1
|
Song CH, Kim N, Nam RH, Choi SI, Jang JY, Kim EH, Ha S, Shin E, Choi H, Kim KW, Jeon S, Oh GT, Seok YJ. Ninjurin1 deficiency differentially mitigates colorectal cancer induced by azoxymethane and dextran sulfate sodium in male and female mice. Int J Cancer 2024. [PMID: 39417611 DOI: 10.1002/ijc.35225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 10/19/2024]
Abstract
This study investigated the role of Ninjurin1 (Ninj1), encoding a small transmembrane protein, in colitis-associated colon tumorigenesis in relation to sex hormones. Male and female wild-type (WT) and Ninj1 knockout (KO) mice were treated with azoxymethane (AOM) and dextran sulfate sodium (DSS), with or without testosterone propionate (TP). At week 2 (acute colitis stage), Ninj1 KO exhibited an alleviation in the colitis symptoms in both male and female mice. The M2 macrophage population increased and CD8+ T cell population decreased only in the female Ninj1 KO than in the female WT AOM/DSS group. In the female AOM/DSS group, TP treatment exacerbated colon shortening in the Ninj1 KO than in the WT. At week 13 (tumorigenesis stage), male Ninj1 KO mice had fewer tumors, but females showed similar tumors. In the WT AOM/DSS group, females had more M2 macrophages and fewer M1 macrophages than males, but this difference was absent in Ninj1 KO mice. In the Ninj1 KO versus WT group, the expression of pro-inflammatory mediators and Ho-1 and CD8+ T cell populations decreased in both female and male Ninj1 KO mice. In the WT group, M2 macrophage populations were increased by AOM/DSS treatment and decreased by TP treatment. However, neither treatment changed the cell populations in the Ninj1 KO group. These results suggest that Ninj1 is involved in colorectal cancer development in a testosterone-dependent manner, which was different in male and female. This highlights the importance of considering sex disparities in understanding Ninj1's role in cancer pathogenesis.
Collapse
Affiliation(s)
- Chin-Hee Song
- Department of Internal Medicine and Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, South Korea
| | - Nayoung Kim
- Department of Internal Medicine and Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, South Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine and Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, South Korea
| | - Soo In Choi
- Department of Internal Medicine and Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, South Korea
| | - Jae Young Jang
- Department of Internal Medicine and Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, South Korea
| | - Eun Hye Kim
- Department of Internal Medicine and Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, South Korea
| | - Sungchan Ha
- Department of Internal Medicine and Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, South Korea
| | - Eun Shin
- Department of Pathology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Gyeonggi-do, South Korea
| | - Hoon Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Kyu-Won Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Sejin Jeon
- Department of Vaccine Biothechnology, Andong National University, Andong, South Korea
| | - Goo Taeg Oh
- Department of Life Sciences, Heart-Immune-Brain Network Research Center, Ewha Womans University, Seoul, South Korea
| | - Yeong-Jae Seok
- Department of Biological Sciences and Institute of Microbiology, Seoul National University, Seoul, South Korea
| |
Collapse
|
2
|
Remke M, Groll T, Metzler T, Urbauer E, Kövilein J, Schnalzger T, Ruland J, Haller D, Steiger K. Histomorphological scoring of murine colitis models: A practical guide for the evaluation of colitis and colitis-associated cancer. Exp Mol Pathol 2024; 140:104938. [PMID: 39418944 DOI: 10.1016/j.yexmp.2024.104938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/19/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND AND AIMS Histomorphology is a powerful and cost-efficient tool for evaluating inflammatory and neoplastic conditions. Inflammatory bowel disease (IBD) is a widespread condition with globally rising incidences, and a lot of research is done to better understand the pathogenesis of IBD and to identify potential therapeutic approaches. However, standardized and reproducible scores for the histomorphological evaluation of murine IBD models are lacking. Therefore, we aimed to develop an easy-to-use and reproducible score for standardized assessment of colitis and associated cancer models. METHODS In this study, samples from three different colitis models with and without associated cancer formation were analyzed to develop a universal, robust, and reproducible score for the grading of murine colitis models using the following three parameters: 1. Extent of leucocyte infiltration, 2. Tissue damage, 3. Architectural disruption of the mucosa. RESULTS A scoring system was established for different kinds of colitis models (genetically induced enterocolitis, genetically induced metabolic injury, and chemically induced colitis-associated cancer) and all stages of the disease, from mild inflammatory changes to severe inflammation with neoplastic changes as the extreme extent of IBD. The scoring scheme is easy to use, can easily be learned, and proves to have a high interrater reliability. CONCLUSIONS We propose a robust histological scoring system for the assessment of murine colitis and colitis-associated cancer models, giving more researchers access to conclusive and reliable histological assessment.
Collapse
Affiliation(s)
- Marianne Remke
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Trogerstr. 18, 81675 Munich, Germany; Comparative Experimental Pathology, School of Medicine and Health, Technical University of Munich, Trogerstr. 18, 81675 Munich, Germany; Member of the German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
| | - Tanja Groll
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Trogerstr. 18, 81675 Munich, Germany; Comparative Experimental Pathology, School of Medicine and Health, Technical University of Munich, Trogerstr. 18, 81675 Munich, Germany
| | - Thomas Metzler
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Trogerstr. 18, 81675 Munich, Germany; Comparative Experimental Pathology, School of Medicine and Health, Technical University of Munich, Trogerstr. 18, 81675 Munich, Germany
| | - Elisabeth Urbauer
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Janine Kövilein
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Theresa Schnalzger
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Trogerstr. 18, 81675 Munich, Germany; Comparative Experimental Pathology, School of Medicine and Health, Technical University of Munich, Trogerstr. 18, 81675 Munich, Germany; Member of the German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
3
|
Zhang S, Nie Q, Sun Y, Zuo S, Chen C, Li S, Yang J, Hu J, Zhou X, Yu Y, Huang P, Lian L, Xie M, Nie S. Bacteroides uniformis degrades β-glucan to promote Lactobacillus johnsonii improving indole-3-lactic acid levels in alleviating colitis. MICROBIOME 2024; 12:177. [PMID: 39300532 DOI: 10.1186/s40168-024-01896-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/30/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Intake of dietary fiber is associated with a reduced risk of inflammatory bowel disease. β-Glucan (BG), a bioactive dietary fiber, has potential health-promoting effects on intestinal functions; however, the underlying mechanism remains unclear. Here, we explore the role of BG in ameliorating colitis by modulating key bacteria and metabolites, confirmed by multiple validation experiments and loss-of-function studies, and reveal a novel bacterial cross-feeding interaction. RESULTS BG intervention ameliorates colitis and reverses Lactobacillus reduction in colitic mice, and Lactobacillus abundance was significantly negatively correlated with the severity of colitis. It was confirmed by further studies that Lactobacillus johnsonii was the most significantly enriched Lactobacillus spp. Multi-omics analysis revealed that L. johnsonii produced abundant indole-3-lactic acid (ILA) leading to the activation of aryl hydrocarbon receptor (AhR) responsible for the mitigation of colitis. Interestingly, L. johnsonii cannot utilize BG but requires a cross-feeding with Bacteroides uniformis, which degrades BG and produces nicotinamide (NAM) to promote the growth of L. johnsonii. A proof-of-concept study confirmed that BG increases L. johnsonii and B. uniformis abundance and ILA levels in healthy individuals. CONCLUSIONS These findings demonstrate the mechanism by which BG ameliorates colitis via L. johnsonii-ILA-AhR axis and reveal the important cross-feeding interaction between L. johnsonii and B. uniformis. Video Abstract.
Collapse
Affiliation(s)
- Shanshan Zhang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Qixing Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Yonggan Sun
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Sheng Zuo
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Chunhua Chen
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Song Li
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Jingrui Yang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Jielun Hu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Xingtao Zhou
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Yongkang Yu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China
| | - Ping Huang
- Department of Nutrition, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lu Lian
- Department of Nutrition, the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mingyong Xie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China.
| | - Shaoping Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang, China.
| |
Collapse
|
4
|
Mertens RT, Misra A, Xiao P, Baek S, Rone JM, Mangani D, Sivanathan KN, Arojojoye AS, Awuah SG, Lee I, Shi GP, Petrova B, Brook JR, Anderson AC, Flavell RA, Kanarek N, Hemberg M, Nowarski R. A metabolic switch orchestrated by IL-18 and the cyclic dinucleotide cGAMP programs intestinal tolerance. Immunity 2024; 57:2077-2094.e12. [PMID: 38906145 DOI: 10.1016/j.immuni.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/10/2024] [Accepted: 06/04/2024] [Indexed: 06/23/2024]
Abstract
Tissues are exposed to diverse inflammatory challenges that shape future inflammatory responses. While cellular metabolism regulates immune function, how metabolism programs and stabilizes immune states within tissues and tunes susceptibility to inflammation is poorly understood. Here, we describe an innate immune metabolic switch that programs long-term intestinal tolerance. Intestinal interleukin-18 (IL-18) stimulation elicited tolerogenic macrophages by preventing their proinflammatory glycolytic polarization via metabolic reprogramming to fatty acid oxidation (FAO). FAO reprogramming was triggered by IL-18 activation of SLC12A3 (NCC), leading to sodium influx, release of mitochondrial DNA, and activation of stimulator of interferon genes (STING). FAO was maintained in macrophages by a bistable switch that encoded memory of IL-18 stimulation and by intercellular positive feedback that sustained the production of macrophage-derived 2'3'-cyclic GMP-AMP (cGAMP) and epithelial-derived IL-18. Thus, a tissue-reinforced metabolic switch encodes durable immune tolerance in the gut and may enable reconstructing compromised immune tolerance in chronic inflammation.
Collapse
Affiliation(s)
- Randall T Mertens
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Aditya Misra
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Harvard-MIT Program in Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peng Xiao
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Seungbyn Baek
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Joseph M Rone
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Davide Mangani
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kisha N Sivanathan
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | - Samuel G Awuah
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA; Center for Pharmaceutical Research and Innovation, College of Pharmacy and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; POSTECH Biotech Center, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Boryana Petrova
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jeannette R Brook
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ana C Anderson
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Howard Hughes Medical Institute, Yale University, New Haven, CT, USA
| | - Naama Kanarek
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Martin Hemberg
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Roni Nowarski
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA; Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
5
|
Bhol NK, Bhanjadeo MM, Singh AK, Dash UC, Ojha RR, Majhi S, Duttaroy AK, Jena AB. The interplay between cytokines, inflammation, and antioxidants: mechanistic insights and therapeutic potentials of various antioxidants and anti-cytokine compounds. Biomed Pharmacother 2024; 178:117177. [PMID: 39053423 DOI: 10.1016/j.biopha.2024.117177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/03/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024] Open
Abstract
Cytokines regulate immune responses essential for maintaining immune homeostasis, as deregulated cytokine signaling can lead to detrimental outcomes, including inflammatory disorders. The antioxidants emerge as promising therapeutic agents because they mitigate oxidative stress and modulate inflammatory pathways. Antioxidants can potentially ameliorate inflammation-related disorders by counteracting excessive cytokine-mediated inflammatory responses. A comprehensive understanding of cytokine-mediated inflammatory pathways and the interplay with antioxidants is paramount for developing natural therapeutic agents targeting inflammation-related disorders and helping to improve clinical outcomes and enhance the quality of life for patients. Among these antioxidants, curcumin, vitamin C, vitamin D, propolis, allicin, and cinnamaldehyde have garnered attention for their anti-inflammatory properties and potential therapeutic benefits. This review highlights the interrelationship between cytokines-mediated disorders in various diseases and therapeutic approaches involving antioxidants.
Collapse
Affiliation(s)
- Nitish Kumar Bhol
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, Odisha 751004, India
| | | | - Anup Kumar Singh
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India
| | - Umesh Chandra Dash
- Environmental Biotechnology Laboratory, KIIT School of Biotechnology, KIIT Deemed to be University, Bhubaneswar, Odisha, India
| | - Rakesh Ranjan Ojha
- Department of Bioinformatics, BJB (A) College, Bhubaneswar, Odisha-751014, India
| | - Sanatan Majhi
- Post Graduate Department of Biotechnology, Utkal University, Bhubaneswar, Odisha 751004, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, Norway.
| | - Atala Bihari Jena
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune, India.
| |
Collapse
|
6
|
Song Y, Lu J, Qin P, Chen H, Chen L. Interferon-I modulation and natural products: Unraveling mechanisms and therapeutic potential in severe COVID-19. Cytokine Growth Factor Rev 2024:S1359-6101(24)00066-2. [PMID: 39261232 DOI: 10.1016/j.cytogfr.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to pose a significant global public health threat, particularly to older adults, pregnant women, and individuals with underlying chronic conditions. Dysregulated immune responses to SARS-CoV-2 infection are believed to contribute to the progression of COVID-19 in severe cases. Previous studies indicates that a deficiency in type I interferon (IFN-I) immunity accounts for approximately 15 %-20 % of patients with severe pneumonia caused by COVID-19, highlighting the potential therapeutic importance of modulating IFN-I signals. Natural products and their derivatives, due to their structural diversity and novel scaffolds, play a crucial role in drug discovery. Some of these natural products targeting IFN-I have demonstrated applications in infectious diseases and inflammatory conditions. However, the immunomodulatory potential of IFN-I in critical COVID-19 pneumonia and the natural compounds regulating the related signal pathway remain not fully understood. In this review, we offer a comprehensive assessment of the association between IFN-I and severe COVID-19, exploring its mechanisms and integrating information on natural compounds effective for IFN-I regulation. Focusing on the primary targets of IFN-I, we also summarize the regulatory mechanisms of natural products, their impact on IFNs, and their therapeutic roles in viral infections. Collectively, by synthesizing these findings, our goal is to provide a valuable reference for future research and to inspire innovative treatment strategies for COVID-19.
Collapse
Affiliation(s)
- Yuheng Song
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiani Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pengcheng Qin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Henan University, Kaifeng 475001, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 200032, China
| | - Lili Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
7
|
Meng Q, Ning J, Lu J, Zhang J, Zu M, Zhang J, Han X, Zheng H, Gong Y, Hao X, Xiong Y, Gu F, Han W, Fu W, Wang J, Ding S. Cmtm4 deficiency exacerbates colitis by inducing gut dysbiosis and S100a8/9 expression. J Genet Genomics 2024; 51:811-823. [PMID: 38575111 DOI: 10.1016/j.jgg.2024.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
The dysfunction of innate immunity components is one of the major drivers for ulcerative colitis (UC), and increasing reports indicate that the gut microbiome serves as an intermediate between genetic mutations and UC development. Here, we find that the IL-17 receptor subunit, CMTM4, is reduced in UC patients and dextran sulfate sodium (DSS)-induced colitis. The deletion of CMTM4 (Cmtm4-/-) in mice leads to a higher susceptibility to DSS-induced colitis than in wild-type, and the gut microbiome significantly changes in composition. The causal role of the gut microbiome is confirmed with a cohousing experiment. We further identify that S100a8/9 is significantly up-regulated in Cmtm4-/- colitis, with the block of its receptor RAGE that reverses the phenotype associated with the CMTM4 deficiency. CMTM4 deficiency rather suppresses S100a8/9 expression in vitro via the IL17 pathway, further supporting that the elevation of S100a8/9 in vivo is most likely a result of microbial dysbiosis. Taken together, the results suggest that CMTM4 is involved in the maintenance of intestinal homeostasis, suppression of S100a8/9, and prevention of colitis development. Our study further shows CMTM4 as a crucial innate immunity component, confirming its important role in UC development and providing insights into potential targets for the development of future therapies.
