1
|
Yan YC, Su L, Zhao WB, Fan Y, Koprich JB, Xiao BG, Song B, Wang J, Yu WB. Bidirectional interaction between IL and 17A/IL-17RA pathway dysregulation and α-synuclein in the pathogenesis of Parkinson's disease. Brain Behav Immun 2024:S0889-1591(24)00666-4. [PMID: 39461385 DOI: 10.1016/j.bbi.2024.10.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 10/08/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024] Open
Abstract
Parkinson's disease (PD) pathogenesis is characterized by α-synuclein (α-syn) pathology, which is influenced by various factors such as neuroinflammation and senescence. Increasing evidence has suggested a pivotal role for Interleukin-17A(IL-17A) and Interleukin-17 Receptor A (IL-17RA) in PD, yet the trigger and impact of IL-17A/IL-17RA activation in PD remains elusive. This study observed an age-related increase in IL-17A and IL-17RA in the human central nervous system, accompanied by increased α-syn and senescence biomarkers. Interestingly, both levels of IL-17A and IL-17RA in PD patients were significantly elevated compared to age-matched controls, wherein the IL-17A was mainly present in neurons. This abnormal neuronal IL-17A activation in the PD brain was recapitulated in α-syn mouse models. Correspondingly, administration of recombinant IL-17A exacerbated pathological α-syn in both neuron and mouse models. Furthermore, IL-17A/IL-17RA pathway interventions via blocking antibody or shRNA-mediated knockdown can mitigate the effects of pathological α-syn. This study reveals an interplay between dysregulation of the IL-17A/IL-17RA pathway and α-syn, suggesting that regulating the IL-17A/IL-17RA pathway could modify PD progression by disrupting the detrimental cycle.
Collapse
Affiliation(s)
- Yu-Chen Yan
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lu Su
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wan-Bing Zhao
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yun Fan
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - James B Koprich
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200040, China
| | - Bin Song
- Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Shanghai 200032, China; Fudan University, Shanghai 200032, China; Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jian Wang
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Wen-Bo Yu
- Department of Neurology and National Research Center for Aging and Medicine & National Center for Neurological Disorders, State Key Laboratory of Medical Neurobiology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
2
|
Guo S, Peng J, Xiao Y, Chen J, Gao R. Synergistic effects of oral inoculation with a recombinant Lactobacillus plantarum NC8 strain co-expressing interleukin-2 and interleukin-17B on the efficacy of the infectious bronchitis vaccine in chickens. Poult Sci 2024; 103:103908. [PMID: 38981363 PMCID: PMC11279255 DOI: 10.1016/j.psj.2024.103908] [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: 04/05/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 07/11/2024] Open
Abstract
Mucosal vaccination strategies are easier to implement than others in large-scale poultry farming. However, the adjuvants that are approved for veterinary use, which are predominantly aluminum- and oil-emulsion-based adjuvants, are not suitable for mucosal vaccination and carry a risk of adverse reactions. In this study, we engineered a novel Lactobacillus plantarum NC8 strain that co-expresses chicken interleukin-2 (IL-2) and IL-17B, which we designated NC8-ChIL2-17B, and evaluated its potential as an oral immunoadjuvant. The immunomodulatory properties of NC8-ChIL2-17B were evidenced by its ability to activate macrophages and inhibit the proliferation of infectious bronchitis virus (IBV) in vitro. We then confirmed its immunoadjuvant activity in vivo by orally administering NC8-ChIL2-17B along with a commercial IBV vaccine to chicks. The results indicated that NC8-ChIL2-17B enhanced the immune response elicited by the IBV vaccine and increased the levels of IBV-specific IgG and sIgA antibodies produced in response to IBV infection. Additionally, administration of NC8-ChIL2-17B promoted weight gain and beneficially modulated the gut microbiota, resulting in improved chicken performance. These findings suggest that oral administration of NC8-ChIL2-17B is a promising strategy to enhance the immune efficacy of the IBV vaccine in chickens, offering an efficacious alternative adjuvant.
Collapse
Affiliation(s)
- Shaohua Guo
- Laboratory of Infectious Diseases and Vaccine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Junjie Peng
- Key Laboratory for Bio-resource and Eco-Environment of Education Ministry, College of Life Sciences, Sichuan University, Chengdu, 610065, PR China; National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
| | - Yongle Xiao
- School of Medicine, Sichuan University of Arts and Science, Dazhou, 635000, PR China
| | - Jianlin Chen
- School of Laboratory Medicine/Collaborative Innovation Center of Sichuan for Elderly Care and Health, Chengdu Medical College, Chengdu, Sichuan 610500, PR China
| | - Rong Gao
- Key Laboratory for Bio-resource and Eco-Environment of Education Ministry, College of Life Sciences, Sichuan University, Chengdu, 610065, PR China.
| |
Collapse
|
3
|
Gonzalez E, Lee MD, Tierney BT, Lipieta N, Flores P, Mishra M, Beckett L, Finkelstein A, Mo A, Walton P, Karouia F, Barker R, Jansen RJ, Green SJ, Weging S, Kelliher J, Singh NK, Bezdan D, Galazska J, Brereton NJB. Spaceflight alters host-gut microbiota interactions. NPJ Biofilms Microbiomes 2024; 10:71. [PMID: 39209868 PMCID: PMC11362537 DOI: 10.1038/s41522-024-00545-1] [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: 02/07/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
The ISS rodent habitat has provided crucial insights into the impact of spaceflight on mammals, inducing symptoms characteristic of liver disease, insulin resistance, osteopenia, and myopathy. Although these physiological responses can involve the microbiome on Earth, host-microbiota interactions during spaceflight are still being elucidated. We explore murine gut microbiota and host gene expression in the colon and liver after 29 and 56 days of spaceflight using multiomics. Metagenomics revealed significant changes in 44 microbiome species, including relative reductions in bile acid and butyrate metabolising bacteria like Extibacter muris and Dysosmobacter welbionis. Functional prediction indicate over-representation of fatty acid and bile acid metabolism, extracellular matrix interactions, and antibiotic resistance genes. Host gene expression described corresponding changes to bile acid and energy metabolism, and immune suppression. These changes imply that interactions at the host-gut microbiome interface contribute to spaceflight pathology and that these interactions might critically influence human health and long-duration spaceflight feasibility.
Collapse
Affiliation(s)
- E Gonzalez
- Microbiome Unit, Canadian Centre for Computational Genomics, Department of Human Genetics, McGill University, Montréal, Canada
- Centre for Microbiome Research, McGill University, Montréal, Canada
| | - M D Lee
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
| | - B T Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - N Lipieta
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - P Flores
- BioServe Space Technologies, University of Colorado Boulder, Boulder, CO, USA
| | - M Mishra
- Grossman School of Medicine, New York University, New York, USA
| | - L Beckett
- University of Nottingham, Nottingham, NG7 2RD, UK
| | - A Finkelstein
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - A Mo
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - P Walton
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - F Karouia
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Centre for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - R Barker
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Yuri GmbH, Wiesentalstr. 40, 88074, Meckenbeuren, Germany
- University of Wisconsin-Madison, Madison, WI, USA
| | - R J Jansen
- Department of Public Health, North Dakota State University, Fargo, ND, USA
- Genomics, Phenomics, and Bioinformatics Program, North Dakota State University, Fargo, ND, USA
| | - S J Green
- Genomics and Microbiome Core Facility, Rush University Medical Centre, 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - S Weging
- Institute of Computer Science, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - J Kelliher
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - N K Singh
- Department of Industrial Relations, Division of Occupational Safety and Health, Oakland, USA
| | - D Bezdan
- University of Wisconsin-Madison, Madison, WI, USA
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- NGS Competence Centre Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| | - J Galazska
- Space Biosciences Research Branch, NASA Ames Research Centre, Moffett Field, CA, USA
| | - N J B Brereton
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
4
|
Lu T, Raju M, Howlader DR, Dietz ZK, Whittier SK, Varisco DJ, Ernst RK, Coghill LM, Picking WD, Picking WL. Vaccination with a Protective Ipa Protein-Containing Nanoemulsion Differentially Alters the Transcriptomic Profiles of Young and Elderly Mice following Shigella Infection. Vaccines (Basel) 2024; 12:618. [PMID: 38932347 PMCID: PMC11209624 DOI: 10.3390/vaccines12060618] [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: 05/04/2024] [Revised: 05/28/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Shigella spp. are responsible for bacillary dysentery or shigellosis transmitted via the fecal-oral route, causing significant morbidity and mortality, especially among vulnerable populations. There are currently no licensed Shigella vaccines. Shigella spp. use a type III secretion system (T3SS) to invade host cells. We have shown that L-DBF, a recombinant fusion of the T3SS needle tip (IpaD) and translocator (IpaB) proteins with the LTA1 subunit of enterotoxigenic E. coli labile toxin, is broadly protective against Shigella spp. challenge in a mouse lethal pulmonary model. Here, we assessed the effect of LDBF, formulated with a unique TLR4 agonist called BECC470 in an oil-in-water emulsion (ME), on the murine immune response in a high-risk population (young and elderly) in response to Shigella challenge. Dual RNA Sequencing captured the transcriptome during Shigella infection in vaccinated and unvaccinated mice. Both age groups were protected by the L-DBF formulation, while younger vaccinated mice exhibited more adaptive immune response gene patterns. This preliminary study provides a step toward identifying the gene expression patterns and regulatory pathways responsible for a protective immune response against Shigella. Furthermore, this study provides a measure of the challenges that need to be addressed when immunizing an aging population.
Collapse
Affiliation(s)
- Ti Lu
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| | - Murugesan Raju
- Bioinformatics and Analytic Core, University of Missouri, Columbia, MO 65211, USA (L.M.C.)
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
| | - Debaki R. Howlader
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| | - Zackary K. Dietz
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| | - Sean K. Whittier
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| | - David J. Varisco
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD 21201, USA
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD 21201, USA
| | - Lyndon M. Coghill
- Bioinformatics and Analytic Core, University of Missouri, Columbia, MO 65211, USA (L.M.C.)
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
| | - William D. Picking
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| | - Wendy L. Picking
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| |
Collapse
|
5
|
Wu Y, Sun A, Chen F, Zhao Y, Zhu X, Zhang T, Ni G, Wang R. Synthesis, structure-activity relationship and biological evaluation of indole derivatives as anti-Candida albicans agents. Bioorg Chem 2024; 146:107293. [PMID: 38507998 DOI: 10.1016/j.bioorg.2024.107293] [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: 11/30/2023] [Revised: 02/29/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024]
Abstract
In this work, we synthesized a series of indole derivatives to cope with the current increasing fungal infections caused by drug-resistant Candida albicans. All compounds were evaluated for antifungal activities against Candida albicans in vitro, and the structure-activity relationships (SARs) were analyzed. The results indicated that indole derivatives used either alone or in combination with fluconazole showed good activities against fluconazole-resistant Candida albicans. Further mechanisms studies demonstrated that compound 1 could inhibit yeast-to-hypha transition and biofilm formation of Candida albicans, increase the activity of the efflux pump, the damage of mitochondrial function, and the decrease of intracellular ATP content. In vivo studies, further proved the anti-Candida albicans activity of compound 1 by histological observation. Therefore, compound 1 could be considered as a novel antifungal agent.
Collapse
Affiliation(s)
- Yandan Wu
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Aimei Sun
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Fei Chen
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Yin Zhao
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Xianhu Zhu
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Tianbao Zhang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Guanghui Ni
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China.
| | - Ruirui Wang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming 650500, Yunnan Province, China.
| |
Collapse
|
6
|
Thomas MF, Slowikowski K, Manakongtreecheep K, Sen P, Samanta N, Tantivit J, Nasrallah M, Zubiri L, Smith NP, Tirard A, Ramesh S, Arnold BY, Nieman LT, Chen JH, Eisenhaure T, Pelka K, Song Y, Xu KH, Jorgji V, Pinto CJ, Sharova T, Glasser R, Chan P, Sullivan RJ, Khalili H, Juric D, Boland GM, Dougan M, Hacohen N, Li B, Reynolds KL, Villani AC. Single-cell transcriptomic analyses reveal distinct immune cell contributions to epithelial barrier dysfunction in checkpoint inhibitor colitis. Nat Med 2024; 30:1349-1362. [PMID: 38724705 DOI: 10.1038/s41591-024-02895-x] [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: 06/06/2022] [Accepted: 03/01/2024] [Indexed: 05/23/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapy has revolutionized oncology, but treatments are limited by immune-related adverse events, including checkpoint inhibitor colitis (irColitis). Little is understood about the pathogenic mechanisms driving irColitis, which does not readily occur in model organisms, such as mice. To define molecular drivers of irColitis, we used single-cell multi-omics to profile approximately 300,000 cells from the colon mucosa and blood of 13 patients with cancer who developed irColitis (nine on anti-PD-1 or anti-CTLA-4 monotherapy and four on dual ICI therapy; most patients had skin or lung cancer), eight controls on ICI therapy and eight healthy controls. Patients with irColitis showed expanded mucosal Tregs, ITGAEHi CD8 tissue-resident memory T cells expressing CXCL13 and Th17 gene programs and recirculating ITGB2Hi CD8 T cells. Cytotoxic GNLYHi CD4 T cells, recirculating ITGB2Hi CD8 T cells and endothelial cells expressing hypoxia gene programs were further expanded in colitis associated with anti-PD-1/CTLA-4 therapy compared to anti-PD-1 therapy. Luminal epithelial cells in patients with irColitis expressed PCSK9, PD-L1 and interferon-induced signatures associated with apoptosis, increased cell turnover and malabsorption. Together, these data suggest roles for circulating T cells and epithelial-immune crosstalk critical to PD-1/CTLA-4-dependent tolerance and barrier function and identify potential therapeutic targets for irColitis.
Collapse
Affiliation(s)
- Molly Fisher Thomas
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA.
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Division of Gastroenterology, Department of Medicine, Oregon Health and Sciences University, Portland, OR, USA.
- Department of Cell, Developmental, and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA.
| | - Kamil Slowikowski
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA.
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Kasidet Manakongtreecheep
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Pritha Sen
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Transplant, Oncology, and Immunocompromised Host Group, Division of Infectious Disease, Department of Medicine, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nandini Samanta
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Jessica Tantivit
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Mazen Nasrallah
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Department of Medicine, North Shore Physicians Group, Mass General Brigham Healthcare Center, Lynn, MA, USA
| | - Leyre Zubiri
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Hematology-Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Neal P Smith
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Alice Tirard
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Swetha Ramesh
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Benjamin Y Arnold
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Linda T Nieman
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jonathan H Chen
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Thomas Eisenhaure
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Karin Pelka
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Yuhui Song
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Katherine H Xu
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Vjola Jorgji
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Tatyana Sharova
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Rachel Glasser
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - PuiYee Chan
- Harvard Medical School, Boston, MA, USA
- Clinical Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Ryan J Sullivan
- Harvard Medical School, Boston, MA, USA
- Division of Hematology-Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Hamed Khalili
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Dejan Juric
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Hematology-Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Genevieve M Boland
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Michael Dougan
- Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Nir Hacohen
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bo Li
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Genentech, South San Francisco, CA, USA
| | - Kerry L Reynolds
- Harvard Medical School, Boston, MA, USA
- Division of Hematology-Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Alexandra-Chloé Villani
- Department of Medicine, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA.
