1
|
Chilukuri A, Kim M, Mitra T, Gubatan JM, Urrete J, Saxon LD, Ablack A, Mikulski Z, Dobaczewska K, Shen Z, Keir M, Yi T, Kaur P, Oliveira P, Murillo-Saich J, Chang EY, Steiner CA, Jedlicka P, Guma M, Rivera-Nieves J. A Similar Mutation in the AAUU-Rich Elements of the Mouse TNF Gene Results in a Distinct Ileocolitic Phenotype: A New Strain of TNF-Overexpressing Mice. Inflamm Bowel Dis 2025; 31:1067-1081. [PMID: 39756463 PMCID: PMC11985683 DOI: 10.1093/ibd/izae307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Tumor necrosis factor (TNF) is a pleiotropic cytokine that plays a critical role in the pathogenesis of immune-mediated diseases including inflammatory bowel disease (IBD). The stability of its mRNA transcript, determined in part by destabilizing sequences in its AAUU repeats (ARE) gene region, is an important regulator of its tissue and systemic levels. A deletion in the ARE region of the gene resulted in IBD and arthritis in mice and pigs, supporting a critical role for the cytokine in human IBD and several human arthritides. A mutation in the same area of the mouse genome by Genentech scientists (T.Y., M.K.) resulted in a similar but not identical phenotype. METHODS Here, we compare histopathological, cellular, and molecular features of the strains and propose reasons for their distinct phenotypes. First, while homozygous TNFΔARE mice develop severe arthritis and die after weaning, homozygous Genentech TNFΔARE (ΔG/ΔG) mice have normal lifespans, and males are often fertile. RESULTS We found that while the ileitic phenotype had peaked at 12 weeks of age in all mice, colitis progressed mostly after 20 weeks of age in heterozygous mice. Their variably penetrant arthritic phenotype progressed mostly after 20 weeks, also in heterozygous mice from both strains. There was expansion of central memory T and B cells in lymphoid organs of TNF-overproducing strains and their transcriptional profile shared well-known pathogenetic pathways with human IBD. Finally, we found differences in the mutated sequences within the ARE regions of the TNF gene and in their microbiota composition and genetic background. These differences likely explain their phenotypic differences. CONCLUSIONS In summary, we describe a different strain of TNF-overproducing mice with an overlapping, yet not identical phenotype, which may have differential applications than the original strain.
Collapse
Affiliation(s)
- Amruth Chilukuri
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
| | - Margaret Kim
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| | - Taniya Mitra
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| | - John M Gubatan
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Josef Urrete
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
| | - Leo D Saxon
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| | - Amber Ablack
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
| | - Zbigniew Mikulski
- Microscopy and Histology Core, La Jolla Institute of Allergy and Immunology, La Jolla, CA, USA
| | - Katarzyna Dobaczewska
- Microscopy and Histology Core, La Jolla Institute of Allergy and Immunology, La Jolla, CA, USA
| | - Zining Shen
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
| | - Mary Keir
- Genentech Pharmaceuticals, South San Francisco, CA, USA
| | - Tangsheng Yi
- Genentech Pharmaceuticals, South San Francisco, CA, USA
| | - Prabhdeep Kaur
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| | - Patricia Oliveira
- Rheumatology Division, University of California San Diego, La Jolla, CA, USA
| | | | - Eric Y Chang
- Radiology Department, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| | - Calen A Steiner
- Division of Gastroenterology, University of Colorado, Denver, CO, USA
| | - Paul Jedlicka
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Mónica Guma
- Rheumatology Division, University of California San Diego, La Jolla, CA, USA
| | - Jesús Rivera-Nieves
- Division of Gastroenterology, University of California San Diego, La Jolla, CA, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
| |
Collapse
|
2
|
Bhutta NK, Xu X, Jian C, Wang Y, Liu Y, Sun J, Han B, Wu S, Javeed A. Gut microbiota mediated T cells regulation and autoimmune diseases. Front Microbiol 2024; 15:1477187. [PMID: 39749132 PMCID: PMC11694513 DOI: 10.3389/fmicb.2024.1477187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025] Open
Abstract
Gut microbiota regulates the immune system, the development and progression of autoimmune diseases (AIDs) and overall health. Recent studies have played a crucial part in understanding the specific role of different gut bacterial strains and their metabolites in different AIDs. Microbial signatures in AIDs are revealed by advanced sequencing and metabolomics studies. Microbes such as Faecalibacterium prausnitzii, Akkermansia muciniphila, Anaerostipes caccae, Bacteroides sp., Roseburia sp., Blautia sp., Blautia faecis, Clostridium lavalense, Christensenellaceae sp., Coprococcus sp., Firmicutes sp., Ruminococcaceae sp., Lachnospiraceae sp., Megamonas sp., Monoglobus sp., Streptococcus pneumoniae and Bifidobacterium sp. help maintain immune homeostasis; whereas, Prevotella copri, Ruminococcus gnavus, Lactobacillus salivarius, Enterococcus gallinarum, Elizabeth menigoseptica, Collinsella sp., Escherichia sp., Fusobacterium sp., Enterobacter ludwigii, Enterobacteriaceae sp., Proteobacteria, Porphyromonas gingivalis, Porphyromonas nigrescens, Dorea sp., and Clostridium sp. cause immuno-pathogenesis. A complex web of interactions is revealed by understanding the influence of gut microbiota on immune cells and various T cell subsets such as CD4+ T cells, CD8+ T cells, natural killer T cells, γδ T cells, etc. Certain AIDs, including rheumatoid arthritis, diabetes mellitus, atopic asthma, inflammatory bowel disease and non-alcoholic fatty liver disease exhibit a state of dysbiosis, characterized by alterations in microbial diversity and relative abundance of specific taxa. This review summarizes recent developments in understanding the role of certain microbiota composition in specific AIDs, and the factors affecting specific regulatory T cells through certain microbial metabolites and also focuses the potential application and therapeutic significance of gut microbiota-based interventions as novel adjunctive therapies for AIDs. Further research to determine the precise association of each gut bacterial strain in specific diseases is required.
Collapse
Affiliation(s)
- Nabeel Khalid Bhutta
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xiujin Xu
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Cuiqin Jian
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yifan Wang
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yi Liu
- Hangzhou Zheda Dixun Biological Gene Engineering Co., Ltd., Hangzhou, China
| | - Jinlyu Sun
- Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment of Allergic Diseases, Department of Allergy, National Clinical Research Center for Dermatologic and Immunologic Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Bingnan Han
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Shandong Wu
- Hangzhou Zheda Dixun Biological Gene Engineering Co., Ltd., Hangzhou, China
| | - Ansar Javeed
- Laboratory of Anti-allergic Functional Molecules, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
3
|
Heidari M, Maleki Vareki S, Yaghobi R, Karimi MH. Microbiota activation and regulation of adaptive immunity. Front Immunol 2024; 15:1429436. [PMID: 39445008 PMCID: PMC11496076 DOI: 10.3389/fimmu.2024.1429436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/30/2024] [Indexed: 10/25/2024] Open
Abstract
In the mucosa, T cells and B cells of the immune system are essential for maintaining immune homeostasis by suppressing reactions to harmless antigens and upholding the integrity of intestinal mucosal barrier functions. Host immunity and homeostasis are regulated by metabolites produced by the gut microbiota, which has developed through the long-term coevolution of the host and the gut biome. This is achieved by the immunological system's tolerance for symbiote microbiota, and its ability to generate a proinflammatory response against invasive organisms. The imbalance of the intestinal immune system with commensal organisms is causing a disturbance in the homeostasis of the gut microbiome. The lack of balance results in microbiota dysbiosis, the weakened integrity of the gut barrier, and the development of inflammatory immune reactions toward symbiotic organisms. Researchers may uncover potential therapeutic targets for preventing or regulating inflammatory diseases by understanding the interactions between adaptive immunity and the microbiota. This discussion will explore the connection between adaptive immunity and microbiota.
Collapse
Affiliation(s)
- Mozhdeh Heidari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saman Maleki Vareki
- Department of Oncology, Western University, London, ON, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada
| | - Ramin Yaghobi
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
4
|
Prasad S, Singh S, Menge S, Mohapatra I, Kim S, Helland L, Singh G, Singh A. Gut redox and microbiome: charting the roadmap to T-cell regulation. Front Immunol 2024; 15:1387903. [PMID: 39234241 PMCID: PMC11371728 DOI: 10.3389/fimmu.2024.1387903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/31/2024] [Indexed: 09/06/2024] Open
Abstract
The gastrointestinal (GI) tract redox environment, influenced by commensal microbiota and bacterial-derived metabolites, is crucial in shaping T-cell responses. Specifically, metabolites from gut microbiota (GM) exhibit robust anti-inflammatory effects, fostering the differentiation and regulation of CD8+ tissue-resident memory (TRM) cells, mucosal-associated invariant T (MAIT) cells, and stabilizing gut-resident Treg cells. Nitric oxide (NO), a pivotal redox mediator, emerges as a central regulator of T-cell functions and gut inflammation. NO impacts the composition of the gut microbiome, driving the differentiation of pro-inflammatory Th17 cells and exacerbating intestinal inflammation, and supports Treg expansion, showcasing its dual role in immune homeostasis. This review delves into the complex interplay between GI redox balance and GM metabolites, elucidating their profound impact on T-cell regulation. Additionally, it comprehensively emphasizes the critical role of GI redox, particularly reactive oxygen species (ROS) and NO, in shaping T-cell phenotype and functions. These insights offer valuable perspectives on disease mechanisms and potential therapeutic strategies for conditions associated with oxidative stress. Understanding the complex cross-talk between GI redox, GM metabolites, and T-cell responses provides valuable insights into potential therapeutic avenues for immune-mediated diseases, underscoring the significance of maintaining GI redox balance for optimal immune health.
Collapse
Affiliation(s)
- Sujata Prasad
- Translational Division, MLM Labs, LLC, Oakdale, MN, United States
| | - Shilpi Singh
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Samuel Menge
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
| | - Iteeshree Mohapatra
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Saint Paul, MN, United States
| | - Stefan Kim
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Logan Helland
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Gatikrushna Singh
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Amar Singh
- Department of Surgery, Schulze Diabetes Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
5
|
Hazime H, Ducasa GM, Santander AM, Brito N, González EE, Ban Y, Kaunitz J, Akiba Y, Fernández I, Burgueño JF, Abreu MT. Intestinal Epithelial Inactivity of Dual Oxidase 2 Results in Microbiome-Mediated Metabolic Syndrome. Cell Mol Gastroenterol Hepatol 2023; 16:557-572. [PMID: 37369278 PMCID: PMC10468370 DOI: 10.1016/j.jcmgh.2023.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/13/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND & AIMS Metabolic syndrome (MetS) is characterized by obesity, glucose intolerance, and hepatic steatosis. Alterations in the gut microbiome play important roles in the development of MetS. However, the mechanisms by which this occurs are poorly understood. Dual oxidase 2 (DUOX2) is an antimicrobial reduced nicotinamide adenine dinucleotide phosphate oxidase expressed in the gut epithelium. Here, we posit that epithelial DUOX2 activity provides a mechanistic link between the gut microbiome and the development of MetS. METHODS Mice carrying an intestinal epithelial-specific deletion of dual oxidase maturation factor 1/2 (DA IEC-KO), and wild-type littermates were fed a standard diet and killed at 24 weeks. Metabolic alterations were determined by glucose tolerance, lipid tests, and body and organ weight measurements. DUOX2 activity was determined by Amplex Red. Intestinal permeability was determined by fluorescein isothiocyanate-dextran, microbial translocation assessments, and portal vein lipopolysaccharide measurements. Metagenomic analysis of the stool microbiome was performed. The role of the microbiome was assessed in antibiotic-treated mice. RESULTS DA IEC-KO males showed increased body and organ weights accompanied by glucose intolerance and increased plasma lipid and liver enzyme levels, and increased adiposity in the liver and adipose tissue. Expression of F4/80, CD68, uncoupling protein 1, carbohydrate response element binding protein, leptin, and adiponectin was altered in the liver and adipose tissue of DA IEC-KO males. DA IEC-KO males produced less epithelial H2O2, had altered relative abundance of Akkermansiaceae and Lachnospiraceae in stool, and showed increased portal vein lipopolysaccharides and intestinal permeability. Females were protected from barrier defects and MetS, despite producing less H2O2. Antibiotic depletion abrogated all MetS phenotypes observed. CONCLUSIONS Intestinal epithelial inactivity of DUOX2 promotes MetS in a microbiome-dependent manner.