Collapse
Affiliation(s)
- Qiao Meng
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China
| | - Jing Ning
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China
| | - Jingjing Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100101, China
| | - Jing Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China
| | - Ming Zu
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China
| | - Jing Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China
| | - Xiurui Han
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China
| | - Huiling Zheng
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China
| | - Yueqing Gong
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China
| | - Xinyu Hao
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China
| | - Ying Xiong
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China
| | - Fang Gu
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China
| | - Wenling Han
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, NHC Key Laboratory of Medical Immunology (Peking University), Beijing 100191, China; Peking University Center for Human Disease Genomics, Beijing 100191, China
| | - Weiwei Fu
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China.
| | - Jun Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shigang Ding
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371), Beijing 100191, China.
| |
Collapse
|
8
|
Huo Z, Li J, Li X, Xiao H, Lin Y, Ma Y, Li J, Yang H, Zhang C. Functional fractions of Astragalus polysaccharides as a potential prebiotic to alleviate ulcerative colitis. Int J Biol Macromol 2024; 271:132580. [PMID: 38788871 DOI: 10.1016/j.ijbiomac.2024.132580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/02/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease of the intestine that is significantly influenced by an imbalance in the gut microbiota. Astragalus membranaceus, particularly its polysaccharide components, has shown therapeutic potential for the treatment of UC, although the specific active constituents and their mechanistic pathways remain to be fully elucidated. In this study, we investigated two molecular weight fractions of Astragalus polysaccharides (APS), APS1 (Mw < 10 kDa) and APS2 (10 kDa < Mw < 50 kDa), isolated by ultrafiltration, focusing on their prebiotic effects, effects on UC, and the underlying mechanism. Our results showed that both APS1 and APS2 exhibit prebiotic properties, with APS1 significantly outperforming APS2 in ameliorating UC symptoms. APS1 significantly attenuated weight loss and UC manifestations, reduced colonic pathology, and improved intestinal mucosal barrier integrity. In addition, APS1 significantly reduced the levels of inflammatory cytokines in the serum and colonic tissue, and downregulated colonic chemokines. Furthermore, APS1 ameliorated dextran sulfate sodium salt (DSS)-induced intestinal dysbiosis by promoting the growth of beneficial microbes and inhibiting the proliferation of potential pathogens, leading to a significant increase in short-chain fatty acids. In conclusion, this study highlights the potential of APS1 as a novel prebiotic for the prevention and treatment of UC.
Collapse
Affiliation(s)
- Zeqi Huo
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China; Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Junxiang Li
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China; Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Xiaofeng Li
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China; Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Han Xiao
- Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Yang Lin
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China; Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Yuchan Ma
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jiaru Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Hui Yang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Chunjiang Zhang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China; Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
9
|
Chang C, Gupta R, Sedighian F, Louie A, Gonzalez DM, Le C, Cho JM, Park SK, Castellanos J, Ting TW, Dong TS, Arias-Jayo N, Lagishetty V, Navab M, Reddy S, Sioutas C, Hsiai T, Jacobs JP, Araujo JA. Subchronic inhalation exposure to ultrafine particulate matter alters the intestinal microbiome in various mouse models. ENVIRONMENTAL RESEARCH 2024; 248:118242. [PMID: 38242419 DOI: 10.1016/j.envres.2024.118242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/13/2024] [Accepted: 01/17/2024] [Indexed: 01/21/2024]
Abstract
Exposure to ultrafine particles (UFPs) has been associated with multiple adverse health effects. Inhaled UFPs could reach the gastrointestinal tract and influence the composition of the gut microbiome. We have previously shown that oral ingestion of UFPs alters the gut microbiome and promotes intestinal inflammation in hyperlipidemic Ldlr-/- mice. Particulate matter (PM)2.5 inhalation studies have also demonstrated microbiome shifts in normolipidemic C57BL/6 mice. However, it is not known whether changes in microbiome precede or follow inflammatory effects in the intestinal mucosa. We hypothesized that inhaled UFPs modulate the gut microbiome prior to the development of intestinal inflammation. We studied the effects of UFP inhalation on the gut microbiome and intestinal mucosa in two hyperlipidemic mouse models (ApoE-/- mice and Ldlr-/- mice) and normolipidemic C57BL/6 mice. Mice were exposed to PM in the ultrafine-size range by inhalation for 6 h a day, 3 times a week for 10 weeks at a concentration of 300-350 μg/m3.16S rRNA gene sequencing was performed to characterize sequential changes in the fecal microbiome during exposures, and changes in the intestinal microbiome at the end. PM exposure led to progressive differentiation of the microbiota over time, associated with increased fecal microbial richness and evenness, altered microbial composition, and differentially abundant microbes by week 10 depending on the mouse model. Cross-sectional analysis of the small intestinal microbiome at week 10 showed significant changes in α-diversity, β-diversity, and abundances of individual microbial taxa in the two hyperlipidemic models. These alterations of the intestinal microbiome were not accompanied, and therefore could not be caused, by increased intestinal inflammation as determined by histological analysis of small and large intestine, cytokine gene expression, and levels of fecal lipocalin. In conclusion, 10-week inhalation exposures to UFPs induced taxonomic changes in the microbiome of various animal models in the absence of intestinal inflammation.
Collapse
Affiliation(s)
- Candace Chang
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA; Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Rajat Gupta
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Farzaneh Sedighian
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Allen Louie
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - David M Gonzalez
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Collin Le
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jae Min Cho
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Seul-Ki Park
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jocelyn Castellanos
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| | - To-Wei Ting
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Tien S Dong
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA; Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, Los Angeles, CA, USA
| | - Nerea Arias-Jayo
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Venu Lagishetty
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Mohamad Navab
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Srinivasa Reddy
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA; Molecular & Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Constantinos Sioutas
- University of Southern California (USC) Viterbi School of Engineering, Los Angeles, CA, USA
| | - Tzung Hsiai
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Henry Samueli School of Engineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Jonathan P Jacobs
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA; Division of Gastroenterology, Hepatology and Parenteral Nutrition, Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, Los Angeles, CA, USA.
| | - Jesus A Araujo
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Department of Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA; Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Maler MD, Zwick S, Kallfass C, Engelhard P, Shi H, Hellig L, Zhengyang P, Hardt A, Zissel G, Ruzsics Z, Jahnen-Dechent W, Martin SF, Nielsen PJ, Stolz D, Lopatecka J, Bastyans S, Beutler B, Schamel WW, Fejer G, Freudenberg MA. Type I Interferon, Induced by Adenovirus or Adenoviral Vector Infection, Regulates the Cytokine Response to Lipopolysaccharide in a Macrophage Type-Specific Manner. J Innate Immun 2024; 16:226-247. [PMID: 38527452 PMCID: PMC11023693 DOI: 10.1159/000538282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
INTRODUCTION While TLR ligands derived from microbial flora and pathogens are important activators of the innate immune system, a variety of factors such as intracellular bacteria, viruses, and parasites can induce a state of hyperreactivity, causing a dysregulated and potentially life-threatening cytokine over-response upon TLR ligand exposure. Type I interferon (IFN-αβ) is a central mediator in the induction of hypersensitivity and is strongly expressed in splenic conventional dendritic cells (cDC) and marginal zone macrophages (MZM) when mice are infected with adenovirus. This study investigates the ability of adenoviral infection to influence the activation state of the immune system and underlines the importance of considering this state when planning the treatment of patients. METHODS Infection with adenovirus-based vectors (Ad) or pretreatment with recombinant IFN-β was used as a model to study hypersensitivity to lipopolysaccharide (LPS) in mice, murine macrophages, and human blood samples. The TNF-α, IL-6, IFN-αβ, and IL-10 responses induced by LPS after pretreatment were measured. Mouse knockout models for MARCO, IFN-αβR, CD14, IRF3, and IRF7 were used to probe the mechanisms of the hypersensitive reaction. RESULTS We show that, similar to TNF-α and IL-6 but not IL-10, the induction of IFN-αβ by LPS increases strongly after Ad infection. This is true both in mice and in human blood samples ex vivo, suggesting that the regulatory mechanisms seen in the mouse are also present in humans. In mice, the scavenger receptor MARCO on IFN-αβ-producing cDC and splenic marginal zone macrophages is important for Ad uptake and subsequent cytokine overproduction by LPS. Interestingly, not all IFN-αβ-pretreated macrophage types exposed to LPS exhibit an enhanced TNF-α and IL-6 response. Pretreated alveolar macrophages and alveolar macrophage-like murine cell lines (MPI cells) show enhanced responses, while bone marrow-derived and peritoneal macrophages show a weaker response. This correlates with the respective absence or presence of the anti-inflammatory IL-10 response in these different macrophage types. In contrast, Ad or IFN-β pretreatment enhances the subsequent induction of IFN-αβ in all macrophage types. IRF3 is dispensable for the LPS-induced IFN-αβ overproduction in infected MPI cells and partly dispensable in infected mice, while IRF7 is required. The expression of the LPS co-receptor CD14 is important but not absolutely required for the elicitation of a TNF-α over-response to LPS in Ad-infected mice. CONCLUSION Viral infections or application of virus-based vaccines induces type I interferon and can tip the balance of the innate immune system in the direction of hyperreactivity to a subsequent exposure to TLR ligands. The adenoviral model presented here is one example of how multiple factors, both environmental and genetic, affect the physiological responses to pathogens. Being able to measure the current reactivity state of the immune system would have important benefits for infection-specific therapies and for the prevention of vaccination-elicited adverse effects.
Collapse
Affiliation(s)
- Mareike D. Maler
- Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Allergy Research Group, Department of Dermatology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Sophie Zwick
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Carsten Kallfass
- Institute of Virology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Peggy Engelhard
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Hexin Shi
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Laura Hellig
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Pang Zhengyang
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Annika Hardt
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Gernot Zissel
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Zsolt Ruzsics
- Institute of Virology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Willi Jahnen-Dechent
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Stefan F. Martin
- Allergy Research Group, Department of Dermatology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Peter Jess Nielsen
- Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Daiana Stolz
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Justyna Lopatecka
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Faculty of Medicine, Aachen, Germany
| | - Sarah Bastyans
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Faculty of Medicine, Aachen, Germany
| | - Bruce Beutler
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wolfgang W. Schamel
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
- School of Biomedical Sciences, Faculty of Health, University of Plymouth, Plymouth, UK
| | - György Fejer
- Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Faculty of Medicine, Aachen, Germany
| | - Marina Alexandra Freudenberg
- Max-Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| |
Collapse
|
11
|
Zhang S, Sun Y, Nie Q, Hu J, Li Y, Shi Z, Ji H, Zhang H, Zhao M, Chen C, Nie S. Effects of four food hydrocolloids on colitis and their regulatory effect on gut microbiota. Carbohydr Polym 2024; 323:121368. [PMID: 37940266 DOI: 10.1016/j.carbpol.2023.121368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/21/2023] [Accepted: 09/04/2023] [Indexed: 11/10/2023]
Abstract
Hydrocolloids are important food additives and have potential regulatory effects on gut microbiota. The development of colitis is closely related to changes in gut microbiota. The effect of food hydrocolloids on the structure of the gut microbiota and their impact on colitis has not been well investigated. Therefore, this study investigated the effects of four hydrocolloids (carrageenan, guar gum, xanthan gum, and pectin) on colitis, and explored their regulatory effects on gut microbiota. The results indicated that pectin and guar effectively alleviated body weight loss and disease activity index, reduced inflammatory cytokine levels, and promoted short-chain fatty acids (SCFAs) production. They increased the abundance of Akkermansia muciniphila, Oscillospira, and Lactobacillus, and Akkermansia abundance had a negative correlation with the severity of colitis. In contrast, carrageenan and xanthan gum did not significantly improve colitis, and carrageenan reduced the production of SCFAs. Both carrageenan and xanthan gum increased the abundance of Ruminococcus gnavus, and Ruminococcus abundance was positively correlated with the severity of colitis. These findings suggest that food additives have an impact on host health and provide guidance for the diet of patients with colitis.
Collapse
Affiliation(s)
- Shanshan Zhang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Yonggan Sun
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Qixing Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Jielun Hu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Yuhao Li
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Zefu Shi
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Haihua Ji
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Mingjiao Zhao
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Chunhua Chen
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
12
|
Silva RCMC, Travassos LH, Dutra FF. The dichotomic role of single cytokines: Fine-tuning immune responses. Cytokine 2024; 173:156408. [PMID: 37925788 DOI: 10.1016/j.cyto.2023.156408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
Cytokines are known for their pleiotropic effects. They can be classified by their function as pro-inflammatory, such as tumor necrosis factor (TNF), interleukin (IL) 1 and IL-12, or anti-inflammatory, like IL-10, IL-35 and transforming growth factor β (TGF-β). Though this type of classification is an important simplification for the understanding of the general cytokine's role, it can be misleading. Here, we discuss recent studies that show a dichotomic role of the so-called pro and anti-inflammatory cytokines, highlighting that their function can be dependent on the microenvironment and their concentrations. Furthermore, we discuss how the back-and-forth interplay between cytokines and immunometabolism can influence the dichotomic role of inflammatory responses as an important target to complement cytokine-based therapies.
Collapse
Affiliation(s)
| | - Leonardo Holanda Travassos
- Laboratório de Receptores e Sinalização intracelular, Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, Brazil
| | - Fabianno Ferreira Dutra
- Laboratório de Imunologia e Inflamação, Instituto de Microbiologia Paulo de Góes, UFRJ, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Paudel D, Nair DVT, Tian S, Hao F, Goand UK, Joseph G, Prodes E, Chai Z, Robert CE, Chassaing B, Patterson AD, Singh V. Dietary fiber guar gum-induced shift in gut microbiota metabolism and intestinal immune activity enhances susceptibility to colonic inflammation. Gut Microbes 2024; 16:2341457. [PMID: 38630030 PMCID: PMC11028019 DOI: 10.1080/19490976.2024.2341457] [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: 08/22/2023] [Accepted: 04/04/2024] [Indexed: 04/19/2024] Open
Abstract
With an increasing interest in dietary fibers (DFs) to promote intestinal health and the growth of beneficial gut bacteria, there is a continued rise in the incorporation of refined DFs in processed foods. It is still unclear how refined fibers, such as guar gum, affect the gut microbiota activity and pathogenesis of inflammatory bowel disease (IBD). Our study elucidated the effect and underlying mechanisms of guar gum, a fermentable DF (FDF) commonly present in a wide range of processed foods, on colitis development. We report that guar gum containing diet (GuD) increased the susceptibility to colonic inflammation. Specifically, GuD-fed group exhibited severe colitis upon dextran sulfate sodium (DSS) administration, as evidenced by reduced body weight, diarrhea, rectal bleeding, and shortening of colon length compared to cellulose-fed control mice. Elevated levels of pro-inflammatory markers in both serum [serum amyloid A (SAA), lipocalin 2 (Lcn2)] and colon (Lcn2) and extensive disruption of colonic architecture further affirmed that GuD-fed group exhibited more severe colitis than control group upon DSS intervention. Amelioration of colitis in GuD-fed group pre-treated with antibiotics suggest a vital role of intestinal microbiota in GuD-mediated exacerbation of intestinal inflammation. Gut microbiota composition and metabolite analysis in fecal and cecal contents, respectively, revealed that guar gum primarily enriches Actinobacteriota, specifically Bifidobacterium. Guar gum also altered multiple genera belonging to phyla Bacteroidota and Firmicutes. Such shift in gut microbiota composition favored luminal accumulation of intermediary metabolites succinate and lactate in the GuD-fed mice. Colonic IL-18 and tight junction markers were also decreased in the GuD-fed group. Importantly, GuD-fed mice pre-treated with recombinant IL-18 displayed attenuated colitis. Collectively, unfavorable changes in gut microbiota activity leading to luminal accumulation of lactate and succinate, reduced colonic IL-18, and compromised gut barrier function following guar gum feeding contributed to increased colitis susceptibility.
