1
|
Holm M, Stepanauskaitė L, Bäckström A, Birgersson M, Socciarelli F, Archer A, Stadler C, Williams C. Spatial profiling of the mouse colonic immune landscape associated with colitis and sex. Commun Biol 2024; 7:1595. [PMID: 39613949 DOI: 10.1038/s42003-024-07276-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/15/2024] [Indexed: 12/01/2024] Open
Abstract
Inflammatory intestinal conditions are a major disease burden. Numerous factors shape the distribution of immune cells in the colon, but a spatial characterization of the homeostatic and inflamed colonic immune microenvironment is lacking. Here, we use the COMET platform for multiplex immunofluorescence to profile the infiltration of nine immune cell populations in mice of both sexes (N = 16) with full spatial context, including in regions of squamous metaplasia. Unsupervised clustering, neighborhood analysis, and manual quantification along the proximal-distal axis characterized the colonic immune landscape, quantified cell-cell interactions, and revealed sex differences. The distal colon was the most affected region during colitis, which was pronounced in males, who exhibited a sex-dependent increase of B cells and reduction of M2-like macrophages. Regions of squamous metaplasia exhibited strong infiltration of numerous immune cell populations, especially in males. Females exhibited more helper T cells and neutrophils at homeostasis and increased M2-like macrophage infiltration in the mid-colon upon colitis. Sex differences were corroborated by plasma cytokine profiles. Our results provide a foundation for future studies of inflammatory intestinal conditions.
Collapse
Affiliation(s)
- Matilda Holm
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Lina Stepanauskaitė
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Anna Bäckström
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Madeleine Birgersson
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Fabio Socciarelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Amena Archer
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Charlotte Stadler
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden
| | - Cecilia Williams
- SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Solna, Sweden.
- Division of Biosciences and Nutrition, Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
2
|
Fontdevila L, Povedano M, Domínguez R, Boada J, Serrano JC, Pamplona R, Ayala V, Portero-Otín M. Examining the complex Interplay between gut microbiota abundance and short-chain fatty acid production in amyotrophic lateral sclerosis patients shortly after onset of disease. Sci Rep 2024; 14:23497. [PMID: 39379597 PMCID: PMC11461871 DOI: 10.1038/s41598-024-75083-z] [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: 06/07/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024] Open
Abstract
This study aimed to assess differences in the enteral microbiome of relatively recent-onset amyotrophic lateral sclerosis (ALS) patients (< 6-15 months since symptom onset) compared to healthy individuals, focusing on short-chain fatty acids (SCFAs) as potential mediators of host metabolism. We included 28 volunteers (16 ALS, 12 controls) with informed consent. No significant effect of ALS on alpha diversity (measuring the variety and abundance of species within a single sample, and indicating the health and complexity of the microbiome) was observed, but ALS patients had higher abundances of Fusobacteria and Acidobacteria. ALS subtypes influenced specific species, with increased Fusobacteria and Tenericutes in spinal ALS compared to bulbar ALS. ALS patients showed increased Enterobacter, Clostridium, Veillonella, Dialister, Turicibacter, and Acidaminococcus species and decreased Prevotella, Lactobacillus, and Butyricimonas. Correlations between species varied between ALS patients and healthy individuals and among ALS subtypes. No significant differences in SCFA concentrations were found, but spinal ALS samples showed a trend towards decreased propionate content. Relationships between SCFAs and phyla colonization differed by disease status. This study suggests distinct enteral microbiome characteristics in ALS patients, though the implications are unclear. Further research is needed to determine if these differences are causative or consequential and to explore their potential as diagnostic or therapeutic targets. The study also underscores the heterogeneity of microbiome constraints in ALS and the need for more research into ALS and SCFA metabolism.
Collapse
Affiliation(s)
- Laia Fontdevila
- Department of Experimental Medicine, School of Medicine, Universitat de Lleida-IRBLleida, Rovira Roure, 80, 25198 Lleida, Spain
| | - Mònica Povedano
- Neurology Service, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Raúl Domínguez
- Neurology Service, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Jordi Boada
- Department of Experimental Medicine, School of Medicine, Universitat de Lleida-IRBLleida, Rovira Roure, 80, 25198 Lleida, Spain
| | - José Ce Serrano
- Department of Experimental Medicine, School of Medicine, Universitat de Lleida-IRBLleida, Rovira Roure, 80, 25198 Lleida, Spain
| | - Reinald Pamplona
- Department of Experimental Medicine, School of Medicine, Universitat de Lleida-IRBLleida, Rovira Roure, 80, 25198 Lleida, Spain
| | - Victòria Ayala
- Department of Experimental Medicine, School of Medicine, Universitat de Lleida-IRBLleida, Rovira Roure, 80, 25198 Lleida, Spain.
| | - Manuel Portero-Otín
- Department of Experimental Medicine, School of Medicine, Universitat de Lleida-IRBLleida, Rovira Roure, 80, 25198 Lleida, Spain.
| |
Collapse
|
3
|
Camarini R, Marianno P, Hanampa-Maquera M, Oliveira SDS, Câmara NOS. Prenatal Stress and Ethanol Exposure: Microbiota-Induced Immune Dysregulation and Psychiatric Risks. Int J Mol Sci 2024; 25:9776. [PMID: 39337263 PMCID: PMC11431796 DOI: 10.3390/ijms25189776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/22/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
Changes in maternal gut microbiota due to stress and/or ethanol exposure can have lasting effects on offspring's health, particularly regarding immunity, inflammation response, and susceptibility to psychiatric disorders. The literature search for this review was conducted using PubMed and Scopus, employing keywords and phrases related to maternal stress, ethanol exposure, gut microbiota, microbiome, gut-brain axis, diet, dysbiosis, progesterone, placenta, prenatal development, immunity, inflammation, and depression to identify relevant studies in both preclinical and human research. Only a limited number of reviews were included to support the arguments. The search encompassed studies from the 1990s to the present. This review begins by exploring the role of microbiota in modulating host health and disease. It then examines how disturbances in maternal microbiota can affect the offspring's immune system. The analysis continues by investigating the interplay between stress and dysbiosis, focusing on how prenatal maternal stress influences both maternal and offspring microbiota and its implications for susceptibility to depression. The review also considers the impact of ethanol consumption on gut dysbiosis, with an emphasis on the effects of prenatal ethanol exposure on both maternal and offspring microbiota. Finally, it is suggested that maternal gut microbiota dysbiosis may be significantly exacerbated by the combined effects of stress and ethanol exposure, leading to immune system dysfunction and chronic inflammation, which could increase the risk of depression in the offspring. These interactions underscore the potential for novel mental health interventions that address the gut-brain axis, especially in relation to maternal and offspring health.
Collapse
Affiliation(s)
- Rosana Camarini
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Priscila Marianno
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Maylin Hanampa-Maquera
- Department of Pharmacology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Samuel Dos Santos Oliveira
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| | - Niels Olsen Saraiva Câmara
- Department of Immunology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-900, Brazil
| |
Collapse
|
4
|
Bonetti L, Horkova V, Grusdat M, Longworth J, Guerra L, Kurniawan H, Franchina DG, Soriano-Baguet L, Binsfeld C, Verschueren C, Spath S, Ewen A, Koncina E, Gérardy JJ, Kobayashi T, Dostert C, Farinelle S, Härm J, Fan YT, Chen Y, Harris IS, Lang PA, Vasiliou V, Waisman A, Letellier E, Becher B, Mittelbronn M, Brenner D. A Th17 cell-intrinsic glutathione/mitochondrial-IL-22 axis protects against intestinal inflammation. Cell Metab 2024; 36:1726-1744.e10. [PMID: 38986617 DOI: 10.1016/j.cmet.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 02/06/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024]
Abstract
The intestinal tract generates significant reactive oxygen species (ROS), but the role of T cell antioxidant mechanisms in maintaining intestinal homeostasis is poorly understood. We used T cell-specific ablation of the catalytic subunit of glutamate cysteine ligase (Gclc), which impaired glutathione (GSH) production, crucially reducing IL-22 production by Th17 cells in the lamina propria, which is critical for gut protection. Under steady-state conditions, Gclc deficiency did not alter cytokine secretion; however, C. rodentium infection induced increased ROS and disrupted mitochondrial function and TFAM-driven mitochondrial gene expression, resulting in decreased cellular ATP. These changes impaired the PI3K/AKT/mTOR pathway, reducing phosphorylation of 4E-BP1 and consequently limiting IL-22 translation. The resultant low IL-22 levels led to poor bacterial clearance, severe intestinal damage, and high mortality. Our findings highlight a previously unrecognized, essential role of Th17 cell-intrinsic GSH in promoting mitochondrial function and cellular signaling for IL-22 protein synthesis, which is critical for intestinal integrity and defense against gastrointestinal infections.
Collapse
Affiliation(s)
- Lynn Bonetti
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Veronika Horkova
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Melanie Grusdat
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Joseph Longworth
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Luana Guerra
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Henry Kurniawan
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Davide G Franchina
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Leticia Soriano-Baguet
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Carole Binsfeld
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Charlène Verschueren
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Sabine Spath
- Institute of Experimental Immunology, Inflammation Research, University of Zurich, 8057 Zurich, Switzerland; Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Anouk Ewen
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Eric Koncina
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, University of Luxembourg, Belval, Luxembourg
| | - Jean-Jacques Gérardy
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg; Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg
| | - Takumi Kobayashi
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Catherine Dostert
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Sophie Farinelle
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Janika Härm
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Yu-Tong Fan
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Isaac S Harris
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Elisabeth Letellier
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, University of Luxembourg, Belval, Luxembourg
| | - Burkhard Becher
- Institute of Experimental Immunology, Inflammation Research, University of Zurich, 8057 Zurich, Switzerland
| | - Michel Mittelbronn
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg; Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg; Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), 1526 Luxembourg, Luxembourg
| | - Dirk Brenner
- Experimental and Molecular Immunology, Department of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
5
|
Jansen SA, Cutilli A, de Koning C, van Hoesel M, Frederiks CL, Saiz Sierra L, Nierkens S, Mokry M, Nieuwenhuis EE, Hanash AM, Mocholi E, Coffer PJ, Lindemans CA. Chemotherapy-induced intestinal epithelial damage directly promotes galectin-9-driven modulation of T cell behavior. iScience 2024; 27:110072. [PMID: 38883813 PMCID: PMC11176658 DOI: 10.1016/j.isci.2024.110072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/05/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
The intestine is vulnerable to chemotherapy-induced damage due to the high rate of intestinal epithelial cell (IEC) proliferation. We have developed a human intestinal organoid-based 3D model system to study the direct effect of chemotherapy-induced IEC damage on T cell behavior. Exposure of intestinal organoids to busulfan, fludarabine, and clofarabine induced damage-related responses affecting both the capacity to regenerate and transcriptional reprogramming. In ex vivo co-culture assays, prior intestinal organoid damage resulted in increased T cell activation, proliferation, and migration. We identified galectin-9 (Gal-9) as a key molecule released by damaged organoids. The use of anti-Gal-9 blocking antibodies or CRISPR/Cas9-mediated Gal-9 knock-out prevented intestinal organoid damage-induced T cell proliferation, interferon-gamma release, and migration. Increased levels of Gal-9 were found early after HSCT chemotherapeutic conditioning in the plasma of patients who later developed acute GVHD. Taken together, chemotherapy-induced intestinal damage can influence T cell behavior in a Gal-9-dependent manner which may provide novel strategies for therapeutic intervention.
Collapse
Affiliation(s)
- Suze A. Jansen
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| | - Alessandro Cutilli
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Coco de Koning
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, 3584GX Utrecht, the Netherlands
| | - Marliek van Hoesel
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| | - Cynthia L. Frederiks
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Leire Saiz Sierra
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, 3584GX Utrecht, the Netherlands
| | - Michal Mokry
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Department of Cardiology, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
| | - Edward E.S. Nieuwenhuis
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- University College Roosevelt, Utrecht University, Middelburg 4331CB, the Netherlands
| | - Alan M. Hanash
- Departments of Medicine and Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY 10065, USA
| | - Enric Mocholi
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Paul J. Coffer
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
- Center of Molecular Medicine, University Medical Center Utrecht, Utrecht 3584CG, the Netherlands
| | - Caroline A. Lindemans
- Division of Pediatrics, University Medical Center Utrecht, Utrecht 3584GX, the Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
- Regenerative Medicine Center, University Medical Center Utrecht, Utrecht 3584CT, the Netherlands
| |
Collapse
|
6
|
Wang J, He M, Yang M, Ai X. Gut microbiota as a key regulator of intestinal mucosal immunity. Life Sci 2024; 345:122612. [PMID: 38588949 DOI: 10.1016/j.lfs.2024.122612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/14/2024] [Accepted: 04/02/2024] [Indexed: 04/10/2024]
Abstract
Gut microbiota is a complex microbial community with the ability of maintaining intestinal health. Intestinal homeostasis largely depends on the mucosal immune system to defense external pathogens and promote tissue repair. In recent years, growing evidence revealed the importance of gut microbiota in shaping intestinal mucosal immunity. Therefore, according to the existing findings, this review first provided an overview of intestinal mucosal immune system before summarizing the regulatory roles of gut microbiota in intestinal innate and adaptive immunity. Specifically, this review delved into the gut microbial interactions with the cells such as intestinal epithelial cells (IECs), macrophages, dendritic cells (DCs), neutrophils, and innate lymphoid cells (ILCs) in innate immunity, and T and B lymphocytes in adaptive immunity. Furthermore, this review discussed the main effects of gut microbiota dysbiosis in intestinal diseases and offered future research prospects. The review highlighted the key regulatory roles of gut microbiota in intestinal mucosal immunity via various host-microbe interactions, providing valuable references for the development of microbial therapy in intestinal diseases.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Mei He
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Ming Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| | - Xiaopeng Ai
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
7
|
McCoy R, Oldroyd S, Yang W, Wang K, Hoven D, Bulmer D, Zilbauer M, Owens RM. In Vitro Models for Investigating Intestinal Host-Pathogen Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306727. [PMID: 38155358 PMCID: PMC10885678 DOI: 10.1002/advs.202306727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/01/2023] [Indexed: 12/30/2023]
Abstract
Infectious diseases are increasingly recognized as a major threat worldwide due to the rise of antimicrobial resistance and the emergence of novel pathogens. In vitro models that can adequately mimic in vivo gastrointestinal physiology are in high demand to elucidate mechanisms behind pathogen infectivity, and to aid the design of effective preventive and therapeutic interventions. There exists a trade-off between simple and high throughput models and those that are more complex and physiologically relevant. The complexity of the model used shall be guided by the biological question to be addressed. This review provides an overview of the structure and function of the intestine and the models that are developed to emulate this. Conventional models are discussed in addition to emerging models which employ engineering principles to equip them with necessary advanced monitoring capabilities for intestinal host-pathogen interrogation. Limitations of current models and future perspectives on the field are presented.
