 37314694 DOI: 10.1007/s12602-023-10106-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2023] [Indexed: 06/15/2023]
Abstract
Capsaicin (CAP) is usually reported to have many biological activities. However, a large intake of CAP may cause heartburn, gastrointestinal pain, and diarrhea. In this study, mice were gavaged with nine lactic acid bacteria (LAB) strains for two weeks, in which the mice were treated with CAP at the second week and lasted for one week. We tried to identify potential probiotics that could prevent CAP-induced intestinal injury and investigate the mechanisms. The modulation of transient receptor potential vanilloid 1 (TRPV1), levels of short-chain fatty acids (SCFAs), and the composition of gut microbiota were analyzed. The results showed that Lactobacillus reuteri CCFM1175 and Lactobacillus paracasei CCFM1176 effectively attenuated CAP-induced injuries to the ileum and colon, including relieving the damage to colonic crypt structures, increasing the number of goblet cells, decreasing levels of interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α), increasing levels of anti-inflammatory factors (IL-10), and reducing levels of substance P (SP) and calcitonin gene-related peptide (CGRP) in serum and colon tissue. Further analysis showed that L. reuteri CCFM1175 increased the relative abundance of Ruminococcaceae UCG_014 and Akkermansia. L. paracasei CCFM1176 downregulated the expression of TRPV1 in the ileal and colonic tissues and promoted the relative abundance of Ruminococcaceae UCG_014 and Lachnospiraceae UCG_006. These results indicate that L. reuteri CCFM1175 and L. paracasei CCFM1176 could prevent CAP-induced intestinal injury and be used as probiotics to improve the gastrointestinal health.
Collapse
Affiliation(s)
- Bingyong Mao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Qunran Xiang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Xin Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
9
|
Gowen R, Gamal A, Di Martino L, McCormick TS, Ghannoum MA. Modulating the Microbiome for Crohn's Disease Treatment. Gastroenterology 2023; 164:828-840. [PMID: 36702360 PMCID: PMC10152883 DOI: 10.1053/j.gastro.2023.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/12/2022] [Accepted: 01/06/2023] [Indexed: 01/28/2023]
Abstract
The central role of the gut microbiota in the regulation of health and disease has been convincingly demonstrated. Polymicrobial interkingdom interactions between bacterial (the bacteriome) and fungal (the mycobiome) communities of the gut have become a prominent focus for development of potential therapeutic approaches. In addition to polymicrobial interactions, the complex gut ecosystem also mediates interactions between the host and the microbiota. These interactions are complex and bidirectional; microbiota composition can be influenced by host immune response, disease-specific therapeutics, antimicrobial drugs, and overall ecosystems. However, the gut microbiota also influences host immune response to a drug or therapy by potentially transforming the drug's structure and altering bioavailability, activity, or toxicity. This is especially true in cases where the gut microbiota has produced a biofilm. The negative ramifications of biofilm formation include alteration of gut permeability, enhanced antimicrobial resistance, and alteration of host immune response effectiveness. Natural modulation of the gut microbiota, using probiotic and prebiotic approaches, may also be used to affect the host microbiome, a type of "natural" modulation of the host microbiota composition. In this review, we discuss potential bidirectional interactions between microbes and host, and we describe the changes in gut microbiota induced by probiotic and prebiotic approaches as well as their potential clinical consequences, including biofilm formation. We outline a systematic approach to designing probiotics capable of altering the host microbiota in disease states, using Crohn's disease as a model chronic disease. Understanding how the effective changes in the microbiome may enhance treatment efficacy may unlock the possibility of modulating the gut microbiome to improve treatment using a natural approach.
Collapse
Affiliation(s)
- Rachael Gowen
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio; University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Ahmed Gamal
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio; University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Luca Di Martino
- University Hospitals Cleveland Medical Center, Cleveland, Ohio; Department of Medicine, Case Western Reserve University, Cleveland, Ohio; Case Digestive Health Research Institute, Case Western Reserve University, Cleveland Ohio
| | - Thomas S McCormick
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio; University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Mahmoud A Ghannoum
- Department of Dermatology, Case Western Reserve University, Cleveland, Ohio; University Hospitals Cleveland Medical Center, Cleveland, Ohio.
| |
Collapse
|
10
|
Yang Y, Li M, Liu Q, Zhao Q, Zeng J, Wang Q, Zhao Y, Du F, Chen Y, Shen J, Luo H, Wang S, Li W, Chen M, Li X, Wang F, Sun Y, Gu L, Xiao Z, Du Y, Wu X. Starch from Pueraria lobata and the amylose fraction alleviates dextran sodium sulfate induced colitis in mice. Carbohydr Polym 2023; 302:120329. [PMID: 36604040 DOI: 10.1016/j.carbpol.2022.120329] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/15/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
Starch from Pueraria lobata (PLS) had polyhedral or spherical granules, displaying a bimodal size distribution within 0.6-30 μm. It showed a trimodal distribution of different molecular weight peaks, with amylose fraction of 18.2 %. PLS had a high crystallinity degree of 37.76 % and consisted of C-type starch, which gelatinized at 64.46-79.61 °C, with a high range of gelatinization (15.15 °C) and high enthalpy (13.98 J/g). A 21-day supplementation of PLS presented a regulative effect on gut microbiota in normal mice, and alleviated DSS-induced murine colitis through attenuating colonic inflammation, maintaining barrier function, preventing gut dysbiosis, increasing the short-chain fatty acids production and inhibiting NF-κB/IL-1β axis. The protective effect of PLS against colitis was in a gut microbiota-dependent manner. Notably, the amylose fraction was responsible for the prebiotic effect of PLS. The results would potentiate new application of PLS and the amylose fraction as functional prebiotics for prevention of colitis.
Collapse
Affiliation(s)
- Yifei Yang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, 646000 Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, 646000 Luzhou, Sichuan, China
| | - Qingsong Liu
- The First People's Hospital of Neijiang, 641000 Neijiang, Sichuan, China
| | - Qianyun Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, 646000 Luzhou, Sichuan, China
| | - Jiuping Zeng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, 646000 Luzhou, Sichuan, China
| | - Qin Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, 646000 Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, 646000 Luzhou, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, 646000 Luzhou, Sichuan, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, 646000 Luzhou, Sichuan, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, 646000 Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, 646000 Luzhou, Sichuan, China
| | - Haoming Luo
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, 646000 Luzhou, Sichuan, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, 999078, Macao
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Department of Oncology, Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China.
| | - Yu Du
- Medical Cosmetology Center, Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, 646000 Luzhou, Sichuan, China.
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, 646000 Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, 646000 Luzhou, Sichuan, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, 999078, Macao.
| |
Collapse
|
11
|
Treeriya R, Ho PN, Titapun A, Klanrit P, Suksawat M, Kulthawatsiri T, Sirirattanakul S, Loilome W, Namwat N, Wangwiwatsin A, Chamadol N, Khuntikeo N, Phetcharaburanin J. 1H NMR fecal metabolic phenotyping of periductal fibrosis- and cholangiocarcinoma-specific metabotypes defining perturbation in gut microbial-host co-metabolism. PeerJ 2023; 11:e15386. [PMID: 37187520 PMCID: PMC10178365 DOI: 10.7717/peerj.15386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Background The liver fluke Opisthorchis viverrini (OV), which subsequently inhabits the biliary system and results in periductal fibrosis (PDF), is one of the primarily causes of cholangiocarcinoma (CCA), a bile duct cancer with an exceptionally high incidence in the northeast of Thailand and other Greater Mekong Subregion (GMS) countries. Insights in fecal metabolic changes associated with PDF and CCA are required for further molecular research related to gut health and potential diagnostic biological marker development. Methods In this study, nuclear magnetic resonance (NMR) metabolomics was applied for fecal metabolic phenotyping from 55 fecal water samples across different study groups including normal bile duct, PDF and CCA groups. Results By using NMR spectroscopy-based metabolomics, fecal metabolic profiles of patients with CCA or PDF and of individuals with normal bile duct have been established with a total of 40 identified metabolites. Further multivariate statistical analysis and hierarchical clustering heat map have demonstrated the PDF- and CCA-specific metabotypes through various altered metabolite groups including amino acids, alcohols, amines, anaerobic glycolytic metabolites, fatty acids, microbial metabolites, sugar, TCA cycle intermediates, tryptophan catabolism substrates, and pyrimidine metabolites. Compared to the normal bile duct group, PDF individuals showed the significantly elevated relative concentrations of fecal ethanol, glycine, tyrosine, and N-acetylglucosamine whereas CCA patients exhibited the remarkable fecal metabolic changes that can be evident through the increased relative concentrations of fecal uracil, succinate, and 5-aminopentanoate. The prominent fecal metabolic alterations between CCA and PDF were displayed by the reduction of relative concentration of methanol observed in CCA. The metabolic alterations associated with PDF and CCA progression have been proposed with the involvement of various metabolic pathways including TCA cycle, ethanol biogenesis, hexamine pathway, methanol biogenesis, pyrimidine metabolism, and lysine metabolism. Among them, ethanol, methanol, and lysine metabolism strongly reflect the association of gut-microbial host metabolic crosstalk in PDF and/or CCA patients. Conclusion The PDF- and CCA-associated metabotypes have been investigated displaying their distinct fecal metabolic patterns compared to that of normal bile duct group. Our study also demonstrated that the perturbation in co-metabolism of host and gut bacteria has been involved from the early step since OV infection to CCA tumorigenesis.
Collapse
Affiliation(s)
- Rujikorn Treeriya
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Phuc N. Ho
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Attapol Titapun
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Poramate Klanrit
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Manida Suksawat
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Thanaporn Kulthawatsiri
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Suphasarang Sirirattanakul
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharin Loilome
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nisana Namwat
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Arporn Wangwiwatsin
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Nittaya Chamadol
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Narong Khuntikeo
- Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Jutarop Phetcharaburanin
- Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Khon Kaen University Phenome Centre, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Department of Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
12
|
Chompre G, Sambolin L, Cruz ML, Sanchez R, Rodriguez Y, Rodríguez-Santiago RE, Yamamura Y, Appleyard CB. A one month high fat diet disrupts the gut microbiome and integrity of the colon inducing adiposity and behavioral despair in male Sprague Dawley rats. Heliyon 2022; 8:e11194. [PMID: 36387539 PMCID: PMC9663868 DOI: 10.1016/j.heliyon.2022.e11194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/17/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
High-fat diet (HFD) is associated with gut microbiome dysfunction and mental disorders. However, the time-dependence as to when this occurs is unclear. We hypothesized that a short-term HFD causes colonic tissue integrity changes resulting in behavioral changes. Rats were fed HFD or low-fat diet (LFD) for a month and gut microbiome, colon, and behavior were evaluated. Behavioral despair was found in the HFD group. Although obesity was absent, the HFD group showed increased percent weight gain, epididymal fat tissue, and leptin expression. Moreover, the HFD group had increased colonic damage, decreased expression of the tight junction proteins, and higher lipopolysaccharides (LPS) in serum. Metagenomic analysis revealed that the HFD group had more Bacteroides and less S24-7 which correlated with the decreased claudin-5. Finally, HFD group showed an increase of microglia percent area, increased astrocytic projections, and decreased phospho-mTOR. In conclusion, HFD consumption in a short period is still sufficient to disrupt gut integrity resulting in LPS infiltration, alterations in the brain, and behavioral despair even in the absence of obesity.
