1
|
Du X, Meng R, Wei H, Fan Z, Wang J, Yuan S, Ge K, Guo H, Wan F, Fu Y, Wang F, Chen X, Zhuang D, Guo H, Zhang H. Nicotinamide Alleviates Synergistic Impairment of Intestinal Barrier Caused by MC-LR and NaNO 2 Coexposure. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24024-24034. [PMID: 39404749 DOI: 10.1021/acs.jafc.4c06756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
Microcystin-LR (MC-LR) and nitrites from the environment and daily life can be ingested and absorbed by humans via the digestive tract. However, their combined effects on intestinal health remain unclear. Here, the combined impact of MC-LR and sodium nitrite (NaNO2) on the intestines of mice was investigated under actual human exposure conditions. After mice were exposed to MC-LR (10, 100 μg/L) and NaNO2 (30, 300 mg/L) individual and in combination for 6 months, it was found that MC-LR and NaNO2 synergistically decreased intestinal permeability and disrupted intestinal physical, chemical, immune, and microbial barriers. In the coexposure groups, the synergistic impairment to the intestinal barrier was noted with increasing concentrations of MC-LR or NaNO2, but this adverse effect was alleviated by nicotinamide supplementation. This study underscores the potential risks of simultaneous ingestion of MC-LR and nitrite on intestinal health. The protective role of nicotinamide suggests avenues for therapeutic intervention against environmental toxin-induced intestinal impairment.
Collapse
Affiliation(s)
- Xingde Du
- College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ruiyang Meng
- College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Houjiang Wei
- School of Henan Medical, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhe Fan
- School of Henan Medical, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jiankang Wang
- School of Henan Medical, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Shumeng Yuan
- College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Kangfeng Ge
- College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Haibin Guo
- The Reproductive Medicine Center, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Feng Wan
- The Reproductive Medicine Center, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Yu Fu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Fufang Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xinghai Chen
- Department of Chemistry and Biochemistry, St Mary's University, San Antonio, Texas 78228, United States
| | - Donggang Zhuang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Hongxiang Guo
- College of Life Sciences, Henan Agricultural University, Zhengzhou, Henan 450002, China
| | - Huizhen Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, China
| |
Collapse
|
2
|
Lin YJ, Li HM, Gao YR, Wu PF, Cheng B, Yu CL, Sheng YX, Xu HM. Environmentally relevant concentrations of benzophenones exposure disrupt intestinal homeostasis, impair the intestinal barrier, and induce inflammation in mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 350:123948. [PMID: 38614423 DOI: 10.1016/j.envpol.2024.123948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/28/2024] [Accepted: 04/08/2024] [Indexed: 04/15/2024]
Abstract
The aim of this study is to investigate the adverse effects of benzophenones (BPs) on the intestinal tract of mice and the potential mechanism. F1-generation ICR mice were exposed to BPs (benzophenone-1, benzophenone-2, and benzophenone-3) by breastfeeding from birth until weaning, and by drinking water after weaning until maturity. The offspring mice were executed on postnatal day 56, then their distal colons were sampled. AB-PAS staining, HE staining, immunofluorescence, Transmission Electron Microscope, immunohistochemistry, Western Blot and RT-qPCR were used to study the effects of BPs exposure on the colonic tissues of offspring mice. The results showed that colonic microvilli appeared significantly deficient in the high-dose group, and the expression of tight junction markers Zo-1 and Occludin was significantly down-regulated and the number of goblet cells and secretions were reduced in all dose groups, and the expression of secretory cell markers MUC2 and KI67 were decreased, as well as the expression of intestinal stem cell markers Lgr5 and Bmi1, suggesting that BPs exposure caused disruption of intestinal barrier and imbalance in the composition of the intestinal stem cell pool. Besides, the expression of cellular inflammatory factors such as macrophage marker F4/80 and tumor necrosis factor TNF-α was elevated in the colonic tissues of all dose groups, and the inflammatory infiltration was observed, which means the exposure of BPs caused inflammatory effects in the intestinal tract of F1-generation mice. In addition, the contents of Notch/Wnt signaling pathway-related genes, such as Dll-4, Notch1, Hes1, Ctnnb1and Sfrp2 were significantly decreased in each high-dose group (P < 0.05), suggesting that BPs may inhibit the regulation of Notch/Wnt signaling pathway. In conclusion, exposure to BPs was able to imbalance colonic homeostasis, disrupt the intestinal barrier, and trigger inflammation in the offspring mice, which might be realized through interfering with the Notch/Wnt signaling pathway.
Collapse
Affiliation(s)
- Yu-Jia Lin
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; The Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Hong-Mei Li
- The Key Laboratory of Fertility Preservation and Maintenance of the Ministry of Education, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yan-Rong Gao
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; The Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Ping-Fan Wu
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; The Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Bin Cheng
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; The Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Chen-Long Yu
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; The Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yu-Xin Sheng
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; The Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Hai-Ming Xu
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, Ningxia, China; The Key Laboratory of Environmental Factors and Chronic Disease Control, Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
3
|
Huang J, Guo F, Abbas W, Hu Z, Liu L, Qiao J, Bi R, Xu T, Zhang K, Huang J, Guo Y, Wang Z. Effects of microencapsulated essential oils and organic acids preparation on growth performance, slaughter characteristics, nutrient digestibility and intestinal microenvironment of broiler chickens. Poult Sci 2024; 103:103655. [PMID: 38537402 PMCID: PMC11067778 DOI: 10.1016/j.psj.2024.103655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/05/2024] [Accepted: 03/10/2024] [Indexed: 05/01/2024] Open
Abstract
To develop effective antibiotics alternatives is getting more and more important to poultry healthy production. The study investigated the effects of a microencapsulated essential oils and organic acids preparation (EOA) on growth performance, slaughter performance, nutrient digestibility and intestinal microenvironment of broilers. A total of 624 1-day-old male Arbor Acres broilers were randomly divided into 6 groups including the control group (T1) fed with basal diet, the antibiotic group (T2) supplemented with basal diet with 45 mg/kg bacitracin methylene disalicylate (BMD), and 4 inclusion levels of EOA-treated groups (T3, T4, T5, T6 groups) chickens given basal diet with 200, 400, 600, and 800 mg EOA/kg of diet, respectively. Results showed that compared with the control, the 200 mg/kg EOA group increased average daily gain (ADG) and average body weight (ABW) during the early stage (P < 0.05). EOA addition decreased crypt depth of the ileum (P < 0.05), but villus height to crypt depth ratio was increased by EOA addition at 200 and 400 mg/kg at d 21 (P < 0.05). Compared with the control, dietary addition EOA at 200, 400 and 600 mg/kg increased the lipase activity in the duodenum at d 21 (P < 0.05). Increased lactic acid bacteria population was found in cecal digesta of the 400 mg/kg EOA group at d 21 (P < 0.05), and higher concentration of butyric acid level was observed in cecal digesta at d 21 and d 42 in the 200 mg/kg EOA group compared with the control (P < 0.05). RT-PCR analysis found that dietary EOA addition decreased the gene expression of IL-1β, COX-2 and TGF-β4 in the ileum at d 21 (P < 0.05), while only the 200 mg/kg EOA increased the gene expression of IL-10, TGF-β4, Claudin-1, ZO-1, CATH-1, CATH-3, AvBD-1, AvBD-9 and AvBD-12 in the ileum at d 42 (P < 0.05) compared with the control. In summary, adding 200 mg/kg and 400 mg/kg of the EOA to the diet could improve the growth performance and intestinal microenvironment through improving intestinal morphology, increasing digestive enzymes activity and cecal lactic acid bacteria abundance and butyric acid content, improving intestinal barrier function as well as maintaining intestinal immune homeostasis. The improving effect induced by EOA addition in the early growth stage was better than that in the later growth stage. Overall, the EOA product might be an effective antibiotic alternative for broiler industry.
Collapse
Affiliation(s)
- Jia Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Fangshen Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Waseem Abbas
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zeqiong Hu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lin Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jianing Qiao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ruichen Bi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Tiantian Xu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Kaichen Zhang
- Shandong Heyi Food Co., Ltd., Zaozhuang City, Shandong Province, China
| | - Jinyu Huang
- Shanghai Meinong Biotechnology Co., Ltd., Shanghai, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhong Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| |
Collapse
|
4
|
Abbas W, Bi R, Hussain MD, Tajdar A, Guo F, Guo Y, Wang Z. Antibiotic Cocktail Effects on Intestinal Microbial Community, Barrier Function, and Immune Function in Early Broiler Chickens. Antibiotics (Basel) 2024; 13:413. [PMID: 38786141 PMCID: PMC11117290 DOI: 10.3390/antibiotics13050413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 04/23/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
This study investigated the effects of an antibiotic cocktail on intestinal microbial composition, mechanical barrier structure, and immune functions in early broilers. One-day-old healthy male broiler chicks were treated with a broad-spectrum antibiotic cocktail (ABX; neomycin, ampicillin, metronidazole, vancomycin, and kanamycin, 0.5 g/L each) or not in drinking water for 7 and 14 days, respectively. Sequencing of 16S rRNA revealed that ABX treatment significantly reduced relative Firmicutes, unclassified Lachnospiraceae, unclassified Oscillospiraceae, Ruminococcus torques, and unclassified Ruminococcaceae abundance in the cecum and relative Firmicutes, Lactobacillus and Baccillus abundance in the ileum, but significantly increased richness (Chao and ACE indices) and relative Enterococcus abundance in the ileum and cecum along with relatively enriched Bacteroidetes, Proteobacteria, Cyanobacteria, and Enterococcus levels in the ileum following ABX treatment for 14 days. ABX treatment for 14 days also significantly decreased intestinal weight and length, along with villus height (VH) and crypt depth (CD) of the small intestine, and remarkably increased serum LPS, TNF-α, IFN-γ, and IgG levels, as well as intestinal mucosa DAO and MPO activity. Moreover, prolonged use of ABX significantly downregulated occludin, ZO-1, and mucin 2 gene expression, along with goblet cell numbers in the ileum. Additionally, chickens given ABX for 14 days had lower acetic acid, butyric acid, and isobutyric acid content in the cecum than the chickens treated with ABX for 7 days and untreated chickens. Spearman correlation analysis found that those decreased potential beneficial bacteria were positively correlated with gut health-related indices, while those increased potential pathogenic strains were positively correlated with gut inflammation and gut injury-related parameters. Taken together, prolonged ABX application increased antibiotic-resistant species abundance, induced gut microbiota dysbiosis, delayed intestinal morphological development, disrupted intestinal barrier function, and perturbed immune response in early chickens. This study provides a reliable lower-bacteria chicken model for further investigation of the function of certain beneficial bacteria in the gut by fecal microbiota transplantation into germ-free or antibiotic-treated chickens.
Collapse
Affiliation(s)
- Waseem Abbas
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100093, China; (W.A.); (R.B.); (F.G.); (Y.G.)
| | - Ruichen Bi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100093, China; (W.A.); (R.B.); (F.G.); (Y.G.)
| | - Muhammad Dilshad Hussain
- MARA-Key Laboratory of Surveillance and Management for Plant Quarantine Pests, College of Plant Protection, China Agricultural University, Beijing 100193, China;
- Key Laboratory of Agricultural Microbiology, College of Agriculture, Guizhou University, Guiyang 550025, China
| | - Alia Tajdar
- Key Laboratory of Insect Behavior and Harmless Management, College of Plant Protection, China Agricultural University, Beijing 100193, China;
| | - Fangshen Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100093, China; (W.A.); (R.B.); (F.G.); (Y.G.)
