1
|
Hoytema van Konijnenburg DP, Nigrovic PA, Zanoni I. Regional specialization within the mammalian respiratory immune system. Trends Immunol 2024; 45:871-891. [PMID: 39438172 PMCID: PMC11560516 DOI: 10.1016/j.it.2024.09.011] [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: 07/17/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
The respiratory tract is exposed to infection from inhaled pathogens, including viruses, bacteria, and fungi. So far, a comprehensive assessment that integrates common and distinct aspects of the immune response along different areas of the respiratory tract has been lacking. Here, we discuss key recent findings regarding anatomical, functional, and microbial factors driving regional immune adaptation in the mammalian respiratory system, how they differ between mice and humans, and the similarities and differences with the gastrointestinal tract. We demonstrate that, under evolutionary pressure, mammals evolved spatially organized immune defenses that vary between the upper and lower respiratory tract. Overall, we propose that the functional specialization of the immune response along the respiratory tract has fundamental implications for the management of infectious or inflammatory diseases.
Collapse
Affiliation(s)
| | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital, and Harvard Medical School, Boston, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Ivan Zanoni
- Division of Immunology, Boston Children's Hospital, and Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
2
|
Fiz-López A, De Prado Á, Arribas-Rodríguez E, García-Alonso FJ, Izquierdo S, Martín-Muñoz Á, Garrote JA, Arranz E, Barrio J, Fernández-Salazar L, Bernardo D. Biological variability of human intraepithelial lymphocytes throughout the human gastrointestinal tract in health and coeliac disease. Eur J Clin Invest 2024:e14304. [PMID: 39210517 DOI: 10.1111/eci.14304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Intraepithelial lymphocytes are the first line of defence of the human intestinal immune system. Besides, their composition is altered on patients with coeliac disease (CD), so they are considered as biomarkers with utility on their diagnose and/or monitoring. Our aim is to address their variability through the human gastrointestinal tract in health and characterized them in further depth in the coeliac duodenum. METHODS Intraepithelial lymphocytes were isolated from human gastric, duodenal, ileal and colonic biopsies, then stained with specific antibodies and acquired by flow cytometry. RESULTS Our results confirmed that the profile of Intraepithelial lymphocytes change through the length of the human gastrointestinal tract. Besides and given the central role that Interleukin-15 (IL-15) elicits on CD pathogenesis; we also assessed the expression of its receptor revealing that there was virtually no functional IL-15 receptor on duodenal Intraepithelial lymphocytes. Nevertheless and contrary to our expectations, the active IL-15 receptor was not increased either on Intraepithelial lymphocytes from CD patients. CONCLUSIONS IL-15 might require additional stimulus to activate intraepithelial lymphocytes. These findings may provide novel tools to aid on a CD diagnosis and/or monitoring, at the time that provide the bases to perform functional studies in order of getting a deeper insight in the specific function that Intraepithelial lymphocytes elicit on CD pathogenesis.
Collapse
Affiliation(s)
- Aida Fiz-López
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Ángel De Prado
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
- Servicio de Gastroenterología, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Elisa Arribas-Rodríguez
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | | | - Sandra Izquierdo
- Servicio de Gastroenterología, Hospital Clínico Universitario, Valladolid, Spain
| | - Álvaro Martín-Muñoz
- Cytometry Facility, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
| | - José A Garrote
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Eduardo Arranz
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Jesús Barrio
- Servicio de Gastroenterología, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Luis Fernández-Salazar
- Servicio de Gastroenterología, Hospital Clínico Universitario, Valladolid, Spain
- Departamento de Medicina, Dermatología y Toxicología, Universidad de Valladolid, Valladolid, Spain
| | - David Bernardo
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
3
|
Pan X, Köberle M, Ghashghaeinia M. Vitamin C-Dependent Uptake of Non-Heme Iron by Enterocytes, Its Impact on Erythropoiesis and Redox Capacity of Human Erythrocytes. Antioxidants (Basel) 2024; 13:968. [PMID: 39199214 PMCID: PMC11352176 DOI: 10.3390/antiox13080968] [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: 07/25/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
In the small intestine, nutrients from ingested food are absorbed and broken down by enterocytes, which constitute over 95% of the intestinal epithelium. Enterocytes demonstrate diet- and segment-dependent metabolic flexibility, enabling them to take up large amounts of glutamine and glucose to meet their energy needs and transfer these nutrients into the bloodstream. During glycolysis, ATP, lactate, and H+ ions are produced within the enterocytes. Based on extensive but incomplete glutamine oxidation large amounts of alanine or lactate are produced. Lactate, in turn, promotes hypoxia-inducible factor-1α (Hif-1α) activation and Hif-1α-dependent transcription of various proton channels and exchangers, which extrude cytoplasmic H+-ions into the intestinal lumen. In parallel, the vitamin C-dependent and duodenal cytochrome b-mediated conversion of ferric iron into ferrous iron progresses. Finally, the generated electrochemical gradient is utilized by the divalent metal transporter 1 for H+-coupled uptake of non-heme Fe2+-ions. Iron efflux from enterocytes, subsequent binding to the plasma protein transferrin, and systemic distribution supply a wide range of cells with iron, including erythroid precursors essential for erythropoiesis. In this review, we discuss the impact of vitamin C on the redox capacity of human erythrocytes and connect enterocyte function with iron metabolism, highlighting its effects on erythropoiesis.
Collapse
Affiliation(s)
- Xia Pan
- Physiological Institute, Department of Vegetative and Clinical Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany
| | - Martin Köberle
- Department of Dermatology and Allergology, School of Medicine and Health, Technical University of Munich, Biedersteinerstr. 29, 80802 München, Germany
| | - Mehrdad Ghashghaeinia
- Physiological Institute, Department of Vegetative and Clinical Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany
| |
Collapse
|
4
|
Wheeler AE, Stoeger V, Owens RM. Lab-on-chip technologies for exploring the gut-immune axis in metabolic disease. LAB ON A CHIP 2024; 24:1266-1292. [PMID: 38226866 DOI: 10.1039/d3lc00877k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The continued rise in metabolic diseases such as obesity and type 2 diabetes mellitus poses a global health burden, necessitating further research into factors implicated in the onset and progression of these diseases. Recently, the gut-immune axis, with diet as a main regulator, has been identified as a possible role player in their development. Translation of conventional 2D in vitro and animal models is however limited, while human studies are expensive and preclude individual mechanisms from being investigated. Lab-on-chip technology therefore offers an attractive new avenue to study gut-immune interactions. This review provides an overview of the influence of diet on gut-immune interactions in metabolic diseases and a critical analysis of the current state of lab-on-chip technology to study this axis. While there has been progress in the development of "immuno-competent" intestinal lab-on-chip models, with studies showing the ability of the technology to provide mechanical cues, support longer-term co-culture of microbiota and maintain in vivo-like oxygen gradients, platforms which combine all three and include intestinal and immune cells are still lacking. Further, immune cell types and inclusion of microenvironment conditions which enable in vivo-like immune cell dynamics as well as host-microbiome interactions are limited. Future model development should focus on combining these conditions to create an environment capable of hosting more complex microbiota and immune cells to allow further study into the effects of diet and related metabolites on the gut-immune ecosystem and their role in the prevention and development of metabolic diseases in humans.
Collapse
Affiliation(s)
- Alexandra E Wheeler
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Verena Stoeger
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, UK.
| |
Collapse
|
5
|
Jiang K, Cao X, Wu H, Xu Y, Liu L, Qian H, Miao Z, Wang H, Ma Y. 2D Nanozymes Modulate Gut Microbiota and T-Cell Differentiation for Inflammatory Bowel Disease Management. Adv Healthc Mater 2024; 13:e2302576. [PMID: 37897434 DOI: 10.1002/adhm.202302576] [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] [Revised: 10/26/2023] [Indexed: 10/30/2023]
Abstract
Intestinal commensal microbiota dysbiosis and immune dysfunction are significant exacerbating factors in inflammatory bowel disease (IBD). To address these problems, Pluronic F-127-coated tungsten diselenide (WSe2 @F127) nanozymes are developed by simple liquid-phase exfoliation. The abundant valence transitions of elemental selenium (Se2- /Se4+ ) and tungsten (W4+ /W6+ ) enable the obtained WSe2 @F127 nanozymes to eliminate reactive oxygen/nitrogen species. In addition, the released tungsten ions are capable of inhibiting the proliferation of Escherichia coli. In a model of dextran sodium sulfate-induced colitis, WSe2 @F127 nanozymes modulate the gut microbiota by increasing the abundance of bacteria S24-7 and significantly reducing the abundance of Enterobacteriaceae. Moreover, WSe2 @F127 nanozymes inhibit T-cell differentiation and improve intestinal immune barrier function in a model of Crohn's disease. The WSe2 @F127 nanozymes effectively alleviate IBD by reducing oxidative stress damage, modulating intestinal microbial populations, and remodeling the immune barrier.
Collapse
Affiliation(s)
- Kai Jiang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Xiangjing Cao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Haitao Wu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yifeng Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Lulu Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230022, China
| | - Zhaohua Miao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yan Ma
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230022, China
| |
Collapse
|
6
|
Li T, Han B, Wang L, Sun L, Cai Y, Yu M, Xiao W, Yang H. Activation of mucosal insulin receptor exacerbates intestinal inflammation by promoting tissue resident memory T cells differentiation through EZH2. J Transl Med 2024; 22:78. [PMID: 38243324 PMCID: PMC10797971 DOI: 10.1186/s12967-023-04789-x] [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: 08/06/2023] [Accepted: 12/09/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Inflammatory Bowel Diseases (IBD), an autoimmune disease characterised by abnormal intestinal immunity, are related to vital morbidity around the world. However, therapeutic agents for IBD have not achieved desired benefit. Exploring new therapeutic targets for IBD, especially based on its abnormally intestinal immunity, could alleviate the flare-up and worsening of IBD. Tissue resident memory T cells (TRM) are core of multiple autoimmune diseases, including IBD. However, the mechanism of TRM differentiation remains to be investigated. METHODS The alterations in mRNA and lncRNA profile of intestinal intraepithelial lymphocytes (IELs), the largest component of intestinal TRM, were analyzed in DSS-induced chronic colitis. Based on it, we examined the function of rectal insulin instillation in a dextran sodium sulfate (DSS) induced chronic colitis. Furthermore, we investigated the downstream-target of the insulin pathway-EZH2 and the crucial role of EZH2 in intestinal tissue resident memory T cell differentiation by utilizing EZH2fl/flCD4cre mice. RESULTS Insulin receptor (INSR) expression was found to be significantly reduced. Activation of mucosal insulin pathway by rectal insulin instillation exacerbated colitis by disrupting IELs subgroups and up-regulating TNF-ɑ and IL-17 expression. Rectal insulin instillation promoted EZH2 expression and EZH2 inhibition alleviated chronic colitis. EZH2fl/flCD4cre mice restored the normal IEL subgroups and suppressed TNF-ɑ and IL-17 expression, exhibiting alleviated colitis. IELs from EZH2fl/flCD4cre mice exhibit significant changes in TRM related phenotype. CD4+TRM was significantly increased in chronic colitis and decreased in EZH2fl/flCD4cre mice. CONCLUSION Insulin receptor of intestinal mucosal T-cells could promote intestinal TRM differentiation via EZH2. Our discoveries suggest that therapies targeting colonic INSR and EZH2 could be potential treatment for IBD based on its regulatory effects on TRM. Insulin receptor inhibitors rather than insulin should be applied during colitis-active phase. In addition, EZH2 shows to be a downstream signal of the insulin pathway and EZH2 inhibitor could alleviating intestinal inflammation. However, the critical role of EZH2 in TRM differentiation restricts the anti-tumor effects of EZH2 inhibitor in vivo.