Collapse
Affiliation(s)
- Gladys Chompre
- Biology and Biotechnology Department, Pontifical Catholic University of Puerto Rico, Ponce, Puerto Rico
- Basic Sciences Department, Division of Physiology, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Lubriel Sambolin
- Basic Sciences Department, Division of Pharmacology, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Myrella L. Cruz
- Basic Sciences Department, Division of Physiology, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Rafael Sanchez
- AIDS Research Infrastructure Program, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Yarelis Rodriguez
- Basic Sciences Department, Division of Physiology, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Ronald E. Rodríguez-Santiago
- AIDS Research Infrastructure Program, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Yasuhiro Yamamura
- AIDS Research Infrastructure Program, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| | - Caroline B. Appleyard
- Basic Sciences Department, Division of Physiology, Ponce Health Sciences University/Ponce Research Institute, Ponce, Puerto Rico
| |
Collapse
|
13
|
Madella AM, Van Bergenhenegouwen J, Garssen J, Masereeuw R, Overbeek SA. Microbial-Derived Tryptophan Catabolites, Kidney Disease and Gut Inflammation. Toxins (Basel) 2022; 14:toxins14090645. [PMID: 36136583 PMCID: PMC9505404 DOI: 10.3390/toxins14090645] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Uremic metabolites, molecules either produced by the host or from the microbiota population existing in the gastrointestinal tract that gets excreted by the kidneys into urine, have significant effects on both health and disease. Tryptophan-derived catabolites are an important group of bacteria-produced metabolites with an extensive contribution to intestinal health and, eventually, chronic kidney disease (CKD) progression. The end-metabolite, indoxyl sulfate, is a key contributor to the exacerbation of CKD via the induction of an inflammatory state and oxidative stress affecting various organ systems. Contrastingly, other tryptophan catabolites positively contribute to maintaining intestinal homeostasis and preventing intestinal inflammation—activities signaled through nuclear receptors in particular—the aryl hydrocarbon receptor (AhR) and the pregnane X receptor (PXR). This review discusses the origins of these catabolites, their effect on organ systems, and how these can be manipulated therapeutically in the future as a strategy to treat CKD progression and gut inflammation management. Furthermore, the use of biotics (prebiotics, probiotics, synbiotics) as a means to increase the presence of beneficial short-chain fatty acids (SCFAs) to achieve intestinal homeostasis is discussed.
Collapse
Affiliation(s)
- Avra Melina Madella
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Correspondence: (A.M.M.); or (S.A.O.); Tel.: +31-30-209-5000 (S.A.O.)
| | - Jeroen Van Bergenhenegouwen
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Danone Nutricia Research, Uppsalalaan 12, Utrecht Science Park, 3584 CT Utrecht, The Netherlands
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Danone Nutricia Research, Uppsalalaan 12, Utrecht Science Park, 3584 CT Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Saskia Adriana Overbeek
- Department of Pharmaceutical Sciences, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Danone Nutricia Research, Uppsalalaan 12, Utrecht Science Park, 3584 CT Utrecht, The Netherlands
- Correspondence: (A.M.M.); or (S.A.O.); Tel.: +31-30-209-5000 (S.A.O.)
| |
Collapse
|
14
|
Xu L, Liu B, Huang L, Li Z, Cheng Y, Tian Y, Pan G, Li H, Xu Y, Wu W, Cui Z, Xie L. Probiotic Consortia and Their Metabolites Ameliorate the Symptoms of Inflammatory Bowel Diseases in a Colitis Mouse Model. Microbiol Spectr 2022; 10:e0065722. [PMID: 35730951 PMCID: PMC9430814 DOI: 10.1128/spectrum.00657-22] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/06/2022] [Indexed: 12/30/2022] Open
Abstract
Inflammatory bowel disease (IBD) has become a global public health problem. Although the pathogenesis of the disease is unknown, a potential association between the gut microbiota and inflammatory signatures has been established. Probiotics, especially Lactobacillus or Bifidobacterium, are orally taken as food supplements or microbial drugs by patients with IBD or gastrointestinal disorders due to their safety, efficacy, and power to restore the gut microenvironment. In the current study, we investigated the comprehensive effects of probiotic bacterial consortia consisting of Lactobacillus reuteri, Lactobacillus gasseri, Lactobacillus acidophilus (Lactobacillus spp.), and Bifidobacterium lactis (Bifidobacterium spp.) or their metabolites in a dextran sodium sulfate (DSS)-induced colitis mouse model. Our data demonstrate that probiotic consortia not only ameliorate the disease phenotype but also restore the composition and structure of the gut microbiota. Moreover, the effect of probiotic consortia is better than that of any single probiotic strain. The results also demonstrate that mixed fermentation metabolites are capable of ameliorating the symptoms of gut inflammation. However, the administration of metabolites is not as effective as probiotic consortia with respect to phenotypic characteristics, such as body weight, disease activity index (DAI), and histological score. In addition, mixed metabolites led only to changes in intestinal flora composition. In summary, probiotic consortia and metabolites could exert protective roles in the DSS-induced colitis mouse model by reducing inflammation and regulating microbial dysbiosis. These findings from the current study provide support for the development of probiotic-based microbial products as an alternative therapeutic strategy for IBD. IMPORTANCE IBD is a chronic nonspecific inflammatory disease. IBD is characterized by a wide range of lesions, often involving the entire colon, and is characterized mainly by ulcers and erosions of the colonic mucosa. In the present study, we investigated the efficacy of probiotics on the recovery of gut inflammation and the restoration of gut microecology. We demonstrate that probiotic consortia have a superior effect in inhibiting inflammation and accelerating recovery compared with the effects observed in the control group or groups administered with a single strain. These results support the utilization of probiotic consortia as an alternative therapeutic approach to treat IBD.
Collapse
Affiliation(s)
- Limin Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Bingdong Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Liujing Huang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ze Li
- School of Public Health, Xinxiang Medical University, Xinxiang, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Yanbo Cheng
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Ye Tian
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Guihua Pan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Huijun Li
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Yinlan Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Zongbin Cui
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Liwei Xie
- School of Public Health, Xinxiang Medical University, Xinxiang, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Zhang Y, Feng D, Zeng Y, Zhang H, Du X, Fu Y, Wang X, Lian D, Wang R, Xiao H, Wei N, Zhai F, Liu H. Xuedan Sustained Release Pellets Ameliorate Dextran Sulfate Sodium-Induced Ulcerative Colitis in Rats by Targeting Gut Microbiota and MAPK Signaling Pathways. Front Pharmacol 2022; 13:833972. [PMID: 35652042 PMCID: PMC9149600 DOI: 10.3389/fphar.2022.833972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022] Open
Abstract
Cucurbitacins have a variety of bioactivities, such as anticancer, anti-inflammatory, antidepressant-like, and antiviral effects, but their pharmacological effect in ulcerative colitis (UC) has not been reported until now. Thus, this study aims to investigate the preventive effects of Xuedan sustained release pellets (XSPs) on UC rats and the underlying mechanisms. XSPs were prepared by extracting cucurbitacins from Hemsleya. Experimental UC rats were induced by the intake of 4% dextran sulfate sodium (DSS) for a week and treated with different doses of XSP (0.95, 1.90, and 3.8 mg/kg). The body weight, colon length, disease activity index (DAI), and histological changes of colonic tissue were measured. In addition, the expressions of pro-inflammatory cytokines were detected by using the enzyme-linked immunosorbent assay. Pathways involved in the intestinal inflammation were targeted by RNA-sequencing. Moreover, the changes of gut microbial diversity and composition were analyzed by the 16SrNA analysis and the contents of short-chain fatty acids (SCFAs) were detected by GC-MS. The results revealed that XSP intervention greatly restored the weight loss and colonic shortening (p < 0.05) and reduced the raised DAI scores, myeloperoxidase, and nitric oxide activities in UC in rats (p < 0.05). XSP administration also downregulated the protein levels of pro-inflammatory factors IL-1β, IL-6, and TNF-α. Notably, it was found that XSP considerably suppressed the activation of the MAPK signaling pathway. In addition, XSP treatment improved the balance of gut microbiota that was disturbed by DSS. The beneficial bacteria Lachnospiraceae_NK4A136 group and Lactobacillus at the genus level significantly increased in the XSP group, which had decreased with the use of DSS (p < 0.05). Pathogenic bacteria including Escherichia-Shigella and Bacteroides in UC in rats were reduced by XSP intervention. Furthermore, XSP significantly elevated the production of SCFAs in UC in rats (p < 0.05). These alterations in inflammatory status were accompanied with changes in gut microbiota diversity and SCFA production. In conclusion, XSP exhibited protective effects against DSS-induced UC in rats. XSP treatment decreased inflammation via modulation of gut microbiota composition and SCFA production.
Collapse
Affiliation(s)
- Yingchun Zhang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Dan Feng
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Yue Zeng
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Hanyu Zhang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xiaohong Du
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Yang Fu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Xinhui Wang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Dingyue Lian
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Ruikang Wang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Hongyu Xiao
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Ning Wei
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| | - Fuqiang Zhai
- Research Institute for New Materials and Technology, Chongqing University of Arts and Sciences, Chongqing, China
| | - Hanru Liu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing, China
| |
Collapse
|
16
|
Du C, Wang K, Zhao Y, Nan X, Chen R, Quan S, Xiong B. Supplementation with Milk-Derived Extracellular Vesicles Shapes the Gut Microbiota and Regulates the Transcriptomic Landscape in Experimental Colitis. Nutrients 2022; 14:nu14091808. [PMID: 35565775 PMCID: PMC9104790 DOI: 10.3390/nu14091808] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022] Open
Abstract
Harboring various proteins, lipids, and RNAs, the extracellular vesicles (EVs) in milk exert vital tissue-specific immune-protective functions in neonates via these bioactive cargos. This study aims to explore the anti-inflammatory effects of bovine milk-derived EVs on a dextran sulfate sodium (DSS)-induced colitis model and to determine the underlying molecular mechanisms. Sixty C57BL/6 mice were divided into the NC group (normal control), DSS group (DSS + PBS), DSS + LOW group (DSS + 1.5 × 108 p/g EVs), DSS + MID group (DSS + 1.5 × 109 p/g EVs), and DSS + HIG group (DSS + 1.0 × 1010 p/g EVs). Histopathological sections, the gut microbiota, and intestinal tissue RNA-Seq were used to comprehensively evaluate the beneficial functions in mitigating colitis. The morphology exhibited that the milk-derived EVs contributed to the integrity of the superficial epithelial structure in the intestine. Additionally, the concentrations of IL-6 and TNF-α in the colon tissues were significantly decreased in the EVs-treated mice. The abundances of the Dubosiella, Bifidobacterium, UCG-007, Lachnoclostridium, and Lachnospiraceae genera were increased in the gut after treatment with the milk-derived EVs. Additionally, the butyrate and acetate production were enriched in feces. In addition, 1659 genes were significantly down-regulated and 1981 genes were significantly up-regulated in the EVs-treated group. Meanwhile, 82 lncRNAs and 6 circRNAs were also differentially expressed. Overall, the milk-derived EVs could attenuate colitis through optimizing gut microbiota abundance and by manipulating intestinal gene expression, implying their application potential for colitis prevention.