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100093, China; (W.A.); (R.B.); (F.G.); (Y.G.)
| | - Zhong Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100093, China; (W.A.); (R.B.); (F.G.); (Y.G.)
| |
Collapse
|
5
|
Zhu M, Zeng R, Wu D, Li Y, Chen T, Wang A. Research progress of the effects of bisphenol analogues on the intestine and its underlying mechanisms: A review. ENVIRONMENTAL RESEARCH 2024; 243:117891. [PMID: 38072107 DOI: 10.1016/j.envres.2023.117891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/26/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023]
Abstract
Bisphenol A (BPA) and its analogues have prompted rising concerns, especially in terms of human safety, due to its broad use and ubiquity throughout the ecosystem. Numerous studies reported various adverse effects of bisphenols, including developmental disorders, reproductive toxicity, cardiovascular toxicity, and so on. There is increasing evidence that bisphenols can enter the gastrointestinal tract. Consequently, it is important to investigate their effects on the intestine. Several in vivo and in vitro studies have examined the impacts of bisphenols on the intestine. Here, we summarized the literature concerning intestinal toxicity of bisphenols over the past decade and presented compelling evidence of the link between bisphenol exposure and intestinal disorders. Experiment studies revealed that even at low levels, bisphenols could promote intestinal barrier dysregulation, disrupt the composition and diversity of intestinal microbiota as well as induce an immunological response. Moreover, possible underlying mechanisms of these effects were discussed. Because of a lack of empirical data, the potential risk of bisphenol exposure in humans is still unidentified, particularly regarding intestinal disorders. Thus, we propose to conduct additional epidemiological investigations and animal experiments to elucidate the associations between bisphenol exposure and human intestinal health and reveal underlying mechanisms to develop preventative and therapeutic techniques.
Collapse
Affiliation(s)
- Min Zhu
- Jiangsu Provincial Key Laboratory of Environmental Engineering, Jiangsu Provincial Academy of Environmental Science, 210036, Nanjing, China; Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 100085, Beijing, China
| | - Ran Zeng
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 100085, Beijing, China; School of Civil and Environmental Engineering, Harbin Institute of Technology, 518055, Shenzhen, China
| | - Dan Wu
- Jiangsu Provincial Key Laboratory of Environmental Engineering, Jiangsu Provincial Academy of Environmental Science, 210036, Nanjing, China
| | - Yuanyuan Li
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 100085, Beijing, China
| | - Ting Chen
- Jiangsu Provincial Key Laboratory of Environmental Engineering, Jiangsu Provincial Academy of Environmental Science, 210036, Nanjing, China.
| | - Aijie Wang
- School of Civil and Environmental Engineering, Harbin Institute of Technology, 518055, Shenzhen, China.
| |
Collapse
|
6
|
Li J, Guo Y, Ma L, Liu Y, Zou C, Kuang H, Han B, Xiao Y, Wang Y. Synergistic effects of alginate oligosaccharide and cyanidin-3-O-glucoside on the amelioration of intestinal barrier function in mice. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.03.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
7
|
Qi X, Zhang Y, Zhang Y, Luo F, Song K, Wang G, Ling F. Vitamin B 12 produced by Cetobacterium somerae improves host resistance against pathogen infection through strengthening the interactions within gut microbiota. MICROBIOME 2023; 11:135. [PMID: 37322528 PMCID: PMC10268390 DOI: 10.1186/s40168-023-01574-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/15/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Pathogen infections seriously affect host health, and the use of antibiotics increases the risk of the emergence of drug-resistant bacteria and also increases environmental and health safety risks. Probiotics have received much attention for their excellent ability to prevent pathogen infections. Particularly, explaining mechanism of action of probiotics against pathogen infections is important for more efficient and rational use of probiotics and the maintenance of host health. RESULTS Here, we describe the impacts of probiotic on host resistance to pathogen infections. Our findings revealed that (I) the protective effect of oral supplementation with B. velezensis against Aeromonas hydrophila infection was dependent on gut microbiota, specially the anaerobic indigenous gut microbe Cetobacterium; (II) Cetobacterium was a sensor of health, especially for fish infected with pathogenic bacteria; (III) the genome resolved the ability of Cetobacterium somerae CS2105-BJ to synthesize vitamin B12 de novo, while in vivo and in vitro metabolism assays also showed the ability of Cetobacterium somerae CS2105-BJ to produce vitamin B12; (IV) the addition of vitamin B12 significantly altered the gut redox status and the gut microbiome structure and function, and then improved the stability of the gut microbial ecological network, and enhanced the gut barrier tight junctions to prevent the pathogen infection. CONCLUSION Collectively, this study found that the effect of probiotics in enhancing host resistance to pathogen infections depended on function of B12 produced by an anaerobic indigenous gut microbe, Cetobacterium. Furthermore, as a gut microbial regulator, B12 exhibited the ability to strengthen the interactions within gut microbiota and gut barrier tight junctions, thereby improving host resistance against pathogen infection. Video Abstract.
Collapse
Affiliation(s)
- Xiaozhou Qi
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yong Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yilin Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Fei Luo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Kaige Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Gaoxue Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| | - Fei Ling
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
8
|
Huang MY, Lo CY, Lai CY, Yu JD, Lee PT. Dietary supplementation of synbiotic Leuconostoc mesenteroide B4 and dextran improves immune regulation and disease resistance of Penaeus vannamei against Vibrio parahaemolyticus. FISH & SHELLFISH IMMUNOLOGY 2023; 132:108498. [PMID: 36539168 DOI: 10.1016/j.fsi.2022.108498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
White shrimp (Penaeus vannamei) is an important culture species in Taiwan but often encounters disease infection by Vibrio parahaemolyticus that cause acute hepatopancreatic necrosis disease (AHPND). This study investigates the effects of dietary supplementation of Leuconostoc mesenteroide B4 and its fermentate (dextran) on the immune response, intestinal morphology, disease resistance, and immune-related gene expression in white shrimp. In comparison to the control group, the shrimp fed with a diet containing B4+dextran (107 CFU B4/g feed and 0.05% dextran) for 14, 28, 42 and 56 days had a significantly higher feed efficiency, weight gain and specific growth rate. A significantly higher villus height in the intestine and higher survival rate after challenging with V. parahaemolyticus was recorded for the B4+dextran group. Flow cytometry analysis demonstrated that the group that had ingested B4+dextran had a higher total hemocyte count and a higher proportion of semi-granulocytes, but a lower percentage of granulocytes compared to the control group. The shotgun metagenomic results in the midgut revealed that Leuco. mesenteroides was barely found in the midgut of the shrimp, suggesting that this microbe and its transient presence in the midgut is not the direct mechanism underlying the improved shrimp growth in the treated sample. Instead, dextran, a key ingredient in the B4 fermentate, on the dynamic of the microbial populations in shrimp, possibly promoting the diversity of gut microbes, especially the beneficial microbes, and thereby rendering protection against AHPND. In terms of comparing the gene expression between the control and synbiotic groups, pre- and post-bacterial challenge, a higher expression level of immune genes was mostly found in the B4+dextran group after challenging it with V. parahaemolyticus (group B4+dextran-VP) in the hepatopancreas and hemocyte. In contrast, the transcript level of immune-related genes was found to be higher in the B4+dextran group than other combinations in the midgut. Taken together, this study found that dietary addition of synbiotic Leuco. mesenteroides B4 and dextran can improve the growth performance, intestinal morphology and microbiome, regulation of immune genes and disease resistance against V. parahaemolyticus infection in white shrimp.
Collapse
Affiliation(s)
- Mei-Ying Huang
- Aquaculture Division, Fisheries Research Institute, Council of Agriculture, Taiwan
| | - Chia-Yi Lo
- Department of Aquaculture, National Taiwan Ocean University, Keelung, Taiwan
| | | | | | - Po-Tsang Lee
- Department of Aquaculture, National Taiwan Ocean University, Keelung, Taiwan.
| |
Collapse
|
9
|
Jiang ZF, Wu W, Hu HB, Li ZY, Zhong M, Zhang L. P2X7 receptor as the regulator of T-cell function in intestinal barrier disruption. World J Gastroenterol 2022; 28:5265-5279. [PMID: 36185635 PMCID: PMC9521516 DOI: 10.3748/wjg.v28.i36.5265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/20/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023] Open
Abstract
The intestinal mucosa is a highly compartmentalized structure that forms a direct barrier between the host intestine and the environment, and its dysfunction could result in a serious disease. As T cells, which are important components of the mucosal immune system, interact with gut microbiota and maintain intestinal homeostasis, they may be involved in the process of intestinal barrier dysfunction. P2X7 receptor (P2X7R), a member of the P2X receptors family, mediates the effects of extracellular adenosine triphosphate and is expressed by most innate or adaptive immune cells, including T cells. Current evidence has demonstrated that P2X7R is involved in inflammation and mediates the survival and differentiation of T lymphocytes, indicating its potential role in the regulation of T cell function. In this review, we summarize the available research about the regulatory role and mechanism of P2X7R on the intestinal mucosa-derived T cells in the setting of intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Zhi-Feng Jiang
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Wei Wu
- Department of Critical Care Medicine, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Han-Bing Hu
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Zheng-Yang Li
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Lin Zhang
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| |
Collapse
|
10
|
Zhu M, Wei R, Li Y, Li J, Dong M, Chen X, Lv L, Qin Z. Bisphenol chemicals disturb intestinal homeostasis via Notch/Wnt signaling and induce mucosal barrier dysregulation and inflammation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 828:154444. [PMID: 35278557 DOI: 10.1016/j.scitotenv.2022.154444] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/22/2022] [Accepted: 03/06/2022] [Indexed: 06/14/2023]
Abstract
Emerging evidence has shown that bisphenol A (BPA) can exert adverse effects on intestinal barrier in rodents, but little is known about its underlying mechanisms. We previously found BPA and its substitute bisphenol F (BPF) disrupted Notch signaling and altered intestinal histological structures in Xenopus laevis tadpoles. The present study aimed to determine whether BPA and BPF could affect intestinal homeostasis via Notch/Wnt signaling and induce intestinal barrier dysregulation in adult mammals, given the fundamental roles of the two conserved signaling pathways in intestinal homeostasis and regulation of intestinal barrier. We found that following 7-day administration with BPA or BPF through drinking water at the reference dose of 50 μg/kg/d and no observed adverse effect level of 5 mg/kg/d (NOAEL) of BPA, adult male mice displayed no alterations at histological and cellular levels in colons, but high dose of both BPA and BPF downregulated the expression of Notch- and Wnt-related genes as well as key genes responsible for intestinal homeostasis. When administration was extended to 14 days, all treatments significantly suppressed the expression of all tested Notch- and Wnt-related genes; correspondingly, administrated colons exhibited downregulated expression of key genes responsible for intestinal homeostasis and reduced cell proliferation in crypts. Importantly, all treatments suppressed secretory cell differentiation, reduced mucin protein levels and downregulated expression of tight junction markers, implicating mucosal barrier dysregulation. Furthermore, inflammatory cell infiltration and upregulated expression of inflammatory cytokine genes in colons, coupled with increased serum inflammatory cytokine levels, were observed in all treatments. All results show that both BPA and BPF at the reference dose disrupted Notch/Wnt signaling and intestinal homeostasis, thereby leading to mucosal barrier dysregulation and intestinal inflammation in mice. This is the first study revealing the adverse influences of BPF on mammal intestines and underlying mechanisms for bisphenol-caused intestinal injury.
