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
|
Rieder F, Mukherjee PK, Massey WJ, Wang Y, Fiocchi C. Fibrosis in IBD: from pathogenesis to therapeutic targets. Gut 2024; 73:854-866. [PMID: 38233198 PMCID: PMC10997492 DOI: 10.1136/gutjnl-2023-329963] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/29/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Intestinal fibrosis resulting in stricture formation and obstruction in Crohn's disease (CD) and increased wall stiffness leading to symptoms in ulcerative colitis (UC) is among the largest unmet needs in inflammatory bowel disease (IBD). Fibrosis is caused by a multifactorial and complex process involving immune and non-immune cells, their soluble mediators and exposure to luminal contents, such as microbiota and environmental factors. To date, no antifibrotic therapy is available. Some progress has been made in creating consensus definitions and measurements to quantify stricture morphology for clinical practice and trials, but approaches to determine the degree of fibrosis within a stricture are still lacking. OBJECTIVE We herein describe the current state of stricture pathogenesis, measuring tools and clinical trial endpoints development. DESIGN Data presented and discussed in this review derive from the past and recent literature and the authors' own research and experience. RESULTS AND CONCLUSIONS Significant progress has been made in better understanding the pathogenesis of fibrosis, but additional studies and preclinical developments are needed to define specific therapeutic targets.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Pranab K Mukherjee
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - William J Massey
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Yan Wang
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Claudio Fiocchi
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
3
|
Chen Q, Wang Y, Shuai J. Current status and future prospects of stomatology research. J Zhejiang Univ Sci B 2023; 24:853-867. [PMID: 37752088 PMCID: PMC10522564 DOI: 10.1631/jzus.b2200702] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/27/2023] [Indexed: 08/08/2023]
Abstract
Research in stomatology (dental medicine) continues to expand globally and is oriented towards solving clinical issues, focusing on clarifying the clinical relevance and potential mechanisms of oral-systemic connections via clinical epidemiology, oral microecological characterization, and the establishment of animal models. Interdisciplinary integration of materials science and tissue engineering with stomatology is expected to lead to the creation of innovative materials and technologies to better resolve the most prevalent and challenging clinical issues such as peri-implantitis, soft and hard tissue defects, and dentin hypersensitivity. With the rapid development of artificial intelligence (AI), 5th generation mobile communication technology (5G), and big data applications, "intelligent stomatology" is emerging to build models for better clinical diagnosis and management, accelerate the reform of education, and support the growth and advancement of scientific research. Here, we summarized the current research status, and listed the future prospects and limitations of these three aspects, aiming to provide a basis for more accurate etiological exploration, novel treatment methods, and abundant big data analysis in stomatology to promote the translation of research achievements into practical applications for both clinicians and the public.
Collapse
Affiliation(s)
- Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China.
| | - Yahui Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Jing Shuai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou 310006, China
| |
Collapse
|
4
|
Garcia-Hernandez V, Raya-Sandino A, Azcutia V, Miranda J, Kelm M, Flemming S, Birkl D, Quiros M, Brazil JC, Parkos CA, Nusrat A. Inhibition of Soluble Stem Cell Factor Promotes Intestinal Mucosal Repair. Inflamm Bowel Dis 2023; 29:1133-1144. [PMID: 36688460 PMCID: PMC10320368 DOI: 10.1093/ibd/izad003] [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: 11/16/2022] [Indexed: 01/24/2023]
Abstract
BACKGROUND Incidences of inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, are escalating worldwide and can be considered a global public health problem. Given that the gold standard approach to IBD therapeutics focuses on reducing the severity of symptoms, there is an urgent unmet need to develop alternative therapies that halt not only inflammatory processes but also promote mucosal repair. Previous studies have identified increased stem cell factor (SCF) expression in inflamed intestinal mucosal tissues. However, the role that SCF plays in mediating intestinal inflammation and repair has not been explored. METHODS Changes in the expression of SCF were evaluated in the colonic tissue of healthy mice and during dextran sodium sulfate (DSS)-induced colitis. Furthermore, mucosal wound healing and colitis severity were analyzed in mice subjected to either mechanical biopsy or DSS treatment, respectively, following intestinal epithelial cell-specific deletion of SCF or anti-SCF antibody administration. RESULTS We report robust expression of SCF by intestinal epithelial cells during intestinal homeostasis with a switch to immune cell-produced SCF during colitis. Data from mice with intestinal epithelial cell-specific deletion of SCF highlight the importance of immune cell-produced SCF in driving the pathogenesis of colitis. Importantly, antibody-mediated neutralization of total SCF or the specific SCF248 isoform decreased immune cell infiltration and enhanced mucosal wound repair following biopsy-induced colonic injury or DSS-induced colitis. CONCLUSIONS These data demonstrate that SCF functions as a pro-inflammatory mediator in mucosal tissues and that specific neutralization of SCF248 could be a viable therapeutic option to reduce intestinal inflammation and promote mucosal wound repair in individuals with IBD.
Collapse
Affiliation(s)
| | - Arturo Raya-Sandino
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Veronica Azcutia
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jael Miranda
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Matthias Kelm
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sven Flemming
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Dorothee Birkl
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Miguel Quiros
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jennifer C Brazil
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
5
|
Biochanin A in murine Schistosoma mansoni infection: effects on inflammation, oxidative stress and fibrosis. J Helminthol 2023; 97:e16. [PMID: 36740983 DOI: 10.1017/s0022149x22000839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Biochanin A (BCA) is a multifunctional natural compound that possesses anti-infective, anti-inflammatory, anti-oxidative and hepatoprotective effects. The aim of the study was to assess the therapeutic efficacy of BCA on Schistosoma mansoni-infected mice. Fifty mice were divided into six different groups as non-infected, non-infected BCA-treated, infected untreated, early infected BCA-treated (seven days post-infection (dpi)), late infected BCA-treated 60 dpi and infected praziquantel (PZQ)-treated groups. Parasitological, histopathological examination and immunohistochemical staining of transforming growth factor (TGF)-β, inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX-2) were investigated in liver sections. Cytochrome P450 (CYP450) gene expression of S. mansoni was evaluated by quantitative real-time polymerase chain reaction (RT-qPCR). A single dose of BCA significantly reduced worm burden in early (82.14%) and late infection (77.74%), mean tissue egg load in early (7.27 ± 0.495) and late BCA administration (7.63 ± 0.435) and decreased granuloma size. CYP450 mRNA expression was significantly reduced in early BCA treatment as compared to late treatment which emphasizes that early administration of BCA had more pronounced effects on worms than late administration. Both early and late BCA administration led to significant reduction in inflammatory cytokines as TGF and iNOS. Although the reduction of TGF and iNOS in BCA-treated mice was superior to PZQ, no statistically significant differences were noted. However, a significant downregulation of COX2 was noted in hepatocytes as compared to both infected control and PZQ-treated mice. BCA has schistosomicidal, anti-inflammatory, antioxidant and anti-fibrotic effects and could be regarded as a potential drug in schistosomiasis treatment.
Collapse
|
6
|
Hayot G, Massonot M, Keime C, Faure E, Golzio C. Loss of autism-candidate CHD8 perturbs neural crest development and intestinal homeostatic balance. Life Sci Alliance 2023; 6:e202201456. [PMID: 36375841 PMCID: PMC9664244 DOI: 10.26508/lsa.202201456] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Individuals with mutations in CHD8 present with gastrointestinal complaints, yet the underlying mechanisms are understudied. Here, using a stable constitutive chd8 mutant zebrafish model, we found that the loss of chd8 leads to a reduced number of vagal neural crest cells (NCCs), enteric neural and glial progenitors, emigrating from the neural tube, and that their early migration capability was altered. At later stages, although the intestinal colonization by NCCs was complete, we found the decreased numbers of both serotonin-producing enterochromaffin cells and NCC-derived serotonergic neurons, suggesting an intestinal hyposerotonemia in the absence of chd8 Furthermore, transcriptomic analyses revealed an altered expression of key receptors and enzymes in serotonin and acetylcholine signaling pathways. The tissue examination of chd8 mutants revealed a thinner intestinal epithelium accompanied by an accumulation of neutrophils and the decreased numbers of goblet cells and eosinophils. Last, single-cell sequencing of whole intestines showed a global disruption of the immune balance with a perturbed expression of inflammatory interleukins and changes in immune cell clusters. Our findings propose a causal developmental link between chd8, NCC development, intestinal homeostasis, and autism-associated gastrointestinal complaints.