Collapse
Affiliation(s)
- Teming Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
- Department of General Surgery, Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Army Medical University, Shigatse, 857000, China
| | - Ben Han
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Liucan Wang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Lihua Sun
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Yujiao Cai
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Min Yu
- Department of General Surgery, Chongqing General Hospital, Chongqing, 401147, China.
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
- Department of General Surgery, Chongqing General Hospital, Chongqing, 401147, China.
| |
Collapse
|
7
|
Lutter L, Ter Linde JJM, Brand EC, Hoytema van Konijnenburg DP, Roosenboom B, Horjus Talabur-Horje C, Oldenburg B, van Wijk F. Compartment-driven imprinting of intestinal CD4 T cells in inflammatory bowel disease and homeostasis. Clin Exp Immunol 2023; 214:235-248. [PMID: 37565620 PMCID: PMC10719222 DOI: 10.1093/cei/uxad095] [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: 03/19/2023] [Revised: 07/16/2023] [Accepted: 08/09/2023] [Indexed: 08/12/2023] Open
Abstract
The mucosal immune system is implicated in the etiology and progression of inflammatory bowel diseases. The lamina propria and epithelium of the gut mucosa constitute two separate compartments, containing distinct T-cell populations. Human CD4 T-cell programming and regulation of lamina propria and epithelium CD4 T cells, especially during inflammation, remain incompletely understood. We performed flow cytometry, bulk, and single-cell RNA-sequencing to profile ileal lamina propria and intraepithelial CD4 T cells (CD4CD8αα, regulatory T cells (Tregs), CD69- and CD69high Trm T cells) in controls and Crohn's disease (CD) patients (paired non-inflamed and inflamed). Inflammation results in alterations of the CD4 T-cell population with a pronounced increase in Tregs and migrating/infiltrating cells. On a transcriptional level, inflammation within the epithelium induced T-cell activation, increased IFNγ responses, and an effector Treg profile. Conversely, few transcriptional changes within the lamina propria were observed. Key regulators including the chromatin remodelers ARID4B and SATB1 were found to drive compartment-specific transcriptional programming of CD4 T(reg) cells. In summary, inflammation in CD patients primarily induces changes within the epithelium and not the lamina propria. Additionally, there is compartment-specific CD4 T-cell imprinting, driven by shared regulators, between the lamina propria and the epithelium. The main consequence of intraepithelial adaptation, irrespective of inflammation, seems to be an overall dampening of broad (pro-inflammatory) responses and tight regulation of lifespan. These data suggest differential regulation of the lamina propria and epithelium, with a specific regulatory role in the inflamed epithelium.
Collapse
Affiliation(s)
- Lisanne Lutter
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - José J M Ter Linde
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Eelco C Brand
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - David P Hoytema van Konijnenburg
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Division of Immunology, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Britt Roosenboom
- Department of Gastroenterology and Hepatology, Rijnstate Hospital, Arnhem, The Netherlands
| | | | - Bas Oldenburg
- Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Femke van Wijk
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
8
|
Vebr M, Pomahačová R, Sýkora J, Schwarz J. A Narrative Review of Cytokine Networks: Pathophysiological and Therapeutic Implications for Inflammatory Bowel Disease Pathogenesis. Biomedicines 2023; 11:3229. [PMID: 38137450 PMCID: PMC10740682 DOI: 10.3390/biomedicines11123229] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/11/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a lifelong inflammatory immune mediated disorder, encompassing Crohn's disease (CD) and ulcerative colitis (UC); however, the cause and specific pathogenesis of IBD is yet incompletely understood. Multiple cytokines produced by different immune cell types results in complex functional networks that constitute a highly regulated messaging network of signaling pathways. Applying biological mechanisms underlying IBD at the single omic level, technologies and genetic engineering enable the quantification of the pattern of released cytokines and new insights into the cytokine landscape of IBD. We focus on the existing literature dealing with the biology of pro- or anti-inflammatory cytokines and interactions that facilitate cell-based modulation of the immune system for IBD inflammation. We summarize the main roles of substantial cytokines in IBD related to homeostatic tissue functions and the remodeling of cytokine networks in IBD, which may be specifically valuable for successful cytokine-targeted therapies via marketed products. Cytokines and their receptors are validated targets for multiple therapeutic areas, we review the current strategies for therapeutic intervention and developing cytokine-targeted therapies. New biologics have shown efficacy in the last few decades for the management of IBD; unfortunately, many patients are nonresponsive or develop therapy resistance over time, creating a need for novel therapeutics. Thus, the treatment options for IBD beyond the immune-modifying anti-TNF agents or combination therapies are expanding rapidly. Further studies are needed to fully understand the immune response, networks of cytokines, and the direct pathogenetic relevance regarding individually tailored, safe and efficient targeted-biotherapeutics.
Collapse
Affiliation(s)
- Marek Vebr
- Departments of Pediatrics, Faculty Hospital, Faculty of Medicine in Pilsen, Charles University of Prague, 323 00 Pilsen, Czech Republic; (R.P.); (J.S.); (J.S.)
| | | | | | | |
Collapse
|
9
|
Liu Y, Duan H, Chen Y, Zhang C, Zhao J, Narbad A, Tian F, Zhai Q, Yu L, Chen W. Intraspecific difference of Latilactobacillus sakei in inflammatory bowel diseases: Insights into potential mechanisms through comparative genomics and metabolomics analyses. IMETA 2023; 2:e136. [PMID: 38868211 PMCID: PMC10989848 DOI: 10.1002/imt2.136] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 06/14/2024]
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory diseases of the gastrointestinal tract that have become a global health burden. Studies have revealed that Latilactobacillus sakei can effectively alleviate various immune diseases, including colitis, rheumatoid arthritis, and metabolic disorders. Here, we obtained 72 strains of L. sakei from 120 fermentation and fecal samples across China. In total, 16 strains from different sources were initially screened in an in vitro Caco-2 model induced by dextran sulfate sodium. Subsequently, six strains (four exhibiting effectiveness and two exhibiting ineffectiveness) were selected for further validation in an in vivo colitis mouse model. The results demonstrated that L. sakei strains exhibited varying degrees of amelioration of the colitis disease process. Notably, L. sakei CCFM1267, the most effective strain, significantly restored colon length and tight-junction protein expression, and reduced the levels of cytokines and associated inflammatory enzymes. Moreover, L. sakei CCFM1267 upregulated the abundance of Enterorhabdus, Alloprevotella, and Roseburia, leading to increased levels of acetic acid and propionic acid. Conversely, the other four strains (L. sakei QJSSZ1L4, QJSSZ4L10, QGZZYRHMT1L6, and QGZZYRHMT2L6) only exhibited a partial remission effect, while L. sakei QJSNT1L10 displayed minimal impact. Therefore, L. sakei CCFM1267 and QJSNT1L10 were selected for further exploration of the mechanisms underlying their differential mitigating effects. Comparative genomics analysis revealed significant variations between the two strains, particularly in genes associated with carbohydrate-active enzymes, such as the glycoside hydrolase family, which potentially contribute to the diverse profiles of short-chain fatty acids in vivo. Additionally, metabolome analysis demonstrated that acetylcholine and indole-3-acetic acid were the main differentiating metabolites of the two strains. Therefore, the strains of L. sakei exhibited varying degrees of effectiveness in alleviating IBD-related symptoms, and the possible reasons for these variations were attributed to discrepancies in the carbohydrate-active enzymes and metabolites among the strains.
Collapse
Affiliation(s)
- Yaru Liu
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Hui Duan
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Ying Chen
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Jianxin Zhao
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
- National Engineering Research Center for Functional FoodJiangnan UniversityWuxiChina
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
| | - Arjan Narbad
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
- Gut Health and Microbiome Institute Strategic ProgrammeQuadram Institute BioscienceNorwichUK
| | - Fengwei Tian
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
| | - Qixiao Zhai
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
| | - Leilei Yu
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
| | - Wei Chen
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
- National Engineering Research Center for Functional FoodJiangnan UniversityWuxiChina
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
| |
Collapse
|
10
|
van Eijs MJ, ter Linde JJ, Baars MJ, Amini M, Laclé MM, Brand EC, Delemarre EM, Drylewicz J, Nierkens S, Verheijden RJ, Oldenburg B, Vercoulen Y, Suijkerbuijk KP, van Wijk F. Highly multiplexed spatial analysis identifies tissue-resident memory T cells as drivers of ulcerative and immune checkpoint inhibitor colitis. iScience 2023; 26:107891. [PMID: 37766980 PMCID: PMC10520880 DOI: 10.1016/j.isci.2023.107891] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Colitis is a prevalent adverse event associated with immune checkpoint inhibitor (ICI) therapy with similarities to inflammatory bowel disease. Incomplete mechanistic understanding of ICI colitis curtails evidence-based treatment. Given the often-overlooked connection between tissue architecture and mucosal immune cell function, we here applied imaging mass cytometry (IMC) to gain spatial proteomic insight in ICI colitis in comparison to ulcerative colitis (UC). Using a cell segmentation pipeline that simultaneously utilizes high-resolution nuclear imaging and high-multiplexity IMC, we show that intra-epithelial CD8+ T cells are significantly more abundant (and numerically dominant) in anti-PD-1 ± anti-CTLA-4-induced colitis compared to anti-CTLA-4-induced colitis and UC. We identified activated, cycling CD8+ tissue-resident memory T(RM) cells at the lamina propria-epithelial interface as drivers of cytotoxicity in ICI colitis and UC. Moreover, we found that combined ICI-induced colitis featured highest granzyme B levels both in tissue and serum. Together, these data reinforce CD8+ TRM cells as potentially targetable drivers of ICI colitis.