Collapse
Affiliation(s)
- Min Zhu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China; Jiangsu Provincial Key Laboratory of Environmental Engineering, Jiangsu Provincial Academy of Environmental Science, Nanjing 210036, China
| | - Rongguo Wei
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Department of Clinical Laboratory, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning 530016, China
| | - Yuanyuan Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinbo Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengqi Dong
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuanyue Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Lv
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhanfen Qin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
11
|
Rastall RA, Diez-Municio M, Forssten SD, Hamaker B, Meynier A, Moreno FJ, Respondek F, Stah B, Venema K, Wiese M. Structure and function of non-digestible carbohydrates in the gut microbiome. Benef Microbes 2022; 13:95-168. [PMID: 35729770 DOI: 10.3920/bm2021.0090] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Together with proteins and fats, carbohydrates are one of the macronutrients in the human diet. Digestible carbohydrates, such as starch, starch-based products, sucrose, lactose, glucose and some sugar alcohols and unusual (and fairly rare) α-linked glucans, directly provide us with energy while other carbohydrates including high molecular weight polysaccharides, mainly from plant cell walls, provide us with dietary fibre. Carbohydrates which are efficiently digested in the small intestine are not available in appreciable quantities to act as substrates for gut bacteria. Some oligo- and polysaccharides, many of which are also dietary fibres, are resistant to digestion in the small intestines and enter the colon where they provide substrates for the complex bacterial ecosystem that resides there. This review will focus on these non-digestible carbohydrates (NDC) and examine their impact on the gut microbiota and their physiological impact. Of particular focus will be the potential of non-digestible carbohydrates to act as prebiotics, but the review will also evaluate direct effects of NDC on human cells and systems.
Collapse
Affiliation(s)
- R A Rastall
- Department of Food and Nutritional Sciences, The University of Reading, P.O. Box 226, Whiteknights, Reading, RG6 6AP, United Kingdom
| | - M Diez-Municio
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), CEI (UAM+CSIC), Nicolás Cabrera 9, 28049 Madrid, Spain
| | - S D Forssten
- IFF Health & Biosciences, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - B Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, 745 Agriculture Mall Drive, West Lafayette, IN 47907-2009, USA
| | - A Meynier
- Nutrition Research, Mondelez France R&D SAS, 6 rue René Razel, 91400 Saclay, France
| | - F Javier Moreno
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), CEI (UAM+CSIC), Nicolás Cabrera 9, 28049 Madrid, Spain
| | - F Respondek
- Tereos, Zoning Industriel Portuaire, 67390 Marckolsheim, France
| | - B Stah
- Human Milk Research & Analytical Science, Danone Nutricia Research, Uppsalalaan 12, 3584 CT Utrecht, the Netherlands.,Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, the Netherlands
| | - K Venema
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University - campus Venlo, St. Jansweg 20, 5928 RC Venlo, the Netherlands
| | - M Wiese
- Department of Microbiology and Systems Biology, TNO, Utrechtseweg 48, 3704 HE, Zeist, the Netherlands
| |
Collapse
|
12
|
Niewiem M, Grzybowska-Chlebowczyk U. Intestinal Barrier Permeability in Allergic Diseases. Nutrients 2022; 14:nu14091893. [PMID: 35565858 PMCID: PMC9101724 DOI: 10.3390/nu14091893] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 02/01/2023] Open
Abstract
The role of intestinal permeability (IP) markers among children and adults with food allergies is not fully understood, and the identification of biological indicators/markers that predict growth retardation in children with allergic diseases and atopy has not been well explained. Studies have shown that patients with atopic diseases respond abnormally to food allergens. Accordingly, differences in the types of immune complexes formed in response to antigen challenges are significant, which seems to underlie the systemic signs of the food allergy. Increased intestinal permeability over the course of a food allergy allows allergens to penetrate through the intestinal barrier and stimulate the submucosal immune system. Additionally, the release of cytokines and inflammatory mediators enhances the degradation of the epithelial barrier and leads to an improper cycle, resulting in increased intestinal permeability. Several studies have also demonstrated increased permeability of the epithelial cells in those afflicted with atopic eczema and bronchial asthma. Ongoing research is aimed at finding various indicators to assess IP in patients with atopic diseases.
Collapse
|
13
|
Zheng H, Cao H, Zhang D, Huang J, Li J, Wang S, Lu J, Li X, Yang G, Shi X. Cordyceps militaris Modulates Intestinal Barrier Function and Gut Microbiota in a Pig Model. Front Microbiol 2022; 13:810230. [PMID: 35369439 PMCID: PMC8969440 DOI: 10.3389/fmicb.2022.810230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/26/2022] [Indexed: 12/13/2022] Open
Abstract
This study investigated the effects of Cordyceps militaris (CM) on intestinal barrier function and gut microbiota in a pig model. A total of 160 pigs were randomly allocated to either a control group (fed the basal diet) or a CM group (fed the basal diet supplemented with 300 mg/kg CM). CM improved intestinal morphology and increased the numbers of goblet cells and intraepithelial lymphocytes. CM also elevated the expression of zona occluden-1, claudin-1, mucin-2 and secretory immunoglobulin A. Furthermore, the mucosal levels of pro-inflammatory cytokines were downregulated while the levels of anti-inflammatory cytokines were upregulated in the CM group. Mechanistically, CM downregulated the expression of key proteins of the TLR4/MyD88/NF-κB signaling pathway. Moreover, CM altered the colonic microbial composition and increased the concentrations of acetate and butyrate. In conclusion, CM can modulate the intestinal barrier function and gut microbiota, which may provide a new strategy for improving intestinal health.
Collapse
|
14
|
Chen Z, Luo J, Li J, Kim G, Chen ES, Xiao S, Snapper SB, Bao B, An D, Blumberg RS, Lin CH, Wang S, Zhong J, Liu K, Li Q, Wu C, Kuchroo VK. Foxo1 controls gut homeostasis and commensalism by regulating mucus secretion. J Exp Med 2021; 218:e20210324. [PMID: 34287641 PMCID: PMC8424467 DOI: 10.1084/jem.20210324] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/18/2021] [Accepted: 06/30/2021] [Indexed: 12/23/2022] Open
Abstract
Mucus produced by goblet cells in the gastrointestinal tract forms a biological barrier that protects the intestine from invasion by commensals and pathogens. However, the host-derived regulatory network that controls mucus secretion and thereby changes gut microbiota has not been well studied. Here, we identify that Forkhead box protein O1 (Foxo1) regulates mucus secretion by goblet cells and determines intestinal homeostasis. Loss of Foxo1 in intestinal epithelial cells (IECs) results in defects in goblet cell autophagy and mucus secretion, leading to an impaired gut microenvironment and dysbiosis. Subsequently, due to changes in microbiota and disruption in microbiome metabolites of short-chain fatty acids, Foxo1 deficiency results in altered organization of tight junction proteins and enhanced susceptibility to intestinal inflammation. Our study demonstrates that Foxo1 is crucial for IECs to establish commensalism and maintain intestinal barrier integrity by regulating goblet cell function.
Collapse
Affiliation(s)
- Zuojia Chen
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jialie Luo
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jian Li
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Girak Kim
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Eric S. Chen
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA
| | - Sheng Xiao
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA
| | - Scott B. Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Boston, MA
| | - Bin Bao
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA
| | - Dingding An
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA
| | - Richard S. Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Cheng-hui Lin
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA
| | - Sui Wang
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA
| | - Jiaxin Zhong
- Department of Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Kuai Liu
- Department of Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Qiyuan Li
- Department of Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Chuan Wu
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Vijay K. Kuchroo
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA
- Klarman Cell Observatory, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| |
Collapse
|
15
|
Hernández-Ramírez JO, Merino-Guzmán R, Téllez-Isaías G, Vázquez-Durán A, Méndez-Albores A. Mitigation of AFB 1-Related Toxic Damage to the Intestinal Epithelium in Broiler Chickens Consumed a Yeast Cell Wall Fraction. Front Vet Sci 2021; 8:677965. [PMID: 34381831 PMCID: PMC8350163 DOI: 10.3389/fvets.2021.677965] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/21/2021] [Indexed: 11/21/2022] Open
Abstract
In vivo experiments were conducted to evaluate the effectiveness of a yeast cell wall fraction (YCW) to reduce the negative impact of aflatoxin B1 (AFB1) to the intestinal epithelium in broiler chickens. Zeta potential (ζ-potential), point of zero charge (pHpzc), Fourier transform infrared spectroscopy (FTIR), and scanning electron microscopy (SEM) techniques were used to characterize the YCW. Two hundred one-day-old male Ross 308 broiler chickens were randomly allocated into four treatments: (1) control, chickens fed an AFB1-free diet; (2) AF, chickens feed an AFB1-contaminated diet (500 ng AFB1/g); (3) YCW, chickens fed an AFB1-free diet + 0.05% YCW; and (4) AF + YCW, chickens fed an AFB1-contaminated diet (500 ng AFB1/g) + 0.05% YCW. At the end of the 21-day feeding period, fluorescein isothiocyanate dextran (FITC-d) was administered to chicks by oral gavage to evaluate gastrointestinal leakage. Blood and duodenum samples were collected to assess serum biochemistry and histomorphology, respectively. Compared to the control group, chicks of the AF group significantly diminished weight gain (WG) and average daily feed intake (ADFI), and increased feed conversion ratio (FCR), mortality rate (MR), and intestinal lesion scores (p < 0.05). Alterations in some serum biochemical parameters, and damage to the intestinal integrity were also evident in the AF-intoxicated birds. YCW supplementation improved WG and FCR and increased villus height, villus area, crypt depth, and the number of goblet cells in villi. The effects of YCW on growth performance were not significant in chicks of the AF + YCW group; however, the treatment decreased MR and significantly ameliorated some biochemical and histomorphological alterations. The beneficial effect of YCW was more evident in promoting gut health since chickens of the AF + YCW group presented a significant reduction in serum FITC-d concentration. This positive effect was mainly related to the changes in negative charges of YCW due to changes in pH, the net negative surface charge above the pHpzc, the higher quantities of negative charged functional groups on the YCW surface, and its ability to form large aggregates. From these results, it can be concluded that YCW at low supplementation level can partially protect broilers' intestinal health from chronic exposure to AFB1.