Collapse
Affiliation(s)
- Chunmei Du
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (C.D.); (K.W.); (Y.Z.); (X.N.); (R.C.)
| | - Kun Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (C.D.); (K.W.); (Y.Z.); (X.N.); (R.C.)
| | - Yiguang Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (C.D.); (K.W.); (Y.Z.); (X.N.); (R.C.)
| | - Xuemei Nan
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (C.D.); (K.W.); (Y.Z.); (X.N.); (R.C.)
| | - Ruipeng Chen
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (C.D.); (K.W.); (Y.Z.); (X.N.); (R.C.)
| | - Suyu Quan
- College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin 300384, China
- Correspondence: (S.Q.); (B.X.); Tel.: +86-022-2378-1297 (S.Q.); +86-010-6281-6017 (B.X.)
| | - Benhai Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (C.D.); (K.W.); (Y.Z.); (X.N.); (R.C.)
- Correspondence: (S.Q.); (B.X.); Tel.: +86-022-2378-1297 (S.Q.); +86-010-6281-6017 (B.X.)
| |
Collapse
|
17
|
Abstract
Environmental chemicals can alter gut microbial community composition, known as dysbiosis. However, the gut microbiota is a highly dynamic system and its functions are still largely underexplored. Likewise, it is unclear whether xenobiotic exposure affects host health through impairing host-microbiota interactions. Answers to this question not only can lead to a more precise understanding of the toxic effects of xenobiotics but also can provide new targets for the development of new therapeutic strategies. Here, we aim to identify the major challenges in the field of microbiota-exposure research and highlight the need to exam the health effects of xenobiotic-induced gut microbiota dysbiosis in host bodies. Although the changes of gut microbiota frequently co-occur with the xenobiotic exposure, the causal relationship of xenobiotic-induced microbiota dysbiosis and diseases is rarely established. The high dynamics of the gut microbiota and the complex interactions among exposure, microbiota, and host, are the major challenges to decipher the specific health effects of microbiota dysbiosis. The next stage of study needs to combine various technologies to precisely assess the xenobiotic-induced gut microbiota perturbation and the subsequent health effects in host bodies. The exposure, gut microbiota dysbiosis, and disease outcomes have to be causally linked. Many microbiota-host interactions are established by previous studies, including signaling metabolites and response pathways in the host, which may use as start points for future research to examine the mechanistic interactions of exposure, gut microbiota, and host health. In conclusion, to precisely understand the toxicity of xenobiotics and develop microbiota-based therapies, the causal and mechanistic links of exposure and microbiota dysbiosis have to be established in the next stage study.
Collapse
Affiliation(s)
- Liang Chi
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, United States
| | - Pengcheng Tu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, United States
| | - Hongyu Ru
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, United States
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, United States,CONTACT Kun Lu Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC27599, United States
| |
Collapse
|
18
|
He W, Gao Y, Guo Z, Yang Z, Wang X, Liu H, Sun H, Shi B. Effects of fermented wheat bran and yeast culture on growth performance, immunity, and intestinal microflora in growing-finishing pigs. J Anim Sci 2021; 99:6409236. [PMID: 34687291 DOI: 10.1093/jas/skab308] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/20/2021] [Indexed: 01/03/2023] Open
Abstract
This study was conducted to evaluate the effects of feeding fermented wheat bran (FWB) and yeast culture (YC) on growth performance, immune levels, and intestinal microflora in growing-finishing pigs. In total, 96 crossbred pigs were randomly distributed into four treatments with four replicates pens and six pigs per pen. This study was performed using a 2 × 2 factor design: 1) CON (basal diet), 2) FWB (basal diet + 5% FWB), 3) YC (basal diet + 2% YC), and 4) FWB + YC (basal diet + 5% FWB + 2% YC). Dietary FWB supplementation significantly increased the average daily gain and significantly decreased the feed gain ratio of growing-finishing pigs (P < 0.05). Supplementation of FWB and YC improved the immune capacity and reduced the inflammation level of growing-finishing pigs (P < 0.05). In addition, pigs fed FWB, YC, and FWB + YC diets showed better intestinal development and morphology compared with those CON pigs. The relative abundance of Streptococcus in the FWB group was significantly lower than that in the CON group (P < 0.05), and the relative abundance of probiotics (unclassified_f_Lachnospiraceae, Turicibacter) increased significantly (P < 0.05). Furthermore, the relative abundance of probiotics (Lactobacillus, norank_f_Muribaculaceae) in the YC group was significantly increased compared with the CON group (P < 0.05). The results of this study observed positive effects of FWB and YC on growing-finishing pigs, which provides insights into the application of biological feed in swine industry.
Collapse
Affiliation(s)
- Wei He
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Yanan Gao
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Zhiqiang Guo
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Zheng Yang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiaoxu Wang
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Honggui Liu
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Haoyang Sun
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| | - Baoming Shi
- Institute of Animal Nutrition, Northeast Agricultural University, Harbin 150030, PR China
| |
Collapse
|
19
|
Zhang W, Cheng H, Gui Y, Zhan Q, Li S, Qiao W, Tong A. Mannose Treatment: A Promising Novel Strategy to Suppress Inflammation. Front Immunol 2021; 12:756920. [PMID: 34646279 PMCID: PMC8502929 DOI: 10.3389/fimmu.2021.756920] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/09/2021] [Indexed: 02/05/2023] Open
Abstract
High glucose and fructose intake have been proven to display pro-inflammatory roles during the progression of inflammatory diseases. However, mannose has been shown to be a special type of hexose that has immune regulatory functions. In this review, we trace the discovery process of the regulatory functions of mannose and summarize some past and recent studies showing the therapeutic functions of mannose in inflammatory diseases. We conclude that treatment with mannose can suppress inflammation by inducing regulatory T cells, suppressing effector T cells and inflammatory macrophages, and increasing anti-inflammatory gut microbiome. By summarizing all the important findings, we highlight that mannose treatment is a safe and promising novel strategy to suppress inflammatory diseases, including autoimmune disease and allergic disease.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanyuan Gui
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qipeng Zhan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Si Li
- Discovery Project Unit, HitGen Inc. Tianfu International Bio-Town, Chengdu, China
| | - Wenliang Qiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Wang N, Wang S, Xu B, Liu F, Huo G, Li B. Alleviation Effects of Bifidobacterium animalis subsp. lactis XLTG11 on Dextran Sulfate Sodium-Induced Colitis in Mice. Microorganisms 2021; 9:microorganisms9102093. [PMID: 34683415 PMCID: PMC8539219 DOI: 10.3390/microorganisms9102093] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 01/11/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-related disease, which can occur through the dysfunction of the immune system caused by the imbalance of gut microbiota. Previous studies have reported the beneficial effects of Bifidobacterium on colitis, while the related mechanisms behind these effects have not been fully elucidated. The aim of our study is to investigate the alleviation effect of Bifidobacterium animalis subsp. lactis XLTG11 (B. lactis) on dextran sulfate sodium (DSS)-induced colitis and its potential mechanism. The results showed that B. lactis XLTG11 significantly decreased weight loss, disease activity index score, colon shortening, myeloperoxide activity, spleen weight, and colon tissue damage. Additionally, B. lactis XLTG11 significantly decreased the levels of pro-inflammatory cytokines and increased the level of anti-inflammatory cytokine. Meanwhile, high doses of B. lactis XLTG11 significantly up-regulated the expression of tight junction proteins and inhibited activation of Toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MYD88)/nuclear factor-κB (NF-κB) signaling pathway. Furthermore, B. lactis XLTG11 increased the gut microbiota diversity and modulated gut microbiota composition caused by DSS. Moreover, Spearman’s correlation analysis also found that several specific gut microbiota were significantly correlated with colitis-related indicators. These results demonstrated that B. lactis XLTG11 can alleviate DSS-induced colitis by inhibiting the activation of the TLR4/MYD88/NF-κB signaling pathway, regulating inflammatory cytokines, improving intestinal barrier function, and modulating the gut microbiota.
Collapse
Affiliation(s)
- Nana Wang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Song Wang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Baofeng Xu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Fei Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
- Correspondence: ; Tel.: +86-451-5519-0426
| |
Collapse
|
21
|
Huang H, Li M, Wang Y, Wu X, Shen J, Xiao Z, Zhao Y, Du F, Chen Y, Wu Z, Ji H, Zhang C, Li J, Wen Q, Kaboli PJ, Cho CH, Wang S, Wang Y, He Y, Wu X. Excessive Intake of Longan Arillus Alters gut Homeostasis and Aggravates Colitis in Mice. Front Pharmacol 2021; 12:640417. [PMID: 33841158 PMCID: PMC8033040 DOI: 10.3389/fphar.2021.640417] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/18/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Longan is the fruit of Dimocarpus longan Lour. and the longan arillus has long been used in traditional Chinese medicine possessing various health benefits. However, the excessive intake of longan is found in daily life to cause "shanghuo" syndrome. "Shanghuo" has been linked to increased disease susceptibility. The present study thus aimed to investigate the toxicological outcomes after excessive longan treatment. Methods: Longan extract at a normal dosage of 4 g/kg and two excess dosages of 8 and 16 g/kg was orally administered to normal C57BL/6J mice for two weeks or to C57BL/6J mice with DSS-induced colitis. Mouse gut microbiome were analyzed by 16S rRNA sequencing. Short chain fatty acid (SCFA) contents in colonic contents were measured by GC-MS. Colon tissue was used for histopathological observation after H and E staining, detection of protein expression by western blot, analysis of gene expression by qPCR, and detection of apoptotic cells by TUNEL assay. ELISA was used for biochemical analysis in serum. Results: In normal mice, repeated longan intake at excess doses, but not the normal dose, increased infiltration of inflammatory cells, elevated serum levels of TNF-α and IL-6 and reduced production of SCFAs. In DSS-induced colitic mice, longan intake at 4 g/kg did not promote colitis in mice, while excessive longan (8 or 16 g/kg) aggravated colitis in mice, showing increased inflammation, more serious histological abnormalities, increased gut permeability, and increased epithelia injury when compared to DSS alone. Excessive longan induced a significant reduction of microbial diversity in colitic mice, accompanied with aggravated alterations of DSS-associated bacteria including the increase of Proteobacteria phylum and genera of Bacteroides, Akkermansia, Turicibacter and Escherchia-Shigella, and the decrease of norank_f__Muribaculaceae. The changed microbial compositions were accompanied with decreased SCFAs when longan was supplemented with DSS. The aggravated colon injury by excessive intake of longan in colitic mice was tightly correlated with the altered microbial communities and decreased SCFAs production. Conclusion: Excessive longan intake disturbs gut homeostasis and aggravates colitis via promoting inflammation and altering gut microbe compositions and associated metabolism in mice. Our findings warrant rational longan arillus consumption as a dietary supplement or herbal medicine.