Collapse
Affiliation(s)
- Mick J.M. van Eijs
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584 EA Utrecht, the Netherlands
- Department of Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands
| | - José J.M. ter Linde
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584 EA Utrecht, the Netherlands
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands
| | - Matthijs J.D. Baars
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Mojtaba Amini
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
- UCyTOF.nl, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Miangela M. Laclé
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands
| | - Eelco C. Brand
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584 EA Utrecht, the Netherlands
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands
| | - Eveline M. Delemarre
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Julia Drylewicz
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| | - Stefan Nierkens
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584 EA Utrecht, the Netherlands
- Princess Máxima Center for Pediatric Oncology, P.O. Box 113, 3720 AC Utrecht, the Netherlands
| | - Rik J. Verheijden
- Department of Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands
| | - Bas Oldenburg
- Department of Gastroenterology and Hepatology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands
| | - Yvonne Vercoulen
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
- UCyTOF.nl, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Karijn P.M. Suijkerbuijk
- Department of Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584 EA Utrecht, the Netherlands
| |
Collapse
|
11
|
Hickey JW, Becker WR, Nevins SA, Horning A, Perez AE, Zhu C, Zhu B, Wei B, Chiu R, Chen DC, Cotter DL, Esplin ED, Weimer AK, Caraccio C, Venkataraaman V, Schürch CM, Black S, Brbić M, Cao K, Chen S, Zhang W, Monte E, Zhang NR, Ma Z, Leskovec J, Zhang Z, Lin S, Longacre T, Plevritis SK, Lin Y, Nolan GP, Greenleaf WJ, Snyder M. Organization of the human intestine at single-cell resolution. Nature 2023; 619:572-584. [PMID: 37468586 PMCID: PMC10356619 DOI: 10.1038/s41586-023-05915-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/02/2023] [Indexed: 07/21/2023]
Abstract
The intestine is a complex organ that promotes digestion, extracts nutrients, participates in immune surveillance, maintains critical symbiotic relationships with microbiota and affects overall health1. The intesting has a length of over nine metres, along which there are differences in structure and function2. The localization of individual cell types, cell type development trajectories and detailed cell transcriptional programs probably drive these differences in function. Here, to better understand these differences, we evaluated the organization of single cells using multiplexed imaging and single-nucleus RNA and open chromatin assays across eight different intestinal sites from nine donors. Through systematic analyses, we find cell compositions that differ substantially across regions of the intestine and demonstrate the complexity of epithelial subtypes, and find that the same cell types are organized into distinct neighbourhoods and communities, highlighting distinct immunological niches that are present in the intestine. We also map gene regulatory differences in these cells that are suggestive of a regulatory differentiation cascade, and associate intestinal disease heritability with specific cell types. These results describe the complexity of the cell composition, regulation and organization for this organ, and serve as an important reference map for understanding human biology and disease.
Collapse
Affiliation(s)
- John W Hickey
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Winston R Becker
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | | | - Aaron Horning
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Almudena Espin Perez
- Department of Biomedical Data Science, Stanford School of Medicine, Stanford, CA, USA
| | - Chenchen Zhu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Bokai Zhu
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Bei Wei
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Roxanne Chiu
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Derek C Chen
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Daniel L Cotter
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Edward D Esplin
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Annika K Weimer
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Chiara Caraccio
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | | | - Christian M Schürch
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Sarah Black
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Maria Brbić
- Department of Computer Science, Stanford University, Stanford, CA, USA
- School of Computer and Communication Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Kaidi Cao
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Shuxiao Chen
- Department of Statistics and Data Science, University of Pennsylvania, Pennsylvania, PA, USA
| | - Weiruo Zhang
- Department of Biomedical Data Science, Stanford School of Medicine, Stanford, CA, USA
| | - Emma Monte
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| | - Nancy R Zhang
- Department of Statistics and Data Science, University of Pennsylvania, Pennsylvania, PA, USA
| | - Zongming Ma
- Department of Statistics and Data Science, University of Pennsylvania, Pennsylvania, PA, USA
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Zhengyan Zhang
- Department of Surgery, Washington University, St Louis, MO, USA
| | - Shin Lin
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Teri Longacre
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | - Sylvia K Plevritis
- Department of Biomedical Data Science, Stanford School of Medicine, Stanford, CA, USA
| | - Yiing Lin
- Department of Surgery, Washington University, St Louis, MO, USA
| | - Garry P Nolan
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA.
| | | | - Michael Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
12
|
Yu C, Chen Y, Ahmadi S, Wu D, Wu J, Ding T, Liu D, Ye X, Chen S, Pan H. Goji berry leaf exerts a comparable effect against colitis and microbiota dysbiosis to its fruit in dextran-sulfate-sodium-treated mice. Food Funct 2023; 14:3026-3037. [PMID: 36861301 DOI: 10.1039/d2fo02886g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
Goji berry and mulberry are both popular berries with anti-colitis effects, but their leaves have received less attention. In this study, the anti-colitis effects of goji berry leaf and mulberry leaf were investigated in dextran-sulfate-sodium-induced colitis C57BL/6N mice compared with their fruits. Goji berry leaf and goji berry reduced colitic symptoms and ameliorated tissue damage, while mulberry leaf did not. ELISA and western blotting analysis suggested that goji berry showed the best performance in inhibiting the overproduction of pro-inflammatory cytokines (TNF-α, IL-6 and IL-10) and improving damaged colonic barrier (occludin and claudin-1). Besides, goji berry leaf and goji berry reversed the gut microbiota dysbiosis by increasing the abundance of beneficial bacteria like Bifidobacterium and Muribaculaceae, and decreasing the abundance of harmful bacteria like Bilophila and Lachnoclostridium. Goji berry, mulberry and goji berry leaf could restore acetate, propionate, butyrate and valerate to ameliorate inflammation, while mulberry leaf could not restore butyrate. To the best of our knowledge, this is the first report on the comparison of the anti-colitis effects of goji berry leaf, mulberry leaf and their fruits, which is meaningful for the rational utilization of goji berry leaf as a functional food.
Collapse
Affiliation(s)
- Chengxiao Yu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang University, Zhejiang, 310058, China.
| | - Yihao Chen
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang University, Zhejiang, 310058, China.
| | - Shokouh Ahmadi
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang University, Zhejiang, 310058, China.
| | - Dongmei Wu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang University, Zhejiang, 310058, China.
| | - Jiaxiong Wu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang University, Zhejiang, 310058, China.
| | - Tian Ding
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang University, Zhejiang, 310058, China.
| | - Donghong Liu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang University, Zhejiang, 310058, China.
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang University, Zhejiang, 310058, China. .,Zhejiang University Zhongyuan Institute, Zhengzhou, 450000, China.,Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi, 276000, China
| | - Shiguo Chen
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang University, Zhejiang, 310058, China. .,Zhejiang University Zhongyuan Institute, Zhengzhou, 450000, China.,Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi, 276000, China
| | - Haibo Pan
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang University, Zhejiang, 310058, China. .,Zhejiang University Zhongyuan Institute, Zhengzhou, 450000, China.,Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi, 276000, China
| |
Collapse
|
13
|
Lechuga S, Braga-Neto MB, Naydenov NG, Rieder F, Ivanov AI. Understanding disruption of the gut barrier during inflammation: Should we abandon traditional epithelial cell lines and switch to intestinal organoids? Front Immunol 2023; 14:1108289. [PMID: 36875103 PMCID: PMC9983034 DOI: 10.3389/fimmu.2023.1108289] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/01/2023] [Indexed: 02/18/2023] Open
Abstract
Disruption of the intestinal epithelial barrier is a hallmark of mucosal inflammation. It increases exposure of the immune system to luminal microbes, triggering a perpetuating inflammatory response. For several decades, the inflammatory stimuli-induced breakdown of the human gut barrier was studied in vitro by using colon cancer derived epithelial cell lines. While providing a wealth of important data, these cell lines do not completely mimic the morphology and function of normal human intestinal epithelial cells (IEC) due to cancer-related chromosomal abnormalities and oncogenic mutations. The development of human intestinal organoids provided a physiologically-relevant experimental platform to study homeostatic regulation and disease-dependent dysfunctions of the intestinal epithelial barrier. There is need to align and integrate the emerging data obtained with intestinal organoids and classical studies that utilized colon cancer cell lines. This review discusses the utilization of human intestinal organoids to dissect the roles and mechanisms of gut barrier disruption during mucosal inflammation. We summarize available data generated with two major types of organoids derived from either intestinal crypts or induced pluripotent stem cells and compare them to the results of earlier studies with conventional cell lines. We identify research areas where the complementary use of colon cancer-derived cell lines and organoids advance our understanding of epithelial barrier dysfunctions in the inflamed gut and identify unique questions that could be addressed only by using the intestinal organoid platforms.
Collapse
Affiliation(s)
- Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Manuel B. Braga-Neto
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Nayden G. Naydenov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| |
Collapse
|
14
|
Chen X, Zhu Y, Wei Y, Fan S, Xia L, Chen Q, Lu Y, Wu D, Liu X, Peng X. Glutamine alleviates intestinal injury in a murine burn sepsis model by maintaining intestinal intraepithelial lymphocyte homeostasis. Eur J Pharmacol 2023; 940:175480. [PMID: 36566008 DOI: 10.1016/j.ejphar.2022.175480] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/05/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Intestinal intraepithelial lymphocytes (IELs) play a sentinel role in the mucosal immune system because of their unique anatomical location in the epithelial layer. The disruption of IEL homeostasis is implicated in driving the intestinal injury of many typical inflammatory disorders, such as inflammatory bowel disease (IBD) and sepsis. Therefore, it is meaningful to alleviate intestinal injury by restoring IEL homeostasis in disease conditions. This study explores the effects of glutamine on intestinal IEL homeostasis in a murine model of burn sepsis. We report that glutamine inhibits inflammatory response and reduces injury in the small intestine of burn septic mice. This effect is attributed to the maintaining of IEL homeostasis by suppressing apoptosis and restoring the disrupted subpopulation balance induced by burn sepsis. Mechanistically, we show that glutamine does not affect the IL-15 dependent mechanisms that drive the maintenance and differentiation of IELs. Instead, glutamine sustains IEL homeostasis by upregulate aryl hydrocarbon receptor (AHR) and interleukin (IL)-22 transcription and expression. Consistently, the protective roles of glutamine in burn septic mice were repressed by further supplement with an AHR antagonist CH-223191. Collectively, our study reveals a new role of glutamine to maintain IEL homeostasis by activating the AHR signaling pathway, which in turn ameliorates intestinal injury in burn sepsis.
Collapse
Affiliation(s)
- Xiaoli Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yuanfeng Zhu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yan Wei
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shijun Fan
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Lin Xia
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Qian Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yongling Lu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Dan Wu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xin Liu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Xi Peng
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
15
|
Zhu D, Wu H, Jiang K, Xu Y, Miao Z, Wang H, Ma Y. Zero-Valence Selenium-Enriched Prussian Blue Nanozymes Reconstruct Intestinal Barrier against Inflammatory Bowel Disease via Inhibiting Ferroptosis and T Cells Differentiation. Adv Healthc Mater 2023; 12:e2203160. [PMID: 36651877 DOI: 10.1002/adhm.202203160] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/13/2023] [Indexed: 01/19/2023]
Abstract
The structural disruption of mechanical barrier and dysfunction of immune barrier in intestinal, are important factors, that aggravate inflammatory bowel disease (IBD). To tackle this challenge, a multifunctional nanozyme capable of scavenging reactive oxygen species (ROS) and inhibiting ferroptosis or T cells differentiation for IBD therapy is here reported. In this work, zero-valence selenium-enriched Prussian blue nanozymes (Se-HMPB nanozymes) are prepared via the hard template method. PB nanozymes with multi-enzyme activities can effectively scavenge various ROS in inflammatory tissues. Meanwhile, the presence of selenium element endows the glutathione peroxidase activity of Se-HMPB nanozymes, which can inhibit ferroptosis and reverse the lipid peroxidation of intestinal epithelial cells to protect the intestinal mechanical barrier in ulcerative colitis (UC) model. In addition, selenium supplementation can realize efficient inhibition on the differentiation of T cells in Crohn's disease (CD) model, regulating the intestinal immune barrier. Thus, the Se-HMPB nanozymes reconstructed intestinal barrier via inhibiting ferroptosis and T cells differentiation in UC and CD models, depicting great potential to alleviate IBD.
