451
|
Liu Y, Zheng JW, Peng XC, Li HY, Huang L, Li DW, Liu JS, Yang WD. Changes in colonic microbiotas in rat after long-term exposure to low dose of okadaic acid. CHEMOSPHERE 2020; 254:126874. [PMID: 32361543 DOI: 10.1016/j.chemosphere.2020.126874] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/05/2020] [Accepted: 04/21/2020] [Indexed: 06/11/2023]
Abstract
Okadaic acid (OA), one of the most important phycotoxins, is widely distributed around the world, concerning diarrheic shellfish poisoning (DSP), and even colorectal cancer. Here, we found that long-term exposure of OA at a low dose (80 μg kg-1 body weight) had certain effects on colonic microbiotas and tract in rat. In the OA-exposed rat, colonic epithelium layer was damaged, and relative abundance of some microbiotas were significantly changed, especially genera in Clostridiales. However, no intestinal inflammation or significant disease was observed. Combined with the increase in relative abundance of some genera in Clostridiales induced by OA in the fermentation experiment, we proposed that OA could cause damage to the intestinal epithelium and increase the relative abundance of pathogenic bacteria, thereby increasing the probability of contact between intestinal epithelium and pathogenic bacteria and leading to an easier pathogenicity.
Collapse
Affiliation(s)
- Yang Liu
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Jian-Wei Zheng
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xi-Chun Peng
- Department of Food Science and Engineering, Jinan University, Guangzhou, 510630, China
| | - Hong-Ye Li
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Lu Huang
- Department of Pathology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Da-Wei Li
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Jie-Sheng Liu
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Wei-Dong Yang
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
452
|
Shini S, Zhang D, Aland RC, Li X, Dart PJ, Callaghan MJ, Speight RE, Bryden WL. Probiotic Bacillus amyloliquefaciens H57 ameliorates subclinical necrotic enteritis in broiler chicks by maintaining intestinal mucosal integrity and improving feed efficiency. Poult Sci 2020; 99:4278-4293. [PMID: 32867972 PMCID: PMC7597999 DOI: 10.1016/j.psj.2020.05.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/10/2020] [Accepted: 05/22/2020] [Indexed: 01/24/2023] Open
Abstract
Subclinical necrotic enteritis (NE) was induced in broiler chicks using a high dose of Eimeria spp. vaccine in the drinking water on day 9, and Clostridium perfringens (Cp) culture mixed in the feed on days 14 and 15. The aim was to evaluate the effects of probiotic Bacillus amyloliquefaciens strain H57 (H57) in preventing NE in chicks. Day-old Ross 308, male broilers were weighed and randomly assigned to 6 treatment groups (6 replicate cages/treatment and 8 birds/cage). Birds in group 1 (control) were fed the basal wheat-soybean diet without H57 or NE infection; in group 2 (Eimeria) were treated with Eimeria alone; in group 3 (Cp) were treated with Cp alone; in group 4 (NE) received both Eimeria and Cp; in group 5 (NE-H57) received NE infection and H57; and group 6 (H57) received H57. The basal diet of chicks in groups 5 and 6 was supplemented with H57 at a density of 2 × 108 spores/g feed from 1 D of age. On day 21, there were no significant treatment effects on BW and feed intake between control and H57 birds. However, on day 21, the feed conversion ratio of NE-H57 birds was significantly improved when compared with NE birds (1.28 vs. 1.36; P < 0.001). Birds challenged with NE had a higher occurrence of pasty vent than birds infected with either Eimeria, Cp, or NE-H57 (41 vs. 27 vs. 29 vs. 19%, respectively; P < 0.001). Intestinal lesion scores of NE birds were also higher than those of Eimeria, Cp, and NE-H57 birds (5.67 vs. 2.56 vs. 2.78 vs. 2.10, respectively; P < 0.001) and correlated with pasty vent (Pearson's r = 0.56; P < 0.001). Microscopic evaluation showed mucosal damage and necrosis in NE birds. In contrast, villi from NE-H57 birds were normal, with no damage or infiltration with Eimeria or Cp. H57 appears to be effective in challenged birds, as it maintained epithelial barrier integrity and improved feed efficiency.
Collapse
Affiliation(s)
- S Shini
- School of Agriculture & Food Sciences, University of Queensland, Gatton Queensland 4343, Australia.
| | - D Zhang
- School of Agriculture & Food Sciences, University of Queensland, Gatton Queensland 4343, Australia
| | - R C Aland
- School of Biomedical Sciences, University of Queensland, St Lucia Queensland 4071, Australia
| | - X Li
- School of Agriculture & Food Sciences, University of Queensland, Gatton Queensland 4343, Australia
| | - P J Dart
- School of Agriculture & Food Sciences, University of Queensland, Gatton Queensland 4343, Australia
| | - M J Callaghan
- Ridley AgriProducts Pty Ltd, Toowong, Queensland 4066, Australia
| | - R E Speight
- Queensland University of Technology, Brisbane, Queensland 4000, Australia
| | - W L Bryden
- School of Agriculture & Food Sciences, University of Queensland, Gatton Queensland 4343, Australia
| |
Collapse
|
453
|
Hassan M, Moghadamrad S, Sorribas M, Muntet SG, Kellmann P, Trentesaux C, Fraudeau M, Nanni P, Wolski W, Keller I, Hapfelmeier S, Shroyer NF, Wiest R, Romagnolo B, De Gottardi A. Paneth cells promote angiogenesis and regulate portal hypertension in response to microbial signals. J Hepatol 2020; 73:628-639. [PMID: 32205193 DOI: 10.1016/j.jhep.2020.03.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/27/2020] [Accepted: 03/13/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Paneth cells (PCs) synthesize and secrete antimicrobial peptides that are key mediators of host-microbe interactions, establishing a balance between intestinal microflora and enteric pathogens. We observed that their number increases in experimental portal hypertension and aimed to investigate the mechanisms by which these cells can contribute to the regulation of portal pressure. METHODS We first treated Math1Lox/LoxVilcreERT2 mice with tamoxifen to induce the complete depletion of intestinal PCs. Subsequently, we performed partial portal vein or bile duct ligation. We then studied the effects of these interventions on hemodynamic parameters, proliferation of blood vessels and the expression of genes regulating angiogenesis. Intestinal organoids were cultured and exposed to different microbial products to study the composition of their secreted products (by proteomics) and their effects on the proliferation and tube formation of endothelial cells (ECs). In vivo confocal laser endomicroscopy was used to confirm the findings on blood vessel proliferation. RESULTS Portal hypertension was significantly attenuated in PC-depleted mice compared to control mice and was associated with a decrease in portosystemic shunts. Depletion of PCs also resulted in a significantly decreased density of blood vessels in the intestinal wall and mesentery. Furthermore, we observed reduced expression of intestinal genes regulating angiogenesis in Paneth cell depleted mice using arrays and next generation sequencing. Tube formation and wound healing responses were significantly decreased in ECs treated with conditioned media from PC-depleted intestinal organoids exposed to intestinal microbiota-derived products. Proteomic analysis of conditioned media in the presence of PCs revealed an increase in factors regulating angiogenesis and additional metabolic processes. In vivo endomicroscopy showed decreased vascular proliferation in the absence of PCs. CONCLUSIONS These results suggest that in response to intestinal flora and microbiota-derived factors, PCs secrete not only antimicrobial peptides, but also pro-angiogenic signaling molecules, thereby promoting intestinal and mesenteric angiogenesis and regulating portal hypertension. LAY SUMMARY Paneth cells are present in the lining of the small intestine. They prevent the passage of bacteria from the intestine into the blood circulation by secreting substances to fight bacteria. In this paper, we discovered that these substances not only act against bacteria, but also increase the quantity of blood vessels in the intestine and blood pressure in the portal vein. This is important, because high blood pressure in the portal vein may result in several complications which could be targeted with novel approaches.
Collapse
Affiliation(s)
- Mohsin Hassan
- Department for Biomedical Research, Hepatology, University of Bern, Switzerland
| | - Sheida Moghadamrad
- Department for Biomedical Research, Hepatology, University of Bern, Switzerland; Inselspital, Hepatology, University of Bern, Switzerland (Clinic of Visceral Surgery and Medicine, Inselspital, Berne, Switzerland)
| | - Marcel Sorribas
- Department for Biomedical Research, Gastroenterology, University of Bern, Switzerland
| | - Sergi G Muntet
- Department for Biomedical Research, Hepatology, University of Bern, Switzerland
| | - Philipp Kellmann
- Department for Biomedical Research, Hepatology, University of Bern, Switzerland
| | - Coralie Trentesaux
- INSERM, U106, Institut Cochin, F-75014 Paris, France; CNRS, UMR8104, F-75014 Paris, France; Université paris Descartes Sorbonne paris Cité, Paris, France
| | - Marie Fraudeau
- INSERM, U106, Institut Cochin, F-75014 Paris, France; CNRS, UMR8104, F-75014 Paris, France; Université paris Descartes Sorbonne paris Cité, Paris, France
| | - Paolo Nanni
- Functional Genomic Centre, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Witold Wolski
- Functional Genomic Centre, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Irene Keller
- Department for Biomedical Research and Swiss Institute of Bioinformatics, University of Bern, Switzerland
| | | | - Noah F Shroyer
- Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - Reiner Wiest
- Department for Biomedical Research, Gastroenterology, University of Bern, Switzerland; Inselspital, Hepatology, University of Bern, Switzerland (Clinic of Visceral Surgery and Medicine, Inselspital, Berne, Switzerland)
| | - Beatrice Romagnolo
- INSERM, U106, Institut Cochin, F-75014 Paris, France; CNRS, UMR8104, F-75014 Paris, France; Université paris Descartes Sorbonne paris Cité, Paris, France
| | - Andrea De Gottardi
- Department for Biomedical Research, Hepatology, University of Bern, Switzerland; Inselspital, Hepatology, University of Bern, Switzerland (Clinic of Visceral Surgery and Medicine, Inselspital, Berne, Switzerland); Gastroenterology and Hepatology, Ente Ospedaliero Cantonale, Università della Svizzera Italiana, Lugano, Switzerland.
| |
Collapse
|
454
|
Zhang Y, Thanou M, Vllasaliu D. Exploiting disease-induced changes for targeted oral delivery of biologics and nanomedicines in inflammatory bowel disease. Eur J Pharm Biopharm 2020; 155:128-138. [PMID: 32853696 DOI: 10.1016/j.ejpb.2020.08.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/21/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic and progressive disorder with destructive inflammation in the gastrointestinal tract (GIT). Biologics have changed the management of IBD, but have serious limitations, which is associated with their systemic administration via injection. Oral administration is the most accepted route of drug administration. However, the physiological barriers of the GIT pose significant challenges for oral administration of biologics, making this route of administration currently unavailable. The status of tissue barriers to oral drug delivery is altered in IBD. This may bring more challenges, but also present opportunities for oral delivery of biologics. This article provides an overview of disease-induced alterations of GIT barriers in IBD and discusses challenges, opportunities and commonly-utilised strategies for oral delivery of complex therapeutics, including biologics and nanomedicines.
Collapse
Affiliation(s)
- Yunyue Zhang
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London SE1 9NH, United Kingdom.
| | - Maya Thanou
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London SE1 9NH, United Kingdom.
| | - Driton Vllasaliu
- Institute of Pharmaceutical Science, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London SE1 9NH, United Kingdom.
| |
Collapse
|
455
|
Chénard T, Prévost K, Dubé J, Massé E. Immune System Modulations by Products of the Gut Microbiota. Vaccines (Basel) 2020; 8:vaccines8030461. [PMID: 32825559 PMCID: PMC7565937 DOI: 10.3390/vaccines8030461] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022] Open
Abstract
The gut microbiota, which consists of all bacteria, viruses, fungus, and protozoa living in the intestine, and the immune system have co-evolved in a symbiotic relationship since the origin of the immune system. The bacterial community forming the microbiota plays an important role in the regulation of multiple aspects of the immune system. This regulation depends, among other things, on the production of a variety of metabolites by the microbiota. These metabolites range from small molecules to large macro-molecules. All types of immune cells from the host interact with these metabolites resulting in the activation of different pathways, which result in either positive or negative responses. The understanding of these pathways and their modulations will help establish the microbiota as a therapeutic target in the prevention and treatment of a variety of immune-related diseases.
Collapse
|
456
|
Xie M, Zhang L, Li L, Fan M, Hou L. MiR-339 attenuates LPS-induced intestinal epithelial cells inflammatory responses and apoptosis by targeting TLR4. Genes Genomics 2020; 42:1097-1105. [PMID: 32757169 DOI: 10.1007/s13258-020-00977-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 07/22/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Intestinal epithelial cells are important for defending against pathogen infection. LPS is an endotoxin that is highly antigenic and cytotoxic produced by bacteria. LPS disrupts the intestine epithelium integrity and induced the intestinal epithelial cell inflammation and apoptosis. Our previous study has predicted the function of exosome miRNAs through bioinformatics analysis, and we found that miR-339 had a potential function in cell inflammation response. To our knowledge, no published paper has demonstrated the miR-339 function in protecting the intestine epithelium against bacterial infection. OBJECTIVE The objective of this study is to evaluate the miR-339 function in regulating intestinal epithelial cells to defend against bacterial infection through biological experiments and bioinformatics analyses. METHODS Through the miR-339 transfection experiment and TLR4 interfering experiment, we evaluated the function of miR-339 and TLR4 in the process of inflammatory responses and apoptosis. Through Bioinformatics analyses and dual-luciferase reporter experiment, we identified the target gene of miR-339. RESULTS miR-339 attenuates LPS-induced intestinal epithelial cells inflammatory responses through the TLR4/NF-κB signaling pathway and inhibited LPS-induced apoptosis through the P53 signaling pathway. TLR4 is the target gene of miR-339. TLR4 reduced LPS-induced proinflammatory responses and apoptosis. CONCLUSIONS In conclusion, miR-339 protected the intestine epithelial cells from LPS-induced cell inflammation and apoptosis through targeting TLR4. This study expanded our understanding of how miRNAs and genes work collaboratively in regulating intestinal epithelial cells to defend against bacterial infection.