Collapse
Affiliation(s)
- Huimin Huang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yi Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Xiaoxiao Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Zhigui Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Huijiao Ji
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Chunyuan Zhang
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jing Li
- Department of Oncology and Hematology, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, China
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, China
| | - Parham Jabbarzadeh Kaboli
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yisheng He
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, China
| |
Collapse
|
22
|
Chen Y, Jin Y, Stanton C, Paul Ross R, Zhao J, Zhang H, Yang B, Chen W. Alleviation effects of Bifidobacterium breve on DSS-induced colitis depends on intestinal tract barrier maintenance and gut microbiota modulation. Eur J Nutr 2021; 60:369-387. [PMID: 32350653 DOI: 10.1007/s00394-020-02252-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 04/14/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE The study aimed to investigate the discrepancy and potential mechanisms of different CLA-producing B. breve on dextran sulphate sodium (DSS)-induced colitis. METHODS Colitis was induced in C57BL/6 J mice using DSS. Disease activity index (DAI), histopathological changes, epithelial barrier integrity and epithelial apoptosis were determined. Gut microbiota were gauged to evaluate the systemic effects of CLA-producing B. breve. RESULTS Oral administration of different B. breve showed different effects, in which B. breve M1 and B. breve M2 alleviated the inflammation induced by DSS as well as significantly increased the concentration of mucin2 (MUC2) and goblet cells, but neither B. breve M3 nor B. breve M4 had those protective effects. Meanwhile, B. breve M1 and B. breve M2 treatments significantly up-regulated the tight junction (TJ) proteins and ameliorated the epithelial apoptosis lead by DSS challenge. Moreover, inflammatory cytokines (TNF-α, IL-6) were modulated by B. breve M1 and B. breve M2, neither B. breve M3 nor B. breve M4. Furthermore, B. breve M1 and B. breve M2 reduced the abundance of Bacteroides and increased the abundance of Odoribacter, then rebalanced the damaged gut microbiota. Colonic CLA concentrations in mice fed with B. breve M1, B. breve M2, B. breve M3 and B. breve M4 decreased successively, which showed significant positive correlation with the effectiveness of relieving colitis. CONCLUSIONS Bifidobacterium breve M1 and B. breve M2 alleviated DSS-induced colitis by producing CLA, inhibiting the inflammatory cytokines, maintaining of the intestinal epithelial barrier and regulating the gut microbiota.
Collapse
Affiliation(s)
- Yang Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu, China
| | - Yan Jin
- Department of Gastroenterology, The Affiliated Wuxi Second People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China.
| | - Catherine Stanton
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - R Paul Ross
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Translational Medicine Research Center, Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, Jiangsu, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu, China.
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China.
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
- Beijing Innovation Center of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, China
| |
Collapse
|
23
|
Akobeng AK, Singh P, Kumar M, Al Khodor S. Role of the gut microbiota in the pathogenesis of coeliac disease and potential therapeutic implications. Eur J Nutr 2020; 59:3369-3390. [PMID: 32651763 PMCID: PMC7669811 DOI: 10.1007/s00394-020-02324-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/01/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE Although genetic predisposition and exposure to dietary gluten are considered necessary triggers for the development of coeliac disease, alterations in the gut microbial composition may also contribute towards the pathogenesis of coeliac disease. This review aims to provide an overview of the available data on the potential mechanisms through which the gut microbiota plays a role in the causation of coeliac disease and to discuss the potential therapeutic strategies that could diminish the consequences of microbial dysbiosis. METHOD A search of the literature was performed using the PubMed, Embase, and JSTOR databases; relevant articles were included. RESULTS Recent studies in patients with coeliac disease have reported an increase in the relative amounts of gram negative bacterial genera such as Bacteroides, Prevotella, and Escherichia, and reduced amounts of protective anti-inflammatory bacteria such as Bifidobacteria and Lactobacilli. Dysbiotic microbiota may lead to a dysregulated immune response that may contribute to the pathogenesis of coeliac disease. In infancy, antibiotic use and certain infant feeding practices may lead to alterations in the developing gut microbiota to influence the immune maturation process and predispose to coeliac disease. CONCLUSION The induction of the intestinal immune system and gluten intolerance may be influenced by the relative abundance of certain microbiota. Factors such as infant feeding practices, diet, antibiotics, and infections, may be involved in the development of coeliac disease due to their influence on gut microbial composition. The efficacy of potential modulators of the gut microbiota such as probiotics, prebiotics, and fecal microbial transplant as adjunctive treatments to gluten-free diet in coeliac disease is unproven and requires further investigation.
Collapse
Affiliation(s)
- Anthony K Akobeng
- Division of Gastroenterology, Hepatology, and Nutrition, Sidra Medicine, Doha, Qatar
- Weill Cornell Medicine, Cornell University, Doha, Qatar
| | - Parul Singh
- Research Department, Sidra Medicine, Doha, Qatar
| | - Manoj Kumar
- Research Department, Sidra Medicine, Doha, Qatar
| | | |
Collapse
|
24
|
Gunasekera DC, Ma J, Vacharathit V, Shah P, Ramakrishnan A, Uprety P, Shen Z, Sheh A, Brayton CF, Whary MT, Fox JG, Bream JH. The development of colitis in Il10 -/- mice is dependent on IL-22. Mucosal Immunol 2020; 13:493-506. [PMID: 31932715 PMCID: PMC7566780 DOI: 10.1038/s41385-019-0252-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/01/2019] [Accepted: 11/27/2019] [Indexed: 02/08/2023]
Abstract
Mice deficient in the IL-10 pathway are the most widely used models of intestinal immunopathology. IL-17A is strongly implicated in gut disease in mice and humans, but conflicting evidence has drawn IL-17's role in the gut into question. IL-22 regulates antimicrobial and repair activities of intestinal epithelial cells (IECs) and is closely associated with IL-17A responses but it's role in chronic disease is uncertain. We report that IL-22, like IL-17A, is aberrantly expressed in colitic Il10-/- mice. While IL-22+ Th17 cells were elevated in the colon, IL-22-producing ILC3s were highly enriched in the small intestines of Il10-/- mice. Remarkably, Il10-/-Il22-/- mice did not develop colitis despite retaining high levels of Th17 cells and remaining colonized with colitogenic Helicobacter spp. Accordant with IL-22-induced IEC proliferation, the epithelia hyperplasia observed in Il10-/- animals was reversed in Il10-/-Il22-/- mice. Also, the high levels of antimicrobial IL-22-target genes, including Reg3g, were normalized in Il10-/-Il22-/- mice. Consistent with a heightened antimicrobial environment, Il10-/- mice had reduced diversity of the fecal microbiome that was reestablished in Il10-/-Il22-/- animals. These data suggest that spontaneous colitis in Il10-/- mice is driven by IL-22 and implicates an underappreciated IL-10/IL-22 axis in regulating intestinal homeostasis.
Collapse
Affiliation(s)
- Dilini C Gunasekera
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jinxia Ma
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Vimvara Vacharathit
- Graduate Program in Immunology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Palak Shah
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Amritha Ramakrishnan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Priyanka Uprety
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Zeli Shen
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alexander Sheh
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Cory F Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mark T Whary
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jay H Bream
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Graduate Program in Immunology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
25
|
Chen Y, Yang B, Ross RP, Jin Y, Stanton C, Zhao J, Zhang H, Chen W. Orally Administered CLA Ameliorates DSS-Induced Colitis in Mice via Intestinal Barrier Improvement, Oxidative Stress Reduction, and Inflammatory Cytokine and Gut Microbiota Modulation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:13282-13298. [PMID: 31690068 DOI: 10.1021/acs.jafc.9b05744] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Dietary supplementation with conjugated linoleic acid (CLA) has been reported to alleviate the effect of colitis in mice, but the mechanisms involved need further exploration. The study aimed to investigate how orally administered CLA alleviates dextran sulfate sodium (DSS)-induced colitis in mice. CLA was administered in five different doses: 40, 20, 10, 5, and 2.5 mg/day. Doses of CLA at 10 mg/day and higher alleviated colitis symptoms and reduced inflammation induced by DSS, in which 40, 20, and 10 mg/day CLA significantly increased the concentration of mucin2 and goblet cells, but neither 5 mg/day CLA nor 2.5 mg/day CLA had any effects. Meanwhile, 40 and 20 mg/day CLA treatments significantly upregulated the concentration of tight junction proteins (ZO-1, occludin, and claudin-3) and ameliorated epithelial apoptosis caused by DSS. Moreover, oxidative-stress-related enzymes (superoxide dismutase, glutathione peroxidase, and catalase) and inflammatory cytokines [tumor necrosis factor-α, interleukin (IL)-10, and IL-6] were modulated by 40 and 20 mg/day CLA. Furthermore, 40 mg/day CLA rebalanced the gut microbiota damaged by DSS, including reducing Bacteroides and increasing Bifidobacterium and Odoribacter. In conclusion, CLA supplementation alleviated DSS-induced colitis in a dose-dependent manner by modulating inflammatory cytokines and oxidation stress, maintaining the mucosal barrier, and reverting microbiota changes.
Collapse
Affiliation(s)
| | | | - R Paul Ross
- APC Microbiome Ireland , University College Cork , Cork T12 K8AF , Ireland
| | - Yan Jin
- Department of Gastroenterology , The Affiliated Wuxi Second People's Hospital of Nanjing Medical University , Wuxi 214023 , China
| | - Catherine Stanton
- Teagasc Food Research Centre , Moorepark, Fermoy, Cork P61 C996 , Ireland
- APC Microbiome Ireland , University College Cork , Cork T12 K8AF , Ireland
| | | | - Hao Zhang
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch , Wuxi 214122 , China
| | - Wei Chen
- Beijing Innovation Center of Food Nutrition and Human Health , Beijing Technology and Business University (BTBU) , Beijing 100048 , China
| |
Collapse
|
26
|
Abstract
There is now a wealth of evidence showing that communication between microbiota and the host is critical to sustain the vital functions of the healthy host, and disruptions of this homeostatic coexistence are known to be associated with a range of diseases including obesity and type 2 diabetes. Microbiota-derived metabolites act both as nutrients and as messenger molecules and can signal to distant organs in the body to shape host pathophysiology. In this review, we provide a new perspective on succinate as a gut microbiota-derived metabolite with a key role governing intestinal homeostasis and energy metabolism. Thus, succinate is not merely a major intermediary of the TCA traditionally considered as an extracellular danger signal in the host, but also a by-product of some bacteria and a primary cross-feeding metabolite between gut resident microbes. In addition to maintain a healthy microbiome, specific functions of microbiota-derived succinate in peripheral tissues regulating host nutrient metabolism should not be rule out. Indeed, recent research point to some probiotic interventions directed to modulate succinate levels in the intestinal lumen, as a new microbiota-based therapies to treat obesity and related co-morbidities. While further research is essential, a large body of evidence point to succinate as a new strategic mediator in the microbiota-host cross-talk, which might provide the basis for new therapeutically approaches in a near future.