Collapse
Affiliation(s)
- Dongdong Zhu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, P. R. China
| | - Haitao Wu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, P. R. China
| | - Kai Jiang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, P. R. China
| | - Yifeng Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, P. R. China
| | - Zhaohua Miao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, P. R. China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, P. R. China
| | - Yan Ma
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, P. R. China
| |
Collapse
|
16
|
Røyset ES, Sahlin Pettersen HP, Xu W, Larbi A, Sandvik AK, Steigen SE, Catalan‐Serra I, Bakke I. Deep learning-based image analysis reveals significant differences in the number and distribution of mucosal CD3 and γδ T cells between Crohn's disease and ulcerative colitis. J Pathol Clin Res 2022; 9:18-31. [PMID: 36416283 PMCID: PMC9732684 DOI: 10.1002/cjp2.301] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/20/2022] [Accepted: 10/26/2022] [Indexed: 11/25/2022]
Abstract
Colon mucosae of ulcerative colitis (UC) and Crohn's disease (CD) display differences in the number and distribution of immune cells that are difficult to assess by eye. Deep learning-based analysis on whole slide images (WSIs) allows extraction of complex quantitative data that can be used to uncover different inflammatory patterns. We aimed to explore the distribution of CD3 and γδ T cells in colon mucosal compartments in histologically inactive and active inflammatory bowel disease. By deep learning-based segmentation and cell detection on WSIs from a well-defined cohort of CD (n = 37), UC (n = 58), and healthy controls (HCs, n = 33), we quantified CD3 and γδ T cells within and beneath the epithelium and in lamina propria in proximal and distal colon mucosa, defined by the Nancy histological index. We found that inactive CD had significantly fewer intraepithelial γδ T cells than inactive UC, but higher total number of CD3 cells in all compartments than UC and HCs. Disease activity was associated with a massive loss of intraepithelial γδ T cells in UC, but not in CD. The total intraepithelial number of CD3 cells remained constant regardless of disease activity in both CD and UC. There were more mucosal CD3 and γδ T cells in proximal versus distal colon. Oral corticosteroids had an impact on γδ T cell numbers, while age, gender, and disease duration did not. Relative abundance of γδ T cells in mucosa and blood did not correlate. This study reveals significant differences in the total number of CD3 and γδ T cells in particularly the epithelial area between CD, UC, and HCs, and demonstrates useful application of deep segmentation to quantify cells in mucosal compartments.
Collapse
Affiliation(s)
- Elin Synnøve Røyset
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences (MH)NTNU – Norwegian University of Science and TechnologyTrondheimNorway,Department of Pathology, St. Olav's HospitalTrondheim University HospitalTrondheimNorway,Clinic of Laboratory Medicine, St. Olav's HospitalTrondheim University HospitalTrondheimNorway
| | - Henrik P Sahlin Pettersen
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences (MH)NTNU – Norwegian University of Science and TechnologyTrondheimNorway,Department of Pathology, St. Olav's HospitalTrondheim University HospitalTrondheimNorway
| | - Weili Xu
- Singapore Immunology Network (SIgN)Agency for Science Technology and Research, BiopolisSingapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN)Agency for Science Technology and Research, BiopolisSingapore
| | - Arne K Sandvik
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences (MH)NTNU – Norwegian University of Science and TechnologyTrondheimNorway,Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's HospitalTrondheim University HospitalTrondheimNorway,Centre of Molecular Inflammation Research (CEMIR)NTNUTrondheimNorway
| | - Sonja E Steigen
- Department of Medical Biology, Faculty of Health SciencesUiT The Arctic University of NorwayTromsøNorway,Department of Clinical PathologyUniversity Hospital of North NorwayTromsøNorway
| | - Ignacio Catalan‐Serra
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences (MH)NTNU – Norwegian University of Science and TechnologyTrondheimNorway,Centre of Molecular Inflammation Research (CEMIR)NTNUTrondheimNorway,Department of Medicine, GastroenterologyLevanger Hospital, Nord‐Trøndelag Hospital TrustLevangerNorway
| | - Ingunn Bakke
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences (MH)NTNU – Norwegian University of Science and TechnologyTrondheimNorway,Clinic of Laboratory Medicine, St. Olav's HospitalTrondheim University HospitalTrondheimNorway
| |
Collapse
|
17
|
Wiarda JE, Trachsel JM, Sivasankaran SK, Tuggle CK, Loving CL. Intestinal single-cell atlas reveals novel lymphocytes in pigs with similarities to human cells. Life Sci Alliance 2022; 5:e202201442. [PMID: 35995567 PMCID: PMC9396248 DOI: 10.26508/lsa.202201442] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 12/13/2022] Open
Abstract
Lymphocytes can heavily influence intestinal health, but resolving intestinal lymphocyte function is challenging as the intestine contains a vastly heterogeneous mixture of cells. Pigs are an advantageous biomedical model, but deeper understanding of intestinal lymphocytes is warranted to improve model utility. Twenty-six cell types were identified in the porcine ileum by single-cell RNA sequencing and further compared with cells in human and murine ileum. Though general consensus of cell subsets across species was revealed, some porcine-specific lymphocyte subsets were identified. Differential tissue dissection and in situ analyses conferred spatial context, revealing similar locations of lymphocyte subsets in Peyer's patches and epithelium in pig-to-human comparisons. Like humans, activated and effector lymphocytes were abundant in the ileum but not periphery of pigs, suggesting tissue-specific and/or activation-associated gene expression. Gene signatures for peripheral and ileal innate lymphoid cells newly discovered in pigs were defined and highlighted similarities to human innate lymphoid cells. Overall, we reveal novel lymphocyte subsets in pigs and highlight utility of pigs for intestinal research applications.
Collapse
Affiliation(s)
- Jayne E Wiarda
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
- Immunobiology Graduate Program, Iowa State University, Ames, IA, USA
- Oak Ridge Institute for Science and Education, Agricultural Research Service Participation Program, Oak Ridge, TN, USA
| | - Julian M Trachsel
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Sathesh K Sivasankaran
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
- Genome Informatics Facility, Iowa State University, Ames, IA, USA
| | | | - Crystal L Loving
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| |
Collapse
|
18
|
Dolan B, Ermund A, Martinez-Abad B, Johansson ME, Hansson GC. Clearance of small intestinal crypts involves goblet cell mucus secretion by intracellular granule rupture and enterocyte ion transport. Sci Signal 2022; 15:eabl5848. [PMID: 36126118 PMCID: PMC9749883 DOI: 10.1126/scisignal.abl5848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Goblet cells in the small intestinal crypts contain large numbers of mucin granules that are rapidly discharged to clean bacteria from the crypt. Because acetylcholine released by neuronal and nonneuronal cells controls many aspects of intestinal epithelial function, we used tissue explants and organoids to investigate the response of the small intestinal crypt to cholinergic stimulation. The activation of muscarinic acetylcholine receptors initiated a coordinated and rapid emptying of crypt goblet cells that flushed the crypt contents into the intestinal lumen. Cholinergic stimulation induced an expansion of the granule contents followed by intracellular rupture of the mucin granules. The mucus expanded intracellularly before the rupture of the goblet cell apical membrane and continued to expand after its release into the crypt lumen. The goblet cells recovered from membrane rupture and replenished their stores of mucin granules. Mucus secretion from the goblet cells depended on Ca2+ signaling and the expansion of the mucus in the crypt depended on gap junctions and on ion and water transport by enterocytes adjacent to the goblet cells. This distinctive mode of mucus secretion, which we refer to as "expanding secretion," efficiently cleans the small intestine crypt through coordinated mucus, ion, and fluid secretion by goblet cells and enterocytes.
Collapse
Affiliation(s)
- Brendan Dolan
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Anna Ermund
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Beatriz Martinez-Abad
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Malin E.V. Johansson
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| | - Gunnar C. Hansson
- Department of Medical Biochemistry and Cell Biology, University of
Gothenburg, 405 30 Gothenburg, Sweden
| |
Collapse
|
19
|
Wang Y, Song W, Yu S, Liu Y, Chen YG. Intestinal cellular heterogeneity and disease development revealed by single-cell technology. CELL REGENERATION 2022; 11:26. [PMID: 36045190 PMCID: PMC9433512 DOI: 10.1186/s13619-022-00127-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022]
Abstract
The intestinal epithelium is responsible for food digestion and nutrient absorption and plays a critical role in hormone secretion, microorganism defense, and immune response. These functions depend on the integral single-layered intestinal epithelium, which shows diversified cell constitution and rapid self-renewal and presents powerful regeneration plasticity after injury. Derailment of homeostasis of the intestine epithelium leads to the development of diseases, most commonly including enteritis and colorectal cancer. Therefore, it is important to understand the cellular characterization of the intestinal epithelium at the molecular level and the mechanisms underlying its homeostatic maintenance. Single-cell technologies allow us to gain molecular insights at the single-cell level. In this review, we summarize the single-cell RNA sequencing applications to understand intestinal cell characteristics, spatiotemporal evolution, and intestinal disease development.
Collapse
|
20
|
Yu C, Hu X, Ahmadi S, Wu D, Xiao H, Zhang H, Ding T, Liu D, Ye X, Chen S, Chen J. Structure and In Vitro Fermentation Characteristics of Polysaccharides Sequentially Extracted from Goji Berry ( Lycium barbarum) Leaves. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:7535-7546. [PMID: 35549264 DOI: 10.1021/acs.jafc.2c01157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Herein, the chelating agent-soluble fraction (CA), sodium carbonate-soluble fraction (SC), and sodium hydroxide-soluble fraction (SH) were sequentially extracted from the cell wall of goji berry (Lycium barbarum) leaves. Furthermore, SC was purified with Q-Sepharose fast flow resin to obtain the neutral sugar fraction (SC-I) and acid sugar fraction (SC-II). Physicochemical properties of polysaccharides were characterized by high-performance anion-exchange chromatography with pulsed amperometry detection, size exclusion chromatography-multi-angle laser light scattering, Fourier transform infrared spectroscopy, nuclear magnetic resonance, and atomic force microscopy analysis. Additionally, the impact of polysaccharides on modulating human gut microbiota was investigated by in vitro fermentation. A high amount of galacturonic acid (GalA) in CA showed that it was an aggregation of linear homogalacturonan. SC was the main pectic polysaccharide fraction and rich in neutral sugars. SC-I was the neutral sugar fraction with an extremely high molecular weight (2.055 × 106 Da), while SC-II was the acid sugar fraction with a low molecular weight (1.766 × 105 Da). SH seemed like a mixture of pectin and hemicellulose. All the five polysaccharides significantly (P < 0.05) increased the abundance of Bacteroides, Bifidobacteria, and Lactobacilli. To the best of our knowledge, this is the first report on the structure and fermentation characteristics of goji berry leaf polysaccharides, which is meaningful to provide a structural basis for further bioactivity research.