Collapse
Affiliation(s)
- Meiying Xie
- Collaborative Innovation Center of Plant Pest Management and Bioenvironmental Health Application Technology, Guangdong Eco-Engineering Polytechnic, 297 Guangshan First Road, Tianhe District, Guangzhou, 510520, Guangdong, China
| | - Lina Zhang
- Collaborative Innovation Center of Plant Pest Management and Bioenvironmental Health Application Technology, Guangdong Eco-Engineering Polytechnic, 297 Guangshan First Road, Tianhe District, Guangzhou, 510520, Guangdong, China
| | - Luoye Li
- Collaborative Innovation Center of Plant Pest Management and Bioenvironmental Health Application Technology, Guangdong Eco-Engineering Polytechnic, 297 Guangshan First Road, Tianhe District, Guangzhou, 510520, Guangdong, China
| | - Minhuan Fan
- Collaborative Innovation Center of Plant Pest Management and Bioenvironmental Health Application Technology, Guangdong Eco-Engineering Polytechnic, 297 Guangshan First Road, Tianhe District, Guangzhou, 510520, Guangdong, China
| | - Lianjie Hou
- Qingyuan City People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, Guangdong, China.
| |
Collapse
|
457
|
Fu R, Niu R, Li R, Yue B, Zhang X, Cao Q, Wang J, Sun Z. Fluoride-Induced Alteration in the Diversity and Composition of Bacterial Microbiota in Mice Colon. Biol Trace Elem Res 2020; 196:537-544. [PMID: 31741202 DOI: 10.1007/s12011-019-01942-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/18/2019] [Indexed: 01/20/2023]
Abstract
Fluoride, as an environmental toxin, causes damage to intestinal mucosa. It may promote pathogen infection by increasing the intestinal mucosa permeability. In this study, the colonic fecal samples from the control group (C group, 0 mg/L NaF for 60 days) and the fluoride group (F group, 100 mg/L NaF for 60 days) were subjected to high-throughput 16S rRNA sequencing to verify the effects of fluoride on the colonic flora of animals. Results revealed a total of 253 operative taxonomical units (OTUs) in two groups, and 22 unique OTUs occurred in the F group. Fluoride increased the microbiota diversity and species richness of the colon. Concretely, the abundance of the Tenericutes was increased at the level of the phyla in the F group. In addition, in the F group, significant differences at the genus level were observed in Faecalibaculum, Alloprevotella, [Eubacterium]_xylanophilum_group, Prevotellaceae_UCG-001, and Ruminiclostridium_9, compared to the C group. Among them, except for the reduction in Faecalibaculum, the other four bacteria were increased in the F group. In summary, the intestinal microbial composition of mice was reconstituted by the presence of fluoride, and the significantly changing bacteria may partly account for the pathogenesis of fluoride-induced intestinal dysfunction.
Collapse
Affiliation(s)
- Rong Fu
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, Shanxi, China
| | - Ruiyan Niu
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, Shanxi, China
| | - Rui Li
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, Shanxi, China
| | - Baijuan Yue
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, Shanxi, China
| | - Xuhua Zhang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, Shanxi, China
| | - Qiqi Cao
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, Shanxi, China
| | - Jundong Wang
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, Shanxi, China
| | - Zilong Sun
- Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, Jinzhong, 030801, Shanxi, China.
| |
Collapse
|
458
|
Bolla BS, Erdei L, Urbán E, Burián K, Kemény L, Szabó K. Cutibacterium acnes regulates the epidermal barrier properties of HPV-KER human immortalized keratinocyte cultures. Sci Rep 2020; 10:12815. [PMID: 32733073 PMCID: PMC7393503 DOI: 10.1038/s41598-020-69677-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022] Open
Abstract
Our skin provides a physical barrier to separate the internal part of our body from the environment. Maintenance of complex barrier functions is achieved through anatomical structures in the skin, the stratified squamous epithelium specialized junctional organelles, called tight junctions (TJs). Several members of our microbial communities are known to affect the differentiation state and function of the colonized organ. Whether and how interactions between skin cells and cutaneous microbes, including Cutibacterium acnes (C. acnes), modify the structure and/or function of our skin is currently only partly understood. Thus, in our studies, we investigated whether C. acnes may affect the epidermal barrier using in vitro model systems. Real-time cellular analysis showed that depending on the keratinocyte differentiation state, the applied C. acnes strains and their dose, the measured impedance values change, together with the expression of selected TJ proteins. These may reflect barrier alterations, which can be partially restored upon antibiotic–antimycotic treatment. Our findings suggest that C. acnes can actively modify the barrier properties of cultured keratinocytes, possibly through alteration of tight cell-to-cell contacts. Similar events may play important roles in our skin, in the maintenance of cutaneous homeostasis.
Collapse
Affiliation(s)
- Beáta Szilvia Bolla
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary
| | - Lilla Erdei
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary
| | - Edit Urbán
- Department of Public Health, University of Szeged, Szeged, Hungary
| | - Katalin Burián
- Institute of Clinical Microbiology, University of Szeged, Szeged, Hungary
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary.,MTA-SZTE Dermatological Research Group, Szeged, Hungary
| | - Kornélia Szabó
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary. .,MTA-SZTE Dermatological Research Group, Szeged, Hungary.
| |
Collapse
|
459
|
Protective action of Bacillus clausii probiotic strains in an in vitro model of Rotavirus infection. Sci Rep 2020; 10:12636. [PMID: 32724066 PMCID: PMC7387476 DOI: 10.1038/s41598-020-69533-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 07/06/2020] [Indexed: 02/06/2023] Open
Abstract
Rotavirus is the most common cause of acute gastroenteritis (AGE) in young children. Bacillus clausii (B. clausii) is a spore-forming probiotic that is able to colonize the gut. A mixture of four B. clausii strains (O/C, T, SIN and N/R) is commonly used for the treatment of AGE, and it has been demonstrated that it can reduce the duration and severity of diarrhea in children with AGE. Few studies have sought to characterize the mechanisms responsible for such beneficial effects. Intestinal effects of probiotics are likely to be strain-specific. We conducted a series of in vitro experiments investigating the activities of this mixture of B. clausii strains on biomarkers of mucosal barrier integrity and immune function in a cellular model of Rotavirus infection. B. clausii protected enterocytes against Rotavirus-induced decrease in trans-epithelial electrical resistance, and up-regulated expression of mucin 5AC and tight junction proteins (occludin and zonula occludens-1), all of which are important for effective mucosal barrier function. B. clausii also inhibited reactive oxygen species production and release of pro-inflammatory cytokines (interleukin-8 and interferon-β) in Rotavirus-infected cells, and down-regulated pro-inflammatory Toll-like receptor 3 pathway gene expression. Such mechanisms likely contributed to the observed protective effects of B. clausii against reduced cell proliferation and increased apoptosis in Rotavirus-infected enterocytes.
Collapse
|
460
|
Diefenbach A, Gnafakis S, Shomrat O. Innate Lymphoid Cell-Epithelial Cell Modules Sustain Intestinal Homeostasis. Immunity 2020; 52:452-463. [PMID: 32187516 DOI: 10.1016/j.immuni.2020.02.016] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023]
Abstract
The intestines have the essential but challenging mission of absorbing nutrients, restricting damage from food-derived toxins, promoting colonization by symbionts, and expelling pathogens. These processes are often incompatible with each other and must therefore be prioritized in view of the most crucial contemporary needs of the host. Recent work has shown that tissue-resident innate lymphoid cells (ILCs) constitute a central sensory module allowing adaptation of intestinal organ function to changing environmental input. Here, we propose a conceptual framework positing that the various types of ILC act in distinct modules with intestinal epithelial cells, collectively safeguarding organ function. Such homeostasis-promoting circuitry has high potential to be plumbed for new therapeutic approaches to the treatment of immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Andreas Diefenbach
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10117 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany.
| | - Stylianos Gnafakis
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10117 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany
| | - Omer Shomrat
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch Strasse 2, 10117 Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
461
|
Le N, Mazahery C, Nguyen K, Levine AD. Regulation of Intestinal Epithelial Barrier and Immune Function by Activated T Cells. Cell Mol Gastroenterol Hepatol 2020; 11:55-76. [PMID: 32659380 PMCID: PMC7596298 DOI: 10.1016/j.jcmgh.2020.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Communication between T cells and the intestinal epithelium is altered in many diseases, causing T-cell activation, depletion, or recruitment, and disruption of the epithelium. We hypothesize that activation of T cells regulates epithelial barrier function by targeting the assembly of the tight junction complex. METHODS In a 3-dimensional and 2-dimensional co-culture model of activated T cells subjacent to the basolateral surface of an epithelial monolayer, the pore, leak, and unrestricted pathways were evaluated using transepithelial resistance and flux of fluorescently labeled tracers. T cells were acutely and chronically activated by cross-linking the T-cell receptor. Tight junction assembly and expression were measured using quantitative polymerase chain reaction, immunoblot, and immunofluorescence confocal microscopy. RESULTS Co-culture with acutely and chronically activated T cells decreased the magnitude of ion flux through the pore pathway, which was maintained in the presence of acutely activated T cells. Chronically activated T cells after 30 hours induced a precipitous increase in the magnitude of both ion and molecular flux, resulting in an increase in the unrestricted pathway, destruction of microvilli, expansion in cell surface area, and cell death. These fluctuations in permeability were the result of changes in the assembly and expression of tight junction proteins, cell morphology, and viability. Co-culture modulated the expression of immune mediators in the epithelium and T cells. CONCLUSIONS Bidirectional communication between T cells and epithelium mediates a biphasic response in barrier integrity that is facilitated by the balance between structural proteins partitioning in the mobile lateral phase vs the tight junction complex and cell morphology.
Collapse
Affiliation(s)
- Nga Le
- Department of Molecular Biology and Microbiology
| | | | - Kien Nguyen
- Department of Molecular Biology and Microbiology
| | - Alan D Levine
- Department of Molecular Biology and Microbiology; Department of Pathology; Department of Pharmacology; Department of Medicine; Department of Pediatrics; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
462
|
Dunne MR, Byrne G, Chirdo FG, Feighery C. Coeliac Disease Pathogenesis: The Uncertainties of a Well-Known Immune Mediated Disorder. Front Immunol 2020; 11:1374. [PMID: 32733456 PMCID: PMC7360848 DOI: 10.3389/fimmu.2020.01374] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/28/2020] [Indexed: 12/21/2022] Open
Abstract
Coeliac disease is a common small bowel enteropathy arising in genetically predisposed individuals and caused by ingestion of gluten in the diet. Great advances have been made in understanding the role of the adaptive immune system in response to gluten peptides. Despite detailed knowledge of these adaptive immune mechanisms, the complete series of pathogenic events responsible for development of the tissue lesion remains less certain. This review contributes to the field by discussing additional mechanisms which may also contribute to pathogenesis. These include the production of cytokines such as interleukin-15 by intestinal epithelial cells and local antigen presenting cells as a pivotal event in the disease process. A subset of unconventional T cells called gamma/delta T cells are also persistently expanded in the coeliac disease (CD) small intestinal epithelium and recent analysis has shown that these cells contribute to pathogenic inflammation. Other unconventional T cell subsets may play a local immunoregulatory role and require further study. It has also been suggested that, in addition to activation of pathogenic T helper cells by gluten peptides, other peptides may directly interact with the intestinal mucosa, further contributing to the disease process. We also discuss how myofibroblasts, a major source of tissue transglutaminase and metalloproteases, may play a key role in intestinal tissue remodeling. Contribution of each of these factors to pathogenesis is discussed to enhance our view of this complex disorder and to contribute to a wider understanding of chronic immune-mediated disease.
Collapse
Affiliation(s)
- Margaret R. Dunne
- Department of Surgery, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Greg Byrne
- School of Biological & Health Sciences, Technological University, Dublin, Ireland
| | - Fernando G. Chirdo
- Instituto de Estudios Inmunologicos y Fisiopatologicos - IIFP (UNLP-CONICET), National University of La Plata, La Plata, Argentina
| | - Conleth Feighery
- Department of Immunology, Trinity College Dublin and St. James's Hospital, Dublin, Ireland
| |
Collapse
|
463
|
Barnhoorn MC, Hakuno SK, Bruckner RS, Rogler G, Hawinkels LJAC, Scharl M. Stromal Cells in the Pathogenesis of Inflammatory Bowel Disease. J Crohns Colitis 2020; 14:995-1009. [PMID: 32160284 PMCID: PMC7392167 DOI: 10.1093/ecco-jcc/jjaa009] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Up till now, research on inflammatory bowel disease [IBD] has mainly been focused on the immune cells present in the gastrointestinal tract. However, recent insights indicate that stromal cells also play an important and significant role in IBD pathogenesis. Stromal cells in the intestines regulate both intestinal epithelial and immune cell homeostasis. Different subsets of stromal cells have been found to play a role in other inflammatory diseases [e.g. rheumatoid arthritis], and these various stromal subsets now appear to carry out also specific functions in the inflamed gut in IBD. Novel potential therapies for IBD utilize, as well as target, these pathogenic stromal cells. Injection of mesenchymal stromal cells [MSCs] into fistula tracts of Crohn's disease patients is already approved and used in clinical settings. In this review we discuss the current knowledge of the role of stromal cells in IBD pathogenesis. We further outline recent attempts to modify the stromal compartment in IBD with agents that target or replace the pathogenic stroma.