Collapse
Affiliation(s)
- Sonia Fernández-Veledo
- Departament of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII-Institut d'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch, 4, 43007, Tarragona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain.
| | - Joan Vendrell
- Departament of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII-Institut d'Investigació Sanitària Pere Virgili (IISPV), c/ Dr. Mallafré Guasch, 4, 43007, Tarragona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM)-Instituto de Salud Carlos III, Madrid, Spain
- Rovira i Virgili University, Tarragona, Spain
| |
Collapse
|
27
|
Kusakabe S, Fukushima K, Maeda T, Motooka D, Nakamura S, Fujita J, Yokota T, Shibayama H, Oritani K, Kanakura Y. Pre- and post-serial metagenomic analysis of gut microbiota as a prognostic factor in patients undergoing haematopoietic stem cell transplantation. Br J Haematol 2019; 188:438-449. [PMID: 31566729 DOI: 10.1111/bjh.16205] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/17/2019] [Indexed: 12/21/2022]
Abstract
The human gut harbours diverse microorganisms, and gut dysbiosis has recently attracted attention because of its possible involvement in various diseases. In particular, the lack of diversity in the gut microbiota has been associated with complications of haematopoietic stem cell transplantation (HSCT), such as infections, acute graft-versus-host disease and relapse of primary disease, which lead to a poor prognosis. However, few studies have serially examined the composition of the intestinal microbiota after HSCT. In this study, we demonstrated, using next-generation sequencing of the bacterial 16S ribosomal RNA gene, combined with uniFrac distance analysis, that the intestinal microbiota of patients undergoing allogeneic HSCT substantially differed from that of healthy controls and recipients of autologous transplants. Faecal samples were obtained daily throughout the clinical course, before and after transplantation. Notably, the proportions of Bifidobacterium and genera categorized as butyrate-producing bacteria were significantly lower in patients with allogeneic HSCT than in healthy controls. Furthermore, among allogeneic transplant recipients, a subgroup with a preserved microbiota composition showed a benign course, whereas patients with a skewed microbiota showed a high frequency of complications and mortality after transplantation. Thus, we conclude that the stability of intestinal microbiota is critically involved in outcomes of HSCT.
Collapse
Affiliation(s)
- Shinsuke Kusakabe
- Department of Haematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kentaro Fukushima
- Department of Haematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tetsuo Maeda
- Department of Haematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Jiro Fujita
- Department of Haematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takafumi Yokota
- Department of Haematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hirohiko Shibayama
- Department of Haematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kenji Oritani
- Department of Haematology, School of Medicine, International University of Health and Welfare, Narita, Chiba, Japan
| | - Yuzuru Kanakura
- Department of Haematology and Oncology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
28
|
Khan I, Ullah N, Zha L, Bai Y, Khan A, Zhao T, Che T, Zhang C. Alteration of Gut Microbiota in Inflammatory Bowel Disease (IBD): Cause or Consequence? IBD Treatment Targeting the Gut Microbiome. Pathogens 2019; 8:pathogens8030126. [PMID: 31412603 PMCID: PMC6789542 DOI: 10.3390/pathogens8030126] [Citation(s) in RCA: 417] [Impact Index Per Article: 83.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/03/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic complex inflammatory gut pathological condition, examples of which include Crohn’s disease (CD) and ulcerative colitis (UC), which is associated with significant morbidity. Although the etiology of IBD is unknown, gut microbiota alteration (dysbiosis) is considered a novel factor involved in the pathogenesis of IBD. The gut microbiota acts as a metabolic organ and contributes to human health by performing various physiological functions; deviation in the gut flora composition is involved in various disease pathologies, including IBD. This review aims to summarize the current knowledge of gut microbiota alteration in IBD and how this contributes to intestinal inflammation, as well as explore the potential role of gut microbiota-based treatment approaches for the prevention and treatment of IBD. The current literature has clearly demonstrated a perturbation of the gut microbiota in IBD patients and mice colitis models, but a clear causal link of cause and effect has not yet been presented. In addition, gut microbiota-based therapeutic approaches have also shown good evidence of their effects in the amelioration of colitis in animal models (mice) and IBD patients, which indicates that gut flora might be a new promising therapeutic target for the treatment of IBD. However, insufficient data and confusing results from previous studies have led to a failure to define a core microbiome associated with IBD and the hidden mechanism of pathogenesis, which suggests that well-designed randomized control trials and mouse models are required for further research. In addition, a better understanding of this ecosystem will also determine the role of prebiotics and probiotics as therapeutic agents in the management of IBD.
Collapse
Affiliation(s)
- Israr Khan
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Naeem Ullah
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Lajia Zha
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Yanrui Bai
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Ashiq Khan
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Probiotics and Biological Feed Research Center, Lanzhou University, Lanzhou 730000, China
| | - Tang Zhao
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Tuanjie Che
- Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou 730000, China
| | - Chunjiang Zhang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China.
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China.
- Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou 730000, China.
| |
Collapse
|
29
|
Vitellio P, Celano G, Bonfrate L, Gobbetti M, Portincasa P, De Angelis M. Effects of Bifidobacterium longum and Lactobacillus rhamnosus on Gut Microbiota in Patients with Lactose Intolerance and Persisting Functional Gastrointestinal Symptoms: A Randomised, Double-Blind, Cross-Over Study. Nutrients 2019; 11:E886. [PMID: 31010241 PMCID: PMC6520754 DOI: 10.3390/nu11040886] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/13/2019] [Accepted: 04/16/2019] [Indexed: 02/07/2023] Open
Abstract
Functional gastrointestinal symptoms are frequent, and may be driven by several pathogenic mechanisms. Symptoms may persist in lactose intolerant (LI) patients (i.e., subjects with intestinal lactase deficiency, lactose malabsorption producing symptoms), after a lactose-free diet. Our hypothesis was that probiotic and vitamin B6 treatment may be useful to alleviate symptoms in LI patients through a positive modulation of gut microbial composition and relative metabolism. We aimed to test the efficacy of a novel formulation of Bifidobacterium longum BB536 and Lactobacillus rhamnosus HN001 plus vitamin B6 (ZR) in 23 LI subjects with persistent symptoms during a lactose-free diet. Symptoms, microbiome, and metabolome were measured at baseline and after 30 days in a crossover, randomized, double-blind study of ZR versus placebo (PL). Compared with PL, the administration of probiotics and vitamin B6 significantly decreased bloating (p = 0.028) and ameliorated constipation (p = 0.045). Fecal microbiome differed between ZR and PL. ZR drove the enrichment of several genera involved in lactose digestion including Bifidobacerium. Moreover, the relative abundance of acetic acid, 2-methyl-propanoic acid, nonenal, and indolizine 3-methyl increased, while phenol decreased. Our findings highlight the importance of selected probiotics and vitamin B6 to alleviate symptoms and gut dysbiosis in lactose intolerant patients with persistent functional gastrointestinal symptoms.
Collapse
Affiliation(s)
- Paola Vitellio
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, via Amendola 165/a, 70126 Bari, Italy.
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70121 Bari, Italy.
| | - Giuseppe Celano
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, via Amendola 165/a, 70126 Bari, Italy.
| | - Leonilde Bonfrate
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70121 Bari, Italy.
| | - Marco Gobbetti
- Faculty of Science and Technology, Free University of Bolzano, piazza Università, 5, 39100 Bolzano, Italy.
| | - Piero Portincasa
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70121 Bari, Italy.
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, via Amendola 165/a, 70126 Bari, Italy.
| |
Collapse
|
30
|
The Role of Succinate in the Regulation of Intestinal Inflammation. Nutrients 2018; 11:nu11010025. [PMID: 30583500 PMCID: PMC6356305 DOI: 10.3390/nu11010025] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/14/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022] Open
Abstract
Succinate is a metabolic intermediate of the tricarboxylic acid (TCA) cycle within host cells. Succinate is also produced in large amounts during bacterial fermentation of dietary fiber. Elevated succinate levels within the gut lumen have been reported in association with microbiome disturbances (dysbiosis), as well as in patients with inflammatory bowel disease (IBD) and animal models of intestinal inflammation. Recent studies indicate that succinate can activate immune cells via its specific surface receptor, succinate receptor 1(SUCNR1), and enhance inflammation. However, the role of succinate in inflammatory processes within the gut mucosal immune system is unclear. This review includes current literature on the association of succinate with intestinal inflammation and the potential role of succinate–SUCNR1 signaling in gut immune functions.
Collapse
|
31
|
Yu LCH. Microbiota dysbiosis and barrier dysfunction in inflammatory bowel disease and colorectal cancers: exploring a common ground hypothesis. J Biomed Sci 2018; 25:79. [PMID: 30413188 PMCID: PMC6234774 DOI: 10.1186/s12929-018-0483-8] [Citation(s) in RCA: 244] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a multifactorial disease which arises as a result of the interaction of genetic, environmental, barrier and microbial factors leading to chronic inflammation in the intestine. Patients with IBD had a higher risk of developing colorectal carcinoma (CRC), of which the subset was classified as colitis-associated cancers. Genetic polymorphism of innate immune receptors had long been considered a major risk factor for IBD, and the mutations were also recently observed in CRC. Altered microbial composition (termed microbiota dybiosis) and dysfunctional gut barrier manifested by epithelial hyperpermeability and high amount of mucosa-associated bacteria were observed in IBD and CRC patients. The findings suggested that aberrant immune responses to penetrating commensal microbes may play key roles in fueling disease progression. Accumulative evidence demonstrated that mucosa-associated bacteria harbored colitogenic and protumoral properties in experimental models, supporting an active role of bacteria as pathobionts (commensal-derived opportunistic pathogens). Nevertheless, the host factors involved in bacterial dysbiosis and conversion mechanisms from lumen-dwelling commensals to mucosal pathobionts remain unclear. Based on the observation of gut leakiness in patients and the evidence of epithelial hyperpermeability prior to the onset of mucosal histopathology in colitic animals, it was postulated that the epithelial barrier dysfunction associated with mucosal enrichment of specific bacterial strains may predispose the shift to disease-associated microbiota. The speculation of leaky gut as an initiating factor for microbiota dysbiosis that eventually led to pathological consequences was proposed as the "common ground hypothesis", which will be highlighted in this review. Overall, the understanding of the core interplay between gut microbiota and epithelial barriers at early subclinical phases will shed light to novel therapeutic strategies to manage chronic inflammatory disorders and colitis-associated cancers.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Suite 1020, #1 Jen-Ai Rd. Sec. 1, Taipei, 100, Taiwan, Republic of China.
| |
Collapse
|
32
|
Matsuoka K, Uemura Y, Kanai T, Kunisaki R, Suzuki Y, Yokoyama K, Yoshimura N, Hibi T. Efficacy of Bifidobacterium breve Fermented Milk in Maintaining Remission of Ulcerative Colitis. Dig Dis Sci 2018; 63:1910-1919. [PMID: 29450747 PMCID: PMC6015104 DOI: 10.1007/s10620-018-4946-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 01/23/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Fermented milk products containing Bifidobacterium breve strain Yakult (BFM) may improve clinical status in ulcerative colitis (UC) patients. AIMS To assess efficacy of BFM in maintaining remission in Japanese patients with quiescent UC. METHODS This double-blind study (B-FLORA) enrolled 195 patients with quiescent UC, randomized to receive one pack of BFM fermented milk per day [Bifidobacterium breve strain Yakult (10 billion bacteria) and Lactobacillus acidophilus (1 billion bacteria)] (n = 98) or matching placebo (n = 97) for 48 weeks. The primary efficacy endpoint was relapse-free survival (relapse: rectal bleeding score ≥ 2 on Sutherland disease activity index scale for 3 consecutive days and/or initiation of remission induction therapy for worsening of UC). RESULTS An interim analysis was conducted after inclusion and follow-up of one-third of patients for the first phase of the study (n = 195). Relapse-free survival was not significantly different between the BFM and placebo groups (P = 0.643; hazard ratio 1.16; 95% CI 0.63-2.14, log-rank test), nor was the incidence of relapse. Therefore, the study was discontinued for lack of efficacy. An exploratory analysis of fecal samples from a subgroup of patients revealed no effects of either study beverage on intestinal microbiota, but there was a significant decrease in Bifidobacterium species before relapse, regardless of treatment group. Three mild adverse events occurred for which a causal relationship with the study beverage could not be ruled out (placebo: abdominal bloating and stress in one patient; BFM: body odor in one patient). CONCLUSIONS BFM had no effect on time to relapse in UC patients compared with placebo. STUDY REGISTRATION UMIN000007593.