Collapse
Affiliation(s)
- Gaëlle Hayot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Mathieu Massonot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Céline Keime
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Elodie Faure
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Christelle Golzio
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
- Centre National de la Recherche Scientifique, Illkirch, France
- Institut National de la Santé et de la Recherche Médicale, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| |
Collapse
|
7
|
Mizukoshi H, Kimura K, Ikemura H, Mori Y, Nagaoka M. Structural determination of the cell wall polysaccharide LCPS-1 in Lacticaseibacillus paracasei strain Shirota YIT 9029. Carbohydr Res 2022; 521:108670. [PMID: 36103733 DOI: 10.1016/j.carres.2022.108670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/02/2022]
Abstract
The neutral polysaccharides LCPS-1 and LCPS-2 play functional roles in the cell wall of the lactic acid bacterium Lacticaseibacillus paracasei strain Shirota YIT 9029 (LcS; formerly Lactobacillus casei strain Shirota YIT 9029), which has long been used as a probiotic food product. Studies have shown that LCPS-1 is associated with the immunomodulatory functions of LcS. We hypothesized that the structure of LCPS-1 is crucial for elucidating the mechanism of action of LcS on host immune responses and aimed to solve the undetermined primary structure of LCPS-1. Our results showed that LCPS-1 has a molecular weight of >400 kDa and is composed of Glc, Rha, Gal, and GlcNAc, with a repeating structure. Using limited degradation reactions, including controlled Smith and deamination degradations, we obtained key fragments with low molecular weight. Subsequently, their structures were analyzed using NMR spectra and other analytical techniques. Further, we integrated the results for each key fragment to derive the complete structure of LCPS-1. Our results indicated that the most probable structure of LCPS-1 is composed of two types of units (X, Y), each with a basic structure of seven sugars in which the C2-position of Rha is substituted with an acetyl group. The structure of X is {6[Glcβ1-2] Galα1-3[2-OAc] Rhaβ1-4Glcβ1-4[Rhaα1-3] [Glcα1-6] Glcβ1-} and that of Y is {6[Glcβ1-2] Galα1-3[2-OAc] Rhaβ1-4Glcβ1-4[Rhaα1-3] [Glcα1-6)] GlcNAcβ1-}, which can be expressed as (X6Y12)n. In this study, we identified the primary structure of LCPS-1, and our results may enable an improved understanding of the immunomodulatory abilities of LcS.
Collapse
Affiliation(s)
- Harumi Mizukoshi
- Yakult Central Institute for Microbiological Research, 5-11 Izumi Kunitachi-shi, Tokyo, 186-8650, Japan.
| | - Kazumasa Kimura
- Yakult Central Institute for Microbiological Research, 5-11 Izumi Kunitachi-shi, Tokyo, 186-8650, Japan
| | - Haruo Ikemura
- Yakult Central Institute for Microbiological Research, 5-11 Izumi Kunitachi-shi, Tokyo, 186-8650, Japan
| | - Yoko Mori
- Yakult Central Institute for Microbiological Research, 5-11 Izumi Kunitachi-shi, Tokyo, 186-8650, Japan
| | - Masato Nagaoka
- Yakult Central Institute for Microbiological Research, 5-11 Izumi Kunitachi-shi, Tokyo, 186-8650, Japan
| |
Collapse
|
8
|
Nonnecke EB, Castillo PA, Akahoshi DT, Goley SM, Bevins CL, Lönnerdal B. Characterization of an intelectin-1 ( Itln1) knockout mouse model. Front Immunol 2022; 13:894649. [PMID: 36072603 PMCID: PMC9441953 DOI: 10.3389/fimmu.2022.894649] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/28/2022] [Indexed: 01/26/2023] Open
Abstract
Intelectins are carbohydrate-binding proteins implicated in innate immunity and highly conserved across chordate evolution, including both ascidians and humans. Human intelectin-1 (ITLN1) is highly abundant within the intestinal mucosa and binds microbial but not host glycans. Genome-wide association studies identified SNPs in ITLN1 that are linked to susceptibility for Crohn's disease. Moreover, ITLN1 has been implicated in the pathophysiology of obesity and associated metabolic disease. To gain insight on biological activities of human ITLN1 in vivo, we developed a C57BL/6 mouse model genetically targeting the gene encoding the functional mouse ortholog. In wild-type C57BL/6 mice, both mRNA and protein analysis showed high expression of Itln1 in the small intestine, but manifold lower levels in colon and other extraintestinal tissues. Whereas intestinal expression of human ITLN1 localizes to goblet cells, our data confirm that mouse Itln1 is expressed in Paneth cells. Compared to wild-type littermate controls, mice homozygous for the Itln1 hypomorphic trapping allele had reduced expression levels of Itln1 expression (~10,000-fold). The knockout mice exhibited increased susceptibility in an acute model of experimentally induced colitis with 2% w/v dextran sulfate sodium (DSS). In a model of chronic colitis using a lower dose of DSS (1.5% w/v), which enabled a detailed view of disease activity across a protracted period, no differences were observed in body weight, fecal texture, hemoccult scores, food/water intake, or colon length at necropsy, but there was a statistically significant genotype over time effect for the combined fecal scores of disease activity. In model of diet-induced obesity, using two western-style diets, which varied in amounts of sugar (as sucrose) and saturated fat (as lard), mice with Itln1 expression ablated showed no increased susceptibility, in terms of weight gain, food intake, plasma markers of obesity compared to wildtype littermates. While the mouse genetic knockout model for Itln1 holds promise for elucidating physiological function(s) for mammalian intelectins, results reported here suggest that Itln1, a Paneth cell product in C57BL/6 mice, likely plays a minor role in the pathophysiology of chemically induced colitis or diet-induced obesity.
Collapse
Affiliation(s)
- Eric B. Nonnecke
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States,*Correspondence: Eric B. Nonnecke, ; Charles L. Bevins,
| | - Patricia A. Castillo
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Douglas T. Akahoshi
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Stephanie M. Goley
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Charles L. Bevins
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States,*Correspondence: Eric B. Nonnecke, ; Charles L. Bevins,
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| |
Collapse
|
9
|
Activity of isoflavone biochanin A in chronic experimental toxoplasmosis: impact on inflammation. Parasitol Res 2022; 121:2405-2414. [PMID: 35710847 PMCID: PMC9279236 DOI: 10.1007/s00436-022-07571-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/03/2022] [Indexed: 11/07/2022]
Abstract
Toxoplasma gondii is a worldwide prevalent parasite. The infection has been linked to variable inflammatory effects including neuroinflammation. Biochanin A (BCA) is an isoflavone, known for its anti-inflammatory and anti-oxidative properties. In this study, we examined the effect of BCA on the brain and liver inflammatory lesions in a murine model with chronic toxoplasmosis. Mice were divided in to six groups: non-infected control, non-infected BCA-treated, and four infected groups with Toxoplasmagondii Me49-type II cystogenic strain: infected control, BCA (50 mg/kg/day)-treated, combined BCA/cotrimoxazole-treated and cotrimoxazole (370 mg/kg/day) alone-treated. Gene expression of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and inducible nitric oxide synthase (iNOS) was evaluated by quantitative real-time PCR in the brain and liver tissues. In the infected control group, an upregulation of TNF-α and IL-1β mRNA expression levels was found. However, a downregulation of iNOS expression was detected in the brain of infected control mice. In both BCA- and combined-treated groups, the brain and liver tissues showed significantly reduced inflammatory lesions compared to the infected control mice with inhibited TNF-α and IL-1β mRNA levels. The iNOS expression levels in the brain tissues of BCA group were significantly higher than the levels of the infected control group. BCA alone or combined significantly reduced T. gondii cyst count in the brain tissues. In conclusion, the anti-inflammatory activity of BCA was demonstrated in the brain tissues of mice with chronic toxoplasmosis with decreased TNF-α and IL-1β expression levels and increased iNOS expression levels.
Collapse
|
10
|
Watanabe Y, Mizushima T, Okumura R, Fujino S, Ogino T, Miyoshi N, Takahashi H, Uemura M, Matsuda C, Yamamoto H, Takeda K, Doki Y, Eguchi H. Fecal Stream Diversion Changes Intestinal Environment, Modulates Mucosal Barrier, and Attenuates Inflammatory Cells in Crohn's Disease. Dig Dis Sci 2022; 67:2143-2157. [PMID: 34041649 DOI: 10.1007/s10620-021-07060-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/11/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND The intestinal environment plays important roles in mucosal barrier homeostasis and intestinal inflammation, as clarified in studies using experimental animals but not in humans. AIMS We investigated whether environmental changes in the fecal stream cause phenotypic changes in the human mucosal barrier. METHODS We obtained human ileal samples after fecal stream diversions in patients with rectal cancer or Crohn's disease. We investigated the bacterial load and diversity in the human defunctioned ileum, defined as the anal side of the ileum relative to the ileostomy. We also examined the epithelium and lamina propria cell phenotypes in the defunctioned ileum. RESULTS After fecal stream diversion, bacterial loads decreased significantly in the defunctioned ileum. Based on the Chao1, Shannon, and observed species indices, the diversity of mucosa-associated microbiota was lower in the defunctioned ileum than in the functional ileum. Moreover, the healthy defunctioned ileum showed reductions in villous height, goblet cell numbers, and Ki-67+ cell numbers. Additionally, interferon-γ+, interleukin-17+, and immunoglobulin A+ cell abundance in the lamina propria decreased. After the intestinal environment was restored with an ileostomy closure, the impaired ileal homeostasis recovered. The defunctioned ileum samples from patients with Crohn's disease also showed reductions in interferon-γ+ and interleukin-17+ cell numbers. CONCLUSIONS Fecal stream diversion reduced the abundance and diversity of intestinal bacteria. It also altered the intestinal mucosal barrier, similar to the alterations observed in germ-free animals. In patients with Crohn's disease, Th1 and Th17 cell numbers were attenuated, which suggests that the host-microbiome interaction is important in disease pathogenesis.