Collapse
Affiliation(s)
- Reece McCoy
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Sophie Oldroyd
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Woojin Yang
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Kaixin Wang
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Darius Hoven
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - David Bulmer
- Department of PharmacologyUniversity of CambridgeCambridgeCB2 1PDUK
| | - Matthias Zilbauer
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Róisín M. Owens
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| |
Collapse
|
8
|
Alaifan MA, Khayat A, Bokhary RY, Ibrahim A, Bin-Taleb Y, Alhussaini BH, Saadah OI. Prevalence of Esophageal Eosinophilia, Eosinophilic Esophagitis, and Lymphocytic Gastritis in Children with Celiac Disease: A Saudi Tertiary Center Experience. Can J Gastroenterol Hepatol 2024; 2024:5541687. [PMID: 38234411 PMCID: PMC10791472 DOI: 10.1155/2024/5541687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/02/2023] [Accepted: 12/21/2023] [Indexed: 01/19/2024] Open
Abstract
Background Celiac disease (CD) is an immune-mediated enteropathy that has been associated with other immune-related gastrointestinal disorders, such as eosinophilic esophagitis (EoE) and lymphocytic gastritis (LG). To our knowledge, this is the first study in Saudi Arabia that has described such an association. Aim To evaluate the prevalence of EoE and LG in children and adolescents with CD. Methods This was a retrospective cross-sectional study of all pediatric patients (aged 0-18 years) with CD following up at King Abdulaziz University Hospital, between January, 2014, and December, 2021. The study examined clinical, demographic, endoscopic, and histopathological data. Results Seventy-five patients with CD were included in the analysis. The median age was 12 years (range, 2-18 years). Male constituted 54.7% of the overall cohort (n = 41). The most common clinical symptoms were short stature (54.7%), weight loss (34.7%), abdominal pain (33.3%), abdominal distension (29.3%), anorexia (29.3%), diarrhea (24%), and vomiting (21.3%). The esophageal biopsy results reported were basal cell hyperplasia in 24 patients (32.9%), esophageal eosinophilia in 23 patients (31.5%), and EoE in 3 patients (4.1%). The gastric biopsy results were normal in 40 patients (53.3%). The most common abnormality was chronic inactive gastritis with no Helicobacter pylori (HP) infection (16%). LG was found in 3 patients (4%). Conclusions The prevalence of EoE in this cohort of patients with CD was lower than the prevalence recorded in a number of other studies. Further studies are needed to determine the effects of a gluten-free diet (GFD) on EOE and LG.
Collapse
Affiliation(s)
- Meshari A. Alaifan
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pediatric Gastroenterology Unit, Department of Paediatrics, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Ammar Khayat
- Pediatric Gastroenterology Unit, Department of Paediatrics, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
- Department of Pediatrics, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Rana Y. Bokhary
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulhameed Ibrahim
- Pediatric Gastroenterology Unit, Department of Paediatrics, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
- Department of Pediatrics, Hera Hospital, Makkah, Saudi Arabia
| | - Yagoub Bin-Taleb
- Pediatric Gastroenterology Unit, Department of Paediatrics, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Bakr H. Alhussaini
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pediatric Gastroenterology Unit, Department of Paediatrics, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| | - Omar I. Saadah
- Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pediatric Gastroenterology Unit, Department of Paediatrics, King Abdulaziz University Hospital, Jeddah, Saudi Arabia
| |
Collapse
|
9
|
Gambirasi M, Safa A, Vruzhaj I, Giacomin A, Sartor F, Toffoli G. Oral Administration of Cancer Vaccines: Challenges and Future Perspectives. Vaccines (Basel) 2023; 12:26. [PMID: 38250839 PMCID: PMC10821404 DOI: 10.3390/vaccines12010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Cancer vaccines, a burgeoning strategy in cancer treatment, are exploring innovative administration routes to enhance patient and medical staff experiences, as well as immunological outcomes. Among these, oral administration has surfaced as a particularly noteworthy approach, which is attributed to its capacity to ignite both humoral and cellular immune responses at systemic and mucosal tiers, thereby potentially bolstering vaccine efficacy comprehensively and durably. Notwithstanding this, the deployment of vaccines through the oral route in a clinical context is impeded by multifaceted challenges, predominantly stemming from the intricacy of orchestrating effective oral immunogenicity and necessitating strategic navigation through gastrointestinal barriers. Based on the immunogenicity of the gastrointestinal tract, this review critically analyses the challenges and recent advances and provides insights into the future development of oral cancer vaccines.
Collapse
Affiliation(s)
- Marta Gambirasi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Amin Safa
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
- Doctoral School in Pharmacological Sciences, University of Padua, 35131 Padova, Italy
- Department of Immunology, School of Medicine, Zabol University of Medical Sciences, Zabol 98616-15881, Iran
| | - Idris Vruzhaj
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
- Doctoral School in Pharmacological Sciences, University of Padua, 35131 Padova, Italy
| | - Aurora Giacomin
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Franca Sartor
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
| |
Collapse
|
10
|
Zaongo SD, Chen Y. PSGL-1, a Strategic Biomarker for Pathological Conditions in HIV Infection: A Hypothesis Review. Viruses 2023; 15:2197. [PMID: 38005875 PMCID: PMC10674231 DOI: 10.3390/v15112197] [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: 09/22/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
P-selectin glycoprotein ligand-1 (PSGL-1) has been established to be a cell adhesion molecule that is involved in the cellular rolling mechanism and the extravasation cascade, enabling the recruitment of immune cells to sites of inflammation. In recent years, researchers have established that PSGL-1 also functions as an HIV restriction factor. PSGL-1 has been shown to inhibit the HIV reverse transcription process and inhibit the infectivity of HIV virions produced by cells expressing PSGL-1. Cumulative evidence gleaned from contemporary literature suggests that PSGL-1 expression negatively affects the functions of immune cells, particularly T-cells, which are critical participants in the defense against HIV infection. Indeed, some researchers have observed that PSGL-1 expression and signaling provokes T-cell exhaustion. Additionally, it has been established that PSGL-1 may also mediate virus capture and subsequent transfer to permissive cells. We therefore believe that, in addition to its beneficial roles, such as its function as a proinflammatory molecule and an HIV restriction factor, PSGL-1 expression during HIV infection may be disadvantageous and may potentially predict HIV disease progression. In this hypothesis review, we provide substantial discussions with respect to the possibility of using PSGL-1 to predict the potential development of particular pathological conditions commonly seen during HIV infection. Specifically, we speculate that PSGL-1 may possibly be a reliable biomarker for immunological status, inflammation/translocation, cell exhaustion, and the development of HIV-related cancers. Future investigations directed towards our hypotheses may help to evolve innovative strategies for the monitoring and/or treatment of HIV-infected individuals.
Collapse
Affiliation(s)
| | - Yaokai Chen
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing 400036, China;
| |
Collapse
|
11
|
Bonetti L, Horkova V, Longworth J, Guerra L, Kurniawan H, Franchina DG, Soriano-Baguet L, Grusdat M, Spath S, Koncina E, Ewen A, Binsfeld C, Verschueren C, Gérardy JJ, Kobayashi T, Dostert C, Farinelle S, Härm J, Chen Y, Harris IS, Lang PA, Vasiliou V, Waisman A, Letellier E, Becher B, Mittelbronn M, Brenner D. A Th17 cell-intrinsic glutathione/mitochondrial-IL-22 axis protects against intestinal inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.06.547932. [PMID: 37489135 PMCID: PMC10363291 DOI: 10.1101/2023.07.06.547932] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Although the intestinal tract is a major site of reactive oxygen species (ROS) generation, the mechanisms by which antioxidant defense in gut T cells contribute to intestinal homeostasis are currently unknown. Here we show, using T cell-specific ablation of the catalytic subunit of glutamate cysteine ligase (Gclc), that the ensuing loss of glutathione (GSH) impairs the production of gut-protective IL-22 by Th17 cells within the lamina propria. Although Gclc ablation does not affect T cell cytokine secretion in the gut of mice at steady-state, infection with C. rodentium increases ROS, inhibits mitochondrial gene expression and mitochondrial function in Gclc-deficient Th17 cells. These mitochondrial deficits affect the PI3K/AKT/mTOR pathway, leading to reduced phosphorylation of the translation repressor 4E-BP1. As a consequence, the initiation of translation is restricted, resulting in decreased protein synthesis of IL-22. Loss of IL-22 results in poor bacterial clearance, enhanced intestinal damage, and high mortality. ROS-scavenging, reconstitution of IL-22 expression or IL-22 supplementation in vivo prevent the appearance of these pathologies. Our results demonstrate the existence of a previously unappreciated role for Th17 cell-intrinsic GSH coupling to promote mitochondrial function, IL-22 translation and signaling. These data reveal an axis that is essential for maintaining the integrity of the intestinal barrier and protecting it from damage caused by gastrointestinal infection.
Collapse
Affiliation(s)
- Lynn Bonetti
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Veronika Horkova
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Joseph Longworth
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Luana Guerra
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Henry Kurniawan
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Davide G. Franchina
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Leticia Soriano-Baguet
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Melanie Grusdat
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Sabine Spath
- Institute of Experimental Immunology, Inflammation Research, University of Zurich, 8057 Zurich, Switzerland
- Center for Fundamental Immunology, Benaroya Research Institute; Seattle, WA 98101, USA
| | - Eric Koncina
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, University of Luxembourg, Belval, Luxembourg
| | - Anouk Ewen
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Carole Binsfeld
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Charlène Verschueren
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Jean-Jacques Gérardy
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, L-3555, Luxembourg
| | - Takumi Kobayashi
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Catherine Dostert
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Sophie Farinelle
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Janika Härm
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Isaac S. Harris
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Philipp A. Lang
- Department of Molecular Medicine II, Medical Faculty Heinrich Heine University Düsseldorf, Germany
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Elisabeth Letellier
- Molecular Disease Mechanisms Group, Department of Life Sciences and Medicine, University of Luxembourg, Belval, Luxembourg
| | - Burkhard Becher
- Institute of Experimental Immunology, Inflammation Research, University of Zurich, 8057 Zurich, Switzerland
| | - Michel Mittelbronn
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), Dudelange, L-3555, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, L-4362, Luxembourg
- Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), Luxembourg, L-1526, Luxembourg
| | - Dirk Brenner
- Experimental and Molecular Immunology, Dept. of Infection and Immunity (DII), Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology & Genetics, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7, Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
12
|
Jensen SB, Sheikh MA, Akkouh IA, Szabo A, O’Connell KS, Lekva T, Engh JA, Agartz I, Elvsåshagen T, Ormerod MBEG, Weibell MA, Johnsen E, Kroken RA, Melle I, Drange OK, Nærland T, Vaaler AE, Westlye LT, Aukrust P, Djurovic S, Eiel Steen N, Andreassen OA, Ueland T. Elevated Systemic Levels of Markers Reflecting Intestinal Barrier Dysfunction and Inflammasome Activation Are Correlated in Severe Mental Illness. Schizophr Bull 2023; 49:635-645. [PMID: 36462169 PMCID: PMC10154716 DOI: 10.1093/schbul/sbac191] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
BACKGROUND AND HYPOTHESIS Gut microbiota alterations have been reported in severe mental illness (SMI) but fewer studies have probed for signs of gut barrier disruption and inflammation. We hypothesized that gut leakage of microbial products due to intestinal inflammation could contribute to systemic inflammasome activation in SMI. STUDY DESIGN We measured plasma levels of the chemokine CCL25 and soluble mucosal vascular addressin cell adhesion molecule-1 (sMAdCAM-1) as markers of T cell homing, adhesion and inflammation in the gut, lipopolysaccharide binding protein (LBP) and intestinal fatty acid binding protein (I-FABP) as markers of bacterial translocation and gut barrier dysfunction, in a large SMI cohort (n = 567) including schizophrenia (SCZ, n = 389) and affective disorder (AFF, n = 178), relative to healthy controls (HC, n = 418). We assessed associations with plasma IL-18 and IL-18BPa and leukocyte mRNA expression of NLRP3 and NLRC4 as markers of inflammasome activation. STUDY RESULTS Our main findings were: (1) higher levels of sMAdCAM-1 (P = .002), I-FABP (P = 7.6E-11), CCL25 (P = 9.6E-05) and LBP (P = 2.6E-04) in SMI compared to HC in age, sex, BMI, CRP and freezer storage time adjusted analysis; (2) the highest levels of sMAdCAM-1 and CCL25 (both P = 2.6E-04) were observed in SCZ and I-FABP (P = 2.5E-10) and LBP (3) in AFF; and (3), I-FABP correlated with IL-18BPa levels and LBP correlated with NLRC4. CONCLUSIONS Our findings support that intestinal barrier inflammation and dysfunction in SMI could contribute to systemic inflammation through inflammasome activation.