Collapse
Affiliation(s)
- Hajar Hazime
- Division of Gastroenterology, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida; Department of Microbiology and Immunology, University of Miami-Miller School of Medicine, Miami, Florida
| | - G Michelle Ducasa
- Division of Gastroenterology, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida
| | - Ana M Santander
- Division of Gastroenterology, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida
| | - Nivis Brito
- Division of Gastroenterology, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida
| | - Eddy E González
- Division of Gastroenterology, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida
| | - Yuguang Ban
- Biostatistics and Bioinformatics Shared Resource, Sylvester Comprehensive Cancer Center, University of Miami-Miller School of Medicine, Miami, Florida
| | - Jonathan Kaunitz
- Medical Service and Research Services, VA Greater Los Angeles Healthcare System, Los Angeles, California; Medical Service, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Yasutada Akiba
- Medical Service and Research Services, VA Greater Los Angeles Healthcare System, Los Angeles, California; Medical Service, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Irina Fernández
- Division of Gastroenterology, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida
| | - Juan F Burgueño
- Division of Gastroenterology, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida
| | - Maria T Abreu
- Division of Gastroenterology, Department of Medicine, University of Miami-Miller School of Medicine, Miami, Florida; Department of Microbiology and Immunology, University of Miami-Miller School of Medicine, Miami, Florida.
| |
Collapse
|
6
|
Deng L, Lee JWJ, Tan KSW. Infection with pathogenic Blastocystis ST7 is associated with decreased bacterial diversity and altered gut microbiome profiles in diarrheal patients. Parasit Vectors 2022; 15:312. [PMID: 36064620 PMCID: PMC9446694 DOI: 10.1186/s13071-022-05435-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
Background Blastocystis is a common protistan parasite inhabiting the gastrointestinal tract of humans and animals. While there are increasing reports characterizing the associations between Blastocystis and the gut microbiome in healthy individuals, only a few studies have investigated the relationships between Blastocystis and the gut microbiota in diarrheal patients. Methods The effects of a specific subtype (ST7) of Blastocystis on the composition of gut microbiota in diarrheal patients were investigated using 16S ribosomal RNA (rRNA) gene sequencing and bioinformatic analyses. Results Compared with diarrheal patients without Blastocystis, diarrheal patients infected with Blastocystis ST7 exhibited lower bacterial diversity. Beta diversity analysis revealed significant differences in bacterial community structure between ST7-infected and Blastocystis-free patients. The proportion of Enterobacteriaceae and Escherichia-Shigella were significantly enriched in ST7-infected patients. In contrast, the abundance of Bacteroides and Parabacteroides were more prevalent in Blastocystis-free patients. Conclusions The results of this study revealed, for the first time, that infection with Blastocystis ST7 is associated with lower bacterial diversity and altered microbial structure in diarrheal patients. Our study on clinical diarrheal patients is also the first to reinforce the notion that ST7 is a pathogenic subtype of Blastocystis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05435-z.
Collapse
Affiliation(s)
- Lei Deng
- Laboratory of Molecular and Cellular Parasitology, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore. .,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Jonathan W J Lee
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore.,Division of Gastroenterology & Hepatology, National University Hospital, Singapore, 119074, Singapore
| | - Kevin S W Tan
- Laboratory of Molecular and Cellular Parasitology, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore. .,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
7
|
Harnessing murine models of Crohn's disease ileitis to advance concepts of pathophysiology and treatment. Mucosal Immunol 2022; 15:10-26. [PMID: 34316007 DOI: 10.1038/s41385-021-00433-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/04/2023]
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are both characterized by chronic inflammation and severe dysfunction of the gastrointestinal tract. These two forms of inflammatory bowel disease (IBD) represent distinct clinical disorders with diverse driving mechanisms; however, this divergence is not reflected in currently approved therapeutics that commonly target general proinflammatory pathways. A compelling need therefore remains to understand factors that differentiate the topology and the distinct clinical manifestations of CD versus UC, in order to develop more effective and specialized therapies. Animal models provide valuable platforms for studying IBD heterogeneity and deciphering disease-specific mechanisms. Both the established and the newly developed ileitis mouse models are characterized by various disease initiating mechanisms and diverse phenotypic outcomes that reflect the complexity of human CD-ileitis. Microbial dysbiosis, destruction of epithelial barrier integrity, immune cell deregulation, as well as the recently described genome instability and stromal cell activation have all been proposed as the triggering factors for the development of ileitis-associated pathology. In this review, we aim to critically evaluate the mechanistic underpinnings of murine models of CD-ileitis, discuss their phenotypic similarities to human disease, and envisage their further exploitation for the development of novel targeted and personalized therapeutics.
Collapse
|
8
|
Raffner Basson A, Gomez-Nguyen A, LaSalla A, Buttó L, Kulpins D, Warner A, Di Martino L, Ponzani G, Osme A, Rodriguez-Palacios A, Cominelli F. Replacing Animal Protein with Soy-Pea Protein in an "American Diet" Controls Murine Crohn Disease-Like Ileitis Regardless of Firmicutes: Bacteroidetes Ratio. J Nutr 2021; 151:579-590. [PMID: 33484150 PMCID: PMC7948210 DOI: 10.1093/jn/nxaa386] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/26/2020] [Accepted: 11/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The current nutritional composition of the "American diet" (AD; also known as Western diet) has been linked to the increasing incidence of chronic diseases, including inflammatory bowel disease (IBD), namely Crohn disease (CD). OBJECTIVES This study investigated which of the 3 major macronutrients (protein, fat, carbohydrates) in the AD has the greatest impact on preventing chronic inflammation in experimental IBD mouse models. METHODS We compared 5 rodent diets designed to mirror the 2011-2012 "What We Eat in America" NHANES. Each diet had 1 macronutrient dietary source replaced. The formulated diets were AD, AD-soy-pea (animal protein replaced by soy + pea protein), AD-CHO ("refined carbohydrate" by polysaccharides), AD-fat [redistribution of the ω-6:ω-3 (n-6:n-3) PUFA ratio; ∼10:1 to 1:1], and AD-mix (all 3 "healthier" macronutrients combined). In 3 separate experiments, 8-wk-old germ-free SAMP1/YitFC mice (SAMP) colonized with human gut microbiota ("hGF-SAMP") from CD or healthy donors were fed an AD, an AD-"modified," or laboratory rodent diet for 24 wk. Two subsequent dextran sodium sulfate-colitis experiments in hGF-SAMP (12-wk-old) and specific-pathogen-free (SPF) C57BL/6 (20-wk-old) mice, and a 6-wk feeding trial in 24-wk-old SPF SAMP were performed. Intestinal inflammation, gut metagenomics, and MS profiles were assessed. RESULTS The AD-soy-pea diet resulted in lower histology scores [mean ± SD (56.1% ± 20.7% reduction)] in all feeding trials and IBD mouse models than did other diets (P < 0.05). Compared with the AD, the AD-soy-pea correlated with increased abundance in Lactobacillaceae and Leuconostraceae (1.5-4.7 log2 and 3.0-5.1 log2 difference, respectively), glutamine (6.5 ± 0.8 compared with 3.9 ± 0.3 ng/μg stool, P = 0.0005) and butyric acid (4:0; 3.3 ± 0.5 compared with 2.54 ± 0.4 ng/μg stool, P = 0.006) concentrations, and decreased linoleic acid (18:2n-6; 5.4 ± 0.4 compared with 8.6 ± 0.3 ng/μL plasma, P = 0.01). CONCLUSIONS Replacement of animal protein in an AD by plant-based sources reduced the severity of experimental IBD in all mouse models studied, suggesting that similar, feasible adjustments to the daily human diet could help control/prevent IBD in humans.
Collapse
Affiliation(s)
- Abigail Raffner Basson
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Adrian Gomez-Nguyen
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Alexandria LaSalla
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ludovica Buttó
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Danielle Kulpins
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Alexandra Warner
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Luca Di Martino
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Gina Ponzani
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Abdullah Osme
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Alexander Rodriguez-Palacios
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Fabio Cominelli
- Division of Gastroenterology & Liver Diseases, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Digestive Health Research Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|
9
|
Burgueño JF, Fritsch J, Gonzalez EE, Landau KS, Santander AM, Fernández I, Hazime H, Davies JM, Santaolalla R, Phillips MC, Diaz S, Dheer R, Brito N, Pignac-Kobinger J, Fernández E, Conner GE, Abreu MT. Epithelial TLR4 Signaling Activates DUOX2 to Induce Microbiota-Driven Tumorigenesis. Gastroenterology 2021; 160:797-808.e6. [PMID: 33127391 PMCID: PMC7879481 DOI: 10.1053/j.gastro.2020.10.031] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 09/28/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Chronic colonic inflammation leads to dysplasia and cancer in patients with inflammatory bowel disease. We have described the critical role of innate immune signaling via Toll-like receptor 4 (TLR4) in the pathogenesis of dysplasia and cancer. In the current study, we interrogate the intersection of TLR4 signaling, epithelial redox activity, and the microbiota in colitis-associated neoplasia. METHODS Inflammatory bowel disease and colorectal cancer data sets were analyzed for expression of TLR4, dual oxidase 2 (DUOX2), and NADPH oxidase 1 (NOX1). Epithelial production of hydrogen peroxide (H2O2) was analyzed in murine colonic epithelial cells and colonoid cultures. Colorectal cancer models were carried out in villin-TLR4 mice, carrying a constitutively active form of TLR4, their littermates, and villin-TLR4 mice backcrossed to DUOXA-knockout mice. The role of the TLR4-shaped microbiota in tumor development was tested in wild-type germ-free mice. RESULTS Activation of epithelial TLR4 was associated with up-regulation of DUOX2 and NOX1 in inflammatory bowel disease and colorectal cancer. DUOX2 was exquisitely dependent on TLR4 signaling and mediated the production of epithelial H2O2. Epithelial H2O2 was significantly increased in villin-TLR4 mice; TLR4-dependent tumorigenesis required the presence of DUOX2 and a microbiota. Mucosa-associated microbiota transferred from villin-TLR4 mice to wild-type germ-free mice caused increased H2O2 production and tumorigenesis. CONCLUSIONS Increased TLR4 signaling in colitis drives expression of DUOX2 and epithelial production of H2O2. The local milieu imprints the mucosal microbiota and imbues it with pathogenic properties demonstrated by enhanced epithelial reactive oxygen species and increased development of colitis-associated tumors. The inter-relationship between epithelial reactive oxygen species and tumor-promoting microbiota requires a 2-pronged strategy to reduce the risk of dysplasia in colitis patients.
Collapse
Affiliation(s)
- Juan F Burgueño
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Julia Fritsch
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA,Department of Microbiology and Immunology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Eddy E Gonzalez
- Biotechnology and Biopharmaceuticals Laboratory, Department of Pathophysiology, School of Biological Science, Universidad de Concepción, Concepción, Chile
| | - Kevin S Landau
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Ana M Santander
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Irina Fernández
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Hajar Hazime
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Julie M Davies
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Rebeca Santaolalla
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Matthew C Phillips
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Sophia Diaz
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Rishu Dheer
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Nivis Brito
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Judith Pignac-Kobinger
- Department of Medicine, Division of Gastroenterology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Ester Fernández
- Animal Physiology Unit, Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gregory E Conner
- Department of Cell Biology, University of Miami – Miller School of Medicine, Miami, FL, USA
| | - Maria T Abreu
- Department of Medicine, Division of Gastroenterology, University of Miami-Miller School of Medicine, Miami, Florida; Department of Microbiology and Immunology, University of Miami-Miller School of Medicine, Miami, Florida.
| |
Collapse
|
10
|
Basson AR, Gomez-Nguyen A, Menghini P, Buttó LF, Di Martino L, Aladyshkina N, Osme A, LaSalla A, Fischer D, Ezeji JC, Erkkila HL, Brennan CJ, Lam M, Rodriguez-Palacios A, Cominelli F. Human Gut Microbiome Transplantation in Ileitis Prone Mice: A Tool for the Functional Characterization of the Microbiota in Inflammatory Bowel Disease Patients. Inflamm Bowel Dis 2020; 26:347-359. [PMID: 31750921 PMCID: PMC7012301 DOI: 10.1093/ibd/izz242] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a lifelong digestive disease characterized by periods of severe inflammation and remission. To our knowledge, this is the first study showing a variable effect on ileitis severity from human gut microbiota isolated from IBD donors in remission and that of healthy controls in a mouse model of IBD. METHODS We conducted a series of single-donor intensive and nonintensive fecal microbiota transplantation (FMT) experiments using feces from IBD patients in remission and healthy non-IBD controls (N = 9 donors) in a mouse model of Crohn's disease (CD)-like ileitis that develops ileitis in germ-free (GF) conditions (SAMP1/YitFC; N = 96 mice). RESULTS Engraftment studies demonstrated that the microbiome of IBD in remission could have variable effects on the ileum of CD-prone mice (pro-inflammatory, nonmodulatory, or anti-inflammatory), depending on the human donor. Fecal microbiota transplantation achieved a 95% ± 0.03 genus-level engraftment of human gut taxa in mice, as confirmed at the operational taxonomic unit level. In most donors, microbiome colonization abundance patterns remained consistent over 60 days. Microbiome-based metabolic predictions of GF mice with Crohn's or ileitic-mouse donor microbiota indicate that chronic amino/fatty acid (valine, leucine, isoleucine, histidine; linoleic; P < 1e-15) alterations (and not bacterial virulence markers; P > 0.37) precede severe ileitis in mice, supporting their potential use as predictors/biomarkers in human CD. CONCLUSION The gut microbiome of IBD remission patients is not necessarily innocuous. Characterizing the inflammatory potential of each microbiota in IBD patients using mice may help identify the patients' best anti-inflammatory fecal sample for future use as an anti-inflammatory microbial autograft during disease flare-ups.