Collapse
Affiliation(s)
- Devendra Paudel
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Divek V. T. Nair
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Sangshan Tian
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Fuhua Hao
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Umesh K. Goand
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Grace Joseph
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Eleni Prodes
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Zhi Chai
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chloé E.M. Robert
- INSERM U1016, team “Mucosal microbiota in chronic inflammatory diseases”, CNRS UMR 8104, Université Paris Cité, Paris, France
- INSERM U1306, Microbiome-Host Interaction group, Institut Pasteur, Université Paris Cité, Paris, France
| | - Benoit Chassaing
- INSERM U1016, team “Mucosal microbiota in chronic inflammatory diseases”, CNRS UMR 8104, Université Paris Cité, Paris, France
- INSERM U1306, Microbiome-Host Interaction group, Institut Pasteur, Université Paris Cité, Paris, France
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Vishal Singh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
14
|
Casado-Bedmar M, Roy M, Berthet L, Hugot JP, Yang C, Manceau H, Peoc'h K, Chassaing B, Merlin D, Viennois E. Fecal let-7b and miR-21 directly modulate the intestinal microbiota, driving chronic inflammation. Gut Microbes 2024; 16:2394249. [PMID: 39224018 PMCID: PMC11376420 DOI: 10.1080/19490976.2024.2394249] [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: 12/07/2023] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Inflammatory bowel diseases (IBD) etiology is multifactorial. Luminal microRNAs (miRNAs) have been suspected to play a role in the promotion of chronic inflammation, but the extent to which fecal miRNAs are interacting with the intestinal ecosystem in a way that contribute to diseases, including IBD, remains unknown. Here, fecal let-7b and miR-21 were found elevated, associated with inflammation, and correlating with multiple bacteria in IBD patients and IL-10-/- mice, model of spontaneous colitis. Using an in vitro microbiota modeling system, we revealed that these two miRNAs can directly modify the composition and function of complex human microbiota, increasing their proinflammatory potential. In vivo investigations revealed that luminal increase of let-7b drastically alters the intestinal microbiota and enhances macrophages' associated proinflammatory cytokines (TNF, IL-6, and IL-1β). Such proinflammatory effects are resilient and dependent on the bacterial presence. Moreover, we identified that besides impairing the intestinal barrier function, miR-21 increases myeloperoxidase and antimicrobial peptides secretion, causing intestinal dysbiosis. More importantly, in vivo inhibition of let-7b and miR-21 with anti-miRNAs significantly improved the intestinal mucosal barrier function and promoted a healthier host-microbiota interaction in the intestinal lining, which altogether conferred protection against colitis. In summary, we provide evidence of the functional significance of fecal miRNAs in host-microbiota communication, highlighting their therapeutic potential in intestinal inflammation and dysbiosis-related conditions, such as IBD.
Collapse
Affiliation(s)
| | - Maryline Roy
- Center for Research on Inflammation, Université Paris Cité, Paris, France
| | - Louis Berthet
- Center for Research on Inflammation, Université Paris Cité, Paris, France
| | - Jean-Pierre Hugot
- Center for Research on Inflammation, Université Paris Cité, Paris, France
- Department of Pediatric Gastroenterology, Hôpital Robert Debré, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Chunhua Yang
- Institute for Biomedical Sciences, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | - Hana Manceau
- Center for Research on Inflammation, Université Paris Cité, Paris, France
- Laboratory of Clinical Biochemistry, Beaujon Hospital, APHP, Clichy, France
| | - Katell Peoc'h
- Center for Research on Inflammation, Université Paris Cité, Paris, France
- Laboratory of Clinical Biochemistry, Beaujon Hospital, APHP, Clichy, France
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- Mucosal Microbiota in Chronic Inflammatory Diseases, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris, France
- CHRU Nancy, IHU Infiny, Nancy, France
| | - Didier Merlin
- Institute for Biomedical Sciences, Center for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
- Veterans Affairs Medical Center, Decatur, GA, USA
| | - Emilie Viennois
- Center for Research on Inflammation, Université Paris Cité, Paris, France
- CHRU Nancy, IHU Infiny, Nancy, France
| |
Collapse
|
15
|
Vasquez Ayala A, Hsu CY, Oles RE, Matsuo K, Loomis LR, Buzun E, Carrillo Terrazas M, Gerner RR, Lu HH, Kim S, Zhang Z, Park JH, Rivaud P, Thomson M, Lu LF, Min B, Chu H. Commensal bacteria promote type I interferon signaling to maintain immune tolerance in mice. J Exp Med 2024; 221:e20230063. [PMID: 38085267 PMCID: PMC10716256 DOI: 10.1084/jem.20230063] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 10/05/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
Type I interferons (IFNs) exert a broad range of biological effects important in coordinating immune responses, which have classically been studied in the context of pathogen clearance. Yet, whether immunomodulatory bacteria operate through IFN pathways to support intestinal immune tolerance remains elusive. Here, we reveal that the commensal bacterium, Bacteroides fragilis, utilizes canonical antiviral pathways to modulate intestinal dendritic cells (DCs) and regulatory T cell (Treg) responses. Specifically, IFN signaling is required for commensal-induced tolerance as IFNAR1-deficient DCs display blunted IL-10 and IL-27 production in response to B. fragilis. We further establish that IFN-driven IL-27 in DCs is critical in shaping the ensuing Foxp3+ Treg via IL-27Rα signaling. Consistent with these findings, single-cell RNA sequencing of gut Tregs demonstrated that colonization with B. fragilis promotes a distinct IFN gene signature in Foxp3+ Tregs during intestinal inflammation. Altogether, our findings demonstrate a critical role of commensal-mediated immune tolerance via tonic type I IFN signaling.
Collapse
Affiliation(s)
| | - Chia-Yun Hsu
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Renee E. Oles
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Kazuhiko Matsuo
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-osaka, Japan
| | - Luke R. Loomis
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Ekaterina Buzun
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | | | - Romana R. Gerner
- TUM School of Life Sciences Weihenstephan, ZIEL Institute for Food & Health, Freising-Weihenstephan, Germany
| | - Hsueh-Han Lu
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
| | - Sohee Kim
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ziyue Zhang
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Jong Hwee Park
- Division of Biology, California Institute of Technology, Pasadena, CA, USA
| | - Paul Rivaud
- Division of Biology, California Institute of Technology, Pasadena, CA, USA
| | - Matt Thomson
- Division of Biology, California Institute of Technology, Pasadena, CA, USA
| | - Li-Fan Lu
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Booki Min
- Department of Microbiology and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hiutung Chu
- Department of Pathology, University of California, San Diego, La Jolla, CA, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines, University of California, San Diego, La Jolla, CA, USA
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, Canada
| |
Collapse
|
16
|
Tao R, Zong G, Pan Y, Li H, Cheng P, Deng R, Chen W, Wang A, Xia S, Tang W, Lu Y, Wei Z. Clostridium butyricum and Clostridium tyrobutyricum: angel or devil for necrotizing enterocolitis? mSystems 2023; 8:e0073223. [PMID: 37921463 PMCID: PMC10734425 DOI: 10.1128/msystems.00732-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/06/2023] [Indexed: 11/04/2023] Open
Abstract
IMPORTANCE This study sheds light on that treatment with Clostridium tyrobutyricum but not Clostridium butyricum is entitled to protect against necrotizing enterocolitis (NEC) development potentially. The mechanisms behind the opposite effect on NEC may result in different modulation on the level of Akkermansia muciniphila, which is deeply associated with intestinal homoeostasis. Briefly, through improving the abundance of A. muciniphila to alleviate intestinal inflammation and enhance intestinal barrier integrity, C. tyrobutyricum supplement may become a promising therapy for NEC.
Collapse
Affiliation(s)
- Ruizhi Tao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Gangfan Zong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yehua Pan
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongxing Li
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Peng Cheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Rui Deng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenxing Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shishan Xia
- Ningbo Women and Children's Hospital, Ningbo, China
| | - Weibing Tang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhonghong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
17
|
de Geus ED, Volaric JS, Matthews AY, Mangan NE, Chang J, Ooi JD, de Weerd NA, Giles EM, Hertzog PJ. Epithelially Restricted Interferon Epsilon Protects Against Colitis. Cell Mol Gastroenterol Hepatol 2023; 17:267-278. [PMID: 37879406 PMCID: PMC10765064 DOI: 10.1016/j.jcmgh.2023.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND & AIMS Type I interferon (T1IFN) signalling is crucial for maintaining intestinal homeostasis. We previously found that the novel T1IFN, IFNε, is highly expressed by epithelial cells of the female reproductive tract, where it protects against pathogens. Its function has not been studied in the intestine. We hypothesize that IFNε is important in maintaining intestinal homeostasis. METHODS We characterized IFNε expression in mouse and human intestine by immunostaining and studied its function in the dextran sulfate sodium (DSS) colitis model using both genetic knockouts and neutralizing antibody. RESULTS We demonstrate that IFNε is expressed in human and mouse intestinal epithelium, and expression is lost in inflammation. Furthermore, we show that IFNε limits intestinal inflammation in mouse models. Regulatory T cell (Treg) frequencies were paradoxically decreased in DSS-treated IFNε-/- mice, suggesting a role for IFNε in maintaining the intestinal Treg compartment. Colitis was ameliorated by transfer of wild-type Tregs into IFNε-/- mice. This demonstrates that IFNε supports intestinal Treg function. CONCLUSIONS Overall, we have shown IFNε expression in intestinal epithelium and its critical role in gut homeostasis. Given its known role in the female reproductive tract, we now show IFNε has a protective role across multiple mucosal surfaces.
Collapse
Affiliation(s)
- Eveline D de Geus
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia.
| | - Jennifer S Volaric
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Antony Y Matthews
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Niamh E Mangan
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Janet Chang
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Joshua D Ooi
- Centre for Inflammatory Diseases, Department of Medicine, Monash Medical Centre, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Nicole A de Weerd
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Edward M Giles
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia; Department of Paediatrics, Monash University, Clayton, VIC, Australia
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
18
|
Masuta Y, Otsuka Y, Minaga K, Honjo H, Kudo M, Watanabe T. Regulation of type I IFN responses by deubiquitinating enzyme A in inflammatory bowel diseases. J Clin Biochem Nutr 2023; 73:103-107. [PMID: 37700847 PMCID: PMC10493212 DOI: 10.3164/jcbn.23-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/21/2023] [Indexed: 09/14/2023] Open
Abstract
The development of Inflammatory bowel disease (IBD) is driven by excessive production of pro-inflammatory cytokines including TNF-α, IL-12, and IL-23. This notion is supported by the remarkable clinical success of biologics targeting these cytokines. Recognition of cell wall components derived from intestinal bacteria by Toll-like receptors (TLRs) induces the production of these pro-inflammatory cytokines by macrophages and dendritic cells in human IBD and experimental colitis model. Although sensing of bacterial nucleic acids by endosomal TLRs, specifically TLR3, TLR7, and TLR9 leads to robust production of type I IFNs, it remains debatable whether TLR-mediated type I IFN responses are pathogenic or protective in IBD patients. Additionally, recent studies identified deubiquitinating enzyme A (DUBA) as a novel negative regulator of TLR-mediated type I IFN responses. In light of these observations and their potential applications, in this review, we summarize recent findings on the roles of type I IFN responses and DUBA-mediated negative regulation of these responses in human IBD and experimental colitis model.
Collapse
Affiliation(s)
- Yasuhiro Masuta
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Yasuo Otsuka
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Kosuke Minaga
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Hajime Honjo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Tomohiro Watanabe
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| |
Collapse
|
19
|
Bretto E, Ribaldone DG, Caviglia GP, Saracco GM, Bugianesi E, Frara S. Inflammatory Bowel Disease: Emerging Therapies and Future Treatment Strategies. Biomedicines 2023; 11:2249. [PMID: 37626745 PMCID: PMC10452708 DOI: 10.3390/biomedicines11082249] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a term used to represent a group of chronic, relapsing inflammatory disorders of the gastrointestinal tract. Crohn's disease (CD) and ulcerative colitis (UC) are the two major clinical forms. The global incidence and prevalence of IBD have increased over the last 2-4 decades. Despite the specific etiopathogenesis of IBD still being unknown, it is widely recognized that immunological, genetic, and environmental factors are implicated. A greater understanding of the multiple signaling pathways involved has led to the development of biologic therapies in the last two decades. Although these treatments have dramatically transformed the course of IBD, there is not a definitive cure and available therapies may cause adverse events (AEs), limiting their use, or have an inadequate effect in some patients. In this context, emerging therapies addressing new specific pathogenetic mechanisms have shown promising efficacy and safety data in early clinical trials. The purpose of this review is to highlight the available clinical trial data for these new drugs, such as more preferential JAK inhibitors, anti-IL-23 antibodies, sphingosine-1-phosphate receptor modulators, anti-integrin therapies, and other small molecules that are currently under research. We will emphasize the potential significance of these agents in shaping future treatment options.
Collapse
Affiliation(s)
- Elisabetta Bretto
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
- Unit of Gastroenterology, Città della Salute e della Scienza di Torino-Molinette Hospital, 10126 Turin, Italy
| | - Davide Giuseppe Ribaldone
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
- Unit of Gastroenterology, Città della Salute e della Scienza di Torino-Molinette Hospital, 10126 Turin, Italy
| | - Gian Paolo Caviglia
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
| | - Giorgio Maria Saracco
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
- Unit of Gastroenterology, Città della Salute e della Scienza di Torino-Molinette Hospital, 10126 Turin, Italy
| | - Elisabetta Bugianesi
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
- Unit of Gastroenterology, Città della Salute e della Scienza di Torino-Molinette Hospital, 10126 Turin, Italy
| | - Simone Frara
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (E.B.)
- Unit of Gastroenterology, Città della Salute e della Scienza di Torino-Molinette Hospital, 10126 Turin, Italy
| |
Collapse
|
20
|
Brabec T, Vobořil M, Schierová D, Valter E, Šplíchalová I, Dobeš J, Březina J, Dobešová M, Aidarova A, Jakubec M, Manning J, Blumberg R, Waisman A, Kolář M, Kubovčiak J, Šrůtková D, Hudcovic T, Schwarzer M, Froňková E, Pinkasová T, Jabandžiev P, Filipp D. IL-17-driven induction of Paneth cell antimicrobial functions protects the host from microbiota dysbiosis and inflammation in the ileum. Mucosal Immunol 2023; 16:373-385. [PMID: 36739089 DOI: 10.1016/j.mucimm.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023]
Abstract
Interleukin (IL)-17 protects epithelial barriers by inducing the secretion of antimicrobial peptides. However, the effect of IL-17 on Paneth cells (PCs), the major producers of antimicrobial peptides in the small intestine, is unclear. Here, we show that the targeted ablation of the IL-17 receptor (IL-17R) in PCs disrupts their antimicrobial functions and decreases the frequency of ileal PCs. These changes become more pronounced after colonization with IL-17 inducing segmented filamentous bacteria. Mice with PCs that lack IL-17R show an increased inflammatory transcriptional profile in the ileum along with the severity of experimentally induced ileitis. These changes are associated with a decrease in the diversity of gut microbiota that induces a severe ileum pathology upon transfer to genetically susceptible mice, which can be prevented by the systemic administration of IL-17a/f in microbiota recipients. In an exploratory analysis of a small cohort of pediatric patients with Crohn's disease, we have found that a portion of these patients exhibits a low number of lysozyme-expressing ileal PCs and a high ileitis severity score, resembling the phenotype of mice with IL-17R-deficient PCs. Our study identifies IL-17R-dependent signaling in PCs as an important mechanism that maintains ileal homeostasis through the prevention of dysbiosis.