Collapse
Affiliation(s)
- Søren B Jensen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Mashhood A Sheikh
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Ibrahim A Akkouh
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Attila Szabo
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kevin S O’Connell
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
| | - Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - John A Engh
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Division of Mental health and Addiction, Vestfold Hospital Trust, Tønsberg, Norway
| | - Ingrid Agartz
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Torbjørn Elvsåshagen
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
| | - Monica B E G Ormerod
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Melissa A Weibell
- Division of Psychiatry, Network for Clinical Psychosis Research, Stavanger University Hospital, Stavanger, Norway
- Network for Medical Sciences, Faculty of Health, University of Stavanger, Stavanger, Norway
| | - Erik Johnsen
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- NORMENT Center of Excellence, University of Bergen and Haukeland University Hospital, Bergen, Norway
| | - Rune A Kroken
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- NORMENT Center of Excellence, University of Bergen and Haukeland University Hospital, Bergen, Norway
| | - Ingrid Melle
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole K Drange
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Østmarka, Division of Mental Health, St. Olavs University Hospital, Trondheim, Norway
- Department of Psychiatry, Sørlandet Hospital, Kristiansand, Norway
| | - Terje Nærland
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Rare Disorders, Division of Child and Adolescent medicine, Oslo University Hospital, Oslo, Norway
| | - Arne E Vaaler
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Østmarka, Division of Mental Health, St. Olavs University Hospital, Trondheim, Norway
| | - Lars T Westlye
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Nils Eiel Steen
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, NORMENT, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Center for Neurodevelopmental disorders, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Norwegian Centre for Mental Disorders Research, NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| |
Collapse
|
13
|
Yu Y, Ollodart J, Contino KF, Shiozawa Y. Immunotherapy as a potential treatment approach for currently incurable bone metastasis. J Bone Miner Metab 2023; 41:371-379. [PMID: 36752903 PMCID: PMC10251738 DOI: 10.1007/s00774-023-01404-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 01/16/2023] [Indexed: 02/09/2023]
Abstract
Once cancer metastasizes to the bone, the prognosis of cancer patients becomes extremely poor. Unfortunately, the current most successful treatment for bone metastasis can extend their survival by only a few months. Although recent studies have revealed promising impacts of cancer immunotherapies, their treatment efficacy on bone metastatic diseases remains controversial. Therefore, in this review, we discussed (i) preclinical and clinical evidence of the immunotherapeutic strategies for cancer bone metastasis, mainly focusing on cell-based immunotherapy, cytokine-based immunotherapy, and immune checkpoint blockade, and (ii) current shortcomings of immunotherapy for bone metastasis and their potential future directions. Although the evidence on treatment efficacy and safety, as well as long-term effects, is limited, immunotherapies could induce partial or complete remissions in a few cancer patients with bone metastasis. However, there are still hurdles, such as the immunosuppressive nature of the bone marrow microenvironment and poor distribution of cell-based immunotherapies to bone, that need to be overcome to enhance the treatment efficacy of immunotherapies on bone metastasis. While it is apparent that further investigation is needed regarding immunotherapeutic treatment efficacy in patients with bone metastasis, this therapy may prove to be clinically novel in this subset of cancer patients.
Collapse
Affiliation(s)
- Yang Yu
- Department of Cancer Biology and Comprehensive Cancer Center, Medical Center Blvd, Wake Forest University Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157-1082, USA
| | - Jenna Ollodart
- Department of Cancer Biology and Comprehensive Cancer Center, Medical Center Blvd, Wake Forest University Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157-1082, USA
| | - Kelly F Contino
- Department of Cancer Biology and Comprehensive Cancer Center, Medical Center Blvd, Wake Forest University Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157-1082, USA
| | - Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Medical Center Blvd, Wake Forest University Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157-1082, USA.
| |
Collapse
|
14
|
Chen C, Liu C, Zhang K, Xue W. The role of gut microbiota and its metabolites short-chain fatty acids in food allergy. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
|
15
|
Jansen SA, Cutilli A, de Koning C, van Hoesel M, Sierra LS, Nierkens S, Mokry M, Nieuwenhuis EES, Hanash AM, Mocholi E, Coffer PJ, Lindemans CA. Chemotherapy-induced intestinal injury promotes Galectin-9-driven modulation of T cell function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.30.538862. [PMID: 37163028 PMCID: PMC10168344 DOI: 10.1101/2023.04.30.538862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The intestine is vulnerable to chemotherapy-induced toxicity due to its high epithelial proliferative rate, making gut toxicity an off-target effect in several cancer treatments, including conditioning regimens for allogeneic hematopoietic cell transplantation (allo-HCT). In allo-HCT, intestinal damage is an important factor in the development of Graft-versus-Host Disease (GVHD), an immune complication in which donor immune cells attack the recipient's tissues. Here, we developed a novel human intestinal organoid-based 3D model system to study the direct effect of chemotherapy-induced intestinal epithelial damage on T cell behavior. Chemotherapy treatment using busulfan, fludarabine, and clofarabine led to damage responses in organoids resulting in increased T cell migration, activation, and proliferation in ex- vivo co-culture assays. We identified galectin-9 (Gal-9), a beta-galactoside-binding lectin released by damaged organoids, as a key molecule mediating T cell responses to damage. Increased levels of Gal-9 were also found in the plasma of allo-HCT patients who later developed acute GVHD, supporting the predictive value of the model system in the clinical setting. This study highlights the potential contribution of chemotherapy-induced epithelial damage to the pathogenesis of intestinal GVHD through direct effects on T cell activation and trafficking promoted by galectin-9.
Collapse
|
16
|
Räisänen LK, Kääriäinen SE, Sund R, Engberg E, Viljakainen HT, Kolho KL. Antibiotic exposures and the development of pediatric autoimmune diseases: a register-based case-control study. Pediatr Res 2023; 93:1096-1104. [PMID: 35854091 PMCID: PMC10033398 DOI: 10.1038/s41390-022-02188-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/27/2022] [Accepted: 06/22/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Antibiotics have been associated with several individual autoimmune diseases (ADs). This study aims to discover whether pre-diagnostic antibiotics are associated with the onset of ADs in general. METHODS From a cohort of 11,407 children, 242 developed ADs (type 1 diabetes, autoimmune thyroiditis, juvenile idiopathic arthritis (JIA), or inflammatory bowel diseases) by a median age of 16 years. Antibiotic purchases from birth until the date of diagnosis (or respective date in the matched controls n = 708) were traced from national registers. RESULTS Total number of antibiotic purchases was not related to the onset of ADs when studied as a group. Of specific diagnoses, JIA was associated with the total number of antibiotics throughout the childhood and with broad-spectrum antibiotics before the age of 3 years. Intriguingly, recent and frequent antibiotic use (within 2 years before diagnosis and ≥3 purchases) was associated with the onset of ADs (OR 1.72, 95% CI 1.08-2.74). Regardless of frequent use in childhood (40% of all antibiotics), penicillin group antibiotics were not related to any ADs. CONCLUSIONS Use of antibiotics was relatively safe regarding the overall development of ADs. However, broad-spectrum antibiotics should be used considerately as they may associate with an increased likelihood of JIA. IMPACT Increasing numbers of antibiotic purchases before the age of 3 years or throughout childhood were not associated with the development of pediatric autoimmune diseases. Broad-spectrum antibiotics were related to the development of autoimmune diseases, especially juvenile idiopathic arthritis in children, while penicillin group antibiotics were not. The use of broad-spectrum antibiotics in children should be cautious as they may carry along a risk for autoimmune disease development.
Collapse
Affiliation(s)
- Laura K Räisänen
- Faculty of Medicine and Health Technology (MET), Tampere University, Tampere, Finland
- Folkhälsan Research Center, Helsinki, Finland
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| | | | - Reijo Sund
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Elina Engberg
- Folkhälsan Research Center, Helsinki, Finland
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Heli T Viljakainen
- Folkhälsan Research Center, Helsinki, Finland
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kaija-Leena Kolho
- Faculty of Medicine and Health Technology (MET), Tampere University, Tampere, Finland.
- Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Children's Hospital, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
17
|
Räisänen L, Viljakainen H, Kolho KL. Exposure to proton pump inhibitors is associated with the development of pediatric autoimmune diseases. Front Pediatr 2023; 11:1157547. [PMID: 37051434 PMCID: PMC10083351 DOI: 10.3389/fped.2023.1157547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/08/2023] [Indexed: 04/14/2023] Open
Abstract
Proton pump inhibitors (PPIs) have been associated with decreased gut microbiota diversity. Disrupted gut microbiota composition has been reported in several autoimmune diseases (ADs), such as type 1 diabetes mellitus (DM), autoimmune thyroiditis (AIT), juvenile idiopathic arthritis (JIA), and inflammatory bowel diseases (IBD). We investigated whether PPIs are associated with the development of ADs in children and concluded that PPI exposures could be related to the onset of ADs, especially IBD and potentially AIT as well.
Collapse
Affiliation(s)
- Laura Räisänen
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
- Correspondence: Laura Räisänen Kaija-Leena Kolho
| | - Heli Viljakainen
- Public Health Research Program, Folkhälsan Research Center, Helsinki, Finland
- Children’s Hospital, University of Helsinki and HUS, Helsinki, Finland
| | - Kaija-Leena Kolho
- Children’s Hospital, University of Helsinki and HUS, Helsinki, Finland
- Faculty of Medicine and Medical Technology, Tampere University, Tampere, Finland
- Correspondence: Laura Räisänen Kaija-Leena Kolho
| |
Collapse
|
18
|
Fecal level of butyric acid, a microbiome-derived metabolite, is increased in patients with severe carotid atherosclerosis. Sci Rep 2022; 12:22378. [PMID: 36572703 PMCID: PMC9792531 DOI: 10.1038/s41598-022-26759-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022] Open
Abstract
The short-chain fatty acid (SCFA) butyric acid maintains a healthy gut barrier and vascular endothelium. We aimed to investigate the association between fecal butyric acid, carotid atherosclerosis and risk factors for ischemic stroke. Patients with severe carotid atherosclerosis (i.e. ≥ 50% stenosis) (n = 43) were compared with healthy controls (n = 38). We analyzed fecal SCFAs by gas chromatography, microbiota composition by 16S rRNA sequencing, markers of gut barrier damage and inflammasome activation by immunoassay, and plasma SCFAs by ultra-high performance liquid chromatography-tandem mass spectroscopy. Patients had higher fecal butyric acid level (p = 0.024), along with increased functional potential of microbial butyric acid production (p = 0.031), compared with controls. Dietary fiber intake was comparable. Patients had higher levels of gut barrier damage markers CCL25 and IFABP, and the inflammasome activation marker IL-18, whereas plasma level of butyric was similar. Increased fecal butyric acid was associated with higher BMI, waist-hip ratio, HbA1c, CRP and leukocyte count. Contrary to our hypothesis, patients with severe carotid atherosclerosis had higher fecal butyric acid level, and increased microbial production, compared with controls. Gut barrier damage in patients might indicate decreased absorption of butyric acid and hence contribute to the higher fecal level.
Collapse
|
19
|
Kayar Y, Dertli R, Konür Ş, Ağın M, Kafee AA, Baran B, Örmeci AÇ, Akyüz F, Demir K, Beşışık F, Kaymakoğlu S, Kaymakoglu S. Mucocutaneous Manifestations and Associated Factors in Patients with Crohn's Disease. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2022; 33:945-954. [PMID: 36098365 PMCID: PMC9797716 DOI: 10.5152/tjg.2022.21750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND One-third of all extraintestinal manifestations are mucocutaneous findings in patients with Crohn's disease and there is a relationship between some risk factors. Our aim is to evaluate factors associated with mucocutaneous manifestations in our cohort of patients with Crohn's disease with a follow-up duration of up to 25 years. METHODS In the study, 336 patients with Crohn's disease who were followed up between March 1986 and October 2011 were included. The demographic characteristics, Crohn's disease-related data, and accompanying mucocutaneous manifestations were recorded. The cumulative probability of mucocutaneous extraintestinal manifestations and possible risk factors were analyzed. RESULTS Oral and skin involvement were detected in 109 (32%) and 31 (9.2%) patients, respectively. The cumulative probability of developing oral and skin manifestations were 43.2% and 20.3%, respectively. Cox regression analysis showed that female gender (odds ratio: 3.28, 95% CI: 1.51-7.14, P = .003) and corticosteroid use (odds ratio: 7.88, 95% CI: 1.07-57.97, P = .043) are independently associated with the development of skin manifestations, while family history (odds ratio: 3.59, 95% CI: 2.18-5.93, P < .001) and inflammatory-type disease (odds ratio: 1.776, 95% CI: 1.21-2.61, P = .004) were independently associated with the development of oral ulcers. CONCLUSION Mucocutaneous extraintestinal manifestations are associated with female gender, corticosteroid use, family history, and disease type in a large cohort of patients with Crohn's disease. Defining the specific relationships of immune-mediated diseases will help to better understand the pathogenesis of Crohn's disease and associated mucocutaneous manifestations and to use more effective treatments.