Collapse
Affiliation(s)
- Abigail R Basson
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Adrian Gomez-Nguyen
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Paola Menghini
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Ludovica F Buttó
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Luca Di Martino
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Natalia Aladyshkina
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Abdullah Osme
- Department of Pathology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alexandria LaSalla
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Derek Fischer
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Jessica C Ezeji
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Hailey L Erkkila
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Connery J Brennan
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Minh Lam
- Division of Gastrointestinal and Liver Disease, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Alexander Rodriguez-Palacios
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA,Division of Gastrointestinal and Liver Disease, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA,Division of Gastrointestinal and Liver Disease, University Hospitals Cleveland Medical Center, Cleveland, OH, USA,Address correspondence to: Fabio Cominelli, Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA; Division of Gastrointestinal and Liver Disease, University Hospitals Cleveland Medical Center, 11100 Euclid Avenue, Cleveland OH 44106-5066, USA.
| |
Collapse
|
11
|
Rogala AR, Oka A, Sartor RB. Strategies to Dissect Host-Microbial Immune Interactions That Determine Mucosal Homeostasis vs. Intestinal Inflammation in Gnotobiotic Mice. Front Immunol 2020; 11:214. [PMID: 32133003 PMCID: PMC7040030 DOI: 10.3389/fimmu.2020.00214] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 01/27/2020] [Indexed: 12/16/2022] Open
Abstract
When identifying the key immunologic-microbial interactions leading to either mucosal homeostasis in normal hosts or intestinal inflammatory responses in genetically susceptible individuals, it is important to not only identify microbial community correlations but to also define the functional pathways involved. Gnotobiotic rodents are a very effective tool for this purpose as they provide a highly controlled environment in which to identify the function of complex intestinal microbiota, their individual components, and metabolic products. Herein we review specific strategies using gnotobiotic mice to functionally evaluate the role of various intestinal microbiota in host responses. These studies include basic comparisons between host responses in germ-free (GF), specific-pathogen-free or conventionally raised wild-type mice or those with underlying genetic susceptibilities to intestinal inflammation. We also discuss what can be learned from studies in which GF mice are colonized with single wild-type or genetically-modified microbial isolates to examine the functions of individual bacteria and their targeted bacterial genes, or colonized by multiple defined isolates to determine interactions between members of defined consortia. Additionally, we discuss studies to identify functions of complex microbial communities from healthy or diseased human or murine hosts via fecal transplant into GF mice. Finally, we conclude by suggesting ways to improve studies of immune-microbial interactions using gnotobiotic mice.
Collapse
Affiliation(s)
- Allison R. Rogala
- Division of Comparative Medicine, Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Akihiko Oka
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - R. Balfour Sartor
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Departments of Medicine, Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
12
|
Menghini P, Corridoni D, Buttó LF, Osme A, Shivaswamy S, Lam M, Bamias G, Pizarro TT, Rodriguez-Palacios A, Dinarello CA, Cominelli F. Neutralization of IL-1α ameliorates Crohn's disease-like ileitis by functional alterations of the gut microbiome. Proc Natl Acad Sci U S A 2019; 116:26717-26726. [PMID: 31843928 PMCID: PMC6936591 DOI: 10.1073/pnas.1915043116] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Crohn's disease and ulcerative colitis are chronic and progressive inflammatory bowel diseases (IBDs) that are attributed to dysregulated interactions between the gut microbiome and the intestinal mucosa-associated immune system. There are limited studies investigating the role of either IL-1α or IL-1β in mouse models of colitis, and no clinical trials blocking either IL-1 have yet to be performed. In the present study, we show that neutralization of IL-1α by a specific monoclonal antibody against murine IL-1α was highly effective in reducing inflammation and damage in SAMP mice, mice that spontaneously develop a Crohn's-like ileitis. Anti-mouse IL-1α significantly ameliorated the established, chronic ileitis and also protected mice from developing acute DSS-induced colitis. Both were associated with taxonomic divergence of the fecal gut microbiome, which was treatment-specific and not dependent on inflammation. Anti-IL-1α administration led to a decreased ratio of Proteobacteria to Bacteroidetes, decreased presence of Helicobacter species, and elevated representation of Mucispirillum schaedleri and Lactobacillus salivarius. Such modification in flora was functionally linked to the antiinflammatory effects of IL-1α neutralization, as blockade of IL-1α was not effective in germfree SAMP mice. Furthermore, preemptive dexamethasone treatment of DSS-challenged SAMP mice led to changes in flora composition without preventing the development of colitis. Thus, neutralization of IL-1α changes specific bacterial species of the intestinal microbiome, which is linked to its antiinflammatory effects. These functional findings may be of significant value for patients with IBD, who may benefit from targeted IL-1α-based therapies.
Collapse
Affiliation(s)
- Paola Menghini
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Daniele Corridoni
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, United Kingdom
| | - Ludovica F. Buttó
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Abdullah Osme
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | | | - Minh Lam
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Giorgos Bamias
- Gastrointestinal Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, 11527 Athens, Greece
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Alexander Rodriguez-Palacios
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | | | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| |
Collapse
|
13
|
Lo Y, Sauve JP, Menzies SC, Steiner TS, Sly LM. Phosphatidylinositol 3-kinase p110δ drives intestinal fibrosis in SHIP deficiency. Mucosal Immunol 2019; 12:1187-1200. [PMID: 31358861 DOI: 10.1038/s41385-019-0191-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 06/23/2019] [Accepted: 07/15/2019] [Indexed: 02/04/2023]
Abstract
Crohn's disease is an immune-mediated disease characterized by inflammation along the gastrointestinal tract. Fibrosis requiring surgery occurs in one-third of people with Crohn's disease but there are no treatments for intestinal fibrosis. Mice deficient in the SH2 domain-containing inositolpolyphosphate 5'-phosphatase (SHIP), a negative regulator of phosphatidylinositol 3-kinase (PI3K) develop spontaneous Crohn's disease-like intestinal inflammation and arginase I (argI)-dependent fibrosis. ArgI is up-regulated in SHIP deficiency by PI3Kp110δ activity. Thus, we hypothesized that SHIP-deficient mice develop fibrosis due to increased PI3Kp110δ activity. In SHIP-deficient mice, genetic ablation or pharmacological inhibition of PI3Kp110δ activity reduced intestinal fibrosis, including muscle thickening, accumulation of vimentin+ mesenchymal cells, and collagen deposition. PI3Kp110δ deficiency or inhibition also reduced ileal inflammation in SHIP-deficient mice suggesting that PI3Kp110δ may contribute to inflammation. Targeting PI3Kp110δ activity may be an effective strategy to reduce intestinal fibrosis, and may be particularly effective in the subset of people with Crohn's disease, who have low SHIP activity.
Collapse
Affiliation(s)
- Young Lo
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Jean Philippe Sauve
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Susan C Menzies
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Theodore S Steiner
- Division of Infectious Diseases, Department of Medicine, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Laura M Sly
- Division of Gastroenterology, Department of Pediatrics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
14
|
Mueller KD, Zhang H, Serrano CR, Billmyre RB, Huh EY, Wiemann P, Keller NP, Wang Y, Heitman J, Lee SC. Gastrointestinal microbiota alteration induced by Mucor circinelloides in a murine model. J Microbiol 2019; 57:509-520. [PMID: 31012059 PMCID: PMC6737537 DOI: 10.1007/s12275-019-8682-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/10/2019] [Accepted: 01/15/2019] [Indexed: 12/12/2022]
Abstract
Mucor circinelloides is a pathogenic fungus and etiologic agent of mucormycosis. In 2013, cases of gastrointestinal illness after yogurt consumption were reported to the US FDA, and the producer found that its products were contaminated with Mucor. A previous study found that the Mucor strain isolated from an open contaminated yogurt exhibited virulence in a murine systemic infection model and showed that this strain is capable of surviving passage through the gastrointestinal tract of mice. In this study, we isolated another Mucor strain from an unopened yogurt that is closely related but distinct from the first Mucor strain and subsequently examined if Mucor alters the gut microbiota in a murine host model. DNA extracted from a ten-day course of stool samples was used to analyze the microbiota in the gastrointestinal tracts of mice exposed via ingestion of Mucor spores. The bacterial 16S rRNA gene and fungal ITS1 sequences obtained were used to identify taxa of each kingdom. Linear regressions revealed that there are changes in bacterial and fungal abundance in the gastrointestinal tracts of mice which ingested Mucor. Furthermore, we found an increased abundance of the bacterial genus Bacteroides and a decreased abundance of the bacteria Akkermansia muciniphila in the gastrointestinal tracts of exposed mice. Measurements of abundances show shifts in relative levels of multiple bacterial and fungal taxa between mouse groups. These findings suggest that exposure of the gastrointestinal tract to Mucor can alter the microbiota and, more importantly, illustrate an interaction between the intestinal mycobiota and bacteriota. In addition, Mucor was able to induce increased permeability in epithelial cell monolayers in vitro, which might be indicative of unstable intestinal barriers. Understanding how the gut microbiota is shaped is important to understand the basis of potential methods of treatment for gastrointestinal illness. How the gut microbiota changes in response to exposure, even by pathogens not considered to be causative agents of food-borne illness, may be important to how commercial food producers prevent and respond to contamination of products aimed at the public. This study provides evidence that the fungal microbiota, though understudied, may play an important role in diseases of the human gut.
Collapse
Affiliation(s)
- Katherine D Mueller
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Hao Zhang
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Christian R Serrano
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, TX, USA
| | - R Blake Billmyre
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Eun Young Huh
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Philipp Wiemann
- Department of Medical Microbiology and Immunology, University of Wisconsin at Madison, Madison, WI, USA
| | - Nancy P Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin at Madison, Madison, WI, USA
| | - Yufeng Wang
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Soo Chan Lee
- South Texas Center for Emerging Infectious Diseases (STCEID), Department of Biology, The University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
15
|
Burgueño JF, Lang JK, Santander AM, Fernández I, Fernández E, Zaias J, Abreu MT. Fluid supplementation accelerates epithelial repair during chemical colitis. PLoS One 2019; 14:e0215387. [PMID: 31002683 PMCID: PMC6474653 DOI: 10.1371/journal.pone.0215387] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/01/2019] [Indexed: 12/28/2022] Open
Abstract
The dextran sulfate sodium (DSS) model of colitis is a common animal model of inflammatory bowel disease that causes pain and distress. In this study, we aimed to determine whether fluid supplementation can be used as a welfare-based intervention to minimize animal suffering. C57Bl/6 females undergoing acute colitis by administration of 3% DSS in drinking water were supplemented with 1 mL intraperitoneal injections of NaCl and compared to non-supplemented control mice. Mouse behavior and locomotive activity were assessed on days 5–6 after DSS initiation by means of tail suspension, novel object recognition and open field activity tests. Mice were euthanized after either the acute (day 7) or the recovery phase (day 12) of colitis and inflammation, epithelial proliferation, and differentiation were assessed by means of histology, immunohistochemistry, quantitative PCR, and western blot. We found that fluid-supplemented mice had reduced signs of colitis with no alterations in behavior or locomotive activity. Furthermore, we observed an accelerated epithelial repair response after fluid hydration during the acute phase of colitis, characterized by increased crypt proliferation, activation of ERK1/2, and modulation of TGF-β1 expression. Consistent with these findings, fluid-supplemented mice had increased numbers of goblet cells, upregulated expression of differentiation markers for absorptive enterocytes, and reduced inflammation during the recovery phase. Our results show that fluid hydration does not reduce stress in DSS-treated mice but alters colitis evolution by reducing clinical signs and accelerating epithelial repair. These results argue against the routine use of fluid supplementation in DSS-treated mice.
Collapse
Affiliation(s)
- Juan F. Burgueño
- Division of Gastroenterology, Department of Medicine, University of Miami–Leonard Miller School of Medicine, Miami, FL, United States of America
- * E-mail:
| | - Jessica K. Lang
- Division of Gastroenterology, Department of Medicine, University of Miami–Leonard Miller School of Medicine, Miami, FL, United States of America
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, United States of America
| | - Ana M. Santander
- Division of Gastroenterology, Department of Medicine, University of Miami–Leonard Miller School of Medicine, Miami, FL, United States of America
| | - Irina Fernández
- Division of Gastroenterology, Department of Medicine, University of Miami–Leonard Miller School of Medicine, Miami, FL, United States of America
| | - Ester Fernández
- Animal Physiology Unit, Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Julia Zaias
- Division of Veterinary Resources, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Pathology and Laboratory Medicine, University of Miami–Leonard Miller School of Medicine, Miami, FL, United States of America
| | - Maria T. Abreu
- Division of Gastroenterology, Department of Medicine, University of Miami–Leonard Miller School of Medicine, Miami, FL, United States of America
| |
Collapse
|
16
|
Schmitz JM, Tonkonogy SL, Dogan B, Leblond A, Whitehead KJ, Kim SC, Simpson KW, Sartor RB. Murine Adherent and Invasive E. coli Induces Chronic Inflammation and Immune Responses in the Small and Large Intestines of Monoassociated IL-10-/- Mice Independent of Long Polar Fimbriae Adhesin A. Inflamm Bowel Dis 2019; 25:875-885. [PMID: 30576451 PMCID: PMC6458545 DOI: 10.1093/ibd/izy386] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Adherent and invasive Escherichia coli (AIEC) is preferentially associated with ileal Crohn's disease (CD). The role of AIEC in the development of inflammation and its regional tropism is unresolved. The presence of long polar fimbriae (LPF) in 71% of ileal CD AIEC suggests a role for LPF in the tropism and virulence of AIEC. The aim of our study is to determine if AIEC, with or without LpfA, induces intestinal inflammation in monoassociated IL-10-/- mice. METHODS We compared murine AIEC strains NC101 (phylogroup B2, LpfA-) and CUMT8 (phylogroup B1, LpfA+), and isogenic mutant CUMT8 lacking lpfA154, with a non-AIEC (E. coli K12), evaluating histologic inflammation, bacterial colonization, mucosal adherence and invasion, and immune activation. RESULTS IL-10-/- mice monoassociated with AIEC (either CUMT8, CUMT8:ΔlpfA, or NC101) but not K12 developed diffuse small intestinal and colonic inflammation. There was no difference in the magnitude and distribution of inflammation in mice colonized with CUMT8:ΔlpfA compared with wild-type CUMT8. Bacterial colonization was similar for all E. coli strains. Fluorescence in situ hybridization revealed mucosal adherence and tissue invasion by AIEC but not K12. Production of the cytokines IL-12/23 p40 by the intestinal tissue and IFN-γ and IL-17 by CD4 T cells correlated with inflammation. CONCLUSIONS IL-10-/- mice monoassociated with murine AIEC irrespective of LpfA expression developed chronic inflammation accompanied by IL-12/23 p40 production in the small and large intestines and IFN-γ/IL-17 production by CD4 T cells that model the interplay between enteric pathosymbionts, host susceptibility, and enhanced immune responses in people with IBD.