Collapse
Affiliation(s)
- Juan Omar Hernández-Ramírez
- Unidad de Investigación Multidisciplinaria L14 (Alimentos, Micotoxinas, y Micotoxicosis), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rubén Merino-Guzmán
- Departamento de Medicina y Zootecnia de Aves, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | | | - Alma Vázquez-Durán
- Unidad de Investigación Multidisciplinaria L14 (Alimentos, Micotoxinas, y Micotoxicosis), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Abraham Méndez-Albores
- Unidad de Investigación Multidisciplinaria L14 (Alimentos, Micotoxinas, y Micotoxicosis), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
16
|
Nagu P, Parashar A, Behl T, Mehta V. Gut Microbiota Composition and Epigenetic Molecular Changes Connected to the Pathogenesis of Alzheimer's Disease. J Mol Neurosci 2021; 71:1436-1455. [PMID: 33829390 DOI: 10.1007/s12031-021-01829-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, and its pathogenesis is not fully known. Although there are several hypotheses, such as neuroinflammation, tau hyperphosphorylation, amyloid-β plaques, neurofibrillary tangles, and oxidative stress, none of them completely explain the origin and progression of AD. Emerging evidence suggests that gut microbiota and epigenetics can directly influence the pathogenesis of AD via their effects on multiple pathways, including neuroinflammation, oxidative stress, and amyloid protein. Various gut microbes such as Actinobacteria, Bacteroidetes, E. coli, Firmicutes, Proteobacteria, Tenericutes, and Verrucomicrobia are known to play a crucial role in the pathogenesis of AD. These microbes and their metabolites modulate various physiological processes that contribute to AD pathogenesis, such as neuroinflammation and other inflammatory processes, amyloid deposition, cytokine storm syndrome, altered BDNF and NMDA signaling, impairing neurodevelopmental processes. Likewise, epigenetic markers associated with AD mainly include histone modifications and DNA methylation, which are under the direct control of a variety of enzymes, such as acetylases and methylases. The activity of these enzymes is dependent upon the metabolites generated by the host's gut microbiome, suggesting the significance of epigenetics in AD pathogenesis. It is interesting to know that both gut microbiota and epigenetics are dynamic processes and show a high degree of variation according to diet, stressors, and environmental factors. The bidirectional relation between the gut microbiota and epigenetics suggests that they might work in synchrony to modulate AD representation, its pathogenesis, and progression. They both also provide numerous targets for early diagnostic biomarkers and for the development of AD therapeutics. This review discusses the gut microbiota and epigenetics connection in the pathogenesis of AD and aims to highlight vast opportunities for diagnosis and therapeutics of AD.
Collapse
Affiliation(s)
- Priyanka Nagu
- Department of Pharmaceutics, Govt. College of Pharmacy, Rohru, Himachal Pradesh, India.,Department of Pharmacy, Shri Jagdishprasad Jhabarmal Tibrewala University, Jhunjhunu, Rajasthan, India
| | - Arun Parashar
- Faculty of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vineet Mehta
- Department of Pharmacology, Govt. College of Pharmacy, Rohru, Himachal Pradesh, India.
| |
Collapse
|
17
|
Perioperative neurocognitive dysfunction: thinking from the gut? Aging (Albany NY) 2020; 12:15797-15817. [PMID: 32805716 PMCID: PMC7467368 DOI: 10.18632/aging.103738] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
With the aging of the world population, and improvements in medical and health technologies, there are increasing numbers of elderly patients undergoing anaesthesia and surgery. Perioperative neurocognitive dysfunction has gradually attracted increasing attention from academics. Very recently, 6 well-known journals jointly recommended that the term perioperative neurocognitive dysfunction (defined according to the Diagnostic and Statistical Manual of Mental Disorders, fifth edition) should be adopted to improve the quality and consistency of academic communications. Perioperative neurocognitive dysfunction currently includes preoperatively diagnosed cognitive decline, postoperative delirium, delayed neurocognitive recovery, and postoperative cognitive dysfunction. Increasing evidence shows that the gut microbiota plays a pivotal role in neuropsychiatric diseases, and in central nervous system functions via the microbiota-gut-brain axis. We recently reported that abnormalities in the composition of the gut microbiota might underlie the mechanisms of postoperative cognitive dysfunction and postoperative delirium, suggesting a critical role for the gut microbiota in perioperative neurocognitive dysfunction. This article therefore reviewed recent findings on the linkage between the gut microbiota and the underlying mechanisms of perioperative neurocognitive dysfunction.
Collapse
|
18
|
Otero C, Arredondo C, Echeverría-Vega A, Gordillo-Fuenzalida F. Penicillium spp. mycotoxins found in food and feed and their health effects. WORLD MYCOTOXIN J 2020. [DOI: 10.3920/wmj2019.2556] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mycotoxins are toxic secondary metabolites produced by fungi. These compounds have different structures and target different organs, acting at different steps of biological processes inside the cell. Around 32 mycotoxins have been identified in fungal Penicillium spp. isolated from food and feed. Some of these species are important pathogens which contaminate food, such as maize, cereals, soybeans, sorghum, peanuts, among others. These microorganisms can be present in different steps of the food production process, such as plant growth, harvest, drying, elaboration, transport, and packaging. Although some Penicillium spp. are pathogens, some of them are used in elaboration of processed foods, such as cheese and sausages. This review summarises the Penicillium spp. mycotoxin toxicity, focusing mainly on the subgenus Penicillium, frequently found in food and feed. Toxicity is reviewed both in animal models and cultured cells. Finally, some aspects of their regulations are discussed.
Collapse
Affiliation(s)
- C. Otero
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andrés Bello, República 252, Santiago, Chile
| | - C. Arredondo
- Laboratorio de Neuroepigenética, Instituto de Ciencias Biomédicas (ICB), Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago, Chile
| | - A. Echeverría-Vega
- Centro de Investigación en Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Postgrado, Universidad Católica del Maule, Talca, Chile
| | - F. Gordillo-Fuenzalida
- Centro de Biotecnología de los Recursos Naturales (CENBIO), Laboratorio de Microbiología Aplicada, Facultad de Ciencias Agrarias y Forestales, Universidad Católica del Maule, Avda. San Miguel 3605, Talca, Chile
| |
Collapse
|
19
|
Luo H, Zheng J, Chen Y, Wang T, Zhang Z, Shan Y, Xu J, Yue M, Fang W, Li X. Utility Evaluation of Porcine Enteroids as PDCoV Infection Model in vitro. Front Microbiol 2020; 11:821. [PMID: 32390999 PMCID: PMC7191032 DOI: 10.3389/fmicb.2020.00821] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 04/06/2020] [Indexed: 12/13/2022] Open
Abstract
Porcine deltacoronavirus (PDCoV) is a novel emerging enteric coronavirus found in pigs. Intestinal enteroids, which partially recreate the structure and function of intestinal villi-crypts, have many physiological similarities to the intestinal tissues in vivo. Enteroids exhibit advantages in studying the interactions between intestines and enteric pathogens. To create a novel infection model for PDCoV, we developed an in vitro system to generate porcine intestinal enteroids from crypts of duodenum, jejunum, and ileum of pigs. Enterocytes, enteroendocrine cells, Paneth cells, stem cells, proliferating cells, and goblet cells were found in the differentiated enteroids. Replication of PDCoV was detected in the cultured enteroids by immunofluorescence and quantitative RT-PCR. Double immunofluorescence labeling demonstrated that PDCoV was present in Sox9-positive intestinal cells and Villin1-positive enterocytes. There were multiple cellular responses shown as changes of transcription of genes related to mucosal immunity, antiviral genes, and marker genes of stem cells and other cells in the enteroids infected with PDCoV. We conclude that the 2-D enteroids derived from porcine jejunum can be used as an in vitro multicellular model for the investigation of pathogenesis and host immune responses to porcine enteric pathogens, such as PDCoV.
Collapse
Affiliation(s)
- Hao Luo
- Zhejiang Provincial Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jingyou Zheng
- Zhejiang Provincial Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yunlu Chen
- Zhejiang Provincial Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Tingjun Wang
- Zhejiang Provincial Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Zhenning Zhang
- Zhejiang Provincial Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Ying Shan
- Zhejiang Provincial Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Jidong Xu
- Zhejiang Provincial Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Min Yue
- Zhejiang Provincial Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Weihuan Fang
- Zhejiang Provincial Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoliang Li
- Zhejiang Provincial Key Lab of Preventive Veterinary Medicine, Institute of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
20
|
Jiao LF, Dai TM, Zhong SQ, Jin M, Sun P, Zhou QC. Vibrio parahaemolyticus infection impaired intestinal barrier function and nutrient absorption in Litopenaeus vannamei. FISH & SHELLFISH IMMUNOLOGY 2020; 99:184-189. [PMID: 32035168 DOI: 10.1016/j.fsi.2020.02.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/14/2020] [Accepted: 02/05/2020] [Indexed: 06/10/2023]
Abstract
The intestine is the primary target of pathogenic microbes during invasion. However, the interaction of Vibrio parahaemolyticus (V. parahaemolyticus) with intestinal epithelial cells and its effects on the intestinal function of Litopenaeus vannamei (L. vannamei) are poorly studied. Therefore, the aim of this study was to investigate the influence of V. parahaemolyticus infection on intestinal barrier function and nutrient absorption in L. vannamei. In the present study, a total of 90 shrimp were randomly divided into two groups including the control group and V. parahaemolyticus infection group (final concentration of 1 × 105 CFU/mL), with three replicates per group. The result showed that compared with the control group, V. parahaemolyticus infection increased (P < 0.05) serum diamine oxidase activity and endotoxin quantification, and down-regulated (P < 0.05) the mRNA levels of intestinal peroxinectin, integrin, midline fasciclin at 48 h and 72 h; V. parahaemolyticus infection decreased (P < 0.05) the mRNA expression of intestinal amino acid transporter (CAT1, EAAT3 and ASCT1) and glucose transporter (SGLT-1, GLUT) at 24 h, 48 h and 72 h, and increased (P < 0.05) serum glucose and amino acid (Asp, Thr, Ser, Glu, Gly, Ala, Val, Ile, Leu, Tyr, Phe, Lys, His and Arg) concentration at 24 h. The results indicated that V. parahaemolyticus infection increased intestinal permeability, inhibited absorption of glucose and amino acid in L. vannamei.
Collapse
Affiliation(s)
- Le Fei Jiao
- Laboratory of Fish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Tian Meng Dai
- Laboratory of Fish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Sun Qian Zhong
- Ningbo Economic Technical Development Area Bolun Marine Surveyors Office, Ningbo, 315800, PR China
| | - Min Jin
- Laboratory of Fish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Peng Sun
- Laboratory of Fish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China
| | - Qi Cun Zhou
- Laboratory of Fish Nutrition, School of Marine Sciences, Ningbo University, Ningbo, 315211, PR China.
| |
Collapse
|
21
|
Segrist E, Cherry S. Using Diverse Model Systems to Define Intestinal Epithelial Defenses to Enteric Viral Infections. Cell Host Microbe 2020; 27:329-344. [PMID: 32164844 DOI: 10.1016/j.chom.2020.02.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The intestine is an essential physical and immunological barrier comprised of a monolayer of diverse and specialized epithelial cells that perform functions ranging from nutrient absorption to pathogen sensing and intestinal homeostasis. The intestinal barrier prevents translocation of intestinal microbes into internal compartments. The microbiota is comprised of a complex community largely populated by diverse bacterial species that provide metabolites, nutrients, and immune stimuli that promote intestinal and organismal health. Although commensal organisms promote health, enteric pathogens, including a diverse plethora of enteric viruses, cause acute and chronic diseases. The barrier epithelium plays fundamental roles in immune defenses against enteric viral infections by integrating diverse signals, including those from the microbiota, to prevent disease. Importantly, many model systems have contributed to our understanding of this complex interface. This review will focus on the antiviral mechanisms at play within the intestinal epithelium and how these responses are shaped by the microbiota.