Collapse
Affiliation(s)
- Yusuf Kayar
- Division of Gastroenterology, Department of Internal Medicine, Van Teaching and Research Hospital, Health Sciences University, Van, Turkey,Corresponding author: Yusuf Kayar, e-mail:
| | - Ramazan Dertli
- Division of Gastroenterology, Department of Internal Medicine, Van Teaching and Research Hospital, Health Sciences University, Van, Turkey
| | - Şevki Konür
- Department of Internal Medicine, Van Teaching and Research Hospital, Health Sciences University, Van, Turkey
| | - Mehmet Ağın
- Division of Gastroenterology, Department of Pediatrics, Van Teaching and Research Hospital, Health Sciences University, Van, Turkey
| | - Abdullah Al Kafee
- Department of Biomedical Engineering, İstanbul University, İstanbul, Turkey
| | - Bülent Baran
- Division of Gastroenterology, Department of Internal Medicine, Koç University, İstanbul, Turkey
| | - Aslı Çiftçibaşı Örmeci
- Division of Gastroenterology, Department of Internal Medicine, İstanbul University, İstanbul, Turkey
| | - Filiz Akyüz
- Division of Gastroenterology, Department of Internal Medicine, İstanbul University, İstanbul, Turkey
| | - Kadir Demir
- Division of Gastroenterology, Department of Internal Medicine, İstanbul University, İstanbul, Turkey
| | - Fatih Beşışık
- Division of Gastroenterology, Department of Internal Medicine, İstanbul University, İstanbul, Turkey
| | - Sabahattin Kaymakoğlu
- Division of Gastroenterology, Department of Internal Medicine, İstanbul University, İstanbul, Turkey
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Azarcoya-Barrera J, Lewis ED, Field CJ, Goruk S, Makarowski A, Pouliot Y, Jacobs RL, Richard C. The Lipid-Soluble Forms of Choline Enhance Ex Vivo Responses from the Gut-Associated Immune System in Young Female Rat Offspring. J Nutr 2022; 152:2604-2614. [PMID: 36774126 DOI: 10.1093/jn/nxac180] [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: 03/29/2022] [Revised: 06/16/2022] [Accepted: 08/10/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND In humans, the development of gut-associated lymphoid tissue (GALT) occurs in the first years of life and can be influenced by diet. OBJECTIVES The objective of this study was to determine the effect of dietary choline on the development of gut-associated lymphoid tissue (GALT). METHODS Three feeding trials were conducted in female Sprague-Dawley rats. Beginning 3 d before parturition (studies 1 and 3) or at day 10 of gestation (study 2), control dams consumed a 100% free choline (FC) diet until the end of the lactation period. In studies 1 and 3, test dams consumed a high-glycerophosphocholine (HGPC) diet [75% glycerophosphocholine (GPC), 12.5% phosphatidylcholine (PC), 12.5% FC] and a 100% PC diet, respectively (both 1 g of choline/kg diet). In study 2, test dams consumed a high-sphingomyelin (SM) and PC (SMPC) diet (34% SM, 37% PC, 17% GPC, 7% FC, 5% phosphocholine) or a 50% PC diet (50% PC, 25% FC, 25% GPC), both 1.7 g of choline/kg diet. Immune cell phenotypes and ex vivo cytokine production by mitogen-stimulated immune cells were measured. RESULTS Feeding of the HGPC diet lowered T-cell IL-2 (44%), IFN-γ (34%), and TNF-α (55%) production in mesenteric lymph nodes (MLNs) compared with control. Feeding both SMPC and 50% PC diets during the lactation and weaning periods increased IL-2 (54%) and TNF-α (46%) production after T-cell stimulation compared with control. There was a lower production of IL-2 (46%), IL-6 (66%), and TNF-α (45%), and a higher production of IL-10 (44%) in both SMPC and 50% PC groups following ovalbumin stimulation compared with control in MLNs. Feeding a diet containing 100% PC increased the production of IFN-γ by 52% after T-cell stimulation compared with control. CONCLUSION Feeding a diet containing a mixture of choline forms with a high content of lipid-soluble forms during both the lactation and weaning periods enhances ex vivo immune responses from the GALT in female Sprague-Dawley offspring.
Collapse
Affiliation(s)
- Jessy Azarcoya-Barrera
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Erin D Lewis
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Susan Goruk
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander Makarowski
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Yves Pouliot
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods, Université Laval, Québec, Canada
| | - René L Jacobs
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Caroline Richard
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
21
|
Ortiz-López N, Fuenzalida C, Dufeu MS, Pinto-León A, Escobar A, Poniachik J, Roblero JP, Valenzuela-Pérez L, Beltrán CJ. The immune response as a therapeutic target in non-alcoholic fatty liver disease. Front Immunol 2022; 13:954869. [PMID: 36300120 PMCID: PMC9589255 DOI: 10.3389/fimmu.2022.954869] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/21/2022] [Indexed: 08/25/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complex and heterogeneous disorder considered a liver-damaging manifestation of metabolic syndrome. Its prevalence has increased in the last decades due to modern-day lifestyle factors associated with overweight and obesity, making it a relevant public health problem worldwide. The clinical progression of NAFLD is associated with advanced forms of liver injury such as fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). As such, diverse pharmacological strategies have been implemented over the last few years, principally focused on metabolic pathways involved in NAFLD progression. However, a variable response rate has been observed in NAFLD patients, which is explained by the interindividual heterogeneity of susceptibility to liver damage. In this scenario, it is necessary to search for different therapeutic approaches. It is worth noting that chronic low-grade inflammation constitutes a central mechanism in the pathogenesis and progression of NAFLD, associated with abnormal composition of the intestinal microbiota, increased lymphocyte activation in the intestine and immune effector mechanisms in liver. This review aims to discuss the current knowledge about the role of the immune response in NAFLD development. We have focused mainly on the impact of altered gut-liver-microbiota axis communication on immune cell activation in the intestinal mucosa and the role of subsequent lymphocyte homing to the liver in NAFLD development. We further discuss novel clinical trials that addressed the control of the liver and intestinal immune response to complement current NAFLD therapies.
Collapse
Affiliation(s)
- Nicolás Ortiz-López
- Laboratory of Immunogastroenterology, Unit of Gastroenterology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Catalina Fuenzalida
- Laboratory of Immunogastroenterology, Unit of Gastroenterology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - María Soledad Dufeu
- Laboratory of Immunogastroenterology, Unit of Gastroenterology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Araceli Pinto-León
- Laboratory of Immunogastroenterology, Unit of Gastroenterology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
| | | | - Jaime Poniachik
- Unit of Gastroenterology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Juan Pablo Roblero
- Unit of Gastroenterology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Lucía Valenzuela-Pérez
- Laboratory of Immunogastroenterology, Unit of Gastroenterology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Caroll J. Beltrán
- Laboratory of Immunogastroenterology, Unit of Gastroenterology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
22
|
Pandrea I, Brooks K, Desai RP, Tare M, Brenchley JM, Apetrei C. I've looked at gut from both sides now: Gastrointestinal tract involvement in the pathogenesis of SARS-CoV-2 and HIV/SIV infections. Front Immunol 2022; 13:899559. [PMID: 36032119 PMCID: PMC9411647 DOI: 10.3389/fimmu.2022.899559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/25/2022] [Indexed: 01/08/2023] Open
Abstract
The lumen of the gastrointestinal (GI) tract contains an incredibly diverse and extensive collection of microorganisms that can directly stimulate the immune system. There are significant data to demonstrate that the spatial localization of the microbiome can impact viral disease pathogenesis. Here we discuss recent studies that have investigated causes and consequences of GI tract pathologies in HIV, SIV, and SARS-CoV-2 infections with HIV and SIV initiating GI pathology from the basal side and SARS-CoV-2 from the luminal side. Both these infections result in alterations of the intestinal barrier, leading to microbial translocation, persistent inflammation, and T-cell immune activation. GI tract damage is one of the major contributors to multisystem inflammatory syndrome in SARS-CoV-2-infected individuals and to the incomplete immune restoration in HIV-infected subjects, even in those with robust viral control with antiretroviral therapy. While the causes of GI tract pathologies differ between these virus families, therapeutic interventions to reduce microbial translocation-induced inflammation and improve the integrity of the GI tract may improve the prognoses of infected individuals.
Collapse
Affiliation(s)
- Ivona Pandrea
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kelsie Brooks
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Rahul P. Desai
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Minali Tare
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jason M. Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Cristian Apetrei
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
23
|
Elucidating the Role of Innate and Adaptive Immune Responses in the Pathogenesis of Canine Chronic Inflammatory Enteropathy-A Search for Potential Biomarkers. Animals (Basel) 2022; 12:ani12131645. [PMID: 35804545 PMCID: PMC9264988 DOI: 10.3390/ani12131645] [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] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/11/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Canine chronic inflammatory enteropathy (CIE) is a chronic disease affecting the small or large intestine and, in some cases, the stomach of dogs. This gastrointestinal disorder is common and is characterized by recurrent vomiting, diarrhea, and weight loss in affected dogs. The pathogenesis of IBD is not completely understood. Similar to human IBD, potential disease factors include genetics, environmental exposures, and dysregulation of the microbiota and the immune response. Some important components of the innate and adaptive immune response involved in CIE pathogenesis have been described. However, the immunopathogenesis of the disease has not been fully elucidated. In this review, we summarized the literature associated with the different cell types and molecules involved in the immunopathogenesis of CIE, with the aim of advancing the search for biomarkers with possible diagnostic, prognostic, or therapeutic utility. Abstract Canine chronic inflammatory enteropathy (CIE) is one of the most common chronic gastrointestinal diseases affecting dogs worldwide. Genetic and environmental factors, as well as intestinal microbiota and dysregulated host immune responses, participate in this multifactorial disease. Despite advances explaining the immunological and molecular mechanisms involved in CIE development, the exact pathogenesis is still unknown. This review compiles the latest reports and advances that describe the main molecular and cellular mechanisms of both the innate and adaptive immune responses involved in canine CIE pathogenesis. Future studies should focus research on the characterization of the immunopathogenesis of canine CIE in order to advance the establishment of biomarkers and molecular targets of diagnostic, prognostic, or therapeutic utility.
Collapse
|
24
|
Zhang R, Zhang Q, Chen Y, Zhao Q, Zhang B, Wang L, Zhou C, Zhang Q, Chen K, Zhang Y, Hou X, Chen H, Liu X, Ni M, Jiang B. Combined treatment with Rg1 and adipose-derived stem cells alleviates DSS-induced colitis in a mouse model. Stem Cell Res Ther 2022; 13:272. [PMID: 35729638 PMCID: PMC9210677 DOI: 10.1186/s13287-022-02940-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/13/2021] [Indexed: 11/27/2022] Open
Abstract
Background Inflammatory bowel diseases, consisting of Crohn’s disease and ulcerative colitis constitute chronic inflammatory conditions that may compromise the whole gastrointestinal tract as well as the colonic mucosa. Currently, there are no curative interventions for IBD, and all available treatments have side effects that limit their use. Adipose-derived stem cell (ADSC) treatment is a prospective treatment option for IBD. Previous findings indicated that ginsenoside (Rg1) dampened inflammatory diseases like colitis by inhibiting the binding of LPS to TLR4 on macrophages and restoring the Th17/Treg ratio. The purpose of this work was to investigate whether Rg1 can increase the influence of ADSC in a mouse model of colitis triggered by dextran sulfate sodium (DSS). Methods ADSC was intravenously inoculated into mice with DSS-triggered colitis, while Rg1 was delivered via oral gavage. Colon inflammation was assessed via body weight, colon length along with H&E staining. Serum cytokine levels were measured using ELISA. Besides, flow cytometry was adopted to determine the percentage, as well as FMI of immune cells in the spleen. The effects of simultaneous Rg1 and ADSC treatment on TLR4-MyD88 signaling were assessed via immunofluorescence. Results Rg1 and ADSC effectively alleviated the impacts of colon inflammation, weight loss, and colon length reduction along with histological score. Treatment with Rg1 and ADSC reduced serum levels of the proinflammatory cytokines, IL-1β, TNF-α, IL-6, IL-4, and IL-17A and upregulated the level of immunosuppressive cytokine, IL-10. Compared with ADSC or Rg1 alone, combined treatment with Rg1 and ADSC significantly improved the structure of microbial community. Additionally, treatment with Rg1 plus ADSC selectively elevated the level of splenic regulatory T (Treg) cells and downregulated the proportion of T helper type 17 (Th17) cells, indicating restoration of intestinal homeostasis. Besides, we established that the combination of ADSC + Rg1 restored immunological balance more effectively than either ADSC or Rg1 alone, illustrating that Rg1's modulatory function on the gut microbiota may boost the impact of ADSCs in restoration of the immune balance. ADSC combined with Rg1 might downregulate the expression of TLR4 and MyD88, thereby suppressing TLR4-MyD8 signaling. The immunofluorescence results also suggested that co-therapy with Rg-1 and ADSC may optimize treatment strategies of IBD. Conclusions Here, we find that the combination of Rg1 and ADSC alleviates DSS-induced colitis in a mouse model more efficiently than ADSC alone, indicating that Rg1 enhances the effect of ADSC against colitis. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02940-x.