Collapse
Affiliation(s)
- Tomáš Brabec
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic; Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Matouš Vobořil
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Dagmar Schierová
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Evgeny Valter
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Iva Šplíchalová
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Dobeš
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Jiří Březina
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martina Dobešová
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Aigerim Aidarova
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Jakubec
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jasper Manning
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Richard Blumberg
- Brigham and Women's Hospital, Gastroenterology Division, Boston, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Michal Kolář
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Kubovčiak
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Dagmar Šrůtková
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Tomáš Hudcovic
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Martin Schwarzer
- Laboratory of Gnotobiology, Institute of Microbiology of the Czech Academy of Sciences, Novy Hradek, Czech Republic
| | - Eva Froňková
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tereza Pinkasová
- Department of Pediatric, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Petr Jabandžiev
- Department of Pediatric, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Dominik Filipp
- Laboratory of Immunobiology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
21
|
Wu D, Wang X, Yang X, Gu L, McGeachy MJ, Liu X. Temporary consumption of western diet trains the immune system to reduce future gut inflammation. iScience 2023; 26:106915. [PMID: 37305694 PMCID: PMC10250831 DOI: 10.1016/j.isci.2023.106915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/23/2023] [Accepted: 05/14/2023] [Indexed: 06/13/2023] Open
Abstract
Urbanization drives the popularity of western diet (WD), which increased burden in metabolic diseases but also in inflammatory diseases. Here, we show continuous WD disrupted the gut barrier, initiating low-grade inflammation and enhancing the colitis response. Nevertheless, transient WD consumption followed by ad libitum normal diet enhanced mucin production and tight junction protein expression in recovered mice. Furthermore, transient WD consumption surprisingly reduced the subsequent inflammatory response in DSS colitis and Citrobacter rodentium-infection induced colitis. The protective effect of WD training was not sex-dependent, and co-housing experiments suggested microbiota changes were not responsible. We identified important roles for cholesterol biosynthesis pathway and macrophages, pointing to innate myeloid training. Together, these data suggest detrimental effects of WD consumption can be reversed on return to a healthier diet. Furthermore, transient WD consumption leads to beneficial immune training, suggesting an evolutionary mechanism to benefit from feasting when abundant food is available.
Collapse
Affiliation(s)
- Dongwen Wu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Xiaotong Wang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Xiang Yang
- Changsha Aier Eye Hospital, Changsha, Hunan, China
| | - Lei Gu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Mandy J. McGeachy
- Department of Microbiology & Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Xiaowei Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
22
|
Li Q, Li S, Yao Y, Ma Z, Huang C. MIA mice exhibit enteric nerve defects and are more susceptible to dextran sulfate sodium-induced colitis. Brain Behav Immun 2023:S0889-1591(23)00158-7. [PMID: 37315701 DOI: 10.1016/j.bbi.2023.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/15/2023] [Accepted: 06/10/2023] [Indexed: 06/16/2023] Open
Abstract
Maternal immune activation (MIA) during pregnancy impairs the development of the central nervous system as well as the peripheral nervous system. Emerging evidence indicates that individuals with MIA suffer more from gastrointestinal disorders. The present study aims to test the hypothesis that MIA-induced susceptibility to inflammatory bowel disease is due to defects in the innervation of mucosal sensory nerves. Acute dextran sulfate sodium (DSS) colitis was induced in MIA and control adult mice. Body weight loss, disease activity index and colonic histological changes were measured during colitis. The study found that MIA mice were hypersusceptible to DSS-induced colitis and that macrophage infiltration and cytokine production were elevated in the colon of MIA mice. In vitro experiments also demonstrated that colonic macrophages from MIA mice presented hyperinflammatory responses to LPS stimulation. Sensory nerve-secreted calcitonin gene-related peptide (CGRP) is an important neuropeptide in modulating enteric inflammation. Intriguingly, we found that CGRP-positive nerves were sparsely distributed in the colon of MIA mice regardless of DSS treatment. And the protein level of CGRP was significantly reduced in colon of MIA mice. However, there was no decrease in the number of CGRP-positive cell bodies in either the DRG or vagal ganglion, suggesting that innervation defects of CGRP mucosal sensory nerves exist in the colon of MIA mice. Critically, administration of recombinant CGRP to MIA mice during DSS colitis significantly reversed their hyperinflammatory pathology. Additionally, the hyperinflammatory phenotype of colonic macrophages of MIA mice could also be reversed by CGRP treatment in vitro. Collectively, these findings suggested that the sensor nerve innervation defect-induced CGRP deficiency in MIA mice participates in their increased susceptibility to colitis. Thus, sensor nerve-secreted CGRP may be a new therapeutic target for autism combined with inflammatory bowel disease.
Collapse
Affiliation(s)
- Qian Li
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| | - Shuang Li
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yiwei Yao
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zhongxiang Ma
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chutian Huang
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
23
|
Thapa B, Pak S, Chung D, Shin HK, Lee SH, Lee K. Cell-penetrating TLR inhibitor peptide alleviates ulcerative colitis by the functional modulation of macrophages. Front Immunol 2023; 14:1165667. [PMID: 37215126 PMCID: PMC10196052 DOI: 10.3389/fimmu.2023.1165667] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Toll-like receptors (TLRs) have a crucial role not only in triggering innate responses against microbes but in orchestrating an appropriate adaptive immunity. However, deregulated activation of TLR signaling leads to chronic inflammatory conditions such as inflammatory bowel disease (IBD). In this study, we evaluated the immunomodulatory potential of a TLR inhibitor in the form of a cell-penetrating peptide using an ulcerative colitis animal model. A peptide derived from the TIR domain of the TLR adaptor molecule TIRAP that was conjugated with a cell-penetrating sequence (cpTLR-i) suppressed the induction of pro-inflammatory cytokines such as TNF-α and IL-1β in macrophages. In DSS-induced colitis mice, cpTLR-i treatment ameliorated colitis symptoms, colonic tissue damage, and mucosal inflammation. Intriguingly, cpTLR-i attenuated the induction of TNF-α-expressing proinflammatory macrophages while promoting that of regulatory macrophages expressing arginase-1 and reduced type 17 helper T cell (Th17) responses in the inflamed colonic lamina propria. An in vitro study validated that cpTLR-i enhanced the differentiation of monocyte-driven macrophages into mature macrophages with a regulatory phenotype in a microbial TLR ligand-independent manner. Furthermore, the cocultivation of CD4 T cells with macrophages revealed that cpTLR-i suppressed the activation of Th17 cells through the functional modulation of macrophages. Taken together, our data show the immunomodulatory potential of the TLR inhibitor peptide and suggest cpTLR-i as a novel therapeutic candidate for the treatment of IBD.
Collapse
Affiliation(s)
- Bikash Thapa
- Institute of Bioscience & Biotechnology, Hallym University, Chuncheon, Republic of Korea
| | - Seongwon Pak
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
| | - Dohyeon Chung
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
| | | | - Seong Ho Lee
- R&D Center, Genesen Co., Ltd, Seoul, Republic of Korea
| | - Keunwook Lee
- Institute of Bioscience & Biotechnology, Hallym University, Chuncheon, Republic of Korea
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
| |
Collapse
|
24
|
Metwaly A, Jovic J, Waldschmitt N, Khaloian S, Heimes H, Häcker D, Ahmed M, Hammoudi N, Le Bourhis L, Mayorgas A, Siebert K, Basic M, Schwerd T, Allez M, Panes J, Salas A, Bleich A, Zeissig S, Schnupf P, Cominelli F, Haller D. Diet prevents the expansion of segmented filamentous bacteria and ileo-colonic inflammation in a model of Crohn's disease. MICROBIOME 2023; 11:66. [PMID: 37004103 PMCID: PMC10064692 DOI: 10.1186/s40168-023-01508-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 02/25/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Crohn's disease (CD) is associated with changes in the microbiota, and murine models of CD-like ileo-colonic inflammation depend on the presence of microbial triggers. Increased abundance of unknown Clostridiales and the microscopic detection of filamentous structures close to the epithelium of Tnf ΔARE mice, a mouse model of CD-like ileitis pointed towards segmented filamentous bacteria (SFB), a commensal mucosal adherent bacterium involved in ileal inflammation. RESULTS We show that the abundance of SFB strongly correlates with the severity of CD-like ileal inflammation in two mouse models of ileal inflammation, including Tnf ΔARE and SAMP/Yit mice. SFB mono-colonization of germ-free Tnf ΔARE mice confirmed the causal link and resulted in severe ileo-colonic inflammation, characterized by elevated tissue levels of Tnf and Il-17A, neutrophil infiltration and loss of Paneth and goblet cell function. Co-colonization of SFB in human-microbiota associated Tnf ΔARE mice confirmed that SFB presence is indispensable for disease development. Screening of 468 ileal and colonic mucosal biopsies from adult and pediatric IBD patients, using previously published and newly designed human SFB-specific primer sets, showed no presence of SFB in human tissue samples, suggesting a species-specific functionality of the pathobiont. Simulating the human relevant therapeutic effect of exclusive enteral nutrition (EEN), EEN-like purified diet antagonized SFB colonization and prevented disease development in Tnf ΔARE mice, providing functional evidence for the protective mechanism of diet in modulating microbiota-dependent inflammation in IBD. CONCLUSIONS We identified a novel pathogenic role of SFB in driving severe CD-like ileo-colonic inflammation characterized by loss of Paneth and goblet cell functions in Tnf ΔARE mice. A purified diet antagonized SFB colonization and prevented disease development in Tnf ΔARE mice in contrast to a fiber-containing chow diet, clearly demonstrating the important role of diet in modulating a novel IBD-relevant pathobiont and supporting a direct link between diet and microbial communities in mediating protective functions. Video Abstract.
Collapse
Affiliation(s)
- Amira Metwaly
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Jelena Jovic
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Nadine Waldschmitt
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Sevana Khaloian
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Helena Heimes
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Deborah Häcker
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Mohamed Ahmed
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany
| | - Nassim Hammoudi
- APHP, Hôpital Saint Louis, Department of Gastroenterology, INSERM UMRS 1160, Paris Diderot, Sorbonne Paris-Cité University, Paris, France
- Université Paris Cité, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France
| | - Lionel Le Bourhis
- Université Paris Cité, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France
| | - Aida Mayorgas
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona CSIC, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Kolja Siebert
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Marijana Basic
- Hannover Medical School, Institute for Laboratory Animal Science, Hannover, Germany
| | - Tobias Schwerd
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Matthieu Allez
- APHP, Hôpital Saint Louis, Department of Gastroenterology, INSERM UMRS 1160, Paris Diderot, Sorbonne Paris-Cité University, Paris, France
- Université Paris Cité, INSERM U1160, EMiLy, Institut de Recherche Saint-Louis, Paris, France
| | - Julian Panes
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona CSIC, IDIBAPS, CIBERehd, Barcelona, Spain
| | - Azucena Salas
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona CSIC, IDIBAPS, CIBERehd, Barcelona, Spain
| | - André Bleich
- Hannover Medical School, Institute for Laboratory Animal Science, Hannover, Germany
| | - Sebastian Zeissig
- Department of Medicine I, University Hospital Dresden, Technische Universität (TU) Dresden, Dresden, Germany
| | - Pamela Schnupf
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015, Paris, France
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Dirk Haller
- Chair of Nutrition and Immunology, Technical University of Munich, Freising, Germany.
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany.
| |
Collapse
|
25
|
Menchaca-Tapia PA, Marín-Rosales M, Salazar-Camarena DC, Cruz A, Oregon-Romero E, Tapia-Llanos R, Muñoz-Valle JF, Palafox-Sánchez CA. Analysis of PTPN22 -1123 G>C, +788 G>A and +1858 C>T Polymorphisms in Patients with Primary Sjögren's Syndrome. Diagnostics (Basel) 2023; 13:diagnostics13050899. [PMID: 36900045 PMCID: PMC10001387 DOI: 10.3390/diagnostics13050899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/01/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Primary Sjögren's syndrome (pSS) is an autoimmune exocrinopathy characterized by lymphocytic infiltration, glandular dysfunction and systemic manifestations. Lyp protein is a negative regulator of the T cell receptor encoded by the tyrosine phosphatase nonreceptor-type 22 (PTPN22) gene. Multiple single-nucleotide polymorphisms (SNPs) in the PTPN22 gene have been associated with susceptibility to autoimmune diseases. This study aimed to investigate the association of PTPN22 SNPs rs2488457 (-1123 G>C), rs33996649 (+788 G>A), rs2476601 (+1858 C>T) with pSS susceptibility in Mexican mestizo subjects. METHODS One hundred fifty pSS patients and 180 healthy controls (HCs) were included. Genotypes of PTPN22 SNPs were identified by PCR-RFLP. PTPN22 expression was evaluated through RT-PCR analysis. Serum anti-SSA/Ro and anti-SSB/La levels were measured using an ELISA kit. RESULTS Allele and genotype frequencies for all SNPs studied were similar in both groups (p > 0.05). pSS patients showed 17-fold higher expression of PTNP22 than HCs, and mRNA levels correlated with SSDAI score (r2 = 0.499, p = 0.008) and levels of anti-SSA/Ro and anti-SSB/La autoantibodies (r2 = 0.200, p = 0.03 and r2 = 0.175, p = 0.04, respectively). Positive anti-SSA/Ro pSS patients expressed higher PTPN22 mRNA levels (p = 0.008), with high focus scores by histopathology (p = 0.02). Moreover, PTPN22 expression had high diagnostic accuracy in pSS patients, with an AUC = 0.985. CONCLUSIONS Our findings demonstrate that the PTPN22 SNPs rs2488457 (-1123 G>C), rs33996649 (+788 G>A) and rs2476601 (+1858 C>T) are not associated with the disease susceptibility in the western Mexican population. Additionally, PTPN22 expression may be helpful as a diagnostic biomarker in pSS.
Collapse
Affiliation(s)
- Paula Annahi Menchaca-Tapia
- Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Miguel Marín-Rosales
- Servicio de Reumatología, Hospital General de Occidente, Secretaria de Salud Jalisco, Guadalajara 45170, Mexico
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Diana Celeste Salazar-Camarena
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Alvaro Cruz
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Edith Oregon-Romero
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Raziel Tapia-Llanos
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - José Francisco Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Claudia Azucena Palafox-Sánchez
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
- Grupo de Inmunología Molecular, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
- Correspondence: ; Tel.: +52-33-3815-0611
| |
Collapse
|
26
|
Yim HCH, Chakrabarti A, Kessler S, Morimoto H, Wang D, Sooraj D, Ahmed AU, de la Motte C, Silverman RH, Williams BRG, Sadler AJ. The protein kinase R modifies gut physiology to limit colitis. Front Immunol 2023; 14:1106737. [PMID: 36875104 PMCID: PMC9981792 DOI: 10.3389/fimmu.2023.1106737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/03/2023] [Indexed: 02/19/2023] Open
Abstract
Here we investigate the function of the innate immune molecule protein kinase R (PKR) in intestinal inflammation. To model a colitogenic role of PKR, we determine the physiological response to dextran sulfate sodium (DSS) of wild-type and two transgenic mice strains mutated to express either a kinase-dead PKR or to ablate expression of the kinase. These experiments recognize kinase-dependent and -independent protection from DSS-induced weight loss and inflammation, against a kinase-dependent increase in the susceptibility to DSS-induced injury. We propose these effects arise through PKR-dependent alteration of gut physiology, evidenced as altered goblet cell function and changes to the gut microbiota at homeostasis that suppresses inflammasome activity by controlling autophagy. These findings establish that PKR functions as both a protein kinase and a signaling molecule in instituting immune homeostasis in the gut.