Collapse
Affiliation(s)
- Chengxiao Yu
- College of Biosystems Engineering and Food Science, Ningbo Research Institute, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xinxin Hu
- College of Biosystems Engineering and Food Science, Ningbo Research Institute, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shokouh Ahmadi
- College of Biosystems Engineering and Food Science, Ningbo Research Institute, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Dongmei Wu
- College of Biosystems Engineering and Food Science, Ningbo Research Institute, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hang Xiao
- College of Biosystems Engineering and Food Science, Ningbo Research Institute, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huiling Zhang
- Ningxia Key Laboratory for Food Microbial-Applications Technology and Safety Control, Ningxia University, Yinchuan 750021, China
| | - Tian Ding
- College of Biosystems Engineering and Food Science, Ningbo Research Institute, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Donghong Liu
- College of Biosystems Engineering and Food Science, Ningbo Research Institute, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xingqian Ye
- College of Biosystems Engineering and Food Science, Ningbo Research Institute, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Zhejiang University Zhongyuan Institute, Zhengzhou 450007, China
- Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi 276000, China
| | - Shiguo Chen
- College of Biosystems Engineering and Food Science, Ningbo Research Institute, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Zhejiang University Zhongyuan Institute, Zhengzhou 450007, China
- Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi 276000, China
| | - Jianle Chen
- College of Biosystems Engineering and Food Science, Ningbo Research Institute, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro Food Processing, Fuli Institute of Food Science, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Zhejiang University Zhongyuan Institute, Zhengzhou 450007, China
- Shandong (Linyi) Institute of Modern Agriculture, Zhejiang University, Linyi 276000, China
| |
Collapse
|
21
|
5-Fluorouracil Treatment of CT26 Colon Cancer Is Compromised by Combined Therapy with IMMODIN. Int J Mol Sci 2022; 23:ijms23126374. [PMID: 35742825 PMCID: PMC9223647 DOI: 10.3390/ijms23126374] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/03/2022] [Accepted: 06/05/2022] [Indexed: 02/01/2023] Open
Abstract
Due to the physiological complexity of the tumour, a single drug therapeutic strategy may not be sufficient for effective treatment. Emerging evidence suggests that combination strategies may be important to achieve more efficient tumour responses. Different immunomodulators are frequently tested to reverse the situation for the purpose of improving immune response and minimizing chemotherapy side effects. Immodin (IM) represents an attractive alternative to complement chemotherapy, which can be used to enhance the immune system after disturbances resulting from the side effects of chemotherapy. In the presented study, a model of CT26 tumor-bearing mice was used to investigate the effect of single IM or its combination with 5-fluorouracil (5-FU) on colon cancer cells. Our results highlight that the beneficial role of IM claimed in previous studies cannot be generalised to all chemotherapeutic drugs, as 5-FU toxicity was not increased. On the contrary, the chemotherapeutic anti-cancer efficacy of 5-FU was greatly compromised when combined with IM. Indeed, the combined treatment was significantly less effective regarding the tumour growth and animal survival, most probably due to the increased number of tumour-associated macrophages, and increased 5-FU cytotoxic effect related to kidneys and the liver.
Collapse
|
22
|
Structure and fermentation characteristics of five polysaccharides sequentially extracted from sugar beet pulp by different methods. Food Hydrocoll 2022. [DOI: 10.1016/j.foodhyd.2021.107462] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
23
|
Mou Y, Du Y, Zhou L, Yue J, Hu X, Liu Y, Chen S, Lin X, Zhang G, Xiao H, Dong B. Gut Microbiota Interact With the Brain Through Systemic Chronic Inflammation: Implications on Neuroinflammation, Neurodegeneration, and Aging. Front Immunol 2022; 13:796288. [PMID: 35464431 PMCID: PMC9021448 DOI: 10.3389/fimmu.2022.796288] [Citation(s) in RCA: 128] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 02/22/2022] [Indexed: 02/05/2023] Open
Abstract
It has been noticed in recent years that the unfavorable effects of the gut microbiota could exhaust host vigor and life, yet knowledge and theory are just beginning to be established. Increasing documentation suggests that the microbiota-gut-brain axis not only impacts brain cognition and psychiatric symptoms but also precipitates neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). How the blood-brain barrier (BBB), a machinery protecting the central nervous system (CNS) from the systemic circulation, allows the risky factors derived from the gut to be translocated into the brain seems paradoxical. For the unique anatomical, histological, and immunological properties underpinning its permeable dynamics, the BBB has been regarded as a biomarker associated with neural pathogenesis. The BBB permeability of mice and rats caused by GM dysbiosis raises the question of how the GM and its metabolites change BBB permeability and causes the brain pathophysiology of neuroinflammation and neurodegeneration (NF&ND) and brain aging, a pivotal multidisciplinary field tightly associated with immune and chronic systemic inflammation. If not all, gut microbiota-induced systemic chronic inflammation (GM-SCI) mainly refers to excessive gut inflammation caused by gut mucosal immunity dysregulation, which is often influenced by dietary components and age, is produced at the interface of the intestinal barrier (IB) or exacerbated after IB disruption, initiates various common chronic diseases along its dispersal routes, and eventually impairs BBB integrity to cause NF&ND and brain aging. To illustrate the immune roles of the BBB in pathophysiology affected by inflammatory or "leaky" IB resulting from GM and their metabolites, we reviewed the selected publications, including the role of the BBB as the immune barrier, systemic chronic inflammation and inflammation influences on BBB permeability, NF&ND, and brain aging. To add depth to the bridging role of systemic chronic inflammation, a plausible mechanism indispensable for BBB corruption was highlighted; namely, BBB maintenance cues are affected by inflammatory cytokines, which may help to understand how GM and its metabolites play a major role in NF&ND and aging.
Collapse
Affiliation(s)
- Yi Mou
- Geroscience and Chronic Disease Department, The Eighth Municipal Hospital for the People, Chengdu, China
| | - Yu Du
- Department of Emergency and Critical Care Medicine, The Fourth West China Hospital, Sichuan University, Chengdu, China
| | - Lixing Zhou
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jirong Yue
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xianliang Hu
- Geroscience and Chronic Disease Department, The Eighth Municipal Hospital for the People, Chengdu, China
| | - Yixin Liu
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Sao Chen
- Geroscience and Chronic Disease Department, The Eighth Municipal Hospital for the People, Chengdu, China
| | - Xiufang Lin
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Gongchang Zhang
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hengyi Xiao
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Birong Dong
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Arnous R, Arshad S, Sandgren K, Cunningham AL, Carnt N, White A. Tissue resident memory T cells inhabit the deep human conjunctiva. Sci Rep 2022; 12:6077. [PMID: 35414674 PMCID: PMC9005529 DOI: 10.1038/s41598-022-09886-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 03/14/2022] [Indexed: 12/20/2022] Open
Abstract
Mucosal linings of the body, including the conjunctiva, are enriched in tissue-resident memory T cells (TRMs) whose defining feature is their continual tissue protection that does not rely on migration to lymphoid organs to elicit immune responses. Hitherto, conjunctival TRMs have only been identified in the superficial epithelium. This work aims to develop a more complete understanding of the conjunctival immunological capacity by investigating the presence of TRMs within the deeper, more stable layers of the healthy human conjunctiva. Using immunofluorescence microscopy and antibodies against CD3, CD4, CD69 and HLA-DR on bulbar conjunctival biopsies obtained from 7 healthy adults (age range = 32-77 years; females = 4), we identified CD69+TRM subsets in all layers of the human conjunctiva: the superficial epithelium, the basal epithelium, the adenoid, and the fibrous layers. Interestingly, the adenoid layer showed significantly higher densities of both CD4 and CD8 TRMs when compared to the fibrous layer and conjunctival epithelia. Additionally, CD4 TRMs predominated significantly over CD8 TRMs in the adenoid layer. The abundance of deep conjunctival CD69+TRMs within the healthy human may suggest the presence of defence mechanisms capable of inducing long-term immunogenic memory. Understanding this spatial distribution of conjunctival CD69+TRMs is essential to improving mucosal vaccine design.
Collapse
Affiliation(s)
- Racha Arnous
- Centre for Vision Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Sana Arshad
- Centre for Vision Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Kerrie Sandgren
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Anthony L Cunningham
- Centre for Virus Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Nicole Carnt
- Centre for Vision Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia.,School of Optometry and Vision Science, University of New South Wales, Kensington, NSW, 2033, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Andrew White
- Centre for Vision Research, The Westmead Institute for Medical Research, 176 Hawkesbury Road, Westmead, NSW, 2145, Australia. .,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia. .,Save Sight Institute, The University of Sydney, Sydney, NSW, 2000, Australia.
| |
Collapse
|
25
|
He P, Yu L, Tian F, Zhang H, Chen W, Zhai Q. Dietary Patterns and Gut Microbiota: The Crucial Actors in Inflammatory Bowel Disease. Adv Nutr 2022; 13:1628-1651. [PMID: 35348593 PMCID: PMC9526834 DOI: 10.1093/advances/nmac029] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 02/06/2023] Open
Abstract
It is widely believed that diet and the gut microbiota are strongly related to the occurrence and progression of inflammatory bowel disease (IBD), but the effects of the interaction between dietary patterns and the gut microbiota on IBD have not been well elucidated. In this article, we aim to explore the complex relation between dietary patterns, gut microbiota, and IBD. We first comprehensively summarized the dietary patterns associated with IBD and found that dietary patterns can modulate the occurrence and progression of IBD through various signaling pathways, including mammalian target of rapamycin (mTOR), mitogen-activated protein kinases (MAPKs), signal transducer and activator of transcription 3 (STAT3), and NF-κB. Besides, the gut microbiota performs a vital role in the progression of IBD, which can affect the expression of IBD susceptibility genes, such as dual oxidase 2 (DUOX2) and APOA-1 , the intestinal barrier (in particular, the expression of tight junction proteins), immune function (especially the homeostasis between effector and regulatory T cells) and the physiological metabolism, in particular, SCFAs, bile acids (BAs), and tryptophan metabolism. Finally, we reviewed the current knowledge on the interaction between dietary patterns and the gut microbiota in IBD and found that dietary patterns modulate the onset and progression of IBD, which is partly attributed to the regulation of the gut microbiota (especially SCFAs-producing bacteria and Escherichia coli). Faecalibacteria as "microbiomarkers" of IBD could be used as a target for dietary interventions to alleviate IBD. A comprehensive understanding of the interplay between dietary intake, gut microbiota, and IBD will facilitate the development of personalized dietary strategies based on the regulation of the gut microbiota in IBD and expedite the era of precision nutritional interventions for IBD.