Collapse
Affiliation(s)
- Rui Zhang
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China.,Graduate School of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Qingqing Zhang
- Department of Pathogen Biology, Key Laboratory of Pathogen of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China
| | - Yanni Chen
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China
| | - Qing Zhao
- Department of Pathogen Biology, Key Laboratory of Pathogen of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China
| | - Bo Zhang
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China
| | - Ling Wang
- Graduate School of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Chungen Zhou
- Colorectal Disease Center of Nanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China
| | - Qi Zhang
- Graduate School of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Kun Chen
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China
| | - Yuqing Zhang
- Graduate School of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Xiaotao Hou
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China
| | - Hao Chen
- Anorectal Surgery of Zhongda Hospital Southeast University, Nanjing, 210009, China
| | - Xingyin Liu
- Department of Pathogen Biology, Key Laboratory of Pathogen of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, China.
| | - Min Ni
- Colorectal Disease Center of Nanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China.
| | - Bin Jiang
- Colorectal Disease Center of Nanjing Hospital of Chinese Medicine, Affiliated to Nanjing University of Chinese Medicine, Nanjing, 210022, China.
| |
Collapse
|
25
|
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: 1.7] [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
|
26
|
Beukema M, Jermendi É, Oerlemans M, Logtenberg M, Akkerman R, An R, van den Berg M, Zoetendal E, Koster T, Kong C, Faas M, Schols H, de Vos P. The level and distribution of methyl-esters influence the impact of pectin on intestinal T cells, microbiota, and Ahr activation. Carbohydr Polym 2022; 286:119280. [DOI: 10.1016/j.carbpol.2022.119280] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/04/2022] [Accepted: 02/19/2022] [Indexed: 12/16/2022]
|
27
|
Pagani K, Lukac D, Bhukhan A, McGee JS. Cutaneous Manifestations of Inflammatory Bowel Disease: A Basic Overview. Am J Clin Dermatol 2022; 23:481-497. [PMID: 35441942 DOI: 10.1007/s40257-022-00689-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2022] [Indexed: 11/30/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal (GI) tract that is subdivided into Crohn's disease (CD) and ulcerative colitis (UC). CD is characterized by involvement of the entire GI tract, while UC mainly affects the distal GI tract. Moreover, both CD and UC can present with extraintestinal manifestations (EIMs) of the disease affecting multiple organ systems including the hepatobiliary tract, kidney, bones, eyes, joints, and skin. These complications can cause significant morbidity and negatively impact the quality of life for IBD patients. Although the pathogenesis of EIMs is not clearly elucidated, it is postulated that the diseased GI mucosa similarly stimulates excess immune responses at the extraintestinal sites. Cutaneous EIMs occur in up to 15% of patients with IBD, often predating their IBD diagnosis. They are categorized into (1) specific, (2) reactive, (3) associated, and (4) treatment-induced. Here, we review the epidemiological, clinical, diagnostic, and histologic features of the most commonly described cutaneous EIMs of IBD along with their respective treatment options.
Collapse
Affiliation(s)
- Kyla Pagani
- Department of Dermatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Danitza Lukac
- Department of Dermatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Aashni Bhukhan
- Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Ft. Lauderdale, FL, USA
| | - Jean S McGee
- Department of Dermatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Gutierrez MW, van Tilburg Bernardes E, Changirwa D, McDonald B, Arrieta MC. "Molding" immunity-modulation of mucosal and systemic immunity by the intestinal mycobiome in health and disease. Mucosal Immunol 2022; 15:573-583. [PMID: 35474360 DOI: 10.1038/s41385-022-00515-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/24/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023]
Abstract
Fungi are important yet understudied contributors to the microbial communities of the gastrointestinal tract. Starting at birth, the intestinal mycobiome undergoes a period of dynamic maturation under the influence of microbial, host, and extrinsic influences, with profound functional implications for immune development in early life, and regulation of immune homeostasis throughout life. Candida albicans serves as a model organism for understanding the cross-talk between fungal colonization dynamics and immunity, and exemplifies unique mechanisms of fungal-immune interactions, including fungal dimorphism, though our understanding of other intestinal fungi is growing. Given the prominent role of the gut mycobiome in promoting immune homeostasis, emerging evidence points to fungal dysbiosis as an influential contributor to immune dysregulation in a variety of inflammatory and infectious diseases. Here we review current knowledge on the factors that govern host-fungi interactions in the intestinal tract and immunological outcomes in both mucosal and systemic compartments.
Collapse
Affiliation(s)
- Mackenzie W Gutierrez
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada
| | - Erik van Tilburg Bernardes
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada
| | - Diana Changirwa
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Braedon McDonald
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,International Microbiome Centre, University of Calgary, Calgary, AB, Canada.,Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Immunology Research Group, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada. .,Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada. .,Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada. .,International Microbiome Centre, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
29
|
Contino KF, Yadav H, Shiozawa Y. The gut microbiota can be a potential regulator and treatment target of bone metastasis. Biochem Pharmacol 2022; 197:114916. [PMID: 35041811 PMCID: PMC8858876 DOI: 10.1016/j.bcp.2022.114916] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 02/08/2023]
Abstract
The gut microbiota, an often forgotten organ, have a tremendous impact on human health. It has long been known that the gut microbiota are implicated in cancer development, and more recently, the gut microbiota have been shown to influence cancer metastasis to distant organs. Although one of the most common sites of distant metastasis is the bone, and the skeletal system has been shown to be a subject of interactions with the gut microbiota to regulate bone homeostasis, little research has been done regarding how the gut microbiota control the development of bone metastasis. This review will discuss the mechanisms through which the gut microbiota and derived microbial compounds (i) regulate gastrointestinal cancer disease progression and metastasis, (ii) influence skeletal remodeling and potentially modulate bone metastasis, and (iii) affect and potentially enhance immunotherapeutic treatments for bone metastasis.
Collapse
Affiliation(s)
- Kelly F Contino
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Hariom Yadav
- Department of Neurosurgery and Brain Repair and Institute for Microbiome, University of South Florida, Tampa, FL 33612, USA
| | - Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA.
| |
Collapse
|
30
|
Pantic I, Jevtic D, Nordstrom CW, Madrid C, Milovanovic T, Dumic I. Clinical Manifestations of Leukocytoclastic Vasculitis, Treatment, and Outcome in Patients with Ulcerative Colitis: A Systematic Review of the Literature. J Clin Med 2022; 11:739. [PMID: 35160187 PMCID: PMC8836768 DOI: 10.3390/jcm11030739] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 12/30/2022] Open
Abstract
Leukocytoclastic vasculitis (LCV) is a rare extraintestinal manifestation (EIM) of ulcerative colitis (UC). Observations about its association with UC stem from case reports and small case series. Due to its rarity, more rigorous cross-sectional studies are scarce and difficult to conduct. The aim of this systematic review was to synthetize the knowledge on this association by reviewing published literature in the form of both case reports and case series; and report the findings according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. In contrast to LCV in Chron disease (CD), which occurs secondary to biologic therapies used for its treatment, LCV in UC is a true reactive skin manifestation. Both genders are equally affected. Palpable purpura (41%) and erythematous plaques (27%) are the most common clinical manifestations. In 41% of patients, the rash is painful, and the lower extremities are most commonly involved (73%). Systemic symptoms such as fever, arthralgias, fatigue, and malaise are seen in 60% of patients. Unlike previous reports, we found that LCV more commonly occurs after the UC diagnosis (59%), and 68% of patients have active intestinal disease at the time of LCV diagnosis. Antineutrophil cytoplasmic antibody (ANCA) is positive in 41% of patients, and 36% of patients have other EIMs present concomitantly with LCV. The majority of patients were treated with corticosteroids (77%), and two (10%) required colectomy to control UC and LCV symptoms. Aside from one patient who died from unrelated causes, all others survived with their rash typically resolving without scarring (82%).
Collapse
Affiliation(s)
- Ivana Pantic
- Clinic of Gastroenterology and Hepatology, University Clinical Centre of Serbia, 11000 Belgrade, Serbia; (I.P.); (T.M.)
| | - Djordje Jevtic
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Charles W. Nordstrom
- Department of Hospital Medicine, Mayo Clinic Health System, Eau Claire, WI 54702, USA; (C.W.N.); (C.M.)
- Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
| | - Cristian Madrid
- Department of Hospital Medicine, Mayo Clinic Health System, Eau Claire, WI 54702, USA; (C.W.N.); (C.M.)
- Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
| | - Tamara Milovanovic
- Clinic of Gastroenterology and Hepatology, University Clinical Centre of Serbia, 11000 Belgrade, Serbia; (I.P.); (T.M.)
- School of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Igor Dumic
- Department of Hospital Medicine, Mayo Clinic Health System, Eau Claire, WI 54702, USA; (C.W.N.); (C.M.)
- Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
31
|
Kottom TJ, Carmona EM, Schaefbauer K, Limper AH. Additional C-type lectin receptors mediate interactions with Pneumocystis organisms and major surface glycoprotein. J Med Microbiol 2021; 70:001470. [PMID: 34889727 PMCID: PMC8744274 DOI: 10.1099/jmm.0.001470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Introduction. Pathogen-associated molecular patterns' (PAMPs) are microbial signatures that are recognized by host myeloid C-type lectin receptors (CLRs). These CLRs interact with micro-organisms via their carbohydrate recognition domains (CRDs) and engage signalling pathways within the cell resulting in pro-inflammatory and microbicidal responses.Gap statement. In this article, we extend our laboratory study of additional CLRs that recognize fungal ligands against Pneumocystis murina and Pneumocystis carinii and their purified major surface glycoproteins (Msgs).Aim. To study the potential of newly synthesized hFc-CLR fusions on binding to Pneumocystis and its Msg.Methods. A library of new synthesized hFc-CLR fusions was screened against Pneumocystis murina and Pneumocystis carinii organisms and their purified major surface glycoproteins (Msgs) found on the respective fungi via modified ELISA. Immunofluorescence assay (IFA) was implemented and quantified to verify results. mRNA expression analysis by quantitative PCR (q-PCR) was employed to detect respective CLRs found to bind fungal organisms in the ELISA and determine their expression levels in the mouse immunosuppressed Pneumocystis pneumonia (PCP) model.Results. We detected a number of the CLR hFc-fusions displayed significant binding with P. murina and P. carinii organisms, and similarly to their respective Msgs. Significant organism and Msg binding was observed for CLR members C-type lectin domain family 12 member A (CLEC12A), Langerin, macrophage galactose-type lectin-1 (MGL-1), and specific intracellular adhesion molecule-3 grabbing non-integrin homologue-related 3 (SIGNR3). Immunofluorescence assay (IFA) with the respective CLR hFc-fusions against whole P. murina life forms corroborated these findings. Lastly, we surveyed the mRNA expression profiles of the respective CLRs tested above in the mouse immunosuppressed Pneumocystis pneumonia (PCP) model and determined that macrophage galactose type C-type lectin (Mgl-1), implicated in recognizing terminal N-acetylgalactosamine (GalNAc) found in the glycoproteins of microbial pathogens was significantly up-regulated during infection.Conclusion. The data herein add to the growing list of CLRs recognizing Pneumocystis and provide insights for further study of organism/host immune cell interactions.
Collapse
Affiliation(s)
- Theodore J. Kottom
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester, MN, USA,*Correspondence: Theodore J. Kottom,
| | - Eva M. Carmona
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Kyle Schaefbauer
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Andrew H. Limper
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester, MN, USA
| |
Collapse
|
32
|
Frigerio S, Lartey DA, D’Haens GR, Grootjans J. The Role of the Immune System in IBD-Associated Colorectal Cancer: From Pro to Anti-Tumorigenic Mechanisms. Int J Mol Sci 2021; 22:12739. [PMID: 34884543 PMCID: PMC8657929 DOI: 10.3390/ijms222312739] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Patients with inflammatory bowel disease (IBD) have increased incidence of colorectal cancer (CRC). IBD-associated cancer follows a well-characterized sequence of intestinal epithelial changes, in which genetic mutations and molecular aberrations play a key role. IBD-associated cancer develops against a background of chronic inflammation and pro-inflammatory immune cells, and their products contribute to cancer development and progression. In recent years, the effect of the immunosuppressive microenvironment in cancer development and progression has gained more attention, mainly because of the unprecedented anti-tumor effects of immune checkpoint inhibitors in selected groups of patients. Even though IBD-associated cancer develops in the background of chronic inflammation which is associated with activation of endogenous anti-inflammatory or suppressive mechanisms, the potential role of an immunosuppressive microenvironment in these cancers is largely unknown. In this review, we outline the role of the immune system in promoting cancer development in chronic inflammatory diseases such as IBD, with a specific focus on the anti-inflammatory mechanisms and suppressive immune cells that may play a role in IBD-associated tumorigenesis.
Collapse
Affiliation(s)
- Sofía Frigerio
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.F.); (D.A.L.); (G.R.D.)
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Dalia A. Lartey
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.F.); (D.A.L.); (G.R.D.)
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Location AMC, 1105 AZ Amsterdam, The Netherlands
| | - Geert R. D’Haens
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.F.); (D.A.L.); (G.R.D.)
| | - Joep Grootjans
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.F.); (D.A.L.); (G.R.D.)
