1
|
Lim YJ, Park SA, Wang D, Jin W, Ku WL, Zhang D, Xu J, Patiño LC, Liu N, Chen W, Kazmi R, Zhao K, Zhang YE, Sun L, Chen W. MicroRNA-19b exacerbates systemic sclerosis through promoting Th9 cells. Cell Rep 2024; 43:114565. [PMID: 39083380 PMCID: PMC11440512 DOI: 10.1016/j.celrep.2024.114565] [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: 02/21/2024] [Revised: 06/19/2024] [Accepted: 07/16/2024] [Indexed: 08/02/2024] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune disease characterized by fibrosis of the skin and multiple vital organs, but the immunological pathogenesis of SSc remains unclear. We show here that miR-19b promotes Th9 cells that exacerbate SSc. Specifically, miR-19b and interleukin (IL)-9 increase in CD4+ T cells in experimental SSc in mice induced with bleomycin. Inhibiting miR-19b reduces Th9 cells and ameliorates the disease. Mechanistically, transforming growth factor beta (TGF-β) plus IL-4 activates pSmad3-Ser213 and TRAF6-K63 ubiquitination by suppressing NLRC3. Activated TRAF6 sequentially promotes TGF-β-activated kinase 1 (TAK1) and nuclear factor κB (NF-κB) p65 phosphorylation, leading to the upregulation of miR-19b. Notably, miR-19b activated Il9 gene expression by directly suppressing atypical E2F family member E2f8. In patients with SSc, higher levels of IL9 and MIR-19B correlate with worse disease progression. Our findings reveal miR-19b as a key factor in Th9 cell-mediated SSc pathogenesis and should have clinical implications for patients with SSc.
Collapse
Affiliation(s)
- Yun-Ji Lim
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Sang-A Park
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Dandan Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Wenwen Jin
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Wai Lim Ku
- Systemic Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 31 Center Drive, Bethesda, MD 20892, USA
| | - Dunfang Zhang
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Junji Xu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Liliana C Patiño
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Na Liu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Weiwei Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Rida Kazmi
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA
| | - Keji Zhao
- Systemic Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 31 Center Drive, Bethesda, MD 20892, USA
| | - Ying E Zhang
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China.
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
2
|
Kyo M, Zhu Z, Shibata R, Ooka T, Mansbach JM, Harmon B, Hahn A, Pérez-Losada M, Camargo CA, Hasegawa K. Nasal microRNA signatures for disease severity in infants with respiratory syncytial virus bronchiolitis: a multicentre prospective study. BMJ Open Respir Res 2024; 11:e002288. [PMID: 39089741 PMCID: PMC11293419 DOI: 10.1136/bmjresp-2023-002288] [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: 12/31/2023] [Accepted: 06/28/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) bronchiolitis contributes to a large morbidity and mortality burden globally. While emerging evidence suggests that airway microRNA (miRNA) is involved in the pathobiology of RSV infection, its role in the disease severity remains unclear. METHODS In this multicentre prospective study of infants (aged<1 year) hospitalised for RSV bronchiolitis, we sequenced the upper airway miRNA and messenger RNA (mRNA) at hospitalisation. First, we identified differentially expressed miRNAs (DEmiRNAs) associated with higher bronchiolitis severity-defined by respiratory support (eg, positive pressure ventilation, high-flow oxygen therapy) use. We also examined the biological significance of miRNAs through pathway analysis. Second, we identified differentially expressed mRNAs (DEmRNAs) associated with bronchiolitis severity. Last, we constructed miRNA-mRNA coexpression networks and determined hub mRNAs by weighted gene coexpression network analysis (WGCNA). RESULTS In 493 infants hospitalised with RSV bronchiolitis, 19 DEmiRNAs were associated with bronchiolitis severity (eg, miR-27a-3p, miR-26b-5p; false discovery rate<0.10). The pathway analysis using miRNA data identified 1291 bronchiolitis severity-related pathways-for example, regulation of cell adhesion mediated by integrin. Second, 1298 DEmRNAs were associated with bronchiolitis severity. Last, of these, 190 DEmRNAs were identified as targets of DEmiRNAs and negatively correlated with DEmiRNAs. By applying WGCNA to DEmRNAs, four disease modules were significantly associated with bronchiolitis severity-for example, microtubule anchoring, cell-substrate junction. The hub genes for each of these modules were also identified-for example, PCM1 for the microtubule anchoring module, LIMS1 for the cell-substrate junction module. CONCLUSIONS In infants hospitalised for RSV bronchiolitis, airway miRNA-mRNA coexpression network contributes to the pathobiology of bronchiolitis severity.