Collapse
Affiliation(s)
- Pandi He
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China,Wuxi Translational Medicine Research Center, Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | | |
Collapse
|
26
|
Lechner K, Zeeshana M, Noack M, Ali H, Neurath M, Weigmanna B. Small but powerful: Will nanoparticles be the future state‐of‐the‐art therapy for IBD? Expert Opin Drug Deliv 2022; 19:235-245. [DOI: 10.1080/17425247.2022.2043847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kristina Lechner
- Medical Clinic I, University Hospital Erlangen, Research Campus, Hartmannstr.14, 91052, Erlangen, 91052 Erlangen, Germany
| | - Mahira Zeeshana
- Department of Pharmacy, Faculty of Biological Sciences, Quaid‐i‐Azam University, Islamabad, 45320, Pakistan
| | - Maxi Noack
- Medical Clinic I, University Hospital Erlangen, Research Campus, Hartmannstr.14, 91052, Erlangen, 91052 Erlangen, Germany
| | - Hussain Ali
- Department of Pharmacy, Faculty of Biological Sciences, Quaid‐i‐Azam University, Islamabad, 45320, Pakistan
| | - Markus Neurath
- Medical Clinic I, University Hospital Erlangen, Ulmenweg 14, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Ulmenweg 18, 91054 Erlangen, Germany
| | - Benno Weigmanna
- Medical Immunology Campus Erlangen, Medical Clinic 1, Friedrich‐Alexander University, Erlangen‐Nürnberg, Erlangen, Germany
| |
Collapse
|
27
|
Pettersen HS, Belevich I, Røyset ES, Smistad E, Simpson MR, Jokitalo E, Reinertsen I, Bakke I, Pedersen A. Code-Free Development and Deployment of Deep Segmentation Models for Digital Pathology. Front Med (Lausanne) 2022; 8:816281. [PMID: 35155486 PMCID: PMC8829033 DOI: 10.3389/fmed.2021.816281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Application of deep learning on histopathological whole slide images (WSIs) holds promise of improving diagnostic efficiency and reproducibility but is largely dependent on the ability to write computer code or purchase commercial solutions. We present a code-free pipeline utilizing free-to-use, open-source software (QuPath, DeepMIB, and FastPathology) for creating and deploying deep learning-based segmentation models for computational pathology. We demonstrate the pipeline on a use case of separating epithelium from stroma in colonic mucosa. A dataset of 251 annotated WSIs, comprising 140 hematoxylin-eosin (HE)-stained and 111 CD3 immunostained colon biopsy WSIs, were developed through active learning using the pipeline. On a hold-out test set of 36 HE and 21 CD3-stained WSIs a mean intersection over union score of 95.5 and 95.3% was achieved on epithelium segmentation. We demonstrate pathologist-level segmentation accuracy and clinical acceptable runtime performance and show that pathologists without programming experience can create near state-of-the-art segmentation solutions for histopathological WSIs using only free-to-use software. The study further demonstrates the strength of open-source solutions in its ability to create generalizable, open pipelines, of which trained models and predictions can seamlessly be exported in open formats and thereby used in external solutions. All scripts, trained models, a video tutorial, and the full dataset of 251 WSIs with ~31 k epithelium annotations are made openly available at https://github.com/andreped/NoCodeSeg to accelerate research in the field.
Collapse
Affiliation(s)
- Henrik Sahlin Pettersen
- Department of Pathology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Ilya Belevich
- Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Elin Synnøve Røyset
- Department of Pathology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Erik Smistad
- Department of Health Research, SINTEF Digital, Trondheim, Norway
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- The Clinical Research Unit for Central Norway, Trondheim, Norway
| | - Eija Jokitalo
- Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Ingerid Reinertsen
- Department of Health Research, SINTEF Digital, Trondheim, Norway
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingunn Bakke
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - André Pedersen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Department of Health Research, SINTEF Digital, Trondheim, Norway
- The Cancer Foundation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
28
|
Sun Q, Du M, Kang Y, Zhu MJ. Prebiotic effects of goji berry in protection against inflammatory bowel disease. Crit Rev Food Sci Nutr 2022:1-25. [PMID: 34991393 DOI: 10.1080/10408398.2021.2015680] [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] [Indexed: 02/08/2023]
Abstract
The prevalence of inflammatory bowel disease (IBD) is increasing, which is concerning because IBD is a known risk factor for the development of colorectal cancer. Emerging evidence highlights environmental factors, particularly dietary factors and gut microbiota dysbiosis, as pivotal inducers of IBD onset. Goji berry, an ancient tonic food and a nutraceutical supplement, contains a range of phytochemicals such as polysaccharides, carotenoids, and polyphenols. Among these phytochemicals, L. barbarum polysaccharides (LBPs) are the most important functional constituents, which have protective effects against oxidative stress, inflammation, and neurodegeneration. Recently, the beneficial effects of goji berry and associated LBPs consumption were linked to prebiotic effects, which can prevent dysbiosis associated with IBD. This review assessed pertinent literature on the protective effects of goji berry against IBD focusing on the gut microbiota and their metabolites in mediating the observed beneficial effects.
Collapse
Affiliation(s)
- Qi Sun
- School of Food Science, Washington State University, Pullman, Washington, USA
| | - Min Du
- Department of Animal Science, Washington State University, Pullman, Washington, USA
| | - Yifei Kang
- School of Food Science, Washington State University, Pullman, Washington, USA
| | - Mei-Jun Zhu
- School of Food Science, Washington State University, Pullman, Washington, USA
| |
Collapse
|
29
|
Harnessing murine models of Crohn's disease ileitis to advance concepts of pathophysiology and treatment. Mucosal Immunol 2022; 15:10-26. [PMID: 34316007 DOI: 10.1038/s41385-021-00433-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/04/2023]
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are both characterized by chronic inflammation and severe dysfunction of the gastrointestinal tract. These two forms of inflammatory bowel disease (IBD) represent distinct clinical disorders with diverse driving mechanisms; however, this divergence is not reflected in currently approved therapeutics that commonly target general proinflammatory pathways. A compelling need therefore remains to understand factors that differentiate the topology and the distinct clinical manifestations of CD versus UC, in order to develop more effective and specialized therapies. Animal models provide valuable platforms for studying IBD heterogeneity and deciphering disease-specific mechanisms. Both the established and the newly developed ileitis mouse models are characterized by various disease initiating mechanisms and diverse phenotypic outcomes that reflect the complexity of human CD-ileitis. Microbial dysbiosis, destruction of epithelial barrier integrity, immune cell deregulation, as well as the recently described genome instability and stromal cell activation have all been proposed as the triggering factors for the development of ileitis-associated pathology. In this review, we aim to critically evaluate the mechanistic underpinnings of murine models of CD-ileitis, discuss their phenotypic similarities to human disease, and envisage their further exploitation for the development of novel targeted and personalized therapeutics.
Collapse
|
30
|
Martchenko SE, Prescott D, Martchenko A, Sweeney ME, Philpott DJ, Brubaker PL. Diurnal changes in the murine small intestine are disrupted by obesogenic Western Diet feeding and microbial dysbiosis. Sci Rep 2021; 11:20571. [PMID: 34663882 PMCID: PMC8523685 DOI: 10.1038/s41598-021-98986-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 09/01/2021] [Indexed: 02/07/2023] Open
Abstract
Intestinal functions demonstrate circadian rhythms thought to be entrained, in part, by an organisms’ intrinsic feeding and fasting periods as well as by the intestinal microbiome. Circadian disruption as a result of ill-timed nutrient exposure and obesogenic feeding poses an increased risk to disease. As such, the aim of this study was to assess the relationships between dietary timing, composition, and the microbiome with regard to rhythmic small intestinal structure and mucosal immunity. Rodent chow (RC)-mice exhibited time-dependent increases in small intestinal weight, villus height, and crypt depth as well as an increased proportion of CD8αα+ cells and concomitant decrease in CD8αβ+ cells at the onset of the feeding period (p < 0.05–0.001). Western diet (WD)-animals displayed disrupted time-dependent patterns in intestinal structure and lymphocyte populations (p < 0.05–0.01). Antibiotic-induced microbial depletion abrogated the time- and diet-dependent patterns in both RC- and WD-mice (p < 0.05–0.001). However, although germ-free-mice displayed altered rhythms, fecal microbial transfer from RC-mice was generally unsuccessful in restoring structural and immune changes in these animals. This study shows that adaptive changes in the small intestine at the onset of the feeding and fasting periods are disrupted by WD-feeding, and that these changes are dependent, in part, on the intestinal microbiome.
Collapse
Affiliation(s)
- Sarah E Martchenko
- Departments of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - David Prescott
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Alexandre Martchenko
- Departments of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Maegan E Sweeney
- Departments of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Patricia L Brubaker
- Departments of Physiology, University of Toronto, Rm 3366 Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada. .,Department of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
31
|
Dai B, Hackney JA, Ichikawa R, Nguyen A, Elstrott J, Orozco LD, Sun KH, Modrusan Z, Gogineni A, Scherl A, Gubatan J, Habtezion A, Deswal M, Somsouk M, Faubion WA, Chai A, Sharafali Z, Hassanali A, Oh YS, Tole S, McBride J, Keir ME, Yi T. Dual targeting of lymphocyte homing and retention through α4β7 and αEβ7 inhibition in inflammatory bowel disease. Cell Rep Med 2021; 2:100381. [PMID: 34467254 PMCID: PMC8385326 DOI: 10.1016/j.xcrm.2021.100381] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 03/09/2021] [Accepted: 07/22/2021] [Indexed: 01/07/2023]
Abstract
Anti-integrins are therapeutically effective for inflammatory bowel disease, yet the relative contribution of α4β7 and αEβ7 to gut lymphocyte trafficking is not fully elucidated. Here, we evaluate the effect of α4β7 and αEβ7 blockade using a combination of murine models of gut trafficking and longitudinal gene expression analysis in etrolizumab-treated patients with Crohn's disease (CD). Dual blockade of α4β7 and αEβ7 reduces CD8+ T cell accumulation in the gut to a greater extent than blockade of either integrin alone. Anti-αEβ7 reduces epithelial:T cell interactions and promotes egress of activated T cells from the mucosa into lymphatics. Inflammatory gene expression is greater in human intestinal αEβ7+ T cells. Etrolizumab-treated patients with CD display a treatment-specific reduction in inflammatory and cytotoxic intraepithelial lymphocytes (IEL) genes. Concurrent blockade of α4β7 and αEβ7 promotes reduction of cytotoxic IELs and inflammatory T cells in the gut mucosa through a stepwise inhibition of intestinal tissue entry and retention.