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Location AMC, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
33
|
Rousta E, Oka A, Liu B, Herzog J, Bhatt AP, Wang J, Habibi Najafi MB, Sartor RB. The Emulsifier Carboxymethylcellulose Induces More Aggressive Colitis in Humanized Mice with Inflammatory Bowel Disease Microbiota Than Polysorbate-80. Nutrients 2021; 13:3565. [PMID: 34684567 PMCID: PMC8540676 DOI: 10.3390/nu13103565] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/27/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022] Open
Abstract
Commonly used synthetic dietary emulsifiers, including carboxymethylcellulose (CMC) and polysorbate-80 (P80), promote intestinal inflammation. We compared abilities of CMC vs. P80 to potentiate colitis and impact human microbiota in an inflammatory environment using a novel colitis model of ex-germ-free (GF) IL10-/- mice colonized by pooled fecal transplant from three patients with active inflammatory bowel diseases. After three days, mice received 1% CMC or P80 in drinking water or water alone for four weeks. Inflammation was quantified by serial fecal lipocalin 2 (Lcn-2) and after four weeks by blinded colonic histologic scores and colonic inflammatory cytokine gene expression. Microbiota profiles in cecal contents were determined by shotgun metagenomic sequencing. CMC treatment significantly increased fecal Lcn-2 levels compared to P80 and water treatment by one week and throughout the experiment. Likewise, CMC treatment increased histologic inflammatory scores and colonic inflammatory cytokine gene expression compared with P80 and water controls. The two emulsifiers differentially affected specific intestinal microbiota. CMC did not impact bacterial composition but significantly decreased Caudoviricetes (bacteriophages), while P80 exposure non-significantly increased the abundance of both Actinobacteria and Proteobacteria. Commonly used dietary emulsifiers have different abilities to induce colitis in humanized mice. CMC promotes more aggressive inflammation without changing bacterial composition.
Collapse
Affiliation(s)
- Esmat Rousta
- Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC 27599, USA; (E.R.); (A.O.); (B.L.); (J.H.); (A.P.B.)
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| | - Akihiko Oka
- Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC 27599, USA; (E.R.); (A.O.); (B.L.); (J.H.); (A.P.B.)
- Department of Internal Medicine II, Faculty of Medicine, Shimane University, 89-1, Enya, Izumo, Shimane 693-8501, Japan
| | - Bo Liu
- Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC 27599, USA; (E.R.); (A.O.); (B.L.); (J.H.); (A.P.B.)
| | - Jeremy Herzog
- Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC 27599, USA; (E.R.); (A.O.); (B.L.); (J.H.); (A.P.B.)
| | - Aadra P. Bhatt
- Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC 27599, USA; (E.R.); (A.O.); (B.L.); (J.H.); (A.P.B.)
- Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC 27599, USA;
| | - Jeremy Wang
- Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC 27599, USA;
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Mohammad B. Habibi Najafi
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| | - Ryan Balfour Sartor
- Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC 27599, USA; (E.R.); (A.O.); (B.L.); (J.H.); (A.P.B.)
- Center for Gastrointestinal Biology and Disease, School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC 27599, USA;
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC 27599, USA
| |
Collapse
|
34
|
da Costa Ferreira S, Sadissou IA, Parra RS, Feitosa MR, Neto FSL, Pretti da Cunha Tirapelli D, Ramalho LNZ, Féres O, da Rocha JJR, Donadi EA, de Almeida Troncon LE. Increased HLA-G Expression in Tissue-Infiltrating Cells in Inflammatory Bowel Diseases. Dig Dis Sci 2021; 66:2610-2618. [PMID: 32839905 DOI: 10.1007/s10620-020-06561-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/11/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Since HLA-G is an immune checkpoint molecule and since Crohn's disease (CD) and ulcerative colitis (UC) exhibit deregulated immune-mediated mechanisms, we aimed to evaluate intestinal HLA-G expression and soluble HLA-G (sHLA-G) levels in CD/UC patients stratified according to the CD phenotype/localization and UC extension. METHODS HLA-G tissue expression was assessed by immunohistochemistry in biopsies collected from 151 patients (90 CD, 61 UC) and in surgical resection specimens (28 CD, 12 UC). Surgical material from 24 healthy controls was also assessed. Plasma sHLA-G levels (97 CD, 81 UC, and 120 controls) were evaluated using ELISA. RESULTS HLA-G expression was similarly observed in the intestinal epithelial cells of control and CD/UC specimens. However, in biopsies, the plasma cells/lymphocytes infiltrating the lamina propria in CD/UC presented (1) increased HLA-G expression compared to controls (P < 0.0001), (2) greater cell staining in UC cells than in CD cells irrespective of disease extent (P = 0.0011), and (3) an increased number of infiltrating cells in the inflammatory CD phenotype compared to that in the stenosing and fistulizing phenotypes (P = 0.0407). In surgical specimens, CD/UC patients exhibited higher infiltrating cell HLA-G expression in lesion areas than in margins. sHLA-G levels were higher in UC/CD patients (P < 0.0001) than in controls, but no difference was observed between diseases. CONCLUSIONS Increased infiltrating cell HLA-G expression associated with increased sHLA-G levels in CD/UC patients may reflect ongoing host strategies to suppress chronic inflammation.
Collapse
Affiliation(s)
- Sandro da Costa Ferreira
- Division of Gastroenterology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brazil.
| | - Ibrahim Abiodoun Sadissou
- Division of Clinical Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rogério Serafim Parra
- Division of Coloproctology, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marley Ribeiro Feitosa
- Division of Coloproctology, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fermino Sanches Lizarte Neto
- Molecular Biology Laboratory, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniela Pretti da Cunha Tirapelli
- Molecular Biology Laboratory, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Omar Féres
- Division of Coloproctology, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - José Joaquim Ribeiro da Rocha
- Division of Coloproctology, Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Eduardo Antônio Donadi
- Division of Clinical Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luiz Ernesto de Almeida Troncon
- Division of Gastroenterology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, SP, 14049-900, Brazil
| |
Collapse
|
35
|
Lazzaro A, Innocenti GP, Santinelli L, Pinacchio C, De Girolamo G, Vassalini P, Fanello G, Mastroianni CM, Ceccarelli G, d’Ettorre G. Antiretroviral Therapy Dampens Mucosal CD4 + T Lamina Propria Lymphocytes Immune Activation in Long-Term Treated People Living with HIV-1. Microorganisms 2021; 9:microorganisms9081624. [PMID: 34442703 PMCID: PMC8402205 DOI: 10.3390/microorganisms9081624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/29/2021] [Accepted: 07/21/2021] [Indexed: 01/08/2023] Open
Abstract
HIV infection is characterized by a severe deterioration of an immune cell-mediated response due to a progressive loss of CD4+ T cells from gastrointestinal tract, with a preferential loss of IL-17 producing Th cells (Th17), a specific CD4+ T cells subset specialized in maintaining mucosal integrity and antimicrobial inflammatory responses. To address the effectiveness of antiretroviral therapy (ART) in reducing chronic immunological dysfunction and immune activation of intestinal mucosa, we conducted a cross-sectional observational study comparing total IFN-γ-expressing (Th1) and IL-17-expressing (Th17) frequencies of CD4+ T lamina propria lymphocytes (LPLs) and their immune activation status between 11 male ART-naïve and 11 male long-term ART-treated people living with HIV-1 (PLWH) who underwent colonoscopy and retrograde ileoscopy for biopsies collection. Flow cytometry for surface and intracellular staining was performed. Long-term ART-treated PLWH showed lower levels of CD38+ and/or HLA-DR+ LPLs compared to ART-naïve PLWH. Frequencies of Th1 and Th17 LPLs did not differ between the two groups. Despite ART failing to restore the Th1 and Th17 levels within the gut mucosa, it is effective in increasing overall CD4+ T LPLs frequencies and reducing mucosal immune activation.
Collapse
Affiliation(s)
- Alessandro Lazzaro
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Policlinico Umberto I of Rome, 00185 Roma, Italy; (L.S.); (C.P.); (G.D.G.); (P.V.); (C.M.M.); (G.C.); (G.d.)
- Correspondence: (A.L.); (G.P.I.)
| | - Giuseppe Pietro Innocenti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Policlinico Umberto I of Rome, 00185 Roma, Italy; (L.S.); (C.P.); (G.D.G.); (P.V.); (C.M.M.); (G.C.); (G.d.)
- Correspondence: (A.L.); (G.P.I.)
| | - Letizia Santinelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Policlinico Umberto I of Rome, 00185 Roma, Italy; (L.S.); (C.P.); (G.D.G.); (P.V.); (C.M.M.); (G.C.); (G.d.)
| | - Claudia Pinacchio
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Policlinico Umberto I of Rome, 00185 Roma, Italy; (L.S.); (C.P.); (G.D.G.); (P.V.); (C.M.M.); (G.C.); (G.d.)
| | - Gabriella De Girolamo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Policlinico Umberto I of Rome, 00185 Roma, Italy; (L.S.); (C.P.); (G.D.G.); (P.V.); (C.M.M.); (G.C.); (G.d.)
| | - Paolo Vassalini
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Policlinico Umberto I of Rome, 00185 Roma, Italy; (L.S.); (C.P.); (G.D.G.); (P.V.); (C.M.M.); (G.C.); (G.d.)
| | - Gianfranco Fanello
- Department of Emergency Surgery-Emergency Endoscopic Unit, Sapienza University of Rome, Policlinico Umberto I, 00185 Roma, Italy;
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Policlinico Umberto I of Rome, 00185 Roma, Italy; (L.S.); (C.P.); (G.D.G.); (P.V.); (C.M.M.); (G.C.); (G.d.)
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Policlinico Umberto I of Rome, 00185 Roma, Italy; (L.S.); (C.P.); (G.D.G.); (P.V.); (C.M.M.); (G.C.); (G.d.)
| | - Gabriella d’Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Policlinico Umberto I of Rome, 00185 Roma, Italy; (L.S.); (C.P.); (G.D.G.); (P.V.); (C.M.M.); (G.C.); (G.d.)
| |
Collapse
|
36
|
Kibbie JJ, Dillon SM, Thompson TA, Purba CM, McCarter MD, Wilson CC. Butyrate directly decreases human gut lamina propria CD4 T cell function through histone deacetylase (HDAC) inhibition and GPR43 signaling. Immunobiology 2021; 226:152126. [PMID: 34365090 DOI: 10.1016/j.imbio.2021.152126] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 07/06/2021] [Accepted: 07/25/2021] [Indexed: 12/14/2022]
Abstract
An important function of the gut microbiome is the fermentation of non-digestible dietary fibers into short chain fatty acids (SCFAs). The three primary SCFAs: acetate, propionate, and butyrate, are key mediators of metabolism and immune cell function in the gut mucosa. We previously demonstrated that butyrate at high concentrations decreased human gut lamina propria (LP) CD4 T cell activation in response to enteric bacteria exposure in vitro. However, to date, the mechanism by which butyrate alters human gut LP CD4 T cell activation remains unknown. In this current study, we sought to better understand how exposure to SCFAs across a concentration range impacted human gut LP CD4 T cell function and activation. LP CD4 T cells were directly activated with T cell receptor (TCR) beads in vitro in the presence of a physiologic concentration range of each of the primary SCFAs. Exposure to butyrate potently inhibited CD4 T cell activation, proliferation, and cytokine (IFNγ, IL-17) production in a concentration dependent manner. Butyrate decreased the proliferation and cytokine production of T helper (Th) 1, Th17 and Th22 cells, with differences noted in the sensitivity of LP versus peripheral blood Th cells to butyrate's effects. Higher concentrations of propionate and acetate relative to butyrate were required to inhibit CD4 T cell activation and proliferation. Butyrate directly increased the acetylation of both unstimulated and TCR-stimulated CD4 T cells, and apicidin, a Class I histone deacetylase inhibitor, phenocopied butyrate's effects on CD4 T cell proliferation and activation. GPR43 agonism phenocopied butyrate's effect on CD4 T cell proliferation whereas a GPR109a agonist did not. Our findings indicate that butyrate decreases in vitro human gut LP CD4 T cell activation, proliferation, and inflammatory cytokine production more potently than other SCFAs, likely through butyrate's ability to increase histone acetylation, and potentially via signaling through GPR43. These findings have relevance in furthering our understanding of how perturbations of the gut microbiome alter local immune responses in the gut mucosa.
Collapse
Affiliation(s)
- Jon J Kibbie
- Department of Medicine, Division of Infectious Disease, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA; Department of Immunology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Stephanie M Dillon
- Department of Medicine, Division of Infectious Disease, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Tezha A Thompson
- Department of Medicine, Division of Infectious Disease, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Christine M Purba
- Department of Medicine, Division of Infectious Disease, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Martin D McCarter
- Department of Surgery, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Cara C Wilson
- Department of Medicine, Division of Infectious Disease, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
37
|
Chen Z, Lv Y, Xu H, Deng L. Herbal Medicine, Gut Microbiota, and COVID-19. Front Pharmacol 2021; 12:646560. [PMID: 34305582 PMCID: PMC8293616 DOI: 10.3389/fphar.2021.646560] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 06/25/2021] [Indexed: 01/08/2023] Open
Abstract
Coronavirus Disease 19 (COVID-19) is a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has grown to a worldwide pandemic with substantial mortality. The symptoms of COVID-19 range from mild flu-like symptoms, including cough and fever, to life threatening complications. There are still quite a number of patients with COVID-19 showed enteric symptoms including nausea, vomiting, and diarrhea. The gastrointestinal tract may be one of the target organs of SARS-CoV-2. Angiotensin converting enzyme 2 (ACE2) is the main receptor of SARS-CoV-2 virus, which is significantly expressed in intestinal cells. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Intestinal flora imbalance and endotoxemia may accelerate the progression of COVID-19. Many herbs have demonstrated properties relevant to the treatment of COVID-19, by supporting organs and systems of the body affected by the virus. Herbs can restore the structure of the intestinal flora, which may further modulate the immune function after SARS-CoV-2 infection. Regulation of intestinal flora by herbal medicine may be helpful for the treatment and recovery of the disease. Understanding the role of herbs that regulate intestinal flora in fighting respiratory virus infections and maintaining intestinal flora balance can provide new ideas for preventing and treating COVID-19.