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Krupka-Olek M, Bożek A, Aebisher D, Bartusik-Aebisher D, Cieślar G, Kawczyk-Krupka A. Potential Aspects of the Use of Cytokines in Atopic Dermatitis. Biomedicines 2024; 12:867. [PMID: 38672221 PMCID: PMC11048200 DOI: 10.3390/biomedicines12040867] [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: 02/18/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Atopic dermatitis (AD) is an abnormal inflammatory response in the skin to food, environmental IgE, or non-IgE allergens. This disease belongs to a group of inflammatory diseases that affect both children and adults. In highly developed countries, AD is diagnosed twice as often in children than in adults, which may possibly be connected to increased urbanization. The immune system's pathomechanisms of AD involve humoral mechanisms with IgE, cellular T lymphocytes, dendritic cells occurring in the dermis, Langerhans cells occurring in the epidermis, and other cells infiltrating the site of inflammation (eosinophils, macrophages, mast cells, neutrophils, and basophils). Cytokines are small proteins that affect the interaction and communication between cells. This review characterizes cytokines and potential aspects of the treatment of atopic dermatitis, as well as new strategies that are currently being developed, including targeting cytokines and their receptors.
Collapse
Affiliation(s)
- Magdalena Krupka-Olek
- Clinical Department of Internal Diseases and Geriatrics, Chair of Internal Diseases, Dermatology and Allergology in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland (A.B.)
- Doctoral School, Medical University of Silesia, 40-055 Katowice, Poland
| | - Andrzej Bożek
- Clinical Department of Internal Diseases and Geriatrics, Chair of Internal Diseases, Dermatology and Allergology in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland (A.B.)
| | - David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland;
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland;
| | - Grzegorz Cieślar
- Department of Internal Diseases, Angiology and Physical Medicine, Centre for Laser Diagnostics and Therapy, Medical University of Silesia in Katowice, Batorego 15, 41-902 Bytom, Poland;
| | - Aleksandra Kawczyk-Krupka
- Department of Internal Diseases, Angiology and Physical Medicine, Centre for Laser Diagnostics and Therapy, Medical University of Silesia in Katowice, Batorego 15, 41-902 Bytom, Poland;
| |
Collapse
|
8
|
Liu X, Wang H, Wang X, Jiang X, Jin Y, Han Y, Zhang Z. Identification and verification of inflammatory biomarkers for primary Sjögren's syndrome. Clin Rheumatol 2024; 43:1335-1352. [PMID: 38376769 PMCID: PMC10944815 DOI: 10.1007/s10067-024-06901-y] [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: 10/04/2023] [Revised: 10/04/2023] [Accepted: 02/01/2024] [Indexed: 02/21/2024]
Abstract
INTRODUCTION Primary Sjögren's syndrome (pSS) is an autoimmune disease characterized by inflammatory infiltration, and dysfunction of the salivary and lacrimal glands. This research aimed to explore the disease pathogenesis and improve the diagnosis and treatment of pSS by mining inflammation-associated biomarkers. METHODS Five pSS-related datasets were retrieved from the Gene Expression Omnibus (GEO) database. Inflammation-associated biomarkers were determined by the least absolute shrinkage and selection operator (LASSO) and support vector machines recursive feature elimination (SVM-RFE). Single sample gene set enrichment analysis (ssGSEA) was implemented to profile the infiltration levels of immune cells. Real-time quantitative PCR (RT-qPCR) verified the expression of biomarkers in clinical samples. RESULTS Four genes (LY6E, EIF2AK2, IL15, and CXCL10) were screened as inflammation-associated biomarkers in pSS, the predictive performance of which were determined among three pSS-related datasets (AUC > 0.7). Functional enrichment results suggested that the biomarkers were involved in immune and inflammation-related pathways. Immune infiltration analysis revealed that biomarkers were notably connected with type 2 T helper cells, regulatory T cells which were significantly expressed between pSS and control. TESTOSTERONE and CYCLOSPORINE were predicted to take effect by targeting CXCL10 and IL15 in pSS, respectively. CONCLUSION Four inflammation-associated biomarkers (LY6E, EIF2AK2, IL15, and CXCL10) were explored, and the underlying regulatory mechanisms and targeted drugs associated with these biomarkers were preliminarily investigated according to a series of bioinformatics methods based on the online datasets of pSS, which provided a reference for understanding the pathogenesis of pSS. Key Points • Inflammation-associated biomarkers (LY6E, EIF2AK2, IL15, and CXCL10) were firstly identified in Sjögren's syndrome based on LASSO and SVM-RFE analyses. • CXCL10, EIF2AK2 and LY6E were prominently positively correlated with immature B cells, while IL15 were significantly negatively correlated with memory B cells in Sjögren's syndrome. • LY6E, EIF2AK2, IL15, and CXCL10 were significantly more highly expressed in clinical Sjögren's syndrome samples compared to healthy control samples, which was consistent with the analysis results of the GEO database. •LY6E, EIF2AK2, IL15, and CXCL10 might be used as the biomarkers for the treatment and diagnosis of Sjögren's syndrome.
Collapse
Affiliation(s)
- Xiaodan Liu
- Department of Stomatology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
| | - Haojie Wang
- Department of Stomatology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
| | - Xiao Wang
- Department of Stomatology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
| | - Xiaodan Jiang
- Department of Ophthalmology, Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Peking University Third Hospital, Beijing, 100191, China
| | - Yinji Jin
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China
| | - Ying Han
- Department of Oral Medicine, Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering, Haidian District, 22 Zhongguancun South Avenue, Beijing, 100081, China.
| | - Zhihui Zhang
- Department of Stomatology, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China.
| |
Collapse
|
9
|
Mohamed NM, Abdelhamid AM, Aref M, Abdelhafeez M, Faris Alotabi H, Mohammed Abdelrahman DS, Elwany NE. Role of cytokines and Th17/Tregs imbalance in the pathogenesis of otitis media with effusion. Modulation of Notch1/Hes1/mTORC1/S6k1 signalling pathway underlies the protective effect of astaxanthin. Int Immunopharmacol 2024; 128:111521. [PMID: 38246005 DOI: 10.1016/j.intimp.2024.111521] [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: 08/22/2023] [Revised: 12/26/2023] [Accepted: 01/06/2024] [Indexed: 01/23/2024]
Abstract
Otitis media with effusion (OME) is a recurrent middle ear inflammatory condition. It may be complicated by acquired hearing loss and speech impairment especially in children. Accordingly, the current study aimed to assess the role of cytokines and the imbalance of Th17/Tregs in the pathogenesis of OME. Additionally, the protective effect of astaxanthin and its mechanisms related to Notch1/ Hes1/mTORC1/S6K1 signalling were investigated. METHODS Forty-eight children were grouped as follow: G1: control healthy group G2: acute otitis media (AOM) group, G3: OME group. In the lipopolysaccharide (LPS) induced OME rat model, 15 rats were randomised into: G1: normal control group, G2: LPS group, and G3: astaxanthin treated group. RESULTS Biochemical analysis of the children's peripheral blood samples showed that IL1β, IL-2, IL-4, IL-6, IL-17, and IL-23 were significantly elevated, while TGF-β was significantly decreased in AOM and OME patients (group 2 and 3). In the LPS- induced OME rat model, astaxanthin treatment resulted in suppression of IL-17, IL-6, TNF-α, Muc5A, TFF3, NICD, Hes1, mTORC1, and S6K1 in rat middle ear mucosa. Furthermore, astaxanthin significantly downregulated RORγ while upregulating FoxP3 and restored the balance between Th17/Tregs. Moreover, astaxanthin improved the histopathological picture of the inflamed middle ear mucosa. CONCLUSIONS Proinflammatory cytokines as well as Th17/Tregs imbalance play a crucial role in the pathogenesis of AOM and OME. Additionally, astaxanthin alleviated LPS- induced OME in rats through suppression of Notch1/ Hes1/mTORC1/S6K1 pathway, and regulation of Th17/Tregs.
Collapse
Affiliation(s)
- Noura Mostafa Mohamed
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, 11671 Riyadh, Saudi Arabia.
| | | | - Mohamed Aref
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Zagazig University, Egypt.
| | - Marwa Abdelhafeez
- Department of Otorhinolaryngology, Faculty of Medicine, Minia University, Minia, Egypt.
| | - Hadil Faris Alotabi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, 11671 Riyadh, Saudi Arabia.
| | - Doaa Sayed Mohammed Abdelrahman
- Department of Clinical Science, College of Medicine, Princess Nourah bint Abdulrahman University, 11671 Riyadh, Saudi Arabia.
| | - Nisreen E Elwany
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Egypt.
| |
Collapse
|
10
|
Wang X, Han C, Yang D, Zhou J, Dong H, Wei Z, Xu S, Xu C, Zhang Y, Sun Y, Ni B, Guo S, Zhang J, Zhao T, Chen X, Luo J, Wu Y, Tian Y. STAT3 and SOX-5 induce BRG1-mediated chromatin remodeling of RORCE2 in Th17 cells. Commun Biol 2024; 7:10. [PMID: 38172644 PMCID: PMC10764326 DOI: 10.1038/s42003-023-05735-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Retinoid-related orphan receptor gamma t (RORγt) is the lineage-specific transcription factor for T helper 17 (Th17) cells. Our previous study demonstrated that STAT3 likely participates in the activation of RORCE2 (a novel enhancer of the RORγt gene) in Th17 cells. However, the detailed mechanism is still unclear. Here, we demonstrate that both STAT3 and SOX-5 mediate the enhancer activity of RORCE2 in vitro. Deletion of the STAT3 binding site (STAT3-BS) in RORCE2 impaired RORγt expression and Th17 differentiation, resulting in reduced severity of experimental autoimmune encephalomyelitis (EAE). Mechanistically, STAT3 and SOX-5 bind the RORCE2 region and recruit the chromatin remodeling factor BRG1 to remodel the nucleosomes positioned at this region. Collectively, our data suggest that STAT3 and SOX-5 mediate the differentiation of Th17 cells through the induction of BRG1-mediated chromatin remodeling of RORCE2 in Th17 cells.
Collapse
Affiliation(s)
- Xian Wang
- Institute of Immunology, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
- Department of Immunology, Medical College of Qingdao University, 266071, Qingdao, Shandong, People's Republic of China
| | - Chao Han
- Institute of Immunology, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
| | - Di Yang
- Institute of Immunology, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
| | - Jian Zhou
- Institute of Immunology, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
| | - Hui Dong
- Institute of Immunology, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
| | - Zhiyuan Wei
- The First Affiliated Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
| | - Shuai Xu
- The Second Affiliated Hospital, Third Military Medical University (Army Medical University), 400037, Chongqing, People's Republic of China
| | - Chen Xu
- Institute of Immunology, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
| | - Yiwei Zhang
- Institute of Immunology, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
| | - Yi Sun
- The First Affiliated Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
| | - Bing Ni
- Department of Pathophysiology, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
| | - Sheng Guo
- Institute of Immunology, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
| | - Jingbo Zhang
- The Second Affiliated Hospital, Third Military Medical University (Army Medical University), 400037, Chongqing, People's Republic of China
| | - Tingting Zhao
- Chongqing International Institute for Immunology, 400030, Chongqing, People's Republic of China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, 100853, Beijing, China
| | - Jie Luo
- The First Affiliated Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China
| | - Yuzhang Wu
- Institute of Immunology, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China.
- Chongqing International Institute for Immunology, 400030, Chongqing, People's Republic of China.
| | - Yi Tian
- Institute of Immunology, Third Military Medical University (Army Medical University), 400038, Chongqing, People's Republic of China.
| |
Collapse
|
11
|
Hu S, Zhao R, Xu Y, Gu Z, Zhu B, Hu J. Orally-administered nanomedicine systems targeting colon inflammation for the treatment of inflammatory bowel disease: latest advances. J Mater Chem B 2023; 12:13-38. [PMID: 38018424 DOI: 10.1039/d3tb02302h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic and idiopathic condition that results in inflammation of the gastrointestinal tract, leading to conditions such as ulcerative colitis and Crohn's disease. Commonly used treatments for IBD include anti-inflammatory drugs, immunosuppressants, and antibiotics. Fecal microbiota transplantation is also being explored as a potential treatment method; however, these drugs may lead to systemic side effects. Oral administration is preferred for IBD treatment, but accurately locating the inflamed area in the colon is challenging due to multiple physiological barriers. Nanoparticle drug delivery systems possess unique physicochemical properties that enable precise delivery to the target site for IBD treatment, exploiting the increased permeability and retention effect of inflamed intestines. The first part of this review comprehensively introduces the pathophysiological environment of IBD, covering the gastrointestinal pH, various enzymes in the pathway, transport time, intestinal mucus, intestinal epithelium, intestinal immune cells, and intestinal microbiota. The second part focuses on the latest advances in the mechanism and strategies of targeted delivery using oral nanoparticle drug delivery systems for colitis-related fields. Finally, we present challenges and potential directions for future IBD treatment with the assistance of nanotechnology.
Collapse
Affiliation(s)
- Shumeng Hu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
| | - Runan Zhao
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yu Xu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| | - Zelin Gu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
| | - Beiwei Zhu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| | - Jiangning Hu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| |
Collapse
|
12
|
Wan T, Wang Y, He K, Zhu S. Microbial sensing in the intestine. Protein Cell 2023; 14:824-860. [PMID: 37191444 PMCID: PMC10636641 DOI: 10.1093/procel/pwad028] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/04/2023] [Indexed: 05/17/2023] Open
Abstract
The gut microbiota plays a key role in host health and disease, particularly through their interactions with the immune system. Intestinal homeostasis is dependent on the symbiotic relationships between the host and the diverse gut microbiota, which is influenced by the highly co-evolved immune-microbiota interactions. The first step of the interaction between the host and the gut microbiota is the sensing of the gut microbes by the host immune system. In this review, we describe the cells of the host immune system and the proteins that sense the components and metabolites of the gut microbes. We further highlight the essential roles of pattern recognition receptors (PRRs), the G protein-coupled receptors (GPCRs), aryl hydrocarbon receptor (AHR) and the nuclear receptors expressed in the intestinal epithelial cells (IECs) and the intestine-resident immune cells. We also discuss the mechanisms by which the disruption of microbial sensing because of genetic or environmental factors causes human diseases such as the inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Tingting Wan
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Yalong Wang
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Kaixin He
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Shu Zhu
- Division of Life Sciences and Medicine, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Institute of Immunology, School of Basic Medical Sciences, University of Science and Technology of China, Hefei 230027, China
- Department of Digestive Disease, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei 230001, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, China
| |
Collapse
|
13
|
Larson EC, Ellis-Connell AL, Rodgers MA, Gubernat AK, Gleim JL, Moriarty RV, Balgeman AJ, Ameel CL, Jauro S, Tomko JA, Kracinovsky KB, Maiello P, Borish HJ, White AG, Klein E, Bucsan AN, Darrah PA, Seder RA, Roederer M, Lin PL, Flynn JL, O'Connor SL, Scanga CA. Intravenous Bacille Calmette-Guérin vaccination protects simian immunodeficiency virus-infected macaques from tuberculosis. Nat Microbiol 2023; 8:2080-2092. [PMID: 37814073 PMCID: PMC10627825 DOI: 10.1038/s41564-023-01503-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/13/2023] [Indexed: 10/11/2023]
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (Mtb), is the most common cause of death in people living with human immunodeficiency virus (HIV). Intra-dermal Bacille Calmette-Guérin (BCG) delivery is the only licensed vaccine against tuberculosis; however, it offers little protection from pulmonary tuberculosis in adults and is contraindicated in people living with HIV. Intravenous BCG confers protection against Mtb infection in rhesus macaques; we hypothesized that it might prevent tuberculosis in simian immunodeficiency virus (SIV)-infected macaques, a model for HIV infection. Here intravenous BCG-elicited robust airway T cell influx and elevated plasma and airway antibody titres in both SIV-infected and naive animals. Following Mtb challenge, all 7 vaccinated SIV-naive and 9 out of 12 vaccinated SIV-infected animals were protected, without any culturable bacteria detected from tissues. Peripheral blood mononuclear cell responses post-challenge indicated early clearance of Mtb in vaccinated animals, regardless of SIV infection. These data support that intravenous BCG is immunogenic and efficacious in SIV-infected animals.