Collapse
Affiliation(s)
- Howard Chi Ho Yim
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Arindam Chakrabarti
- Department of Cancer Biology, Lerner Research Institute, Cleveland, OH, United States
| | - Sean Kessler
- Department of Pathobiology, Lerner Research Institute, Cleveland, OH, United States
| | - Hiroyuki Morimoto
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Anatomy, School of Medicine, the University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - Die Wang
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Dhanya Sooraj
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Afsar U. Ahmed
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Carol de la Motte
- Department of Pathobiology, Lerner Research Institute, Cleveland, OH, United States
| | - Robert H. Silverman
- Department of Cancer Biology, Lerner Research Institute, Cleveland, OH, United States
| | - Bryan RG. Williams
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Anthony J. Sadler
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
| |
Collapse
|
27
|
Cheng P, Wu J, Zong G, Wang F, Deng R, Tao R, Qian C, Shan Y, Wang A, Zhao Y, Wei Z, Lu Y. Capsaicin shapes gut microbiota and pre-metastatic niche to facilitate cancer metastasis to liver. Pharmacol Res 2023; 188:106643. [PMID: 36608780 DOI: 10.1016/j.phrs.2022.106643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/18/2022] [Accepted: 12/31/2022] [Indexed: 01/05/2023]
Abstract
Dietary factors are fundamental in tumorigenesis throughout our lifetime. A spicy diet has been ambiguous on the development of cancers, especially in the study of colon cancer metastasis. Here, we utilized a mouse metastasis model to test the potential role of capsaicin in influencing metastasis. Long-term continuous administration of capsaicin diet (300 mg/kg) to mice promotes the formation of liver pre-metastatic niche to facilitate the metastasis of colon cancer cells. Bacteria translocation to liver is clearly observed. Capsaicin increases intestinal barrier permeability and disrupts gut vascular barrier by altering the composition of gut microbiota. Capsaicin not only changes the abundance of mucin-related bacteria like Akkermanisa and Muribaculaceae, but also bacteria involved in bile acids metabolism. Dysregulated bile acids profile is related to the recruitment of natural killer T (NKT) cells in pre-metastatic niche, primary bile acid α-Muricholic acid can enhance the recruitment of NKT cells, while secondary bile acids Glycoursodeoxycholic acid and Taurohyodeoxycholic acid impair the recruitment of NKT cells. These findings reveal long term consumption of capsaicin increases the risk of cancer metastasis through modulating the gut microbiota. Capsaicin (300 mg/kg) disrupts gut barrier and promotes the translocation of bacteria to liver, while altered bile acids metabolism affects the recruitment of NKT cells in liver, forming a pre-metastatic niche and promoting cancer metastasis.
Collapse
Affiliation(s)
- Peng Cheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiawei Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Gangfan Zong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feihui Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Rui Deng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ruizhi Tao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Cheng Qian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yang Zhao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhonghong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
28
|
Choi Y, Choi SI, Kim N, Nam RH, Jang JY, Na HY, Shin CM, Lee DH, Min H, Kim YR, Seok YJ. Effect of Clostridium butyricum on High-Fat Diet-Induced Intestinal Inflammation and Production of Short-Chain Fatty Acids. Dig Dis Sci 2023; 68:2427-2440. [PMID: 36670324 DOI: 10.1007/s10620-023-07835-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/09/2023] [Indexed: 01/22/2023]
Abstract
BACKGROUND/AIMS A high-fat diet (HFD) can cause intestinal inflammation and alter the gut microbiota; probiotics, however, are known to have anti-inflammatory effects. This study aimed to investigate the response of rat colon to HFD and the effect of Clostridium butyricum on HFD-induced intestinal inflammation and production of short-chain fatty acids (SCFAs) according to sex. METHODS Male and female 6-week-old Fischer-344 rats were fed a chow diet or HFD for 8 weeks, and Biovita or three different concentrations of C. butyricum were orally gavaged. The levels of tight junction proteins (TJPs), inflammatory markers in the ascending colonic mucosa, and bile acids (BAs) and SCFAs in stool were measured. RESULTS HFD significantly increased the histological inflammation scores and fat proportions. Fecal BA levels were higher in the HFD group than in the control group, with a more prominent increase in deoxycholic acid/cholic acid after probiotics administration in females; however, no statistically significant differences were observed. TJPs showed an opposite response to HFD depending on sex, and tended to increase and decrease after HFD in males and females, respectively. The HFD-reduced TJPs were recovered by probiotics, with some statistical significance in females. HFD-decreased butyric acid in stools appeared to be recovered by probiotics in males, but not in females. The expression of inflammatory markers (TNF-α) was increased by HFD in males and decreased with medium-concentration probiotic supplementation. The opposite was observed in females. MPO was increased by HFD in both sexes and decreased by probiotic supplementation. CONCLUSIONS The probiotic C. butyricum improved indicators of HFD-induced colonic inflammation such as levels of inflammatory markers and increased the production of SCFAs and the expression of TJPs. These effects tended to be more pronounced in male rats, showing sex difference.
Collapse
Affiliation(s)
- Yonghoon Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Soo In Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea. .,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jae Young Jang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hee Young Na
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Huitae Min
- Department of Biological Sciences and Institute of Microbiology, Seoul National University, Seoul, South Korea
| | - Yeon-Ran Kim
- Department of Biological Sciences and Institute of Microbiology, Seoul National University, Seoul, South Korea
| | - Yeong-Jae Seok
- Department of Biological Sciences and Institute of Microbiology, Seoul National University, Seoul, South Korea
| |
Collapse
|
29
|
Song CH, Kim N, Nam RH, Choi SI, Jang JY, Lee HN. Changes in Gut Microbiome upon Orchiectomy and Testosterone Administration in AOM/DSS-Induced Colon Cancer Mouse Model. Cancer Res Treat 2023; 55:196-218. [PMID: 35790194 PMCID: PMC9873319 DOI: 10.4143/crt.2022.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/30/2022] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Sex hormones are known to affect the gut microbiota. Previously, we reported that endogenous and exogenous testosterone are associated with colorectal cancer (CRC) development and submucosal invasion. In the present study, we investigated whether the gut microbiota is affected by orchiectomy (ORX) and testosterone propionate (TP) administration using an azoxymethane/dextran sulfate sodium (AOM/DSS)-induced CRC mouse model. MATERIALS AND METHODS Gut microbiota was evaluated by means of 16S rRNA gene sequencing of stool DNA extracted from feces that were obtained at 13 weeks after AOM injection (from 22-week-old animals) and stored in a gas-generating pouch. RESULTS The increase in microbial diversity (Chao1 and Phylogenetic Diversity index) and Firmicutes/Bacteroidetes (F/B) ratio upon AOM/DSS treatment in ORX mice was significantly decreased by TP supplementation. The ratio of commensal bacteria to opportunistic pathogens was lower in the TP-administered females and ORX mice than in the AOM/DSS group. Opportunistic pathogens (Mucispirillum schaedleri or Akkermansia muciniphila) were identified only in the TP group. In addition, microbial diversity and F/B ratio were higher in male controls than in female and ORX controls. Flintibacter butyricus, Ruminococcus bromii, and Romboutsia timonensis showed similar changes in the male control group as those in the female and ORX controls. CONCLUSION In conclusion, testosterone determines the dysbiosis of gut microbiota, which suggests that it plays a role in the sex-related differences in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Chin-Hee Song
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam,
Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam,
Korea,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul,
Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam,
Korea
| | - Soo In Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam,
Korea
| | - Jae Young Jang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam,
Korea
| | - Ha-Na Lee
- Laboratory of Immunology, Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD,
USA
| |
Collapse
|
30
|
Kennel KB, Burmeister J, Radhakrishnan P, Giese NA, Giese T, Salfenmoser M, Gebhardt JM, Strowitzki MJ, Taylor CT, Wielockx B, Schneider M, Harnoss JM. The HIF-prolyl hydroxylases have distinct and nonredundant roles in colitis-associated cancer. JCI Insight 2022; 7:153337. [PMID: 36509284 DOI: 10.1172/jci.insight.153337] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/11/2022] [Indexed: 11/22/2022] Open
Abstract
Colitis-associated colorectal cancer (CAC) is a severe complication of inflammatory bowel disease (IBD). HIF-prolyl hydroxylases (PHD1, PHD2, and PHD3) control cellular adaptation to hypoxia and are considered promising therapeutic targets in IBD. However, their relevance in the pathogenesis of CAC remains elusive. We induced CAC in Phd1-/-, Phd2+/-, Phd3-/-, and WT mice with azoxymethane (AOM) and dextran sodium sulfate (DSS). Phd1-/- mice were protected against chronic colitis and displayed diminished CAC growth compared with WT mice. In Phd3-/- mice, colitis activity and CAC growth remained unaltered. In Phd2+/- mice, colitis activity was unaffected, but CAC growth was aggravated. Mechanistically, Phd2 deficiency (i) increased the number of tumor-associated macrophages in AOM/DSS-induced tumors, (ii) promoted the expression of EGFR ligand epiregulin in macrophages, and (iii) augmented the signal transducer and activator of transcription 3 and extracellular signal-regulated kinase 1/2 signaling, which at least in part contributed to aggravated tumor cell proliferation in colitis-associated tumors. Consistently, Phd2 deficiency in hematopoietic (Vav:Cre-Phd2fl/fl) but not in intestinal epithelial cells (Villin:Cre-Phd2fl/fl) increased CAC growth. In conclusion, the 3 different PHD isoenzymes have distinct and nonredundant effects, promoting (PHD1), diminishing (PHD2), or neutral (PHD3), on CAC growth.
Collapse
Affiliation(s)
- Kilian B Kennel
- Department of General, Visceral and Transplantation Surgery and
| | | | | | | | - Thomas Giese
- Institute of Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | | | - Cormac T Taylor
- School of Medicine, Systems Biology Ireland, and the Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Dresden University of Technology, Dresden, Germany
| | | | | |
Collapse
|
31
|
Wang MY, Wang ZX, Huang LJ, Yang RX, Zou ZY, Ge WS, Ren TY, Fan JG. Premorbid Steatohepatitis Increases the Seriousness of Dextran Sulfate Sodium-induced Ulcerative Colitis in Mice. J Clin Transl Hepatol 2022; 10:847-859. [PMID: 36304494 PMCID: PMC9547267 DOI: 10.14218/jcth.2021.00315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/13/2021] [Accepted: 11/30/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS The concurrence of nonalcoholic steatohepatitis (NASH) and ulcerative colitis (UC) is increasingly seen in clinical practice, but the underlying mechanisms remain unclear. This study aimed to develop a mouse model of the phenomenon by combining high-fat high-cholesterol diet (HFHCD)-induced NASH and dextran sulfate sodium (DSS)-induced UC, that would support mechanistic studies. METHODS Male C57BL/6 mice were randomly assigned to two groups receiving either a chow diet or HFHCD for 12 weeks of NASH modeling. The mice were the divided into four subgroups for UC modeling: (1) A control group given a chow diet with normal drinking water; (2) A colitis group given chow diet with 2% DSS in drinking water; (3) A steatohepatitis group given HFHCD with normal drinking water; and (4) A steatohepatitis + colitis group given HFHCD with 2% DSS in drinking water. RESULTS NASH plus UC had high mortality (58.3%). Neither NASH nor UC alone were fatal. Although DSS-induced colitis did not exacerbate histological liver injury in HFHCD-fed mice, premorbid NASH significantly increased UC-related gut injury compared with UC alone. It was characterized by a significantly shorter colon, more colonic congestion, and a higher histopathological score (p<0.05). Inflammatory (tumor necrosis factor-alpha, interleukin 1 beta, C-C motif chemokine ligand 2, and nuclear factor kappa B) and apoptotic (Bcl2, Bad, Bim, and Bax) signaling pathways were significantly altered in distal colon tissues collected from mice with steatohepatitis + colitis compared with the other experimental groups. CONCLUSIONS Premorbid steatohepatitis significantly aggravated DSS-induced colitis and brought about a lethal phenotype. Potential links between NASH and UC pathogeneses can be investigated using this model.
Collapse
Affiliation(s)
- Meng-Yu Wang
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zi-Xuan Wang
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei-Jie Huang
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui-Xu Yang
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zi-Yuan Zou
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Song Ge
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian-Yi Ren
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Correspondence to: Jian-Gao Fan, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China. ORCID: https://orcid.org/0000-0002-8618-6402. Tel: +86-21-25077340, E-mail: ; Tian-Yi Ren, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China. ORCID: https://orcid.org/0000-0002-0844-7072. Tel: +86-18204314931, E-mail:
| | - Jian-Gao Fan
- Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai, China
- Correspondence to: Jian-Gao Fan, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China. ORCID: https://orcid.org/0000-0002-8618-6402. Tel: +86-21-25077340, E-mail: ; Tian-Yi Ren, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China. ORCID: https://orcid.org/0000-0002-0844-7072. Tel: +86-18204314931, E-mail:
| |
Collapse
|
32
|
The intestinal clock drives the microbiome to maintain gastrointestinal homeostasis. Nat Commun 2022; 13:6068. [PMID: 36241650 PMCID: PMC9568547 DOI: 10.1038/s41467-022-33609-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 09/23/2022] [Indexed: 12/24/2022] Open
Abstract
Diurnal (i.e., 24-hour) oscillations of the gut microbiome have been described in various species including mice and humans. However, the driving force behind these rhythms remains less clear. In this study, we differentiate between endogenous and exogenous time cues driving microbial rhythms. Our results demonstrate that fecal microbial oscillations are maintained in mice kept in the absence of light, supporting a role of the host's circadian system rather than representing a diurnal response to environmental changes. Intestinal epithelial cell-specific ablation of the core clock gene Bmal1 disrupts rhythmicity of microbiota. Targeted metabolomics functionally link intestinal clock-controlled bacteria to microbial-derived products, in particular branched-chain fatty acids and secondary bile acids. Microbiota transfer from intestinal clock-deficient mice into germ-free mice altered intestinal gene expression, enhanced lymphoid organ weights and suppressed immune cell recruitment. These results highlight the importance of functional intestinal clocks for microbiota composition and function, which is required to balance the host's gastrointestinal homeostasis.
Collapse
|
33
|
Adamkova P, Hradicka P, Kupcova Skalnikova H, Cizkova V, Vodicka P, Farkasova Iannaccone S, Kassayova M, Gancarcikova S, Demeckova V. Dextran Sulphate Sodium Acute Colitis Rat Model: A Suitable Tool for Advancing Our Understanding of Immune and Microbial Mechanisms in the Pathogenesis of Inflammatory Bowel Disease. Vet Sci 2022; 9:238. [PMID: 35622766 PMCID: PMC9147231 DOI: 10.3390/vetsci9050238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 01/27/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a group of disorders causing inflammation in the digestive tract. Recent data suggest that dysbiosis may play a pivotal role in the IBD pathogenesis. As microbiome-based therapeutics that modulate the gut ecology have been proposed as a novel strategy for preventing IBD, the aim of presenting study was to evaluate the dextran sulphate sodium (DSS) rat model mainly in terms of microbial shifts to confirm its suitability for dysbiosis study in IBD. Acute colitis was induced using 5% DSS solution for seven days and rats were euthanized five days after DSS removal. The faecal/caecal microbiota was analyzed by next generation sequencing. Disease activity index (DAI) score was evaluated daily. Blood and colon tissue immunophenotyping was assessed by flow cytometry and histological, haematological, and biochemical parameters were also evaluated. The colitis induction was reflected in a significantly higher DAI score and changes in all parameters measured. This study demonstrated significant shifts in the colitis-related microbial species after colitis induction. The characteristic inflammation-associated microbiota could be detected even after a five day-recovery period. Moreover, the DSS-model might contribute to an understanding of the effect of different treatments on extraintestinal organ impairments. The observation that certain bacterial species in the gut microbiota are associated with colitis raises the question of whether these organisms are contributors to, or a consequence of the disease. Despite some limitations, we confirmed the suitability of DSS-induced colitis model to monitor microbial changes during acute colitis, in order to test attractive new microbiome-based therapies.