Collapse
Affiliation(s)
- Elisha Segrist
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
22
|
Zhao X, Yang J, Ju Z, Wu J, Wang L, Lin H, Sun S. Clostridium butyricum Ameliorates Salmonella Enteritis Induced Inflammation by Enhancing and Improving Immunity of the Intestinal Epithelial Barrier at the Intestinal Mucosal Level. Front Microbiol 2020; 11:299. [PMID: 32180765 PMCID: PMC7059641 DOI: 10.3389/fmicb.2020.00299] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/10/2020] [Indexed: 12/20/2022] Open
Abstract
This study was aimed to investigate the effects of Clostridium butyricum (C. butyricum) immunity and intestinal epithelial barrier function at the intestinal mucosal level, by using Salmonella enteritidis (S. enteritidis) to infect specific-pathogen-free (SPF) chickens and intestinal epithelial cells (IEC). We found that C. butyricum could decrease cytokine levels (IFN-γ, IL-1β, IL-8, and TNF-α) via the TLR4-, MyD88-, and NF-κB-dependent pathways in intestinal tissues and intestinal epithelial cells. Additionally, C. butyricum could attenuate bacteria-induced intestinal damage and increase the expression level of muc-2 and ZO-1 in the intestine and intestinal epithelial cells. Furthermore, C. butyricum altered the intestinal microbial composition, increased the diversity of the bacterial communities in the cecum of Salmonella-infected chickens. In conclusion, C. butyricum effectively attenuated inflammation and epithelial barrier damage, altered the intestinal microbial composition, increased the diversity of the bacterial communities in the intestine of Salmonella-infected chickens. The result suggests that C. butyricum might be an effective and safe therapy for the treatment of Salmonella infection.
Collapse
Affiliation(s)
- Xiaonan Zhao
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China.,Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jie Yang
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Zijing Ju
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Jianmin Wu
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Lili Wang
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Hai Lin
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| | - Shuhong Sun
- College of Animal Science and Technology, Shandong Agricultural University, Tai'an, China
| |
Collapse
|
23
|
Kumar M, Singh P, Murugesan S, Vetizou M, McCulloch J, Badger JH, Trinchieri G, Al Khodor S. Microbiome as an Immunological Modifier. Methods Mol Biol 2020; 2055:595-638. [PMID: 31502171 PMCID: PMC8276114 DOI: 10.1007/978-1-4939-9773-2_27] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Humans are living ecosystems composed of human cells and microbes. The microbiome is the collection of microbes (microbiota) and their genes. Recent breakthroughs in the high-throughput sequencing technologies have made it possible for us to understand the composition of the human microbiome. Launched by the National Institutes of Health in USA, the human microbiome project indicated that our bodies harbor a wide array of microbes, specific to each body site with interpersonal and intrapersonal variabilities. Numerous studies have indicated that several factors influence the development of the microbiome including genetics, diet, use of antibiotics, and lifestyle, among others. The microbiome and its mediators are in a continuous cross talk with the host immune system; hence, any imbalance on one side is reflected on the other. Dysbiosis (microbiota imbalance) was shown in many diseases and pathological conditions such as inflammatory bowel disease, celiac disease, multiple sclerosis, rheumatoid arthritis, asthma, diabetes, and cancer. The microbial composition mirrors inflammation variations in certain disease conditions, within various stages of the same disease; hence, it has the potential to be used as a biomarker.
Collapse
Affiliation(s)
- Manoj Kumar
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar
| | - Parul Singh
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar
| | - Selvasankar Murugesan
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar
| | - Marie Vetizou
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - John McCulloch
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan H Badger
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Souhaila Al Khodor
- Division of Translational Medicine, Research Department, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
24
|
Wu C, Gao Y, Li S, Huang X, Bao X, Wang J, Zheng N. Modulation of intestinal epithelial permeability and mucin mRNA (MUC2, MUC5AC, and MUC5B) expression and protein secretion in Caco-2/HT29-MTX co-cultures exposed to aflatoxin M1, ochratoxin A, and zearalenone individually or collectively. Toxicol Lett 2019; 309:1-9. [DOI: 10.1016/j.toxlet.2019.03.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/25/2019] [Accepted: 03/18/2019] [Indexed: 12/24/2022]
|
25
|
Grabinger T, Glaus Garzon JF, Hausmann M, Geirnaert A, Lacroix C, Hennet T. Alleviation of Intestinal Inflammation by Oral Supplementation With 2-Fucosyllactose in Mice. Front Microbiol 2019; 10:1385. [PMID: 31275292 PMCID: PMC6593069 DOI: 10.3389/fmicb.2019.01385] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/03/2019] [Indexed: 12/30/2022] Open
Abstract
Milk oligosaccharides exert a prebiotic action that contributes to the development of the infant gut microbiota during lactation. Given that milk oligosaccharides remain intact after passage through stomach and small intestine, they can potentially influence the composition of the gut microbiota when ingested as dietary supplements after weaning. To address the regulatory effects of specific oligosaccharides in colitis linked to the microbiota composition, we have supplemented interleukin-10 null (Il10 -/-) mice with four fucosylated and sialylated oligosaccharides. We found that oral supplementation with 2-fucosyllactose significantly decreased the severity of colitis as displayed by reduced inflammatory marker expression, histological and diarrhea scores, an increased epithelial integrity and less pronounced colon shortening. Oral supplementation with 2-fucosyllactose led to a marked expansion of the commensal Ruminococcus gnavus, which was accompanied by an enhanced cecal concentration of propionate. Decreased activation of immune cells by R. gnavus was confirmed by reconstitution of antibiotic-treated Il10 -/- mice and by stimulation of dendritic cells in vitro. This study demonstrates that post-weaning administration of specific oligosaccharides can shift the composition of the gut microbiota to lessen chronic inflammation as observed in Il10 -/- mice. The expansion of R. gnavus sets a positive microbial environment at the cost of pro-inflammatory Gram-negative bacteria, thereby lowering intestinal inflammation.
Collapse
Affiliation(s)
- Thomas Grabinger
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Martin Hausmann
- Department of Gastroenterology and Hepatology, University Hospital Zurich - University of Zurich, Zurich, Switzerland
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Thierry Hennet
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
Ren Z, Guo C, Yu S, Zhu L, Wang Y, Hu H, Deng J. Progress in Mycotoxins Affecting Intestinal Mucosal Barrier Function. Int J Mol Sci 2019; 20:E2777. [PMID: 31174254 PMCID: PMC6600655 DOI: 10.3390/ijms20112777] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/12/2022] Open
Abstract
Mycotoxins, which are widely found in feed ingredients and human food, can exert harmful effects on animals and pose a serious threat to human health. As the first barrier against external pollutants, the intestinal mucosa is protected by a mechanical barrier, chemical barrier, immune barrier, and biological barrier. Firstly, mycotoxins can disrupt the mechanical barrier function of the intestinal mucosa, by destroying the morphology and tissue integrity of the intestinal epithelium. Secondly, mycotoxins can cause changes in the composition of mucin monosaccharides and the expression of intestinal mucin, which in turn affects mucin function. Thirdly, mycotoxins can cause damage to the intestinal mucosal immune barrier function. Finally, the microbiotas of animals closely interact with ingested mycotoxins. Based on existing research, this article reviews the effects of mycotoxins on the intestinal mucosal barrier and its mechanisms.
Collapse
Affiliation(s)
- Zhihua Ren
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Chaoyue Guo
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Shumin Yu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Ling Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Ya Wang
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Hui Hu
- The College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China.
| | - Junliang Deng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
27
|
Host NLRP6 exacerbates graft-versus-host disease independent of gut microbial composition. Nat Microbiol 2019; 4:800-812. [PMID: 30858572 PMCID: PMC6689241 DOI: 10.1038/s41564-019-0373-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 01/17/2019] [Indexed: 02/03/2023]
Abstract
Host NOD-like receptor family pyrin domain-containing 6 (NLRP6) regulates innate immune responses and gastrointestinal homeostasis. Its protective role in intestinal colitis and tumorigenesis is dependent on the host microbiome. Host innate immunity and microbial diversity also play a role in the severity of allogeneic immune-mediated gastrointestinal graft-versus-host disease (GVHD), the principal toxicity after allogeneic haematopoietic cell transplantation. Here, we examined the role of host NLRP6 in multiple murine models of allogeneic bone marrow transplantation. In contrast to its role in intestinal colitis, host NLRP6 aggravated gastrointestinal GVHD. The impact of host NLRP6 deficiency in mitigating GVHD was observed regardless of co-housing, antibiotic treatment or colonizing littermate germ-free wild-type and NLRP6-deficient hosts with faecal microbial transplantation from specific pathogen-free wild-type and Nlrp6-/- animals. Chimaera studies were performed to assess the role of NLRP6 expression on host haematopoietic and non-haematopoietic cells. The allogeneic [B6Ly5.2 → Nlrp6-/-] animals demonstrated significantly improved survival compared to the allogeneic [B6Ly5.2 → B6] animals, but did not alter the therapeutic graft-versus-tumour effects after haematopoietic cell transplantation. Our results unveil an unexpected, pathogenic role for host NLRP6 in gastrointestinal GVHD that is independent of variations in the intestinal microbiome and in contrast to its well-appreciated microbiome-dependent protective role in intestinal colitis and tumorigenesis.
Collapse
|
28
|
Modulation of Mucin ( MUC2, MUC5AC and MUC5B) mRNA Expression and Protein Production and Secretion in Caco-2/HT29-MTX Co-Cultures Following Exposure to Individual and Combined Aflatoxin M1 and Ochratoxin A. Toxins (Basel) 2019; 11:toxins11020132. [PMID: 30813459 PMCID: PMC6409803 DOI: 10.3390/toxins11020132] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/12/2019] [Accepted: 02/19/2019] [Indexed: 12/12/2022] Open
Abstract
Aflatoxin M1 (AFM1) and ochratoxin A (OTA), which widely coexist in milk, may pose a serious threat to human health. Mucin is a major component of the intestinal mucus layer, which plays an important role in maintaining intestinal mucosal homeostasis. However, the effect of mycotoxins AFM1 and OTA on intestinal mucin production is still not clear. This study aimed to investigate individual and interactive effects of mycotoxins AFM1 and OTA on the intestinal barrier and the mRNA expression of intestinal mucin (MUC2, MUC5AC and MUC5B) and on protein production in Caco-2/HT29-MTX cultures after 48 h of exposure. Our results show that individual mycotoxins and their mixtures significantly reduced intestinal cell viability and transepithelial electrical resistance (TEER) values, as well as significantly altered intestinal mucin mRNA expression and protein abundance. Moreover, OTA showed toxicity similar to AFM1 in cell viability and TEER value at the same concentration. When the two mycotoxins acted in combination, the synergistic effects observed in the assessment of cell viability and protein abundance in all mono- and co-cultures. In general, this study provides evidence that AFM1 and OTA can damage the intestine, and it contributes to optimized maximum permissible limits of mycotoxins in milk.