Collapse
Affiliation(s)
- Bingbing Dai
- Departments of Immunology Discovery, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jason A. Hackney
- OMNI Biomarker Development, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ryan Ichikawa
- Biomarker Discovery OMNI, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Allen Nguyen
- OMNI Biomarker Development, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Justin Elstrott
- Biomedical Imaging, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Luz D. Orozco
- Bioinformatics, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kai-Hui Sun
- Molecular Biology, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zora Modrusan
- Molecular Biology, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Alvin Gogineni
- Biomedical Imaging, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Alexis Scherl
- Pathology, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - John Gubatan
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Monika Deswal
- University of California, San Francisco (UCSF), San Francisco, CA 94143, USA
| | - Ma Somsouk
- University of California, San Francisco (UCSF), San Francisco, CA 94143, USA
| | - William A. Faubion
- Department of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Akiko Chai
- Product Development, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zaineb Sharafali
- Product Development, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Azra Hassanali
- Product Development, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Young S. Oh
- Product Development, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Swati Tole
- Product Development, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jacqueline McBride
- OMNI Biomarker Development, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Mary E. Keir
- Biomarker Discovery OMNI, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Tangsheng Yi
- Departments of Immunology Discovery, Genentech, Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
32
|
Lutter L, Roosenboom B, Brand EC, ter Linde JJ, Oldenburg B, van Lochem EG, Horjus Talabur Horje CS, van Wijk F. Homeostatic Function and Inflammatory Activation of Ileal CD8 + Tissue-Resident T Cells Is Dependent on Mucosal Location. Cell Mol Gastroenterol Hepatol 2021; 12:1567-1581. [PMID: 34224909 PMCID: PMC8551698 DOI: 10.1016/j.jcmgh.2021.06.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 12/10/2022]
Abstract
BACKGROUND & AIMS Tissue-resident memory T (Trm) cells, both of the CD4 and CD8 lineage, have been implicated in disease flares in inflammatory bowel disease. However, data are conflicting regarding the profile of human CD8+ Trm cells, with studies suggesting both proinflammatory and regulatory functions. It is crucial to understand the functional profile of these cells in the context of (new) therapeutic strategies targeting (trafficking of) gut Trm cells. METHODS Here, we performed imaging mass cytometry, flow cytometry, and RNA-sequencing to compare lamina propria and intraepithelial CD103+/-CD69+CD8+ Trm cells in healthy control subjects and patients with active ileal Crohn's disease. RESULTS Our data revealed that lamina propria CD103+CD69+CD8+ T cells have a classical Trm cell profile with active pathways for regulating cell survival/death and cytokine signaling, whereas intraepithelial CD103+CD69+CD8+ T cells display tightly regulated innate-like cytotoxic profile. Furthermore, within lamina propria CD8+CD103- Trm cells, an Itgb2+GzmK+KLRG1+ population distinct from CD103+ CD8+ Trm cells is found. During chronic inflammation, especially intraepithelial CD103+CD69+CD8+ T cells displayed an innate proinflammatory profile with concurrent loss of homeostatic functions. CONCLUSIONS Altogether, these compartmental and inflammation-induced differences indicate that therapeutic strategies could have a different impact on the same immune cells depending on the local compartment and presence of an inflammatory milieu, and should be taken into account when investigating short- and long-term effects of new gut T cell-targeting drugs.
Collapse
Affiliation(s)
- Lisanne Lutter
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, the Netherlands,Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Britt Roosenboom
- Department of Gastroenterology and Hepatology, Rijnstate Hospital, Arnhem, the Netherlands
| | - Eelco C. Brand
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, the Netherlands,Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - José J. ter Linde
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, the Netherlands,Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Bas Oldenburg
- Department of Gastroenterology and Hepatology, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Ellen G. van Lochem
- Department of Microbiology and Immunology, Rijnstate Hospital, Arnhem, the Netherlands
| | | | - Femke van Wijk
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht, the Netherlands,Correspondence Address correspondence to: Femke van Wijk, PhD, Centre for Translational Immunology, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, the Netherlands. fax: (088) 755-4305.
| |
Collapse
|
33
|
Chengxiao Y, Dongmei W, Kai Z, Hou L, Xiao H, Ding T, Liu D, Ye X, Linhardt RJ, Chen S. Challenges of pectic polysaccharides as a prebiotic from the perspective of fermentation characteristics and anti-colitis activity. Carbohydr Polym 2021; 270:118377. [PMID: 34364621 DOI: 10.1016/j.carbpol.2021.118377] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/26/2022]
Abstract
Several studies are described that contribute to the systematic exploration of new aspects of digestion, fermentation, and biological activities of pectic polysaccharides (PPS) leading to a better understanding of prebiotics. Inflammatory bowel disease (IBD) is thought to be associated with the dysbacteriosis induced by different environmental agents in genetically susceptible persons. PPS are considered as an indispensable gut-microbiota-accessible carbohydrate that play a dominant role in maintaining gut microbiota balance and show a better effect in ameliorating IBD than some traditional prebiotics. The aim of this review is to summarize the fermentation characteristics of PPS, highlight its role in improving IBD, and propose a view that PPS may be a new and effective prebiotic.
Collapse
Affiliation(s)
- Yu Chengxiao
- College of Biosystems, Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Wu Dongmei
- College of Biosystems, Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Zhu Kai
- College of Biosystems, Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Lijuan Hou
- College of Biosystems, Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Hang Xiao
- College of Biosystems, Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Tian Ding
- College of Biosystems, Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Donghong Liu
- College of Biosystems, Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Xingqian Ye
- College of Biosystems, Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China
| | - Robert J Linhardt
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Shiguo Chen
- College of Biosystems, Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Ningbo Research Institute, Zhejiang Engineering Laboratory of Food Technology and Equipment, Zhejiang University, Hangzhou, China.
| |
Collapse
|
34
|
Muntjewerff EM, Tang K, Lutter L, Christoffersson G, Nicolasen MJT, Gao H, Katkar GD, Das S, ter Beest M, Ying W, Ghosh P, El Aidy S, Oldenburg B, van den Bogaart G, Mahata SK. Chromogranin A regulates gut permeability via the antagonistic actions of its proteolytic peptides. Acta Physiol (Oxf) 2021; 232:e13655. [PMID: 33783968 DOI: 10.1111/apha.13655] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022]
Abstract
AIM A "leaky" gut barrier has been implicated in the initiation and progression of a multitude of diseases, for example, inflammatory bowel disease (IBD), irritable bowel syndrome and celiac disease. Here we show how pro-hormone Chromogranin A (CgA), produced by the enteroendocrine cells, and Catestatin (CST: hCgA352-372 ), the most abundant CgA-derived proteolytic peptide, affect the gut barrier. METHODS Colon tissues from region-specific CST-knockout (CST-KO) mice, CgA-knockout (CgA-KO) and WT mice were analysed by immunohistochemistry, western blot, ultrastructural and flowcytometry studies. FITC-dextran assays were used to measure intestinal barrier function. Mice were supplemented with CST or CgA fragment pancreastatin (PST: CgA250-301 ). The microbial composition of cecum was determined. CgA and CST levels were measured in blood of IBD patients. RESULTS Plasma levels of CST were elevated in IBD patients. CST-KO mice displayed (a) elongated tight, adherens junctions and desmosomes similar to IBD patients, (b) elevated expression of Claudin 2, and (c) gut inflammation. Plasma FITC-dextran measurements showed increased intestinal paracellular permeability in the CST-KO mice. This correlated with a higher ratio of Firmicutes to Bacteroidetes, a dysbiotic pattern commonly encountered in various diseases. Supplementation of CST-KO mice with recombinant CST restored paracellular permeability and reversed inflammation, whereas CgA-KO mice supplementation with CST and/or PST in CgA-KO mice showed that intestinal paracellular permeability is regulated by the antagonistic roles of these two peptides: CST reduces and PST increases permeability. CONCLUSION The pro-hormone CgA regulates the intestinal paracellular permeability. CST is both necessary and sufficient to reduce permeability and primarily acts by antagonizing PST.
Collapse
Affiliation(s)
- Elke M. Muntjewerff
- Department of Tumor Immunology Radboud Institute for Molecular Life SciencesRadboud University Medical Center Nijmegen the Netherlands
| | - Kechun Tang
- VA San Diego Healthcare System San Diego CA USA
| | - Lisanne Lutter
- Center for Translational Immunology Utrecht University Medical Center Utrecht the Netherlands
- Department of Gastroenterology and Hepatology Utrecht University Medical Center Utrecht the Netherlands
| | - Gustaf Christoffersson
- Science for Life Laboratory Uppsala University Uppsala Sweden
- Department of Medical Cell biology Uppsala University Uppsala Sweden
| | - Mara J. T. Nicolasen
- Department of Tumor Immunology Radboud Institute for Molecular Life SciencesRadboud University Medical Center Nijmegen the Netherlands
| | - Hong Gao
- Department of Medicine University of California San Diego La Jolla CA USA
| | - Gajanan D. Katkar
- Department of Cellular and Molecular Medicine University of California San Diego La Jolla CA USA
| | - Soumita Das
- Department of Pathology University of California San Diego La Jolla CA USA
| | - Martin ter Beest
- Department of Tumor Immunology Radboud Institute for Molecular Life SciencesRadboud University Medical Center Nijmegen the Netherlands
| | - Wei Ying
- Department of Medicine University of California San Diego La Jolla CA USA
| | - Pradipta Ghosh
- Department of Medicine University of California San Diego La Jolla CA USA
- Department of Cellular and Molecular Medicine University of California San Diego La Jolla CA USA
| | - Sahar El Aidy
- Department of Molecular Immunology and Microbiology Groningen Biomolecular Sciences and Biotechnology Institute University of Groningen Groningen the Netherlands
| | - Bas Oldenburg
- Department of Gastroenterology and Hepatology Utrecht University Medical Center Utrecht the Netherlands
| | - Geert van den Bogaart
- Department of Tumor Immunology Radboud Institute for Molecular Life SciencesRadboud University Medical Center Nijmegen the Netherlands
- Department of Molecular Immunology and Microbiology Groningen Biomolecular Sciences and Biotechnology Institute University of Groningen Groningen the Netherlands
| | - Sushil K. Mahata
- VA San Diego Healthcare System San Diego CA USA
- Department of Medicine University of California San Diego La Jolla CA USA
| |
Collapse
|
35
|
Revisiting the gut-joint axis: links between gut inflammation and spondyloarthritis. Nat Rev Rheumatol 2020; 16:415-433. [PMID: 32661321 DOI: 10.1038/s41584-020-0454-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2020] [Indexed: 02/07/2023]
Abstract
Gut inflammation is strongly associated with spondyloarthritis (SpA), as exemplified by the high prevalence of inflammatory bowel disease (IBD) and the even higher occurrence of subclinical gut inflammation in patients with SpA. The gut-joint axis of inflammation in SpA is further reinforced by similarities in immunopathogenesis at both anatomical sites and by the clinical success of therapies blocking TNF and IL-23 in IBD and in some forms of SpA. Many genetic risk factors are shared between SpA and IBD, and changes in the composition of gut microbiota are seen in both diseases. Current dogma is that inflammation in SpA initiates in the gut and leads to joint inflammation; however, although conceptually attractive, some research does not support this causal relationship. For example, therapies targeting IL-17A are efficacious in the joint but not the gut, and interfering with gut trafficking by targeting molecules such as α4β7 in IBD can lead to onset or flares of SpA. Several important knowledge gaps remain that must be addressed in future studies. Determining the true nature of the gut-joint axis has real-world implications for the treatment of patients with co-incident IBD and SpA and for the repurposing of therapeutics from one disease to the other.