Collapse
Affiliation(s)
- Erica C Larson
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Amy L Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Mark A Rodgers
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Abigail K Gubernat
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Janelle L Gleim
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan V Moriarty
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Alexis J Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| | - Cassaundra L Ameel
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Solomon Jauro
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jaime A Tomko
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kara B Kracinovsky
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - H Jake Borish
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexander G White
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Edwin Klein
- Division of Laboratory Animal Resources, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Allison N Bucsan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patricia A Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Philana Ling Lin
- Department of Pediatrics, Children's Hospital of Pittsburgh, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shelby L O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI, USA
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI, USA
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
14
|
Losol P, Ji MH, Kim JH, Choi JP, Yun JE, Seo JH, Kim BK, Chang YS, Kim SH. Bronchial epithelial cells release inflammatory markers linked to airway inflammation and remodeling in response to TLR5 ligand flagellin. World Allergy Organ J 2023; 16:100786. [PMID: 37332524 PMCID: PMC10276272 DOI: 10.1016/j.waojou.2023.100786] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/25/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
Background/Aims Flagellin, which is abundant in gram-negative bacteria, including Pseudomonas, is reported to influence on inflammatory responses in various lung diseases. However, its effect on airway epithelial cells in contribution to asthma pathogenesis is not elucidated yet. We aimed to investigate the effect of TLR5 ligand flagellin on the transcriptomic profile of primary human epithelial cells and to determine the markers of airway inflammation. Methods Normal human bronchial epithelial (NHBE) cells were grown and differentiated in air-liquid interface (ALI) culture for 14-16 days. The cells were treated with flagellin in vitro at 10 and 100 ng/ml for 3 and 24 h. The conditioned media and cells were harvested to validate inflammatory markers involved in airway inflammation using ELISA, Western blot, and quantitative PCR methods. RNA-sequencing was performed to investigate the transcriptional response to flagellin in ALI-NHBE cells. Results Altered transcriptional responses to flagellin in differentiated bronchial epithelial cells were determined, including genes encoding chemokines, matrix metalloproteinases, and antimicrobial biomolecules. Pathway analysis of the transcriptionally responsive genes revealed enrichment of signaling pathways. Flagellin induced the mRNA expressions of proinflammatory cytokines and chemokines, and secretion of GM-CSF, CXCL5, CCL5 and CXCL10. Flagellin enhanced the protein expression of MMP-13 in TGF-β1 and TGF-β2 pretreated cell lysates and Wnt/β-catenin signaling. Conclusions These findings suggest that flagellin could be a potent inducer of inflammatory markers that may contribute to airway inflammation and remodeling.
Collapse
Affiliation(s)
- Purevsuren Losol
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Medical Research Center, Seoul National University, Seoul, South Korea
| | - Mi-Hong Ji
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jin Hee Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jun-Pyo Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jeong-Eun Yun
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Jang-Ho Seo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Byung-Keun Kim
- Department of Internal Medicine, Korea University Medical Center Anam Hospital, Seoul, South Korea
| | - Yoon-Seok Chang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Medical Research Center, Seoul National University, Seoul, South Korea
| | - Sae-Hoon Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
15
|
Larson EC, Ellis-Connell AL, Rodgers MA, Gubernat AK, Gleim JL, Moriarty RV, Balgeman AJ, Ameel CL, Jauro S, Tomko JA, Kracinovsky KB, Maiello P, Borish HJ, White AG, Klein E, Bucsan AN, Darrah PA, Seder RA, Roederer M, Lin PL, Flynn JL, O'Connor SL, Scanga CA. Vaccination with intravenous BCG protects macaques with pre-existing SIV infection from tuberculosis. RESEARCH SQUARE 2023:rs.3.rs-2802306. [PMID: 37090620 PMCID: PMC10120779 DOI: 10.21203/rs.3.rs-2802306/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Tuberculosis (TB) is the most common cause of death in people living with HIV. BCG delivered intradermally (ID) is the only licensed vaccine to prevent TB. However, it offers little protection from pulmonary TB in adults. Intravenous (IV) BCG, but not ID BCG, confers striking protection against Mycobacterium tuberculosis (Mtb) infection and disease in rhesus macaques. We investigated whether IV BCG could protect against TB in macaques with a pre-existing SIV infection. There was a robust influx of airway T cells following IV BCG in both SIV-infected and SIV-naïve animals, with elevated antibody titers in plasma and airways. Following Mtb challenge, all 7 SIV-naïve and 9 out of 12 SIV-infected vaccinated animals were completely protected, without any culturable bacilli in their tissues. PBMC responses post-challenge indicated early clearance of Mtb in vaccinated animals regardless of SIV infection. These data support that IV BCG is immunogenic and efficacious in SIV-infected animals.
Collapse
Affiliation(s)
- Erica C Larson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Amy L Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Mark A Rodgers
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Abigail K Gubernat
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Janelle L Gleim
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Ryan V Moriarty
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Alexis J Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Madison, WI, USA
| | - Cassaundra L Ameel
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Solomon Jauro
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Jaime A Tomko
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Kara B Kracinovsky
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Pauline Maiello
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - H Jake Borish
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Alexander G White
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Edwin Klein
- Division of Laboratory Animal Resources, School of Medicine, University of Pittsburgh, PA, USA
| | - Allison N Bucsan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Patricia A Darrah
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Mario Roederer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Philana Ling Lin
- Department of Pediatrics, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - JoAnne L Flynn
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Shelby L O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin - Madison, Madison, WI, USA
- Wisconsin National Primate Research Center, University of Wisconsin - Madison, Madison, WI, USA
| | - Charles A Scanga
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
16
|
Network-Based Assessment of Minimal Change Disease Identifies Glomerular Response to IL-7 and IL-12 Pathways Activation as Innovative Treatment Target. Biomedicines 2023; 11:biomedicines11010226. [PMID: 36672735 PMCID: PMC9856051 DOI: 10.3390/biomedicines11010226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/20/2022] [Accepted: 01/07/2023] [Indexed: 01/18/2023] Open
Abstract
Background: Minimal change disease (MCD), a major cause of nephrotic syndrome, is usually treated by corticosteroid administration. MCD unresponsiveness to therapy and recurrences are nonetheless frequently observed, particularly in adults. To explore MCD-related pathogenetic mechanisms and to identify novel drug targets ultimately contributing to novel therapeutic avenues with a certain specificity for MCD, we compared glomerular transcriptomes from MCD with membranous nephropathy (MN) patients and healthy controls. Methods: Renal biopsies from adult patients with MCD (n = 14) or MN (n = 12), and non-diseased controls (n = 8) were selected from the Norwegian Kidney Biopsy Registry. RNA for 75 base-pair paired-end RNASeq were obtained from laser capture micro-dissected (LCM) glomeruli from FFPE sections. Transcriptional landscapes were computed by combining pathway-centered analyses and network science methodologies that integrate multiple bioinformatics resources. Results: Compared to normal glomeruli, cells from MCD displayed an inflammatory signature apparently governed by the IL1 and IL7 systems. While enrichment of IL1 production and secretion was a shared feature of MCD and MN compared to normal tissue, responses involving IL7 pathway activation were unique to MCD. Indeed, IL7R expressed by glomeruli was the most upregulated gene of the interleukin family in MCD versus normal controls. IL7 pathway activation was paralleled by significant enrichment in adaptive immune system processes and transcriptional regulation and depletion in pathways related to energy metabolism and transcription. Downregulation of these organ function-related themes again occurred predominately in MCD and was significantly less pronounced in MN. Immunofluorescence and immunohistochemistry, respectively, confirmed the expression of phosphorylated IL-7 receptor alpha (IL7RA, CD127) and IL12 receptor beta 1 (IL12RB1) proteins. Conclusions: Gene expression profiling of archival FFPE-biopsies identifies MCD-specific signatures with IL7RA and IL12RB1 as novel targets for MCD treatment.
Collapse
|
17
|
Lin SJH, Helm ET, Gabler NK, Burrough ER. Acute infection with Brachyspira hyodysenteriae affects mucin expression, glycosylation, and fecal MUC5AC. Front Cell Infect Microbiol 2023; 12:1042815. [PMID: 36683692 PMCID: PMC9852840 DOI: 10.3389/fcimb.2022.1042815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/28/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction Infection with strongly β-hemolytic strains of Brachyspira hyodysenteriae leads to swine dysentery (SD), a production-limiting disease that causes mucohemorrhagic diarrhea and typhlocolitis in pigs. This pathogen has strong chemotactic activity toward mucin, and infected pigs often have a disorganized mucus layer and marked de novo expression of MUC5AC, which is not constitutively expressed in the colon. It has been shown that fucose is chemoattractant for B. hyodysenteriae, and a highly fermentable fiber diet can mitigate and delay the onset of SD. Methods We used lectins targeting sialic acids in α-2,6 or α-2,3 linkages, N-acetylglucosamine (GlcNAc), α-linked L-fucose, and an immunohistochemical stain targeting N-glycolylneuraminic acid (NeuGc) to investigate the local expression of these mucin glycans in colonic tissues of pigs with acute SD. We used a commercial enzyme-linked immunosorbent assay (ELISA) to quantify fecal MUC5AC in infected pigs and assess its potential as a diagnostic monitoring tool and RNA in situ hybridization to detect IL-17A in the colonic mucosa. Results Colonic mucin glycosylation during SD has an overall increase in fucose, a spatially different distribution of GlcNAc with more expression within the crypt lumens of the upper colonic mucosa, and decreased expression or a decreased trend of sialic acids in α-2,6 or α-2,3 linkages, and NeuGc compared to the controls. The degree of increased fucosylation was less in the colonic mucosa of pigs with SD and fed the highly fermentable fiber diet. There was a significant increase in MUC5AC in fecal and colonic samples of pigs with SD at the endpoint compared to the controls, but the predictive value for disease progression was limited. Discussion Fucosylation and the impact of dietary fiber may play important roles in the pathogenesis of SD. The lack of predictive value for fecal MUC5AC quantification by ELISA is possibly due to the presence of other non-colonic sources of MUC5AC in the feces. The moderate correlation between IL-17A, neutrophils and MUC5AC confirms its immunoregulatory and mucin stimulatory role. Our study characterizes local alteration of mucin glycosylation in the colonic mucosa of pigs with SD after B. hyodysenteriae infection and may provide insight into host-pathogen interaction.
Collapse
Affiliation(s)
- Susanne Je-Han Lin
- Department of Veterinary Pathology, Iowa State University, Ames, IA, United States
| | - Emma T Helm
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Nicholas K Gabler
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Eric R Burrough
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
18
|
Zhang X, Zhang X, Song X, Xiang C, He C, Xie Y, Zhou Y, Wang N, Guo G, Zhang W, Li Y, Liu K, Zou Q, Guo H, Shi Y. Interleukin 17 B regulates colonic myeloid cell infiltration in a mouse model of DSS-induced colitis. Front Immunol 2023; 14:1055256. [PMID: 36814913 PMCID: PMC9940313 DOI: 10.3389/fimmu.2023.1055256] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/16/2023] [Indexed: 02/08/2023] Open
Abstract
Cytokines play vital roles in the pathogenesis of inflammatory bowel disease. IL17B is protective in the development of colitis. However, how IL17B regulates intestinal inflammation and what cells are regulated by IL17B is still unknown. Here, we aimed to illustrate the IL17B dependent cellular and molecular changes in colon tissue in a mouse colitis model. The results showed that IL17B expression in colon tissues was elevated in inflamed tissues than non-inflamed tissues of IBD patients. Wild type (WT) and Il17b deficient (Il17b -/-) mice were given 2.5% dextran sodium sulfate (DSS) water, and in some case, Il17b -/- mice were treated with recombinant mouse IL17B. IL17B deficiency resulted in severe DSS-induced colitis with exaggerated weight loss, shorter colon length, and elevated proinflammatory cytokines in colon. Reconstitution of Il17b -/- mice with recombinant IL17B alleviated the severity of DSS-induced colitis. Single cell transcriptional analyses of CD45+ immune cells in colonic lamina propria revealed that loss of IL17B resulted in an increased neutrophil infiltration and enhanced inflammatory cytokines in intestinal macrophages in colitis, which were confirmed by real-time PCR and flow cytometry. IL17B treatment also inhibited lipopolysaccharide-induced inflammation in bone marrow-derived macrophages and mice. IL17B inhibits colitis by regulating colonic myeloid cell response. It might represent a novel potential therapeutic approach to treat the colitis.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Institute of Biopharmaceutical Research, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaokai Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiaomei Song
- Department of Gastroenterology, Chongqing General Hospital, Chongqing, China
| | - Chuanying Xiang
- Institute of Biopharmaceutical Research, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunmei He
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yu Xie
- Institute of Biopharmaceutical Research, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yangyang Zhou
- Institute of Biopharmaceutical Research, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ning Wang
- Institute of Biopharmaceutical Research, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Gang Guo
- Institute of Biopharmaceutical Research, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yan Li
- Institute of Biopharmaceutical Research, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kaiyun Liu
- Institute of Biopharmaceutical Research, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hong Guo
- Department of Gastroenterology, Chongqing General Hospital, Chongqing, China
| | - Yun Shi
- Institute of Biopharmaceutical Research, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
19
|
Ramirez-Diaz J, Cenadelli S, Bornaghi V, Bongioni G, Montedoro SM, Achilli A, Capelli C, Rincon JC, Milanesi M, Passamonti MM, Colli L, Barbato M, Williams JL, Marsan PA. Identification of genomic regions associated with total and progressive sperm motility in Italian Holstein bulls. J Dairy Sci 2023; 106:407-420. [PMID: 36400619 DOI: 10.3168/jds.2021-21700] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 08/10/2022] [Indexed: 11/17/2022]
Abstract
Sperm motility is directly related to the ability of sperm to move through the female reproductive tract to reach the ovum. Sperm motility is a complex trait that is influenced by environmental and genetic factors and is associated with male fertility, oocyte penetration rate, and reproductive success of cattle. In this study we carried out a GWAS in Italian Holstein bulls to identify candidate regions and genes associated with variations in progressive and total motility (PM and TM, respectively). After quality control, the final data set consisted of 5,960 records from 949 bulls having semen collected in 10 artificial insemination stations and genotyped at 412,737 SNPs (call rate >95%; minor allele frequency >5%). (Co)variance components were estimated using single trait mixed models, and associations between SNPs and phenotypes were assessed using a genomic BLUP approach. Ten windows that explained the greatest percentage of genetic variance were located on Bos taurus autosomes 1, 2, 4, 6, 7, 23, and 26 for TM and Bos taurus autosomes 1, 2, 4, 6, 8, 16, 23, and 26 for PM. A total of 150 genes for TM and 72 genes for PM were identified within these genomic regions. Gene Ontology enrichment analyses identified significant Gene Ontology terms involved with energy homeostasis, membrane functions, sperm-egg interactions, protection against oxidative stress, olfactory receptors, and immune system. There was significant enrichment of quantitative trait loci for fertility, calving ease, immune response, feed intake, and carcass weight within the candidate windows. These results contribute to understanding the architecture of the genetic control of sperm motility and may aid in the development of strategies to identify subfertile bulls and improve reproductive success.