Collapse
Affiliation(s)
- Katsuyoshi Matsuoka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan ,Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519 Japan
| | - Yukari Uemura
- Biostatistics Division, Central Coordinating Unit, Clinical Research Support Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033 Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Reiko Kunisaki
- Inflammatory Bowel Disease Center, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama, 232-0024 Japan
| | - Yasuo Suzuki
- Internal Medicine, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura, Chiba 285-8741 Japan
| | - Kaoru Yokoyama
- Department of Gastroenterology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0374 Japan
| | - Naoki Yoshimura
- Department of Internal Medicine, Division of IBD, Tokyo Yamate Medical Center, 3-22-1 Hyakunincho, Shinjuku-ku, Tokyo, 169-0073 Japan
| | - Toshifumi Hibi
- Center for Advanced Inflammatory Bowel Disease Research and Treatment, Kitasato University Kitasato Institute Hospital, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8642 Japan
| |
Collapse
|
33
|
Zhang SL, Wang SN, Miao CY. Influence of Microbiota on Intestinal Immune System in Ulcerative Colitis and Its Intervention. Front Immunol 2017; 8:1674. [PMID: 29234327 PMCID: PMC5712343 DOI: 10.3389/fimmu.2017.01674] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 11/14/2017] [Indexed: 01/07/2023] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease (IBD) with chronic and recurrent characteristics caused by multiple reasons. Although the pathogenic factors have not been clarified yet, recent studies have demonstrated that intestinal microbiota plays a major role in UC, especially in the immune system. This review focuses on the description of several major microbiota communities that affect UC and their interactions with the host. In this review, eight kinds of microbiota that are highly related to IBD, including Faecalibacterium prausnitzii, Clostridium clusters IV and XIVa, Bacteroides, Roseburia species, Eubacterium rectale, Escherichia coli, Fusobacterium, and Candida albicans are demonstrated on the changes in amount and roles in the onset and progression of IBD. In addition, potential therapeutic targets for UC involved in the regulation of microbiota, including NLRPs, vitamin D receptor as well as secreted proteins, are discussed in this review.
Collapse
Affiliation(s)
- Sai-Long Zhang
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Shu-Na Wang
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| |
Collapse
|
34
|
Effects of monobutyrin and tributyrin on liver lipid profile, caecal microbiota composition and SCFA in high-fat diet-fed rats. J Nutr Sci 2017; 6:e51. [PMID: 29152255 PMCID: PMC5672331 DOI: 10.1017/jns.2017.54] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/11/2017] [Accepted: 08/02/2017] [Indexed: 01/05/2023] Open
Abstract
Butyric acid has been shown to have suppressive effects on inflammation and diseases related to the intestinal tract. The aim of the present study was to investigate whether supplementation of two glycerol esters, monobutyrin (MB) and tributyrin (TB), would reach the hindgut of rats, thus having an effect on the caecal profile of SCFA, microbiota composition and some risk markers associated with chronic inflammation. For this purpose, rats were fed high-fat diets after adding MB (1 and 5 g/kg) and TB (5 g/kg) to a diet without any supplementation (high-fat control; HFC). A low-fat (LF) diet was also included. In the liver, total cholesterol concentrations, LDL-cholesterol concentrations, LDL:HDL ratio, and succinic acid concentrations were reduced in rats given the MB and TB (5 g/kg) diets, compared with the group fed the HFC diet. These effects were more pronounced in MB than TB groups as also expressed by down-regulation of the gene Cyp8b1. The composition of the caecal microbiota in rats fed MB and TB was separated from the group fed the HFC diet, and also the LF diet, as evidenced by the absence of the phylum TM7 and reduced abundance of the genera Dorea (similar to LF-fed rats) and rc4-4. Notably, the caecal abundance of Mucispirillum was markedly increased in the MB group compared with the HFC group. The results suggest that dietary supplementation of MB and TB can be used to counteract disturbances associated with a HFC diet, by altering the gut microbiota, and decreasing liver lipids and succinic acid concentrations.
Collapse
|
35
|
Tao JH, Duan JA, Jiang S, Feng NN, Qiu WQ, Ling Y. Polysaccharides from Chrysanthemum morifolium Ramat ameliorate colitis rats by modulating the intestinal microbiota community. Oncotarget 2017; 8:80790-80803. [PMID: 29113344 PMCID: PMC5655239 DOI: 10.18632/oncotarget.20477] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
The gut microflora dysbiosis has been closely related with the inflammatory bowel disease (IBD). In this study, the effect of polysaccharides from Chrysanthemum morifolium Ramat on the gut microbiota was evaluated by ulcerative colitis (UC) rat model. Physiological and pathological analyses suggested that Chrysanthemum polysaccharides possessed notably protective effects on UC in vivo. Based on the Illumina MiSeq platform, 16S rRNA sequencing of the rat colonic contents indicated that the intestinal flora structure remarkably changed in the model rats and the tendency was alleviated to a certain degree by treatment with different dosages of Chrysanthemum polysaccharides. In normal groups, there were more Firmicutes than Bacteroidetes, but this change lost at the pathological state. Following Chrysanthemum polysaccharides, rising Firmicutes/Bacteroidetes ratio was validated. Besides the microbial diversity and the community richness of the UC rats were improved by Chrysanthemum polysaccharides, the composition of intestinal microflora in the model group were also restored after oral administration of Chrysanthemum polysaccharides. The abundance of opportunistic pathogens was decreased (Escherichia, Enterococcus and Prevotella), while the levels of protective bacteria such as Butyricicoccus and Clostridium (butyrate-producing bacteria), Lactobacillus and Bifidobacterium (probiotics), Lachnospiraceae and Rikenellaceae elevated in various degrees. Correlation analysis between intestinal flora and biochemical factors suggested that the relative abundance of protective bacteria was positively correlated with the levels of anti-inflammatory cytokines such as IL-4, IL-10 and IL-11, while aggressive bacteria were positively correlated with proinflammatory cytokine such as IL-23、IL-6、 IF-17、TNF-α、IL-1β and IFN-γ. The above results showed that the intestinal flora were closely related to the secretion and expression of cytokines in the body, and they interacted with each other to regulate immune function. Thus, Chrysanthemum polysaccharides could ameliorate ulcerative colitis by fostering beneficial intestinal flora growth, modulating the balance of intestinal microecology and restoring the immune system.
Collapse
Affiliation(s)
- Jin-Hua Tao
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Nan-Nan Feng
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Wen-Qian Qiu
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Yong Ling
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| |
Collapse
|
36
|
The gut bacterium and pathobiont Bacteroides vulgatus activates NF-κB in a human gut epithelial cell line in a strain and growth phase dependent manner. Anaerobe 2017; 47:209-217. [DOI: 10.1016/j.anaerobe.2017.06.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 05/17/2017] [Accepted: 06/01/2017] [Indexed: 01/06/2023]
|
37
|
Shigemori S, Shimosato T. Applications of Genetically Modified Immunobiotics with High Immunoregulatory Capacity for Treatment of Inflammatory Bowel Diseases. Front Immunol 2017; 8:22. [PMID: 28179904 PMCID: PMC5263139 DOI: 10.3389/fimmu.2017.00022] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
Inflammatory bowel diseases (IBDs), including ulcerative colitis and Crohn’s disease, are chronic inflammatory diseases characterized by dysregulated immune responses of the gastrointestinal tract. In recent years, the incidence of IBDs has increased in developed nations, but their prophylaxis/treatment is not yet established. Site-directed delivery of molecules showing anti-inflammatory properties using genetically modified (gm)-probiotics shows promise as a new strategy for the prevention and treatment of IBD. Advantages of gm-probiotics include (1) the ability to use bacteria as a delivery vehicle, enabling safe and long-term use by humans, (2) decreased risks of side effects, and (3) reduced costs. The intestinal delivery of anti-inflammatory proteins such as cytokines and enzymes using Lactococcus lactis has been shown to regulate host intestinal homeostasis depending on the delivered protein-specific machinery. Additionally, clinical experience using interleukin 10-secreting Lc. lactis has been shown to be safe and to facilitate biological containment in IBD therapy. On the other hand, some preclinical studies have demonstrated that gm-strains of immunobiotics (probiotic strains able to beneficially regulate the mucosal immunity) provide beneficial effects on intestinal inflammation as a result of the synergy between the immunoregulatory effects of the bacterium itself and the anti-inflammatory effects of the delivered recombinant proteins. In this review, we discuss the rapid progression in the development of strategies for the prophylaxis and treatment of IBD using gm-probiotics that exhibit immune regulation effects (gm-immunobiotics). In particular, we discuss the type of strains used as delivery agents.
Collapse
Affiliation(s)
- Suguru Shigemori
- Department of Bioscience and Food Production Science, Interdisciplinary Graduate School of Science and Technology, Shinshu University, Nagano, Japan; Japan Society for the Promotion of Science, Tokyo, Japan
| | - Takeshi Shimosato
- Department of Interdisciplinary Genome Sciences and Cell Metabolism, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan; Supramolecular Complexes Unit, Research Center for Fungal and Microbial Dynamism, Shinshu University, Nagano, Japan
| |
Collapse
|
38
|
Li D, Hu N, Sui Y, Ding D, Li K, Li G, Wang Y. Influence of bicarbonate on the abundance of microbial communities capable of reducing U(vi) in groundwater. RSC Adv 2017. [DOI: 10.1039/c7ra09795f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
7 experiments amended with 0, 5, 10, 15, 20, 25 and 30 mM initial concentrations of bicarbonate were conducted to investigate the influence of different concentrations of bicarbonate on the abundance of microbial communities capable of reducing U(vi) in groundwater.