Collapse
Affiliation(s)
- M C Barnhoorn
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands,Corresponding author: Prof. Dr Michael Scharl, Department of Gastroenterology and Hepatology, University Hospital Zurich, Rämistrasse 100, Zurich 8091, Switzerland. Tel: 41 44 255 3419; Fax: 41 44 255 9497;
| | - S K Hakuno
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - R S Bruckner
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands,Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - G Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - L J A C Hawinkels
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - M Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
464
|
Østvik AE, Svendsen TD, Granlund AVB, Doseth B, Skovdahl HK, Bakke I, Thorsvik S, Afroz W, Walaas GA, Mollnes TE, Gustafsson BI, Sandvik AK, Bruland T. Intestinal Epithelial Cells Express Immunomodulatory ISG15 During Active Ulcerative Colitis and Crohn's Disease. J Crohns Colitis 2020; 14:920-934. [PMID: 32020185 PMCID: PMC7392169 DOI: 10.1093/ecco-jcc/jjaa022] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS Intestinal epithelial cells [IECs] secrete cytokines that recruit immune cells to the mucosa and regulate immune responses that drive inflammation in inflammatory bowel disease [IBD]. However, experiments in patient-derived IEC models are still scarce. Here, we aimed to investigate how innate immunity and IEC-specific pattern recognition receptor [PRR] signalling can be involved in an enhanced type I interferon [IFN] gene signature observed in colon epithelium of patients with active IBD, with a special focus on secreted ubiquitin-like protein ISG15. METHODS Gene and protein expression in whole mucosa biopsies and in microdissected human colonic epithelial lining, in HT29 human intestinal epithelial cells and primary 3D colonoids treated with PRR-ligands and cytokines, were detected by transcriptomics, in situ hybridisation, immunohistochemistry, western blots, and enzyme-linked immunosorbent assay [ELISA]. Effects of IEC-secreted cytokines were examined in human peripheral blood mononuclear cells [PBMCs] by multiplex chemokine profiling and ELISA. RESULTS The type I IFN gene signature in human mucosal biopsies was mimicked in Toll-like receptor TLR3 and to some extent tumour necrosis factor [TNF]-treated human IECs. In intestinal biopsies, ISG15 expression correlated with expression of the newly identified receptor for extracellular ISG15, LFA-1 integrin. ISG15 was expressed and secreted from HT29 cells and primary 3D colonoids through both JAK1-pSTAT-IRF9-dependent and independent pathways. In experiments using PBMCs, we show that ISG15 releases IBD-relevant proinflammatory cytokines such as CXCL1, CXCL5, CXCL8, CCL20, IL1, IL6, TNF, and IFNγ. CONCLUSIONS ISG15 is secreted from primary IECs upon extracellular stimulation, and mucosal ISG15 emerges as an intriguing candidate for immunotherapy in IBD.
Collapse
Affiliation(s)
- Ann Elisabet Østvik
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway,Department of G2astroenterology and Hepatology, Clinic of Medicine, St. Olav’s University Hospital, Trondheim, Norway
| | - Tarjei Dahl Svendsen
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Atle van Beelen Granlund
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway,Centre of Molecular Inflammation Research, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Berit Doseth
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Helene Kolstad Skovdahl
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingunn Bakke
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway,Clinic of Medicine, St Olav’s University Hospital, Trondheim, Norway,Clinic of Laboratory Medicine, St. Olav’s University Hospital, Trondheim, Norway
| | - Silje Thorsvik
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway,Department of G2astroenterology and Hepatology, Clinic of Medicine, St. Olav’s University Hospital, Trondheim, Norway,Centre of Molecular Inflammation Research, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Wahida Afroz
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Gunnar Andreas Walaas
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Tom Eirik Mollnes
- Centre of Molecular Inflammation Research, NTNU-Norwegian University of Science and Technology, Trondheim, Norway,Research Laboratory, Nordland Hospital, Bodo, Norway,K.G. Jebsen Thrombosis Research and Expertise Center, Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway,Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Björn Inge Gustafsson
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway,Department of G2astroenterology and Hepatology, Clinic of Medicine, St. Olav’s University Hospital, Trondheim, Norway
| | - Arne Kristian Sandvik
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway,Department of G2astroenterology and Hepatology, Clinic of Medicine, St. Olav’s University Hospital, Trondheim, Norway,Centre of Molecular Inflammation Research, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Torunn Bruland
- Department of Clinical and Molecular Medicine, NTNU-Norwegian University of Science and Technology, Trondheim, Norway,Clinic of Medicine, St Olav’s University Hospital, Trondheim, Norway,Corresponding author: Torunn Bruland, PhD, Department of Clinical and Molecular Medicine [IKOM], Faculty of Medicine and Health Sciences [MH], NTNU-Norwegian University of Science and Technology, Prinsesse Kristinas gate 1, NO-7489 Trondheim, Norway. Tel.: +47 72825324; E-mail
| |
Collapse
|
465
|
Kayama H, Takeda K. Manipulation of epithelial integrity and mucosal immunity by host and microbiota-derived metabolites. Eur J Immunol 2020; 50:921-931. [PMID: 32511746 DOI: 10.1002/eji.201948478] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/08/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022]
Abstract
The human intestinal tract contains a large number of microbes, their metabolites, and potentially harmful food antigens. The intestinal epithelium separates the mucosa where immune cells are located from luminal microbes by expressing various factors that assemble into physical and chemical barriers. In addition to epithelial cells, immune cells are essential for enforcing mucosal barriers through production of inflammatory and anti-inflammatory mediators. Intestinal microbiota, represented by gut ecological communities of living microorganisms, influences maturation and homeostasis of host immune system and contributes to the maintenance of the epithelial integrity with small molecules derived from their metabolism, termed metabolites. In turn, immune cells receive signals from microbiota, and may play key role in maintenance of a healthy bacterial composition and reinforcement of epithelial barrier functions, leading to the establishment of a host-bacterial mutualism. Alterations in the microbiota community and metabolome profiles are observed in patients with various disorders including inflammatory bowel disease. In this review, we will discuss physiological functions of the microbiota and its metabolites in regulating host immune system and reinforcing epithelial barrier functions. Further understanding of these processes will aid in identification of novel therapeutic targets and subsequent development of therapeutic interventions in a range of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Hisako Kayama
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Institute for Advanced Co-Creation Studies, Osaka University, Osaka, Japan
| | - Kiyoshi Takeda
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan.,WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| |
Collapse
|
466
|
Roodsant T, Navis M, Aknouch I, Renes IB, van Elburg RM, Pajkrt D, Wolthers KC, Schultsz C, van der Ark KCH, Sridhar A, Muncan V. A Human 2D Primary Organoid-Derived Epithelial Monolayer Model to Study Host-Pathogen Interaction in the Small Intestine. Front Cell Infect Microbiol 2020; 10:272. [PMID: 32656095 PMCID: PMC7326037 DOI: 10.3389/fcimb.2020.00272] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Gut organoids are stem cell derived 3D models of the intestinal epithelium that are useful for studying interactions between enteric pathogens and their host. While the organoid model has been used for both bacterial and viral infections, this is a closed system with the luminal side being inaccessible without microinjection or disruption of the organoid polarization. In order to overcome this and simplify their applicability for transepithelial studies, permeable membrane based monolayer approaches are needed. In this paper, we demonstrate a method for generating a monolayer model of the human fetal intestinal polarized epithelium that is fully characterized and validated. Proximal and distal small intestinal organoids were used to generate 2D monolayer cultures, which were characterized with respect to epithelial cell types, polarization, barrier function, and gene expression. In addition, viral replication and bacterial translocation after apical infection with enteric pathogens Enterovirus A71 and Listeria monocytogenes were evaluated, with subsequent monitoring of the pro-inflammatory host response. This human 2D fetal intestinal monolayer model will be a valuable tool to study host-pathogen interactions and potentially reduce the use of animals in research.
Collapse
Affiliation(s)
- Thomas Roodsant
- Department of Global Health-Amsterdam Institute for Global Health and Development, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Marit Navis
- Tytgat Institute for Intestinal and Liver Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Ikrame Aknouch
- Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Viroclinics Xplore, Schaijk, Netherlands
| | - Ingrid B Renes
- Danone Nutricia Research, Utrecht, Netherlands.,Department of Pediatrics, Amsterdam University Medical Center (UMC), Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Ruurd M van Elburg
- Department of Pediatrics, Amsterdam University Medical Center (UMC), Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Dasja Pajkrt
- Department of Pediatric Infectious Diseases, Amsterdam University Medical Center (UMC), Emma Children's Hospital, University of Amsterdam, Amsterdam, Netherlands
| | - Katja C Wolthers
- Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Constance Schultsz
- Department of Global Health-Amsterdam Institute for Global Health and Development, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Kees C H van der Ark
- Department of Global Health-Amsterdam Institute for Global Health and Development, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Adithya Sridhar
- Department of Medical Microbiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Vanesa Muncan
- Tytgat Institute for Intestinal and Liver Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
467
|
Hausmann A, Russo G, Grossmann J, Zünd M, Schwank G, Aebersold R, Liu Y, Sellin ME, Hardt W. Germ-free and microbiota-associated mice yield small intestinal epithelial organoids with equivalent and robust transcriptome/proteome expression phenotypes. Cell Microbiol 2020; 22:e13191. [PMID: 32068945 PMCID: PMC7317401 DOI: 10.1111/cmi.13191] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
Intestinal epithelial organoids established from gut tissue have become a widely used research tool. However, it remains unclear how environmental cues, divergent microbiota composition and other sources of variation before, during and after establishment confound organoid properties, and how these properties relate to the original tissue. While environmental influences cannot be easily addressed in human organoids, mice offer a controlled assay-system. Here, we probed the effect of donor microbiota differences, previously identified as a confounding factor in murine in vivo studies, on organoids. We analysed the proteomes and transcriptomes of primary organoid cultures established from two colonised and one germ-free mouse colony of C57BL/6J genetic background, and compared them to their tissue of origin and commonly used cell lines. While an imprint of microbiota-exposure was observed on the proteome of epithelial samples, the long-term global impact of donor microbiota on organoid expression patterns was negligible. Instead, stochastic culture-to-culture differences accounted for a moderate variability between independently established organoids. Integration of transcriptome and proteome datasets revealed an organoid-typic expression signature comprising 14 transcripts and 10 proteins that distinguished organoids across all donors from murine epithelial cell lines and fibroblasts and closely mimicked expression patterns in the gut epithelium. This included the inflammasome components ASC, Naip1-6, Nlrc4 and Caspase-1, which were highly expressed in all organoids compared to the reference cell line m-ICc12 or mouse embryonic fibroblasts. Taken together, these results reveal that the donor microbiota has little effect on the organoid phenotype and suggest that organoids represent a more suitable culture model than immortalised cell lines, in particular for studies of intestinal epithelial inflammasomes.
Collapse
Affiliation(s)
- Annika Hausmann
- Institute of Microbiology, Department of BiologyETH ZurichZurichSwitzerland
| | - Giancarlo Russo
- Functional Genomics Center ZurichUniversity of ZurichZurichSwitzerland
| | - Jonas Grossmann
- Functional Genomics Center ZurichUniversity of ZurichZurichSwitzerland
| | - Mirjam Zünd
- Institute of Microbiology, Department of BiologyETH ZurichZurichSwitzerland
| | - Gerald Schwank
- Institute of Pharmacology and ToxicologyUniversity of ZurichZurichSwitzerland
| | - Ruedi Aebersold
- Institute of Systems Biology, Department of BiologyETH ZurichZurichSwitzerland
| | - Yansheng Liu
- Institute of Systems Biology, Department of BiologyETH ZurichZurichSwitzerland
- Department of Pharmacology, Cancer Biology InstituteYale University School of MedicineWest HavenConnecticutUSA
| | - Mikael E. Sellin
- Institute of Microbiology, Department of BiologyETH ZurichZurichSwitzerland
- Science for Life Laboratory, Department of Medical Biochemistry and MicrobiologyUppsala UniversityUppsalaSweden
| | | |
Collapse
|
468
|
Papoutsopoulou S, Satsangi J, Campbell BJ, Probert CS. Review article: impact of cigarette smoking on intestinal inflammation-direct and indirect mechanisms. Aliment Pharmacol Ther 2020; 51:1268-1285. [PMID: 32372449 DOI: 10.1111/apt.15774] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/13/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The inflammatory bowel diseases, Crohn's disease and ulcerative colitis are related multifactorial diseases. Their pathogenesis is influenced by each individual's immune system, the environmental factors within exposome and genetic predisposition. Smoking habit is the single best-established environmental factor that influences disease phenotype, behaviour and response to therapy. AIM To assess current epidemiological, experimental and clinical evidence that may explain how smoking impacts on the pathogenesis of inflammatory bowel disease. METHODS A Medline search for 'cigarette smoking', in combination with terms including 'passive', 'second-hand', 'intestinal inflammation', 'Crohn's disease', 'ulcerative colitis', 'colitis'; 'intestinal epithelium', 'immune system', 'intestinal microbiota', 'tight junctions', 'mucus', 'goblet cells', 'Paneth cells', 'autophagy'; 'epigenetics', 'genes', 'DNA methylation', 'histones', 'short noncoding/long noncoding RNAs'; 'carbon monoxide/CO' and 'nitric oxide/NO' was performed. RESULTS Studies found evidence of direct and indirect effects of smoking on various parameters, including oxidative damage, impairment of intestinal barrier and immune cell function, epigenetic and microbiota composition changes, that contribute to the pathogenesis of inflammatory bowel disease. CONCLUSIONS Cigarette smoking promotes intestinal inflammation by affecting the function and interactions among intestinal epithelium, immune system and microbiota/microbiome.