Collapse
Affiliation(s)
- J Ramirez-Diaz
- Department of Animal Sciences, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, Piacenza, Italy 29122; Institute of Agricultural Biology and Biotechnology (IBBA), Consiglio Nazionale di Ricerca, Milano, Italy.
| | - S Cenadelli
- Institute Lazzaro Spallanzani, Rivolta d'Adda (CR), Cremona, Italy
| | - V Bornaghi
- Institute Lazzaro Spallanzani, Rivolta d'Adda (CR), Cremona, Italy
| | - G Bongioni
- Institute Lazzaro Spallanzani, Rivolta d'Adda (CR), Cremona, Italy
| | - S M Montedoro
- Institute Lazzaro Spallanzani, Rivolta d'Adda (CR), Cremona, Italy
| | - A Achilli
- Department of Biology and Biotechnology, Università degli Studi di Pavia, Pavia, Italy
| | - C Capelli
- Department of Chemical, Life and Environmental Sustainability Sciences, Università degli Studi di Parma, Parma, Italy
| | - J C Rincon
- Department of Animal Science, Universidad Nacional de Colombia, Palmira, Valle del Cauca, Colombia
| | - M Milanesi
- Department for Innovation in Biological, Agri-food and Forestry Systems (DIBAF), Università degli Studi della Tuscia, Viterbo, Italy
| | - M M Passamonti
- Department of Animal Sciences, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, Piacenza, Italy 29122
| | - L Colli
- Department of Animal Sciences, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, Piacenza, Italy 29122
| | - M Barbato
- Department of Animal Sciences, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, Piacenza, Italy 29122
| | - J L Williams
- Department of Animal Sciences, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, Piacenza, Italy 29122
| | - P Ajmone Marsan
- Department of Animal Sciences, Food and Nutrition (DIANA), Università Cattolica del Sacro Cuore, Piacenza, Italy 29122
| |
Collapse
|
20
|
Ramos A, Granzotto N, Kremer R, Boeder AM, de Araújo JFP, Pereira AG, Izídio GS. Hunting for Genes Underlying Emotionality in the Laboratory Rat: Maps, Tools and Traps. Curr Neuropharmacol 2023; 21:1840-1863. [PMID: 36056863 PMCID: PMC10514530 DOI: 10.2174/1570159x20666220901154034] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/13/2022] [Accepted: 07/28/2022] [Indexed: 11/22/2022] Open
Abstract
Scientists have systematically investigated the hereditary bases of behaviors since the 19th century, moved by either evolutionary questions or clinically-motivated purposes. The pioneer studies on the genetic selection of laboratory animals had already indicated, one hundred years ago, the immense complexity of analyzing behaviors that were influenced by a large number of small-effect genes and an incalculable amount of environmental factors. Merging Mendelian, quantitative and molecular approaches in the 1990s made it possible to map specific rodent behaviors to known chromosome regions. From that point on, Quantitative Trait Locus (QTL) analyses coupled with behavioral and molecular techniques, which involved in vivo isolation of relevant blocks of genes, opened new avenues for gene mapping and characterization. This review examines the QTL strategy applied to the behavioral study of emotionality, with a focus on the laboratory rat. We discuss the challenges, advances and limitations of the search for Quantitative Trait Genes (QTG) playing a role in regulating emotionality. For the past 25 years, we have marched the long journey from emotionality-related behaviors to genes. In this context, our experiences are used to illustrate why and how one should move forward in the molecular understanding of complex psychiatric illnesses. The promise of exploring genetic links between immunological and emotional responses are also discussed. New strategies based on humans, rodents and other animals (such as zebrafish) are also acknowledged, as they are likely to allow substantial progress to be made in the near future.
Collapse
Affiliation(s)
- André Ramos
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Natalli Granzotto
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Rafael Kremer
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Developmental and Cellular Biology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Ariela Maína Boeder
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Julia Fernandez Puñal de Araújo
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Developmental and Cellular Biology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Aline Guimarães Pereira
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Developmental and Cellular Biology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Geison Souza Izídio
- Behavior Genetics Laboratory, Department of Cell Biology, Embryology and Genetics, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Pharmacology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
- Graduate Program of Developmental and Cellular Biology, Center of Biological Sciences, Federal University of Santa Catarina, Florianopolis, Brazil
| |
Collapse
|
21
|
Swedik SM, Madola A, Cruz MA, Llorens-Bonilla BJ, Levine AD. Th17-Derived Cytokines Synergistically Enhance IL-17C Production by the Colonic Epithelium. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1768-1777. [PMID: 36130829 PMCID: PMC9588696 DOI: 10.4049/jimmunol.2200125] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 08/29/2022] [Indexed: 11/06/2022]
Abstract
Tightly regulated communication between the gastrointestinal epithelium and immune cells in the underlying lamina propria is critical for immune homeostasis and inflammation. IL-17C, produced by epithelial cells after exposure to inflammatory stimuli, facilitates cell-to-cell communication by promoting inflammatory responses in Th17 cells. In this study, we demonstrate that Th17-derived cytokines TNF-α, IL-17A, and IL-22 synergistically enhance IL-17C expression in both human-transformed colonic epithelial cell lines and primary non-inflammatory bowel disease colonic epithelial spheroids. This synergistic expression requires activation of the transcription factor NF-κB downstream of the TNF-α stimulus, evidenced by the reduction of IL-17C expression in the presence of an IκBα inhibitor. IL-17A and IL-22 enhance IL-17C expression through the activation of the transcription factor AP-1 in a p38 MAPK-dependent manner. Colonic spheroids derived from uninvolved epithelial of ulcerative colitis patients stimulated with TNF-α, IL-17A, and IL-22 show muted responses compared with non-inflammatory bowel disease spheroids, and inflamed spheroids yielded more IL-17C expression in the presence of TNF-α, and no response to IL-22 stimulation. Altogether, a role for IL-17C in activating Th17 cells combined with our findings of Th17-derived cytokine-driven synergy in the expression of IL-17C identifies a novel inflammatory amplification loop in the gastrointestinal tract between epithelial cells and Th17 cells.
Collapse
Affiliation(s)
- Stephanie M Swedik
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH
| | - Abson Madola
- Department of Biology, Case Western Reserve University, Cleveland, OH
| | - Michelle A Cruz
- Department of Pathology, Case Western Reserve University, Cleveland, OH
| | | | - Alan D Levine
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, OH;
- Department of Pathology, Case Western Reserve University, Cleveland, OH
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH
- Department of Medicine, Case Western Reserve University, Cleveland, OH; and
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
22
|
Howell SJ, Lee CA, Zapadka TE, Lindstrom SI, Taylor BE, Taylor ZRR, Barber KG, Taylor PR. Inhibition of CD40-TRAF6-dependent inflammatory activity halts the onset of diabetic retinopathy in streptozotocin-diabetic mice. Nutr Diabetes 2022; 12:46. [PMID: 36309487 PMCID: PMC9617859 DOI: 10.1038/s41387-022-00225-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 10/11/2022] [Accepted: 10/20/2022] [Indexed: 01/11/2023] Open
Abstract
Diabetes initiates inflammation that can impair the retinal vasculature, and lead to diabetic retinopathy; one of the leading causes of blindness. Inflammatory pathways have been examined as potential therapeutic targets for diabetic retinopathy, but there is still a need for early-stage treatments. We hypothesized that the CD40-TNF Receptor Associated Factor 6 (TRAF6) axis plays a pivotal role in the onset of diabetic retinopathy, and that the CD40-TRAF6 axis would be a prime therapeutic target for early-stage non-proliferative diabetic retinopathy. The CD40-TRAF6 complex can initiate NFκB activation, inflammation, and tissue damage. Further, CD40 and TRAF6 are constitutively expressed on Muller glia, and upregulated in the diabetic retina. Yet the role of the CD40-TRAF6 complex in the onset of diabetic retinopathy is still unclear. In the current study, we examined the CD40-TRAF6 axis in diabetic retinopathy using a small molecule inhibitor (SMI-6877002) on streptozotocin-induced diabetic mice. When CD40-TRAF6-dependent inflammation was inhibited, retinal vascular leakage and capillary degeneration was ameliorated in diabetic mice. Collectively, these data suggest that the CD40-TRAF6 axis plays a pivotal role in the onset of diabetic retinopathy, and could be a novel therapeutic target for early diabetic retinopathy.
Collapse
Affiliation(s)
- Scott J. Howell
- grid.67105.350000 0001 2164 3847Department of Ophthalmology and Visual Science Case Western Reserve University, School of Medicine, Cleveland, USA ,grid.410349.b0000 0004 5912 6484Louis Stokes Cleveland VA Medical Center, Cleveland, OH USA
| | - Chieh A. Lee
- grid.67105.350000 0001 2164 3847Department of Ophthalmology and Visual Science Case Western Reserve University, School of Medicine, Cleveland, USA
| | - Thomas E. Zapadka
- grid.67105.350000 0001 2164 3847Department of Ophthalmology and Visual Science Case Western Reserve University, School of Medicine, Cleveland, USA ,grid.410349.b0000 0004 5912 6484Louis Stokes Cleveland VA Medical Center, Cleveland, OH USA
| | - Sarah I. Lindstrom
- grid.67105.350000 0001 2164 3847Department of Ophthalmology and Visual Science Case Western Reserve University, School of Medicine, Cleveland, USA
| | - Brooklyn E. Taylor
- grid.67105.350000 0001 2164 3847Department of Ophthalmology and Visual Science Case Western Reserve University, School of Medicine, Cleveland, USA
| | - Zakary R. R. Taylor
- grid.67105.350000 0001 2164 3847Department of Ophthalmology and Visual Science Case Western Reserve University, School of Medicine, Cleveland, USA
| | - Katherine G. Barber
- grid.410349.b0000 0004 5912 6484Louis Stokes Cleveland VA Medical Center, Cleveland, OH USA
| | - Patricia R. Taylor
- grid.67105.350000 0001 2164 3847Department of Ophthalmology and Visual Science Case Western Reserve University, School of Medicine, Cleveland, USA ,grid.410349.b0000 0004 5912 6484Louis Stokes Cleveland VA Medical Center, Cleveland, OH USA ,grid.67105.350000 0001 2164 3847Present Address: Department of Ophthalmology, Case Western Reserve University, Institute of Pathology, 2085 Adelbert Rd., Room 101, Cleveland, OH USA
| |
Collapse
|
23
|
Neuroimmune Responses in a New Experimental Animal Model of Cerebral Aspergillosis. mBio 2022; 13:e0225422. [PMID: 36040029 PMCID: PMC9600342 DOI: 10.1128/mbio.02254-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Exposure of immunosuppressed individuals to the opportunistic fungal pathogen Aspergillus fumigatus may result in invasive pulmonary aspergillosis (IPA), which can lead to the development of cerebral aspergillosis (CA), a highly lethal infection localized in the central nervous system (CNS). There are no experimental models of CA that effectively mimic human disease, resulting in a considerable knowledge gap regarding mechanisms of neurological pathogenicity and neuroimmune responses during infection. In this report, immunosuppressed mice (via acute, high-dose corticosteroid administration) challenged with A. fumigatus resting conidia intranasally, followed a day later by a 70-fold lower inoculum of pre-swollen conidia intravenously (IN + IV + steroid), demonstrated increased weight loss, signs of severe clinical disease, increased fungal burden in the brain, and significant reduction in survival compared to immunosuppressed mice challenged intranasally only (IN + steroid) or non-immunosuppressed mice challenged both intranasally and intravenously (IN + IV). The IN + IV + steroid group demonstrated significant decreases in monocytes, eosinophils, dendritic cells (DCs), and invasive natural killer T (iNKT) cells, but not neutrophils or γδ T cells, in the brain compared to the IN + IV group. Likewise, the IN + IV + steroid group had significantly lower levels of interleukin (IL)-1β, IL-6, IL-17A, CC motif chemokine ligand 3 (CCL3), CXC chemokine ligand 10 (CXCL10), and vascular endothelial growth factor (VEGF) in the brain compared to the IN + IV group. IN + IV + steroid was superior to both IN + IV + chemotherapy (cytarabine + daunorubicin) and IN + IV + neutropenia for the development of CA. In conclusion, we have developed a well-defined, physiologically relevant model of disseminated CA in corticosteroid-induced immunosuppressed mice with a primary pulmonary infection. This model will serve to advance understanding of disease mechanisms, identify immunopathogenic processes, and help define the protective neuroinflammatory response to CA. IMPORTANCE Invasive fungal infections (IFIs) result in significant mortality in immunosuppressed individuals. Of these, invasive pulmonary aspergillosis (IPA), caused by the opportunistic mold Aspergillus fumigatus, is the most lethal. Lethality in IPA is due to two main factors: destruction of the lung leading to compromised pulmonary function, and dissemination of the organism to extrapulmonary organs. Of these, the CNS is the most common site of dissemination. However, very little is known regarding the pathogenesis of or immune response during cerebral aspergillosis, which is directly due to the lack of an animal model that incorporates immunosuppression, lung infection, and consistent dissemination to the CNS/brain. In this report, we have developed a new experimental animal model of CA which includes the above parameters and characterized the neuroimmune response. We further compared this disseminated CA model to two additional immunosuppressive strategies. Overall, this model of disseminated CA following IPA in an immunosuppressed host provides a novel platform for studying the efficacy of antifungal drugs and immunotherapies for improving disease outcomes.
Collapse
|
24
|
Evaluation of Cardiac Biomarkers and Expression Analysis of IL-1, IL-6, IL-10, IL-17, and IL-25 among COVID-19 Patients from Pakistan. Viruses 2022; 14:v14102149. [PMID: 36298704 PMCID: PMC9610190 DOI: 10.3390/v14102149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/24/2022] [Accepted: 09/25/2022] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 19 (COVID-19) is caused by viral infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Where upregulation of several important biomarkers and multiple organ dysfunction occurs, this study aimed to evaluate the association of cardiac biomarkers and CS induced acute lung damage with disease severity and mortality in survival of COVID-19 patients. A total of 500 COVID-19 patients with elevated cardiac biomarkers were studied for the analysis of myocardial abnormality through cardiac enzymes, inflammatory biomarkers, and the expression analysis of various cytokines, including IL-1, IL-6, IL-10, IL-17, and IL-25 genes. The elevation of various cardiac enzymes including LDH (87%), CK (78.4%), TNI (80.4%), CK-MB (83%), and D-dimer (80.8%) were found correlated (p < 0.001) with COVID-19 infection. Cardiac enzyme elevation was highly associated with an increased level of inflammatory biomarkers such as CRP (14.2%), SAA (11.4%) and erythrocyte sedimentation rate (ESR) (7.8%) (p = 0.001 for all). The quantitative expression analysis of IL-10, 1L-17, and 1L-25 were found to be high, while those of IL-1 and IL-6 were moderately elevated. The death-to-live ratio of COVID-19 patients was 457:43 indicating that the patients having elevated levels of both CKMB, D-dimer, CK and IL-1, IL-6, IL-10 and D-dimer, Troponin, CK and IL-1, IL-10 had high fatality rate (73% and 12% respectively). The current finding concludes that the evaluation of cardiac biomarkers with cytokine storm plays a significant role in COVID-19-associated anatomical organ damage, myocardial injury, and mortality. Physicians should pay special attention to cardiac biomarkers in patients with old age, inflammation, and comorbidities among COVID-19 infections.