Collapse
Affiliation(s)
- Dianxin Li
- Key Discipline Laboratory for National Defence for Biotechnology in Uranium Mining and Hydrometallurgy
- University of South China
- 421001 Hengyang
- China
| | - Nan Hu
- Key Discipline Laboratory for National Defence for Biotechnology in Uranium Mining and Hydrometallurgy
- University of South China
- 421001 Hengyang
- China
| | - Yang Sui
- Key Discipline Laboratory for National Defence for Biotechnology in Uranium Mining and Hydrometallurgy
- University of South China
- 421001 Hengyang
- China
| | - Dexin Ding
- Key Discipline Laboratory for National Defence for Biotechnology in Uranium Mining and Hydrometallurgy
- University of South China
- 421001 Hengyang
- China
| | - Ke Li
- Key Discipline Laboratory for National Defence for Biotechnology in Uranium Mining and Hydrometallurgy
- University of South China
- 421001 Hengyang
- China
| | - Guangyue Li
- Key Discipline Laboratory for National Defence for Biotechnology in Uranium Mining and Hydrometallurgy
- University of South China
- 421001 Hengyang
- China
| | - Yongdong Wang
- Key Discipline Laboratory for National Defence for Biotechnology in Uranium Mining and Hydrometallurgy
- University of South China
- 421001 Hengyang
- China
| |
Collapse
|
39
|
Machiels K, Sabino J, Vandermosten L, Joossens M, Arijs I, de Bruyn M, Eeckhaut V, Van Assche G, Ferrante M, Verhaegen J, Van Steen K, Van Immerseel F, Huys G, Verbeke K, Wolthuis A, de Buck Van Overstraeten A, D'Hoore A, Rutgeerts P, Vermeire S. Specific members of the predominant gut microbiota predict pouchitis following colectomy and IPAA in UC. Gut 2017; 66:79-88. [PMID: 26423113 DOI: 10.1136/gutjnl-2015-309398] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Pouchitis is the most common complication after colectomy with ileal pouch-anal anastomosis (IPAA) for UC and the risk is the highest within the 1st year after surgery. The pathogenesis is not completely understood but clinical response to antibiotics suggests a role for gut microbiota. We hypothesised that the risk for pouchitis can be predicted based on the faecal microbial composition before colectomy. DESIGN Faecal samples from 21 patients with UC undergoing IPAA were prospectively collected before colectomy and at predefined clinical visits at 1 month, 3 months, 6 months and 12 months after IPAA. The predominant microbiota was analysed using community profiling with denaturing gradient gel electrophoresis followed by quantitative real-time PCR validation. RESULTS Cluster analysis before colectomy distinguished patients with pouchitis from those with normal pouch during the 1st year of follow-up. In patients developing pouchitis, an increase of Ruminococcus gnavus (p<0.001), Bacteroides vulgatus (p=0.043), Clostridium perfringens (p=0.011) and a reduction of two Lachnospiraceae genera (Blautia (p=0.04), Roseburia (p=0.008)) was observed. A score combining these five bacterial risk factors was calculated and presence of at least two risk factors showed a sensitivity and specificity of 100% and 63.6%, respectively. CONCLUSIONS Presence of R. gnavus, B. vulgatus and C. perfringens and absence of Blautia and Roseburia in faecal samples of patients with UC before surgery is associated with a higher risk of pouchitis after IPAA. Our findings suggest new predictive and therapeutic strategies in patients undergoing colectomy with IPAA.
Collapse
Affiliation(s)
- Kathleen Machiels
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - João Sabino
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Leen Vandermosten
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Marie Joossens
- Department Microbiology and Immunology, KU Leuven, Leuven, Belgium.,Center for the Biology of Disease, VIB, Leuven, Belgium.,Faculty of Sciences and Bioengineering Sciences, Microbiology Unit, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ingrid Arijs
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Magali de Bruyn
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Venessa Eeckhaut
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Merelbeke, Belgium
| | - Gert Van Assche
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Marc Ferrante
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Jan Verhaegen
- Department of Microbiology and Immunology, University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Kristel Van Steen
- Department of Electrical Engineering and Computer Science, Montefiore Institute, Liège, Belgium
| | - Filip Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Merelbeke, Belgium
| | - Geert Huys
- Laboratory of Microbiology & BCCM/LMG Bacteria Collection, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Kristin Verbeke
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Albert Wolthuis
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | | | - Andre D'Hoore
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Paul Rutgeerts
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Translational Research Center for Gastrointestinal Disorders (TARGID), University Hospital Leuven, KU Leuven, Leuven, Belgium
| |
Collapse
|
40
|
De Angelis M, Vannini L, Di Cagno R, Cavallo N, Minervini F, Francavilla R, Ercolini D, Gobbetti M. Salivary and fecal microbiota and metabolome of celiac children under gluten-free diet. Int J Food Microbiol 2016; 239:125-132. [PMID: 27452636 DOI: 10.1016/j.ijfoodmicro.2016.07.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 07/10/2016] [Accepted: 07/18/2016] [Indexed: 02/06/2023]
|
41
|
The Gut Microbiome of Pediatric Crohn's Disease Patients Differs from Healthy Controls in Genes That Can Influence the Balance Between a Healthy and Dysregulated Immune Response. Inflamm Bowel Dis 2016; 22:2607-2618. [PMID: 27760077 DOI: 10.1097/mib.0000000000000949] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Exclusive enteral nutrition (EEN) is a first-line therapy in pediatric Crohn's disease (CD) thought to induce remission through changes in the gut microbiome. With microbiome assessment largely focused on microbial taxonomy and diversity, it remains unclear to what extent EEN induces functional changes that thereby contribute to its therapeutic effect. METHODS Fecal samples were collected from 15 pediatric CD patients prior to and after EEN treatment, as well as from 5 healthy controls. Metagenomic data were obtained via next-generation sequencing, and nonhuman reads were mapped to KEGG pathways, where possible. Pathway abundance was compared between CD patients and controls, and between CD patients that sustained remission (SR) and those that did not sustain remission (NSR). RESULTS Of 132 KEGG pathways identified, 8 pathways differed significantly between baseline CD patients and controls. Examination of these eight pathways showed SR patients had greater similarity to controls than NSR patients in all cases. Pathways fell into one of three groups: 1) no prior connection to IBD, 2) previously reported connection to IBD, and 3) known roles in innate immunity and immunoregulation. CONCLUSIONS The microbiota of CD patients and controls represent alternative ecological states that have broad differences in functional capabilities, including xenobiotic and environmental pollutant degradation, succinate metavolism, and bacterial HtpG, all of which can affect barrier integrity and immune regulation. Moreover, our finding that SR patients were more similar to healthy controls suggests that community microbial function, as inferred from fecal microbiomes, could serve as a valuable diagnostic tool.
Collapse
|
42
|
Effects of two whole-grain barley varieties on caecal SCFA, gut microbiota and plasma inflammatory markers in rats consuming low- and high-fat diets. Br J Nutr 2015; 113:1558-70. [DOI: 10.1017/s0007114515000793] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mixed-linkage β-glucans are fermented by the colon microbiota that give rise to SCFA. Propionic and butyric acids have been found to play an important role in colonic health, as well as they may have extraintestinal metabolic effects. The aim of the present study was to investigate how two whole-grain barley varieties differing in dietary fibre and β-glucan content affected caecal SCFA, gut microbiota and some plasma inflammatory markers in rats consuming low-fat (LF) or high-fat (HF) diets. Barley increased the caecal pool of SCFA in rats fed the LF and HF diets compared with those fed the control diet, and the effect was generally dependent on fibre content, an exception was butyric acid in the LF setting. Furthermore, whole-grain barley reduced plasma lipopolysaccharide-binding protein and monocyte chemoattractant protein-1, increased the caecal abundance of Lactobacillus and decreased the Bacteroides fragilis group, but increased the number of Bifidobacterium only when dietary fat was consumed at a low level. Fat content influenced the effects of barley: rats fed the HF diets had a higher caecal pool of acetic and propionic acids, higher concentrations of amino acids and higher amounts of lipids in the portal plasma and liver than rats fed the LF diets; however, less amounts of butyric acid were generally formed. Interestingly, there was an increase in the caecal abundance of Akkermansia and the caecal pool of succinic acid, and a decrease in the proportion of Bifidobacterium and the Clostridium leptum group. In summary, whole-grain barley decreased HF diet-induced inflammation, which was possibly related to the formation of SCFA and changes in microbiota composition. High β-glucan content in the diet was associated with reduced plasma cholesterol levels.
Collapse
|
43
|
Abstract
Celiac disease (CD) is a common chronic autoimmune enteropathy caused by gluten intake. To date, the only therapy for CD is the complete exclusion of dietary sources of grains and any food containing gluten. It has been hypothesized that the intestinal microbiota is somehow involved in CD. For this reason, probiotics are appearing as an interesting adjuvant in the dietetic management of CD. This review aims to discuss the characteristics of the microbiota in CD subjects and the use of probiotics as a novel therapy for CD. Comparisons between children with CD and controls show that their microbiota profiles differ; the former have fewer lactobacilli and bifidobacteria. Specific probiotics have been found to digest or alter gluten polypeptides. It has also been demonstrated that some bacterial species belonging to the genera Lactobacillus and Bifidobacterium exert protective properties on epithelial cells from damage caused by gliadin.
Collapse
|
44
|
McMullen L, T Leach S, A Lemberg D, S Day A. Current roles of specific bacteria in the pathogenesis of inflammatory bowel disease. AIMS Microbiol 2015. [DOI: 10.3934/microbiol.2015.1.82] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
45
|
Davis-Richardson AG, Ardissone AN, Dias R, Simell V, Leonard MT, Kemppainen KM, Drew JC, Schatz D, Atkinson MA, Kolaczkowski B, Ilonen J, Knip M, Toppari J, Nurminen N, Hyöty H, Veijola R, Simell T, Mykkänen J, Simell O, Triplett EW. Bacteroides dorei dominates gut microbiome prior to autoimmunity in Finnish children at high risk for type 1 diabetes. Front Microbiol 2014; 5:678. [PMID: 25540641 PMCID: PMC4261809 DOI: 10.3389/fmicb.2014.00678] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/19/2014] [Indexed: 01/15/2023] Open
Abstract
The incidence of the autoimmune disease, type 1 diabetes (T1D), has increased dramatically over the last half century in many developed countries and is particularly high in Finland and other Nordic countries. Along with genetic predisposition, environmental factors are thought to play a critical role in this increase. As with other autoimmune diseases, the gut microbiome is thought to play a potential role in controlling progression to T1D in children with high genetic risk, but we know little about how the gut microbiome develops in children with high genetic risk for T1D. In this study, the early development of the gut microbiomes of 76 children at high genetic risk for T1D was determined using high-throughput 16S rRNA gene sequencing. Stool samples from children born in the same hospital in Turku, Finland were collected at monthly intervals beginning at 4-6 months after birth until 2.2 years of age. Of those 76 children, 29 seroconverted to T1D-related autoimmunity (cases) including 22 who later developed T1D, the remaining 47 subjects remained healthy (controls). While several significant compositional differences in low abundant species prior to seroconversion were found, one highly abundant group composed of two closely related species, Bacteroides dorei and Bacteroides vulgatus, was significantly higher in cases compared to controls prior to seroconversion. Metagenomic sequencing of samples high in the abundance of the B. dorei/vulgatus group before seroconversion, as well as longer 16S rRNA sequencing identified this group as Bacteroides dorei. The abundance of B. dorei peaked at 7.6 months in cases, over 8 months prior to the appearance of the first islet autoantibody, suggesting that early changes in the microbiome may be useful for predicting T1D autoimmunity in genetically susceptible infants. The cause of increased B. dorei abundance in cases is not known but its timing appears to coincide with the introduction of solid food.