Collapse
|
36
|
Ceramide Synthase 5 Deficiency Aggravates Dextran Sodium Sulfate-Induced Colitis and Colon Carcinogenesis and Impairs T-Cell Activation. Cancers (Basel) 2020; 12:cancers12071753. [PMID: 32630271 PMCID: PMC7409364 DOI: 10.3390/cancers12071753] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
Ceramide synthase 5 is one of six enzymes that catalyze the production of ceramides from sphingosine or sphinganine. Ceramides are important components of cell membranes and act as signaling molecules. Previously it has been shown that ceramide synthase 6 and 2 influence colitis in several animal models with sometimes opposite effects. Here, we investigated the disease course of dextran sodium sulfate-induced acute colitis and azoxymethane/dextran sodium sulfate-induced colitis-associated colon cancer in mice with global ceramide synthase 5 knockout (CerS5-ko) or with ceramide synthase 5 knockout restricted to the colon epithelium (CerS5fl/fl VilCre). We monitored disease development and analyzed colon barrier function as well as the immune cell status in these mice. CerS5-ko mice but not CerS5fl/fl-VilCre mice were more susceptible to acute and chronic inflammation. However, the cell barrier function of colon epithelial cells was not disturbed by downregulation of ceramide synthase 5. Instead, untreated CerS5-ko mice displayed reduced numbers of CD3+ immune cells in the spleen, colon, and blood, especially of intraepithelial CD8+ T-cells, which was not obvious in CerS5fl/fl Vil Cre mice. Reduced T-cell number in colon tissue of CerS5-ko mice was accompanied by a reduced expression of IL-1β, IFNγ, and IL-4. In vitro investigations revealed that knockdown of ceramide synthase 5 in T-cells impaired T-cell activation. In summary, we show that CerS5-ko mice were more susceptible to dextran sodium sulfate-induced colitis and azoxymethane/dextran sodium sulfate-induced colitis-associated colon cancer. A reduced number of T-cells in the colon epithelium that was already the case in untreated CerS5-ko mice might have contributed to this effect.
Collapse
|
37
|
Huang B, Chen Z, Geng L, Wang J, Liang H, Cao Y, Chen H, Huang W, Su M, Wang H, Xu Y, Liu Y, Lu B, Xian H, Li H, Li H, Ren L, Xie J, Ye L, Wang H, Zhao J, Chen P, Zhang L, Zhao S, Zhang T, Xu B, Che D, Si W, Gu X, Zeng L, Wang Y, Li D, Zhan Y, Delfouneso D, Lew AM, Cui J, Tang WH, Zhang Y, Gong S, Bai F, Yang M, Zhang Y. Mucosal Profiling of Pediatric-Onset Colitis and IBD Reveals Common Pathogenics and Therapeutic Pathways. Cell 2020; 179:1160-1176.e24. [PMID: 31730855 DOI: 10.1016/j.cell.2019.10.027] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/25/2019] [Accepted: 10/23/2019] [Indexed: 12/17/2022]
Abstract
Pediatric-onset colitis and inflammatory bowel disease (IBD) have significant effects on the growth of infants and children, but the etiopathogenesis underlying disease subtypes remains incompletely understood. Here, we report single-cell clustering, immune phenotyping, and risk gene analysis for children with undifferentiated colitis, Crohn's disease, and ulcerative colitis. We demonstrate disease-specific characteristics, as well as common pathogenesis marked by impaired cyclic AMP (cAMP)-response signaling. Specifically, infiltration of PDE4B- and TNF-expressing macrophages, decreased abundance of CD39-expressing intraepithelial T cells, and platelet aggregation and release of 5-hydroxytryptamine at the colonic mucosae were common in colitis and IBD patients. Targeting these pathways by using the phosphodiesterase inhibitor dipyridamole restored immune homeostasis and improved colitis symptoms in a pilot study. In summary, comprehensive analysis of the colonic mucosae has uncovered common pathogenesis and therapeutic targets for children with colitis and IBD.
Collapse
Affiliation(s)
- Bing Huang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Zhanghua Chen
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, 100871, China
| | - Lanlan Geng
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jun Wang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Huiying Liang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yujie Cao
- Department of Pediatrics and Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Huan Chen
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wanming Huang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Meiling Su
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Hanqing Wang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yanhui Xu
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yukun Liu
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Bingtai Lu
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Huifang Xian
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Huiwen Li
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Huilin Li
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Lu Ren
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Jing Xie
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Liping Ye
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Hongli Wang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Junhong Zhao
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Peiyu Chen
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Li Zhang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Shanmeizi Zhao
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ting Zhang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Banglao Xu
- Department of Laboratory Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Di Che
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wenyue Si
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Xiaoqiong Gu
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Liang Zeng
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yong Wang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Dingyou Li
- Division of Gastroenterology, Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Yifan Zhan
- Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, University of Melbourne, Parkville, Melbourne, VIC 3052, Australia
| | - David Delfouneso
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Andrew M Lew
- Walter and Eliza Hall Institute of Medical Research and Department of Medical Biology, University of Melbourne, Parkville, Melbourne, VIC 3052, Australia
| | - Jun Cui
- School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510006, China
| | - Wai Ho Tang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yan Zhang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Fan Bai
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, 100871, China; Center for Translational Cancer Research, First Hospital, Peking University, Beijing 100871, China.
| | - Min Yang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Yuxia Zhang
- Department of Gastroenterology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China; The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
38
|
Shonyela SM, Feng B, Yang W, Yang G, Wang C. The regulatory effect of Lactobacillus rhamnosus GG on T lymphocyte and the development of intestinal villi in piglets of different periods. AMB Express 2020; 10:76. [PMID: 32303860 PMCID: PMC7165236 DOI: 10.1186/s13568-020-00980-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022] Open
Abstract
The maturation and development of T cells were not completed until T cells were selected in thymus. It was not until the early 1960s that j.f.a.p. discovered the importance of thymus in T cell development. Twelve healthy piglets were randomly divided into two groups, the experimental group (LGG group) and the control group (saline group). The LGG group piglets were given 1 ml LGG (6 × 109 CFU/ml) per day. The saline group was given 1 ml of normal saline per day. The piglets were slaughtered at 30 days and 45 days, respectively, and the MLN, jejunum and ileum PPs, LP of the piglets were taken. The expression of CD3+CD4+ T lymphocytes was detected by flow cytometry, and intestinal villi development was observed by intestinal paraffin section. The results showed that the flow cytometry results at 30 days and 45 days showed that the CD3+CD4+ T lymphocytes in the MLN group were significantly different from those in the saline group (P < 0.05, P < 0.01).The CD3+CD4+ T lymphocytes in the jejunum PP of piglets in LGG group were significantly different from those in saline group (P < 0.05). The CD3+CD4+ T lymphocytes in the ileum PP of the LGG group were significantly different from those in the saline group (P < 0.05, P < 0.01). CD3+CD4+ T lymphocytes and normal saline in the piglets of the LGG group There was a significant difference between the two groups (P < 0.001, P < 0.05). P < 0.001). HE staining results showed the length of the LGG group ileal villi in piglets at 30 days, 45 days was significantly different from that in normal saline group (P < 0.01, P < 0.01). LGG can also regulate the proliferation of T lymphocytes in the intestine of early weaned piglets at 30 days and 45 days increase the number of CD3+CD4+ T lymphocytes.
Collapse
|
39
|
Hou G, Bishu S. Th17 Cells in Inflammatory Bowel Disease: An Update for the Clinician. Inflamm Bowel Dis 2020; 26:653-661. [PMID: 31970388 PMCID: PMC11491631 DOI: 10.1093/ibd/izz316] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Indexed: 12/11/2022]
Abstract
Studies in humans strongly implicate Th17 cells in the pathogenesis of inflammatory bowel disease. Thus, Th17 cells are major targets of approved and emerging biologics. Herein, we review the role of Th17 in IBD with a clinical focus.
Collapse
Affiliation(s)
- Guoqing Hou
- Division of Gastroenterology, Department of Medicine, University of Michigan, MI, USA
| | - Shrinivas Bishu
- Crohn's and Colitis Center, Division of Gastroenterology, Department of Medicine, University of Michigan, MI, USA
| |
Collapse
|
40
|
Caër C, Wick MJ. Human Intestinal Mononuclear Phagocytes in Health and Inflammatory Bowel Disease. Front Immunol 2020; 11:410. [PMID: 32256490 PMCID: PMC7093381 DOI: 10.3389/fimmu.2020.00410] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/21/2020] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a complex immune-mediated disease of the gastrointestinal tract that increases morbidity and negatively influences the quality of life. Intestinal mononuclear phagocytes (MNPs) have a crucial role in maintaining epithelial barrier integrity while controlling pathogen invasion by activating an appropriate immune response. However, in genetically predisposed individuals, uncontrolled immune activation to intestinal flora is thought to underlie the chronic mucosal inflammation that can ultimately result in IBD. Thus, MNPs are involved in fine-tuning mucosal immune system responsiveness and have a critical role in maintaining homeostasis or, potentially, the emergence of IBD. MNPs include monocytes, macrophages and dendritic cells, which are functionally diverse but highly complementary. Despite their crucial role in maintaining intestinal homeostasis, specific functions of human MNP subsets are poorly understood, especially during diseases such as IBD. Here we review the current understanding of MNP ontogeny, as well as the recently identified human intestinal MNP subsets, and discuss their role in health and IBD.
Collapse
Affiliation(s)
- Charles Caër
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mary Jo Wick
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
41
|
Barra NG, Anhê FF, Schertzer JD. Immunometabolism Sentinels: Gut Surface T-Cells Regulate GLP-1 Availability. Endocrinology 2019; 160:1177-1178. [PMID: 30958533 PMCID: PMC6480433 DOI: 10.1210/en.2019-00215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 04/02/2019] [Indexed: 11/19/2022]
Affiliation(s)
- Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Fernando F Anhê
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
- Correspondence: Jonathan D. Schertzer, PhD, Biochemistry and Biomedical Studies, McMaster University, 1280 Main Street West, HSC 4H19, Hamilton, Ontario L8S 4K1, Canada. E-mail:
| |
Collapse
|
42
|
Li Y, Sun H, Du B, Xu H. Gut Microbiota Shifts in Pup Athymic BALB/c Mice: An Updated Identification in Nude Mice. Animals (Basel) 2019; 9:ani9040151. [PMID: 30965569 PMCID: PMC6523615 DOI: 10.3390/ani9040151] [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: 03/13/2019] [Revised: 04/01/2019] [Accepted: 04/01/2019] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Mammal gut microbiota has been gradually considered to be related to innate and adaptive immunity. Incredibly, although athymic nude mouse is one of the most popular animals for modeling immunodeficiency and tumors, a basic understanding of its gut microbiota has still not been attained, and current relevant conclusions are controversial. In this 30-day study, based on high-throughput sequencing technology, we compared the differences in gut microbial community structures and functions between normal and nude pup mice, and concluded that gut microbiota shifts did occur in nude mice. These findings provide updated insight for the nude mouse tumor model. Abstract It is commonly recognized that immunodeficiency modifies the gut microbiota in mammals. However, little information on the gut microbiota is available for athymic nude mice; one of the most popular animals for modeling immunodeficiency and tumors. In this study, 16S rDNA amplicon sequencing was performed to investigate the gut microbial composition of pup nude BALB/c mice during a 30-day experimental period. In contrast to pup normal mice, pup nude mice showed a significant variation in gut microbiota. Continuously decreased dynamics of the gut bacterial Shannon index, abnormal Firmicutes/Bacteroidetes ratio, the rarity of Bifidobacterium and Lactobacillus species, and a developmental lag of gut bacterial functions were observed in nude mice. The shift in gut microbiota and abnormal colonization of beneficial bacterial species in nude mice provide an updated insight into the nude mouse tumor model and a new perspective for establishing an animal model for study on dysbacteriosis.
Collapse
Affiliation(s)
- Yuting Li
- Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Hao Sun
- Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China.
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China.
| | - Beibei Du
- Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Hui Xu
- Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China.
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China.
| |
Collapse
|