Collapse
Affiliation(s)
- Yoshifumi Watanabe
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tsunekazu Mizushima
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Department of Therapeutics for Inflammatory Bowel Diseases, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan.
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 1-1 Yamadaoka, Suita, Osaka, Japan.
| | - Ryu Okumura
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
- Immunology Frontier Research Center, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Shiki Fujino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takayuki Ogino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Norikatsu Miyoshi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hidekazu Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mamoru Uemura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Chu Matsuda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hirofumi Yamamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kiyoshi Takeda
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 1-1 Yamadaoka, Suita, Osaka, Japan
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
- Immunology Frontier Research Center, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
11
|
Watanabe D, Kamada N. Contribution of the Gut Microbiota to Intestinal Fibrosis in Crohn's Disease. Front Med (Lausanne) 2022; 9:826240. [PMID: 35198577 PMCID: PMC8859331 DOI: 10.3389/fmed.2022.826240] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 12/16/2022] Open
Abstract
In Crohn's disease (CD), intestinal fibrosis is a critical determinant of a patient's prognosis. Although inflammation may be a prerequisite for the initiation of intestinal fibrosis, research shows that the progression or continuation of intestinal fibrosis can occur independently of inflammation. Thus, once initiated, intestinal fibrosis may persist even if medical treatment controls inflammation. Clearly, an understanding of the pathophysiological mechanisms of intestinal fibrosis is required to diminish its occurrence. Accumulating evidence suggests that the gut microbiota contributes to the pathogenesis of intestinal fibrosis. For example, the presence of antibodies against gut microbes can predict which CD patients will have intestinal complications. In addition, microbial ligands can activate intestinal fibroblasts, thereby inducing the production of extracellular matrix. Moreover, in various animal models, bacterial infection can lead to the development of intestinal fibrosis. In this review, we summarize the current knowledge of the link between intestinal fibrosis in CD and the gut microbiota. We highlight basic science and clinical evidence that the gut microbiota can be causative for intestinal fibrosis in CD and provide valuable information about the animal models used to investigate intestinal fibrosis.
Collapse
Affiliation(s)
- Daisuke Watanabe
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| |
Collapse
|
12
|
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
|
13
|
An Update of Research Animal Models of Inflammatory Bowel Disease. ScientificWorldJournal 2021; 2021:7479540. [PMID: 34938152 PMCID: PMC8687830 DOI: 10.1155/2021/7479540] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 11/28/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a group of chronic disorders that includes two main disease forms, Crohn's disease, and ulcerative colitis. The understanding of the intestinal inflammation occurring in IBD has been immeasurably advanced by the development of the now numerous murine models of intestinal inflammation. The usefulness of this research tool in IBD arises from a convergence of underlying genetic susceptibility, immune system dysfunction, environmental factors, and shifts in gut microbiota. Due to the multifactorial feature of these diseases, different animal models have been used to investigate the underlying mechanisms and develop potential therapeutic strategies. The results of preclinical efficacy studies often inform the progression of therapeutic strategies. This review describes the distinct feature and limitations of each murine IBD model and discusses the previous and current lessons from the IBD models.
Collapse
|
14
|
Suarez-Lopez L, Shui B, Brubaker DK, Hill M, Bergendorf A, Changelian PS, Laguna A, Starchenko A, Lauffenburger DA, Haigis KM. Cross-species transcriptomic signatures predict response to MK2 inhibition in mouse models of chronic inflammation. iScience 2021; 24:103406. [PMID: 34849469 PMCID: PMC8609096 DOI: 10.1016/j.isci.2021.103406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/28/2021] [Accepted: 11/03/2021] [Indexed: 11/30/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) are genetically complex and exhibit significant inter-patient heterogeneity in disease presentation and therapeutic response. Here, we show that mouse models of IBD exhibit variable responses to inhibition of MK2, a pro-inflammatory serine/threonine kinase, and that MK2 inhibition suppresses inflammation by targeting inflammatory monocytes and neutrophils in murine models. Using a computational approach (TransComp-R) that allows for cross-species comparison of transcriptomic features, we identified an IBD patient subgroup that is predicted to respond to MK2 inhibition, and an independent preclinical model of chronic intestinal inflammation predicted to be non-responsive, which we validated experimentally. Thus, cross-species mouse-human translation approaches can help to identify patient subpopulations in which to deploy new therapies. MK2 kinase inhibition shows variable efficacy in different IBD mouse models TCT and TNFΔARE mice express distinct inflammatory and MK2-responsive genes “Response to MK2i” signature is enriched in monocytes and neutrophils Cross-species modeling identifies patient groups potentially responsive to MK2i
Collapse
Affiliation(s)
- Lucia Suarez-Lopez
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Bing Shui
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Douglas K. Brubaker
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47906, USA
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, IN 47906, USA
| | - Marza Hill
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Alexander Bergendorf
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47906, USA
| | - Paul S. Changelian
- Aclaris Therapeutics, Inc., 4320 Forest Park Avenue, St. Louis, MO 63108, USA
| | - Aisha Laguna
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Alina Starchenko
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kevin M. Haigis
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
- Corresponding author
| |
Collapse
|
15
|
Bandyopadhyay C, Schecterson L, Gumbiner BM. E-cadherin activating antibodies limit barrier dysfunction and inflammation in mouse inflammatory bowel disease. Tissue Barriers 2021; 9:1940741. [PMID: 34402758 PMCID: PMC8794503 DOI: 10.1080/21688370.2021.1940741] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 01/21/2023] Open
Abstract
Deficits in gastrointestinal (GI) paracellular permeability has been implicated in etiology of Inflammatory Bowel Disease (IBD), and E-cadherin, a key component of the epithelial junctional complex, has been implicated in both barrier function and IBD. We have previously described antibodies against E-cadherin that activate cell adhesion, and in this study, we show that they increase transepithelial electrical resistance in epithelial cell monolayers in vitro. We therefore tested the hypothesis that adhesion activating E-cadherin mAbs will enhance epithelial barrier function in vivo and limit progression of inflammation in IBD. Activating mAbs to mouse E-cadherin were tested in different mouse models of IBD including the IL10-/- and adoptive T cell transfer models of colitis. Previously established histological and biomarker measures of inflammation were evaluated to monitor disease progression. Mouse E-cadherin activating mAb treatment reduced total colitis score, individual histological measures of inflammation, and other hallmarks of inflammation compared to control treatment. Activating mAbs also reduced the fecal accumulation lipocalin2 and albumin content, consistent with enhanced barrier function. Therefore, E-cadherin activation could be a potential strategy for limiting inflammation in UC.
Collapse
Affiliation(s)
- Chirosree Bandyopadhyay
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States
| | - Leslayann Schecterson
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States
| | - Barry M Gumbiner
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States
- Department of Pediatrics, University of Washington, Seattle, United States
- Department of Biochemistry, University of Washington, Seattle, United States
| |
Collapse
|
16
|
Abstract
Mouse models are essential for investigation of underlying disease mechanisms that drive intestinal fibrosis, as well as assessment of potential therapeutic approaches to either prevent or resolve fibrosis. Here we describe several common mouse models of intestinal inflammation and fibrosis, including chemically driven colitis models, a bacterially triggered colitis model, and spontaneous intestinal inflammation in genetically susceptible mouse strains. Detailed protocols are provided for dextran sodium sulfate (DSS) colitis, 2,4,6-trinitro-benzene sulfonic acid (TNBS) colitis, adherent-invasive Escherichia coli (AIEC)-triggered colitis, the interleukin-10 knockout (IL-10KO) mouse model of spontaneous colitis, and the SAMP/YitFc model of spontaneous ileocolitis.
Collapse
|
17
|
Hansen AK, Hansen CHF. The microbiome and rodent models of immune mediated diseases. Mamm Genome 2021; 32:251-262. [PMID: 33792799 PMCID: PMC8012743 DOI: 10.1007/s00335-021-09866-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/12/2021] [Indexed: 02/07/2023]
Abstract
Over the last six decades production of laboratory rodents have been refined with the aim of eliminating all pathogens, which could influence research results. This has, however, also created rodents with little diversity in their microbiota. Until 10 years ago the impact of the microbiota on the outcome of rodent studies was ignored, but today it is clear that the phenotype of rodent models differs essentially in relation to the environment of origin, i.e. different breeders or different rooms. In this review, we outline the mechanisms behind gut bacterial impact on rodent models of immune mediated diseases, and how differences in environment of origin leads to phenotypic model differences within research areas such as infectious diseases and vaccine development, the metabolic syndrome, gut immunity and inflammation, autoimmunity and allergy. Finally, we sum up some tools to handle this impact to increase reproducibility and translatability of rodent models.