Collapse
Affiliation(s)
- Stamatia Papoutsopoulou
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Jack Satsangi
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Experimental Medicine Division, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Barry J Campbell
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Chris S Probert
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
469
|
Abstract
Recent years have witnessed an emergence of interest in understanding metabolic changes associated with immune responses, termed immunometabolism. As oxygen is central to all aerobic metabolism, hypoxia is now recognized to contribute fundamentally to inflammatory and immune responses. Studies from a number of groups have implicated a prominent role for oxygen metabolism and hypoxia in innate immunity of healthy tissue (physiologic hypoxia) and during active inflammation (inflammatory hypoxia). This inflammatory hypoxia emanates from a combination of recruited inflammatory cells (e.g., neutrophils, eosinophils, and monocytes), high rates of oxidative metabolism, and the activation of multiple oxygen-consuming enzymes during inflammation. These localized shifts toward hypoxia have identified a prominent role for the transcription factor hypoxia-inducible factor (HIF) in the regulation of innate immunity. Such studies have provided new and enlightening insight into our basic understanding of immune mechanisms, and extensions of these findings have identified potential therapeutic targets. In this review, we summarize recent literature around the topic of innate immunity and mucosal hypoxia with a focus on transcriptional responses mediated by HIF.
Collapse
Affiliation(s)
- Sean P Colgan
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, USA;
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Glenn T Furuta
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Cormac T Taylor
- UCD Conway Institute, Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
470
|
Cheng L, Akkerman R, Kong C, Walvoort MTC, de Vos P. More than sugar in the milk: human milk oligosaccharides as essential bioactive molecules in breast milk and current insight in beneficial effects. Crit Rev Food Sci Nutr 2020; 61:1184-1200. [PMID: 32329623 DOI: 10.1080/10408398.2020.1754756] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Human milk is the gold standard for newborn infants. Breast milk not only provides nutrients, it also contains bioactive components that guide the development of the infant's intestinal immune system, which can have a lifelong effect. The bioactive molecules in breast milk regulate microbiota development, immune maturation and gut barrier function. Human milk oligosaccharides (hMOs) are the most abundant bioactive molecules in human milk and have multiple beneficial functions such as support of growth of beneficial bacteria, anti-pathogenic effects, immune modulating effects, and stimulation of intestine barrier functions. Here we critically review the current insight into the benefits of bioactive molecules in mother milk that contribute to neonatal development and focus on current knowledge of hMO-functions on microbiota and the gastrointestinal immune barrier. hMOs produced via genetically engineered microorganisms are now applied in infant formulas to mimic the nutritional composition of breast milk as closely as possible, and their prospects and scientific challenges are discussed in depth.
Collapse
Affiliation(s)
- Lianghui Cheng
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Renate Akkerman
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Chunli Kong
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marthe T C Walvoort
- Stratingh Institute for Chemistry, Faculty of Science and Engineering, University of Groningen, Groningen, Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
471
|
Iyer N, Grizotte-Lake M, Duncan K, Gordon SR, Palmer ACS, Calvin C, Zhong G, Isoherranen N, Vaishnava S. Epithelium intrinsic vitamin A signaling co-ordinates pathogen clearance in the gut via IL-18. PLoS Pathog 2020; 16:e1008360. [PMID: 32330185 PMCID: PMC7202665 DOI: 10.1371/journal.ppat.1008360] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 05/06/2020] [Accepted: 01/27/2020] [Indexed: 01/17/2023] Open
Abstract
Intestinal epithelial cells (IECs) are at the forefront of host-pathogen interactions, coordinating a cascade of immune responses to protect against pathogens. Here we show that IEC-intrinsic vitamin A signaling restricts pathogen invasion early in the infection and subsequently activates immune cells to promote pathogen clearance. Mice blocked for retinoic acid receptor (RAR) signaling selectively in IECs (stopΔIEC) showed higher Salmonella burden in colonic tissues early in the infection that associated with higher luminal and systemic loads of the pathogen at later stages. Higher pathogen burden in stopΔIEC mice correlated with attenuated mucosal interferon gamma (IFNγ) production by underlying immune cells. We found that, at homeostasis, the intestinal epithelium of stopΔIEC mice produced significantly lower amounts of interleukin 18 (IL-18), a potent inducer of IFNγ. Regulation of IL-18 by vitamin A was also observed in a dietary model of vitamin A supplementation. IL-18 reconstitution in stopΔIEC mice restored resistance to Salmonella by promoting epithelial cell shedding to eliminate infected cells and limit pathogen invasion early in infection. Further, IL-18 augmented IFNγ production by underlying immune cells to restrict pathogen burden and systemic spread. Our work uncovers a critical role for vitamin A in coordinating a biphasic immune response to Salmonella infection by regulating IL-18 production by IECs. Epithelial cells line the intestinal lumen, forming a barrier between the body and dietary and microbial contents in the lumen. Apart from absorbing nutrients from diet, these epithelial cells help mediate a stable, symbiotic relationship between commensal bacteria and the immune cells. During infection, they help co-ordinate the immune response to counter the infection. How dietary micronutrients, such as vitamin A, inform epithelial cell function during infection is poorly understood. Using a model where epithelial cells in the gut cannot respond to vitamin A signals, we find that epithelial vitamin A signaling promotes resistance to Salmonella infection. We show that, vitamin A increases the production of a key cytokine, interleukin 18, by epithelial cells. IL-18 promotes shedding of infected epithelial cells to reduce the pathogen invasion while also inducing the production of interferon gamma by immune cells to mediate pathogen clearance. Thus, epithelial cells dynamically respond to dietary vitamin A to regulate interleukin 18 production and potentiate resistance to infection.
Collapse
Affiliation(s)
- Namrata Iyer
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States of America
| | - Mayara Grizotte-Lake
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States of America
| | - Kellyanne Duncan
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States of America
| | - Sarah R. Gordon
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States of America
| | - Ana C. S. Palmer
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States of America
| | - Crystle Calvin
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States of America
| | - Guo Zhong
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
| | - Nina Isoherranen
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States of America
| | - Shipra Vaishnava
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, United States of America
- * E-mail:
| |
Collapse
|
472
|
Farré R, Fiorani M, Abdu Rahiman S, Matteoli G. Intestinal Permeability, Inflammation and the Role of Nutrients. Nutrients 2020; 12:nu12041185. [PMID: 32340206 PMCID: PMC7231157 DOI: 10.3390/nu12041185] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/10/2020] [Accepted: 04/17/2020] [Indexed: 12/17/2022] Open
Abstract
The interaction between host and external environment mainly occurs in the gastrointestinal tract, where the mucosal barrier has a critical role in many physiologic functions ranging from digestion, absorption, and metabolism. This barrier allows the passage and absorption of nutrients, but at the same time, it must regulate the contact between luminal antigens and the immune system, confining undesirable products to the lumen. Diet is an important regulator of the mucosal barrier, and the cross-talk among dietary factors, the immune system, and microbiota is crucial for the modulation of intestinal permeability and for the maintenance of gastrointestinal tract (GI) homeostasis. In the present review, we will discuss the role of a number of dietary nutrients that have been proposed as regulators of inflammation and epithelial barrier function. We will also consider the metabolic function of the microbiota, which is capable of elaborating the diverse nutrients and synthesizing products of great interest. Better knowledge of the influence of dietary nutrients on inflammation and barrier function can be important for the future development of new therapeutic approaches for patients with mucosal barrier dysfunction, a critical factor in the pathogenesis of many GI and non-GI diseases.
Collapse
Affiliation(s)
- Ricard Farré
- Translational Research Center for Gastrointestinal Disorders (TARGID) Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (M.F.); (S.A.R.); (G.M.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +32-16-34-57-52
| | - Marcello Fiorani
- Translational Research Center for Gastrointestinal Disorders (TARGID) Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (M.F.); (S.A.R.); (G.M.)
| | - Saeed Abdu Rahiman
- Translational Research Center for Gastrointestinal Disorders (TARGID) Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (M.F.); (S.A.R.); (G.M.)
| | - Gianluca Matteoli
- Translational Research Center for Gastrointestinal Disorders (TARGID) Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (M.F.); (S.A.R.); (G.M.)
| |
Collapse
|
473
|
Kaur A, Goggolidou P. Ulcerative colitis: understanding its cellular pathology could provide insights into novel therapies. JOURNAL OF INFLAMMATION-LONDON 2020; 17:15. [PMID: 32336953 PMCID: PMC7175540 DOI: 10.1186/s12950-020-00246-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 04/07/2020] [Indexed: 12/16/2022]
Abstract
Dynamic interactions between the gastrointestinal epithelium and the mucosal immune system normally contribute to ensuring intestinal homeostasis and optimal immunosurveillance, but destabilisation of these interactions in genetically predisposed individuals can lead to the development of chronic inflammatory diseases. Ulcerative colitis is one of the main types of inflammatory diseases that affect the bowel, but its pathogenesis has yet to be completely defined. Several genetic factors and other inflammation-related genes are implicated in mediating the inflammation and development of the disease. Some susceptibility loci associated with increased risk of ulcerative colitis are found to be implicated in mucosal barrier function. Different biomarkers that cause damage to the colonic mucosa can be detected in patients, including perinuclear ANCA, which is also useful in distinguishing ulcerative colitis from other colitides. The choice of treatment for ulcerative colitis depends on disease severity. Therapeutic strategies include anti-tumour necrosis factor alpha (TNF-α) monoclonal antibodies used to block the production of TNF-α that mediates intestinal tract inflammation, an anti-adhesion drug that prevents lymphocyte infiltration from the blood into the inflamed gut, inhibitors of JAK1 and JAK3 that suppress the innate immune cell signalling and interferons α/β which stimulate the production of anti-inflammatory cytokines, as well as faecal microbiota transplantation. Although further research is still required to fully dissect the pathophysiology of ulcerative colitis, understanding its cellular pathology and molecular mechanisms has already proven beneficial and it has got the potential to identify further novel, effective targets for therapy and reduce the burden of this chronic disease.
Collapse
Affiliation(s)
- Amandip Kaur
- Department of Biomedical Science and Physiology, Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton, WV1 1LY UK
| | - Paraskevi Goggolidou
- Department of Biomedical Science and Physiology, Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton, WV1 1LY UK
| |
Collapse
|
474
|
Mechanisms of Endogenous HIV-1 Reactivation by Endocervical Epithelial Cells. J Virol 2020; 94:JVI.01904-19. [PMID: 32051273 DOI: 10.1128/jvi.01904-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 02/01/2020] [Indexed: 12/23/2022] Open
Abstract
Pharmacological HIV-1 reactivation to reverse latent infection has been extensively studied. However, HIV-1 reactivation also occurs naturally, as evidenced by occasional low-level viremia ("viral blips") during antiretroviral treatment (ART). Clarifying where blips originate from and how they happen could provide clues to stimulate latency reversal more effectively and safely or to prevent viral rebound following ART cessation. We studied HIV-1 reactivation in the female genital tract, a dynamic anatomical target for HIV-1 infection throughout all disease stages. We found that primary endocervical epithelial cells from several women reactivated HIV-1 from latently infected T cells. The endocervical cells' HIV-1 reactivation capacity further increased upon Toll-like receptor 3 stimulation with poly(I·C) double-stranded RNA or infection with herpes simplex virus 2 (HSV-2). Notably, acyclovir did not eliminate HSV-2-induced HIV-1 reactivation. While endocervical epithelial cells secreted large amounts of several cytokines and chemokines, especially tumor necrosis factor alpha (TNF-α), CCL3, CCL4, and CCL20, their HIV-1 reactivation capacity was almost completely blocked by TNF-α neutralization alone. Thus, immunosurveillance activities by columnar epithelial cells in the endocervix can cause endogenous HIV-1 reactivation, which may contribute to viral blips during ART or rebound following ART interruption.IMPORTANCE A reason that there is no universal cure for HIV-1 is that the virus can hide in the genome of infected cells in the form of latent proviral DNA. This hidden provirus is protected from antiviral drugs until it eventually reactivates to produce new virions. It is not well understood where in the body or how this reactivation occurs. We studied HIV-1 reactivation in the female genital tract, which is often the portal of HIV-1 entry and which remains a site of infection throughout the disease. We found that the columnar epithelial cells lining the endocervix, the lower part of the uterus, are particularly effective in reactivating HIV-1 from infected T cells. This activity was enhanced by certain microbial stimuli, including herpes simplex virus 2, and blocked by antibodies against the inflammatory cytokine TNF-α. Avoiding HIV-1 reactivation could be important for maintaining a functional HIV-1 cure when antiviral therapy is stopped.
Collapse
|
475
|
Crowley SM, Han X, Allaire JM, Stahl M, Rauch I, Knodler LA, Vallance BA. Intestinal restriction of Salmonella Typhimurium requires caspase-1 and caspase-11 epithelial intrinsic inflammasomes. PLoS Pathog 2020; 16:e1008498. [PMID: 32282854 PMCID: PMC7179941 DOI: 10.1371/journal.ppat.1008498] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 04/23/2020] [Accepted: 03/25/2020] [Indexed: 12/22/2022] Open
Abstract
We investigated the role of the inflammasome effector caspases-1 and -11 during Salmonella enterica serovar Typhimurium infection of murine intestinal epithelial cells (IECs). Salmonella burdens were significantly greater in the intestines of caspase-1/11 deficient (Casp1/11−/−), Casp1−/− and Casp11−/− mice, as compared to wildtype mice. To determine if this reflected IEC-intrinsic inflammasomes, enteroid monolayers were derived and infected with Salmonella. Casp11−/− and wildtype monolayers responded similarly, whereas Casp1−/− and Casp1/11−/− monolayers carried significantly increased intracellular burdens, concomitant with marked decreases in IEC shedding and death. Pretreatment with IFN-γ to mimic inflammation increased caspase-11 levels and IEC death, and reduced Salmonella burdens in Casp1−/− monolayers, while high intracellular burdens and limited cell shedding persisted in Casp1/11−/− monolayers. Thus caspase-1 regulates inflammasome responses in IECs at baseline, while proinflammatory activation of IECs reveals a compensatory role for caspase-11. These results demonstrate the importance of IEC-intrinsic canonical and non-canonical inflammasomes in host defense against Salmonella. Intestinal epithelial cells (IECs) are located at the interface between the gut lumen and the mucosal immune system and form the first layer of defense against the invasive enteric pathogen Salmonella enterica serovar Typhimurium. To prevent Salmonella, and other pathogens from establishing a foothold in the gut, the host mobilizes the inflammasome to selectively eject infected/compromised IECs from the epithelial layer into the intestinal lumen. This involves the activation of the inflammatory caspases; caspase-1 and -11. The individual contributions of each caspase to intestinal host defense, as well as the importance of IEC-intrinsic inflammasomes have not been previously defined, due to the lack of Casp1−/− mice as well as appropriate IEC-intrinsic defense models. Here, we determined that both caspases contribute to controlling Salmonella pathogen burdens and IEC shedding in the mouse intestine. Caspase-1 appears to play a larger role at baseline since caspase-11 expression must be first induced through proinflammatory signalling. Our data also highlights that IEC-intrinsic caspase activation is sufficient for infection-induced cell shedding and that the intestinal epithelium is a key site for inflammasome-mediated immune defense.