Collapse
MESH Headings
- Animals
- Bacterial Adhesion
- Escherichia coli/immunology
- Escherichia coli Infections/immunology
- Escherichia coli Infections/metabolism
- Escherichia coli Infections/microbiology
- Escherichia coli Infections/pathology
- Escherichia coli Proteins/metabolism
- Fimbriae Proteins/metabolism
- Fimbriae, Bacterial/immunology
- Fimbriae, Bacterial/pathology
- Inflammation/etiology
- Inflammation/metabolism
- Inflammation/pathology
- Interleukin-10/physiology
- Intestine, Large/immunology
- Intestine, Large/metabolism
- Intestine, Large/microbiology
- Intestine, Large/pathology
- Intestine, Small/immunology
- Intestine, Small/metabolism
- Intestine, Small/microbiology
- Intestine, Small/pathology
- Mice
- Mice, Knockout
Collapse
Affiliation(s)
- Julia M Schmitz
- Center for Gastrointestinal Biology and Disease, University of North Carolina and North Carolina State University, Chapel Hill and Raleigh, North Carolina
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Susan L Tonkonogy
- Center for Gastrointestinal Biology and Disease, University of North Carolina and North Carolina State University, Chapel Hill and Raleigh, North Carolina
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Belgin Dogan
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Anna Leblond
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Kristi J Whitehead
- Center for Gastrointestinal Biology and Disease, University of North Carolina and North Carolina State University, Chapel Hill and Raleigh, North Carolina
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sandra C Kim
- Center for Gastrointestinal Biology and Disease, University of North Carolina and North Carolina State University, Chapel Hill and Raleigh, North Carolina
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kenneth W Simpson
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - R Balfour Sartor
- Center for Gastrointestinal Biology and Disease, University of North Carolina and North Carolina State University, Chapel Hill and Raleigh, North Carolina
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
17
|
Buttó LF, Jia LG, Arseneau KO, Tamagawa H, Rodriguez-Palacios A, Li Z, De Salvo C, Pizarro TT, Bamias G, Cominelli F. Death-Domain-Receptor 3 Deletion Normalizes Inflammatory Gene Expression and Prevents Ileitis in Experimental Crohn's Disease. Inflamm Bowel Dis 2019; 25:14-26. [PMID: 30295722 PMCID: PMC6290789 DOI: 10.1093/ibd/izy305] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND TNF-like cytokine 1A (TL1A) and its functional receptor, death-domain-receptor-3 (DR3), are multifunctional mediators of effector and regulatory immunity. We aimed to evaluate the functional role and therapeutic potential of TL1A/DR3 signaling in Crohn's disease-like ileitis. METHODS Ileitis-prone SAMP1/YitFc (SAMP) and TNFΔARE/+ mice were rendered deficient for DR3 or TL1A by microsatellite marker-assisted backcrossing. Pathological and immunological characteristics were compared between control and knockout mice, and mucosal immunophenotype was analyzed by Nanostring microarray assay. The therapeutic effect of pharmacological TL1A neutralization was also investigated. RESULTS DR3 deficiency was associated with restoration of a homeostatic mucosal immunostat in SAMP mice through the regulation of several pro- and anti-inflammatory genes. This led to suppression of effector immunity, amelioration of ileitis severity, and compromised ability of either unfractionated CD4+ or CD4+CD45RBhi mucosal lymphocytes to transfer ileitis to severe combined immunodeficient mice recipients. TNF-driven ileitis was also prevented in TNFΔARE/+xDR3-/- mice, in association with decreased expression of the pro-inflammatory cytokines TNF and IFN-γ. In contrast to DR3, TL1A was dispensable for the development of ileitis although it affected the kinetics of inflammation, as TNFΔARE/+xTL1A-/- demonstrated delayed onset of inflammation, whereas administration of a neutralizing, anti-TL1A antibody ameliorated early but not late TNFΔARE/+ ileitis. CONCLUSION We found a prominent pro-inflammatory role of DR3 in chronic ileitis, which is only partially mediated via interaction with TL1A, raising the possibility for additional DR3 ligands. Death-domain-receptor-3 appears to be a master regulator of mucosal homeostasis and inflammation and may represent a candidate therapeutic target for chronic inflammatory conditions of the bowel.
Collapse
Affiliation(s)
- Ludovica F Buttó
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Li-Guo Jia
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Kristen O Arseneau
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | - Alex Rodriguez-Palacios
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Zhaodong Li
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Carlo De Salvo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Giorgos Bamias
- GI-Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
18
|
Rios-Arce ND, Collins FL, Schepper JD, Steury MD, Raehtz S, Mallin H, Schoenherr DT, Parameswaran N, McCabe LR. Epithelial Barrier Function in Gut-Bone Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1033:151-183. [PMID: 29101655 DOI: 10.1007/978-3-319-66653-2_8] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The intestinal epithelial barrier plays an essential role in maintaining host homeostasis. The barrier regulates nutrient absorption as well as prevents the invasion of pathogenic bacteria in the host. It is composed of epithelial cells, tight junctions, and a mucus layer. Several factors, such as cytokines, diet, and diseases, can affect this barrier. These factors have been shown to increase intestinal permeability, inflammation, and translocation of pathogenic bacteria. In addition, dysregulation of the epithelial barrier can result in inflammatory diseases such as inflammatory bowel disease. Our lab and others have also shown that barrier disruption can have systemic effects including bone loss. In this chapter, we will discuss the current literature to understand the link between intestinal barrier and bone. We will discuss how inflammation, aging, dysbiosis, and metabolic diseases can affect intestinal barrier-bone link. In addition, we will highlight the current suggested mechanism between intestinal barrier and bone.
Collapse
Affiliation(s)
- Naiomy Deliz Rios-Arce
- Comparative Medicine and Integrative Biology Program, East Lansing, MI, USA.,Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Fraser L Collins
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | | | - Michael D Steury
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Sandi Raehtz
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Heather Mallin
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Danny T Schoenherr
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Narayanan Parameswaran
- Comparative Medicine and Integrative Biology Program, East Lansing, MI, USA. .,Department of Physiology, Michigan State University, East Lansing, MI, USA.
| | - Laura R McCabe
- Department of Physiology and Department of Radiology, Biomedical Imaging Research Centre, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
19
|
Barko P, McMichael M, Swanson K, Williams D. The Gastrointestinal Microbiome: A Review. J Vet Intern Med 2018; 32:9-25. [PMID: 29171095 PMCID: PMC5787212 DOI: 10.1111/jvim.14875] [Citation(s) in RCA: 408] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 08/30/2017] [Accepted: 10/12/2017] [Indexed: 12/14/2022] Open
Abstract
The gastrointestinal microbiome is a diverse consortium of bacteria, archaea, fungi, protozoa, and viruses that inhabit the gut of all mammals. Studies in humans and other mammals have implicated the microbiome in a range of physiologic processes that are vital to host health including energy homeostasis, metabolism, gut epithelial health, immunologic activity, and neurobehavioral development. The microbial genome confers metabolic capabilities exceeding those of the host organism alone, making the gut microbiome an active participant in host physiology. Recent advances in DNA sequencing technology and computational biology have revolutionized the field of microbiomics, permitting mechanistic evaluation of the relationships between an animal and its microbial symbionts. Changes in the gastrointestinal microbiome are associated with diseases in humans and animals including inflammatory bowel disease, asthma, obesity, metabolic syndrome, cardiovascular disease, immune-mediated conditions, and neurodevelopmental conditions such as autism spectrum disorder. While there remains a paucity of data regarding the intestinal microbiome in small animals, recent studies have helped to characterize its role in host animal health and associated disease states. This review is intended to familiarize small animal veterinarians with recent advances in the field of microbiomics and to prime them for a future in which diagnostic tests and therapies will incorporate these developments into clinical practice.
Collapse
Affiliation(s)
- P.C. Barko
- Veterinary Clinical MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL
| | - M.A. McMichael
- Veterinary Clinical MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL
| | - K.S. Swanson
- Veterinary Clinical MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL
- Department of Animal SciencesUniversity of Illinois at Urbana‐ChampaignUrbanaIL
| | - D.A. Williams
- Veterinary Clinical MedicineUniversity of Illinois at Urbana‐ChampaignUrbanaIL
| |
Collapse
|
20
|
Rogler G. Challenges of Translation of Anti-Fibrotic Therapies into Clinical Practice in IBD. FIBROSTENOTIC INFLAMMATORY BOWEL DISEASE 2018:295-305. [DOI: 10.1007/978-3-319-90578-5_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
21
|
Corridoni D, Rodriguez-Palacios A, Di Stefano G, Di Martino L, Antonopoulos DA, Chang EB, Arseneau KO, Pizarro TT, Cominelli F. Genetic deletion of the bacterial sensor NOD2 improves murine Crohn's disease-like ileitis independent of functional dysbiosis. Mucosal Immunol 2017; 10:971-982. [PMID: 27848951 PMCID: PMC5433921 DOI: 10.1038/mi.2016.98] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 09/23/2016] [Indexed: 02/07/2023]
Abstract
Although genetic polymorphisms in NOD2 (nucleotide-binding oligomerization domain-containing 2) have been associated with the pathogenesis of Crohn's disease (CD), little is known regarding the role of wild-type (WT) NOD2 in the gut. To date, most murine studies addressing the role of WT Nod2 have been conducted using healthy (ileitis/colitis-free) mouse strains. Here, we evaluated the effects of Nod2 deletion in a murine model of spontaneous ileitis, i.e., the SAMP1Yit/Fc (SAMP) strain, which closely resembles CD. Remarkably, Nod2 deletion improved both chronic cobblestone ileitis (by 50% assessed, as the % of abnormal mucosa at 24 wks of age), as well as acute dextran sodium sulfate (DSS) colitis. Mechanistically, Th2 cytokine production and Th2-transcription factor activation (i.e., STAT6 phosphorylation) were reduced. Microbiologically, the effects of Nod2 deletion appeared independent of fecal microbiota composition and function, assessed by 16S rRNA and metatranscriptomics. Our findings indicate that pharmacological blockade of NOD2 signaling in humans could improve health in Th2-driven chronic intestinal inflammation.
Collapse
Affiliation(s)
- D Corridoni
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Case Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, USA
| | - A Rodriguez-Palacios
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Case Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, USA
| | - G Di Stefano
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Case Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, USA
| | - L Di Martino
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Case Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, USA
| | - D A Antonopoulos
- Biosciences Division, Argonne National Laboratory, Argonne, Illinois, USA
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - E B Chang
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - K O Arseneau
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Case Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, USA
| | - T T Pizarro
- Case Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - F Cominelli
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Case Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
22
|
Cominelli F, Arseneau KO, Rodriguez-Palacios A, Pizarro TT. Uncovering Pathogenic Mechanisms of Inflammatory Bowel Disease Using Mouse Models of Crohn's Disease-Like Ileitis: What is the Right Model? Cell Mol Gastroenterol Hepatol 2017; 4:19-32. [PMID: 28560286 PMCID: PMC5439236 DOI: 10.1016/j.jcmgh.2017.02.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 02/15/2017] [Indexed: 02/06/2023]
Abstract
Crohn's disease and ulcerative colitis, together known as inflammatory bowel disease, are debilitating chronic disorders of unknown cause and cure. Our evolving understanding of these pathologies is enhanced greatly by the use of animal models of intestinal inflammation that allow in vivo mechanistic studies, preclinical evaluation of new therapies, and investigation into the causative factors that underlie disease pathogenesis. Several animal models, most commonly generated in mice, exist for the study of colitis. The appropriateness of their use often can be determined by their mode of generation (ie, chemical induction, T-cell transfer, targeted genetic manipulation, spontaneously occurring, and so forth), the type of investigation (mechanistic studies, pathogenic experiments, preclinical evaluations, and so forth), and the type of inflammation that occurs in the model (acute vs chronic colitis, tissue injury/repair, and so forth). Although most murine models of inflammatory bowel disease develop inflammation in the colon, Crohn's disease most commonly occurs in the terminal ileum, where a very limited number of mouse models manifest disease. This review discusses appropriate experimental applications for different mouse models of colitis, and highlights the particular utility of 2 highly relevant models of Crohn's-like ileitis-the spontaneous SAMP1/YitFc inbred mouse strain and the genetically engineered TnfΔAU-rich element/+ mouse model of tumor necrosis factor overexpression, both of which bear strong resemblance to the human condition. Similar to patients with Crohn's disease, SAMP1/YitFc ileitis develops spontaneously, without chemical, genetic, or immunologic manipulation, making this model particularly relevant for studies aimed at identifying the primary defect underlying the occurrence of Crohn's ileitis, as well as preclinical testing of novel treatment modalities.