Collapse
Affiliation(s)
- Michihito Kyo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhaozhong Zhu
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ryohei Shibata
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tadao Ooka
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Health Science, University of Yamanashi, Kofu, Yamanashi, Japan
| | - Jonathan M Mansbach
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Brennan Harmon
- Centre for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
| | - Andrea Hahn
- Centre for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
- Department of Paediatrics, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
- Division of Infectious Diseases, Children’s National Hospital, Washington, District of Columbia, USA
| | - Marcos Pérez-Losada
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, The George Washington University, Washington, District of Columbia, USA
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Liao K, Chen P, Zhang M, Wang J, Hatzihristidis T, Lin X, Yang L, Yao N, Liu C, Hong Y, Li X, Liu H, Zúñiga-Pflücker JC, Love PE, Chen X, Liu WH, Zhao B, Xiao C. Critical roles of the miR-17∼92 family in thymocyte development, leukemogenesis, and autoimmunity. Cell Rep 2024; 43:114261. [PMID: 38776224 DOI: 10.1016/j.celrep.2024.114261] [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: 11/15/2023] [Revised: 03/24/2024] [Accepted: 05/07/2024] [Indexed: 05/24/2024] Open
Abstract
Thymocyte development requires precise control of PI3K-Akt signaling to promote proliferation and prevent leukemia and autoimmune disorders. Here, we show that ablating individual clusters of the miR-17∼92 family has a negligible effect on thymocyte development, while deleting the entire family severely impairs thymocyte proliferation and reduces thymic cellularity, phenocopying genetic deletion of Dicer. Mechanistically, miR-17∼92 expression is induced by Myc-mediated pre-T cell receptor (TCR) signaling, and miR-17∼92 promotes thymocyte proliferation by suppressing the translation of Pten. Retroviral expression of miR-17∼92 restores the proliferation and differentiation of Myc-deficient thymocytes. Conversely, partial deletion of the miR-17∼92 family significantly delays Myc-driven leukemogenesis. Intriguingly, thymocyte-specific transgenic miR-17∼92 expression does not cause leukemia or lymphoma but instead aggravates skin inflammation, while ablation of the miR-17∼92 family ameliorates skin inflammation. This study reveals intricate roles of the miR-17∼92 family in balancing thymocyte development, leukemogenesis, and autoimmunity and identifies those microRNAs (miRNAs) as potential therapeutic targets for leukemia and autoimmune diseases.
Collapse
Affiliation(s)
- Kunyu Liao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Pengda Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Mengdi Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Furong Laboratory, Changsha, China
| | - Jiazhen Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Teri Hatzihristidis
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Xiaoxi Lin
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Liang Yang
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Nan Yao
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Chenfeng Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Yazhen Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hong Liu
- Furong Laboratory, Changsha, China; Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan Carlos Zúñiga-Pflücker
- Department of Immunology, University of Toronto, Toronto, ON, Canada; Sunnybrook Research Institute, Toronto, ON, Canada
| | - Paul E Love
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Xiang Chen
- Furong Laboratory, Changsha, China; Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Wen-Hsien Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China.
| | - Bin Zhao
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China; Furong Laboratory, Changsha, China.
| | - Changchun Xiao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
4
|
Gaál Z. Role of microRNAs in Immune Regulation with Translational and Clinical Applications. Int J Mol Sci 2024; 25:1942. [PMID: 38339220 PMCID: PMC10856342 DOI: 10.3390/ijms25031942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 01/31/2024] [Accepted: 02/03/2024] [Indexed: 02/12/2024] Open
Abstract
MicroRNAs (miRNAs) are 19-23 nucleotide long, evolutionarily conserved noncoding RNA molecules that regulate gene expression at the post-transcriptional level. In this review, involvement of miRNAs is summarized in the differentiation and function of immune cells, in anti-infective immune responses, immunodeficiencies and autoimmune diseases. Roles of miRNAs in anticancer immunity and in the transplantation of solid organs and hematopoietic stem cells are also discussed. Major focus is put on the translational clinical applications of miRNAs, including the establishment of noninvasive biomarkers for differential diagnosis and prediction of prognosis. Patient selection and response prediction to biological therapy is one of the most promising fields of application. Replacement or inhibition of miRNAs has enormous therapeutic potential, with constantly expanding possibilities. Although important challenges still await solutions, evaluation of miRNA fingerprints may contribute to an increasingly personalized management of immune dysregulation with a remarkable reduction in toxicity and treatment side effects. More detailed knowledge of the molecular effects of physical exercise and nutrition on the immune system may facilitate self-tailored lifestyle recommendations and advances in prevention.