Collapse
Affiliation(s)
- Axel Kornerup Hansen
- Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark.
| | - Camilla Hartmann Friis Hansen
- Section of Experimental Animal Models, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Ridebanevej 9, 1870, Frederiksberg C, Denmark.
| |
Collapse
|
18
|
Lu S, Zhu K, Guo Y, Wang E, Huang J. Evaluation of animal models of Crohn's disease with anal fistula (Review). Exp Ther Med 2021; 22:974. [PMID: 34335916 PMCID: PMC8290422 DOI: 10.3892/etm.2021.10406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/09/2021] [Indexed: 12/28/2022] Open
Abstract
Anal fistula is a common and serious complication of Crohn's disease (CD). A sufficiently suitable animal model that may be used to simulate this disease is yet to be established. The aim of the present review was to summarize the different characteristics and experimental methods of commonly used animal models of CD with anal fistula. Electronic databases were searched for studies reporting on the use of this type of animal model. A total of 234 related articles were retrieved, of which six articles met the inclusion criteria; these were used as references for the present review article. The characteristics of the animal models, the advantages and disadvantages of the modeling methods and the similarities with patients with CD and anal fistula were summarized and analyzed. The evidence suggests that a sufficiently suitable animal preclinical model requires to be established.
Collapse
Affiliation(s)
- Shuangshuang Lu
- Department of Internal Medicine, School of Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Gastrointestinal Center, Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Keyuan Zhu
- Department of Internal Medicine, School of Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Gastrointestinal Center, Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Yongxin Guo
- Department of Internal Medicine, School of Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Gastrointestinal Center, Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Enjing Wang
- Gastrointestinal Center, Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China.,Department of Internal Medicine, School of Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Jin Huang
- Department of Internal Medicine, School of Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China.,Gastrointestinal Center, Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
19
|
Lee C, Hong SN, Kim ER, Chang DK, Kim YH. Depletion of Intestinal Stem Cell Niche Factors Contributes to the Alteration of Epithelial Differentiation in SAMP1/YitFcsJ Mice With Crohn Disease-Like Ileitis. Inflamm Bowel Dis 2021; 27:667-676. [PMID: 33274375 DOI: 10.1093/ibd/izaa314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND SAMP1/YitFcsJ (SAMP1) mice spontaneously develop terminal ileitis resembling human Crohn disease. SAMP1 mice have exhibited alteration of epithelial cell lineage distribution and an overall proliferation of the crypt cell population; however, it has not been evaluated whether epithelial differentiation is impaired because of dysfunction of intestinal stem cells (ISCs) or their niche factors. METHODS Using the intestine of SAMP1 mice aged 10 to 14 weeks, morphometric alterations in the crypt-villus architecture, ISCs, crypt cells, and differentiated cells; organoid formation capacity of intestinal crypts; and niche signaling pathways were analyzed and compared with those of age-matched control AKR/J (AKR) mice. RESULTS The ileum of SAMP1 mice showed increased depth of intestinal crypts and decreased surface area of the villi compared with those in the ileum of AKR mice. The number of ISCs in the ileal crypts did not differ between SAMP1 and AKR mice; however, the number of Paneth cells decreased and the number of transient amplifying cells increased. The organoid formation rate of the ileal crypts of SAMP1 mice decreased significantly compared with that of AKR mice. The performance of RNA sequencing for intestinal crypts found that the expression of ISC niche factors, such as Wnt3, Dll1, and Dll4, was decreased significantly in the ileal crypts of SAMP1 mice compared with those of AKR mice. Among the ISC niche signals, the Notch signaling-related genes tended to be downregulated. In particular, immunocytochemistry revealed that the expression of Paneth cell-expressing Notch ligand Dll4 was significantly decreased in the intestinal tissue and organoids of SAMP1 mice compared with those of AKR mice. CONCLUSIONS Depletion of niche factors for ISCs contributes to the alteration of epithelial differentiation in SAMP1 mice.
Collapse
Affiliation(s)
- Chansu Lee
- Samsung Biomedical Research Institute, Samsung Medical Center, Seoul, Korea.,Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Noh Hong
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun Ran Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong Kyung Chang
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Ho Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
20
|
DeHaan RK, Sarvestani SK, Huang EH. Organoid Models of Colorectal Pathology: Do They Hold the Key to Personalized Medicine? A Systematic Review. Dis Colon Rectum 2020; 63:1559-1569. [PMID: 32868555 PMCID: PMC7547902 DOI: 10.1097/dcr.0000000000001806] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Colorectal cancer and IBD account for a large portion of the practice of colorectal surgery. Historical research models have provided insights into the underlying causes of these diseases but come with many limitations. OBJECTIVE The aim of this study was to systematically review the literature regarding the advantage of organoid models in modeling benign and malignant colorectal pathology. DATA SOURCES Sources included PubMed, Ovid-Medline, and Ovid Embase STUDY SELECTION:: Two reviewers completed a systematic review of the literature between January 2006 and January of 2020 for studies related to colon and intestinal organoids. Reviews, commentaries, protocols, and studies not performed in humans or mice were excluded. RESULTS A total of 73 articles were included. Organoid models of colorectal disease have been rising in popularity to further elucidate the genetic, transcriptomic, and treatment response of these diseases at the individual level. Increasingly complex models utilizing coculture techniques are being rapidly developed that allow in vitro recapitulation of the disease microenvironment. LIMITATIONS This review is only qualitative, and the lack of well utilized nomenclature in the organoid community may have resulted in the exclusion of articles. CONCLUSIONS Historical disease models including cell lines, patient-derived tumor xenografts, and animal models have created a strong foundation for our understanding of colorectal pathology. Recent advances in 3-dimensional cell cultures, in the form of patient-derived epithelial organoids and induced human intestinal organoids have opened a new avenue for high-resolution analysis of pathology at the level of an individual patient. Recent research has shown the potential of organoids as a tool for personalized medicine with their ability to retain patient characteristics, including treatment response.
Collapse
|
21
|
Mirzaei F, Meshkini A, Habibi B, Salehpour F, Rafei E, Fathi W, Alavi SHN, Majdi A, Rahigh Aghasan S, Naseri Alavi SA. Ceftriaxone Plus Methylprednisolone Combination Therapy Versus Methylprednisolone Monotherapy in Patients With Acute Spinal Cord Injury: A Randomized, Triple-Blind Clinical Trial. Int J Spine Surg 2020; 14:706-712. [PMID: 33077437 PMCID: PMC7671452 DOI: 10.14444/7102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Guidelines do not suggest the routine use of methylprednisolone (MP) in patients with acute traumatic spinal cord injury (SCI). We tested the hypothesis regarding whether combination therapy with ceftriaxone and MP is superior to MP monotherapy in patients with acute traumatic SCI. METHODS In a randomized, triple-blind clinical trial, 60 patients with acute (first 8 hours of the injury) traumatic SCI were enrolled at the Tabriz University of Medical Sciences, Tabriz, Iran, between December 2016 and June 2017. Accordingly, the patients were randomly divided into 2 case and control groups (n = 30 each). Upon admission, all included patients received a bolus dose of MP at 33 mg/kg intravenously (IV) for 15 minutes. Then, after 45 minutes, MP infusion was continued for 24 to 48 hours at a 5.4 mg/kg IV dose. The case group received an additional dose of ceftriaxone at 1 g 2 times a day for 7 days through an IV route. Erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) were checked and compared between case and control groups upon admission and on the fourth and eighth days. Also, sensory and motor functions were evaluated according to the American Spinal Injury Association (ASIA) grading score upon admission, on the third and seventh days, upon discharge and 6 months after admission. RESULTS Analyses showed a significant statistical difference between groups in the changes in CRP levels during days 1 and 4 (P = .001) and also during days 4 and 8 (P = .001). However, no significant statistical difference was detected in ESR levels changes between groups during days 1 and 4 (P = .073), and during days 4 and 8 (P = .069). ASIA scale was found to be significantly different between the MP plus ceftriaxone group and MP monotherapy upon admission and 6 months after treatment (P = .001 for both comparisons). However, the number of variations in the ASIA score had no significant statistical difference between groups 6 months after intervention (P = .465). CONCLUSION The addition of ceftriaxone to the routine therapeutic protocol of acute SCI is accompanied by improved inflammation markers and functional outcomes 6 months after the intervention.
Collapse
Affiliation(s)
- Farhad Mirzaei
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Meshkini
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bohlool Habibi
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Firooz Salehpour
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ebrahim Rafei
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Wouria Fathi
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Alireza Majdi
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Rahigh Aghasan
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Ahmad Naseri Alavi
- Department of Neurosurgery, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Relationship between T cells and microbiota in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 171:95-129. [PMID: 32475529 DOI: 10.1016/bs.pmbts.2020.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the past decades, the fields of microbiology and immunology have largely advanced by using germ-free animals and next-generation sequencing. Many studies revealed the relationship among gut microbiota, activation of immune system, and various diseases. Especially, some gut commensals can generate their antigen-specific T cells. It is becoming clear that commensal bacteria have important roles in various autoimmune and inflammatory diseases, such as autism, rheumatoid arthritis (RA), and inflammatory bowel diseases (IBD). Recently, it was reported that commensals contribute to the cancer immune therapy. However, how commensal-specific T cells contribute to the disease development and cancer treatment are not fully understood yet. In this chapter, we will summarize the decade history of the studies associated with commensal-induced T cells and commensal-causing diseases.