Collapse
Affiliation(s)
- Shauna M Crowley
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xiao Han
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joannie M Allaire
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Martin Stahl
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Isabella Rauch
- Department of Medical Microbiology & Immunology, School of Medicine, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Leigh A Knodler
- Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, Washington, United States of America
| | - Bruce A Vallance
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
476
|
Caër C, Wick MJ. Human Intestinal Mononuclear Phagocytes in Health and Inflammatory Bowel Disease. Front Immunol 2020; 11:410. [PMID: 32256490 PMCID: PMC7093381 DOI: 10.3389/fimmu.2020.00410] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/21/2020] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a complex immune-mediated disease of the gastrointestinal tract that increases morbidity and negatively influences the quality of life. Intestinal mononuclear phagocytes (MNPs) have a crucial role in maintaining epithelial barrier integrity while controlling pathogen invasion by activating an appropriate immune response. However, in genetically predisposed individuals, uncontrolled immune activation to intestinal flora is thought to underlie the chronic mucosal inflammation that can ultimately result in IBD. Thus, MNPs are involved in fine-tuning mucosal immune system responsiveness and have a critical role in maintaining homeostasis or, potentially, the emergence of IBD. MNPs include monocytes, macrophages and dendritic cells, which are functionally diverse but highly complementary. Despite their crucial role in maintaining intestinal homeostasis, specific functions of human MNP subsets are poorly understood, especially during diseases such as IBD. Here we review the current understanding of MNP ontogeny, as well as the recently identified human intestinal MNP subsets, and discuss their role in health and IBD.
Collapse
Affiliation(s)
- Charles Caër
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mary Jo Wick
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
477
|
Johnson MD, Witherden DA, Havran WL. The Role of Tissue-resident T Cells in Stress Surveillance and Tissue Maintenance. Cells 2020; 9:E686. [PMID: 32168884 PMCID: PMC7140644 DOI: 10.3390/cells9030686] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
While forming a minor population in the blood and lymphoid compartments, T cells are significantly enriched within barrier tissues. In addition to providing protection against infection, these tissue-resident T cells play critical roles in tissue homeostasis and repair. T cells in the epidermis and intestinal epithelium produce growth factors and cytokines that are important for the normal turnover and maintenance of surrounding epithelial cells and are additionally required for the efficient recognition of, and response to, tissue damage. A role for tissue-resident T cells is emerging outside of the traditional barrier tissues as well, with recent research indicating that adipose tissue-resident T cells are required for the normal maintenance and function of the adipose tissue compartment. Here we review the functions of tissue-resident T cells in the epidermis, intestinal epithelium, and adipose tissue, and compare the mechanisms of their activation between these sites.
Collapse
Affiliation(s)
| | - Deborah A. Witherden
- Department of Immunology and Microbiology, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, USA; (M.D.J.); (W.L.H.)
| | | |
Collapse
|
478
|
Jang H, Kwak SY, Park S, Kim K, Kim YH, Na J, Kim H, Jang WS, Lee SJ, Kim MJ, Myung JK, Shim S. Pravastatin Alleviates Radiation Proctitis by Regulating Thrombomodulin in Irradiated Endothelial Cells. Int J Mol Sci 2020; 21:ijms21051897. [PMID: 32164317 PMCID: PMC7084904 DOI: 10.3390/ijms21051897] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/07/2020] [Accepted: 03/08/2020] [Indexed: 12/23/2022] Open
Abstract
Although radiotherapy plays a crucial in the management of pelvic tumors, its toxicity on surrounding healthy tissues such as the small intestine, colon, and rectum is one of the major limitations associated with its use. In particular, proctitis is a major clinical complication of pelvic radiotherapy. Recent evidence suggests that endothelial injury significantly affects the initiation of radiation-induced inflammation. The damaged endothelial cells accelerate immune cell recruitment by activating the expression of endothelial adhesive molecules, which participate in the development of tissue damage. Pravastatin, a cholesterol lowering drug, exerts persistent anti-inflammatory and anti-thrombotic effects on irradiated endothelial cells and inhibits the interaction of leukocytes and damaged endothelial cells. Here, we aimed to investigate the effects of pravastatin on radiation-induced endothelial damage in human umbilical vein endothelial cell and a murine proctitis model. Pravastatin attenuated epithelial damage and inflammatory response in irradiated colorectal lesions. In particular, pravastatin improved radiation-induced endothelial damage by regulating thrombomodulin (TM) expression. In addition, exogenous TM inhibited leukocyte adhesion to the irradiated endothelial cells. Thus, pravastatin can inhibit endothelial damage by inducing TM, thereby alleviating radiation proctitis. Therefore, we suggest that pharmacological modulation of endothelial TM may limit intestinal inflammation after irradiation.
Collapse
Affiliation(s)
- Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Seo-Young Kwak
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
| | - Kyuchang Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Young-heon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Jiyoung Na
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Hyewon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Won-Suk Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Sun-Joo Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Min Jung Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Jae Kyung Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
- Correspondence: ; Tel.: +82-2-3399-5873
| |
Collapse
|
479
|
Yang W, Men P, Xue H, Jiang M, Luo Q. Risk of Gastrointestinal Adverse Events in Cancer Patients Treated With Immune Checkpoint Inhibitor Plus Chemotherapy: A Systematic Review and Meta-Analysis. Front Oncol 2020; 10:197. [PMID: 32211312 PMCID: PMC7076172 DOI: 10.3389/fonc.2020.00197] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 02/05/2020] [Indexed: 12/19/2022] Open
Abstract
Background: The combination of immune checkpoint inhibitors (ICIs) and chemotherapy can improve clinical outcomes in the treatment of various tumors, but may also be associated with more adverse events (AEs). We performed a systematic review and meta-analysis to characterize the risk of gastrointestinal AEs in cancer patients treated with ICI plus chemotherapy. Methods: This review was based on comprehensive search through PubMed, EMBASE, and the Cochrane Library for randomized controlled trials (RCTs) that reported gastrointestinal AEs following the use of ICI plus chemotherapy. Literature screening, data extraction, and quality evaluation were performed by two individual reviewers. Revman (version 5.3) was used for meta-analysis. Risk ratios (RR) with 95% confidence interval (CI) were calculated. Meta-analysis was conducted according to different types of ICIs [programmed death 1 (PD-1), programmed death ligand 1 (PD-L1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors]. Results: After a full-text review, 10 trials involving 5,142 patients were included in the study. Compared with chemotherapy alone, PD-1 inhibitor plus chemotherapy significantly increased the risk of diarrhea (RR = 1.38, 95% CI, 1.13–1.68, P = 0.001; I2 = 0%) and colitis (RR = 2.90, 95% CI, 1.02–8.21, P = 0.050; I2 = 0%), PD-L1 inhibitor plus chemotherapy significantly increased the risk of nausea (RR = 1.17, 95% CI, 1.02-1.35, P = 0.020; I2 = 0%), while CTLA-4 inhibitor plus chemotherapy significantly increased the risk of decreased appetite (RR = 1.49, 95% CI, 1.17–1.90, P = 0.001; I2 = 0%), diarrhea (RR = 2.23, 95% CI, 1.90–2.63, P < 0.00001; I2 = 0%), and colitis (RR = 28.39, 95% CI, 5.59–144.24, P < 0.001; I2 = 0%). Conclusions: This meta-analysis demonstrated that ICI plus chemotherapy is associated with a higher risk of gastrointestinal AEs. However, combining different ICIs may lead to diverse gastrointestinal toxicities. Clinicians should be aware of these AEs in the application of ICI plus chemotherapy.
Collapse
Affiliation(s)
- Wenhan Yang
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang, China.,School of Pharmacy, China Medical University, Shenyang, China
| | - Peng Men
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| | - Huimin Xue
- School of Pharmacy, China Medical University, Shenyang, China
| | - Mingyan Jiang
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang, China.,School of Pharmacy, China Medical University, Shenyang, China
| | - Qiuhua Luo
- Department of Pharmacy, The First Hospital of China Medical University, Shenyang, China.,School of Pharmacy, China Medical University, Shenyang, China
| |
Collapse
|
480
|
Fan L, Huang R, Wu C, Cao Y, Du T, Pu G, Wang H, Zhou W, Li P, Kim SW. Defatted Rice Bran Supplementation in Diets of Finishing Pigs: Effects on Physiological, Intestinal Barrier, and Oxidative Stress Parameters. Animals (Basel) 2020; 10:ani10030449. [PMID: 32182669 PMCID: PMC7143537 DOI: 10.3390/ani10030449] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Most studies on dietary fiber mainly focus on the digestibility of feed nutrients and microbial flora, etc. However, insufficient attention has been paid to the regulation of immune and oxidative stress of the intestinal tract by dietary fiber. This study investigated the effects of varying levels of defatted rice bran replacing corn on physiological, intestinal barrier, and oxidative stress parameters in finishing pigs. Based on the current findings, a high diet of rice bran will not only reduce the level of inflammatory factors in the peripheral blood of finishing pigs, but also enhance the healthy level of the colon through mucin2 and keap1-Nrf2 pathways. Our results can be used as reference for dietary rice bran to improve intestinal health in finishing pigs. Abstract Rice bran is a waste product with low cost and high fiber content, giving it an added advantage over corn and soybean meal, which have to be purchased and always at a relatively higher cost. Under the background of increased attention to sustainable agriculture, it is significant to find alternative uses for this byproduct. A total of 35 finishing pigs were allotted to five dietary treatments: a control group with basal diet and four experimental diets where corn was equivalently substituted by 7%, 14%, 21%, and 28% defatted rice bran (DFRB), respectively. With increasing levels of DFRB, the neutrophil to lymphocyte ratio (NLR) linearly decreased (p < 0.05). In the jejunum, the mRNA level of nuclear factor erythroid-2 related factor-2 (Nrf2) exhibited a quadratic response (p < 0.01) with incremental levels of DFRB. In the colon, the mRNA levels of mucin 2 (MUC2), Nrf2, and NAD(P)H: quinone oxidoreductase 1 (NQO1) were upregulated (linear, p < 0.05) and heme oxygenase-1 (HO-1) was upregulated (linear, p < 0.01). Overall, using DFRB to replace corn decreased the inflammatory biomarkers of serum and showed potential function in modulating the intestinal barrier by upregulating the mRNA expression levels of MUC2 and downregulating that of Nrf2, NQO1, and HO-1 in the colon.
Collapse
Affiliation(s)
- Lijuan Fan
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China; (L.F.); (R.H.); (C.W.); (Y.C.); (T.D.); (G.P.); (H.W.); (W.Z.)
- Huaian Academy, Nanjing Agricultural University, Huaian 223003, China
| | - Ruihua Huang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China; (L.F.); (R.H.); (C.W.); (Y.C.); (T.D.); (G.P.); (H.W.); (W.Z.)
- Huaian Academy, Nanjing Agricultural University, Huaian 223003, China
- Industrial Technology System Integration Innovation Center of Jiangsu Modern Agriculture (PIG), Nanjing 210095, China
| | - Chengwu Wu
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China; (L.F.); (R.H.); (C.W.); (Y.C.); (T.D.); (G.P.); (H.W.); (W.Z.)
- Huaian Academy, Nanjing Agricultural University, Huaian 223003, China
| | - Yang Cao
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China; (L.F.); (R.H.); (C.W.); (Y.C.); (T.D.); (G.P.); (H.W.); (W.Z.)
- Huaian Academy, Nanjing Agricultural University, Huaian 223003, China
| | - Taoran Du
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China; (L.F.); (R.H.); (C.W.); (Y.C.); (T.D.); (G.P.); (H.W.); (W.Z.)
- Huaian Academy, Nanjing Agricultural University, Huaian 223003, China
| | - Guang Pu
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China; (L.F.); (R.H.); (C.W.); (Y.C.); (T.D.); (G.P.); (H.W.); (W.Z.)
- Huaian Academy, Nanjing Agricultural University, Huaian 223003, China
| | - Huan Wang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China; (L.F.); (R.H.); (C.W.); (Y.C.); (T.D.); (G.P.); (H.W.); (W.Z.)
- Huaian Academy, Nanjing Agricultural University, Huaian 223003, China
| | - Wuduo Zhou
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China; (L.F.); (R.H.); (C.W.); (Y.C.); (T.D.); (G.P.); (H.W.); (W.Z.)
- Industrial Technology System Integration Innovation Center of Jiangsu Modern Agriculture (PIG), Nanjing 210095, China
| | - Pinghua Li
- Institute of Swine Science, Nanjing Agricultural University, Nanjing 210095, China; (L.F.); (R.H.); (C.W.); (Y.C.); (T.D.); (G.P.); (H.W.); (W.Z.)