Collapse
|
25
|
Malik J, Ahmed S, Yaseen Z, Alanazi M, Alharby TN, Alshammari HA, Anwar S. Association of SARS-CoV-2 and Polypharmacy with Gut-Lung Axis: From Pathogenesis to Treatment. ACS OMEGA 2022; 7:33651-33665. [PMID: 36164411 PMCID: PMC9491241 DOI: 10.1021/acsomega.2c02524] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/29/2022] [Indexed: 06/12/2023]
Abstract
SARS-CoV-2 is a novel infectious contagion leading to COVID-19 disease. The virus has affected the lives of millions of people across the globe with a high mortality rate. It predominantly affects the lung (respiratory system), but it also affects other organs, including the cardiovascular, psychological, and gastrointestinal (GIT) systems. Moreover, elderly and comorbid patients with compromised organ functioning and pre-existing polypharmacy have worsened COVID-19-associated complications. Microbiota (MB) of the lung plays an important role in developing COVID-19. The extent of damage mainly depends on the predominance of opportunistic pathogens and, inversely, with the predominance of advantageous commensals. Changes in the gut MB are associated with a bidirectional shift in the interaction among the gut with a number of vital human organs, which leads to severe disease symptoms. This review focuses on dysbiosis in the gut-lung axis, COVID-19-induced worsening of comorbidities, and the influence of polypharmacy on MB.
Collapse
Affiliation(s)
- Jonaid
Ahmad Malik
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research, Guwahati, Assam 781101, India
- Department
of Biomedical Engineering, Indian Institute
of Technology Rupnagar 140001, India
| | - Sakeel Ahmed
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat 382355, India
| | - Zahid Yaseen
- Department
of Pharmaceutical Biotechnology, Delhi Pharmaceutical
Sciences and Research University, New Delhi, Delhi 110017, India
| | - Muteb Alanazi
- Department
of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail 81422, Saudi Arabia
| | - Tareq Nafea Alharby
- Department
of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail 81422, Saudi Arabia
| | | | - Sirajudheen Anwar
- Department
of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 81422, Saudi Arabia
| |
Collapse
|
26
|
Ritzmann F, Lunding LP, Bals R, Wegmann M, Beisswenger C. IL-17 Cytokines and Chronic Lung Diseases. Cells 2022; 11:2132. [PMID: 35883573 PMCID: PMC9318387 DOI: 10.3390/cells11142132] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/27/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
IL-17 cytokines are expressed by numerous cells (e.g., gamma delta (γδ) T, innate lymphoid (ILC), Th17, epithelial cells). They contribute to the elimination of bacteria through the induction of cytokines and chemokines which mediate the recruitment of inflammatory cells to the site of infection. However, IL-17-driven inflammation also likely promotes the progression of chronic lung diseases, such as chronic obstructive pulmonary disease (COPD), lung cancer, cystic fibrosis, and asthma. In this review, we highlight the role of IL-17 cytokines in chronic lung diseases.
Collapse
Affiliation(s)
- Felix Ritzmann
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
- Helmholtz Institute for Pharmaceutical Research, 66123 Saarbrücken, Germany
| | - Lars Peter Lunding
- Division of Lung Immunology, Priority Area Asthma and Allergy, Research Center Borstel—Leibniz Lung Center, 23845 Borstel, Germany; (L.P.L.); (M.W.)
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Robert Bals
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
- Helmholtz Institute for Pharmaceutical Research, 66123 Saarbrücken, Germany
| | - Michael Wegmann
- Division of Lung Immunology, Priority Area Asthma and Allergy, Research Center Borstel—Leibniz Lung Center, 23845 Borstel, Germany; (L.P.L.); (M.W.)
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
| |
Collapse
|
27
|
Shi JL, Zheng ZM, Chen M, Shen HH, Li MQ, Shao J. IL-17: an important pathogenic factor in endometriosis. Int J Med Sci 2022; 19:769-778. [PMID: 35582411 PMCID: PMC9108413 DOI: 10.7150/ijms.71972] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 03/31/2022] [Indexed: 11/05/2022] Open
Abstract
Interleukin-17 (IL-17) is known as a Th17-cell-derived proinflammatory cytokine, which plays a pivotal role in several inflammatory and autoimmune diseases such as systemic lupus erythematosus (SLE), rheumatoid arthritis, and psoriasis. Emerging evidence has shown that IL-17 is linked to endometriosis, although the etiology of endometriosis is still unknown. The IL-17 expression is up-regulated in serum, peritoneal fluid (PF) and endometriotic lesions from patients with endometriosis but the related regulation mechanisms are complex and obscure. Meanwhile, the specific roles of IL-17 in endometriosis are also worthy of further exploration. Through the integration and summary of literature, we conclude that the secretion of IL-17 increases under the regulation of ectopic microenvironment and other factors, and then IL-17 is deeply involved in endometriosis in the regulation of immune microenvironment, the invasion and growth of ectopic lesions, and so on, which implies its therapeutic value in this disorder.
Collapse
Affiliation(s)
- Jia-Lu Shi
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Zi-Meng Zheng
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Min Chen
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Hui-Hui Shen
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
- NHC Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Jun Shao
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| |
Collapse
|
28
|
Azadeh H, Alizadeh-Navaei R, Rezaiemanesh A, Rajabinejad M. Immune-related adverse events (irAEs) in ankylosing spondylitis (AS) patients treated with interleukin (IL)-17 inhibitors: a systematic review and meta-analysis. Inflammopharmacology 2022; 30:435-451. [PMID: 35188599 DOI: 10.1007/s10787-022-00933-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/03/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Ankylosing spondylitis (AS) is a chronic inflammatory rheumatic disease characterized by immune system dysregulation and inflammation in the joints. Interleukin (IL)-17 inhibitors are new biological drugs used to treat AS. In this study, we aimed to assess the risk of immune system-related AEs due to targeting IL-17 or IL-17R. METHODS The CENTRAL, PubMed, Scopus, Google Scholar, Clinical Trials Registry, and ICTRP were searched for randomized clinical trials (RCTs) and non-RCTs until February 2021. The risk of irAEs in patients treated with IL-17 inhibitors compared to the placebo or a drug-free control was evaluated. In studies that reported AEs of the IL-17 inhibitors at several different time points, we compared the number of cases/100 patient-year in which irAEs were reported. Subgroup analyses were also performed based on the dose and type of drugs. RESULTS Thirteen studies of 1848 AS patients treated by IL-17 inhibitors (secukinumab, ixekizumab, bimekizumab, and netakimab) and 764 participants who received a placebo were included. The risk of some AEs related to immune function in patients under IL-17 inhibitors treatment was significantly higher than that of the placebo group, including infection and infestation (risk difference RD = 0.09, P = 0.02), nasopharyngitis (RD = 0.04, P < 0.001), opportunistic infections (RD = 0.01, P = 0.04), and neutropenia (RD = 0.04, P = 0.03). Besides, the results of the Cochran Q test showed that there were significant differences between the occurrence of some AEs over time, including infection and infestations (p < 0.001, RCTs), upper respiratory tract infections (p < 0.001, non-RCTs), urinary tract infections (p < 0.001, non-RCTs), and diarrhea (p < 0.01, RCTs). CONCLUSIONS The most common immune system-related AEs in patients treated with IL-17 inhibitors are mucosal and opportunistic infections.
Collapse
Affiliation(s)
- Hossein Azadeh
- Department of Internal Medicine, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Alizadeh-Navaei
- Gastrointestinal Cancer Research Center, Non-Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Rezaiemanesh
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Misagh Rajabinejad
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, 18 km khazarabad, Sari, Iran.
| |
Collapse
|
29
|
Nada S, Kahaleh B, Altorok N. Genome-wide DNA methylation pattern in systemic sclerosis microvascular endothelial cells: Identification of epigenetically affected key genes and pathways. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2022; 7:71-81. [PMID: 35386944 PMCID: PMC8922681 DOI: 10.1177/23971983211033772] [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: 03/22/2021] [Accepted: 06/13/2021] [Indexed: 02/03/2023]
Abstract
Background The etiology of systemic sclerosis is not clear, but there is evidence suggesting a critical role for epigenetic alterations in disease pathogenesis and clinical expression. We sought, in this study, to characterize the genome-wide DNA methylation signature in systemic sclerosis microvascular endothelial cells. Methods We performed a genome-wide DNA methylation study in microvascular endothelial cells derived from seven diffuse cutaneous systemic sclerosis patients compared to seven age-, sex-, and ethnicity-matched healthy controls. We paired matched samples on Illumina HumanMethylation450 (three diffuse cutaneous systemic sclerosis microvascular endothelial cells and three controls), and reproduced the results in an independent set of matched patient and controls using Illumina Infinium MethylationEPIC (four diffuse cutaneous systemic sclerosis patients and four controls) to identify differentially methylated genes. Results We identified 71,353 differentially methylated CpG sites in systemic sclerosis microvascular endothelial cells using Infinium MethylationEPIC microarray in the first group (0.081% of representative probes) and 33,170 CpG sites in the second group using HumanMethylation450 microarray (0.073% of representative probes) in diffuse cutaneous systemic sclerosis microvascular endothelial cells. Among the two groups of subjects, we identified differential methylation of 2455 CpG sites, representing 1301 genes. Most of the differentially methylated CpG sites were hypermethylated (1625 CpG), corresponding to 910 genes. Common hypermethylated genes in systemic sclerosis microvascular endothelial cells include NOS1, DNMT3A, DNMT3B, HDAC4, and ANGPT2. We also identified hypomethylation of IL17RA, CTNNA3, ICAM2, and SDK1 in systemic sclerosis microvascular endothelial cells. Furthermore, we demonstrate significant inverse correlation between DNA methylation status and gene expression in the majority of genes evaluated. Gene ontology analysis of hypermethylated genes demonstrated enrichment of genes involved in angiogenesis (p = 0.0006). Pathway analysis of hypomethylated genes includes genes involved in vascular smooth muscle contraction (p = 0.014) and adherens junctions (p = 0.013). Conclusion Our data suggest the presence of significant genome-wide DNA methylation aberrancies in systemic sclerosis microvascular endothelial cells, and identify novel affected genes and pathways in systemic sclerosis microvascular endothelial cells.
Collapse
Affiliation(s)
- Shadia Nada
- Division of Rheumatology, University of Toledo, Toledo, OH, USA,Department of Internal Medicine, University of Toledo, Toledo, OH, USA
| | - Bashar Kahaleh
- Division of Rheumatology, University of Toledo, Toledo, OH, USA,Department of Internal Medicine, University of Toledo, Toledo, OH, USA
| | - Nezam Altorok
- Division of Rheumatology, University of Toledo, Toledo, OH, USA,Department of Internal Medicine, University of Toledo, Toledo, OH, USA,Nezam Altorok, Department of Internal Medicine, University of Toledo, 3000 Arlington Avenue, Mailstop 1186, Toledo, OH 43614, USA.
| |
Collapse
|
30
|
Zhang N, Qian T, Sun S, Cao W, Wang Z, Liu D, Li P, Wu J, Li H, Yang J. IL-17 is a Potential Therapeutic Target in a Rodent Model of Otitis Media with Effusion. J Inflamm Res 2022; 15:635-648. [PMID: 35140496 PMCID: PMC8818970 DOI: 10.2147/jir.s338598] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/21/2022] [Indexed: 11/23/2022] Open
Abstract
Background Otitis media with effusion (OME) is a non-suppurative inflammation of the middle ear that is characterized by middle ear effusion and hearing loss. However, the mechanisms of OME are not fully understood. The aim of this study was to determine the function and the mechanism of the IL-17 cytokine in the pathogenesis of OME and to investigate IL-17 as a potential strategy for the treatment of OME. Methods In this study, the OME rat model was induced by ovalbumin (OVA) as previously described. The severity of OME was determined with an oto-endoscope, by histochemical analysis, and by acoustic immittance. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of RNA-sequencing (RNA-seq) data was carried out to analyze the signaling pathways related to the pathogenesis of OME, which indicated that IL-17 is involved in OME. The anti-IL-17A monoclonal antibody was administrated by nasal drip to block IL-17 to treat OME in the rat model. The rats were finally injected intraperitoneally with the inhibitor of Notch signaling pathway to study the mechanisms of IL-17-induced inflammation. Serum and lavage fluid were collected for the detection of related cytokines, and middle ear tissue was collected for Western blot, quantitative real-time PCR (qRT-PCR), and immunohistochemical and immunofluorescence analysis. Results KEGG analysis of RNA-seq data suggested that the IL-17 signaling pathway might be involved in the onset of OME. IL-17 expression was confirmed to be increased in both the serum and the middle ear of the rat model. The monoclonal antibody against IL-17 neutralized IL-17, inhibited the inflammation in the middle ear, and reduced the overall severity of OME in vivo. Furthermore, the Notch signaling pathway was activated upon IL-17 upregulation in OME and was suppressed by IL-17 blockage. However, there was no change in IL-17 expression after Notch inhibitor treatment, which reduced the severity of OME in the rat middle ear. Conclusion IL-17 plays a key role in the pathogenesis of the OVA-induced OME rat model. IL-17 induced inflammatory responses via the Notch signaling pathway and targeting IL-17 might be an effective approach for OME therapy.
Collapse
Affiliation(s)
- Nanfeng Zhang
- Department of ENT, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230031, People’s Republic of China
| | - Tingting Qian
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Shan Sun
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Wei Cao
- Department of ENT, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230031, People’s Republic of China
| | - Zhixian Wang
- Department of ENT, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230031, People’s Republic of China
| | - Danling Liu
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Peifan Li
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Jingfang Wu
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Huawei Li
- Department of ENT, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230031, People’s Republic of China
- Department of ENT Institute and Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, NHC Key Laboratory of Hearing Medicine Research, Fudan University, Shanghai, 200032, People’s Republic of China
| | - Jianming Yang
- Department of ENT, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230031, People’s Republic of China
- Correspondence: Jianming Yang; Huawei Li, Email ;
| |
Collapse
|
31
|
Bae JS, Oh SB, Kim J, Kim H, Kim JH, Kim EH, Cho KJ, Mo JH. Particulate Matter Exposure Aggravates IL-17-Induced Eye and Nose Inflammation in an OVA/Poly(I:C) Mouse Model. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2022; 14:59-72. [PMID: 34983107 PMCID: PMC8724832 DOI: 10.4168/aair.2022.14.1.59] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/23/2021] [Accepted: 10/12/2021] [Indexed: 11/20/2022]
Abstract
PURPOSE Data on the effects of direct particulate matter (PM) exposure on the eyes and the nose are limited. Here, an interleukin (IL)-17/neutrophil-dominant ovalbumin (OVA)/polyinosinic-polycytidylic acid (Poly(I:C)) mouse model was used to evaluate the effect of different-sized titanium dioxide (TiO2) particles on the eyes and the nose. We also examined whether IL-17-neutralizing antibody (IL-17Ab) treatment could reverse TiO2 effects. METHODS The nasal cavities and conjunctival sacs of each mouse were challenged with OVA and Poly(I:C) to induce neutrophil-dominant inflammation and then exposed to micro- and nano-TiO2. Subsequently, IL-17Ab was administered to investigate the role of IL-17 and inflammatory parameters. RESULTS Micro- and nano-TiO2 resulted in significant decreases in tear-break-up time and increases in corneal damage. Airborne micro-TiO2 also increased nasal rubbing and sneezing counts compared with the OVA/Poly(I:C). Micro-TiO2 exposure increased infiltration of neutrophils and IL-17A+ cells in the conjunctival tissues and the nasal mucosae. In addition, these increased symptoms and inflammation in the eyes and the nose by micro-TiO2 exposure were inhibited by the IL-17Ab, suggesting IL-17 dependency. CONCLUSIONS TiO2 aggravated IL-17-induced eye and nose inflammation and the IL-17Ab alleviated inflammation in the OVA/Poly(I:C) mouse model. These results may help develop a therapeutic modality for PM exposure and provide evidence for PM-associated diseases.