Collapse
|
23
|
Abstract
Inflammatory bowel diseases (IBD), including Crohn's disease, ulcerative colitis, and pouchitis, are chronic, relapsing intestinal inflammatory disorders mediated by dysregulated immune responses to resident microbiota. Current standard therapies that block immune activation with oral immunosuppressives or biologic agents are generally effective, but each therapy induces a sustained remission in only a minority of patients. Furthermore, these approaches can have severe adverse events. Recent compelling evidence of a role of unbalanced microbiota (dysbiosis) driving immune dysfunction and inflammation in IBD supports the therapeutic rationale for manipulating the dysbiotic microbiota. Traditional approaches using currently available antibiotics, probiotics, prebiotics, and synbiotics have not produced optimal results, but promising outcomes with fecal microbiota transplant provide a proof of principle for targeting the resident microbiota. Rationally designed oral biotherapeutic products (LBPs) composed of mixtures of protective commensal bacterial strains demonstrate impressive preclinical results. Resident microbial-based and microbial-targeted therapies are currently being studied with increasing intensity for IBD primary therapy with favorable early results. This review presents current evidence and therapeutic mechanisms of microbiota modulation, emphasizing clinical studies, and outlines prospects for future IBD treatment using new approaches, such as LBPs, bacteriophages, bacterial function-editing substrates, and engineered bacteria. We believe that the optimal clinical use of microbial manipulation may be as adjuvants to immunosuppressive for accelerated and improved induction of deep remission and as potential safer solo approaches to sustained remission using personalized regimens based on an individual patient's microbial profile.
Collapse
Affiliation(s)
- Akihiko Oka
- Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC, 27599, USA
| | - R Balfour Sartor
- Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC, 27599, USA.
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, 125 Mason Farm Road, Chapel Hill, NC, 27599, USA.
- National Gnotobiotic Rodent Resource Center, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC, 27514, USA.
| |
Collapse
|
24
|
Newman KM, Vaughn BP. Efficacy of intestinal microbiota transplantation in ulcerative colitis: a review of current literature and knowledge. MINERVA GASTROENTERO 2020; 65. [DOI: 10.23736/s1121-421x.19.02610-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
25
|
Lovisa S, Genovese G, Danese S. Role of Epithelial-to-Mesenchymal Transition in Inflammatory Bowel Disease. J Crohns Colitis 2019; 13:659-668. [PMID: 30520951 DOI: 10.1093/ecco-jcc/jjy201] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intestinal fibrosis is an inevitable complication in patients with inflammatory bowel disease [IBD], occurring in its two major clinical manifestations: ulcerative colitis and Crohn's disease. Fibrosis represents the final outcome of the host reaction to persistent inflammation, which triggers a prolonged wound healing response resulting in the excessive deposition of extracellular matrix, eventually leading to intestinal dysfunction. The process of epithelial-to-mesenchymal transition [EMT] represents an embryonic program relaunched during wound healing, fibrosis and cancer. Here we discuss the initial observations and the most recent findings highlighting the role of EMT in IBD-associated intestinal fibrosis and fistulae formation. In addition, we briefly review knowledge on the cognate process of endothelial-to-mesenchymal transition [EndMT]. Understanding EMT functionality and the molecular mechanisms underlying the activation of this mesenchymal programme will permit designing new therapeutic strategies to halt the fibrogenic response in the intestine.
Collapse
Affiliation(s)
- Sara Lovisa
- Department of Cancer Biology, Metastasis Research Center, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Giannicola Genovese
- Department of Genomic Medicine, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA.,Department of Genitourinary Medical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Silvio Danese
- IBD Center, Department of Gastroenterology, Humanitas Research Hospital, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
| |
Collapse
|
26
|
Intestinal Organoids as a Novel Complementary Model to Dissect Inflammatory Bowel Disease. Stem Cells Int 2019; 2019:8010645. [PMID: 31015842 PMCID: PMC6444246 DOI: 10.1155/2019/8010645] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/04/2019] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) include colitis ulcerosa and Crohn's disease, besides the rare microscopic colitis. Both diseases show a long-lasting, relapsing-remitting, or even chronic active course with tremendous impact on quality of life. IBDs frequently cause disability, surgical interventions, and high costs; as in other autoimmune diseases, their prevalent occurrence at an early phase of life raises the burden on health care systems. Unfortunately, our understanding of the pathogenesis is still incomplete and treatment therefore largely focuses on suppressing the resulting excessive inflammation. One obstacle for deciphering the causative processes is the scarcity of models that parallel the development of the disease, since intestinal inflammation is mostly induced artificially; moreover, the intestinal epithelium, which strongly contributes to IBD pathogenesis, is difficult to assess. Recently, the development of intestinal epithelial organoids has overcome many of those problems. Here, we give an overview on the current understanding of the pathogenesis of IBDs with reference to the limitations of previous well-established experimental models. We highlight the advantages and detriments of recent organoid-based experimental setups within the IBD field and suggest possible future applications.
Collapse
|
27
|
Mikami Y, Takada Y, Hagihara Y, Kanai T. Innate lymphoid cells in organ fibrosis. Cytokine Growth Factor Rev 2018; 42:27-36. [PMID: 30104153 DOI: 10.1016/j.cytogfr.2018.07.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 02/06/2023]
Abstract
Innate lymphoid cells (ILCs) are a recently identified family of lymphoid effector cells. ILCs are mainly clustered into 3 groups based on their unique cytokine profiles and transcription factors typically attributed to the subsets of T helper cells. ILCs have a critical role in the mucosal immune response through promptly responding to pathogens and producing large amount of effector cytokines of type 1, 2, or 3 responses. In addition to the role of early immune responses against infections, ILCs, particularly group 2 ILCs (ILC2), have recently gained attention for modulating remodeling and fibrosis especially in the mucosal tissues. Herein, we overview the current knowledge in this area, highlighting roles of ILCs on fibrosis in the mucosal tissues, especially focusing on the gut and lung. We also discuss some new directions for future research by extrapolating from knowledge derived from studies on Th cells.
Collapse
Affiliation(s)
- Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan.
| | - Yoshiaki Takada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Yuya Hagihara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan
| |
Collapse
|
28
|
Jeschke JC, Mayne CG, Ziegelbauer J, DeCiantis CL, Singh S, Kumar SN, Suchi M, Iwakura Y, Drobyski WR, Salzman NH, Williams CB. A model of TH17-associated ileal hyperplasia that requires both IL-17A and IFNγ to generate self-tolerance and prevent colitis. Mucosal Immunol 2018; 11:1127-1137. [PMID: 29728642 PMCID: PMC6571016 DOI: 10.1038/s41385-018-0023-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 03/03/2018] [Accepted: 03/09/2018] [Indexed: 02/04/2023]
Abstract
Homeostasis in the ileum, which is commonly disrupted in patients with Crohn's disease, involves ongoing immune responses. To study how homeostatic processes of the ileum impact CD4+T cell responses, we used TCR transgenic tools to breed mice that spontaneously produced CD4+T cells reactive to an antigen expressed in the ileum. At an early age, the ilea of these mice exhibit crypt hyperplasia and accumulate increased numbers of TH17 cells bearing non-transgenic clonotypes. Half of these mice subsequently developed colitis linked to broad mucosal infiltration by TH17 and TH1 cells expressing non-transgenic clonotypes, chronic wasting disease and loss of ileal crypt hyperplasia. By contrast, adult mice with normal growth continued to exhibit TH17-associated ileal crypt hyperplasia and additionally accumulated ileal-reactive Treg cells. Both IL-17A and IFNγ were protective, as their deficiency precluded ileal-reactive Treg accumulation and exacerbated colitic disease. IL-23R blockade prevented progression to colitis, whereas nTreg cell transfers prevented colitic disease, ileal crypt hyperplasia and ileal-reactive Treg accumulation. Thus, our studies identify an IL-17A and IFNγ-dependent homeostatic process that mobilizes ileal-reactive Treg cells and is disrupted by IL-23.