Collapse
Affiliation(s)
- Ziqi Chen
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
- Medical College, Sun Yat-sen University, Guangzhou, China
| | - Yiwen Lv
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Huachong Xu
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Li Deng
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
38
|
Sharma VK, Singh TG, Garg N, Dhiman S, Gupta S, Rahman MH, Najda A, Walasek-Janusz M, Kamel M, Albadrani GM, Akhtar MF, Saleem A, Altyar AE, Abdel-Daim MM. Dysbiosis and Alzheimer's Disease: A Role for Chronic Stress? Biomolecules 2021; 11:biom11050678. [PMID: 33946488 PMCID: PMC8147174 DOI: 10.3390/biom11050678] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is an incurable, neuropsychiatric, pathological condition that deteriorates the worth of geriatric lives. AD is characterized by aggregated senile amyloid plaques, neurofibrillary tangles, neuronal loss, gliosis, oxidative stress, neurotransmitter dysfunction, and bioenergetic deficits. The changes in GIT composition and harmony have been recognized as a decisive and interesting player in neuronal pathologies including AD. Microbiota control and influence the oxidoreductase status, inflammation, immune system, and the endocrine system through which it may have an impact on the cognitive domain. The altered and malfunctioned state of microbiota is associated with minor infections to complicated illnesses that include psychosis and neurodegeneration, and several studies show that microbiota regulates neuronal plasticity and neuronal development. The altered state of microbiota (dysbiosis) may affect behavior, stress response, and cognitive functions. Chronic stress-mediated pathological progression also has a well-defined role that intermingles at various physiological levels and directly impacts the pathological advancement of AD. Chronic stress-modulated alterations affect the well-established pathological markers of AD but also affect the gut–brain axis through the mediation of various downstream signaling mechanisms that modulate the microbial commensals of GIT. The extensive literature reports that chronic stressors affect the composition, metabolic activities, and physiological role of microbiota in various capacities. The present manuscript aims to elucidate mechanistic pathways through which stress induces dysbiosis, which in turn escalates the neuropathological cascade of AD. The stress–dysbiosis axis appears a feasible zone of work in the direction of treatment of AD.
Collapse
Affiliation(s)
- Vivek Kumar Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (V.K.S.); (N.G.); (S.D.); (S.G.)
- Goverment College of Pharmacy, District Shimla, Rohru 171207, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (V.K.S.); (N.G.); (S.D.); (S.G.)
- Correspondence: or (T.G.S.); (M.M.A.-D.); Tel.: +91-98-1595-1171 (T.G.S.); +20-96-65-8019-2142 (M.M.A.-D.)
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (V.K.S.); (N.G.); (S.D.); (S.G.)
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (V.K.S.); (N.G.); (S.D.); (S.G.)
| | - Saurabh Gupta
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (V.K.S.); (N.G.); (S.D.); (S.G.)
| | - Md. Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh;
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland; (A.N.); (M.W.-J.)
| | - Magdalena Walasek-Janusz
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland; (A.N.); (M.W.-J.)
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia;
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore 54950, Pakistan;
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Ahmed E. Altyar
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia;
| | - Mohamed M. Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
- Correspondence: or (T.G.S.); (M.M.A.-D.); Tel.: +91-98-1595-1171 (T.G.S.); +20-96-65-8019-2142 (M.M.A.-D.)
| |
Collapse
|
39
|
Käser T. Swine as biomedical animal model for T-cell research-Success and potential for transmittable and non-transmittable human diseases. Mol Immunol 2021; 135:95-115. [PMID: 33873098 DOI: 10.1016/j.molimm.2021.04.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/23/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Swine is biologically one of the most relevant large animal models for biomedical research. With its use as food animal that can be exploited as a free cell and tissue source for research and its high susceptibility to human diseases, swine additionally represent an excellent option for both the 3R principle and One Health research. One of the previously most limiting factors of the pig model was its arguably limited immunological toolbox. Yet, in the last decade, this toolbox has vastly improved including the ability to study porcine T-cells. This review summarizes the swine model for biomedical research with focus on T cells. It first contrasts the swine model to the more commonly used mouse and non-human primate model before describing the current capabilities to characterize and extend our knowledge on porcine T cells. Thereafter, it not only reflects on previous biomedical T-cell research but also extends into areas in which more in-depth T-cell analyses could strongly benefit biomedical research. While the former should inform on the successes of biomedical T-cell research in swine, the latter shall inspire swine T-cell researchers to find collaborations with researchers working in other areas - such as nutrition, allergy, cancer, transplantation, infectious diseases, or vaccine development.
Collapse
Affiliation(s)
- Tobias Käser
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, 27607 Raleigh, NC, USA.
| |
Collapse
|
40
|
Cevaal PM, Ali A, Czuba-Wojnilowicz E, Symons J, Lewin SR, Cortez-Jugo C, Caruso F. In Vivo T Cell-Targeting Nanoparticle Drug Delivery Systems: Considerations for Rational Design. ACS NANO 2021; 15:3736-3753. [PMID: 33600163 DOI: 10.1021/acsnano.0c09514] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
T cells play an important role in immunity and repair and are implicated in diseases, including blood cancers, viral infections, and inflammation, making them attractive targets for the treatment and prevention of diseases. Over recent years, the advent of nanomedicine has shown an increase in studies that use nanoparticles as carriers to deliver therapeutic cargo to T cells for ex vivo and in vivo applications. Nanoparticle-based delivery has several advantages, including the ability to load and protect a variety of drugs, control drug release, improve drug pharmacokinetics and biodistribution, and site- or cell-specific targeting. However, the delivery of nanoparticles to T cells remains a major technological challenge, which is primarily due to the nonphagocytic nature of T cells. In this review, we discuss the physiological barriers to effective T cell targeting and describe the different approaches used to deliver cargo-loaded nanoparticles to T cells for the treatment of disease such as T cell lymphoma and human immunodeficiency virus (HIV). In particular, engineering strategies that aim to improve nanoparticle internalization by T cells, including ligand-based targeting, will be highlighted. These nanoparticle engineering approaches are expected to inspire the development of effective nanomaterials that can target or manipulate the function of T cells for the treatment of T cell-related diseases.
Collapse
Affiliation(s)
| | | | - Ewa Czuba-Wojnilowicz
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | | | - Sharon R Lewin
- Victorian Infectious Diseases, Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria 3000, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Victoria 3004, Australia
| | - Christina Cortez-Jugo
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
41
|
Gu BH, Kim M, Yun CH. Regulation of Gastrointestinal Immunity by Metabolites. Nutrients 2021; 13:nu13010167. [PMID: 33430497 PMCID: PMC7826526 DOI: 10.3390/nu13010167] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/01/2021] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal tract contains multiple types of immune cells that maintain the balance between tolerance and activation at the first line of host defense facing non-self antigens, including dietary antigens, commensal bacteria, and sometimes unexpected pathogens. The maintenance of homeostasis at the gastrointestinal tract requires stringent regulation of immune responses against various environmental conditions. Dietary components can be converted into gut metabolites with unique functional activities through host as well as microbial enzymatic activities. Accumulating evidence demonstrates that gastrointestinal metabolites have significant impacts on the regulation of intestinal immunity and are further integrated into the immune response of distal mucosal tissue. Metabolites, especially those derived from the microbiota, regulate immune cell functions in various ways, including the recognition and activation of cell surface receptors, the control of gene expression by epigenetic regulation, and the integration of cellular metabolism. These mucosal immune regulations are key to understanding the mechanisms underlying the development of gastrointestinal disorders. Here, we review recent advancements in our understanding of the role of gut metabolites in the regulation of gastrointestinal immunity, highlighting the cellular and molecular regulatory mechanisms by macronutrient-derived metabolites.
Collapse
Affiliation(s)
- Bon-Hee Gu
- Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Korea;
| | - Myunghoo Kim
- Life and Industry Convergence Research Institute, Pusan National University, Miryang 50463, Korea;
- Department of Animal Science, College of Natural Resources & Life Science, Pusan National University, Miryang 50463, Korea
- Correspondence: (M.K.); (C.-H.Y.)
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
- Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Korea
- Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang-gun, Gangwon-do 25354, Korea
- Correspondence: (M.K.); (C.-H.Y.)
| |
Collapse
|
42
|
Riva A, Gray EH, Azarian S, Zamalloa A, McPhail MJ, Vincent RP, Williams R, Chokshi S, Patel VC, Edwards LA. Faecal cytokine profiling as a marker of intestinal inflammation in acutely decompensated cirrhosis. JHEP Rep 2020; 2:100151. [PMID: 32838247 PMCID: PMC7391986 DOI: 10.1016/j.jhepr.2020.100151] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/26/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND & AIMS Gut dysbiosis and inflammation perpetuate loss of gut barrier integrity (GBI) and pathological bacterial translocation (BT) in cirrhosis, contributing to infection risk. Little is known about gut inflammation in cirrhosis and how this differs in acute decompensation (AD). We developed a novel approach to characterise intestinal immunopathology by quantifying faecal cytokines (FCs) and GBI markers. METHODS Faeces and plasma were obtained from patients with stable cirrhosis (SC; n = 16), AD (n = 47), and healthy controls (HCs; n = 31). A panel of 15 cytokines and GBI markers, including intestinal fatty-acid-binding protein-2 (FABP2), d-lactate, and faecal calprotectin (FCAL), were quantified by electrochemiluminescence/ELISA. Correlations between analytes and clinical metadata with univariate and multivariate analyses were performed. RESULTS Faecal (F) IL-1β, interferon gamma, tumour necrosis factor alpha, IL-21, IL-17A/F, and IL-22 were significantly elevated in AD vs. SC (q <0.01). F-IL-23 was significantly elevated in AD vs. HC (p = 0.0007). FABP2/d-lactate were significantly increased in faeces in AD vs. SC and AD vs. HC (p <0.0001) and in plasma (p = 0.0004; p = 0.011). F-FABP2 correlated most strongly with disease severity (Spearman's rho: Child-Pugh 0.466; p <0.0001; model for end-stage liver disease 0.488; p <0.0001). FCAL correlated with plasma IL-21, IL-1β, and IL-17F only and none of the faecal analytes. F-cytokines and F-GBI markers were more accurate than plasma in discriminating AD from SC. CONCLUSIONS FC profiling represents an innovative approach to investigating the localised intestinal cytokine micro-environment in cirrhosis. These data reveal that AD is associated with a highly inflamed and permeable gut barrier. FC profiles are very different from the classical innate-like features of systemic inflammation. There is non-specific upregulation of TH1/TH17 effector cytokines and those known to mediate intestinal barrier damage. This prevents mucosal healing in AD and further propagates BT and systemic inflammation. LAY SUMMARY The gut barrier is crucial in cirrhosis in preventing infection-causing bacteria that normally live in the gut from accessing the liver and other organs via the bloodstream. Herein, we characterised gut inflammation by measuring different markers in stool samples from patients at different stages of cirrhosis and comparing this to healthy people. These markers, when compared with equivalent markers usually measured in blood, were found to be very different in pattern and absolute levels, suggesting that there is significant gut inflammation in cirrhosis related to different immune system pathways to that seen outside of the gut. This provides new insights into gut-specific immune disturbances that predispose to complications of cirrhosis, and emphasises that a better understanding of the gut-liver axis is necessary to develop better targeted therapies.
Collapse
Key Words
- ACLF, acute-on-chronic liver failure
- AD, acute decompensation
- AUROC, area under the receiver operating characteristic
- BT, bacterial translocation
- Bacterial translocation
- CLIF-C AD, Chronic Liver Failure Consortium-acute decompensation
- Chronic liver disease
- Cytokines
- DS, discriminant score
- FABP2, fatty-acid-binding protein-2
- FCAL, faecal calprotectin
- FDR, false discovery rate
- FL, faecal lysate
- FWER, family-wise error rate
- GVB, gut vascular barrier
- Gut inflammation
- HC, healthy control
- IBD, inflammatory bowel disease
- IEC, intestinal epithelial cell
- Intestinal barrier function
- MELD, model for end-stage liver disease
- OPLS-DA, orthogonal projection to latent structures discriminant analysis
- PAMP, pathogen-associated molecular pattern
- PCA, principal component analysis
- ROC, receiver operating characteristic
- SC, stable cirrhosis
- UKELD, United Kingdom model for end-stage liver disease
Collapse
Affiliation(s)
- Antonio Riva
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Elizabeth H. Gray
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Sarah Azarian
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Ane Zamalloa
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London, UK
| | - Mark J.W. McPhail
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London, UK
| | - Royce P. Vincent
- Department of Clinical Biochemistry, King's College Hospital NHS Foundation Trust, London, UK
- Department of Nutritional Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Roger Williams
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Shilpa Chokshi
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Vishal C. Patel
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London, UK
| | - Lindsey A. Edwards
- Institute of Hepatology London, Foundation for Liver Research, London, UK
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
43
|
Bhattacharya P, Ellegård R, Khalid M, Svanberg C, Govender M, Keita ÅV, Söderholm JD, Myrelid P, Shankar EM, Nyström S, Larsson M. Complement opsonization of HIV affects primary infection of human colorectal mucosa and subsequent activation of T cells. eLife 2020; 9:e57869. [PMID: 32876566 PMCID: PMC7492089 DOI: 10.7554/elife.57869] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
HIV transmission via genital and colorectal mucosa are the most common routes of dissemination. Here, we explored the effects of free and complement-opsonized HIV on colorectal tissue. Initially, there was higher antiviral responses in the free HIV compared to complement-opsonized virus. The mucosal transcriptional response at 24 hr revealed the involvement of activated T cells, which was mirrored in cellular responses observed at 96 hr in isolated mucosal T cells. Further, HIV exposure led to skewing of T cell phenotypes predominantly to inflammatory CD4+ T cells, that is Th17 and Th1Th17 subsets. Of note, HIV exposure created an environment that altered the CD8+ T cell phenotype, for example expression of regulatory factors, especially when the virions were opsonized with complement factors. Our findings suggest that HIV-opsonization alters the activation and signaling pathways in the colorectal mucosa, which promotes viral establishment by creating an environment that stimulates mucosal T cell activation and inflammatory Th cells.