Collapse
Affiliation(s)
- Petra Adamkova
- Faculty of Science, Institute of Biology and Ecology, Pavol Jozef Safarik University in Kosice, 041 54 Kosice, Slovakia; (P.A.); (P.H.); (M.K.)
| | - Petra Hradicka
- Faculty of Science, Institute of Biology and Ecology, Pavol Jozef Safarik University in Kosice, 041 54 Kosice, Slovakia; (P.A.); (P.H.); (M.K.)
| | - Helena Kupcova Skalnikova
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 277 21 Libechov, Czech Republic; (H.K.S.); (V.C.); (P.V.)
| | - Veronika Cizkova
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 277 21 Libechov, Czech Republic; (H.K.S.); (V.C.); (P.V.)
| | - Petr Vodicka
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, 277 21 Libechov, Czech Republic; (H.K.S.); (V.C.); (P.V.)
| | - Silvia Farkasova Iannaccone
- Department of Forensic Medicine, Faculty of Medicine, Pavol Jozef Safarik University in Kosice, 040 11 Kosice, Slovakia;
| | - Monika Kassayova
- Faculty of Science, Institute of Biology and Ecology, Pavol Jozef Safarik University in Kosice, 041 54 Kosice, Slovakia; (P.A.); (P.H.); (M.K.)
| | - Sona Gancarcikova
- Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Kosice, 041 81 Kosice, Slovakia;
| | - Vlasta Demeckova
- Faculty of Science, Institute of Biology and Ecology, Pavol Jozef Safarik University in Kosice, 041 54 Kosice, Slovakia; (P.A.); (P.H.); (M.K.)
| |
Collapse
|
34
|
Experimental Intestinal Stenosis Alters Crohn's Disease-Like Intestinal Inflammation in Ileitis-Prone Mice. Dig Dis Sci 2022; 67:1783-1793. [PMID: 34350516 DOI: 10.1007/s10620-021-07161-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 06/08/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Clinical observations indicate that mechanical factors contribute to the expression or recurrence of Crohn's disease. We investigated whether the creation of an intestinal stenosis could alter the severity of the expected Crohn-like ileitis, in a Crohn's disease animal model, the TNFΔare/+ mouse. METHODS Thirty-six, 6-weeks-old TNFΔare/+ mice, were divided into 3 intervention groups: triple suture, single suture and sham. In the terminal ileum, in the first group, a triple suture stenosis was created, whereas, in the second, a loose suture was placed. Same triple-suture stenosis was performed on twelve wild type mice. All animals were sacrificed at 6 weeks post-operatively and the ileum parts were evaluated histopathologically. A summative total ileitis score was applied in each sample using a bespoke semiquantitative histological scoring system for the Crohn-like changes. RESULTS The triple suture stenosis induced significant muscular hypertrophy proximal to interventional site which was more prominent in TNFΔare/+ than wild type mice. In triple suture group, the total ileitis score was significantly increased proximal to the intervention as compared to the single suture (P: 0.004) and the sham groups (P: 0.013). The total ileitis score distally, was unaffected, regardless of the experimental intervention. Intestinal stenosis did not induce intestinal inflammation in wild type mice. CONCLUSION The creation of a stenosis in the terminal ileum of TNFΔare/+ mice alters Crohn-like inflammation. We assume that mechanical forces, such as intraluminal pressure, may contribute as important co-factors to the pathophysiology of Crohn's disease in genetically predisposed subjects.
Collapse
|
35
|
Khan S, Mentrup HL, Novak EA, Siow VS, Wang Q, Crawford EC, Schneider C, Comerford TE, Firek B, Rogers MB, Loughran P, Morowitz MJ, Mollen KP. Cyclic GMP-AMP synthase contributes to epithelial homeostasis in intestinal inflammation via Beclin-1-mediated autophagy. FASEB J 2022; 36:e22282. [PMID: 35344224 PMCID: PMC9040047 DOI: 10.1096/fj.202200138r] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/04/2022] [Accepted: 03/15/2022] [Indexed: 12/29/2022]
Abstract
Inflammatory bowel disease (IBD) represents a set of idiopathic and chronic inflammatory diseases of the gastrointestinal tract. Central to the pathogenesis of IBD is a dysregulation of normal intestinal epithelial homeostasis. cGAS is a DNA-sensing receptor demonstrated to promote autophagy, a mechanism that removes dysfunctional cellular components. Beclin-1 is a crucial protein involved in the initiation of autophagy. We hypothesized that cGAS plays a key role in intestinal homeostasis by upregulating Beclin-1-mediated autophagy. We evaluated intestinal cGAS levels in humans with IBD and in murine colonic tissue after performing a 2% dextran sulfate sodium (DSS) colitis model. Autophagy and cell death mechanisms were studied in cGAS KO and WT mice via qPCR, WB analysis, H&E, IF, and TUNEL staining. Autophagy was measured in stimulated intestinal epithelial cells (IECs) via WB analysis. Our data demonstrates cGAS to be upregulated during human and murine colitis. Furthermore, cGAS deficiency leads to worsened colitis and decreased levels of autophagy proteins including Beclin-1 and LC3-II. Co-IP demonstrates a direct binding between cGAS and Beclin-1 in IECs. Transfection of cGAS in stimulated HCT-116 cells leads to increased autophagy. IECs isolated from cGAS KO have diminished autophagic flux. cGAS KO mice subjected to DSS have increased cell death and cleaved caspase-3. Lastly, treatment of cGAS KO mice with rapamycin decreased the severity of colitis. Our data suggest that cGAS maintains intestinal epithelial homeostasis during human IBD and murine colitis by upregulating Beclin-1-mediated autophagy and preventing IEC death. Rescue of autophagy can attenuate the severity of colitis associated with cGAS deficiency.
Collapse
Affiliation(s)
- Sidrah Khan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Heather L Mentrup
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Division of Pediatric Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Elizabeth A Novak
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Division of Pediatric Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Vei Shaun Siow
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Qian Wang
- Department of Pathology, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Erin C Crawford
- Division of Gastroenterology, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Corinne Schneider
- Division of Pediatric Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Thomas E Comerford
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Brian Firek
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Division of Pediatric Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Matt B Rogers
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Division of Pediatric Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael J Morowitz
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Division of Pediatric Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Kevin P Mollen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Division of Pediatric Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
36
|
Park Y, Kim T, Ham J, Choi J, Lee H, Yeon YJ, Choi SI, Kim N, Kim Y, Seok Y. Physiological activity of E. coli engineered to produce butyric acid. Microb Biotechnol 2022; 15:832-843. [PMID: 33729711 PMCID: PMC8913873 DOI: 10.1111/1751-7915.13795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/10/2021] [Accepted: 02/23/2021] [Indexed: 11/29/2022] Open
Abstract
Faecalibacterium prausnitzii (F. prausnitzii) is one of the most abundant bacteria in the human intestine, with its anti-inflammatory effects establishing it as a major effector in human intestinal health. However, its extreme sensitivity to oxygen makes its cultivation and physiological study difficult. F. prausnitzii produces butyric acid, which is beneficial to human gut health. Butyric acid is a short-chain fatty acid (SCFA) produced by the fermentation of carbohydrates, such as dietary fibre in the large bowel. The genes encoding butyryl-CoA dehydrogenase (BCD) and butyryl-CoA:acetate CoA transferase (BUT) in F. prausnitzii were cloned and expressed in E. coli to determine the effect of butyric acid production on intestinal health using DSS-induced colitis model mice. The results from the E. coli Nissle 1917 strain, expressing BCD, BUT, or both, showed that BCD was essential, while BUT was dispensable for producing butyric acid. The effects of different carbon sources, such as glucose, N-acetylglucosamine (NAG), N-acetylgalactosamine (NAGA), and inulin, were compared with results showing that the optimal carbon sources for butyric acid production were NAG, a major component of mucin in the human intestine, and glucose. Furthermore, the anti-inflammatory effects of butyric acid production were tested by administering these strains to DSS-induced colitis model mice. The oral administration of the E. coli Nissle 1917 strain, carrying the expression vector for BCD and BUT (EcN-BCD-BUT), was found to prevent DSS-induced damage. Introduction of the BCD expression vector into E. coli Nissle 1917 led to increased butyric acid production, which improved the strain's health-beneficial effects.
Collapse
Affiliation(s)
- Young‐Tae Park
- Department of Biological Sciences and Institute of MicrobiologySeoul National UniversitySeoulKorea
- Korea Institute of Science and Technology Natural Products Research InstituteGangneungKorea
| | - Taejung Kim
- Korea Institute of Science and Technology Natural Products Research InstituteGangneungKorea
| | - Jungyeob Ham
- Korea Institute of Science and Technology Natural Products Research InstituteGangneungKorea
| | - Jaeyoung Choi
- Korea Institute of Science and Technology Green City Technology InstituteSeoulKorea
| | - Hoe‐Suk Lee
- Department of Biochemical EngineeringGangneung‐Wonju National UniversityGangneungKorea
| | - Young Joo Yeon
- Department of Biochemical EngineeringGangneung‐Wonju National UniversityGangneungKorea
| | - Soo In Choi
- Seoul National University Bundang HospitalSeongnamKorea
| | - Nayoung Kim
- Seoul National University Bundang HospitalSeongnamKorea
| | - Yeon‐Ran Kim
- Department of Biological Sciences and Institute of MicrobiologySeoul National UniversitySeoulKorea
| | - Yeong‐Jae Seok
- Department of Biological Sciences and Institute of MicrobiologySeoul National UniversitySeoulKorea
| |
Collapse
|
37
|
Xu J, Li X, Du Y. Antibody-Pattern Recognition Receptor Agonist Conjugates: A Promising Therapeutic Strategy for Cancer. Adv Biol (Weinh) 2022; 6:e2101065. [PMID: 35122418 DOI: 10.1002/adbi.202101065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/27/2021] [Indexed: 01/15/2023]
Abstract
Antibody-drug conjugates (ADCs) are composed of monoclonal antibodies linked to cytotoxic payload drugs, each of which can be diversely designed in accordance with pharmacological and clinical requirements. The use of ADCs is effective for the treatment of different diseases, including cancers, and is gaining widespread attention. To date, 12 ADCs have been approved by the U.S. Food and Drug Administration for treating cancer and improving the quality of life of patients. To expand the application of ADCs and improve their treatment efficiency, various formats have recently been manufactured, including pattern recognition receptor (PRR) agonist-based ADCs. The antibody has a unique structure that enables the specific delivery of PRR agonists to the tumor area, and this improves the therapeutic efficacy while minimizing systemic toxicity. This review briefly discusses the current landscape and future perspectives of antibody-PRR agonist conjugates for cancer therapy.
Collapse
Affiliation(s)
- Jian Xu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiang Li
- Beijing Kawin Technology Share-Holding Co., Ltd, BDA, Beijing, 100176, China
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| |
Collapse
|
38
|
Mills RH, Dulai PS, Vázquez-Baeza Y, Sauceda C, Daniel N, Gerner RR, Batachari LE, Malfavon M, Zhu Q, Weldon K, Humphrey G, Carrillo-Terrazas M, Goldasich LD, Bryant M, Raffatellu M, Quinn RA, Gewirtz AT, Chassaing B, Chu H, Sandborn WJ, Dorrestein PC, Knight R, Gonzalez DJ. Multi-omics analyses of the ulcerative colitis gut microbiome link Bacteroides vulgatus proteases with disease severity. Nat Microbiol 2022; 7:262-276. [PMID: 35087228 PMCID: PMC8852248 DOI: 10.1038/s41564-021-01050-3] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022]
Abstract
Ulcerative colitis (UC) is driven by disruptions in host-microbiota homoeostasis, but current treatments exclusively target host inflammatory pathways. To understand how host-microbiota interactions become disrupted in UC, we collected and analysed six faecal- or serum-based omic datasets (metaproteomic, metabolomic, metagenomic, metapeptidomic and amplicon sequencing profiles of faecal samples and proteomic profiles of serum samples) from 40 UC patients at a single inflammatory bowel disease centre, as well as various clinical, endoscopic and histologic measures of disease activity. A validation cohort of 210 samples (73 UC, 117 Crohn's disease, 20 healthy controls) was collected and analysed separately and independently. Data integration across both cohorts showed that a subset of the clinically active UC patients had an overabundance of proteases that originated from the bacterium Bacteroides vulgatus. To test whether B. vulgatus proteases contribute to UC disease activity, we first profiled B. vulgatus proteases found in patients and bacterial cultures. Use of a broad-spectrum protease inhibitor improved B. vulgatus-induced barrier dysfunction in vitro, and prevented colitis in B. vulgatus monocolonized, IL10-deficient mice. Furthermore, transplantation of faeces from UC patients with a high abundance of B. vulgatus proteases into germfree mice induced colitis dependent on protease activity. These results, stemming from a multi-omics approach, improve understanding of functional microbiota alterations that drive UC and provide a resource for identifying other pathways that could be inhibited as a strategy to treat this disease.