- Huaian Academy, Nanjing Agricultural University, Huaian 223003, China
- Industrial Technology System Integration Innovation Center of Jiangsu Modern Agriculture (PIG), Nanjing 210095, China
- Nanjing Agricultural University’s New Rural Research and Development Corporation of Huaian City, Huaian 223003, China
- Correspondence:
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA;
| |
Collapse
|
481
|
Teichman EM, O'Riordan KJ, Gahan CGM, Dinan TG, Cryan JF. When Rhythms Meet the Blues: Circadian Interactions with the Microbiota-Gut-Brain Axis. Cell Metab 2020; 31:448-471. [PMID: 32130879 DOI: 10.1016/j.cmet.2020.02.008] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 12/18/2019] [Accepted: 02/10/2020] [Indexed: 12/19/2022]
Abstract
The microbiota-gut-brain axis encompasses a bidirectional mode of communication between the microorganisms residing in our gut, and our brain function and behavior. The composition of the gut microbiota is subject to diurnal variation and is entrained by host circadian rhythms. In turn, a diverse microbiota is essential for optimal regulation of host circadian pathways. Disruption of the cyclical nature of this microbe-host interaction profoundly influences disease pathology and severity. This review aims to summarize current knowledge on this bidirectional relationship. Indeed, the past few years have revealed promising data regarding the relationship between the microbiota-gut-brain axis and circadian rhythms and how they act in concert to influence disease, but further research needs to be done to examine how they coalesce to modulate severity of, and risk for, certain diseases. Moreover, there is a need for a greater understanding of the molecular mechanisms underlying the close relationship between circadian-microbiome-brain interactions.
Collapse
Affiliation(s)
| | | | - Cormac G M Gahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; School of Microbiology, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
482
|
Xing PY, Pettersson S, Kundu P. Microbial Metabolites and Intestinal Stem Cells Tune Intestinal Homeostasis. Proteomics 2020; 20:e1800419. [PMID: 31994831 DOI: 10.1002/pmic.201800419] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/07/2020] [Indexed: 12/13/2022]
Abstract
Microorganisms that colonize the gastrointestinal tract, collectively known as the gut microbiota, are known to produce small molecules and metabolites that significantly contribute to host intestinal development, functions, and homeostasis. Emerging insights from microbiome research reveal that gut microbiota-derived signals and molecules influence another key player maintaining intestinal homeostasis-the intestinal stem cell niche, which regulates epithelial self-renewal. In this review, the literature on gut microbiota-host crosstalk is surveyed, highlighting the effects of gut microbial metabolites on intestinal stem cells. The production of various classes of metabolites, their actions on intestinal stem cells are discussed and, finally, how the production and function of metabolites are modulated by aging and dietary intake is commented upon.
Collapse
Affiliation(s)
- Peter Yuli Xing
- Singapore Centre for Environmental Life Sciences Engineering, 60 Nanyang Drive, Singapore, 637551, Singapore.,Interdisciplinary Graduate School, Nanyang Technological University, 50 Nanyang Avenue, South Spine, Level B3, Block S2-B3a, Singapore, 639798, Singapore
| | - Sven Pettersson
- Singapore Centre for Environmental Life Sciences Engineering, 60 Nanyang Drive, Singapore, 637551, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore.,Department of Neurobiology, Care Sciences and Society, Karolinska Institute, SE, 17 177, Stockholm, Sweden
| | - Parag Kundu
- Singapore Centre for Environmental Life Sciences Engineering, 60 Nanyang Drive, Singapore, 637551, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore.,The Center for Microbes, Development and Health, Laboratory for Microbiota-Host Interactions, Institute Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Life Science Research Building, Shanghai, 200031, China
| |
Collapse
|
483
|
Wang S, Ahmadi S, Nagpal R, Jain S, Mishra SP, Kavanagh K, Zhu X, Wang Z, McClain DA, Kritchevsky SB, Kitzman DW, Yadav H. Lipoteichoic acid from the cell wall of a heat killed Lactobacillus paracasei D3-5 ameliorates aging-related leaky gut, inflammation and improves physical and cognitive functions: from C. elegans to mice. GeroScience 2020; 42:333-352. [PMID: 31814084 PMCID: PMC7031475 DOI: 10.1007/s11357-019-00137-4] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/12/2019] [Indexed: 12/22/2022] Open
Abstract
Increased inflammation associated with leaky gut is a major risk factor for morbidity and mortality in older adults; however, successful preventive and therapeutic strategies against these conditions are not available. In this study, we demonstrate that a human-origin Lactobacillus paracasei D3-5 strain (D3-5), even in the non-viable form, extends life span of Caenorhabditis elegans. In addition, feeding of heat-killed D3-5 to old mice (> 79 weeks) prevents high- fat diet-induced metabolic dysfunctions, decreases leaky gut and inflammation, and improves physical and cognitive functions. D3-5 feeding significantly increases mucin production, and proportionately, the abundance of mucin-degrading bacteria Akkermansia muciniphila also increases. Mechanistically, we show that the lipoteichoic acid (LTA), a cell wall component of D3-5, enhances mucin (Muc2) expression by modulating TLR-2/p38-MAPK/NF-kB pathway, which in turn reduces age-related leaky gut and inflammation. The findings indicate that the D3-5 and its LTA can prevent/treat age-related leaky gut and inflammation.
Collapse
Affiliation(s)
- Shaohua Wang
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Shokouh Ahmadi
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ravinder Nagpal
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Shalini Jain
- Department of Endocrinology and Metabolism, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Mouse Metabolic Phenotyping Core, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sidharth P Mishra
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kylie Kavanagh
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Pathology-Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Biomedical Sciences, University of Tasmania, Hobart, Australia
| | - Xuewei Zhu
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Zhan Wang
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Donald A McClain
- Department of Endocrinology and Metabolism, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Mouse Metabolic Phenotyping Core, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Stephen B Kritchevsky
- Department of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Dalane W Kitzman
- Department of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Cardiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Hariom Yadav
- Department of Internal Medicine-Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
484
|
Min S, Kim S, Cho SW. Gastrointestinal tract modeling using organoids engineered with cellular and microbiota niches. Exp Mol Med 2020; 52:227-237. [PMID: 32103122 PMCID: PMC7062772 DOI: 10.1038/s12276-020-0386-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/21/2019] [Accepted: 12/27/2019] [Indexed: 02/08/2023] Open
Abstract
The recent emergence of organoid technology has attracted great attention in gastroenterology because the gastrointestinal (GI) tract can be recapitulated in vitro using organoids, enabling disease modeling and mechanistic studies. However, to more precisely emulate the GI microenvironment in vivo, several neighboring cell types and types of microbiota need to be integrated into GI organoids. This article reviews the recent progress made in elucidating the crosstalk between GI organoids and components of their microenvironment. We outline the effects of stromal cells (such as fibroblasts, neural cells, immune cells, and vascular cells) on the gastric and intestinal epithelia of organoids. Because of the important roles that microbiota play in the physiology and function of the GI tract, we also highlight interactions between organoids and commensal, symbiotic, and pathogenic microorganisms and viruses. GI organoid models that contain niche components will provide new insight into gastroenterological pathophysiology and disease mechanisms.
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Suran Kim
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
485
|
Kim HW, Ju DB, Kye YC, Ju YJ, Kim CG, Lee IK, Park SM, Choi IS, Cho KK, Lee SH, Kim SC, Jung ID, Han SH, Yun CH. Galectin-9 Induced by Dietary Probiotic Mixture Regulates Immune Balance to Reduce Atopic Dermatitis Symptoms in Mice. Front Immunol 2020; 10:3063. [PMID: 32038618 PMCID: PMC6987441 DOI: 10.3389/fimmu.2019.03063] [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: 08/20/2019] [Accepted: 12/16/2019] [Indexed: 12/22/2022] Open
Abstract
Probiotics can be an effective treatment for atopic dermatitis (AD), while their mechanism of action is still unclear. Here, we induced AD in mice with 2,4-dinitrochlorobenzene and administrated YK4, a probiotic mixture consisting of Lactobacillus acidophilus CBT LA1, L. plantarum CBT LP3, Bifidobacterium breve CBT BR3, and B. lactis CBT BL3. Then, we have validated the underlying mechanism for the alleviation of AD by YK4 from the intestinal and systematic immunological perspectives. Administration of YK4 in AD mice alleviated the symptoms of AD by suppressing the expression of skin thymic stromal lymphopoietin and serum immunoglobulin E eliciting excessive T-helper (Th) 2 cell-mediated responses. YK4 inhibited Th2 cell population through induce the proportion of Th1 cells in spleen and Treg cells in Peyer's patches and mesenteric lymph node (mLN). CD103+ dendritic cells (DCs) in mLN and the spleen were significantly increased in AD mice administered with YK4 when compared to AD mice. Furthermore, galectin-9 was significantly increased in the gut of AD mice administered with YK4. In vitro experiments were performed using bone marrow-derived DCs (BMDC) and CD4+ T cells to confirm the immune mechanisms of YK4 and galectin-9. The expression of CD44, a receptor of galectin-9, together with programmed death-ligand 1 was significantly upregulated in BMDCs following treatment with YK4. IL-10 and IL-12 were upregulated when BMDCs were treated with YK4. Cytokines together with co-receptors from DCs play a major role in the differentiation and activation of CD4+ T cells. Proliferation of Tregs and Th1 cell activation were enhanced when CD4+T cells were co-cultured with YK4-treated BMDCs. Galectin-9 appeared to contribute at least partially to the proliferation of Tregs. The results further suggested that DCs treated with YK4 induced the differentiation of naïve T cells toward Th1 and Tregs. At the same time, YK4 alleviated AD symptoms by inhibiting Th2 response. Thus, the present study suggested a potential role of YK4 as an effective immunomodulatory agent in AD patients.
Collapse
Affiliation(s)
- Han Wool Kim
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Do Bin Ju
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Yoon-Chul Kye
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Young-Jun Ju
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Cheol Gyun Kim
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - In Kyu Lee
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - Sung-Moo Park
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea
| | - In Soon Choi
- Department of Biological Science, College of Medical and Life Sciences, Silla University, Busan, South Korea
| | - Kwang Keun Cho
- Department of Animal Resources Technology, Gyeongnam National University of Science and Technology, Jinju, South Korea
| | - Seung Ho Lee
- Department of Nano-Bioengineering, Incheon National University, Incheon, South Korea
| | - Sung Chan Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon, South Korea
| | - In Duk Jung
- Laboratory of Dendritic Cell Differentiation and Regulation, Department of Immunology, School of Medicine, Konkuk University, Chungju, South Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, Dental Research Institute and Brain Korea 21 Plus Program, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, South Korea.,Institute of Green Bio Science Technology, Seoul National University, Seoul, South Korea.,Center for Food and Bioconvergence, Seoul National University, Seoul, South Korea
| |
Collapse
|
486
|
González-Miguel J, Becerro-Recio D, Sotillo J, Simón F, Siles-Lucas M. Set up of an in vitro model to study early host-parasite interactions between newly excysted juveniles of Fasciola hepatica and host intestinal cells using a quantitative proteomics approach. Vet Parasitol 2020; 278:109028. [PMID: 31986420 DOI: 10.1016/j.vetpar.2020.109028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 12/20/2022]
Abstract
Fasciola hepatica is the causative agent of fasciolosis, a parasitic zoonosis of global distribution causing significant economic losses in animal production and a human public health problem in low-income countries. Hosts are infected by ingestion of aquatic plants carrying metacercariae. Once ingested, the juvenile parasites excyst in the small intestine and, after crossing it, they follow a complex migratory route that lead the parasites to their definitive location in the bile ducts. Despite being a critical event in the progression of the infection, the available data on the cross-talk relationships between the parasite and the host at an early stage of the infection are scarce. The objective of the present work is to characterize the proteomic changes occurring in both the parasite and the host, through the development of a novel in vitro model, to shed light on the molecular pathways of communication between the newly excysted juveniles (NEJ) from F. hepatica and the host's intestinal epithelium. For this, in vitro excystation of F. hepatica metacercariae was carried out and NEJ were obtained. Additionally, optimal conditions of growth and expansion of mouse primary small intestinal epithelial cells (MPSIEC) in culture were fine-tuned. Tegumentary and somatic parasite antigens (NEJ-Teg and NEJ-Som), as well as host cell protein lysate (MPSIEC-Lys) were obtained before and after 24 h co-culture of NEJ with MPSIEC. We used an isobaric tags for relative and absolute quantitation (iTRAQ)-based strategy to detect 191 and 62 up-regulated, and 112 and 57 down-regulated proteins in the NEJ-Teg and NEJ-Som extracts, respectively. Similarly, 87 up-regulated and 73 down-regulated proteins in the MPSIEC-Lys extract were identified. Taking into account the biological processes in which these proteins were involved, interesting mechanisms related to parasite development, invasion and evasion, as well as manipulation of the host intestinal epithelial cell adhesion, immunity and apoptosis pathways, among others, could be inferred, taking place at the host-parasite interface. The further understanding of these processes could constitute promising therapeutic targets in the future against fasciolosis.
Collapse
Affiliation(s)
- Javier González-Miguel
- Institute of Natural Resources and Agrobiology (IRNASA, CSIC), Sustainable Development Department, C/Cordel de Merinas, 52, 37008, Salamanca, Spain; Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov University, Moscow, Russia.
| | - David Becerro-Recio
- Institute of Natural Resources and Agrobiology (IRNASA, CSIC), Sustainable Development Department, C/Cordel de Merinas, 52, 37008, Salamanca, Spain
| | - Javier Sotillo
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain; Centre for Molecular Therapeutics, Australian Institute for Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Fernando Simón
- Laboratory of Parasitology, Faculty of Pharmacy, University of Salamanca, 37007, Salamanca, Spain
| | - Mar Siles-Lucas
- Institute of Natural Resources and Agrobiology (IRNASA, CSIC), Sustainable Development Department, C/Cordel de Merinas, 52, 37008, Salamanca, Spain
| |
Collapse
|
487
|
Targeting NLRP3 Inflammasome in Inflammatory Bowel Disease: Putting out the Fire of Inflammation. Inflammation 2020; 42:1147-1159. [PMID: 30937839 DOI: 10.1007/s10753-019-01008-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Inflammatory bowel disease (IBD) is a group of inflammatory conditions of the colon and small intestine, comprised of ulcerative colitis and Crohn's disease. Among the complicated pathogenic factors of IBD, the overaction of inflammatory and immune reaction serves as an important factor. Inflammasome is a form of innate immunity as well as inflammation. Among all kinds of inflammasomes, the NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome is the most studied one, and has been revealed to be involved in the pathogenesis and progression of IBD. Here, in this review, the association between the NLRP3 inflammasome and IBD will be discussed. Furthermore, several NLRP3 inflammasome inhibitors which have been demonstrated to be effective in the alleviation of IBD will be described in this review.