Collapse
Affiliation(s)
- Pradyot Bhattacharya
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Rada Ellegård
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Mohammad Khalid
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Cecilia Svanberg
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Melissa Govender
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Åsa V Keita
- Division of Surgery, Orthopedics and Oncology, Linköping UniversityLinköpingSweden
| | - Johan D Söderholm
- Division of Surgery, Orthopedics and Oncology, Linköping UniversityLinköpingSweden
| | - Pär Myrelid
- Division of Surgery, Orthopedics and Oncology, Linköping UniversityLinköpingSweden
| | - Esaki M Shankar
- Center of Excellence for Research in AIDS (CERiA), University of Malaya, Lembah PantaiKuala LumpurMalaysia
- Division of Infection Biology and Medical Microbiology, Department of Life Sciences, Central University of Tamil NaduThiruvarurIndia
| | - Sofia Nyström
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
- Department of Clinical Immunology and Transfusion Medicine and Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| | - Marie Larsson
- Division of Molecular Medicine and Virology, Department of Clinical and Experimental Medicine, Linköping UniversityLinköpingSweden
| |
Collapse
|
44
|
Wu N, Chen D, Sun H, Tan J, Zhang Y, Zhang T, Han Y, Liu H, Ouyang X, Yang XD, Niu X, Zhong J, Wang Z, Su B. MAP3K2 augments Th1 cell differentiation via IL-18 to promote T cell-mediated colitis. SCIENCE CHINA-LIFE SCIENCES 2020; 64:389-403. [PMID: 32737854 DOI: 10.1007/s11427-020-1720-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022]
Abstract
T cell-mediated immunity in the intestine is stringently controlled to ensure proper immunity against pathogenic microbes and to prevent autoimmunity, a known cause of inflammatory bowel disease. However, precisely how T cells regulate intestine immunity remains to be fully understood. In this study, we found that mitogen-activated protein kinase kinase kinase 2 (MAP3K2) is required for the CD4+ T cell-mediated inflammation in the intestine. Using a T cell transfer colitis model, we found that MAP3K2-deficient naïve CD4 T cells had a dramatically reduced ability to induce colitis compared to wild type T cells. In addition, significantly fewer IFN-γ- but more IL-17A-producing CD4+ T cells in the intestines of mice receiving MAP3K2-deficient T cells than in those from mice receiving wild type T cells was observed. Interestingly, under well-defined in vitro differentiation conditions, MAP3K2-deficient naïve T cells were not impaired in their ability to differentiate into Th1, Th17 and Treg. Furthermore, the MAP3K2-regulated colitis severity was mediated by Th1 but not Th17 cells in the intestine. At the molecular level, we showed that MAP3K2-mediated Th1 cell differentiation in the intestine was regulated by IL-18 and required specific JNK activation. Together, our study reveals a novel regulatory role of MAP3K2 in intestinal T cell immunity via the IL-18-MAP3K2-JNK axis and may provide a novel target for intervention in T cell-mediated colitis.
Collapse
Affiliation(s)
- Ningbo Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Dongping Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Hongxiang Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Jianmei Tan
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Yao Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Tianyu Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Yuheng Han
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Hongzhi Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Xinxing Ouyang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Xiao-Dong Yang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyin Niu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Jie Zhong
- Department of Gastroenterology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China
| | - Zhengting Wang
- Department of Gastroenterology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China.
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China.
- Shanghai JiaoTong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai JiaoTong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
45
|
Cordes F, Foell D, Ding JN, Varga G, Bettenworth D. Differential regulation of JAK/STAT-signaling in patients with ulcerative colitis and Crohn's disease. World J Gastroenterol 2020; 26:4055-4075. [PMID: 32821070 PMCID: PMC7403801 DOI: 10.3748/wjg.v26.i28.4055] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/24/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
In 2018, the pan-Janus kinase (JAK) inhibitor tofacitinib was launched for the treatment of ulcerative colitis (UC). Although tofacitinib has proven efficacious in patients with active UC, it failed in patients with Crohn's disease (CD). This finding strongly hints at a different contribution of JAK signaling in both entities. Here, we review the current knowledge on the interplay between the JAK/signal transducer and activator of transcription (STAT) pathway and inflammatory bowel diseases (IBD). In particular, we provide a detailed overview of the differences and similarities of JAK/STAT-signaling in UC and CD, highlight the impact of the JAK/STAT pathway in experimental colitis models and summarize the published evidence on JAK/STAT-signaling in immune cells of IBD as well as the genetic association between the JAK/STAT pathway and IBD. Finally, we describe novel treatment strategies targeting JAK/STAT inhibition in UC and CD and comment on the limitations and challenges of the new drug class.
Collapse
Affiliation(s)
- Friederike Cordes
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster D-48149, Germany
| | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children’s Hospital Münster, Münster D-48149, Germany
| | - John Nik Ding
- Department of Gastroenterology, St. Vincent’s Hospital, Melbourne 3002, Australia
- Department of Medicine, University of Melbourne, East Melbourne 3002, Australia
| | - Georg Varga
- Department of Pediatric Rheumatology and Immunology, University Children’s Hospital Münster, Münster D-48149, Germany
| | - Dominik Bettenworth
- Department of Medicine B, Gastroenterology and Hepatology, University Hospital Münster, Münster D-48149, Germany
| |
Collapse
|
46
|
Le N, Mazahery C, Nguyen K, Levine AD. Regulation of Intestinal Epithelial Barrier and Immune Function by Activated T Cells. Cell Mol Gastroenterol Hepatol 2020; 11:55-76. [PMID: 32659380 PMCID: PMC7596298 DOI: 10.1016/j.jcmgh.2020.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Communication between T cells and the intestinal epithelium is altered in many diseases, causing T-cell activation, depletion, or recruitment, and disruption of the epithelium. We hypothesize that activation of T cells regulates epithelial barrier function by targeting the assembly of the tight junction complex. METHODS In a 3-dimensional and 2-dimensional co-culture model of activated T cells subjacent to the basolateral surface of an epithelial monolayer, the pore, leak, and unrestricted pathways were evaluated using transepithelial resistance and flux of fluorescently labeled tracers. T cells were acutely and chronically activated by cross-linking the T-cell receptor. Tight junction assembly and expression were measured using quantitative polymerase chain reaction, immunoblot, and immunofluorescence confocal microscopy. RESULTS Co-culture with acutely and chronically activated T cells decreased the magnitude of ion flux through the pore pathway, which was maintained in the presence of acutely activated T cells. Chronically activated T cells after 30 hours induced a precipitous increase in the magnitude of both ion and molecular flux, resulting in an increase in the unrestricted pathway, destruction of microvilli, expansion in cell surface area, and cell death. These fluctuations in permeability were the result of changes in the assembly and expression of tight junction proteins, cell morphology, and viability. Co-culture modulated the expression of immune mediators in the epithelium and T cells. CONCLUSIONS Bidirectional communication between T cells and epithelium mediates a biphasic response in barrier integrity that is facilitated by the balance between structural proteins partitioning in the mobile lateral phase vs the tight junction complex and cell morphology.
Collapse
Affiliation(s)
- Nga Le
- Department of Molecular Biology and Microbiology
| | | | - Kien Nguyen
- Department of Molecular Biology and Microbiology
| | - Alan D Levine
- Department of Molecular Biology and Microbiology; Department of Pathology; Department of Pharmacology; Department of Medicine; Department of Pediatrics; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
47
|
Zhang E, Wang J, Li Y, Huang L, Wang Y, Yang Q. Comparison of oral and nasal immunization with inactivated porcine epidemic diarrhea virus on intestinal immunity in piglets. Exp Ther Med 2020; 20:1596-1606. [PMID: 32742391 PMCID: PMC7388329 DOI: 10.3892/etm.2020.8828] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 02/20/2020] [Indexed: 12/11/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) has proven to be a major problem for the porcine industry worldwide. Conventional injectable vaccines induce effective systemic immune responses but are less effective in preventing PEDV at mucosal invasion sites, including the nasal or oral mucosa. Additionally, antigens delivered orally are easily degraded. Nasal immunization induces intestinal mucosal immune responses, which can aid in blocking viral invasion, and requires fewer antigen inoculation doses. Therefore, nasal immunizations are considered to be a potential approach to overcome viral infections. In the present study, nasal immunization of piglets was performed using inactivated PEDV combined with Bacillus subtilis as an immunopotentiator and the efficacy of nasal immunization was assessed. The results demonstrated that compared with oral immunization, piglets from the nasal immunization group exhibited higher levels of neutralizing antibodies (P<0.01) in the intestine, PEDV-specific immunoglobulin (Ig)G (P<0.01) in serum and PEDV-specific secretory IgA (SIgA) in saliva (P<0.01) and nasal secretions (P<0.01). An increased number of intestinal CD3+ T cells, IgA-secreting cells and intraepithelial lymphocytes (P<0.05) were also observed. Furthermore, the protein expression levels of interleukin-6 and interferon-γ, relative to the control PEDV infection, were also significantly elevated (P<0.05). The results of the present study indicate that nasal immunization is more effective at inducing the intestinal mucosal immune response, and provide new insights into a novel vaccination strategy that may be used to decrease the incidence of PEDV infections.
Collapse
Affiliation(s)
- En Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Jialu Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Yuchen Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Lulu Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Yongheng Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| |
Collapse
|
48
|
Ion Transport Basis of Diarrhea in a Mouse Model of Adoptive T Cell Transfer Colitis. Dig Dis Sci 2020; 65:1700-1709. [PMID: 31741140 PMCID: PMC7230007 DOI: 10.1007/s10620-019-05945-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Diarrhea, a major pathological hallmark of inflammatory bowel disease, is characterized by a significant reduction in the expression and function of key intestinal ion transporters. The adoptive naïve CD4+ T cell transfer colitis is an immune-based, chronic colitis mouse model which resembles human Crohn's disease. Although mice with T cell transfer colitis demonstrate diarrhea, the ion transporter basis of this phenotype has not been explored. AIMS/METHODS In the current studies, we aimed to determine the mRNA and protein levels of the key NaCl transporters DRA and NHE3 along with the mRNA expression of other transporters in the inflamed intestine. RESULTS Naïve CD4+ T cells, transferred to Rag2 knockout mice, induced severe colonic inflammation characterized by histological damage and increased mRNA levels of cytokines in the colon with no effect in the ileum. Diarrheal phenotype was a key feature of the excised colons of mice where loose stools were evident. Our results demonstrated that the key chloride transporter DRA, mRNA, and protein levels were significantly reduced in the inflamed colon. However, expression of the key sodium hydrogen exchanger NHE3 was unaffected. The mRNA expression of other important transporters was also determined; in this regard, the sodium channel ENACα and the monocarboxylate transporters MCT1 and SMCT1 mRNA levels were also significantly lower compared to control mice. However, CFTR mRNA was not altered in the colon or ileum. CONCLUSIONS The studies conducted herein for the first time demonstrate the downregulation of important intestinal ion transporters in proximal and distal colon in T cell transfer colitis mouse model, providing valuable evidence for the ion transporter basis of diarrhea in this chronic model of inflammation.
Collapse
|
49
|
The role of host molecules in communication with the resident and pathogenic microbiota: A review. MEDICINE IN MICROECOLOGY 2020. [DOI: 10.1016/j.medmic.2020.100005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
50
|
Effect of Probiotics and Herbal Products on Intestinal Histomorphological and Immunological Development in Piglets. Vet Med Int 2020; 2020:3461768. [PMID: 32373310 PMCID: PMC7196157 DOI: 10.1155/2020/3461768] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/25/2020] [Indexed: 01/21/2023] Open
Abstract
The aim of the study was to evaluate the effect of probiotics and herbal products on the intestinal histomorphological and immunological development in piglets. Accordingly, 2-week-old piglets were allocated in 4 groups: C (basal diet), Pro (basal diet + probiotics), Pro+B (basal diet + probiotics + buckwheat bran), and H (powder of herbs). After 6 weeks of the experiment, 4 piglets from each experimental group were randomly selected and slaughtered at a slaughterhouse. Samples of tissue and digestive content from the jejunum and colon were collected for bacteriological, histological, and immunohistochemical examination. The results showed that probiotics increased the number of Lactobacillus spp. in the small (p < 0.05) and large intestines. The intestinal histomorphology was improved (p < 0.05) in all experimental groups by an increased villus height, VH : CD ration, colon crypt depth, and number of Ki-67+ epithelial cells. A higher number (p < 0.05) of goblet cells and their acidification were observed in group Pro, while the density of goblet cells was decreased by the herbs. Probiotics increased (p < 0.05) the number of intraepithelial lymphocytes (IELs), density of CD3+ cells in Peyer's patches (PPs), and lamina propria (LP). In group H, a dual effect on the CD3+ cell distribution was observed. The herbs reduced (p < 0.05) the number of IELs and CD3+ in LP but increased the distribution of CD3+ cells in PPs. In the colon, herbs increased CD3+ cells in LP as well. It suggests that probiotics and herbs had influence on the intestinal histomorphology and the ability to modulate the mucosal immune system; however, the combination of probiotics and buckwheat bran was not so convincing, probably due to the inhibitory effect of the buckwheat bran on the probiotics used.
Collapse
|