Collapse
Affiliation(s)
- Robert H Mills
- Department of Pharmacology, University of California, San Diego, CA, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, USA.,Department of Pediatrics, University of California, San Diego, CA, USA
| | - Parambir S Dulai
- Division of Gastroenterology, University of California, San Diego, CA, USA
| | - Yoshiki Vázquez-Baeza
- Department of Pediatrics, University of California, San Diego, CA, USA.,Department of Computer Science and Engineering, University of California, San Diego, CA, USA.,Center for Microbiome Innovation, University of California, San Diego, CA, USA
| | - Consuelo Sauceda
- Department of Pharmacology, University of California, San Diego, CA, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, USA
| | - Noëmie Daniel
- INSERM U1016, team Mucosal microbiota in chronic inflammatory diseases, CNRS UMR 8104, Université de Paris, Paris, France
| | - Romana R Gerner
- Department of Pediatrics, University of California, San Diego, CA, USA.,Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, CA, USA
| | | | - Mario Malfavon
- Department of Pharmacology, University of California, San Diego, CA, USA
| | - Qiyun Zhu
- Department of Pediatrics, University of California, San Diego, CA, USA.,School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Kelly Weldon
- Center for Microbiome Innovation, University of California, San Diego, CA, USA
| | - Greg Humphrey
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - Marvic Carrillo-Terrazas
- Department of Pharmacology, University of California, San Diego, CA, USA.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, USA.,Department of Pathology, University of California, San Diego, CA, USA
| | | | - MacKenzie Bryant
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - Manuela Raffatellu
- Center for Microbiome Innovation, University of California, San Diego, CA, USA.,Division of Host-Microbe Systems and Therapeutics, University of California, San Diego, CA, USA
| | - Robert A Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Benoit Chassaing
- INSERM U1016, team Mucosal microbiota in chronic inflammatory diseases, CNRS UMR 8104, Université de Paris, Paris, France
| | - Hiutung Chu
- Department of Pathology, University of California, San Diego, CA, USA
| | - William J Sandborn
- Division of Gastroenterology, University of California, San Diego, CA, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, USA.,Department of Pediatrics, University of California, San Diego, CA, USA.,Center for Microbiome Innovation, University of California, San Diego, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego, CA, USA. .,Department of Computer Science and Engineering, University of California, San Diego, CA, USA. .,Center for Microbiome Innovation, University of California, San Diego, CA, USA.
| | - David J Gonzalez
- Department of Pharmacology, University of California, San Diego, CA, USA. .,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, USA. .,Center for Microbiome Innovation, University of California, San Diego, CA, USA.
| |
Collapse
|
39
|
Deng F, Wu Z, Xu M, Xia P. YAP Activates STAT3 Signalling to Promote Colonic Epithelial Cell Proliferation in DSS-Induced Colitis and Colitis Associated Cancer. J Inflamm Res 2022; 15:5471-5482. [PMID: 36164660 PMCID: PMC9508680 DOI: 10.2147/jir.s377077] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/15/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND AIMS Yes-associated protein (YAP) is a key transcriptional coactivator of cell proliferation and differentiation. In this study, we sought to identify the roles of YAP in colonic epithelial regeneration and tumourigenesis. METHODS Murine DSS-induced colitis and YAP overexpression models were constructed via lentiviral intraperitoneal injection. Stable YAP-overexpressing cells, protein immunoprecipitation, and ChIP were used to deeply explore the molecular mechanism. RESULTS We found that the expression of YAP was dramatically diminished in the colonic crypts during the acute colitis phase, while YAP was strikingly enhanced to initiate tissue repair after DSS withdrawal. Overexpressing YAP in mice drastically accelerated epithelial regeneration, presenting with more intact structural integrity and reduced inflammatory cell infiltration in the mucosa. Further mechanistic studies showed that the expression of YAP in the nucleus was significantly increased by 2 h post-DSS removal, accompanied by upregulated protein levels of activated STAT3. Overexpression of YAP (YAPWT) elevated the expression of activated STAT3 and its transcriptional targets and strengthened the proliferation and "wound healing" ability of colonic cells. However, these effects were reversed when STAT3 was silenced in YAPWT cells. Moreover, YAP could directly interact with STAT3 in the nucleus, and c-Myc and CyclinD1 were the transcriptional targets. Finally, during colitis-associated cancer (CAC), YAPWT promoted the progression of CAC, while the phosphomimetic YAP downregulated the expression of STAT3 and inhibited the development and progression of CAC. CONCLUSION YAP activates STAT3 signalling to facilitate mucosal regeneration after DSS-induced colitis. However, excessive YAP activation in the colonic epithelium promotes CAC development.
Collapse
Affiliation(s)
- Feihong Deng
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
- Correspondence: Feihong Deng, Department of Gastroenterology, the Second Xiangya Hospital of Central South University; Research Center of Digestive Disease, Central South University, Changsha, Hunan, 410011, People’s Republic of China, Email
| | - Zengrong Wu
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
| | - Mengmeng Xu
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
| | - Pianpian Xia
- Department of Gastroenterology, the Second Xiangya Hospital of Central South University, Changsha, 410011, People’s Republic of China
- Research Center of Digestive Disease, Central South University, Changsha, 410011, People’s Republic of China
| |
Collapse
|
40
|
Krammer S, Sicorschi Gutu C, Grund JC, Chiriac MT, Zirlik S, Finotto S. Regulation and Function of Interferon-Lambda (IFNλ) and Its Receptor in Asthma. Front Immunol 2021; 12:731807. [PMID: 34899691 PMCID: PMC8660125 DOI: 10.3389/fimmu.2021.731807] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/12/2021] [Indexed: 12/22/2022] Open
Abstract
Asthma is a chronic respiratory disease affecting people of all ages, especially children, worldwide. Origins of asthma are suggested to be placed in early life with heterogeneous clinical presentation, severity and pathophysiology. Exacerbations of asthma disease can be triggered by many factors, including viral respiratory tract infections. Rhinovirus (RV) induced respiratory infections are the predominant cause of the common cold and also play a crucial role in asthma development and exacerbations. Rhinovirus mainly replicates in epithelial cells lining the upper and lower respiratory tract. Type III interferons, also known as interferon-lambda (IFNλ), are potent immune mediators of resolution of infectious diseases but they are known to be involved in autoimmune diseases as well. The protective role of type III IFNs in antiviral, antibacterial, antifungal and antiprotozoal functions is of major importance for our innate immune system. The IFNλ receptor (IFNλR) is expressed in selected types of cells like epithelial cells, thus orchestrating a specific immune response at the site of viruses and bacteria entry into the body. In asthma, IFNλ restricts the development of TH2 cells, which are induced in the airways of asthmatic patients. Several studies described type III IFNs as the predominant type of interferon increased after infection caused by respiratory viruses. It efficiently reduces viral replication, viral spread into the lungs and viral transmission from infected to naive individuals. Several reports showed that bronchial epithelial cells from asthmatic subjects have a deficient response of type III interferon after RV infection ex vivo. Toll like Receptors (TLRs) recognize pathogen-associated molecular patterns (PAMPs) expressed on infectious agents, and induce the development of antiviral and antibacterial immunity. We recently discovered that activation of TLR7/8 resulted in enhanced IFNλ receptor mRNA expression in PBMCs of healthy and asthmatic children, opening new therapeutic frontiers for rhinovirus-induced asthma. This article reviews the recent advances of the literature on the regulated expression of type III Interferons and their receptor in association with rhinovirus infection in asthmatic subjects.
Collapse
Affiliation(s)
- Susanne Krammer
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Cristina Sicorschi Gutu
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Janina C Grund
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mircea T Chiriac
- Medical Clinic 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sabine Zirlik
- Medical Clinic 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany.,Medical Clinic 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
41
|
Zhou Y, Duan L, Zeng Y, Niu L, Pu Y, Jacobs JP, Chang C, Wang J, Khalique A, Pan K, Fang J, Jing B, Zeng D, Ni X. The Panda-Derived Lactobacillus plantarum G201683 Alleviates the Inflammatory Response in DSS-Induced Panda Microbiota-Associated Mice. Front Immunol 2021; 12:747045. [PMID: 34956180 PMCID: PMC8692892 DOI: 10.3389/fimmu.2021.747045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/15/2021] [Indexed: 11/23/2022] Open
Abstract
Intestinal diseases are one of the main causes of captive giant panda death. Their special dietary habits and gastrointestinal tract structure often lead to intestinal epithelium damage and secondary intestinal infection. The captive giant panda is predisposed to suffer from microbiota dysbiosis due to long-term artificial feeding and antibiotic misuse. However, there are few reported probiotics to treat giant panda enteritis and the associated dysbiosis. This study aims to elucidate the mechanism by which Lactobacillus plantarum G201683 (L. plantarum G83), a promising panda-derived probiotic, exerts a protective effect on intestinal inflammation in the dextran sulfate sodium- (DSS) induced panda microbiota-associated (DPMA) mouse model. The DPMA mouse was generated by antibiotic treatment and 5% DSS drinking water administration to assess the effect of L. plantarum G83 on intestinal inflammation and microbiota in vivo. Our results demonstrated the successful generation of a DPMA mouse model with Enterobacteriaceae enrichment, consistent with the giant panda intestinal microbiota. L. plantarum G83 decreased clinical and histological severity of intestinal inflammation, enhanced intestinal tight junction protein expression (ZO-1, Occludin) and alleviated inflammatory cytokine production (TNF-) in the colon of DPMA mice. The administration of L. plantarum G83 altered the microbiota composition by decreasing pathogen associated taxa such as E. coli and increasing abundance of beneficial bacteria including Bifidobacterium spp. These changes in microbiota composition were associated with an increased concentration of short chain fatty acids (SCFA), reduced NF-κB signaling, and an altered balance of T helper cell subsets. Our findings support L. plantarum G83 as a promising probiotic to treat intestinal inflammation in the giant panda.
Collapse
Affiliation(s)
- Yi Zhou
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ling Duan
- Central Station of Animal Feed Affairs of Sichuan Province, Sichuan Provincial Department of Agriculture and Rural Affairs, Chengdu, China
| | - Yan Zeng
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lili Niu
- Chengdu Wildlife Institute, Chengdu Zoo, Chengdu, China
| | - Yang Pu
- Chengdu Wildlife Institute, Chengdu Zoo, Chengdu, China
| | - Jonathan P. Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Candace Chang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles (UCLA), Los Angeles, CA, United States
| | - Jie Wang
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Abdul Khalique
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Kangcheng Pan
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Jing Fang
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bo Jing
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Dong Zeng
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xueqin Ni
- Animal Microecology Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
42
|
Ntunzwenimana JC, Boucher G, Paquette J, Gosselin H, Alikashani A, Morin N, Beauchamp C, Thauvette L, Rivard MÈ, Dupuis F, Deschênes S, Foisy S, Latour F, Lavallée G, Daly MJ, Xavier RJ, Charron G, Goyette P, Rioux JD. Functional screen of inflammatory bowel disease genes reveals key epithelial functions. Genome Med 2021; 13:181. [PMID: 34758847 PMCID: PMC8582123 DOI: 10.1186/s13073-021-00996-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/21/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Genetic studies have been tremendously successful in identifying genomic regions associated with a wide variety of phenotypes, although the success of these studies in identifying causal genes, their variants, and their functional impacts has been more limited. METHODS We identified 145 genes from IBD-associated genomic loci having endogenous expression within the intestinal epithelial cell compartment. We evaluated the impact of lentiviral transfer of the open reading frame (ORF) of these IBD genes into the HT-29 intestinal epithelial cell line via transcriptomic analyses. By comparing the genes in which expression was modulated by each ORF, as well as the functions enriched within these gene lists, we identified ORFs with shared impacts and their putative disease-relevant biological functions. RESULTS Analysis of the transcriptomic data for cell lines expressing the ORFs for known causal genes such as HNF4a, IFIH1, and SMAD3 identified functions consistent with what is already known for these genes. These analyses also identified two major clusters of genes: Cluster 1 contained the known IBD causal genes IFIH1, SBNO2, NFKB1, and NOD2, as well as genes from other IBD loci (ZFP36L1, IRF1, GIGYF1, OTUD3, AIRE and PITX1), whereas Cluster 2 contained the known causal gene KSR1 and implicated DUSP16 from another IBD locus. Our analyses highlight how multiple IBD gene candidates can impact on epithelial structure and function, including the protection of the mucosa from intestinal microbiota, and demonstrate that DUSP16 acts a regulator of MAPK activity and contributes to mucosal defense, in part via its regulation of the polymeric immunoglobulin receptor, involved in the protection of the intestinal mucosa from enteric microbiota. CONCLUSIONS This functional screen, based on expressing IBD genes within an appropriate cellular context, in this instance intestinal epithelial cells, resulted in changes to the cell's transcriptome that are relevant to their endogenous biological function(s). This not only helped in identifying likely causal genes within genetic loci but also provided insight into their biological functions. Furthermore, this work has highlighted the central role of intestinal epithelial cells in IBD pathophysiology, providing a scientific rationale for a drug development strategy that targets epithelial functions in addition to the current therapies targeting immune functions.
Collapse
Affiliation(s)
- Jessy Carol Ntunzwenimana
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Gabrielle Boucher
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Jean Paquette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Hugues Gosselin
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Azadeh Alikashani
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Nicolas Morin
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Claudine Beauchamp
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Louise Thauvette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Marie-Ève Rivard
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Frédérique Dupuis
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Sonia Deschênes
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Sylvain Foisy
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Frédéric Latour
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Geneviève Lavallée
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Mark J Daly
- Massachusetts General Hospital, Boston, MA, USA
- The Broad Institute, Cambridge, MA, USA
| | - Ramnik J Xavier
- Massachusetts General Hospital, Boston, MA, USA
- The Broad Institute, Cambridge, MA, USA
| | - Guy Charron
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - Philippe Goyette
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada
| | - John D Rioux
- Montreal Heart Institute Research Centre, 5000 rue Bélanger, S-6201, Montreal, Quebec, Canada.
- Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
43
|
Lu J, Liu D, Tan Y, Li R, Wang X, Deng F. Thalidomide Attenuates Colitis and Is Associated with the Suppression of M1 Macrophage Polarization by Targeting the Transcription Factor IRF5. Dig Dis Sci 2021; 66:3803-3812. [PMID: 34085173 DOI: 10.1007/s10620-021-07067-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 05/17/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory bowel disease. The TNF-α inhibitor thalidomide is reported to be effective for inducing remission in pediatric Crohn's disease (CD) and adults with refractory CD. The mechanisms underlying the immunomodulatory and anti-inflammatory properties of thalidomide are unclear. METHODS Histological assessments were firstly performed in thalidomide treated UC patients. Then the effect of thalidomide in vivo was detected in DSS-induced murine colitis. The mechanism involving IRF5, and M1 macrophage polarization was investigated by using plasmid transfection, western blotting, and real-time PCR. Finally, AOM/DSS model was used to detect the role of thalidomide in colitis associated cancer. RESULTS We first found that treatment with thalidomide could ameliorate colon inflammation for 8 weeks and promote mucosal healing in human UC. Moreover, treatment with thalidomide protected mice from dextran sodium sulfate (DSS)-induced acute colitis, with treated mice presenting with a higher body weight, lower histological score, and lower DAI. Concomitantly, in comparison with control mice, mice treated with thalidomide showed accelerated recovery following colitis after 10 days of thalidomide treatment. Mechanistically, we observed that thalidomide could increase epithelial cell self-renewal capacity and modulate M1/M2 polarization by decreasing M1 markers CD86 and CCR7 and increasing M2 protein signatures CD206 and Arg-1. Thalidomide controls M1 macrophage polarization by targeting the transcription factor IRF5. Finally, by using the classical AOM/DSS model, we found that thalidomide-treated mice presented with a lower incidence and growth of colitis-associated carcinoma (CAC) than negative control mice. CONCLUSIONS In summary, thalidomide suppresses M1 polarization in the inflammatory microenvironment, which not only attenuates colonic inflammation to facilitate mucosal healing after DSS-induced injury but also represses the progression of CAC.
Collapse
Affiliation(s)
- Jiaxi Lu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Center of Digestive Disease, Central South University, Changsha, 410011, Hunan, China
| | - Deliang Liu
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Center of Digestive Disease, Central South University, Changsha, 410011, Hunan, China
| | - Yuyong Tan
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Center of Digestive Disease, Central South University, Changsha, 410011, Hunan, China
| | - Rong Li
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Center of Digestive Disease, Central South University, Changsha, 410011, Hunan, China
| | - Xuehong Wang
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.,Research Center of Digestive Disease, Central South University, Changsha, 410011, Hunan, China
| | - Feihong Deng
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China. .,Research Center of Digestive Disease, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
44
|
Li J, Yu S, Pan X, Zhang M, Lv Z, Pan LL, Sun J. Recombinant CRAMP-producing Lactococcus lactis attenuates dextran sulfate sodium-induced colitis by colonic colonization and inhibiting p38/NF-κB signaling. Food Nutr Res 2021; 65:5570. [PMID: 34650393 PMCID: PMC8494263 DOI: 10.29219/fnr.v65.5570] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/05/2021] [Accepted: 01/28/2021] [Indexed: 11/23/2022] Open
Abstract
Background Inflammatory bowel diseases (IBDs) are generally characterized by persistent abdominal pain and diarrhea caused by chronic inflammation in the intestine. Cathelicidins are antimicrobial peptides with pleiotropic roles in anti-infection, wound healing, and immune modulation. However, the sensitivity to the acidic environment and short half-life of cathelicidins limit their application in IBD treatment. Recombinant cathelicidin-related antimicrobial peptide (CRAMP)-producing Lactococcus lactis may represent a potential approach for IBD therapy. Objective The aim of this study was to develop recombinant CRAMP-producing L. lactis NZ9000 and explore the role and mechanism of recombinant L. lactis NZ9000 expressing CRAMP in colitis. Design We constructed two strains of CRAMP-producing L. lactis NZ9000 with different plasmids pMG36e (L.L-pMU45CR) or pNZ8148 (L.L-pNU45CR), which use a Usp45 secretion signal to drive the secretion of CRAMP. Bacterial suspensions were orally supplemented to mice with a syringe for 4 days after dextran sodium sulfate (DSS) treatment. Body weight change, disease active score, colon length, and colonic histology were determined. The expression of tight junction (ZO-1, ZO-2, and Occludin) and cytokines (IL-6, IL-1β, TNF-α, and IL-10) in colon was performed by qPCR. The expression of p-ERK, p-p38, and p-p65 was determined by Western blot analysis. Results Both CRAMP-producing L. lactis NZ9000 strains protected against colitis, as shown by reduced weight loss and disease activity score, improved colon shortening, and histopathological injury. In addition, CRAMP-producing L. lactis NZ9000 restored gut barrier by upregulating ZO-1, ZO-2, and occludin. Moreover, CRAMP-producing L. lactis NZ9000 regulated the colonic cytokines profile with reduced IL-6, IL-1β, and TNF-α production, and increased IL-10 production. By further analysis, we found that CRAMP-producing L. lactis NZ9000 reduced the expression of p-p38 and p-p65. Conclusions Together, our data suggested that CRAMP-secreting L. lactis NZ9000 attenuated dextran sulfate sodium-induced colitis by colonic colonization and inhibiting p38/NF-κB signaling. Orally administered recombinant CRAMP-secreting L. lactis NZ9000 represents a potential strategy for colitis therapy.