Collapse
|
488
|
Liu Q, Liu Q, Meng H, Lv H, Liu Y, Liu J, Wang H, He L, Qin J, Wang Y, Dai Y, Otto M, Li M. Staphylococcus epidermidis Contributes to Healthy Maturation of the Nasal Microbiome by Stimulating Antimicrobial Peptide Production. Cell Host Microbe 2020; 27:68-78.e5. [PMID: 31866425 PMCID: PMC10988655 DOI: 10.1016/j.chom.2019.11.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 08/19/2019] [Accepted: 11/07/2019] [Indexed: 12/18/2022]
Abstract
The composition of the human microbiome profoundly impacts human well-being. However, the mechanisms underlying microbiome maturation are poorly understood. The nasal microbiome is of particular importance as a source of many respiratory infections. Here, we performed a large sequencing and culture-based analysis of the human nasal microbiota from different age groups. We observed a significant decline of pathogenic bacteria before adulthood, with an increase of the commensal Staphylococcus epidermidis. In seniors, this effect was partially reversed. In vitro, many S. epidermidis isolates stimulated nasal epithelia to produce antimicrobial peptides, killing pathogenic competitors, while S. epidermidis itself proved highly resistant owing to its exceptional capacity to form biofilms. Furthermore, S. epidermidis isolates with high antimicrobial peptide-inducing and biofilm-forming capacities outcompeted pathogenic bacteria during nasal colonization in vivo. Our study identifies a pivotal role of S. epidermidis in healthy maturation of the nasal microbiome, which is achieved at least in part by symbiotic cooperation with innate host defense.
Collapse
Affiliation(s)
- Qian Liu
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Qingyun Liu
- Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China
| | - Hongwei Meng
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Huiying Lv
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yao Liu
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Junlan Liu
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Hua Wang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Lei He
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Juanxiu Qin
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yanan Wang
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Yingxin Dai
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD 20814, USA.
| | - Min Li
- Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China.
| |
Collapse
|
489
|
González-Quilen C, Rodríguez-Gallego E, Beltrán-Debón R, Pinent M, Ardévol A, Blay MT, Terra X. Health-Promoting Properties of Proanthocyanidins for Intestinal Dysfunction. Nutrients 2020; 12:E130. [PMID: 31906505 PMCID: PMC7019584 DOI: 10.3390/nu12010130] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/20/2019] [Accepted: 12/31/2019] [Indexed: 12/22/2022] Open
Abstract
The intestinal barrier is constantly exposed to potentially harmful environmental factors, including food components and bacterial endotoxins. When intestinal barrier function and immune homeostasis are compromised (intestinal dysfunction), inflammatory conditions may develop and impact overall health. Evidence from experimental animal and cell culture studies suggests that exposure of intestinal mucosa to proanthocyanidin (PAC)-rich plant products, such as grape seeds, may contribute to maintaining the barrier function and to ameliorating the pathological inflammation present in diet-induced obesity and inflammatory bowel disease. In this review, we aim to update the current knowledge on the bioactivity of PACs in experimental models of intestinal dysfunction and in humans, and to provide insights into the underlying biochemical and molecular mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | - M Teresa Blay
- MoBioFood Research Group, Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43007 Tarragona, Spain; (C.G.-Q.); (E.R.-G.); (R.B.-D.); (M.P.); (A.A.); (X.T.)
| | | |
Collapse
|
490
|
Yang J, Zhang J, Zhao C, Gai Z, Mu X, Wang Y, Zhang C, Su Z, Gao L, Zhu D, Zuo Z, Heng X, Zhang L. Blood Loss Leads to Increase in Relative Abundance of Opportunistic Pathogens in the Gut Microbiome of Rabbits. Curr Microbiol 2020; 77:415-424. [PMID: 31894374 DOI: 10.1007/s00284-019-01825-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/25/2019] [Indexed: 01/26/2023]
Abstract
Massive blood loss, a common pathological complication in the clinic, is often accompanied by altered gut integrity and intestinal wall damage. Little is known to what extent the gut microbiome could be correlated with this process. The gut microbiome plays a crucial role in human health, especially in immune and inflammatory responses. This study aims to determine whether acute blood loss affects the gut microbiome and the dynamic variation of the gut microbiome following the loss of blood. We used New Zealand rabbits to mimic the blood loss complication and designed a five-time-point fecal sampling strategy including 24-h pre-blood loss procedure, 24 h, 36 h, 48 h, and 1-week post-blood loss procedure. Gut microbiome composition and diversity were analyzed using 16S rRNA gene sequencing and downstream α-diversity, β-diversity, and taxonomy analysis. The gut microbiome changed dramatically after blood loss procedure. There was a significant increase in diversity and richness of the gut microbiome at 24-h post-procedure (P = 0.038). Based on an analysis of similarities, the composition of gut microbiome in the samples collected at 24-h post-procedure was significantly different from that of pre-procedure samples (r = 0.79, P = 0.004 weighted unifrac distance; r = 0.99, P = 0.002, unweighted unifrac distance). The relative abundance of Lactobacillus was significantly decreased in the post-procedure samples (P = 0.0006), while the relative abundance of Clostridiales (P = 0.018) and Bacteroidales (P = 0.015) was significantly increased after procedure. We also found the relative abundance of Bacilli, Lactobacillus, Myroides, and Prevotella decreased gradually at different time points after blood loss. The relative abundance of the Clostridia, Alphaproteobacteria, and Sporosarcina increased at 24-h post-procedure and decreased thereafter. This preliminary study discovered potential connections between blood loss and dysbiosis of gut microbiome. The diversity and abundance of the gut microbiome was affected to various extents after acute blood loss and unable to be restored to the original microbiome profile even after one week. The increase in relative abundance of opportunistic pathogens after blood loss could be an important indication to reconsider immune and inflammatory responses after acute blood loss from the perspective of gut microbiome.
Collapse
Affiliation(s)
- Junjie Yang
- Microbiological Laboratory, Department of Infection Management, Department of Neurosurgery, Lin Yi People's Hospital, Linyi, 276000, Shandong, China.,College of Life Science, Qilu Normal University, Jinan, 250200, Shandong, China
| | - Jiaming Zhang
- Microbiome-X Group, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Chemistry and Environment, Beihang University, Beijing, 100191, China.,Shandong Institutes for Food and Drug Control, Jinan, 250101, Shandong, China
| | - Changying Zhao
- Shandong Children's Microbiome Center, Qilu Children's Hospital of Shandong University, Jinan, 250022, Shandong, China.,Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, Jinan, 250100, Shandong, China
| | - Zhongtao Gai
- Shandong Children's Microbiome Center, Qilu Children's Hospital of Shandong University, Jinan, 250022, Shandong, China.,Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, Jinan, 250100, Shandong, China
| | - Xiaofeng Mu
- Clinical Laboratory and Core Research Laboratory, Qingdao Human Microbiome Center, The Affiliated Central Hospital of QingdaoUniversity, Qingdao, 266042, Shandong, China
| | - Ye Wang
- Clinical Laboratory and Core Research Laboratory, Qingdao Human Microbiome Center, The Affiliated Central Hospital of QingdaoUniversity, Qingdao, 266042, Shandong, China
| | - Chunling Zhang
- Department of Respiratory Medicine, The Affiliated Central Hospital of Qingdao University, Qingdao, 266042, Shandong, China
| | - Zhenzhen Su
- Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, Jinan, 250100, Shandong, China
| | - Lihe Gao
- Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, Jinan, 250100, Shandong, China
| | - Dequan Zhu
- Microbiological Laboratory, Department of Infection Management, Department of Neurosurgery, Lin Yi People's Hospital, Linyi, 276000, Shandong, China
| | - Zhiwen Zuo
- Microbiological Laboratory, Department of Infection Management, Department of Neurosurgery, Lin Yi People's Hospital, Linyi, 276000, Shandong, China
| | - Xueyuan Heng
- Microbiological Laboratory, Department of Infection Management, Department of Neurosurgery, Lin Yi People's Hospital, Linyi, 276000, Shandong, China.
| | - Lei Zhang
- Microbiome-X Group, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Chemistry and Environment, Beihang University, Beijing, 100191, China. .,Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, Jinan, 250100, Shandong, China.
| |
Collapse
|
491
|
Capdevila C, Calderon RI, Bush EC, Sheldon-Collins K, Sims PA, Yan KS. Single-Cell Transcriptional Profiling of the Intestinal Epithelium. Methods Mol Biol 2020; 2171:129-153. [PMID: 32705639 DOI: 10.1007/978-1-0716-0747-3_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Emerging single-cell technologies, like single-cell RNA sequencing (scRNA-seq), enable the study of heterogeneous biological systems at cellular resolution. By profiling the set of expressed transcripts in each cell, single-cell transcriptomics has allowed for the cataloging of the cellular constituents of multiple organs and tissues, both in health and disease. In addition, these technologies have provided mechanistic insights into cellular function, cell state transitions, developmental trajectories and lineage relationships, as well as helped to dissect complex, population-level responses to environmental perturbations. scRNA-seq is particularly useful for characterizing the intestinal epithelium because it is a dynamic, rapidly self-renewing tissue comprised of more than a dozen specialized cell types. Here we discuss the fundamentals of single-cell transcriptomics of the murine small intestinal epithelium. We review the principles of proper experimental design and provide methods for the dissociation of the small intestinal epithelium into single cells followed by fluorescence-activated cell sorting (FACS) and for scRNA-seq using the 10× Genomics Chromium platform.
Collapse
Affiliation(s)
- Claudia Capdevila
- Columbia Center for Human Development, Columbia Stem Cell Initiative, Division of Digestive & Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Ruben I Calderon
- Columbia Center for Human Development, Columbia Stem Cell Initiative, Division of Digestive & Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Erin C Bush
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Kismet Sheldon-Collins
- Columbia Center for Human Development, Columbia Stem Cell Initiative, Division of Digestive & Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Peter A Sims
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biochemistry & Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Kelley S Yan
- Columbia Center for Human Development, Columbia Stem Cell Initiative, Division of Digestive & Liver Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Genetics & Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
492
|
Li HZ, Li N, Wang JJ, Li H, Huang X, Guo L, Zheng HW, He ZL, Zhao Y, Yang ZN, Fan HT, Chu MM, Yang JX, Wu QW, Liu LD. Dysbiosis of gut microbiome affecting small intestine morphology and immune balance: a rhesus macaque model. Zool Res 2020; 41:20-31. [PMID: 31930784 PMCID: PMC6956715 DOI: 10.24272/j.issn.2095-8137.2020.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
There is a growing appreciation for the specific health benefits conferred by commensal microbiota on their hosts. Clinical microbiota analysis and animal studies in germ-free or antibiotic-treated mice have been crucial for improving our understanding of the role of the microbiome on the host mucosal surface; however, studies on the mechanisms involved in microbiome-host interactions remain limited to small animal models. Here, we demonstrated that rhesus monkeys under short-term broad-spectrum antibiotic treatment could be used as a model to study the gut mucosal host-microbiome niche and immune balance with steady health status. Results showed that the diversity and community structure of the gut commensal bacteria in rhesus monkeys were both disrupted after antibiotic treatment. Furthermore, the 16S rDNA amplicon sequencing results indicated that Escherichia-Shigella were predominant in stool samples 9 d of treatment, and the abundances of bacterial functional genes and predicted KEGG pathways were significantly changed. In addition to inducing aberrant morphology of small intestinal villi, the depletion of gut commensal bacteria led to increased proportions of CD3 + T, CD4 + T, and CD16 + NK cells in peripheral blood mononuclear cells (PBMCs), but decreased numbers of Treg and CD20 + B cells. The transcriptome of PBMCs from antibiotic-treated monkeys showed that the immune balance was affected by modulation of the expression of many functional genes, including IL-13, VCAM1, and LGR4.
Collapse
Affiliation(s)
- Hong-Zhe Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Nan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Jing-Jing Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Heng Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Xing Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Lei Guo
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Hui-Wen Zheng
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Zhan-Long He
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China
| | - Yuan Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China
| | - Ze-Ning Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Hai-Tao Fan
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Man-Man Chu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Jin-Xi Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Qiong-Wen Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China
| | - Long-Ding Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan 650118, China.,Key Laboratory of Systemic Innovative Research on Virus Vaccine, Chinese Academy of Medical Sciences, Kunming, Yunnan 650118, China. E-mail:
| |
Collapse
|
493
|
Xiang Y, Wen H, Yu Y, Li M, Fu X, Huang S. Gut-on-chip: Recreating human intestine in vitro. J Tissue Eng 2020; 11:2041731420965318. [PMID: 33282173 PMCID: PMC7682210 DOI: 10.1177/2041731420965318] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/22/2020] [Indexed: 01/04/2023] Open
Abstract
The human gut is important for food digestion and absorption, as well as a venue for a large number of microorganisms that coexist with the host. Although numerous in vitro models have been proposed to study intestinal pathology or interactions between intestinal microbes and host, they are far from recapitulating the real intestinal microenvironment in vivo. To assist researchers in further understanding gut physiology, the intestinal microbiome, and disease processes, a novel technology primarily based on microfluidics and cell biology, called "gut-on-chip," was developed to simulate the structure, function, and microenvironment of the human gut. In this review, we first introduce various types of gut-on-chip systems, then highlight their applications in drug pharmacokinetics, host-gut microbiota crosstalk, and nutrition metabolism. Finally, we discuss challenges in this field and prospects for better understanding interactions between intestinal flora and human hosts, and then provide guidance for clinical treatment of related diseases.