Collapse
Affiliation(s)
- Jun-Sang Bae
- Department of Otorhinolaryngology, College of Medicine, Dankook University, Cheonan, Korea.,Beckman Laser Institute Korea, Dankook University, Cheonan, Korea.,Medical Laser Research Center, Dankook University, Cheonan, Korea
| | - Soo Bin Oh
- Department of Ophthalmology, College of Medicine, Dankook University, Cheonan, Korea.,Catholic Institute for Visual Science, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jeongyun Kim
- Department of Physics, College of Natural Sciences, Dankook University, Cheonan, Korea
| | - Hoon Kim
- Department of Ophthalmology, College of Medicine, Dankook University, Cheonan, Korea
| | - Ji Hye Kim
- Department of Otorhinolaryngology, College of Medicine, Dankook University, Cheonan, Korea.,Beckman Laser Institute Korea, Dankook University, Cheonan, Korea
| | - Eun-Hee Kim
- Department of Otorhinolaryngology, College of Medicine, Dankook University, Cheonan, Korea.,Beckman Laser Institute Korea, Dankook University, Cheonan, Korea
| | - Kyong Jin Cho
- Department of Ophthalmology, College of Medicine, Dankook University, Cheonan, Korea.
| | - Ji-Hun Mo
- Department of Otorhinolaryngology, College of Medicine, Dankook University, Cheonan, Korea.,Beckman Laser Institute Korea, Dankook University, Cheonan, Korea.,Medical Laser Research Center, Dankook University, Cheonan, Korea.
| |
Collapse
|
32
|
Pu X, Li F, Lin X, Wang R, Chen Z. Oxidative stress and expression of inflammatory factors in lung tissue of acute mountain sickness rats. Mol Med Rep 2021; 25:49. [PMID: 34913080 PMCID: PMC8711020 DOI: 10.3892/mmr.2021.12565] [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: 10/16/2019] [Accepted: 01/27/2021] [Indexed: 11/12/2022] Open
Abstract
The aim of the present study was to investigate the changes in lung histomorphology and oxidative stress, as well as the expression of interleukin (IL)-17C and other inflammatory factors during acute mountain sickness (AMS) in male Sprague-Dawley rats and to explore the underlying mechanism. Rats were randomly divided into a control group (0 h) and three hypoxia stress groups, exposed to low-pressure oxygen storage at a simulated altitude of 6,000 m for 24, 48 and 72 h, respectively. Morphological changes in lung tissue were observed by hematoxylin and eosin staining under light microscopy and transmission electron microscopy. The expression of inflammatory factors IL-17C, nuclear factor-κB (NF-κB), IL-1β, IL-6 and tumor necrosis factor-α (TNF-α) in lung tissue was assessed by RNA sequencing and verified by reverse transcription-quantitative PCR (RT-qPCR) and western blotting (WB). Superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) enzyme activity and malondialdehyde (MDA) expression were also measured. Experimental groups were compared to the control group following 24, 48 and 72 h of hypoxic stress. Lung tissue suffered from different degrees of injury, and the damage was the most severe after 48 h of hypoxic stress. RNA sequencing data from the lung tissue of rats from each group suggested that the expression of IL-17C, NF-κB, IL-1β, IL-6, and TNF-α increased significantly after hypoxic stress. RT-qPCR and WB demonstrated that the expression of IL-17C and NF-κB increased significantly after hypoxia lasting 48 and 72 h. IL-1β expression increased significantly after hypoxia stress lasting 24 and 48 h, and the expressions of TNF-α and IL-6 increased significantly after hypoxia stress lasting 24, 48 and 72 h (P<0.01). The enzyme activity of SOD and GSH-Px decreased significantly after lasting 24, 48 and 72 h of hypoxia (P<0.01), and MDA increased significantly after hypoxic stress lasting 48 and 72 h (P<0.01). In conclusion, under hypoxic stress, rats quickly initiate oxidative stress and immune responses. However, with prolonged hypoxic stress time, excessive oxidative stress can further stimulate the immune system in vivo, and release a large quantity of inflammatory factors accumulating in the body. This, in turn, may lead to the occurrence of inflammatory storms and further damage the lung tissue resulting in AMS.
Collapse
Affiliation(s)
- Xiaoyan Pu
- Qinghai Normal University, Xining, Qinghai 810001, P.R. China
| | - Fuxin Li
- College of Medicine, Qinghai University, Xining, Qinghai 810001, P.R. China
| | - Xue Lin
- College of Medicine, Qinghai University, Xining, Qinghai 810001, P.R. China
| | - Rong Wang
- College of Medicine, Qinghai University, Xining, Qinghai 810001, P.R. China
| | - Zhi Chen
- Qinghai Normal University, Xining, Qinghai 810001, P.R. China
| |
Collapse
|
33
|
Chung SH, Ye XQ, Iwakura Y. Interleukin-17 family members in health and disease. Int Immunol 2021; 33:723-729. [PMID: 34611705 PMCID: PMC8633656 DOI: 10.1093/intimm/dxab075] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/25/2021] [Indexed: 12/15/2022] Open
Abstract
The interleukin-17 (IL-17) family consists of six family members (IL-17A-IL-17F) and all the corresponding receptors have been identified recently. This family is mainly involved in the host defense mechanisms against bacteria, fungi and helminth infection by inducing cytokines and chemokines, recruiting neutrophils, inducing anti-microbial proteins and modifying T-helper cell differentiation. IL-17A and some other family cytokines are also involved in the development of psoriasis, psoriatic arthritis and ankylosing spondylitis by inducing inflammatory cytokines and chemokines, and antibodies against IL-17A as well as the receptor IL-17RA are being successfully used for the treatment of these diseases. Involvement in the development of inflammatory bowel disease, multiple sclerosis, rheumatoid arthritis and tumors has also been suggested in animal disease models. In this review, we will briefly review the mechanisms by which IL-17 cytokines are involved in the development of these diseases and discuss possible treatment of inflammatory diseases by targeting IL-17 family members.
Collapse
Affiliation(s)
- Soo-Hyun Chung
- Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba, Japan
| | - Xiao-Qi Ye
- Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba, Japan
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba, Japan
| |
Collapse
|
34
|
Kathamuthu GR, Kumar NP, Moideen K, Menon PA, Babu S. Decreased Frequencies of Gamma/Delta T Cells Expressing Th1/Th17 Cytokine, Cytotoxic, and Immune Markers in Latent Tuberculosis-Diabetes/Pre-Diabetes Comorbidity. Front Cell Infect Microbiol 2021; 11:756854. [PMID: 34765568 PMCID: PMC8577793 DOI: 10.3389/fcimb.2021.756854] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/01/2021] [Indexed: 11/13/2022] Open
Abstract
Antigen-specific gamma-delta (γδ) T cells are important in exhibiting anti-mycobacterial immunity, but their role in latent tuberculosis (LTB) with diabetes mellitus (DM) or pre-DM (PDM) and non-DM comorbidities have not been studied. Thus, we have studied the baseline, mycobacterial (PPD, WCL), and positive control antigen-stimulated γδ T cells expressing Th1 (IFNγ, TNFα, IL-2) and Th17 (IL-17A, IL-17F, IL-22) cytokine as well as cytotoxic (perforin [PFN], granzyme [GZE B], granulysin [GNLSN]) and immune (GMCSF, PD-1, CD69) markers in LTB (DM, PDM, NDM) comorbidities by flow cytometry. In the unstimulated (UNS) condition, we did not observe any significant difference in the frequencies of γδ T cells expressing Th1 and Th17 cytokine, cytotoxic, and immune markers. In contrast, upon PPD antigen stimulation, the frequencies of γδ T cells expressing Th1 (IFNγ, TNFα) and Th17 (IL-17F, IL-22) cytokine, cytotoxic (PFN, GZE B, GNLSN), and immune (CD69) markers were significantly diminished in LTB DM and/or PDM individuals compared to LTB NDM individuals. Similarly, upon WCL antigen stimulation, the frequencies of γδ T cells expressing Th1 (TNFα) and Th17 (IL-17A, IL-22) cytokine, cytotoxic (PFN), and immune (PD-1, CD69) markers were significantly diminished in LTB DM and/or PDM individuals compared to LTB NDM individuals. Finally, upon P/I stimulation we did not observe any significant difference in the γδ T cell frequencies expressing cytokine, cytotoxic, and immune markers between the study populations. The culture supernatant levels of IFNγ, TNFα, and IL-17A cytokines were significantly increased in LTB DM and PDM after stimulation with Mtb antigens compared to LTB NDM individuals. Therefore, diminished γδ T cells expressing cytokine, cytotoxic, and other immune markers and elevated levels of cytokines in the supernatants is a characteristic feature of LTB PDM/DM co-morbidities.
Collapse
Affiliation(s)
- Gokul Raj Kathamuthu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India.,Indian Council of Medical Research-National Institute for Research in Tuberculosis (ICMR-NIRT), Chennai, India
| | - Nathella Pavan Kumar
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India.,Indian Council of Medical Research-National Institute for Research in Tuberculosis (ICMR-NIRT), Chennai, India
| | - Kadar Moideen
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India
| | - Pradeep A Menon
- Indian Council of Medical Research-National Institute for Research in Tuberculosis (ICMR-NIRT), Chennai, India
| | - Subash Babu
- National Institutes of Health-NIRT-International Center for Excellence in Research, Chennai, India.,Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
35
|
Sinha P, Dash M, Bhatkoti B, Krishnan L. Epithelial herpes simplex keratitis in a patient on treatment with secukinumab for psoriasis: An effect of interleukin-17 blockade? Indian J Dermatol Venereol Leprol 2021; 88:225-227. [PMID: 34877851 DOI: 10.25259/ijdvl_496_2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 08/01/2021] [Indexed: 01/18/2023]
Affiliation(s)
- Preema Sinha
- Department of Dermatology, Base Hospital, Lucknow, Uttar Pradesh, India
| | - Mahashweta Dash
- Department of Dermatology, Base Hospital, Lucknow, Uttar Pradesh, India
| | - Bhupesh Bhatkoti
- Department of Ophthalmology, Command Hospital, Lucknow, Uttar Pradesh, India
| | | |
Collapse
|
36
|
Gabriel EM, Wiche Salinas TR, Gosselin A, Larouche-Anctil E, Durand M, Landay AL, El-Far M, Tremblay CL, Routy JP, Ancuta P. Overt IL-32 isoform expression at intestinal level during HIV-1 infection is negatively regulated by IL-17A. AIDS 2021; 35:1881-1894. [PMID: 34101628 PMCID: PMC8416712 DOI: 10.1097/qad.0000000000002972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Untreated HIV infection was previously associated with IL-32 overexpression in gut/intestinal epithelial cells (IEC). Here, we explored IL-32 isoform expression in the colon of people with HIV (PWH) receiving antiretroviral therapy (ART) and IL-32 triggers/modulators in IEC. DESIGN Sigmoid colon biopsies (SCB) and blood were collected from ART-treated PWH (HIV + ART; n = 17; mean age: 56 years; CD4+ cell counts: 679 cells/μl; time on ART: 72 months) and age-matched HIV-uninfected controls (HIVneg; n = 5). The IEC line HT-29 was used for mechanistic studies. METHODS Cells from SCB and blood were isolated by enzymatic digestion and/or gradient centrifugation. HT-29 cells were exposed to TLR1-9 agonists, TNF-α, IL-17A and HIV. IL-32α/β/γ/D/ε/θ and IL-17A mRNA levels were quantified by real-time RT-PCR. IL-32 protein levels were quantified by ELISA. RESULTS IL-32β/γ/ε isoform transcripts were detectable in the blood and SCB, with IL-32β mRNA levels being predominantly expressed in both compartments and at significantly higher levels in HIV + ART compared to HIVneg. IL-17A transcripts were only detectable in SCB, with increased IL-17A levels in HIVneg compared with HIV + ART and negatively correlated with IL-32β mRNA levels. IL-32β/γ/ε isoform mRNA were detected in HT-29 cells upon exposure to TNF-α, Poly I:C (TLR3 agonist), Flagellin (TLR-5 agonist) and HIV. IL-17A significantly decreased both IL-32 β/γ/ε mRNA and cell-associated IL-32 protein levels induced upon TNF-α and Poly I:C triggering. CONCLUSION We document IL-32 isoforms abundant in the colon of ART-treated PWH and reveal the capacity of the Th17 hallmark cytokine IL-17A to attenuate IL-32 overexpression in a model of inflamed IEC.
Collapse
Affiliation(s)
- Etiene Moreira Gabriel
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Tomas Raul Wiche Salinas
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | | | | | - Madeleine Durand
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | | | | | - Cécile L. Tremblay
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Pierre Routy
- McGill University Health Centre, Montreal, Québec, Canada
- Chronic Viral Illness Service and Hematology Department, McGill University Health Centre, Montréal, Québec, Canada
| | - Petronela Ancuta
- CHUM Research Centre, Montréal, Québec, Canada
- Department de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
37
|
Pan N, Liu B, Bao X, Zhang H, Sheng S, Liang Y, Pan H, Wang X. Oral Delivery of Novel Recombinant Lactobacillus Elicit High Protection against Staphylococcus aureus Pulmonary and Skin Infections. Vaccines (Basel) 2021; 9:vaccines9090984. [PMID: 34579221 PMCID: PMC8473125 DOI: 10.3390/vaccines9090984] [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: 08/06/2021] [Revised: 08/26/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
Staphylococcus aureus is a leading cause of nosocomial and community-associated infection worldwide; however, there is no licensed vaccine available. S. aureus initiates infection via the mucosa; therefore, a mucosal vaccine is likely to be a promising approach against S. aureus infection. Lactobacilli, a non-pathogenic bacterium, has gained increasing interest as a mucosal delivery vehicle. Hence, we attempted to develop an oral S. aureus vaccine based on lactobacilli to cushion the stress of drug resistance and vaccine needs. In this study, we designed, constructed, and evaluated recombinant Lactobacillus strains synthesizing S. aureus nontoxic mutated α-hemolysins (HlaH35L). The results from animal clinical trials showed that recombinant Lactobacillus can persist for at least 72 h and can stably express heterologous protein in vivo. Recombinant L. plantarum WXD234 (pNZ8148-Hla) could induce robust mucosal immunity in the GALT, as evidenced by a significant increase in IgA and IL-17 production and the strong proliferation of T-lymphocytes derived from Peyer’s patches. WXD234 (pNZ8148-Hla) conferred up to 83% protection against S. aureus pulmonary infection and significantly reduced the abscess size in a S. aureus skin infection model. Of particular interest is the sharp reduction of the protective effect offered by WXD234 (pNZ8148-Hla) vaccination in γδ T cell-deficient or IL-17-deficient mice. In conclusion, for the first time, genetically engineered Lactobacillus WXD234 (pNZ8148-Hla) as an oral vaccine induced superior mucosal immunity, which was associated with high protection against pulmonary and skin infections caused by S. aureus. Taken together, our findings suggest the great potential for a delivery system based on lactobacilli and provide experimental data for the development of mucosal vaccines for S. aureus.