Collapse
Affiliation(s)
- Jiahong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Shiwen Yu
- Department of Obstetrics, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, P. R. China
| | - Xiaohua Pan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Ming Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Zhuwu Lv
- Department of Obstetrics, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, P. R. China
| | - Li-Long Pan
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| |
Collapse
|
45
|
Li J, Pan X, Ren Z, Li B, Liu H, Wu C, Dong X, de Vos P, Pan LL, Sun J. Protein arginine methyltransferase 2 (PRMT2) promotes dextran sulfate sodium-induced colitis by inhibiting the SOCS3 promoter via histone H3R8 asymmetric dimethylation. Br J Pharmacol 2021; 179:141-158. [PMID: 34599829 DOI: 10.1111/bph.15695] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE There is emerging evidence for a critical role for epigenetic modifiers in the development of inflammatory bowel disease (IBD). Protein arginine methyltransferase 2 (PRMT2) is responsible for the methylation of arginine residues on histones and targets transcription factors involved in many cellular processes, including gene transcription, mRNA splicing, cell proliferation, and cell differentiation. In this study, the role and underlying mechanisms of PRMT2 in colitis were studied. EXPERIMENTAL APPROACH A mouse dextran sulfate sodium (DSS)-induced experimental colitis model was used to study PRMT2 in colitis. Lentivirus-induced PRMT2 silencing or overexpression in vivo was applied to address the role of PRMT2 in colitis. Detailed western blot and expression analysis were done to understand epigenetic changes induced by PRMT2 in colitis. KEY RESULTS PRMT2 is highly expressed in inflammatory bowel disease patients, in inflamed murine colon and in TNF-α stimulated murine gut epithelial cells. PRMT2 overexpression aggravates, while knockdown alleviates DSS-induced colitis, suggesting that PRMT2 is a pivotal mediator of colitis in mice. Mechanistically, PRMT2 mediates colitis by increasing repressive histone mark H3R8 asymmetric methylation (H3R8me2a) at the promoter region of the suppressor of cytokine signalling 3 promoter (SOCS3). Resultant inhibition of SOCS3 expression and inhibition of SOCS3-mediated degradation of TNF receptor associated factor 5 (TRAF5) via ubiquitination led to elevated TRAF5 expression and TRAF5-mediated downstream NF-κB/MAPK activation. CONCLUSION AND IMPLICATIONS Our study demonstrates that PRMT2 acts as a transcriptional co-activator for proinflammatory genes during colitis. Hence, targeting PRMT2 may provide a novel therapeutic approach for colitis.
Collapse
Affiliation(s)
- Jiahong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xiaohua Pan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Zhengnan Ren
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Binbin Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - He Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Chengfei Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xiaoliang Dong
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Li-Long Pan
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| |
Collapse
|
46
|
Liu CY, Cham CM, Chang EB. Epithelial wound healing in inflammatory bowel diseases: the next therapeutic frontier. Transl Res 2021; 236:35-51. [PMID: 34126257 PMCID: PMC8380699 DOI: 10.1016/j.trsl.2021.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/25/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023]
Abstract
Patients with one of the many chronic inflammatory disorders broadly classified as inflammatory bowel disease (IBD) now have a diverse set of immunomodulatory therapies at their disposal. Despite these recent medical advances, complete sustained remission of disease remains elusive for most patients. The full healing of the damaged intestinal mucosa is the primary goal of all therapies. Achieving this requires not just a reduction of the aberrant immunological response, but also wound healing of the epithelium. No currently approved therapy directly targets the epithelium. Epithelial repair is compromised in IBD and normally facilitates re-establishment of the homeostatic barrier between the host and the microbiome. In this review, we summarize the evidence that epithelial wound healing represents an important yet underdeveloped therapeutic modality for IBD. We highlight 3 general approaches that are promising for developing a new class of epithelium-targeted therapies: epithelial stem cells, cytokines, and microbiome engineering. We also provide a frank discussion of some of the challenges that must be overcome for epithelial repair to be therapeutically leveraged. A concerted approach by the field to develop new therapies targeting epithelial wound healing will offer patients a game-changing, complementary class of medications and could dramatically improve outcomes.
Collapse
Affiliation(s)
- Cambrian Y Liu
- Department of Medicine, The University of Chicago, Chicago, Illinois.
| | - Candace M Cham
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Eugene B Chang
- Department of Medicine, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
47
|
Zhang Y, Liu W, Zhang D, Yang Y, Wang X, Li L. Fermented and Germinated Processing Improved the Protective Effects of Foxtail Millet Whole Grain Against Dextran Sulfate Sodium-Induced Acute Ulcerative Colitis and Gut Microbiota Dysbiosis in C57BL/6 Mice. Front Nutr 2021; 8:694936. [PMID: 34395495 PMCID: PMC8358663 DOI: 10.3389/fnut.2021.694936] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/07/2021] [Indexed: 12/30/2022] Open
Abstract
This study investigated the effects of foxtail millet whole grain flours obtained through different processing methods on alleviating symptoms and gut microbiota dysbiosis in a dextran sulfate sodium (DSS)-induced murine colitis model. Sixty C57BL/6 mice were divided into six groups (n = 10 in each group), including one control group (CTRL) without DSS treatment and five DSS-treated groups receiving one of the following diets: AIN-93M standard diet (93MD), whole grain foxtail millet flour (FM), fermented (F-FM), germinated (G-FM), and fermented-germinated foxtail millet flour (FG-FM). A comparison of the disease activity index (DAI) demonstrated that foxtail millet whole grain-based diets could alleviate the symptoms of enteritis to varying degrees. In addition, 16S rRNA gene sequencing revealed that FG-FM almost completely alleviated DSS-induced dysbiosis. Mice on the FG-FM diet also had the lowest plasma IL-6 levels and claudin2 expression levels in the colon, indicating reduced systemic inflammation and improved gut barrier function. This study suggested that foxtail millet whole grain is an attractive choice for the intervention of IBD and gut microbiota dysbiosis, and its prebiotic properties are highly affected by the processing methods.
Collapse
Affiliation(s)
- Yuhan Zhang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China.,Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China.,Qilu Hospital, Shandong University, Jinan, China
| | - Wei Liu
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Di Zhang
- Qilu Hospital, Shandong University, Jinan, China
| | - Yanbing Yang
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Xianshu Wang
- Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Lingfei Li
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
48
|
Haring E, Zeiser R, Apostolova P. Interfering With Inflammation: Heterogeneous Effects of Interferons in Graft- Versus-Host Disease of the Gastrointestinal Tract and Inflammatory Bowel Disease. Front Immunol 2021; 12:705342. [PMID: 34249014 PMCID: PMC8264264 DOI: 10.3389/fimmu.2021.705342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
The intestine can be the target of several immunologically mediated diseases, including graft-versus-host disease (GVHD) and inflammatory bowel disease (IBD). GVHD is a life-threatening complication that occurs after allogeneic hematopoietic stem cell transplantation. Involvement of the gastrointestinal tract is associated with a particularly high mortality. GVHD development starts with the recognition of allo-antigens in the recipient by the donor immune system, which elicits immune-mediated damage of otherwise healthy tissues. IBD describes a group of immunologically mediated chronic inflammatory diseases of the intestine. Several aspects, including genetic predisposition and immune dysregulation, are responsible for the development of IBD, with Crohn’s disease and ulcerative colitis being the two most common variants. GVHD and IBD share multiple key features of their onset and development, including intestinal tissue damage and loss of intestinal barrier function. A further common feature in the pathophysiology of both diseases is the involvement of cytokines such as type I and II interferons (IFNs), amongst others. IFNs are a family of protein mediators produced as a part of the inflammatory response, typically to pathogens or malignant cells. Diverse, and partially paradoxical, effects have been described for IFNs in GVHD and IBD. This review summarizes current knowledge on the role of type I, II and III IFNs, including basic concepts and controversies about their functions in the context of GVHD and IBD. In addition, therapeutic options, research developments and remaining open questions are addressed.
Collapse
Affiliation(s)
- Eileen Haring
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Petya Apostolova
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
49
|
Song CH, Kim N, Nam RH, Choi SI, Yu JE, Nho H, Shin E, Lee HN, Surh YJ. Testosterone strongly enhances azoxymethane/dextran sulfate sodium-induced colorectal cancer development in C57BL/6 mice. Am J Cancer Res 2021; 11:3145-3162. [PMID: 34249451 PMCID: PMC8263677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/25/2021] [Indexed: 06/13/2023] Open
Abstract
Colorectal cancer (CRC) is known to occur more frequently in males than in females, with sex hormones reportedly influencing the development. The purpose of the study was to investigate whether orchiectomy in C57BL/6 male mice reduces colorectal tumorigenesis and whether testosterone administration increases tumorigenesis after orchiectomy in an azoxymethane (AOM)/dextran sulfate sodium (DSS) mouse model. Clinical symptoms, including colitis and tumor incidence, were evaluated in the absence or presence of testosterone in AOM/DSS-treated male, as well as orchiectomized (ORX) male and female mice. The levels of serum testosterone and colonic myeloperoxidase, interleukin (IL)-1β, and IL-6 were measured by ELISA. Target mRNA expression was assessed by quantitative real-time PCR. Orchiectomy significantly diminished the AOM/DSS-induced colitis indices, including disease activity index, colon shortening, and histological severity at week 2, and decreased tumor numbers and incidence rates in the distal part of the colon increased following AOM/DSS administration at week 13; this reduction was reversed by testosterone supplementation. Furthermore, it was confirmed that the ELISA level (MPO and IL-1β) and the mRNA expression of the inflammatory mediators (COX-2 and iNOS) were maintained at high levels in the tumors of the testosterone-treated group compared with AOM/DSS groups. Interestingly, both endogenous and exogenous testosterone administrations were associated with tumor development (> 2 mm in size) and submucosal invasive cancer. Based on multivariate logistic regression analysis, testosterone was identified as a reasonable hazard factor for the progression of submucosal invasive cancer of the distal colon. In conclusion, endogenous and exogenous testosterone presented a stimulating effect on AOM/DSS-induced colitis and carcinogenicity.
Collapse
Affiliation(s)
- Chin-Hee Song
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnam, Gyeonggi-do, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnam, Gyeonggi-do, South Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of MedicineSeoul, South Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnam, Gyeonggi-do, South Korea
| | - Soo In Choi
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnam, Gyeonggi-do, South Korea
| | - Jeong Eun Yu
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnam, Gyeonggi-do, South Korea
| | - Heewon Nho
- Department of Internal Medicine, Seoul National University Bundang HospitalSeongnam, Gyeonggi-do, South Korea
| | - Eun Shin
- Department of Pathology, Hallym University Dongtan Sacred Heart HospitalHwaseong, Gyeonggi-do, South Korea
| | - Ha-Na Lee
- Laboratory of Immunology, Division of Biotechnology Review and Research-III, Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug AdministrationSilver Spring, MD 20993, USA
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center, Seoul National University College of PharmacySeoul, South Korea
- Cancer Research Institute, Seoul National UniversitySeoul, South Korea
| |
Collapse
|
50
|
McElrath C, Espinosa V, Lin JD, Peng J, Sridhar R, Dutta O, Tseng HC, Smirnov SV, Risman H, Sandoval MJ, Davra V, Chang YJ, Pollack BP, Birge RB, Galan M, Rivera A, Durbin JE, Kotenko SV. Critical role of interferons in gastrointestinal injury repair. Nat Commun 2021; 12:2624. [PMID: 33976143 PMCID: PMC8113246 DOI: 10.1038/s41467-021-22928-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
The etiology of ulcerative colitis is poorly understood and is likely to involve perturbation of the complex interactions between the mucosal immune system and the commensal bacteria of the gut, with cytokines acting as important cross-regulators. Here we use IFN receptor-deficient mice in a dextran sulfate sodium (DSS) model of acute intestinal injury to study the contributions of type I and III interferons (IFN) to the initiation, progression and resolution of acute colitis. We find that mice lacking both types of IFN receptors exhibit enhanced barrier destruction, extensive loss of goblet cells and diminished proliferation of epithelial cells in the colon following DSS-induced damage. Impaired mucosal healing in double IFN receptor-deficient mice is driven by decreased amphiregulin expression, which IFN signaling can up-regulate in either the epithelial or hematopoietic compartment. Together, these data underscore the pleiotropic functions of IFNs and demonstrate that these critical antiviral cytokines also support epithelial regeneration following acute colonic injury.
Collapse
Affiliation(s)
- Constance McElrath
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
- School of Graduate Studies, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Vanessa Espinosa
- Pediatrics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Jian-Da Lin
- School of Graduate Studies, Rutgers-The State University of New Jersey, Newark, NJ, USA
- Pathology, Immunology and Laboratory Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
- Department of Biochemical Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Jianya Peng
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
- School of Graduate Studies, Rutgers-The State University of New Jersey, Newark, NJ, USA
- Department of Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Raghavendra Sridhar
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
- School of Graduate Studies, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Orchi Dutta
- School of Graduate Studies, Rutgers-The State University of New Jersey, Newark, NJ, USA
- Pediatrics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Hsiang-Chi Tseng
- School of Graduate Studies, Rutgers-The State University of New Jersey, Newark, NJ, USA
- Pathology, Immunology and Laboratory Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Sergey V Smirnov
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Heidi Risman
- Pathology, Immunology and Laboratory Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Marvin J Sandoval
- Department of Pathology, New York University School of Medicine, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA
| | - Viralkumar Davra
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
- School of Graduate Studies, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Yun-Juan Chang
- Office of Advance Research Computing, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Brian P Pollack
- Atlanta Veterans Affairs Medical Center, Atlanta, GA, USA
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Raymond B Birge
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
- Center for Cell Signaling, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
- Cancer Institute of New Jersey, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Mark Galan
- Pathology, Immunology and Laboratory Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Amariliz Rivera
- Pediatrics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Joan E Durbin
- Pathology, Immunology and Laboratory Medicine, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA
| | - Sergei V Kotenko
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA.
- Center for Cell Signaling, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA.
- Cancer Institute of New Jersey, Rutgers-The State University of New Jersey, Newark, NJ, USA.
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey, Newark, NJ, USA.
| |
Collapse
|