Collapse
Affiliation(s)
- Yunqing Xiang
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hui Wen
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yue Yu
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiongfei Fu
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuqiang Huang
- CAS Key Laboratory of Quantitative Engineering Biology, Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
494
|
Haupeltshofer S, Leichsenring T, Berg S, Pedreiturria X, Joachim SC, Tischoff I, Otte JM, Bopp T, Fantini MC, Esser C, Willbold D, Gold R, Faissner S, Kleiter I. Smad7 in intestinal CD4 + T cells determines autoimmunity in a spontaneous model of multiple sclerosis. Proc Natl Acad Sci U S A 2019; 116:25860-25869. [PMID: 31796589 PMCID: PMC6926056 DOI: 10.1073/pnas.1905955116] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Environmental triggers acting at the intestinal barrier are thought to contribute to the initiation of autoimmune disorders. The transforming growth factor beta inhibitor Smad7 determines the phenotype of CD4+ T cells. We hypothesized that Smad7 in intestinal CD4+ T cells controls initiation of opticospinal encephalomyelitis (OSE), a murine model of multiple sclerosis (MS), depending on the presence of gut microbiota. Smad7 was overexpressed or deleted in OSE CD4+ T cells to determine the effect on clinical progression, T cell differentiation, and T cell migration from the intestine to the central nervous system (CNS). Smad7 overexpression worsened the clinical course of OSE and increased CNS inflammation and demyelination. It favored expansion of intestinal CD4+ T cells toward an inflammatory phenotype and migration of intestinal CD4+ T cells to the CNS. Intestinal biopsies from MS patients revealed decreased transforming growth factor beta signaling with a shift toward inflammatory T cell subtypes. Smad7 in intestinal T cells might represent a valuable therapeutic target for MS to achieve immunologic tolerance in the intestine and suppress CNS inflammation.
Collapse
Affiliation(s)
- Steffen Haupeltshofer
- St. Josef-Hospital, Department of Neurology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Teresa Leichsenring
- St. Josef-Hospital, Department of Neurology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Sarah Berg
- St. Josef-Hospital, Department of Neurology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Xiomara Pedreiturria
- St. Josef-Hospital, Department of Neurology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Stephanie C Joachim
- University Eye Clinic, Experimental Eye Research Institute, Ruhr-University Bochum, 44892 Bochum, Germany
| | - Iris Tischoff
- Institut für Pathologie, Bergmannsheil, 44789 Bochum, Germany
| | - Jan-Michel Otte
- Department of Internal Medicine I, Klinikum Links der Weser, 28277 Bremen, Germany
| | - Tobias Bopp
- Institute for Immunology, Universitätsmedizin Mainz, 55131 Mainz, Germany
- Research Center for Immunotherapy (FZI), Universitätsmedizin Mainz, 55131 Mainz, Germany
| | - Massimo C Fantini
- Department of Systems Medicine, University of Rome "Tor Vergata," 00133 Roma RM, Italy
| | - Charlotte Esser
- Leibniz-Institut für Umweltmedizinische Forschung, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Dieter Willbold
- Institut für Physikalische Biologie, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- Institute of Complex Systems (ICS-6), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Ralf Gold
- St. Josef-Hospital, Department of Neurology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Simon Faissner
- St. Josef-Hospital, Department of Neurology, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Ingo Kleiter
- St. Josef-Hospital, Department of Neurology, Ruhr-University Bochum, 44791 Bochum, Germany;
- Marianne-Strauss-Klinik, Behandlungszentrum Kempfenhausen für Multiple Sklerose Kranke gGmbH, 82335 Berg, Germany
| |
Collapse
|
495
|
Mechanisms Underlying Bone Loss Associated with Gut Inflammation. Int J Mol Sci 2019; 20:ijms20246323. [PMID: 31847438 PMCID: PMC6940820 DOI: 10.3390/ijms20246323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/29/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
Patients with gastrointestinal diseases frequently suffer from skeletal abnormality, characterized by reduced bone mineral density, increased fracture risk, and/or joint inflammation. This pathological process is characterized by altered immune cell activity and elevated inflammatory cytokines in the bone marrow microenvironment due to disrupted gut immune response. Gastrointestinal disease is recognized as an immune malfunction driven by multiple factors, including cytokines and signaling molecules. However, the mechanism by which intestinal inflammation magnified by gut-residing actors stimulates bone loss remains to be elucidated. In this article, we discuss the main risk factors potentially contributing to intestinal disease-associated bone loss, and summarize current animal models, illustrating gut-bone axis to bridge the gap between intestinal inflammation and skeletal disease.
Collapse
|
496
|
Xie S, Zhao S, Jiang L, Lu L, Yang Q, Yu Q. Lactobacillus reuteri Stimulates Intestinal Epithelial Proliferation and Induces Differentiation into Goblet Cells in Young Chickens. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:13758-13766. [PMID: 31789514 DOI: 10.1021/acs.jafc.9b06256] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Probiotics, such as Lactobacillus, have been proven to be effective in maintaining intestinal homeostasis. The modulatory effect of Lactobacillus on intestinal epithelial development in early life is still unclear. In this study, Lactobacillus isolates with good probiotic abilities were screened and orally administered to detect their regulatory effect on intestinal development in chickens. L. reuteri 22 was isolated from chickens and chosen for subsequent chicken experiments due to its strong acid and bile salt resistance and ability to adhere to epithelial cells. The 3-day-old chickens were orally administrated with 108 CFU L. reuteri 22 for consecutive 7 days. L. reuteri 22 increased Lgr5 mRNA expression (3.23 ± 0.40, P = 0.001) and activated the Wnt/β-catenin signaling pathway, with increasing expression of proliferating cell nuclear antigen (PCNA) (49.27 ± 9.81, P = 0.021) to support the proliferation of chicken intestinal epithelial cells. Moreover, L. reuteri 22 also inhibited the Notch signaling pathway to induce intestinal stem cell differentiation into goblet cells with increased mucin 2 (Muc-2) expression (1.72 ± 0.34, P = 0.047). L. reuteri 22 significantly enhanced lysozyme mRNA expression (2.32 ± 0.55, P = 0.019) to improve intestinal innate mucosal immunity. This study demonstrated that L. reuteri administration could regulate chicken intestinal epithelium development to ensure the function of the intestinal mucosal barrier, which is beneficial for newborn animals.
Collapse
Affiliation(s)
- Shuang Xie
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine , Nanjing Agricultural University , Weigang 1 , Nanjing , Jiangsu 210095 , P.R. China
| | - Shiyi Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine , Nanjing Agricultural University , Weigang 1 , Nanjing , Jiangsu 210095 , P.R. China
| | - Lan Jiang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine , Nanjing Agricultural University , Weigang 1 , Nanjing , Jiangsu 210095 , P.R. China
| | - Linhao Lu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine , Nanjing Agricultural University , Weigang 1 , Nanjing , Jiangsu 210095 , P.R. China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine , Nanjing Agricultural University , Weigang 1 , Nanjing , Jiangsu 210095 , P.R. China
| | - Qinghua Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine , Nanjing Agricultural University , Weigang 1 , Nanjing , Jiangsu 210095 , P.R. China
| |
Collapse
|
497
|
Thoo L, Noti M, Krebs P. Keep calm: the intestinal barrier at the interface of peace and war. Cell Death Dis 2019; 10:849. [PMID: 31699962 PMCID: PMC6838056 DOI: 10.1038/s41419-019-2086-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 10/11/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022]
Abstract
Epithelial barriers have to constantly cope with both harmless and harmful stimuli. The epithelial barrier therefore serves as a dynamic and not static wall to safeguard its proper physiological function while ensuring protection. This is achieved through multiple defence mechanisms involving various cell types - epithelial and non-epithelial - that work in an integrated manner to build protective barriers at mucosal sites. Damage may nevertheless occur, due to pathogens, physical insults or dysregulated immune responses, which trigger a physiologic acute or a pathologic chronic inflammatory cascade. Inflammation is often viewed as a pathological condition, particularly due to the increasing prevalence of chronic inflammatory (intestinal) diseases. However, inflammation is also necessary for wound healing. The aetiology of chronic inflammatory diseases is incompletely understood and identification of the underlying mechanisms would reveal additional therapeutic approaches. Resolution is an active host response to end ongoing inflammation but its relevance is under-appreciated. Currently, most therapies aim at dampening inflammation at damaged mucosal sites, yet these approaches do not efficiently shut down the inflammation process nor repair the epithelial barrier. Therefore, future treatment strategies should also promote the resolution phase. Yet, the task of repairing the barrier can be an arduous endeavour considering its multiple integrated layers of defence - which is advantageous for damage prevention but becomes challenging to repair at multiple levels. In this review, using the intestines as a model epithelial organ and barrier paradigm, we describe the consequences of chronic inflammation and highlight the importance of the mucosae to engage resolving processes to restore epithelial barrier integrity and function. We further discuss the contribution of pre-mRNA alternative splicing to barrier integrity and intestinal homeostasis. Following discussions on current open questions and challenges, we propose a model in which resolution of inflammation represents a key mechanism for the restoration of epithelial integrity and function.
Collapse
Affiliation(s)
- Lester Thoo
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Mario Noti
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland.,Department of Gastro-Intestinal Health, Immunology, Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Philippe Krebs
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
498
|
Li XV, Leonardi I, Iliev ID. Gut Mycobiota in Immunity and Inflammatory Disease. Immunity 2019; 50:1365-1379. [PMID: 31216461 DOI: 10.1016/j.immuni.2019.05.023] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/01/2019] [Accepted: 05/29/2019] [Indexed: 02/08/2023]
Abstract
The mammalian intestine is colonized by a wealth of microorganisms-including bacteria, viruses, protozoa, and fungi-that are all integrated into a functional trans-kingdom community. Characterization of the composition of the fungal community-the mycobiota-has advanced further than the much-needed mechanistic studies. Recent findings have revealed roles for the gut mycobiota in the regulation of host immunity and in the development and progression of human diseases of inflammatory origin. We review these findings here while placing them in the context of the current understanding of the pathways and cellular networks that induce local and systemic immune responses to fungi in the gastrointestinal tract. We discuss gaps in knowledge and argue for the importance of considering bacteria-fungal interactions as we aim to define the roles of mycobiota in immune homeostasis and immune-associated pathologies.
Collapse
Affiliation(s)
- Xin V Li
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Irina Leonardi
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Iliyan D Iliev
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
499
|
Sun X, Yao L, Liang H, Wang D, He Y, Wei Y, Ye L, Wang K, Li L, Chen J, Zhang CY, Xu G, Wang F, Zen K. Intestinal epithelial PKM2 serves as a safeguard against experimental colitis via activating β-catenin signaling. Mucosal Immunol 2019; 12:1280-1290. [PMID: 31462699 DOI: 10.1038/s41385-019-0197-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/22/2019] [Accepted: 08/08/2019] [Indexed: 02/04/2023]
Abstract
The pyruvate kinase M2 (PKM2)-mediated aerobic glycolysis has been shown to play a critical role in promoting cell survival and proliferation. However, little is known about the function of intestinal epithelial PKM2 in intestine homeostasis. Here we investigate whether and how intestinal epithelial PKM2 modulates the morphology and function of the adult intestine in experimental colitis. Analyzing colonoscopic biopsies from Crohn's disease and ulcerative colitis patients, we found significantly decreased level of intestinal epithelial PKM2 in patients compared to that in non-inflamed tissues. Similar reduction of intestinal epithelial PKM2 was observed in mice with dextran sulfate sodium-induced colitis. Moreover, intestinal epithelial-specific PKM2-knockout (Pkm2-/-) mice displayed more severe intestinal inflammation, as evidenced by a shortened colon, disruption of epithelial tight junctions, an increase in inflammatory cytokine levels, and immune cell infiltration, when compared to wild-type mice. Gene profiling, western blot, and function analyses indicated that cell survival signals, particularly the Wnt/β-catenin pathways, were associated with PKM2 activity. Increasing mouse intestinal epithelial PKM2 expression via delivery of a PKM2-expressing plasmid attenuated experimental colitis. In conclusion, our studies demonstrate that intestinal epithelial PKM2 increases cell survival and wound healing under the colitic condition via activating the Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Xinlei Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Li Yao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Hongwei Liang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Dong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Yueqin He
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Yao Wei
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, China
| | - Lei Ye
- Department of Gastroenterology and Hepatology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210093, China
| | - Kai Wang
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, No. 305 East Zhongshan Road, Nanjing, Jiangsu, 210002, China
| | - Limin Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Jiangning Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Chen-Yu Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China
| | - Guifang Xu
- Department of Gastroenterology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210008, China.
| | - Fangyu Wang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210093, China.
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, 210093, China.
| |
Collapse
|
500
|
Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O'Connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, Dinan TG. The Microbiota-Gut-Brain Axis. Physiol Rev 2019; 99:1877-2013. [DOI: 10.1152/physrev.00018.2018] [Citation(s) in RCA: 1243] [Impact Index Per Article: 248.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kenneth J. O'Riordan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitlin S. M. Cowan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kiran V. Sandhu
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Thomaz F. S. Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Martin G. Codagnone
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Christine Fulling
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Anna V. Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Katherine E. Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Minal Jaggar
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitriona M. Long-Smith
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joshua M. Lyte
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Jason A. Martin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Alicia Molinero-Perez
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Moloney
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emanuela Morelli
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Enrique Morillas
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Rory O'Connor
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joana S. Cruz-Pereira
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Veronica L. Peterson
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Eoin Sherwin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Simon Spichak
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emily M. Teichman
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Shauna E. Wallace-Fitzsimons
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Niall Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Timothy G. Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|