Collapse
Affiliation(s)
- Jonathan C. Jeschke
- Section of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee WI, 53226
| | - Christopher G. Mayne
- Section of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee WI, 53226
| | - Jennifer Ziegelbauer
- Section of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee WI, 53226
| | - Christopher L. DeCiantis
- Section of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee WI, 53226
| | - Selina Singh
- Section of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee WI, 53226
| | - Suresh N. Kumar
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee WI, 53226
| | - Mariko Suchi
- Division of Pediatric Pathology, Department of Pathology, Medical College of Wisconsin, Milwaukee WI, 53226
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan; Core Research for Evolutionary Science and Technology, Japan Science and Technology Agency, Saitama 332-0012, Japan
| | - William R. Drobyski
- Bone Marrow Transplant Program, Department of Medicine, Medical College of Wisconsin, Milwaukee WI, 53226
| | - Nita H Salzman
- Section of Gastroenterology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee WI, 53226
| | - Calvin B. Williams
- Section of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, Milwaukee WI, 53226,Correspondence should be addressed to C.B.W. (), Calvin B. Williams, M.D., Ph.D., Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, MFRC Room 5052, Milwaukee WI 53226, Phone: 414-456-4343, Fax: 414-266-6695
| |
Collapse
|
29
|
Chen J, Fan J, Wang S, Sun Z. Secreted Klotho Attenuates Inflammation-Associated Aortic Valve Fibrosis in Senescence-Accelerated Mice P1. Hypertension 2018; 71:877-885. [PMID: 29581213 DOI: 10.1161/hypertensionaha.117.10560] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/09/2017] [Accepted: 03/01/2018] [Indexed: 01/04/2023]
Abstract
Senescence-accelerated mice P1 (SAMP1) is an aging model characterized by shortened lifespan and early signs of senescence. Klotho is an aging-suppressor gene. The purpose of this study is to investigate whether in vivo expression of secreted klotho (Skl) gene attenuates aortic valve fibrosis in SAMP1 mice. SAMP1 mice and age-matched (AKR/J) control mice were used. SAMP1 mice developed obvious fibrosis in aortic valves, namely fibrotic aortic valve disease. Serum level of Skl was decreased drastically in SAMP1 mice. Expression of MCP-1 (monocyte chemoattractant protein 1), ICAM-1 (intercellular adhesion molecule 1), F4/80, and CD68 was increased in aortic valves of SAMP1 mice, indicating inflammation. An increase in expression of α-smooth muscle actin (myofibroblast marker), transforming growth factorβ-1, and scleraxis (a transcription factor of collagen synthesis) was also found in aortic valves of SAMP1 mice, suggesting that accelerated aging is associated with myofibroblast transition and collagen gene activation. We constructed adeno-associated virus 2 carrying mouse Skl cDNA for in vivo expression of Skl. Skl gene delivery effectively increased serum Skl of SAMP1 mice to the control level. Skl gene delivery inhibited inflammation and myofibroblastic transition in aortic valves and attenuated fibrotic aortic valve disease in SAMP1 mice. It is concluded that senescence-related fibrotic aortic valve disease in SAMP1 mice is associated with a decrease in serum klotho leading to inflammation, including macrophage infiltration and transforming growth factorβ-1/scleraxis-driven myofibroblast differentiation in aortic valves. Restoration of serum Skl levels by adeno-associated virus 2 carrying mouse Skl cDNA effectively suppresses inflammation and myofibroblastic transition and attenuates aortic valve fibrosis. Skl may be a potential therapeutic target for fibrotic aortic valve disease.
Collapse
Affiliation(s)
- Jianglei Chen
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Jun Fan
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Shirley Wang
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City
| | - Zhongjie Sun
- From the Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City.
| |
Collapse
|
30
|
Worthington JJ, Reimann F, Gribble FM. Enteroendocrine cells-sensory sentinels of the intestinal environment and orchestrators of mucosal immunity. Mucosal Immunol 2018; 11:3-20. [PMID: 28853441 DOI: 10.1038/mi.2017.73] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 07/14/2017] [Indexed: 02/06/2023]
Abstract
The intestinal epithelium must balance efficient absorption of nutrients with partitioning commensals and pathogens from the bodies' largest immune system. If this crucial barrier fails, inappropriate immune responses can result in inflammatory bowel disease or chronic infection. Enteroendocrine cells represent 1% of this epithelium and have classically been studied for their detection of nutrients and release of peptide hormones to mediate digestion. Intriguingly, enteroendocrine cells are the key sensors of microbial metabolites, can release cytokines in response to pathogen associated molecules and peptide hormone receptors are expressed on numerous intestinal immune cells; thus enteroendocrine cells are uniquely equipped to be crucial and novel orchestrators of intestinal inflammation. In this review, we introduce enteroendocrine chemosensory roles, summarize studies correlating enteroendocrine perturbations with intestinal inflammation and describe the mechanistic interactions by which enteroendocrine and mucosal immune cells interact during disease; highlighting this immunoendocrine axis as a key aspect of innate immunity.
Collapse
Affiliation(s)
- J J Worthington
- Lancaster University, Faculty of Health and Medicine, Division of Biomedical and Life Sciences, Lancaster, Lancashire, UK
| | - F Reimann
- University of Cambridge, Metabolic Research Laboratories, Wellcome Trust/MRC Institute of Metabolic Science & MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Cambridge, UK
| | - F M Gribble
- University of Cambridge, Metabolic Research Laboratories, Wellcome Trust/MRC Institute of Metabolic Science & MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
31
|
Bailey J, Vince V, Williams N, Cogan T. Streptococcus thermophilus NCIMB 41856 ameliorates signs of colitis in an animal model of inflammatory bowel disease. Benef Microbes 2017; 8:605-614. [DOI: 10.3920/bm2016.0110] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Treatment of inflammatory bowel disease (IBD) is mainly based on suppression of symptoms, often with numerous side effects. Trials of probiotics in IBD have frequently produced disappointing results. The majority of probiotics are unusual, since they do not require iron for growth, unlike many bacteria resident in the intestine. The IBD intestine is iron-rich due to bleeding and use of oral iron supplements; conventional probiotics would be rapidly outcompeted. We have evaluated an iron-responsive Streptococcus thermophilus strain for its potential to reduce signs of colitis. Efficacy of S. thermophilus was evaluated in the dextran sodium sulphate mouse model of colitis. Treated animals were given 1×108 cfu S. thermophilus per day and clinical observations were taken daily. At termination, gross and histopathological signs of disease, cellular infiltration, location of bacteria, and cytokine expression in the intestine were determined. S. thermophilus delayed onset of colitis and reduced clinical signs of disease, including bodyweight loss and gastrointestinal bleeding. It reduced bacterial translocation into the colonic tissue. Increased numbers of CD8+ intraepithelial lymphocytes were seen in control animals treated with S. thermophilus. S. thermophilus had no effect on gross pathology, histopathology or cytokine production in either colitic or control animals. We propose that S. thermophilus promotes maintenance of mucosal barrier function which reduces bacterial translocation, thereby reducing immune stimulation and associated inflammation. This allows mucosal healing, reducing gastrointestinal bleeding and weight loss. This could be studied as a locally-acting adjunct or alternative to current IBD treatments.
Collapse
Affiliation(s)
- J.R. Bailey
- Mucosal Microbiology, School of Veterinary Sciences, University of Bristol, BS40 5DU, United Kingdom
| | - V. Vince
- KWS BioTest Ltd., Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, United Kingdom
| | - N.A. Williams
- KWS BioTest Ltd., Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, United Kingdom
| | - T.A. Cogan
- Mucosal Microbiology, School of Veterinary Sciences, University of Bristol, BS40 5DU, United Kingdom
| |
Collapse
|
32
|
Celiberto LS, Bedani R, Rossi EA, Cavallini DCU. Probiotics: The scientific evidence in the context of inflammatory bowel disease. Crit Rev Food Sci Nutr 2017; 57:1759-1768. [PMID: 25996176 DOI: 10.1080/10408398.2014.941457] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Inflammatory bowel disease (IBD) generally comprises Crohn's disease (CD) and ulcerative colitis (UC), and their main characteristic is the intestinal mucosa inflammation. Although its origin is not yet fully known, there is growing evidence related to genetics, intestinal microbiota composition, and the immune system factors such as precursors for the initiation and progression of intestinal conditions. The use of certain probiotic microorganisms has been touted as a possible and promising therapeutic approach in reducing the risk of inflammatory bowel disease, specifically ulcerative colitis. Several mechanisms have been proposed to explain the benefits of probiotics, indicating that some bacterial strains are able to positively modulate the intestinal microbiota and the immune system, and to produce metabolites with anti-inflammatory properties. The aim of this paper is to bring together the various results and information, based on scientific evidence, that are related to probiotics and inflammatory bowel disease, emphasizing the possible mechanisms involved in this action.
Collapse
Affiliation(s)
- Larissa Sbaglia Celiberto
- a Department of Food & Nutrition , Faculty of Pharmaceutical Sciences, São Paulo State University (UNESP) , Araraquara , SP , Brazil
| | - Raquel Bedani
- b Departament of Biochemical and Pharmaceutical Technology , Faculty of Pharmaceutical Sciences, University of São Paulo (USP) Properties , SP , Brazil
| | - Elizeu Antonio Rossi
- a Department of Food & Nutrition , Faculty of Pharmaceutical Sciences, São Paulo State University (UNESP) , Araraquara , SP , Brazil
| | | |
Collapse
|
33
|
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: 50] [Impact Index Per Article: 6.3] [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
|
34
|
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
|
35
|
Fiebiger U, Bereswill S, Heimesaat MM. Dissecting the Interplay Between Intestinal Microbiota and Host Immunity in Health and Disease: Lessons Learned from Germfree and Gnotobiotic Animal Models. Eur J Microbiol Immunol (Bp) 2016; 6:253-271. [PMID: 27980855 PMCID: PMC5146645 DOI: 10.1556/1886.2016.00036] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/21/2016] [Indexed: 02/06/2023] Open
Abstract
This review elaborates the development of germfree and gnotobiotic animal models and their application in the scientific field to unravel mechanisms underlying host-microbe interactions and distinct diseases. Strictly germfree animals are raised in isolators and not colonized by any organism at all. The germfree state is continuously maintained by birth, raising, housing and breeding under strict sterile conditions. However, isolator raised germfree mice are exposed to a stressful environment and exert an underdeveloped immune system. To circumvent these physiological disadvantages depletion of the bacterial microbiota in conventionally raised and housed mice by antibiotic treatment has become an alternative approach. While fungi and parasites are not affected by antibiosis, the bacterial microbiota in these "secondary abiotic mice" have been shown to be virtually eradicated. Recolonization of isolator raised germfree animals or secondary abiotic mice results in a gnotobiotic state. Both, germfree and gnotobiotic mice have been successfully used to investigate biological functions of the conventional microbiota in health and disease. Particularly for the development of novel clinical applications germfree mice are widely used tools, as summarized in this review further focusing on the modulation of bacterial microbiota in laboratory mice to better mimic conditions in the human host.