Collapse
|
29
|
Pisanu S, Cubeddu T, Cacciotto C, Pilicchi Y, Pagnozzi D, Uzzau S, Rocca S, Addis MF. Characterization of paucibacillary ileal lesions in sheep with subclinical active infection by Mycobacterium avium subsp. paratuberculosis. Vet Res 2018; 49:117. [PMID: 30514405 PMCID: PMC6278003 DOI: 10.1186/s13567-018-0612-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/13/2018] [Indexed: 01/10/2023] Open
Abstract
Paratuberculosis (PTB) or Johne's disease is a contagious enteritis of ruminants caused by Mycobacterium avium subsp. paratuberculosis (MAP). Ovine PTB is less understood than bovine PTB, especially concerning paucibacillary infection and its evolution into clinical disease. We combined shotgun proteomics, histopathology and immunohistochemistry for the characterization of ileal tissues collected from seven asymptomatic sheep negative to serum ELISA, positive to feces and tissue MAP IS900 and F57 PCR, histologically classified as paucibacillary, actively infected, together with 3 MAP-free controls (K). Following shotgun proteomics with label-free quantitation and differential analysis, 96 proteins were significantly changed in PTB vs K, and were mostly involved in immune defense processes and in the macrophage-MAP interaction. Principal component analysis (PCA) of protein abundances highlighted two PTB sample clusters, PTB1 and PTB2, indicating a dichotomy in their proteomic profiles. This was in line with the PCA of histopathology data and was related to features of type 2 (PTB1) and type 3a (PTB2) lesions, respectively. PTB2 proteomes differed more than PTB1 proteomes from K: 43 proteins changed significantly only in PTB2 and 11 only in PTB1. The differential proteins cathelicidin, haptoglobin, S100A8 and S100A9 were evaluated by immunohistochemistry. K tissues were negative to cathelicidin and haptoglobin and sparsely positive to S100A8 and S100A9. PTB tissues were positive to all four proteins, with significantly more cells in PTB2 than in PTB1. In conclusion, we described several pathways altered in paucibacillary PTB, highlighted some proteomic differences among paucibacillary PTB cases, and identified potential markers for disease understanding, staging, and detection.
Collapse
Affiliation(s)
- Salvatore Pisanu
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041, Alghero, Italy
| | - Tiziana Cubeddu
- Dipartimento di Medicina Veterinaria, Università degli Studi di Sassari, Via Vienna 2, 07100, Sassari, Italy
| | - Carla Cacciotto
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041, Alghero, Italy
| | - Ylenia Pilicchi
- Dipartimento di Medicina Veterinaria, Università degli Studi di Sassari, Via Vienna 2, 07100, Sassari, Italy
| | - Daniela Pagnozzi
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041, Alghero, Italy
| | - Sergio Uzzau
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041, Alghero, Italy.,Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Viale S. Pietro 43/B, 07100, Sassari, Italy
| | - Stefano Rocca
- Dipartimento di Medicina Veterinaria, Università degli Studi di Sassari, Via Vienna 2, 07100, Sassari, Italy
| | - Maria Filippa Addis
- Porto Conte Ricerche, SP 55 Porto Conte/Capo Caccia, Km 8.400, Loc. Tramariglio, 07041, Alghero, Italy. .,Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Via Celoria 10, 20133, Milan, Italy.
| |
Collapse
|
30
|
Implications of Inflammasomes in Human Diseases: NLRP3 Inflammasome and Animal Models. Cornea 2018; 37 Suppl 1:S86-S90. [DOI: 10.1097/ico.0000000000001717] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Kang SH, Hong SJ, Lee YK, Cho S. Oral Vaccine Delivery for Intestinal Immunity-Biological Basis, Barriers, Delivery System, and M Cell Targeting. Polymers (Basel) 2018; 10:E948. [PMID: 30960873 PMCID: PMC6403562 DOI: 10.3390/polym10090948] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/19/2022] Open
Abstract
Most currently available commercial vaccines are delivered by systemic injection. However, needle-free oral vaccine delivery is currently of great interest for several reasons, including the ability to elicit mucosal immune responses, ease of administration, and the relatively improved safety. This review summarizes the biological basis, various physiological and immunological barriers, current delivery systems with delivery criteria, and suggestions for strategies to enhance the delivery of oral vaccines. In oral vaccine delivery, basic requirements are the protection of antigens from the GI environment, targeting of M cells and activation of the innate immune response. Approaches to address these requirements aim to provide new vaccines and delivery systems that mimic the pathogen's properties, which are capable of eliciting a protective mucosal immune response and a systemic immune response and that make an impact on current oral vaccine development.
Collapse
Affiliation(s)
- Sung Hun Kang
- Department of Medical Sciences, College of Medicine, Hallym University, Chuncheon 24252, Korea.
| | - Seok Jin Hong
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong 18450, Korea.
| | - Yong-Kyu Lee
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Korea.
- 4D Biomaterials Center, Korea National University of Transportation, Jeungpyeong 27909, Korea.
| | - Sungpil Cho
- 4D Biomaterials Center, Korea National University of Transportation, Jeungpyeong 27909, Korea.
| |
Collapse
|
32
|
Pham VT, Seifert N, Richard N, Raederstorff D, Steinert RE, Prudence K, Mohajeri MH. The effects of fermentation products of prebiotic fibres on gut barrier and immune functions in vitro. PeerJ 2018; 6:e5288. [PMID: 30128177 PMCID: PMC6089210 DOI: 10.7717/peerj.5288] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 07/02/2018] [Indexed: 12/18/2022] Open
Abstract
The beneficial effects of prebiotic fibres on human health have been related to their capacities to alter the gut microbiota and modify the growth of beneficial microorganisms. It is long appreciated that bacterial metabolites affect the host’s physiology. The inner lining of the intestinal tract is the first level of interaction between the host and bacteria and their metabolites. Therefore, we set out to test the effects of five common dietary fibres (oat β-glucan 28%; oat β-glucan 94%; dried chicory root containing inulin 75%; xylo-oligosaccharide; inulin 90%) and maltodextrin, after fermentation by human gut microbiota in vitro, on measures of gut barrier integrity using a Caco-2/HT29-MTX co-culture as well as mucus production and immune parameters using HT29-MTX and HT29 cell models, respectively. Our data show that all fibres, fermentation products increased the tightness of the gut barrier with oat β-glucan 28% having the largest effect. Fermentation supernatants were tested also in models of the compromised gut barrier (leaky gut). After the addition of ethanol as basolateral stressor, only fermentation supernatant of oat β-glucan 28%, oat β-glucan 94% and maltodextrin improved the gut barrier integrity, while oat β-glucan 28% and dried chicory root containing inulin 75% significantly improved the gut barrier integrity after addition of rhamnolipids as apical stressor. Using the Luminex Technology, we demonstrated an important role of oat β-glucan fermentation products in modulating cytokine and chemokine productions. Furthermore, treating the goblet cells with effluent from xylo-oligosaccharide fermentation significantly increased mucus production. In summary, our data emphasize the potential positive effects of fermentation supernatant of dietary fibres on gut-related physiological outcomes and show that prebiotic fibres may have promising potential to induce specific gut health benefits.
Collapse
Affiliation(s)
- Van T Pham
- R&D Human Nutrition and Health, DSM Nutritional Products Ltd., Basel, Switzerland
| | - Nicole Seifert
- R&D Human Nutrition and Health, DSM Nutritional Products Ltd., Basel, Switzerland
| | - Nathalie Richard
- R&D Human Nutrition and Health, DSM Nutritional Products Ltd., Basel, Switzerland
| | - Daniel Raederstorff
- R&D Human Nutrition and Health, DSM Nutritional Products Ltd., Basel, Switzerland
| | - Robert E Steinert
- R&D Human Nutrition and Health, DSM Nutritional Products Ltd., Basel, Switzerland
| | - Kevin Prudence
- R&D Human Nutrition and Health, DSM Nutritional Products Ltd., Basel, Switzerland
| | - M Hasan Mohajeri
- R&D Human Nutrition and Health, DSM Nutritional Products Ltd., Basel, Switzerland
| |
Collapse
|
33
|
Freedman JC, Navarro MA, Morrell E, Beingesser J, Shrestha A, McClane BA, Uzal FA. Evidence that Clostridium perfringens Enterotoxin-Induced Intestinal Damage and Enterotoxemic Death in Mice Can Occur Independently of Intestinal Caspase-3 Activation. Infect Immun 2018; 86:e00931-17. [PMID: 29685988 PMCID: PMC6013662 DOI: 10.1128/iai.00931-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/15/2018] [Indexed: 01/13/2023] Open
Abstract
Clostridium perfringens enterotoxin (CPE) is responsible for the gastrointestinal symptoms of C. perfringens type A food poisoning and some cases of nonfoodborne gastrointestinal diseases, such as antibiotic-associated diarrhea. In the presence of certain predisposing medical conditions, this toxin can also be absorbed from the intestines to cause enterotoxemic death. CPE action in vivo involves intestinal damage, which begins at the villus tips. The cause of this CPE-induced intestinal damage is unknown, but CPE can induce caspase-3-mediated apoptosis in cultured enterocyte-like Caco-2 cells. Therefore, the current study evaluated whether CPE activates caspase-3 in the intestines and, if so, whether this effect is required for the development of intestinal tissue damage or enterotoxemic lethality. Using a mouse ligated small intestinal loop model, CPE was shown to cause intestinal caspase-3 activation in a dose- and time-dependent manner. Most of this caspase-3 activation occurred in epithelial cells shed from villus tips. However, CPE-induced caspase-3 activation occurred after the onset of tissue damage. Furthermore, inhibition of intestinal caspase-3 activity did not affect the onset of intestinal tissue damage. Similarly, inhibition of intestinal caspase-3 activity did not reduce CPE-induced enterotoxemic lethality in these mice. Collectively, these results demonstrate that caspase-3 activation occurs in the CPE-treated intestine but that this effect is not necessary for the development of CPE-induced intestinal tissue damage or enterotoxemic lethality.
Collapse
Affiliation(s)
- John C Freedman
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mauricio A Navarro
- California Animal Health and Food Safety Laboratory, San Bernardino Branch, School of Veterinary Medicine, University of California-Davis, San Bernardino, California, USA
| | - Eleonora Morrell
- California Animal Health and Food Safety Laboratory, San Bernardino Branch, School of Veterinary Medicine, University of California-Davis, San Bernardino, California, USA
| | - Juliann Beingesser
- California Animal Health and Food Safety Laboratory, San Bernardino Branch, School of Veterinary Medicine, University of California-Davis, San Bernardino, California, USA
| | - Archana Shrestha
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bruce A McClane
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Francisco A Uzal
- California Animal Health and Food Safety Laboratory, San Bernardino Branch, School of Veterinary Medicine, University of California-Davis, San Bernardino, California, USA
| |
Collapse
|
34
|
Chiu L, Bazin T, Truchetet ME, Schaeverbeke T, Delhaes L, Pradeu T. Protective Microbiota: From Localized to Long-Reaching Co-Immunity. Front Immunol 2017; 8:1678. [PMID: 29270167 PMCID: PMC5725472 DOI: 10.3389/fimmu.2017.01678] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/15/2017] [Indexed: 12/17/2022] Open
Abstract
Resident microbiota do not just shape host immunity, they can also contribute to host protection against pathogens and infectious diseases. Previous reviews of the protective roles of the microbiota have focused exclusively on colonization resistance localized within a microenvironment. This review shows that the protection against pathogens also involves the mitigation of pathogenic impact without eliminating the pathogens (i.e., “disease tolerance”) and the containment of microorganisms to prevent pathogenic spread. Protective microorganisms can have an impact beyond their niche, interfering with the entry, establishment, growth, and spread of pathogenic microorganisms. More fundamentally, we propose a series of conceptual clarifications in support of the idea of a “co-immunity,” where an organism is protected by both its own immune system and components of its microbiota.