Collapse
Affiliation(s)
- Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio,Correspondence Address correspondence to: Fabio Cominelli, MD, PhD, Division of Gastroenterology, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Cleveland, Ohio 44106-5066. fax: (216) 844-7371.Division of GastroenterologyCase Western Reserve University School of Medicine11100 Euclid AvenueClevelandOhio 44106-5066
| | - Kristen O. Arseneau
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Alexander Rodriguez-Palacios
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Theresa T. Pizarro
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
23
|
Epithelial-specific Toll-like Receptor (TLR)5 Activation Mediates Barrier Dysfunction in Experimental Ileitis. Inflamm Bowel Dis 2017; 23:392-403. [PMID: 28146004 DOI: 10.1097/mib.0000000000001035] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND A large body of evidence supports a central role of TLR5 and its natural ligand, flagellin, in Crohn's disease (CD), with the precise mechanism(s) still unresolved. METHODS We investigated the role of flagellin/TLR5 in SAMP1/YitFc (SAMP) mice, a spontaneous model of Crohn's disease-like ileitis. RESULTS Ileal Tlr5 and serum antiflagellin IgG antibodies were increased in SAMP before the onset of inflammation and during established disease; these trends were abrogated in the absence of colonizing commensal bacteria. Irradiated SAMP receiving either wild-type (AKR) or SAMP bone marrow (BM) developed severe ileitis and displayed increased ileal Tlr5 compared with AKR recipients of either SAMP or AKR bone marrow, neither of which conferred ileitis, suggesting that elevated TLR5 in native SAMP is derived primarily from a nonhematopoietic (e.g., epithelial) source. Indeed, ileal epithelial TLR5 in preinflamed SAMP was increased compared with age-matched AKR and germ-free SAMP. TLR5-specific ex vivo activation of SAMP ileal tissues decreased epithelial barrier resistance, indicative of increased permeability, and was accompanied by altered expression of the tight junction proteins, claudin-3, occludin, and zonula occludens-1. CONCLUSIONS Our results provide evidence that aberrant, elevated TLR5 expression is present in the ileal epithelium of SAMP mice, is augmented in the presence of the gut microbiome, and that TLR5 activation in response to bacterial flagellin results in a deficiency to maintain appropriate epithelial barrier integrity. Together, these findings represent a potential mechanistic pathway leading to the exacerbation and perpetuation of chronic gut inflammation in experimental ileitis and possibly, in patients with Crohn's disease.
Collapse
|
24
|
Do JS, Kim S, Keslar K, Jang E, Huang E, Fairchild RL, Pizarro TT, Min B. γδ T Cells Coexpressing Gut Homing α4β7 and αE Integrins Define a Novel Subset Promoting Intestinal Inflammation. THE JOURNAL OF IMMUNOLOGY 2016; 198:908-915. [PMID: 27927968 DOI: 10.4049/jimmunol.1601060] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 11/10/2016] [Indexed: 02/07/2023]
Abstract
γδ T lymphocytes, dominant T cell subsets in the intestine, mediate both regulatory and pathogenic roles, yet the mechanisms underlying such opposing effects remain unclear. In this study, we identified a unique γδ T cell subset that coexpresses high levels of gut-homing integrins, CD103 and α4β7. They were exclusively found in the mesenteric lymph node after T cell-mediated colitis induction, and their appearance preceded the inflammation. Adoptive transfer of the CD103+α4β7high subsets enhanced Th1/Th17 T cell generation and accumulation in the intestine, and the disease severity. The level of generation correlated with the disease severity. Moreover, these cells were also found to be elevated in a spontaneous mouse model of ileitis. Based on the procolitogenic function, we referred to this subset as "inflammatory" γδ T cells. Targeting inflammatory γδ T cells may open a novel strategy to treat inflammatory diseases where γδ T cells play a pathogenic role including inflammatory bowel disease.
Collapse
Affiliation(s)
- Jeong-Su Do
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Sohee Kim
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Karen Keslar
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Eunjung Jang
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Emina Huang
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195; and
| | - Robert L Fairchild
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44116
| | - Booki Min
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195;
| |
Collapse
|
25
|
Hernández-Chirlaque C, Aranda CJ, Ocón B, Capitán-Cañadas F, Ortega-González M, Carrero JJ, Suárez MD, Zarzuelo A, Sánchez de Medina F, Martínez-Augustin O. Germ-free and Antibiotic-treated Mice are Highly Susceptible to Epithelial Injury in DSS Colitis. J Crohns Colitis 2016; 10:1324-1335. [PMID: 27117829 DOI: 10.1093/ecco-jcc/jjw096] [Citation(s) in RCA: 189] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 04/22/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Intestinal microbiota is required to maintain immune homeostasis and intestinal barrier function. At the same time, intraluminal bacteria are considered to be involved in inflammatory bowel disease and are required for colitis induction in animal models, with the possible exception of dextran sulphate sodium [DSS] colitis. This study was carried out to ascertain the mechanism underlying the induction of colitis by DSS in the absence of bacteria. METHODS Conventional and germ-free [GF] Naval Medical Research Institute [NMRI] mice were used, plus conventional mice treated with an antibiotic cocktail to deplete the intestinal microbiota ['pseudo-GF' or PGF mice]. The differential response to DSS was assessed. RESULTS Conventional mice developed DSS-induced colitis normally, whereas GF mice showed only minimal inflammation [no colonic thickening, lower myeloperoxidase activity, IL-6, IL-17, TNF-α, and IFN-γ secretion by splenocytes and mesenteric cell cultures, etc.]. However, these mice suffered enhanced haemorrhage, epithelial injury and mortality as a consequence of a weakened intestinal barrier, as shown by lower occludin, claudin 4, TFF3, MUC3, and IL-22. In contrast, PGF mice had a relatively normal, albeit attenuated, inflammatory response, but were less prone to haemorrhage and epithelial injury than GF mice. This was correlated with an increased expression of IL-10 and Foxp3 and preservation barrier-related markers. CONCLUSIONS We conclude that enteric bacteria are essential for the development of normal DSS-induced colitis. The absence of microbiota reduces DSS colonic inflammation dramatically but it also impairs barrier function, whereas subtotal microbiota depletion has intermediate effects at both levels.
Collapse
Affiliation(s)
- Cristina Hernández-Chirlaque
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas [CIBERehd], University of Granada, Granada, Spain
| | - Carlos J Aranda
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas [CIBERehd], University of Granada, Granada, Spain
| | - Borja Ocón
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas [CIBERehd], University of Granada, Granada, Spain
| | - Fermín Capitán-Cañadas
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas [CIBERehd], University of Granada, Granada, Spain
| | - Mercedes Ortega-González
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas [CIBERehd], University of Granada, Granada, Spain
| | | | - María Dolores Suárez
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas [CIBERehd], University of Granada, Granada, Spain
| | - Antonio Zarzuelo
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas [CIBERehd], University of Granada, Granada, Spain
| | - Fermín Sánchez de Medina
- Department of Pharmacology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas [CIBERehd], University of Granada, Granada, Spain
| | - Olga Martínez-Augustin
- Department of Biochemistry and Molecular Biology II, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas [CIBERehd], University of Granada, Granada, Spain
| |
Collapse
|
26
|
Hörmannsperger G, Schaubeck M, Haller D. Intestinal Microbiota in Animal Models of Inflammatory Diseases. ILAR J 2016; 56:179-91. [PMID: 26323628 DOI: 10.1093/ilar/ilv019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The intestinal microbiota has long been known to play an important role in the maintenance of health. In addition, alterations of the intestinal microbiota have recently been associated with a range of immune-mediated and metabolic disorders. Characterizing the composition and functionality of the intestinal microbiota, unravelling relevant microbe-host interactions, and identifying disease-relevant microbes are therefore currently of major interest in scientific and medical communities. Experimental animal models for the respective diseases of interest are pivotal in order to address functional questions on microbe-host interaction and to clarify the clinical relevance of microbiome alterations associated with disease initiation and development. This review presents an overview of the outcomes of highly sophisticated experimental studies on microbe-host interaction in animal models of inflammatory diseases, with a focus on inflammatory bowel disease (IBD). We will address the advantages and drawbacks of analyzing microbe-host interaction in complex colonized animal models compared with gnotobiotic animal models using monoassociation, simplified microbial consortia (SMC), or microbial humanization.
Collapse
Affiliation(s)
- G Hörmannsperger
- Gabriele Hörmannsperger, PhD, is a molecular biologist researcher, Monika Schaubeck, MSc, is a PhD student, and Dirk Haller, PhD, is full professor and head of the Chair of Nutrition and Immunology at the Technische Universität München, Freising-Weihenstephan, Germany
| | - M Schaubeck
- Gabriele Hörmannsperger, PhD, is a molecular biologist researcher, Monika Schaubeck, MSc, is a PhD student, and Dirk Haller, PhD, is full professor and head of the Chair of Nutrition and Immunology at the Technische Universität München, Freising-Weihenstephan, Germany
| | - D Haller
- Gabriele Hörmannsperger, PhD, is a molecular biologist researcher, Monika Schaubeck, MSc, is a PhD student, and Dirk Haller, PhD, is full professor and head of the Chair of Nutrition and Immunology at the Technische Universität München, Freising-Weihenstephan, Germany
| |
Collapse
|
27
|
Roulis M, Bongers G, Armaka M, Salviano T, He Z, Singh A, Seidler U, Becker C, Demengeot J, Furtado GC, Lira SA, Kollias G. Host and microbiota interactions are critical for development of murine Crohn's-like ileitis. Mucosal Immunol 2016; 9:787-97. [PMID: 26487367 PMCID: PMC5027991 DOI: 10.1038/mi.2015.102] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 08/31/2015] [Indexed: 02/04/2023]
Abstract
Deregulation of host-microbiota interactions in the gut is a pivotal characteristic of Crohn's disease. It remains unclear, however, whether commensals and/or the dysbiotic microbiota associated with pathology in humans are causally involved in Crohn's pathogenesis. Here, we show that Crohn's-like ileitis in Tnf(ΔARE/+) mice is microbiota-dependent. Germ-free Tnf(ΔARE/+) mice are disease-free and the microbiota and its innate recognition through Myd88 are indispensable for tumor necrosis factor (TNF) overexpression and disease initiation in this model. The epithelium of diseased mice shows no major defects in mucus barrier and paracellular permeability. However, Tnf(ΔARE/+) ileitis associates with the reduction of lysozyme-expressing Paneth cells, mediated by adaptive immune effectors. Furthermore, we show that established but not early ileitis in Tnf(ΔARE/+) mice involves defective expression of antimicrobials and dysbiosis, characterized by Firmicutes expansion, including epithelial-attaching segmented filamentous bacteria, and decreased abundance of Bacteroidetes. Microbiota modulation by antibiotic treatment at an early disease stage rescues ileitis. Our results suggest that the indigenous microbiota is sufficient to drive TNF overexpression and Crohn's ileitis in the genetically susceptible Tnf(ΔARE/+) hosts, whereas dysbiosis in this model results from disease-associated alterations including loss of lysozyme-expressing Paneth cells.
Collapse
Affiliation(s)
- M Roulis
- Institute of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Vari 16672, Greece
| | - G Bongers
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - M Armaka
- Institute of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Vari 16672, Greece
| | - T Salviano
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Z He
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - A Singh
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - U Seidler
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - C Becker
- Department of Medicine 1, Universitätsklinikum der Friedrich-Alexander-Universität, Erlangen, Germany
| | - J Demengeot
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - GC Furtado
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - SA Lira
- Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA,Address correspondence to: Sergio Lira, Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, Box 1630, New York, NY 10029-6574. Phone: 1-212-659-9404; Fax: 1-212-849-2525; and George Kollias, Biomedical Sciences Research Center “Alexander Fleming”, 34, Al. Fleming Street, 16672 Vari, Greece. Phone: +302109656507; Fax: +302109656563; and Department of Physiology, Medical School, National & Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27, Greece. Phone: +302107462507; Fax: +30210-7462571;
| | - G Kollias
- Institute of Immunology, Biomedical Sciences Research Center “Alexander Fleming”, Vari 16672, Greece,Second address: Department of Physiology, Medical School, National & Kapodistrian University of Athens, Athens 11527, Greece,Address correspondence to: Sergio Lira, Immunology Institute, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, Box 1630, New York, NY 10029-6574. Phone: 1-212-659-9404; Fax: 1-212-849-2525; and George Kollias, Biomedical Sciences Research Center “Alexander Fleming”, 34, Al. Fleming Street, 16672 Vari, Greece. Phone: +302109656507; Fax: +302109656563; and Department of Physiology, Medical School, National & Kapodistrian University of Athens, 75 Micras Asias, Goudi-Athens, 115 27, Greece. Phone: +302107462507; Fax: +30210-7462571;
| |
Collapse
|
28
|
De Salvo C, Wang XM, Pastorelli L, Mattioli B, Omenetti S, Buela KA, Chowdhry S, Garg RR, Goodman WA, Rodriguez-Palacios A, Smith DE, Abbott DW, Cominelli F, Bamias G, Xin W, Lee JJ, Vecchi M, Pizarro TT. IL-33 Drives Eosinophil Infiltration and Pathogenic Type 2 Helper T-Cell Immune Responses Leading to Chronic Experimental Ileitis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:885-898. [PMID: 26908008 PMCID: PMC5807926 DOI: 10.1016/j.ajpath.2015.11.028] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/03/2015] [Accepted: 11/19/2015] [Indexed: 02/08/2023]
Abstract
Although a clear association has been established between IL-33 and inflammatory bowel disease, mechanistic studies to date, primarily using acute murine models of colitis, have yielded contradicting results, demonstrating both pathogenic and protective roles. We used a well-characterized, spontaneous model of inflammatory bowel disease [ie, SAMP1/YitFc (SAMP) mice] to investigate the role of IL-33 during chronic intestinal inflammation. Our results showed marked eosinophil infiltration into the gut mucosa with increased levels of eotaxins and type 2 helper T-cell (Th2) cytokines as disease progressed and became more severe, which could be reversed upon either eosinophil depletion or blockade of IL-33 signaling. Exogenous IL-33 administration recapitulated these effects in ilea of uninflamed (parental) control AKR/J mice. Human data supported these findings, showing colocalization and up-regulation of IL-33 and eosinophils in the colonic mucosa of inflammatory bowel disease patients versus noninflamed controls. Finally, colonization of commensal flora by fecal material transplantation into germ-free SAMP and the presence of the gut microbiome induced IL-33, subsequent eosinophil infiltration, and mounting of Th2 immune responses, leading to exacerbation of chronic intestinal inflammation characteristic of SAMP mice. These data demonstrate a pathogenic role for IL-33-mediated eosinophilia and activation of Th2 immunity in chronic intestinal inflammation that is dependent on the gut microbiome. Targeting IL-33 may represent a novel therapeutic approach to treat patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Carlo De Salvo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, San Donato Milanese and Department of Biomedical Sciences, University of Milan, Milan, Italy
| | - Xiao-Ming Wang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Luca Pastorelli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, San Donato Milanese and Department of Biomedical Sciences, University of Milan, Milan, Italy
| | - Benedetta Mattioli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Sara Omenetti
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Kristine A Buela
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Saleem Chowdhry
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Rekha R Garg
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Wendy A Goodman
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | | | - Dirk E Smith
- Inflammation Research, Amgen, Seattle, Washington
| | - Derek W Abbott
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Giorgos Bamias
- Academic Department of Gastroenterology, Kapodistrian University of Athens and Laikon Hospital, Athens, Greece
| | - Wei Xin
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - James J Lee
- Division of Pulmonary Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, Arizona
| | - Maurizio Vecchi
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, San Donato Milanese and Department of Biomedical Sciences, University of Milan, Milan, Italy
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio.