Collapse
Affiliation(s)
| | | | - Markus M. Heimesaat
- Gastrointestinal Microbiology Research Group, Institute of Microbiology and Hygiene, Charité – University Medicine Berlin, Campus Benjamin Franklin
| |
Collapse
|
36
|
Gubernatorova EO, Tumanov AV. Tumor Necrosis Factor and Lymphotoxin in Regulation of Intestinal Inflammation. BIOCHEMISTRY. BIOKHIMIIA 2016; 81:1309-1325. [PMID: 27914457 DOI: 10.1134/s0006297916110092] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Ulcerative colitis and Crohn's disease are the major forms of inflammatory bowel disease. Cytokines of the tumor necrosis factor (TNF) family play an important role in the regulation of intestinal inflammation. In this review, we discuss the function of key cytokines of this family - TNF and lymphotoxin (LT) - in mucosal healing, IgA production, and in control of innate lymphoid cells (ILCs), novel regulators of mucosal homeostasis in the gut. TNF plays a central role in the pathogenesis of inflammatory bowel diseases (IBD). LT regulates group 3 of ILCs and IL-22 production and protects the epithelium against damage by chemicals and mucosal bacterial pathogens. In addition, we discuss major mouse models employed to study the mechanism of intestinal inflammation, their advantages and limitations, as well as application of TNF blockers in the therapy for IBD.
Collapse
Affiliation(s)
- E O Gubernatorova
- Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
| | | |
Collapse
|
37
|
Abstract
The cause of Crohn’s disease (CD) has posed a conundrum for at least a century. A large body of work coupled with recent technological advances in genome research have at last started to provide some of the answers. Initially this review seeks to explain and to differentiate between bowel inflammation in the primary immunodeficiencies that generally lead to very early onset diffuse bowel inflammation in humans and in animal models, and the real syndrome of CD. In the latter, a trigger, almost certainly enteric infection by one of a multitude of organisms, allows the faeces access to the tissues, at which stage the response of individuals predisposed to CD is abnormal. Direct investigation of patients’ inflammatory response together with genome-wide association studies (GWAS) and DNA sequencing indicate that in CD the failure of acute inflammation and the clearance of bacteria from the tissues, and from within cells, is defective. The retained faecal products result in the characteristic chronic granulomatous inflammation and adaptive immune response. In this review I will examine the contemporary evidence that has led to this understanding, and look for explanations for the recent dramatic increase in the incidence of this disease.
Collapse
|
38
|
Abstract
The cause of Crohn's disease (CD) has posed a conundrum for at least a century. A large body of work coupled with recent technological advances in genome research have at last started to provide some of the answers. Initially this review seeks to explain and to differentiate between bowel inflammation in the primary immunodeficiencies that generally lead to very early onset diffuse bowel inflammation in humans and in animal models, and the real syndrome of CD. In the latter, a trigger, almost certainly enteric infection by one of a multitude of organisms, allows the faeces access to the tissues, at which stage the response of individuals predisposed to CD is abnormal. Direct investigation of patients' inflammatory response together with genome-wide association studies (GWAS) and DNA sequencing indicate that in CD the failure of acute inflammation and the clearance of bacteria from the tissues, and from within cells, is defective. The retained faecal products result in the characteristic chronic granulomatous inflammation and adaptive immune response. In this review I will examine the contemporary evidence that has led to this understanding, and look for explanations for the recent dramatic increase in the incidence of this disease.
Collapse
|
39
|
de Bruyn M, Vandooren J, Ugarte-Berzal E, Arijs I, Vermeire S, Opdenakker G. The molecular biology of matrix metalloproteinases and tissue inhibitors of metalloproteinases in inflammatory bowel diseases. Crit Rev Biochem Mol Biol 2016; 51:295-358. [PMID: 27362691 DOI: 10.1080/10409238.2016.1199535] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
40
|
RETRACTED ARTICLE: Mouse models of intestinal inflammation and cancer. Arch Toxicol 2016; 90:2109-2130. [DOI: 10.1007/s00204-016-1747-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 06/01/2016] [Indexed: 12/19/2022]
|
41
|
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: 63] [Impact Index Per Article: 7.0] [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
|
42
|
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: 304] [Impact Index Per Article: 33.8] [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
|
43
|
Taghipour N, Molaei M, Mosaffa N, Rostami-Nejad M, Asadzadeh Aghdaei H, Anissian A, Azimzadeh P, Zali MR. An experimental model of colitis induced by dextran sulfate sodium from acute progresses to chronicity in C57BL/6: correlation between conditions of mice and the environment. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2016; 9:45-52. [PMID: 26744614 PMCID: PMC4702041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AIM To induce acute colitis progresses to chronicity in C57BL/6 mice by dextran sulfate sodium. BACKGROUND Murine models are essential tools to understand IBD pathogenesis. Among different types of chemically induced colitis models, the dextran sulfate sodium (DSS)-induced colitis model is the most common model of IBD, due to its simplicity. PATIENTS AND METHODS Male C57BL/6 mice 6-8 weeks old, were collected and matched by age with controls. C57BL/6 mice treated with 2 cycles of 3.5% DSS for 4 days and 4 days of pure water between each cycle. After that, mice were sacrificed and the entire colon was removed. Small sections of the colon were fixed in formaldehyde, embedded in paraffin and sectioned with a microtome. Sections were stained with hematoxylin eosin to analyses the degree of inflammation. RESULTS After the first cycle oral administration of DSS, mice with severe and visible rectal bleeding and diarrhea entered into the acute phase. After day 4-5, bleeding and diarrhea were improved and mice entered into the chronic phase with peak levels of weight loss. Macroscopically, the inflammation was predominantly located in the distal colon. Microscopically, examination of the distal colon sections showed a decrease number of goblet cells, loss of crypts, signs of surface epithelial regeneration and moderate to severe infiltration of inflammatory cells in the mucosa. CONCLUSION In order to achieve an experimental colitis model, our protocol is recommended for future therapies in IBD experimental modeling.
Collapse
Affiliation(s)
- Niloofar Taghipour
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Molaei
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nariman Mosaffa
- Immunology Department, Faculty of Medicine,Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rostami-Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Anissian
- Department of veterinary, College of Agriculture, Islamic Azad University, Abhar branch, Abhar, Iran
| | - Pedram Azimzadeh
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Wanner A, Sandhaus RA. Alpha-1 Antitrypsin as a Therapeutic Agent for Conditions not Associated with Alpha-1 Antitrypsin Deficiency. ALPHA-1 ANTITRYPSIN 2016. [PMCID: PMC7121596 DOI: 10.1007/978-3-319-23449-6_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Alpha-1 antitrypsin is a positive acute phase reactant whose serum level rises in response to inflammatory stress, presumably to balance pro-inflammatory processes. In addition to its serine protease inhibitory action, alpha-1 antitrypsin exhibits broader anti-inflammatory and immunomodulatory activity, and increasing its serum concentration by the administration of exogenous alpha-1 antitrypsin to above-normal levels potentially could be therapeutic in conditions other than alpha-1 antitrypsin deficiency. In vitro observations, studies in animal models and in some instances early human trials suggest that intravenous or inhaled alpha-1 antitrypsin has beneficial effects in type 1 diabetes, viral infections, graft-versus-host disease, cystic fibrosis, and alpha-1 antitrypsin-replete chronic obstructive pulmonary disease among others. While the results of pivotal clinical trials have not been reported to date, new indications for alpha-1 antitrypsin therapy are likely to emerge in the future based on currently available scientific data.