Collapse
Affiliation(s)
- Lynn Chiu
- University of Bordeaux, CNRS, ImmunoConcept, UMR 5164, Bordeaux, France
| | - Thomas Bazin
- University of Bordeaux, INRA, Mycoplasmal and Chlamydial Infections in Humans, EA 3671, Bordeaux, France.,Department of Hepato-Gastroenterology, Bordeaux Hospital University Center, Pessac, France
| | | | - Thierry Schaeverbeke
- University of Bordeaux, INRA, Mycoplasmal and Chlamydial Infections in Humans, EA 3671, Bordeaux, France.,Department of Rheumatology, Bordeaux Hospital University Center, Bordeaux, France
| | - Laurence Delhaes
- Department of Parasitology and Mycology, Bordeaux Hospital University Center, Bordeaux, France.,University of Bordeaux, INSERM, Cardio-Thoracic Research Centre of Bordeaux, U1045, Bordeaux, France
| | - Thomas Pradeu
- University of Bordeaux, CNRS, ImmunoConcept, UMR 5164, Bordeaux, France
| |
Collapse
|
35
|
NLRP6: A Multifaceted Innate Immune Sensor. Trends Immunol 2017; 38:248-260. [PMID: 28214100 DOI: 10.1016/j.it.2017.01.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 12/31/2016] [Accepted: 01/12/2017] [Indexed: 01/27/2023]
Abstract
NLRP6, a member of the nucleotide-binding domain, leucine-rich repeat-containing (NLR) innate immune receptor family, regulates inflammation and host defense against microorganisms. Similar to other NLRs, NLRP6 not only participates in inflammasome formation, but is also involved in nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling regulation and facilitation of gastrointestinal antiviral effector functions. Additionally, NLRP6 contributes to the regulation of mucus secretion and antimicrobial peptide production, thereby impacting intestinal microbial colonization and associated microbiome-related infectious, autoinflammatory, metabolic, and neoplastic diseases. However, several of the mechanisms attributed to the functions of NLRP6 remain debatable, leaving open questions as to the relevant molecular mechanisms and interacting partners, and putative human relevance. We herein discuss recent findings related to NLRP6 activity, while highlighting outstanding questions and future perspectives in elucidating its roles in health and disease.
Collapse
|
36
|
Escherichia albertii, a novel human enteropathogen, colonizes rat enterocytes and translocates to extra-intestinal sites. PLoS One 2017; 12:e0171385. [PMID: 28178312 PMCID: PMC5298312 DOI: 10.1371/journal.pone.0171385] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/18/2017] [Indexed: 01/01/2023] Open
Abstract
Diarrhea is the second leading cause of death of children up to five years old in the developing countries. Among the etiological diarrheal agents are atypical enteropathogenic Escherichia coli (aEPEC), one of the diarrheagenic E. coli pathotypes that affects children and adults, even in developed countries. Currently, genotypic and biochemical approaches have helped to demonstrate that some strains classified as aEPEC are actually E. albertii, a recently recognized human enteropathogen. Studies on particular strains are necessary to explore their virulence potential in order to further understand the underlying mechanisms of E. albertii infections. Here we demonstrated for the first time that infection of fragments of rat intestinal mucosa is a useful tool to study the initial steps of E. albertii colonization. We also observed that an E. albertii strain can translocate from the intestinal lumen to Mesenteric Lymph Nodes and liver in a rat model. Based on our finding of bacterial translocation, we investigated how E. albertii might cross the intestinal epithelium by performing infections of M-like cells in vitro to identify the potential in vivo translocation route. Altogether, our approaches allowed us to draft a general E. albertii infection route from the colonization till the bacterial spreading in vivo.
Collapse
|
37
|
Salvo Romero E, Alonso Cotoner C, Pardo Camacho C, Casado Bedmar M, Vicario M. The intestinal barrier function and its involvement in digestive disease. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2017; 107:686-96. [PMID: 26541659 DOI: 10.17235/reed.2015.3846/2015] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The gastrointestinal mucosal surface is lined with epithelial cells representing an effective barrier made up with intercellular junctions that separate the inner and the outer environments, and block the passage of potentially harmful substances. However, epithelial cells are also responsible for the absorption of nutrients and electrolytes, hence a semipermeable barrier is required that selectively allows a number of substances in while keeping others out. To this end, the intestine developed the "intestinal barrier function", a defensive system involving various elements, both intra- and extracellular, that work in a coordinated way to impede the passage of antigens, toxins, and microbial byproducts, and simultaneously preserves the correct development of the epithelial barrier, the immune system, and the acquisition of tolerance against dietary antigens and the intestinal microbiota. Disturbances in the mechanisms of the barrier function favor the development of exaggerated immune responses; while exact implications remain unknown, changes in intestinal barrier function have been associated with the development of inflammatory conditions in the gastrointestinal tract. This review details de various elements of the intestinal barrier function, and the key molecular and cellular changes described for gastrointestinal diseases associated with dysfunction in this defensive mechanism.
Collapse
Affiliation(s)
| | | | | | | | - María Vicario
- Gastroenteroogia, Vall d'Hebron Institut de REcerca, España
| |
Collapse
|
38
|
Miyazaki T. Protective effects of lactic acid bacteria on influenza A virus infection. AIMS ALLERGY AND IMMUNOLOGY 2017. [DOI: 10.3934/allergy.2017.3.138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
39
|
Abstract
The cause of Crohn’s disease (CD) has posed a conundrum for at least a century. A large body of work coupled with recent technological advances in genome research have at last started to provide some of the answers. Initially this review seeks to explain and to differentiate between bowel inflammation in the primary immunodeficiencies that generally lead to very early onset diffuse bowel inflammation in humans and in animal models, and the real syndrome of CD. In the latter, a trigger, almost certainly enteric infection by one of a multitude of organisms, allows the faeces access to the tissues, at which stage the response of individuals predisposed to CD is abnormal. Direct investigation of patients’ inflammatory response together with genome-wide association studies (GWAS) and DNA sequencing indicate that in CD the failure of acute inflammation and the clearance of bacteria from the tissues, and from within cells, is defective. The retained faecal products result in the characteristic chronic granulomatous inflammation and adaptive immune response. In this review I will examine the contemporary evidence that has led to this understanding, and look for explanations for the recent dramatic increase in the incidence of this disease.
Collapse
|
40
|
Abstract
The cause of Crohn's disease (CD) has posed a conundrum for at least a century. A large body of work coupled with recent technological advances in genome research have at last started to provide some of the answers. Initially this review seeks to explain and to differentiate between bowel inflammation in the primary immunodeficiencies that generally lead to very early onset diffuse bowel inflammation in humans and in animal models, and the real syndrome of CD. In the latter, a trigger, almost certainly enteric infection by one of a multitude of organisms, allows the faeces access to the tissues, at which stage the response of individuals predisposed to CD is abnormal. Direct investigation of patients' inflammatory response together with genome-wide association studies (GWAS) and DNA sequencing indicate that in CD the failure of acute inflammation and the clearance of bacteria from the tissues, and from within cells, is defective. The retained faecal products result in the characteristic chronic granulomatous inflammation and adaptive immune response. In this review I will examine the contemporary evidence that has led to this understanding, and look for explanations for the recent dramatic increase in the incidence of this disease.
Collapse
|
41
|
Sansone CL, Cohen J, Yasunaga A, Xu J, Osborn G, Subramanian H, Gold B, Buchon N, Cherry S. Microbiota-Dependent Priming of Antiviral Intestinal Immunity in Drosophila. Cell Host Microbe 2016; 18:571-81. [PMID: 26567510 DOI: 10.1016/j.chom.2015.10.010] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/31/2015] [Accepted: 10/15/2015] [Indexed: 01/16/2023]
Abstract
Enteric pathogens must overcome intestinal defenses to establish infection. In Drosophila, the ERK signaling pathway inhibits enteric virus infection. The intestinal microflora also impacts immunity but its role in enteric viral infection is unknown. Here we show that two signals are required to activate antiviral ERK signaling in the intestinal epithelium. One signal depends on recognition of peptidoglycan from the microbiota, particularly from the commensal Acetobacter pomorum, which primes the NF-kB-dependent induction of a secreted factor, Pvf2. However, the microbiota is not sufficient to induce this pathway; a second virus-initiated signaling event involving release of transcriptional paused genes mediated by the kinase Cdk9 is also required for Pvf2 production. Pvf2 stimulates antiviral immunity by binding to the receptor tyrosine kinase PVR, which is necessary and sufficient for intestinal ERK responses. These findings demonstrate that sensing of specific commensals primes inflammatory signaling required for epithelial responses that restrict enteric viral infections.
Collapse
Affiliation(s)
- Christine L Sansone
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Jonathan Cohen
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Ari Yasunaga
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Jie Xu
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Greg Osborn
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Harry Subramanian
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Beth Gold
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Nicolas Buchon
- Department of Entomology, Cornell University, Ithaca, NY 14853, USA
| | - Sara Cherry
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
42
|
Yarandi SS, Peterson DA, Treisman GJ, Moran TH, Pasricha PJ. Modulatory Effects of Gut Microbiota on the Central Nervous System: How Gut Could Play a Role in Neuropsychiatric Health and Diseases. J Neurogastroenterol Motil 2016; 22:201-12. [PMID: 27032544 PMCID: PMC4819858 DOI: 10.5056/jnm15146] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 01/12/2016] [Accepted: 01/27/2016] [Indexed: 12/11/2022] Open
Abstract
Gut microbiome is an integral part of the Gut-Brain axis. It is becoming increasingly recognized that the presence of a healthy and diverse gut microbiota is important to normal cognitive and emotional processing. It was known that altered emotional state and chronic stress can change the composition of gut microbiome, but it is becoming more evident that interaction between gut microbiome and central nervous system is bidirectional. Alteration in the composition of the gut microbiome can potentially lead to increased intestinal permeability and impair the function of the intestinal barrier. Subsequently, neuro-active compounds and metabolites can gain access to the areas within the central nervous system that regulate cognition and emotional responses. Deregulated inflammatory response, promoted by harmful microbiota, can activate the vagal system and impact neuropsychological functions. Some bacteria can produce peptides or short chain fatty acids that can affect gene expression and inflammation within the central nervous system. In this review, we summarize the evidence supporting the role of gut microbiota in modulating neuropsychological functions of the central nervous system and exploring the potential underlying mechanisms.