Collapse
Affiliation(s)
- Na Pan
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010070, China; (N.P.); (B.L.); (X.B.); (H.Z.); (S.S.); (Y.L.); (H.P.)
| | - Bohui Liu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010070, China; (N.P.); (B.L.); (X.B.); (H.Z.); (S.S.); (Y.L.); (H.P.)
| | - Xuemei Bao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010070, China; (N.P.); (B.L.); (X.B.); (H.Z.); (S.S.); (Y.L.); (H.P.)
| | - Haochi Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010070, China; (N.P.); (B.L.); (X.B.); (H.Z.); (S.S.); (Y.L.); (H.P.)
| | - Shouxin Sheng
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010070, China; (N.P.); (B.L.); (X.B.); (H.Z.); (S.S.); (Y.L.); (H.P.)
| | - Yanchen Liang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010070, China; (N.P.); (B.L.); (X.B.); (H.Z.); (S.S.); (Y.L.); (H.P.)
| | - Haiting Pan
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010070, China; (N.P.); (B.L.); (X.B.); (H.Z.); (S.S.); (Y.L.); (H.P.)
- Basic Medical College, Inner Mongolia Medical University, Hohhot 010110, China
| | - Xiao Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010070, China; (N.P.); (B.L.); (X.B.); (H.Z.); (S.S.); (Y.L.); (H.P.)
- Correspondence:
| |
Collapse
|
38
|
Wu Q, Cui D, Chao X, Chen P, Liu J, Wang Y, Su T, Li M, Xu R, Zhu Y, Zhang Y. Transcriptome Analysis Identifies Strategies Targeting Immune Response-Related Pathways to Control Enterotoxigenic Escherichia coli Infection in Porcine Intestinal Epithelial Cells. Front Vet Sci 2021; 8:677897. [PMID: 34447800 PMCID: PMC8383179 DOI: 10.3389/fvets.2021.677897] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/05/2021] [Indexed: 11/13/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is an important cause of post-weaning diarrhea (PWD) worldwide, resulting in huge economic losses to the swine industry worldwide. In this study, to understand the pathogenesis, the transcriptomic analysis was performed to explore the biological processes (BP) in porcine intestinal epithelial J2 cells infected with an emerging ETEC strain isolated from weaned pigs with diarrhea. Under the criteria of |fold change| (FC) ≥ 2 and P < 0.05 with false discovery rate < 0.05, a total of 131 referenced and 19 novel differentially expressed genes (DEGs) were identified after ETEC infection, including 96 upregulated DEGs and 54 downregulated DEGs. The Gene Ontology (GO) analysis of DEGs showed that ETEC evoked BP specifically involved in response to lipopolysaccharide (LPS) and negative regulation of intracellular signal transduction. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that immune response-related pathways were mainly enriched in J2 cells after ETEC infection, in which tumor necrosis factor (TNF), interleukin 17, and mitogen-activated protein kinase (MAPK) signaling pathways possessed the highest rich factor, followed by nucleotide-binding and oligomerization domain-like receptor (NLRs), C-type lectin receptor (CLR), cytokine–cytokine receptor interaction, and Toll-like receptor (TLR), and nuclear factor kappa-B (NF-κB) signaling pathways. Furthermore, 30 of 131 referenced DEGs, especially the nuclear transcription factor AP-1 and NF-κB, participate in the immune response to infection through an integral signal cascade and can be target molecules for prevention and control of enteric ETEC infection by probiotic Lactobacillus reuteri. Our data provide a comprehensive insight into the immune response of porcine intestinal epithelial cells (IECs) to ETEC infection and advance the identification of targets for prevention and control of ETEC-related PWD.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China.,Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Defeng Cui
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China.,Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Xinyu Chao
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Peng Chen
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Jiaxuan Liu
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Yiding Wang
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Tongjian Su
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Meng Li
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Ruyu Xu
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Yaohong Zhu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yonghong Zhang
- Department of Animal Medicine, College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China.,Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| |
Collapse
|
39
|
Gallbladder Interleukins in Children with Calculous Cholecystitis. Pediatr Rep 2021; 13:470-482. [PMID: 34449702 PMCID: PMC8396171 DOI: 10.3390/pediatric13030054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/06/2021] [Accepted: 08/02/2021] [Indexed: 11/16/2022] Open
Abstract
Calculous cholecystitis connects to inflammation and various complications. It is a common disease in the paediatric population, yet it is still uncertain how inflammation factors are involved in its morphopathogenesis. Twenty calculous cholecystitis surgery tissue samples were obtained from 20 children. As a control, seven unaffected gallbladders were used. Tissues were immunohistochemically stained for IL-1α, IL-4, IL-6, IL-7, IL-8, IL-10, and IL-17A, and the slides were inspected by light microscopy. To evaluate statistical differences and correlations between interleukins, Mann-Whitney U and Spearman's tests were used. Statistically significant difference between patient and control gallbladder epithelium was for IL-1α and IL-17A, but connective tissue-IL-1α, IL-4, IL-6, IL-7, IL-8, and IL-17A positive structures. A strong positive correlation in patients was detected between epithelial IL-1α and IL-1α in connective tissue, epithelial IL-6 and IL-7, IL-6 and IL-17A, IL-7 and IL-10, IL-7 and IL-17A, as well as between IL-6 and IL-7, IL-7 and IL-10 in connective tissue. The increase of IL-1α, IL-4, IL-6, IL-7, IL-8 and IL-17A positive structures suggests their role in the morphopathogenesis of calculous cholecystitis. The correlations between interleukins in epithelium and in connective tissues prove that the epithelial barrier function and inflammatory response in deeper layers are sustained through intercellular signalling pathways.
Collapse
|
40
|
Swedik S, Madola A, Levine A. IL-17C in human mucosal immunity: More than just a middle child. Cytokine 2021; 146:155641. [PMID: 34293699 DOI: 10.1016/j.cyto.2021.155641] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023]
Abstract
Interleukin-17C (IL-17C) is an understudied member of the IL-17 family of cytokines. Its synthesis is induced by both cytokines and pathogenic stimuli in a variety of cell types, most often expressed at mucosal and barrier surfaces. IL-17C expression is dysregulated in a variety of autoinflammatory and autoimmune diseases including inflammatory bowel disease, psoriasis, and atopic dermatitis, yet it is protective against bacterial infections of the gut, skin, and lungs. In this review we highlight studies on IL-17C regulation and its function at human mucosal surfaces. Understanding the relationship between IL-17C and autoinflammatory and autoimmune diseases of the mucosa and defining the beneficial and pathogenic functions of the cytokine in inflammatory responses are the first steps in determining the potential for IL-17C as a therapeutic target.
Collapse
Affiliation(s)
- Stephanie Swedik
- Department of Molecular Biology and Microbiology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, United States
| | - Abson Madola
- Department of Biology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, United States
| | - Alan Levine
- Department of Molecular Biology and Microbiology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, United States; Departments of Pathology, Pharmacology, Medicine, and Pediatrics, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, United States.
| |
Collapse
|
41
|
Lv Y, Zhang J, Wang C. Self-assembled chitosan nanoparticles for intranasal delivery of recombinant protein interleukin-17 receptor C (IL-17RC): preparation and evaluation in asthma mice. Bioengineered 2021; 12:3029-3039. [PMID: 34180764 PMCID: PMC8806589 DOI: 10.1080/21655979.2021.1940622] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Asthma is mentioned as a chronic airway inflammatory disease, whose pathogenesis is complicated. The promotion of inflammation in asthma by IL-17A and IL-17F has been confirmed. In addition to covalent homodimers, both cytokines are also able to form heterodimers, further inducing downstream pathways via binding to the IL-17RA and IL-17RC receptor complex. In recent years, IL-17RA and its signal transduction pathway have been extensively researched. IL-17RC, however, remains relatively unexplored. In the present study, we self-assembled chitosan (CS) nanoparticles for intranasal delivery of recombinant protein IL-17RC (rIL-17RC) and preliminarily investigated its effect on a murine model of allergic asthma induced by ovalbumin (OVA). rIL-17RC was produced by the prokaryotic expression system and encapsulated into the CS nanoparticles via ionic cross-linking technique. The results showed that CS-RC nanoparticles via intranasal intervention significantly caused inhibition of mucus secretion and airway inflammatory cell infiltration, and reduced IL-4, IL-17, IL-17F levels in BALF. Hence, delivering receptor proteins such as IL-17RC, through CS nanoparticles as a carrier, could be an attractive therapeutic intervention for asthma.
Collapse
Affiliation(s)
- Yongli Lv
- Department of Paediatrics, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School Of Medicine, Shanghai, China
| | - Jianhua Zhang
- Department of Paediatrics, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School Of Medicine, Shanghai, China
| | - Chaoying Wang
- Department of Paediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
42
|
Yun Y, Baek A, Kim DE. Autophagy down-regulates NLRP3-dependent inflammatory response of intestinal epithelial cells under nutrient deprivation. BMB Rep 2021. [PMID: 33407996 PMCID: PMC8167250 DOI: 10.5483/bmbrep.2021.54.5.211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dysregulation of inflammation induced by noninfectious stress conditions, such as nutrient deprivation, causes tissue damage and intestinal permeability, resulting in the development of inflammatory bowel diseases. We studied the effect of autophagy on cytokine secretion related to intestinal permeability under nutrient deprivation. Autophagy removes NLRP3 inflammasomes via ubiquitin-mediated degradation under starvation. When autophagy was inhibited, starvation-induced NLRP3 inflammasomes and their product, IL-1β, were significantly enhanced. A prolonged nutrient deprivation resulted in an increased epithelial mesenchymal transition (EMT), leading to intestinal permeability. Under nutrient deprivation, IL-17E/25, which is secreted by IL-1β, demolished the intestinal epithelial barrier. Our results suggest that an upregulation of autophagy maintains the intestinal barrier by suppressing the activation of NLRP3 inflammasomes and the release of their products, including proinflammatory cytokines IL-1β and IL-17E/25, under nutrient deprivation.
Collapse
Affiliation(s)
- Yewon Yun
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Ahruem Baek
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
43
|
Munoz K, Wasnik S, Abdipour A, Bi H, Wilson SM, Tang X, Ghahramanpouri M, Baylink DJ. The Effects of Insulin-Like Growth Factor I and BTP-2 on Acute Lung Injury. Int J Mol Sci 2021; 22:ijms22105244. [PMID: 34063554 PMCID: PMC8170877 DOI: 10.3390/ijms22105244] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/06/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
Acute lung injury (ALI) afflicts approximately 200,000 patients annually and has a 40% mortality rate. The COVID-19 pandemic has massively increased the rate of ALI incidence. The pathogenesis of ALI involves tissue damage from invading microbes and, in severe cases, the overexpression of inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). This study aimed to develop a therapy to normalize the excess production of inflammatory cytokines and promote tissue repair in the lipopolysaccharide (LPS)-induced ALI. Based on our previous studies, we tested the insulin-like growth factor I (IGF-I) and BTP-2 therapies. IGF-I was selected, because we and others have shown that elevated inflammatory cytokines suppress the expression of growth hormone receptors in the liver, leading to a decrease in the circulating IGF-I. IGF-I is a growth factor that increases vascular protection, enhances tissue repair, and decreases pro-inflammatory cytokines. It is also required to produce anti-inflammatory 1,25-dihydroxyvitamin D. BTP-2, an inhibitor of cytosolic calcium, was used to suppress the LPS-induced increase in cytosolic calcium, which otherwise leads to an increase in proinflammatory cytokines. We showed that LPS increased the expression of the primary inflammatory mediators such as toll like receptor-4 (TLR-4), IL-1β, interleukin-17 (IL-17), TNF-α, and interferon-γ (IFN-γ), which were normalized by the IGF-I + BTP-2 dual therapy in the lungs, along with improved vascular gene expression markers. The histologic lung injury score was markedly elevated by LPS and reduced to normal by the combination therapy. In conclusion, the LPS-induced increases in inflammatory cytokines, vascular injuries, and lung injuries were all improved by IGF-I + BTP-2 combination therapy.
Collapse
Affiliation(s)
- Kevin Munoz
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
| | - Samiksha Wasnik
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
| | - Amir Abdipour
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
- Division of Nephrology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | - Hongzheng Bi
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China;
| | - Sean M. Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA;
| | - Xiaolei Tang
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
- Department of Veterinary Biomedical Sciences, College of Veterinary Medicine, Long Island University, Brookville, NY 11548, USA
| | - Mahdis Ghahramanpouri
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
| | - David J. Baylink
- Department of Medicine, Division of Regenerative Medicine, Loma Linda University, Loma Linda, CA 92354, USA; (K.M.); (S.W.); (A.A.); (X.T.); (M.G.)
- Correspondence: ; Tel.: +909-558-4000-49796; Fax: +(909)-558-0428
| |
Collapse
|
44
|
Shiomitsu S, Gillen J, Frasca S, Santoro D. Evaluation of the cutaneous expression of IL-17, IL-22, IL-31, and their receptors in canine atopic dermatitis. Res Vet Sci 2021; 136:74-80. [PMID: 33588097 DOI: 10.1016/j.rvsc.2020.12.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/08/2020] [Accepted: 12/22/2020] [Indexed: 11/29/2022]
Abstract
Interleukins (IL)-17, IL-22, and IL-31 play roles in human atopic dermatitis (AD), but scant information is available on canine AD. Histopathological assessment for interleukin expression is a challenge due to a lack of canine specific antibodies. To evaluate the mRNA and protein expression of IL-17 and IL-22, and mRNA expression of IL-31 and their receptors in the skin of healthy and atopic dogs, seventeen atopic (10 with and 7 without an active infection) and 13 healthy privately owned dogs were sampled. RNAscope® In situ hybridization (ISH) for IL-17, IL-22, IL-31, and their receptors was performed on archived canine skin samples. Simultaneously, indirect immunofluorescence (IIF) was performed for IL-17 and IL-22. RNAscope® ISH probes were validated by RT-PCR and RNAscope® ISH on cytospin preparations of peripheral blood mononuclear cells from atopic dogs. IL-17, IL-22, IL-31, and their receptors were successfully detected by RNAscope® ISH and by IIF (IL-17 and IL-22) in both atopic and healthy canine skin. There was no significant difference in the expression of interleukins and their receptors between healthy and atopic skin with or without active infection. Data from both methodologies were similar. The role and the relationship among those proteins in atopic skin is unclear from this study results. Data from IIF and ISH were overlapping and support each other. Fresh skin samples taken at different times during the development of atopic dermatitis might better assess the role that interleukins and their receptors play in AD.