| |
Collapse
|
29
|
Wu W, Liu HP, Chen F, Liu H, Cao AT, Yao S, Sun M, Evans-Marin HL, Zhao Y, Zhao Q, Duck LW, Elson CO, Liu Z, Cong Y. Commensal A4 bacteria inhibit intestinal Th2-cell responses through induction of dendritic cell TGF-β production. Eur J Immunol 2016; 46:1162-7. [PMID: 26840377 DOI: 10.1002/eji.201546160] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/31/2015] [Accepted: 01/26/2016] [Indexed: 12/19/2022]
Abstract
It has been shown that while commensal bacteria promote Th1, Th17 and Treg cells in lamina propria (LP) in steady-state conditions, they suppress mucosal Th2 cells. However, it is still unclear whether there are specific commensal organisms down-regulating Th2 responses, and the mechanism involved. Here we demonstrate that commensal A4 bacteria, a member of the Lachnospiraceae family, which produce an immunodominant microbiota CBir1 antigen, inhibits LP Th2-cell development. When transferred into the intestines of RAG(-/-) mice, CBir1-specific T cells developed predominately towards Th1 cells and Th17 cells, but to a lesser extent into Th2 cells. The addition of A4 bacterial lysates to CD4(+) T-cell cultures inhibited production of IL-4. A4 bacteria stimulated dendritic cell production of TGF-β, and blockade of TGF-β abrogated A4 bacteria inhibition of Th2-cell development in vitro and in vivo. Collectively, our data show that A4 bacteria inhibit Th2-cell differentiation by inducing dendritic cell production of TGF-β.
Collapse
Affiliation(s)
- Wei Wu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Hou-Pu Liu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Feidi Chen
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Han Liu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Department of Gastroenterology, The Qilu Hospital, Shandong University, Shandong, China
| | - Anthony T Cao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mingming Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Heather L Evans-Marin
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ye Zhao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Qing Zhao
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - L Wayne Duck
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles O Elson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhanju Liu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
30
|
Schaubeck M, Clavel T, Calasan J, Lagkouvardos I, Haange SB, Jehmlich N, Basic M, Dupont A, Hornef M, von Bergen M, Bleich A, Haller D. Dysbiotic gut microbiota causes transmissible Crohn's disease-like ileitis independent of failure in antimicrobial defence. Gut 2016; 65:225-37. [PMID: 25887379 PMCID: PMC4752651 DOI: 10.1136/gutjnl-2015-309333] [Citation(s) in RCA: 293] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 03/21/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Dysbiosis of the intestinal microbiota is associated with Crohn's disease (CD). Functional evidence for a causal role of bacteria in the development of chronic small intestinal inflammation is lacking. Similar to human pathology, TNF(deltaARE) mice develop a tumour necrosis factor (TNF)-driven CD-like transmural inflammation with predominant ileal involvement. DESIGN Heterozygous TNF(deltaARE) mice and wildtype (WT) littermates were housed under conventional (CONV), specific pathogen-free (SPF) and germ-free (GF) conditions. Microbial communities were analysed by high-throughput 16S ribosomal RNA gene sequencing. Metaproteomes were measured using LC-MS. Temporal and spatial resolution of disease development was followed after antibiotic treatment and transfer of microbial communities into GF mice. Granulocyte infiltration and Paneth cell function was assessed by immunofluorescence and gene expression analysis. RESULTS GF-TNF(deltaARE) mice were free of inflammation in the gut and antibiotic treatment of CONV-TNF(deltaARE) mice attenuated ileitis but not colitis, demonstrating that disease severity and location are microbiota-dependent. SPF-TNF(deltaARE) mice developed distinct ileitis-phenotypes associated with gradual loss of antimicrobial defence. 16S analysis and metaproteomics revealed specific compositional and functional alterations of bacterial communities in inflamed mice. Transplantation of disease-associated but not healthy microbiota transmitted CD-like ileitis to GF-TNF(deltaARE) recipients and triggered loss of lysozyme and cryptdin-2 expression. Monoassociation of GF-TNF(deltaARE) mice with the human CD-related Escherichia coli LF82 did not induce ileitis. CONCLUSIONS We provide clear experimental evidence for the causal role of gut bacterial dysbiosis in the development of chronic ileal inflammation with subsequent failure of Paneth cell function.
Collapse
Affiliation(s)
- Monika Schaubeck
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Thomas Clavel
- ZIEL-Institute for Food and Health, Technische Universität München, Freising-Weihenstephan, Germany
| | - Jelena Calasan
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany
| | - Ilias Lagkouvardos
- ZIEL-Institute for Food and Health, Technische Universität München, Freising-Weihenstephan, Germany
| | - Sven Bastiaan Haange
- Department of Proteomics, Helmholtz-Centre for Environmental Research—UFZ, Leipzig, Germany
| | - Nico Jehmlich
- Department of Proteomics, Helmholtz-Centre for Environmental Research—UFZ, Leipzig, Germany
| | - Marijana Basic
- Institut for Medical Microbiology, RWTH University, Aachen, Germany
| | - Aline Dupont
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany,Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Mathias Hornef
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany,Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Martin von Bergen
- Department of Proteomics, Helmholtz-Centre for Environmental Research—UFZ, Leipzig, Germany,UFZ, Department of Metabolomics, Helmholtz-Centre for Environmental Research, Leipzig, Germany,Department of Biotechnology, Chemistry and Environmental Engineering, University of Aalborg, Aalborg, Denmark
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technische Universität München, Freising-Weihenstephan, Germany,ZIEL-Institute for Food and Health, Technische Universität München, Freising-Weihenstephan, Germany
| |
Collapse
|
31
|
Buttó LF, Schaubeck M, Haller D. Mechanisms of Microbe-Host Interaction in Crohn's Disease: Dysbiosis vs. Pathobiont Selection. Front Immunol 2015; 6:555. [PMID: 26635787 PMCID: PMC4652232 DOI: 10.3389/fimmu.2015.00555] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/16/2015] [Indexed: 12/11/2022] Open
Abstract
Crohn’s disease (CD) is a systemic chronic inflammatory condition mainly characterized by discontinuous transmural pathology of the gastrointestinal tract and frequent extraintestinal manifestations with intermittent episodes of remission and relapse. Genome-wide association studies identified a number of risk loci that, catalyzed by environmental triggers, result in the loss of tolerance toward commensal bacteria based on dysregulated innate effector functions and antimicrobial defense, leading to exacerbated adaptive immune responses responsible for chronic immune-mediated tissue damage. In this review, we discuss the inter-related role of changes in the intestinal microbiota, epithelial barrier integrity, and immune cell functions on the pathogenesis of CD, describing the current approaches available to investigate the molecular mechanisms underlying the disease. Substantial effort has been dedicated to define disease-associated changes in the intestinal microbiota (dysbiosis) and to link pathobionts to the etiology of inflammatory bowel diseases. A cogent definition of dysbiosis is lacking, as well as an agreement of whether pathobionts or complex shifts in the microbiota trigger inflammation in the host. Among the rarely available animal models, SAMP/Yit and TNFdeltaARE mice are the best known displaying a transmural CD-like phenotype. New hypothesis-driven mouse models, e.g., epithelial-specific Caspase8−/−, ATG16L1−/−, and XBP1−/− mice, validate pathway-focused function of specific CD-associated risk genes highlighting the role of Paneth cells in antimicrobial defense. To study the causal role of bacteria in initiating inflammation in the host, the use of germ-free mouse models is indispensable. Unraveling the interactions of genes, immune cells and microbes constitute a criterion for the development of safe, reliable, and effective treatment options for CD.
Collapse
Affiliation(s)
- Ludovica F Buttó
- Chair of Nutrition and Immunology, Technische Universität München , Freising-Weihenstephan , Germany
| | - Monika Schaubeck
- Chair of Nutrition and Immunology, Technische Universität München , Freising-Weihenstephan , Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technische Universität München , Freising-Weihenstephan , Germany
| |
Collapse
|
32
|
Singh V, Yeoh BS, Carvalho F, Gewirtz AT, Vijay-Kumar M. Proneness of TLR5 deficient mice to develop colitis is microbiota dependent. Gut Microbes 2015; 6:279-83. [PMID: 26067589 PMCID: PMC4615783 DOI: 10.1080/19490976.2015.1060390] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Alterations in the gut microbiota have been implicated to play a role in potentiating inflammatory bowel diseases in both humans and mice. Mice lacking the flagellin receptor, toll-like receptor 5 (TLR5), are prone to develop spontaneous gut inflammation, but are significantly protected when treated with antibiotics or maintained in germ-free conditions. However, given that the incidence of spontaneous inflammation in TLR5KO mice is quite variable in conventional conditions (typically ∼10% show clear colitis), this result is far from definitive and does not rule out that TLR5KO mice might be prone to develop inflammation even in the absence of a microbiota. Herein, we demonstrate that neutralization of IL10 signaling induces colitis in 100% of TLR5KO mice which provide a more rigorous approach to evaluate the role of microbiota in gut inflammation. Mice treated with antibiotics or maintained in germ-free condition are substantially protected against IL-10R neutralization-induced colitis, underscoring that gut inflammation in TLR5KO mice is dependent upon the presence of a gut microbiota.
Collapse
Affiliation(s)
- Vishal Singh
- Department of Nutritional Sciences; The Pennsylvania State University; University Park, PA USA
| | - Beng San Yeoh
- Department of Nutritional Sciences; The Pennsylvania State University; University Park, PA USA
| | | | - Andrew T Gewirtz
- Institute of Biomedical Sciences; Georgia State University; Atlanta, GA USA
| | - Matam Vijay-Kumar
- Department of Nutritional Sciences; The Pennsylvania State University; University Park, PA USA,Department of Medicine; The Pennsylvania State University Medical Center; Hershey, PA USA,Correspondence to: Matam Vijay-Kumar;
| |
Collapse
|
33
|
Omenetti S, Brogi M, Goodman WA, Croniger CM, Eid S, Huang AY, Laffi G, Roskams T, Cominelli F, Pinzani M, Pizarro TT. Dysregulated intrahepatic CD4 + T-cell activation drives liver inflammation in ileitis-prone SAMP1/YitFc mice. Cell Mol Gastroenterol Hepatol 2015; 1. [PMID: 26213712 PMCID: PMC4511857 DOI: 10.1016/j.jcmgh.2015.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Liver inflammation is a common extraintestinal manifestation of inflammatory bowel disease (IBD); however, whether liver involvement is a consequence of a primary intestinal defect or results from alternative pathogenic processes remains unclear. Therefore, we sought to determine the potential pathogenic mechanism(s) of concomitant liver inflammation in an established murine model of IBD. METHODS Liver inflammation and immune cell subsets were characterized in ileitis-prone SAMP1/YitFc (SAMP) and AKR/J (AKR) control mice, lymphocyte-depleted SAMP (SAMPxRag-1-/-), and immunodeficient SCID recipient mice receiving SAMP or AKR donor CD4+ T-cells. Proliferation and suppressive capacity of CD4+ T-effector (Teff) and T-regulatory (Treg) cells from gut-associated lymphoid tissue (GALT) and livers of SAMP and AKR mice were measured. RESULTS Surprisingly, prominent inflammation was detected in 4-wk-old SAMP livers, prior to histologic evidence of ileitis, while both disease phenotypes were absent in age-matched AKRs. SAMP liver disease was characterized by abundant infiltration of lymphocytes, required for hepatic inflammation to occur, a Th1-skewed environment, and phenotypically-activated CD4+ T-cells. SAMP intrahepatic CD4+ T-cells also had the ability to induce liver and ileal inflammation when adoptively transferred into SCID recipients, whereas GALT-derived CD4+ T-cells produced milder ileitis, but not liver inflammation. Interestingly, SAMP intrahepatic CD4+ Teff cells showed increased proliferation compared to both SAMP GALT- and AKR liver-derived CD4+ Teff cells, while SAMP intrahepatic Tregs were decreased among CD4+ T-cells and impaired in in vitro suppressive function compared to AKR. CONCLUSIONS Activated intrahepatic CD4+ T-cells induce liver inflammation and contribute to experimental ileitis via locally-impaired hepatic immunosuppressive function.