Collapse
Affiliation(s)
- Shadi S Yarandi
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daniel A Peterson
- Division of Immunology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Glen J Treisman
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Timothy H Moran
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Pankaj J Pasricha
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
43
|
Lactobacillus rhamnosus GG modulates intestinal mucosal barrier and inflammation in mice following combined dietary exposure to deoxynivalenol and zearalenone. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.01.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
44
|
Vijendravarma RK, Narasimha S, Chakrabarti S, Babin A, Kolly S, Lemaitre B, Kawecki TJ. Gut physiology mediates a trade‐off between adaptation to malnutrition and susceptibility to food‐borne pathogens. Ecol Lett 2015; 18:1078-86. [DOI: 10.1111/ele.12490] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/28/2015] [Accepted: 07/12/2015] [Indexed: 12/12/2022]
Affiliation(s)
| | - Sunitha Narasimha
- Department of Ecology and Evolution University of Lausanne CH 1015 Lausanne Switzerland
| | | | - Aurelie Babin
- Department of Ecology and Evolution University of Lausanne CH 1015 Lausanne Switzerland
| | - Sylvain Kolly
- Department of Ecology and Evolution University of Lausanne CH 1015 Lausanne Switzerland
| | - Bruno Lemaitre
- Global Health Institute EPFL CH 1015 Lausanne Switzerland
| | - Tadeusz J. Kawecki
- Department of Ecology and Evolution University of Lausanne CH 1015 Lausanne Switzerland
| |
Collapse
|
45
|
Abstract
BACKGROUND The role of muscarinic receptors in mucosal homeostasis, response to enteric pathogens, and modulation of immune cell function is undefined. METHODS The contribution of type 3 muscarinic receptors (M3R) to mucosal homeostasis within the colon and host defense against Citrobacter rodentium was determined in uninfected and C. rodentium-infected WT and M3R-deficient (Chrm3) mice. In addition, WT and Chrm3 bone marrow-derived macrophages were studied to determine the ability of M3R to modulate macrophage phenotype and function. RESULTS In Chrm3 mice, clearance of C. rodentium was delayed despite an amplified TH1/TH17 response. Delayed clearance of C. rodentium from Chrm3 mice was associated with prolonged adherence of bacteria to colonic mucosa, decreased goblet cell number, and decreased mucin 2 gene expression. Treatment of bone marrow-derived macrophages with bethanechol, a muscarinic-selective agonist, induced a classically activated macrophage phenotype, which was dependent on M3R expression. Chrm3 bone marrow-derived macrophages retained their ability to attain a classically activated macrophage phenotype when treated with the TH1 cytokine IFN-γ. CONCLUSIONS In Chrm3 mice, mucin production is attenuated and is associated with prolonged adherence of C. rodentium to colonic mucosa. The immune response, as characterized by production of TH1/TH17 cytokines, in C. rodentium-infected Chrm3 mice is intact. In addition, M3R activity promotes the development of classically activated macrophages. Our data establish a role for M3R in host defense against C. rodentium through effects on goblet cell mucus production and in the modulation of macrophage phenotype and function.
Collapse
|
46
|
Forbester JL, Goulding D, Vallier L, Hannan N, Hale C, Pickard D, Mukhopadhyay S, Dougan G. Interaction of Salmonella enterica Serovar Typhimurium with Intestinal Organoids Derived from Human Induced Pluripotent Stem Cells. Infect Immun 2015; 83:2926-34. [PMID: 25964470 PMCID: PMC4468523 DOI: 10.1128/iai.00161-15] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/24/2015] [Indexed: 11/20/2022] Open
Abstract
The intestinal mucosa forms the first line of defense against infections mediated by enteric pathogens such as salmonellae. Here we exploited intestinal "organoids" (iHOs) generated from human induced pluripotent stem cells (hIPSCs) to explore the interaction of Salmonella enterica serovar Typhimurium with iHOs. Imaging and RNA sequencing were used to analyze these interactions, and clear changes in transcriptional signatures were detected, including altered patterns of cytokine expression after the exposure of iHOs to bacteria. S. Typhimurium microinjected into the lumen of iHOs was able to invade the epithelial barrier, with many bacteria residing within Salmonella-containing vacuoles. An S. Typhimurium invA mutant defective in the Salmonella pathogenicity island 1 invasion apparatus was less capable of invading the iHO epithelium. Hence, we provide evidence that hIPSC-derived organoids are a promising model of the intestinal epithelium for assessing interactions with enteric pathogens.
Collapse
Affiliation(s)
- Jessica L Forbester
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - David Goulding
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Ludovic Vallier
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory, Department of Surgery, West Forvie Site, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas Hannan
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory, Department of Surgery, West Forvie Site, University of Cambridge, Cambridge, United Kingdom
| | - Christine Hale
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Derek Pickard
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | | | - Gordon Dougan
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, United Kingdom
| |
Collapse
|
47
|
Increasing fat content from 20 to 45 wt% in a complex diet induces lower endotoxemia in parallel with an increased number of intestinal goblet cells in mice. Nutr Res 2015; 35:346-56. [PMID: 25687164 DOI: 10.1016/j.nutres.2015.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 01/12/2015] [Accepted: 01/15/2015] [Indexed: 12/18/2022]
Abstract
The impacts of high-fat diets (HFDs) on the onset of metabolic endotoxemia and low-grade inflammation are well established in rodent models. However, the dose-effect of dietary lipid intakes on these parameters is not known. We hypothesized that increasing dietary lipid amounts could be linked to parallel increases of endotoxemia, low-grade inflammation, and metabolic and intestinal alterations. Six-week-old male C57BL/6J mice were fed a low-fat diet (LFD, 2.6 wt% of lipids), a moderate HFD (mHFD, 22 wt% of lipids), or a very HFD (vHFD, 45 wt% of lipids) formulated mainly using chow ingredients and milk fat. After 12 weeks, white adipose tissues, liver, intestine, distal colon contents, and plasma were collected. Only vHFD mice significantly increased body weight and fat mass vs LFD mice. This was associated with increases of plasma concentrations of triglycerides, leptin and adiponectin, and liver lipids. No such differences were observed between LFD and mHFD mice. However, mHFD developed metabolic endotoxemia and inflammation, unlike vHFD mice. In turn, vHFD mice showed more goblet cells in all intestine segments vs both other groups and a decrease of Bacteroides-Prevotella in their microbiota vs LFD mice. Finally, mHFD mice colon exhibited a decrease in lactobacilli and in the levels of occludin phosphorylation. Altogether, using complex HFD, no associations were observed between dietary lipid amounts and the magnitude of endotoxemia, inflammation, and physiological alterations developed. These results reveal the impact of the diet composition on intestinal goblet cells and mucus coat, bringing new insights about further consequences on HFD-induced metabolic disorders.
Collapse
|
48
|
Intestinal barrier function and the brain-gut axis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 817:73-113. [PMID: 24997030 DOI: 10.1007/978-1-4939-0897-4_4] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The luminal-mucosal interface of the intestinal tract is the first relevant location where microorganism-derived antigens and all other potentially immunogenic particles face the scrutiny of the powerful mammalian immune system. Upon regular functioning conditions, the intestinal barrier is able to effectively prevent most environmental and external antigens to interact openly with the numerous and versatile elements that compose the mucosal-associated immune system. This evolutionary super system is capable of processing an astonishing amount of antigens and non-immunogenic particles, approximately 100 tons in one individual lifetime, only considering food-derived components. Most important, to develop oral tolerance and proper active immune responses needed to prevent disease and inflammation, this giant immunogenic load has to be managed in a way that physiological inflammatory balance is constantly preserved. Adequate functioning of the intestinal barrier involves local and distant regulatory networks integrating the so-called brain-gut axis. Along this complex axis both brain and gut structures participate in the processing and execution of response signals to external and internal changes coming from the digestive tract, using multidirectional pathways to communicate. Dysfunction of brain-gut axis facilitates malfunctioning of the intestinal barrier, and vice versa, increasing the risk of uncontrolled immunological reactions that may trigger mucosal and brain low-grade inflammation, a putative first step to the initiation of more permanent gut disorders. In this chapter, we describe the structure, function and interactions of intestinal barrier, microbiota and brain-gut axis in both healthy and pathological conditions.
Collapse
|
49
|
NLRP6 inflammasome orchestrates the colonic host-microbial interface by regulating goblet cell mucus secretion. Cell 2014; 156:1045-59. [PMID: 24581500 PMCID: PMC4017640 DOI: 10.1016/j.cell.2014.01.026] [Citation(s) in RCA: 539] [Impact Index Per Article: 53.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 11/12/2013] [Accepted: 01/10/2014] [Indexed: 02/07/2023]
Abstract
Mucus production by goblet cells of the large intestine serves as a crucial antimicrobial protective mechanism at the interface between the eukaryotic and prokaryotic cells of the mammalian intestinal ecosystem. However, the regulatory pathways involved in goblet cell-induced mucus secretion remain largely unknown. Here, we demonstrate that the NLRP6 inflammasome, a recently described regulator of colonic microbiota composition and biogeographical distribution, is a critical orchestrator of goblet cell mucin granule exocytosis. NLRP6 deficiency leads to defective autophagy in goblet cells and abrogated mucus secretion into the large intestinal lumen. Consequently, NLRP6 inflammasome-deficient mice are unable to clear enteric pathogens from the mucosal surface, rendering them highly susceptible to persistent infection. This study identifies an innate immune regulatory pathway governing goblet cell mucus secretion, linking nonhematopoietic inflammasome signaling to autophagy and highlighting the goblet cell as a critical innate immune player in the control of intestinal host-microbial mutualism. PAPERCLIP:
Collapse
|
50
|
Rogier EW, Frantz AL, Bruno MEC, Kaetzel CS. Secretory IgA is Concentrated in the Outer Layer of Colonic Mucus along with Gut Bacteria. Pathogens 2014; 3:390-403. [PMID: 25437806 PMCID: PMC4243452 DOI: 10.3390/pathogens3020390] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 04/22/2014] [Accepted: 04/24/2014] [Indexed: 12/15/2022] Open
Abstract
Antibodies of the secretory IgA (SIgA) class comprise the first line of antigen-specific immune defense, preventing access of commensal and pathogenic microorganisms and their secreted products into the body proper. In addition to preventing infection, SIgA shapes the composition of the gut microbiome. SIgA is transported across intestinal epithelial cells into gut secretions by the polymeric immunoglobulin receptor (pIgR). The epithelial surface is protected by a thick network of mucus, which is composed of a dense, sterile inner layer and a loose outer layer that is colonized by commensal bacteria. Immunofluorescence microscopy of mouse and human colon tissues demonstrated that the SIgA co-localizes with gut bacteria in the outer mucus layer. Using mice genetically deficient for pIgR and/or mucin-2 (Muc2, the major glycoprotein of intestinal mucus), we found that Muc2 but not SIgA was necessary for excluding gut bacteria from the inner mucus layer in the colon. Our findings support a model whereby SIgA is anchored in the outer layer of colonic mucus through combined interactions with mucin proteins and gut bacteria, thus providing immune protection against pathogens while maintaining a mutually beneficial relationship with commensals.
Collapse
Affiliation(s)
- Eric W Rogier
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA.
| | - Aubrey L Frantz
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA.
| | - Maria E C Bruno
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA.
| | - Charlotte S Kaetzel
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|