Collapse
Affiliation(s)
- Austin G Davis-Richardson
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Alexandria N Ardissone
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Raquel Dias
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Ville Simell
- Department of Pediatrics, Turku University Hospital and University of Turku Turku, Finland
| | - Michael T Leonard
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Kaisa M Kemppainen
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Jennifer C Drew
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Desmond Schatz
- Department of Pediatrics, University of Florida Gainesville, FL, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Gainesville, FL, USA
| | - Bryan Kolaczkowski
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| | - Jorma Ilonen
- Department of Clinical Microbiology, University of Eastern Finland Kuopio, Finland ; Immunogenetics Laboratory, University of Turku Turku, Finland
| | - Mikael Knip
- Department of Pediatrics, Children's Hospital, University of Helsinki and Helsinki University Central Hospital Helsinki, Finland ; Diabetes and Obesity Research Program, University of Helsinki Helsinki, Finland ; Department of Pediatrics, Tampere University Hospital Tampere, Finland
| | - Jorma Toppari
- Department of Pediatrics, Turku University Hospital and University of Turku Turku, Finland
| | - Noora Nurminen
- School of Medicine, University of Tampere Tampere, Finland
| | - Heikki Hyöty
- School of Medicine, University of Tampere Tampere, Finland
| | - Riitta Veijola
- Department of Pediatrics, University of Oulu, and Oulu University Hospital Oulu, Finland
| | - Tuula Simell
- Department of Pediatrics, Turku University Hospital and University of Turku Turku, Finland
| | - Juha Mykkänen
- Department of Pediatrics, Turku University Hospital and University of Turku Turku, Finland
| | - Olli Simell
- Department of Pediatrics, Turku University Hospital and University of Turku Turku, Finland
| | - Eric W Triplett
- Department of Microbiology and Cell Science, Institute of Food and Agricultural Sciences, University of Florida Gainesville, FL, USA
| |
Collapse
|
46
|
Downs R, Perna J, Vitelli A, Cook D, Dhurjati P. Model-based hypothesis of gut microbe populations and gut/brain barrier permeabilities in the development of regressive autism. Med Hypotheses 2014; 83:649-55. [DOI: 10.1016/j.mehy.2014.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/29/2014] [Accepted: 09/10/2014] [Indexed: 01/15/2023]
|
47
|
Chandrasekaran NC, Weir C, Alfraji S, Grice J, Roberts MS, Barnard RT. Effects of magnesium deficiency--more than skin deep. Exp Biol Med (Maywood) 2014; 239:1280-91. [PMID: 24928863 DOI: 10.1177/1535370214537745] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Dead Sea and magnesium salt therapy are two of the oldest forms of treatment for skin disease and several other disorders, supported by a body of largely anecdotal evidence. In this paper we review possible pathways for penetration of magnesium ions through the epidermis to reach the circulation, in turn replenishing cellular magnesium levels. We also discuss mechanisms for intercellular movement of magnesium ions and possible mechanisms for the interaction between magnesium ions and inflammatory mediators. Upon addition of magnesium ions in vitro, the expression of inflammatory mediators such as tumour necrosis factor α (TNFα) and nuclear factor κβ (NFκβ) is down regulated. Dysregulation of these and other inflammatory mediators has been linked to several inflammatory disorders, including asthma, arthritis, atherosclerosis and neuroinflammation.
Collapse
Affiliation(s)
- Navin Chandrakanth Chandrasekaran
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia School of Medicine, Translational Research Institute, The University of Queensland, Wooloongabba, Queensland 4102, Australia
| | - Christopher Weir
- Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Sumaya Alfraji
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| | - Jeff Grice
- School of Medicine, Translational Research Institute, The University of Queensland, Wooloongabba, Queensland 4102, Australia
| | - Michael S Roberts
- School of Medicine, Translational Research Institute, The University of Queensland, Wooloongabba, Queensland 4102, Australia
| | - Ross T Barnard
- School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre, The University of Queensland, Queensland 4072, Australia
| |
Collapse
|
48
|
Chen LL, Zou YY, Lu FG, Li FJ, Lian GH. Efficacy profiles for different concentrations of Lactobacillus acidophilus in experimental colitis. World J Gastroenterol 2013; 19:5347-5356. [PMID: 23983440 PMCID: PMC3752571 DOI: 10.3748/wjg.v19.i32.5347] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 05/29/2013] [Accepted: 07/19/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine the efficacy profiles of different concentrations of Lactobacillus acidophilus (L. acidophilus) for treating colitis using an experimental murine model.
METHODS: Colitis was established in 64 BALB/c mice by adding 5% dextran sodium sulfate (DSS) to the drinking water and allowing ad libitum access for 7 d. The mice were then randomly divided into the following control and experimental model groups (n = 8 each; day 0): untreated model control; negative-treatment model control (administered gavage of 1 mL/10 g normal saline); experimental-treatment models C4-C8 (administered gavage of 104, 105, 106, 107, or 108 CFU/10 g L. acidophilus, respectively); positive-treatment model control (administration of the anti-inflammatory agent prednisone acetate at 45 μg/10 g). Eight mice given regular water (no DSS) and no subsequent treatments served as the normal control group. Body weight, fecal traits, and presence of fecal occult blood were assessed daily. All animals were sacrificed on post-treatment day 7 to measure colonic length, perform histological scoring, and quantify the major bacteria in the proximal and distal colon. Intergroup differences were determined by one-way ANOVA and post-hoc Student-Newman-Keuls comparison.
RESULTS: All treatments (L. acidophilus and prednisone acetate) protected against colitis-induced weight loss (P < 0.05 vs model and normal control groups). The extent of colitis-induced colonic shortening was significantly reduced by all treatments (prednisone acetate > C4 > C5 > C7 > C8 > C6; P < 0.05 vs untreated model group), and the C6 group showed colonic length similar to that of the normal control group (P > 0.05). The C6 group also had the lowest disease activity index scores among the model groups. The bacterial profiles in the proximal colon were similar between all of the experimental-treatment model groups (all P > 0.05). In contrast, the bacterial profile in the distal colon of the C6 group showed the distinctive features (P < 0.05 vs all other experimental-treatment model groups) of Lactobacillus sp. and Bifidobacterium sp. being the most abundant bacteria and Staphylococcus aureus being the least abundant bacteria.
CONCLUSION: The most therapeutically efficacious concentration of L. acidophilus (106 CFU/10 g) may exert its effects by modulating the bacterial profile in the distal colon.
Collapse
|
49
|
Yao J, Wang JY, Lai MG, Li YX, Zhu HM, Shi RY, Mo J, Xun AY, Jia CH, Feng JL, Wang LS, Zeng WS, Liu L. Treatment of mice with dextran sulfate sodium-induced colitis with human interleukin 10 secreted by transformed Bifidobacterium longum. Mol Pharm 2011; 8:488-97. [PMID: 21271712 DOI: 10.1021/mp100331r] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease (IBD) the etiology of which has not yet been fully clarified. Cytokine interleukin-10 (IL-10) plays a central role in downregulating inflammatory cascade in UC and is likely a candidate for therapeutic intervention. However, its intravenous administration is costly and inconvenient. Therefore, we established a novel IL-10 delivery system by transforming a hIL-10-containing plasmid into B. longum (BL-hIL-10) and investigated its effects on 5% dextran sulfate sodium (DSS)-induced ulcerative colitis in mice and the possible underlying mechanism. Our results show that (1) hIL-10 was expressed and secreted into the culture supernatant of BL-hIL-10 after L-arabinose induction in vitro as examined by Western blot, enzyme-linked immunosorbent assay (ELISA) and RT-PCR; (2) addition of BL-hIL-10 culture supernatant had no cytotoxic effect and morphological alteration, but significantly inhibited the enhancement of proinflammatory cytokines by lipopolysaccharide (LPS) in THP-1 cells; (3) oral administration of BL-hIL-10 alleviated colitis syndrome of the model mice, attenuated colitis-activated NF-κB pathway measured by DNA-binding assay and colitis-elevated expression of proinflammatory cytokines examined with CCK cytotoxic kits, and upregulated CD4+CD25+Foxp3+ Treg in blood and mesenteric lymph nodes measured by flow cytometry. In conclusion, BL-hIL-10 as a novel oral hIL-10 delivery system has been successfully established and oral administration of BL-hIL-10 alleviated inflammatory damage of colonic tissue in the model mice by blocking the colitis-activated NF-κB pathway and upregulating CD4+CD25+Foxp3+ Treg in blood and mesenteric lymph nodes in mice.
Collapse
Affiliation(s)
- Jun Yao
- Department of Gastroenterology, Jinan University of Medical Sciences, Shenzhen Municipal People's Hospital, Shenzhen 518020, Guangdong Province, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Goto H, Takemura N, Ogasawara T, Sasajima N, Watanabe J, Ito H, Morita T, Sonoyama K. Effects of fructo-oligosaccharide on DSS-induced colitis differ in mice fed nonpurified and purified diets. J Nutr 2010; 140:2121-7. [PMID: 20943955 DOI: 10.3945/jn.110.125948] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We investigated whether feeding a purified compared with nonpurified diet supplemented with or without fructo-oligosaccharide (FOS; 50 g/kg diet) altered the response of C57BL/6 mice to DSS-induced diarrhea. In Expt. 1, we examined disease severity in mice receiving DSS (2% in drinking water) for 5 d. In Expt. 2, we measured cecal organic acid concentrations and fecal water-holding capacity (WHC). In Expts. 3 and 4, we tested whether polycarbophil calcium (PC), a water-absorbing polymer, altered fecal WHC and disease severity. FOS exacerbated diarrhea and weight loss in mice fed the purified diet and reduced fecal bleeding in mice fed the nonpurified diet (P < 0.05). Without DSS administration, cecal acetate and butyrate concentrations were higher in mice fed the nonpurified diet than in mice fed the purified diet (P < 0.05). Fecal WHC was higher in mice fed the nonpurified diet than in mice fed the purified diet (P < 0.05). One day after starting DSS administration, cecal succinate concentrations were higher in mice fed the FOS-supplemented purified diet than in mice fed the other 3 diets, whereas SCFA concentrations were higher in mice fed the nonpurified diet than in mice fed the purified diet (P < 0.05). PC supplementation increased fecal WHC and prevented FOS exacerbation of diarrhea in mice fed the purified diet (P < 0.05). We conclude that the effects of FOS on DSS-induced diarrhea differ in mice fed the purified and nonpurified diets. The protective effect of nonpurified diet was associated with increased production of organic acids and WHC in the intestinal contents.
Collapse
Affiliation(s)
- Haruka Goto
- Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | | | |
Collapse
|