Collapse
Affiliation(s)
- Adam Wanner
- University of Miami Miller School of Medicine, Miami, Florida USA
| | | |
Collapse
|
45
|
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
|
46
|
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
|
47
|
Han D, Walsh MC, Kim KS, Hong SW, Lee J, Yi J, Rivas G, Surh CD, Choi Y. Microbiota-Independent Ameliorative Effects of Antibiotics on Spontaneous Th2-Associated Pathology of the Small Intestine. PLoS One 2015; 10:e0118795. [PMID: 25689829 PMCID: PMC4331505 DOI: 10.1371/journal.pone.0118795] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/15/2015] [Indexed: 11/18/2022] Open
Abstract
We have previously generated a mouse model of spontaneous Th2-associated disease of the small intestine called TRAF6ΔDC, in which dendritic cell (DC)-intrinsic expression of the signaling mediator TRAF6 is ablated. Interestingly, broad-spectrum antibiotic treatment ameliorates TRAF6ΔDC disease, implying a role for commensal microbiota in disease development. However, the relationship between the drug effects and commensal microbiota status remains to be formally demonstrated. To directly assess this relationship, we have now generated TRAF6ΔDC bone marrow chimera mice under germ-free (GF) conditions lacking commensal microbiota, and found, unexpectedly, that Th2-associated disease is actually exacerbated in GF TRAF6ΔDC mice compared to specific pathogen-free (SPF) TRAF6ΔDC mice. At the same time, broad-spectrum antibiotic treatment of GF TRAF6ΔDC mice has an ameliorative effect similar to that observed in antibiotics-treated SPF TRAF6ΔDC mice, implying a commensal microbiota-independent effect of broad-spectrum antibiotic treatment. We further found that treatment of GF TRAF6ΔDC mice with broad-spectrum antibiotics increases Foxp3+ Treg populations in lymphoid organs and the small intestine, pointing to a possible mechanism by which treatment may directly exert an immunomodulatory effect. To investigate links between the exacerbated phenotype of the small intestines of GF TRAF6ΔDC mice and local microbiota, we performed microbiotic profiling of the luminal contents specifically within the small intestines of diseased TRAF6ΔDC mice, and, when compared to co-housed control mice, found significantly increased total bacterial content characterized by specific increases in Firmicutes Lactobacillus species. These data suggest a protective effect of Firmicutes Lactobacillus against the spontaneous Th2-related inflammation of the small intestine of the TRAF6ΔDC model, and may represent a potential mechanism for related disease phenotypes.
Collapse
Affiliation(s)
- Daehee Han
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, 790–784, Republic of Korea
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 790–784, Republic of Korea
| | - Matthew C. Walsh
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, 19104, United States of America
| | - Kwang Soon Kim
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, 790–784, Republic of Korea
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 790–784, Republic of Korea
| | - Sung-Wook Hong
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, 790–784, Republic of Korea
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 790–784, Republic of Korea
| | - Junyoung Lee
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, 790–784, Republic of Korea
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 790–784, Republic of Korea
| | - Jaeu Yi
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, 790–784, Republic of Korea
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 790–784, Republic of Korea
| | - Gloriany Rivas
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, 19104, United States of America
| | - Charles D. Surh
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang, 790–784, Republic of Korea
- Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 790–784, Republic of Korea
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California, 92037, United States of America
- * E-mail: (YC); (CDS)
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, 19104, United States of America
- * E-mail: (YC); (CDS)
| |
Collapse
|
48
|
Abstract
The understanding of the intestinal inflammation occurring in the inflammatory bowel diseases (IBD) has been immeasurably advanced by the development of the now numerous murine models of intestinal inflammation. The usefulness of this research tool in IBD studies has been enabled by our improved knowledge of mucosal immunity and thus our improved ability to interpret the complex responses of mice with various causes of colitis; in addition, it has been powered by the availability of models in which the mice have specific genetic and/or immunologic defects that can be related to the origin of the inflammation. Finally, and more recently, it has been enhanced by our newly acquired ability to define the intestinal microbiome under various conditions and thus to understand how intestinal microorganisms impact on inflammation. In this brief review of murine models of intestinal inflammation we focus mainly on the most often used models that are, not incidentally, also the models that have yielded major insights into IBD pathogenesis.
Collapse
Affiliation(s)
| | | | - Warren Strober
- Correspondence Address correspondence to: Warren Strober, MD, National Institutes of Health, Mucosal Immunity Section, 10 Center Drive, CRC Bldg. 10 5west-3940, Bethesda, Maryland 20892. fax: (301) 402-2240.
| |
Collapse
|
49
|
Park JH, Kwon JG, Kim SJ, Song DK, Lee SG, Kim ES, Kim ES, Cho KB, Jang BI, Kim DH, Sin JI, Kim TW, Song IH, Park KS. Alterations of colonic contractility in an interleukin-10 knockout mouse model of inflammatory bowel disease. J Neurogastroenterol Motil 2015; 21:51-61. [PMID: 25537671 PMCID: PMC4288096 DOI: 10.5056/jnm14008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 09/25/2014] [Accepted: 09/27/2014] [Indexed: 12/18/2022] Open
Abstract
Background/Aims Inflammatory bowel disease is commonly accompanied by colonic dysmotility and causes changes in intestinal smooth muscle contractility. In this study, colonic smooth muscle contractility in a chronic inflammatory condition was investigated using smooth muscle tissues prepared from interleukin-10 knockout (IL-10−/−) mice. Methods Prepared smooth muscle sections were placed in an organ bath system. Cholinergic and nitrergic neuronal responses were observed using carbachol and electrical field stimulation with L-NG-nitroarginine methyl ester (L-NAME). The expression of interstitial cells of Cajal (ICC) networks, muscarinic receptors, neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS) was observed via immunofluorescent staining. Results The spontaneous contractility and expression of ICC networks in the proximal and distal colon was significantly decreased in IL-10−/− mice compared to IL-10+/+ mice. The contractility in response to carbachol was significantly decreased in the proximal colon of IL-10−/− mice compared to IL-10+/+ mice, but no significant difference was found in the distal colon. In addition, the expression of muscarinic receptor type 2 was reduced in the proximal colon of IL-10−/− mice. The nictric oxide-mediated relaxation after electrical field stimulation was significantly decreased in the proximal and distal colon of IL-10−/− mice. In inflamed colon, the expression of nNOS decreased, whereas the expression of iNOS increased. Conclusions These results suggest that damage to the ICC network and NOS system in the proximal and distal colon, as well as damage to the smooth muscle cholinergic receptor in the proximal colon may play an important role in the dysmotility of the inflamed colon.
Collapse
Affiliation(s)
- Jae Hyung Park
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Joong Goo Kwon
- Department of Internal Medicine, Catholic University of Daegu School of Medicine, Daegu, Korea
| | - Sun Joo Kim
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Dae Kyu Song
- Department of Physiology, Keimyung University School of Medicine, Daegu, Korea
| | - Seok Guen Lee
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | | | - Eun Su Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Kwang Bum Cho
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Byung Ik Jang
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Dae Hwan Kim
- Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Korea
| | - Jeong-Im Sin
- Department of Microbiology, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Tae Wan Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Kyungpook National University, Daegu, Korea
| | - In Hwan Song
- Department of Anatomy, Yeungnam University College of Medicine, Daegu, Korea
| | - Kyung Sik Park
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| |
Collapse
|
50
|
Henderson AL, Brand MW, Darling RJ, Maas KJ, Detzel CJ, Hostetter J, Wannemuehler MJ, Weaver EM. Attenuation of Colitis by Serum-Derived Bovine Immunoglobulin/Protein Isolate in a Defined Microbiota Mouse Model. Dig Dis Sci 2015; 60:3293-303. [PMID: 26026602 PMCID: PMC4621698 DOI: 10.1007/s10620-015-3726-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 05/21/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND The pathogenesis of inflammatory bowel disease (IBD) is complex and multifaceted including genetic predisposition, environmental components, microbial dysbiosis, and inappropriate immune activation to microbial components. Pathogenic bacterial provocateurs like adherent and invasive E. coli have been reported to increase susceptibility to Crohn's disease. Serum-derived bovine immunoglobulin/protein isolate (SBI) is comprised primarily of immunoglobulins (Igs) that bind to conserved microbial components and neutralize exotoxins. AIM To demonstrate that oral administration of SBI may modulate mucosal inflammation following colonization with E. coli, LF82, and exposure to dextran sodium sulfate (DSS). METHODS Defined microbiota mice harboring the altered Schaedler flora (ASF) were administered SBI or hydrolyzed collagen twice daily starting 7 days prior to challenge with E. coli LF82 and continuing for the remainder of the experiment. Mice were treated with DSS for 7 days and then evaluated for evidence of local and peripheral inflammation. RESULTS Igs within SBI bound multiple antigens from all eight members of the ASF and E. coli LF82 by western blot analysis. Multiple parameters of LF82/DSS-induced colitis were reduced following administration of SBI, including histological lesion scores, secretion of cytokines and chemokines from cecal biopsies, intestinal fatty acid binding protein (I-FABP) and serum amyloid A from plasma. CONCLUSIONS Oral administration of SBI attenuated clinical signs of LF82/DSS-induced colitis in mice. The data are consistent with the hypothesis that SBI immunoglobulin binding of bacterial antigens in the intestinal lumen may inhibit the inflammatory cascades that contribute to IBD, thus attenuating DSS-induced colitis.
Collapse
Affiliation(s)
| | - Meghan Wymore Brand
- />Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011 USA
| | - Ross J. Darling
- />Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011 USA
| | - Kenneth J. Maas
- />Entera Health, Inc., 2575 SE Oak Tree Ct., Ankeny, IA 50023 USA , />Entera Health, Inc., Cary, NC 27518 USA
| | | | - Jesse Hostetter
- />Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA 50011 USA
| | - Michael J. Wannemuehler
- />Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011 USA
| | - Eric M. Weaver
- />Entera Health, Inc., 2575 SE Oak Tree Ct., Ankeny, IA 50023 USA
| |
Collapse
|