Collapse
Affiliation(s)
- Sara Omenetti
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,“DENOThe” Center, University of Florence, Florence, Italy
| | - Marco Brogi
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,“DENOThe” Center, University of Florence, Florence, Italy
| | - Wendy A. Goodman
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Colleen M. Croniger
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Saada Eid
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Alex Y. Huang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Giacomo Laffi
- “DENOThe” Center, University of Florence, Florence, Italy
| | - Tania Roskams
- Department of Morphology and Molecular Pathology, University of Leuven, Leuven, Belgium
| | - Fabio Cominelli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,“DENOThe” Center, University of Florence, Florence, Italy,Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Massimo Pinzani
- “DENOThe” Center, University of Florence, Florence, Italy,Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, United Kingdom
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio,“DENOThe” Center, University of Florence, Florence, Italy,Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio,Correspondence Address correspondence to: Theresa T. Pizarro, PhD, Department of Pathology, Case Western Reserve University School of Medicine, 2103 Cornell Road, WRB 5534, Cleveland, Ohio 44106. fax: (216) 368-0494.
| |
Collapse
|
34
|
Rogler G. New therapeutic avenues for treatment of fibrosis: can we learn from other diseases? Dig Dis 2014; 32 Suppl 1:39-49. [PMID: 25531352 DOI: 10.1159/000367825] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Crohn's disease (CD) is characterized by the frequent occurrence of complications, such as fibrotic strictures and subsequently the need for CD-related surgery. Chronic or recurrent inflammation is generally regarded to be a necessary precondition for the initiation of intestinal fibrosis. In this view, fibrosis is a pathologically augmented healing response to inflammation-induced mucosal tissue destruction and injury. At present, there are no approved or effective medical therapies aimed specifically at fibrosis or stricture in IBD. Indirect benefits may occur from anti-inflammatory therapies, although there is no consensus on this. Therapy for fibrosis is complicated by the fact that a wound-healing response is essential in CD and ulcerative colitis. Several pharmaceutical companies are now working on the therapy of fibrosis in other diseases. Strategies interfering with TGF-β expression and activation are promising. Pirfenidone has been studied in several clinical trials. Further therapeutic options are second-generation and wide-spectrum tyrosine kinase inhibitors. These inhibit growth factor receptor signaling, thus reducing fibrosis in animal models and some patients with tumor-associated fibrosis. At present, the development of antifibrotic therapies takes place in other diseases such as lung and liver fibrosis. This is partially due to a lack of experimental models for gut fibrosis and the fact that reliable readouts (MRI, serum markers) in patients are lacking. It will be important to test the above-mentioned newly available treatment strategies in IBD to profit from progress in other fibrotic diseases.
Collapse
Affiliation(s)
- Gerhard Rogler
- Division of Gastroenterology and Hepatology, University Hospital Zurich, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Abstract
Intestinal mucosal barrier function is the capacity of the intestine to provide adequate containment of luminal microorganisms and molecules while preserving the ability to absorb nutrients. The central element is the epithelial layer, which physically separates the lumen and the internal milieu and is in charge of vectorial transport of ions, nutrients, and other substances. The secretion of mucus-forming mucins, sIgA, and antimicrobial peptides reinforces the mucosal barrier on the extraepithelial side, while a variety of immune cells contributes to mucosal defense in the inner side. Thus, the mucosal barrier is of physical, biochemical, and immune nature. In addition, the microbiota may be viewed as part of this system because of the mutual influence occurring between the host and the luminal microorganisms. Alteration of the mucosal barrier function with accompanying increased permeability and/or bacterial translocation has been linked with a variety of conditions, including inflammatory bowel disease. Genetic and environmental factors may converge to evoke a defective function of the barrier, which in turn may lead to overt inflammation of the intestine as a result of an exacerbated immune reaction toward the microbiota. According to this hypothesis, inflammatory bowel disease may be both precipitated and treated by either stimulation or downregulation of the different elements of the mucosal barrier, with the outcome depending on timing, the cell type affected, and other factors. In this review, we cover briefly the elements of the barrier and their involvement in functional defects and the resulting phenotype.
Collapse
|
36
|
Ray S, De Salvo C, Pizarro TT. Central role of IL-17/Th17 immune responses and the gut microbiota in the pathogenesis of intestinal fibrosis. Curr Opin Gastroenterol 2014; 30:531-8. [PMID: 25255234 PMCID: PMC4512208 DOI: 10.1097/mog.0000000000000119] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Intestinal fibrosis is a serious, yet common, outcome in patients with inflammatory bowel disease (IBD). Despite advances in developing novel treatment modalities to control chronic gut inflammation characteristic of IBD, no effective antifibrotic therapies exist to date. As such, a deeper understanding of the molecular mechanisms underlying intestinal fibrosis and the availability of relevant animal models are critical to move this area of investigation forward. RECENT FINDINGS Emerging concepts in the pathogenesis of intestinal fibrosis include the central role of interleukin (IL)-17 and Th17 immune responses, although their precise contribution to chronic inflammation and IBD remains controversial. Other novel mediators of intestinal fibrosis, such as tumor necrosis factor-like ligand 1A and components of the renin-angiotensin system, support the importance of IL-17. Additionally, recent studies utilizing novel mouse models highlight the significance of the gut microbiota and link components of bacterial sensing, including nucleotide-binding oligomerization domain-containing protein 2, to IL-17/Th17 immune responses in the development of inflammation-associated intestinal fibrosis. SUMMARY Recent progress in identifying key mediators, novel animal models, and important mechanistic pathways in the pathogenesis of intestinal fibrosis holds promise for the development of effective antifibrotics in an area of significant, unmet clinical need.
Collapse
Affiliation(s)
- Shuvra Ray
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Department of Gastroenterology, Guy’s & St. Thomas’ Hospitals/University College London, London, United Kingdom
| | - Carlo De Salvo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
37
|
Goodman WA, Garg RR, Reuter BK, Mattioli B, Rissman EF, Pizarro TT. Loss of estrogen-mediated immunoprotection underlies female gender bias in experimental Crohn's-like ileitis. Mucosal Immunol 2014; 7:1255-65. [PMID: 24621993 PMCID: PMC4139459 DOI: 10.1038/mi.2014.15] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 02/01/2014] [Accepted: 02/10/2014] [Indexed: 02/04/2023]
Abstract
The incidence and severity of Crohn's disease (CD) are increased in female patients. Using SAMP1/YitFc (SAMP) mice, a spontaneous model of chronic intestinal inflammation that displays histologic and pathogenic similarities to human CD, we investigated the potential mechanism(s) contributing to sex differences observed in CD. Similar to gender differences observed in CD patients, SAMP female (SAMP-F) mice displayed an earlier onset and more severe ileitis compared with SAMP male (SAMP-M) mice. Furthermore, T-regulatory cells (Tregs) from gut-associated lymphoid tissue (GALT) of SAMP-F mice were reduced in frequency and impaired in their in vitro and in vivo suppressive functions compared with that of SAMP-M mice. Given the interaction between sex hormones and Treg function, we investigated the possible role of estrogen (E2) in SAMP ileitis. SAMP-M mice responded to exogenous E2 administration by expanding Treg frequency and reducing ileal inflammation, whereas SAMP-F mice were resistant. Conventional T cells and Tregs responded differentially to estrogen signaling, leading to distinct immunoprotective effects mediated by distinct estrogen receptor (ER) isoforms. These mechanisms were impaired in T cells from SAMP-F mice. Thus, hormone signaling influences the expansion and function of GALT Tregs in an ER-dependent manner and contributes to gender-based differences in experimental CD.
Collapse
Affiliation(s)
- Wendy A. Goodman
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Rekha R. Garg
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Brian K. Reuter
- Centre of Excellence for Gastrointestinal Inflammation and Immunity Research, University of Alberta, Edmonton, Alberta, Canada T6G 2X8
| | - Benedetta Mattioli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Emilie F. Rissman
- Department of Biochemistry & Molecular Genetics, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
38
|
Jones-Hall YL, Grisham MB. Immunopathological characterization of selected mouse models of inflammatory bowel disease: Comparison to human disease. ACTA ACUST UNITED AC 2014; 21:267-88. [PMID: 24935242 DOI: 10.1016/j.pathophys.2014.05.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/16/2014] [Accepted: 05/17/2014] [Indexed: 12/19/2022]
Abstract
Inflammatory bowel diseases (IBD) are chronic, relapsing conditions of multifactorial etiology. The two primary diseases of IBD are Crohn's disease (CD) and ulcerative colitis (UC). Both entities are hypothesized to occur in genetically susceptible individuals due to microbial alterations and environmental contributions. The exact etiopathogenesis, however, is not known for either disease. A variety of mouse models of CD and UC have been developed to investigate the pathogenesis of these diseases and evaluate treatment modalities. Broadly speaking, the mouse models can be divided into 4 categories: genetically engineered, immune manipulated, spontaneous and erosive/chemically induced. No one mouse model completely recapitulates the immunopathology of CD or UC, however each model possesses particular similarities to human IBD and offers advantageous for specific details of IBD pathogenesis. Here we discuss the more commonly used models in each category and critically evaluate how the immunopathology induced compares to CD or UC, as well as the advantages and disadvantages associated with each model.
Collapse
Affiliation(s)
- Yava L Jones-Hall
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47906, United States.
| | - Matthew B Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States
| |
Collapse
|
39
|
The role of Klebsiella in Crohn's disease with a potential for the use of antimicrobial measures. Int J Rheumatol 2013; 2013:610393. [PMID: 24223596 PMCID: PMC3810322 DOI: 10.1155/2013/610393] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 09/04/2013] [Indexed: 12/23/2022] Open
Abstract
There is a general consensus that Crohn's disease (CD) develops as the result of immune-mediated tissue damage triggered by infections with intestinal microbial agents. Based on the results of existing microbiological, molecular, and immunological studies, Klebsiella microbe seems to have a key role in the initiation and perpetuation of the pathological damage involving the gut and joint tissues in patients with CD. Six different gastroenterology centres in the UK have reported elevated levels of antibodies to Klebsiella in CD patients. There is a relationship between high intake of starch-containing diet, enhanced growth of gut microbes, and the production of pullulanases by Klebsiella. It is proposed that eradication of these microbes by the use of antibiotics and low starch diet, in addition to the currently used treatment, could help in alleviating or halting the disease process in CD.
Collapse
|
40
|
|
41
|
Ishikawa D, Okazawa A, Corridoni D, Jia LG, Wang ZM, Xin W, Arseneau KO, Pizarro TT, Cominelli F. Tregs are dysfunctional in vivo in a spontaneous murine model of Crohn's disease. Mucosal Immunol 2013; 6:267-75. [PMID: 22785225 PMCID: PMC3606708 DOI: 10.1038/mi.2012.67] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although regulatory T cells (Tregs) have been implicated in inflammatory bowel disease, Tregs from Crohn's disease (CD) patients are increased in number and function normally in vitro. To clarify this disparity, we studied Treg function in vivo using a spontaneous model of CD-like ileitis. We first administered anti-CD25-depleting antibodies to SAMP1/YitFc (SAMP) mice to assess ileitis; mesenteric lymph node cells were then transferred into SCID (severe combined immunodeficient) recipients to induce colitis. CD25 depletion increased the severity of both spontaneous ileitis and adoptively transferred colitis. Interestingly, a second transfer of CD4(+)CD25(+) cells from untreated AKR control mice was able to ameliorate the induced colitis, whereas CD4(+)CD25(+) cells from untreated SAMP mice were not, suggesting a functional abnormality in SAMP Tregs. Anti-CD25 treatment in SAMP mice also induced proliferation of CD25(-)Foxp3(+) Tregs, which had a proinflammatory intestinal T helper type 1/ T helper type 2 (Th1/Th2) effector phenotype. These studies demonstrate Treg dysfunction in a spontaneous model of CD-like ileitis.
Collapse
Affiliation(s)
- Dai Ishikawa
- Departments of Medicine and Pathology, Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| | - Akira Okazawa
- Departments of Medicine and Pathology, Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| | - Daniele Corridoni
- Departments of Medicine and Pathology, Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| | - Li Guo Jia
- Departments of Medicine and Pathology, Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| | - Ziao Ming Wang
- Departments of Medicine and Pathology, Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| | - Wei Xin
- Departments of Medicine and Pathology, Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| | - Kristen O. Arseneau
- Departments of Medicine and Pathology, Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| | - Theresa T. Pizarro
- Departments of Medicine and Pathology, Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| | - Fabio Cominelli
- Departments of Medicine and Pathology, Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| |
Collapse
|
42
|
Abstract
Chronic inflammation of the intestine is detrimental to mammals. Similarly, constant activation of the immune response in the gut by the endogenous flora is suspected to be harmful to Drosophila. Therefore, the innate immune response in the gut of Drosophila melanogaster is tightly balanced to simultaneously prevent infections by pathogenic microorganisms and tolerate the endogenous flora. Here we describe the role of the big bang (bbg) gene, encoding multiple membrane-associated PDZ (PSD-95, Discs-large, ZO-1) domain-containing protein isoforms, in the modulation of the gut immune response. We show that in the adult Drosophila midgut, BBG is present at the level of the septate junctions, on the apical side of the enterocytes. In the absence of BBG, these junctions become loose, enabling the intestinal flora to trigger a constitutive activation of the anterior midgut immune response. This chronic epithelial inflammation leads to a reduced lifespan of bbg mutant flies. Clearing the commensal flora by antibiotics prevents the abnormal activation of the gut immune response and restores a normal lifespan. We now provide genetic evidence that Drosophila septate junctions are part of the gut immune barrier, a function that is evolutionarily conserved in mammals. Collectively, our data suggest that septate junctions are required to maintain the subtle balance between immune tolerance and immune response in the Drosophila gut, which represents a powerful model to study inflammatory bowel diseases.