Collapse
Affiliation(s)
- Sayaka Shiomitsu
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610, USA
| | - James Gillen
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Salvatore Frasca
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Domenico Santoro
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
45
|
Azevedo MLV, Zanchettin AC, Vaz de Paula CB, Motta Júnior JDS, Malaquias MAS, Raboni SM, Neto PC, Zeni RC, Prokopenko A, Borges NH, Godoy TM, Benevides APK, de Souza DG, Baena CP, Machado-Souza C, de Noronha L. Lung Neutrophilic Recruitment and IL-8/IL-17A Tissue Expression in COVID-19. Front Immunol 2021; 12:656350. [PMID: 33868301 PMCID: PMC8044579 DOI: 10.3389/fimmu.2021.656350] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
The new SARS-CoV-2 virus differs from the pandemic Influenza A virus H1N1 subtype (H1N1pmd09) how it induces a pro-inflammatory response in infected patients. This study aims to evaluate the involvement of SNPs and tissue expression of IL-17A and the neutrophils recruitment in post-mortem lung samples from patients who died of severe forms of COVID-19 comparing to those who died by H1N1pdm09. Twenty lung samples from patients SARS-CoV-2 infected (COVID-19 group) and 10 lung samples from adults who died from a severe respiratory H1N1pdm09 infection (H1N1 group) were tested. The tissue expression of IL-8/IL-17A was identified by immunohistochemistry, and hematoxylin and eosin (H&E) stain slides were used for neutrophil scoring. DNA was extracted from paraffin blocks, and genotyping was done in real time-PCR for two IL17A target polymorphisms. Tissue expression increasing of IL-8/IL-17A and a higher number of neutrophils were identified in samples from the H1N1 group compared to the COVID-19 group. The distribution of genotype frequencies in the IL17A gene was not statistically significant between groups. However, the G allele (GG and GA) of rs3819025 was correlated with higher tissue expression of IL-17A in the COVID-19 group. SARS-CoV-2 virus evokes an exacerbated response of the host’s immune system but differs from that observed in the H1N1pdm09 infection since the IL-8/IL-17A tissue expression, and lung neutrophilic recruitment may be decreased. In SNP rs3819025 (G/A), the G allele may be considered a risk allele in the patients who died for COVID-19.
Collapse
Affiliation(s)
- Marina Luise Viola Azevedo
- Laboratory of Experimental Pathology, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Aline Cristina Zanchettin
- Postgraduate Program in Biotechnology Applied to Child and Adolescent Health, Faculdades Pequeno Príncipe, Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | - Caroline Busatta Vaz de Paula
- Laboratory of Experimental Pathology, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Jarbas da Silva Motta Júnior
- Hospital Marcelino Champagnat, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Mineia Alessandra Scaranello Malaquias
- Laboratory of Experimental Pathology, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Sonia Mara Raboni
- Virology Laboratory, Universidade Federal do Paraná, Hospital de Clínicas, Curitiba, Brazil
| | - Plínio Cezar Neto
- Laboratory of Experimental Pathology, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Rafaela Chiuco Zeni
- Laboratory of Experimental Pathology, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Amanda Prokopenko
- Laboratory of Experimental Pathology, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Nícolas Henrique Borges
- Laboratory of Experimental Pathology, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Thiago Mateus Godoy
- Laboratory of Experimental Pathology, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Ana Paula Kubaski Benevides
- Laboratory of Experimental Pathology, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Daiane Gavlik de Souza
- Virology Laboratory, Universidade Federal do Paraná, Hospital de Clínicas, Curitiba, Brazil
| | - Cristina Pellegrino Baena
- Hospital Marcelino Champagnat, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Cleber Machado-Souza
- Postgraduate Program in Biotechnology Applied to Child and Adolescent Health, Faculdades Pequeno Príncipe, Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, Brazil
| | - Lucia de Noronha
- Laboratory of Experimental Pathology, Postgraduate Program of Health Sciences, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| |
Collapse
|
46
|
Ritzmann F, Beisswenger C. Preclinical studies and the function of IL-17 cytokines in COPD. Ann Anat 2021; 237:151729. [PMID: 33798693 DOI: 10.1016/j.aanat.2021.151729] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is among the leading causes of death worldwide and imposes a high economic burden to the health systems. COPD is characterized by chronic inflammation of the lung leading to airflow limitation, alveolar tissue destruction, and emphysema. Therefore, anti-inflammatory therapies for the treatment of COPD are of interest. In this review, we focus on the function of the IL-17 cytokines IL-17A and IL-17C, both known to mediate the recruitment of inflammatory cells, in the pathogenesis of COPD. We highlight that the expression of IL-17A and IL-17C is induced by pathogens frequently found in lungs of COPD patients and that targeting IL-17-signaling is an interesting option for the treatment of acute exacerbation of COPD.
Collapse
Affiliation(s)
- Felix Ritzmann
- Department of Internal Medicine V - Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V - Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany.
| |
Collapse
|
47
|
Grossman PC, Schneider DA, Herndon DR, Knowles DP, Highland MA. Differential pulmonary immunopathology of domestic sheep (Ovis aries) and bighorn sheep (Ovis canadensis) with Mycoplasma ovipneumoniae infection: A retrospective study. Comp Immunol Microbiol Infect Dis 2021; 76:101641. [PMID: 33689940 DOI: 10.1016/j.cimid.2021.101641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023]
Abstract
Mycoplasma ovipneumoniae is a respiratory pathogen that impacts domestic sheep (Ovis aries; DS) and bighorn sheep (Ovis canadensis; BHS). BHS are reported to be more susceptible than DS to developing polymicrobial pneumonia associated with M. ovipneumoniae infection. Using formalin-fixed paraffin-embedded tissues, we performed a retrospective study investigating the pulmonary immune response of DS and BHS to M. ovipneumoniae infection. M. ovipneumoniae infected DS exhibited a more robust and well-organized BALT formation as compared to BHS. Digital analysis of immunohistochemical chromogen deposition in lung tissue was used to quantitate T cell marker CD3, B cell markers CD20 and CD79a, macrophage markers CD163 and Iba1, and cytokine IL-17. A significant interaction of species and infection status was identified for CD3, CD163, and IL-17. BHS had a greater increase in bronchiolar CD3 and bronchiolar and alveolar CD163 with infection, as compared to DS. BHS had an increase in bronchiolar associated lymph tissue (BALT) and alveolar IL-17 with infection, while these remained similar in DS regardless of infection status. IL-17 in respiratory epithelium of bronchi and bronchioles comparatively decreased in DS and increased in BHS with infection. These data begin to define the interspecies differential immune response to pulmonary M. ovipneumoniae infection in DS and BHS and provide the first investigations of respiratory epithelium-associated IL-17 in ovine.
Collapse
Affiliation(s)
- Paige C Grossman
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - David A Schneider
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA; United States Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, WA, 99164, USA
| | - David R Herndon
- United States Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, WA, 99164, USA
| | - Donald P Knowles
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA; United States Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, WA, 99164, USA
| | - Margaret A Highland
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA; United States Department of Agriculture, Agricultural Research Service, Animal Disease Research Unit, Pullman, WA, 99164, USA.
| |
Collapse
|
48
|
Madbouly N, El Amir A, Abdel Kader A, Rabee I, Farid A. The immunomodulatory activity of secnidazole-nitazoxanide in a murine cryptosporidiosis model. J Med Microbiol 2021; 70. [PMID: 33625354 DOI: 10.1099/jmm.0.001327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. Cryptosporidium parvum causes intestinal parasitic infections affecting both immunosuppressed and immunocompetent individuals.Gap statement. Given the absence of effective treatments for cryptosporidiosis, especially in immunodeficient patients, the present study was designed to assess the therapeutic efficacy of secnidazole (SEC) and its combination with nitazoxanide (NTZ) in comparison to single NTZ treatment in relation to the immune status of a murine model of C. parvum infection.Methodology. The infected groups were administered NTZ, SEC or NTZ-SEC for three or five successive doses. At days 10 and 12 post-infection (p.i.), the mice were sacrificed, and the efficacy of the applied drugs was evaluated by comparing the histopathological alterations in ileum and measuring the T helper Th1 (interferon gamma; IFN-γ), Th2 [interleukin (IL)-4 and IL-10] and Th17 (IL-17) cytokine profiles in serum.Results. The NTZ-SEC combination recorded the maximal reduction of C. parvum oocyst shedding, endogenous stages count and intestinal histopathology, regardless of the immune status of the infected mice. The efficacy of NTZ-SEC was dependent on the period of administration, as the 5 day-based treatment protocol was also more effective than the 3 day-based one in terms of immunocompetence and immunosuppression. The present treatment schedule induced an immunomodulatory effect from SEC that developed a protective immune response against C. parvum infection with reduced production of serum IL-17, IFN-γ, IL-4 and IL-10.Conclusions. Application of NTZ-SEC combined therapy may be useful in treatment of C. parvum, especially in cases involving immunosuppression.
Collapse
Affiliation(s)
- Neveen Madbouly
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Azza El Amir
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Asmaa Abdel Kader
- Department of Parasitology, Theodore Bilharz Research Institute, Giza, Egypt
| | - Ibraheem Rabee
- Department of Parasitology, Theodore Bilharz Research Institute, Giza, Egypt
| | - Alyaa Farid
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
49
|
Rimachi Hidalgo MA, Cirelli T, da Silva BR, Nicchio IG, Nepomuceno R, Orrico SRP, Cirelli JA, Theodoro LH, Barros SP, Scarel-Caminaga RM. Polymorphisms and haplotypes in the Interleukin 17 Alfa gene: potential effect of smoking habits in the association with periodontitis and type 2 diabetes mellitus. Mol Biol Rep 2021; 48:1103-1114. [PMID: 33559820 DOI: 10.1007/s11033-021-06172-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 01/19/2021] [Indexed: 11/26/2022]
Abstract
Few studies evaluate interrelationships between periodontitis (P) and Type 2 Diabetes Mellitus (T2DM). The aim of this study is to investigate the genetic susceptibility to periodontitis alone, or concomitant with T2DM (as comorbidities), analyzing single nucleotide polymorphisms (SNPs) in the Interleukin 17 alpha (IL17A) gene, considering the biochemical profile and smoking habits on the subjects' periodontal status. We investigated 879 individuals divided into: T2DM subjects also affected by severe or moderate periodontitis (T2DM-P, n = 199); non-diabetics with severe or moderate periodontitis (PERIODONTITIS, n = 342); and healthy subjects (HEALTHY, n = 338). Subjects underwent complete periodontal examination, history of smoking habits, glycemic and lipid biochemical evaluation. DNA from buccal cells was utilized to genotype the SNPs rs2275913, rs3819024 and rs10484879. The impact of the subjects' biochemical profile was analyzed in their periodontal status. Each SNP was analyzed independently, and as haplotypes, by multiple logistic regressions, adjusted for covariates, and also stratifying the groups by age, sex and smoking habits. Independently of the periodontitis degree, poorly-controlled T2DM subjects showed worse glycemic and lipid profile. Multiple logistic regressions demonstrated that smokers and former-smokers carrying the GG genotype of rs3819024 seemed to have higher risk for T2DM-Periodontitis (OR = 6.33; 95% CI = 1.26-31.77, p = 0.02), and mainly for T2DM alone (OR = 5.11; 95% CI = 1.37-19.06, p = 0.01), than never smokers. We found the potential effect of smoking habits in the association of IL17A-rs3819024-GG with diseased phenotypes. Because the observed wide confidence intervals, further studies enrolling larger populations, and SNPs' functional evaluations are needed to better understand our findings.
Collapse
Affiliation(s)
- Marco A Rimachi Hidalgo
- Department of Diagnosis and Surgery, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
- Department of Morphology and Pediatric Clinics, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
| | - Thamiris Cirelli
- Department of Diagnosis and Surgery, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
- Department of Morphology and Pediatric Clinics, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
| | - Bárbara Roque da Silva
- Department of Diagnosis and Surgery, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
- Department of Morphology and Pediatric Clinics, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
| | - Ingra Gagno Nicchio
- Department of Diagnosis and Surgery, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
- Department of Morphology and Pediatric Clinics, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
| | - Rafael Nepomuceno
- Department of Diagnosis and Surgery, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
- Department of Morphology and Pediatric Clinics, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
| | - Silvana R P Orrico
- Department of Diagnosis and Surgery, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
- Advanced Research Center in Medicine, Union of the Colleges of the Great Lakes (UNILAGO), São José do Rio Preto, SP, 15030-070, Brazil
| | - Joni A Cirelli
- Department of Diagnosis and Surgery, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil
| | - Letícia Helena Theodoro
- Department of Diagnosis and Surgery, São Paulo State University - UNESP, School of Dentistry at Araçatuba, Araçatuba, SP, Brazil
| | - Silvana P Barros
- Department of Periodontology, University of North Carolina at Chapel Hill - UNC, School of Dentistry, Chapel Hill, NC, USA
| | - Raquel M Scarel-Caminaga
- Department of Morphology and Pediatric Clinics, São Paulo State University - UNESP, School of Dentistry at Araraquara, Araraquara, SP, Brazil.
| |
Collapse
|
50
|
Kang J, Loh K, Belyayev L, Cha P, Sadat M, Khan K, Gusev Y, Bhuvaneshwar K, Ressom H, Moturi S, Kaiser J, Hawksworth J, Robson SC, Matsumoto CS, Zasloff M, Fishbein TM, Kroemer A. Type 3 innate lymphoid cells are associated with a successful intestinal transplant. Am J Transplant 2021; 21:787-797. [PMID: 32594614 PMCID: PMC8049507 DOI: 10.1111/ajt.16163] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 01/25/2023]
Abstract
Although innate lymphoid cells (ILCs) play fundamental roles in mucosal barrier functionality and tissue homeostasis, ILC-related mechanisms underlying intestinal barrier function, homeostatic regulation, and graft rejection in intestinal transplantation (ITx) patients have yet to be thoroughly defined. We found protective type 3 NKp44+ ILCs (ILC3s) to be significantly diminished in newly transplanted allografts, compared to allografts at 6 months, whereas proinflammatory type 1 NKp44- ILCs (ILC1s) were higher. Moreover, serial immunomonitoring revealed that in healthy allografts, protective ILC3s repopulate by 2-4 weeks postoperatively, but in rejecting allografts they remain diminished. Intracellular cytokine staining confirmed that NKp44+ ILC3 produced protective interleukin-22 (IL-22), whereas ILC1s produced proinflammatory interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α). Our findings about the paucity of protective ILC3s immediately following transplant and their repopulation in healthy allografts during the first month following transplant were confirmed by RNA-sequencing analyses of serial ITx biopsies. Overall, our findings show that ILCs may play a key role in regulating ITx graft homeostasis and could serve as sentinels for early recognition of allograft rejection and be targets for future therapies.
Collapse
Affiliation(s)
- Jiman Kang
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007
| | - Katrina Loh
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007,Children’s National Medical Center, 111 Michigan Avenue NW, Washington DC, 20010
| | - Leonid Belyayev
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007,Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda MD, 20814
| | - Priscilla Cha
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007,Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda MD, 20814
| | - Mohammed Sadat
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007
| | - Khalid Khan
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007
| | - Yuriy Gusev
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, 2115 Wisconsin Ave NW, Suite 110, Washington DC, 20007
| | - Krithika Bhuvaneshwar
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, 2115 Wisconsin Ave NW, Suite 110, Washington DC, 20007
| | - Habtom Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 4000 Reservoir Road NW, Washington DC, 20007
| | - Sangeetha Moturi
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007
| | - Jason Kaiser
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007,Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda MD, 20814
| | - Jason Hawksworth
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007,Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda MD, 20814
| | - Simon C. Robson
- Departments of Anesthesiology and Medicine, CLS 612, 330 Brookline Avenue, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA, 02115
| | - Cal S. Matsumoto
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007
| | - Michael Zasloff
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007
| | - Thomas M. Fishbein
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007
| | - Alexander Kroemer
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington DC, 20007
| |
Collapse
|