Collapse
|
43
|
Aomatsu T, Imaeda H, Takahashi K, Fujimoto T, Kasumi E, Ban H, Bamba S, Yoden A, Tamai H, Fujiyama Y, Andoh A. Neutralization of complement component C5 ameliorates the development of dextran sulfate sodium (DSS)-colitis in mice. J Clin Biochem Nutr 2012; 52:72-5. [PMID: 23341701 PMCID: PMC3541422 DOI: 10.3164/jcbn.12-63] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 07/23/2012] [Indexed: 01/08/2023] Open
Abstract
The complement system is a potent effector of innate immunity. To elucidate the pathophysiological role of the complement system in inflammatory bowel disease, we evaluated the effects of anti-C5 antibodies on the development of dextran sulfate sodium-induced colitis in mice. Dextran sulfate sodium-colitis was induced in BALB/c mice with intraperitoneal administrations of anti-C5 antibodies (1 mg/body [DOSAGE ERROR CORRECTED]) every 48 h. Tissue samples were evaluated by standard histological procedures. The mucosal mRNA expression of the inflammatory cytokines was analyzed by real-time PCR. Body weight loss in the mice was completely blocked by the administration of anti-C5 antibody. The disease activity index was significantly lower in the anti-C5 antibody-treated mice than the dextran sulfate sodium mice. The colonic weight/length ratio, histological colitis score and mucosal myeloperoxidase activity were significantly lower in the anti-C5 antibody-treated mice than the dextran sodium sulfate mice. The administration of the anti-C5 antibody significantly reduced the mucosal expression of mRNAs for tumor necrosis factor-α, interleukin-1β and interleukin-6. In conclusion, the complement system plays a role in the development of dextran sodium sulfate-induced experimental colitis.
Collapse
Affiliation(s)
- Tomoki Aomatsu
- Department of Medicine, Shiga University of Medical Science, Seta Tsukinowa, Otsu 520-2192, Japan ; Department of Pediatrics, Osaka Medical College, 2-7 Daigaku-cho, Takatsuki 569-0801, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Natividad JMM, Verdu EF. Modulation of intestinal barrier by intestinal microbiota: pathological and therapeutic implications. Pharmacol Res 2012; 69:42-51. [PMID: 23089410 DOI: 10.1016/j.phrs.2012.10.007] [Citation(s) in RCA: 324] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 10/11/2012] [Accepted: 10/13/2012] [Indexed: 02/07/2023]
Abstract
Mammals and their intestinal microbiota peacefully coexist in a mutualistic relationship. Commensal bacteria play an active role in shaping and modulating physiological processes in the host, which include, but are not restricted to, the immune system and the intestinal barrier. Both play a crucial role in containing intestinal bacteria and other potentially noxious luminal antigens within the lumen and mucosal compartment. Although mutualism defines the relationship between the host and the intestinal microbiota, disruptions in this equilibrium may promote disease. Thus, alterations in gut microbiota (dysbiosis) have been linked to the recent increased expression of obesity, allergy, autoimmunity, functional and inflammatory disorders such as irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD). In this article, we review the evidence supporting a role of gut microbiota in regulating intestinal barrier function. We discuss the hypothesis that microbial factors can modulate the barrier in ways that can prevent or promote gastrointestinal disease. A better understanding of the role of the intestinal microbiota in maintaining a functional intestinal barrier may help develop targeted strategies to prevent and treat disease.
Collapse
Affiliation(s)
- Jane M M Natividad
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | | |
Collapse
|
45
|
Maxwell JR, Viney JL. Overview of mouse models of inflammatory bowel disease and their use in drug discovery. ACTA ACUST UNITED AC 2012; Chapter 5:Unit5.57. [PMID: 22294403 DOI: 10.1002/0471141755.ph0557s47] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Inflammatory bowel disease (IBD), a condition that affects millions of individuals, encompasses two distinct conditions: Crohn's disease (CD) and ulcerative colitis (UC). CD is an inflammatory condition affecting any part of the digestive tract between the mouth and anus, but, most commonly, the ileum and colon. It is distinguished by the presence of granulomas in the mucosal tissue and patchy areas of transmural inflammation. UC is restricted to the colon and is manifest as continuous inflammation starting from the rectum and extending back towards the cecum. Inflammation in UC is primarily restricted to mucosal layers. Research is ongoing to understand the causality of these two diseases, and advances in understanding of their pathology have resulted from the variety of mouse models of IBD that have emerged since the early 1990s. Described in this unit are contemporary mouse models of these conditions and examples of their use in drug discovery.
Collapse
|
46
|
Ernst PB, Erickson LD, Loo WM, Scott KG, Wiznerowicz EB, Brown CC, Torres-Velez FJ, Alam MS, Black SG, McDuffie M, Feldman SH, Wallace JL, McKnight GW, Padol IT, Hunt RH, Tung KS. Spontaneous autoimmune gastritis and hypochlorhydria are manifest in the ileitis-prone SAMP1/YitFcs mice. Am J Physiol Gastrointest Liver Physiol 2012; 302:G105-15. [PMID: 21921286 PMCID: PMC3345967 DOI: 10.1152/ajpgi.00194.2011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
SAMP1/YitFcs mice serve as a model of Crohn's disease, and we have used them to assess gastritis. Gastritis was compared in SAMP1/YitFcs, AKR, and C57BL/6 mice by histology, immunohistochemistry, and flow cytometry. Gastric acid secretion was measured in ligated stomachs, while anti-parietal cell antibodies were assayed by immunofluorescence and enzyme-linked immunosorbent spot assay. SAMP1/YitFcs mice display a corpus-dominant, chronic gastritis with multifocal aggregates of mononuclear cells consisting of T and B lymphocytes. Relatively few aggregates were observed elsewhere in the stomach. The infiltrates in the oxyntic mucosa were associated with the loss of parietal cell mass. AKR mice, the founder strain of the SAMP1/YitFcs, also have gastritis, although they do not develop ileitis. Genetic studies using SAMP1/YitFcs-C57BL/6 congenic mice showed that the genetic regions regulating ileitis had comparable effects on gastritis. The majority of the cells in the aggregates expressed the T cell marker CD3 or the B cell marker B220. Adoptive transfer of SAMP1/YitFcs CD4(+) T helper cells, with or without B cells, into immunodeficient recipients induced a pangastritis and duodenitis. SAMP1/YitFcs and AKR mice manifest hypochlorhydria and anti-parietal cell antibodies. These data suggest that common genetic factors controlling gastroenteric disease in SAMP1/YitFcs mice regulate distinct pathogenic mechanisms causing inflammation in separate sites within the digestive tract.
Collapse
Affiliation(s)
| | | | | | - K. G. Scott
- 6Department of Biology, University of Manitoba, Winnipeg, Manitoba;
| | | | - C. C. Brown
- 7Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia;
| | - F. J. Torres-Velez
- 8Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, Bethesda; and
| | - M. S. Alam
- 9Immunobiology Branch, Center for Food Safety and Nutrition, US Food and Drug Administration, Laurel, Maryland
| | | | | | - S. H. Feldman
- 5Center for Comparative Medicine, University of Virginia, Charlottesville, Virginia;
| | - J. L. Wallace
- 10Department of Medicine and The Farncombe Institute, McMaster University, Hamilton, Ontario, Canada;
| | - G. W. McKnight
- 10Department of Medicine and The Farncombe Institute, McMaster University, Hamilton, Ontario, Canada;
| | - I. T. Padol
- 10Department of Medicine and The Farncombe Institute, McMaster University, Hamilton, Ontario, Canada;
| | - R. H. Hunt
- 10Department of Medicine and The Farncombe Institute, McMaster University, Hamilton, Ontario, Canada;
| | | |
Collapse
|
47
|
Mizoguchi A. Animal models of inflammatory bowel disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 105:263-320. [PMID: 22137435 DOI: 10.1016/b978-0-12-394596-9.00009-3] [Citation(s) in RCA: 179] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory condition that is medicated by genetic, immune, and environmental factors. At least 66 different kinds of animal models have been established to study IBD, which are classified primarily into chemically induced, cell-transfer, congenial mutant, and genetically engineered models. These IBD models have provided significant contributions to not only dissect the mechanism but also develop novel therapeutic strategies for IBD. In addition, recent advances on genetically engineered techniques such as cell-specific and inducible knockout as well as knockin mouse systems have brought novel concepts on IBD pathogenesis to the fore. Further, mouse models, which lack some IBD susceptibility genes, have suggested more complicated mechanism of IBD than previously predicted. This chapter summarizes the distinct feature of each murine IBD model and discusses the previous and current lessons from the IBD models.
Collapse
Affiliation(s)
- Atsushi Mizoguchi
- Department of Pathology, Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
48
|
Pizarro TT, Pastorelli L, Bamias G, Garg RR, Reuter BK, Mercado JR, Chieppa M, Arseneau KO, Ley K, Cominelli F. SAMP1/YitFc mouse strain: a spontaneous model of Crohn's disease-like ileitis. Inflamm Bowel Dis 2011; 17:2566-2584. [PMID: 21557393 PMCID: PMC3154989 DOI: 10.1002/ibd.21638] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 01/03/2011] [Indexed: 12/18/2022]
Abstract
The SAMP1/YitFc mouse strain represents a model of Crohn's disease (CD)-like ileitis that is ideal for investigating the pathogenesis of chronic intestinal inflammation. Different from the vast majority of animal models of colitis, the ileal-specific phenotype characteristic of SAMP1/YitFc mice occurs spontaneously, without genetic, chemical, or immunological manipulation. In addition, SAMP1/YitFc mice possess remarkable similarities to the human condition with regard to disease location, histologic features, incidence of extraintestinal manifestations, and response to conventional therapies. SAMP1/YitFc mice also display a well-defined time course of a predisease state and phases of acute and chronic ileitis. As such, the SAMP1/YitFc model is particularly suitable for elucidating pathways that precede the clinical phenotype that may lead to preventive, and therefore more efficacious, intervention with the natural course of disease, or alternatively, for the development of therapeutic strategies directed against chronic, established ileitis. In this review we summarize important contributions made by our group and others that uncover potential mechanisms in the pathogenesis of CD using this unique murine model of chronic intestinal inflammation.
Collapse
Affiliation(s)
- Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Collins CB, Aherne CM, Kominsky D, McNamee EN, Lebsack MD, Eltzschig H, Jedlicka P, Rivera-Nieves J. Retinoic acid attenuates ileitis by restoring the balance between T-helper 17 and T regulatory cells. Gastroenterology 2011; 141:1821-31. [PMID: 22027263 PMCID: PMC3684415 DOI: 10.1053/j.gastro.2011.05.049] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 05/11/2011] [Accepted: 05/26/2011] [Indexed: 12/23/2022]
Abstract
BACKGROUND & AIMS Retinoic acid (RA), produced by intestinal epithelial cells (IECs) and dendritic cells (DCs) promotes the induction of regulatory T cells (Tregs) and decreases the induction of T-helper (Th)17 cells. METHODS We studied the roles of RA in mice that overproduce tumor necrosis factor (TNF) and develop chronic ileitis (TNF_ARE mice). We assessed the frequency and function of CD103+ DCs, Th17 cells, and Tregs by flow cytometry, and we measured expression of cytokines and retinaldehyde dehydrogenase (RALDH) enzymes in ileum samples, DCs, and IECs by real-time polymerase chain reaction. We quantified RA by electrochemical analysis and examined the effect of RA supplementation on TNF-induced ileitis using histologic, coculture, and suppression assays and flow cytometry. RESULTS Numbers of CD103+ DCs decreased in the inflamed ilea of mice with chronic disease; RA synthetic machinery (RALDH1,2) was down-regulated. Nevertheless, the proportion of CD4+, CD25+, FoxP3+ Tregs increased, indicating an alternate source for RA. IECs responded to reduced levels of RA by up-regulating RALDH3 in vivo and in vitro. Net tissue levels of RA remained lower in TNF+ARE than wild-type mice, indicating that epithelial up-regulation of RALDH3 could not maintain adequate concentrations of RA, probably because of loss of IEC mass. RA supplementation significantly attenuated disease by increasing the number and function of CD103+ DCs and Tregs and reducing Th17 cells. CONCLUSIONS Reduced levels of RA appear to induce IECs to up-regulate synthesis of RA. RA supplementation attenuates ileitis through its effects on CD103+ DCs, Tregs, and Th17 cells. RA supplementation might offer therapeutic benefit in Crohn's disease.
Collapse
Affiliation(s)
- Colm B. Collins
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Carol M. Aherne
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045,Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Douglas Kominsky
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045,Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Eóin N. McNamee
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Matthew D.P. Lebsack
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Holger Eltzschig
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045,Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Paul Jedlicka
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045
| | - Jesús Rivera-Nieves
- Mucosal Inflammation Program, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045,Division of Gastroenterology and Hepatology, University of California San Diego
| |
Collapse
|
50
|
Yi P, Li L. The germfree murine animal: an important animal model for research on the relationship between gut microbiota and the host. Vet Microbiol 2011; 157:1-7. [PMID: 22079217 DOI: 10.1016/j.vetmic.2011.10.024] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 10/17/2011] [Accepted: 10/19/2011] [Indexed: 12/26/2022]
Abstract
Scientific findings in recent decades have demonstrated that the commensal intestinal microbiota has profound effects on the physiology and diseases of the host. It is estimated that the human microbiota is composed of 10(14) bacterial cells, a number 10 times greater than the total number of human cells. The variety and the complex interactions of the intestinal microbiota are associated with physiological details that remain largely unknown. Germfree hosts, especially murine (rat or mouse) animals that have been maintained free from demonstrable microbial associates such as bacteria, viruses, fungi, and parasites throughout life, have become a powerful tool for exploring the interplay between the host and microorganisms inhabiting the human intestine. This review and survey of recent findings will argue that the germfree mouse model can produce its greatest potential benefits in the study of the metabolism and immunity of the host.
Collapse
Affiliation(s)
- Ping Yi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou 310003, China
| | | |
Collapse
|