Collapse
Affiliation(s)
- Zsuzsanna Gaál
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 98 Nagyerdei krt, 4032 Debrecen, Hungary
| |
Collapse
|
5
|
Jafarzadeh A, Chauhan P, Nemati M, Jafarzadeh S, Yoshimura A. Aberrant expression of suppressor of cytokine signaling (SOCS) molecules contributes to the development of allergic diseases. Clin Exp Allergy 2023; 53:1147-1161. [PMID: 37641429 DOI: 10.1111/cea.14385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/20/2023] [Accepted: 08/12/2023] [Indexed: 08/31/2023]
Abstract
Suppressor of cytokine signalling (SOCS) proteins bind to certain cytokine receptors, Janus kinases and signalling molecules to regulate signalling pathways, thus controlling immune and inflammatory responses. Dysregulated expression of various types of SOCS molecules was indicated in multiple types of allergic diseases. SOCS1, SOCS2, SOCS3, SOCS5, and cytokine-inducible SH2 domain protein (CISH) can differentially exert anti-allergic impacts through different mechanisms, such as suppressing Th2 cell development and activation, reducing eosinophilia, decreasing IgE production, repressing production of pro-allergic chemokines, promoting Treg cell differentiation and activation, suppressing Th17 cell differentiation and activation, increasing anti-allergic Th1 responses, inhibiting M2 macrophage polarization, modulating survival and development of mast cells, reducing pro-allergic activity of keratinocytes, and suppressing pulmonary fibrosis. Although some anti-allergic effects were attributed to SOCS3, it can perform pro-allergic impacts through several pathways, such as promoting Th2 cell development and activation, supporting eosinophilia, boosting pro-allergic activity of eosinophils, increasing IgE production, enhancing the expression of the pro-allergic chemokine receptor, reducing Treg cell differentiation, increasing pro-allergic Th9 responses, as well as supporting mucus secretion and collagen deposition. In this review, we discuss the contrasting roles of SOCS proteins in contexts of allergic disorders to provide new insights regarding the pathophysiology of these diseases and possibly explore SOCS proteins as potential therapeutic targets for alleviating allergies.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Prashant Chauhan
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Maryam Nemati
- Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Khayati S, Dehnavi S, Sadeghi M, Tavakol Afshari J, Esmaeili SA, Mohammadi M. The potential role of miRNA in regulating macrophage polarization. Heliyon 2023; 9:e21615. [PMID: 38027572 PMCID: PMC10665754 DOI: 10.1016/j.heliyon.2023.e21615] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Macrophage polarization is a dynamic process determining the outcome of various physiological and pathological situations through inducing pro-inflammatory responses or resolving inflammation via exerting anti-inflammatory effects. The miRNAs are epigenetic regulators of different biologic pathways that target transcription factors and signaling molecules to promote macrophage phenotype transition and regulate immune responses. Modulating the macrophage activation, differentiation, and polarization by miRNAs is crucial for immune responses in response to microenvironmental signals and under various physiological and pathological conditions. In term of clinical significance, regulating macrophage polarization via miRNAs could be utilized for inflammation control. Also, understanding the role of miRNAs in macrophage polarization can provide insights into diagnostic strategies associated with dysregulated miRNAs and for developing macrophage-centered therapeutic methods. In this case, targeting miRNAs to further regulate of macrophage polarization may become an efficient strategy for treating immune-associated disorders. The current review investigated and categorized various miRNAs directly or indirectly involved in macrophage polarization by targeting different transcription factors and signaling pathways. In addition, prospects for regulating macrophage polarization via miRNA as a therapeutic choice that could be implicated in various pathological conditions, including cancer or inflammation-mediated injuries, were discussed.
Collapse
Affiliation(s)
- Shaho Khayati
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sajad Dehnavi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahvash Sadeghi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojgan Mohammadi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Johansson K, Gagnon JD, Zhou SK, Fassett MS, Schroeder AW, Kageyama R, Bautista RA, Pham H, Woodruff PG, Ansel KM. An essential role for miR-15/16 in Treg suppression and restriction of proliferation. Cell Rep 2023; 42:113298. [PMID: 37862171 PMCID: PMC10664750 DOI: 10.1016/j.celrep.2023.113298] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/07/2023] [Accepted: 10/02/2023] [Indexed: 10/22/2023] Open
Abstract
The miR-15/16 family targets a large network of genes in T cells to restrict their cell cycle, memory formation, and survival. Upon T cell activation, miR-15/16 are downregulated, allowing rapid expansion of differentiated effector T cells to mediate a sustained response. Here, we used conditional deletion of miR-15/16 in regulatory T cells (Tregs) to identify immune functions of the miR-15/16 family in T cells. miR-15/16 are indispensable to maintain peripheral tolerance by securing efficient suppression by a limited number of Tregs. miR-15/16 deficiency alters expression of critical Treg proteins and results in accumulation of functionally impaired FOXP3loCD25loCD127hi Tregs. Excessive proliferation in the absence of miR-15/16 shifts Treg fate and produces an effector Treg phenotype. These Tregs fail to control immune activation, leading to spontaneous multi-organ inflammation and increased allergic inflammation in a mouse model of asthma. Together, our results demonstrate that miR-15/16 expression in Tregs is essential to maintain immune tolerance.
Collapse
Affiliation(s)
- Kristina Johansson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medical Biochemistry and Cell Biology, University of Gothenburg, 40530 Gothenburg, Sweden; Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, 40530 Gothenburg, Sweden
| | - John D Gagnon
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Simon K Zhou
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Marlys S Fassett
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Andrew W Schroeder
- Department of Medicine, Genomics CoLab, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Robin Kageyama
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Rodriel A Bautista
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hewlett Pham
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Prescott G Woodruff
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - K Mark Ansel
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
8
|
Chen J, Zhu Q, Mo Y, Ling H, Wang Y, Xie H, Li L. Exploring the action mechanism of Jinxin oral liquid on asthma by network pharmacology, molecular docking, and microRNA recognition. Medicine (Baltimore) 2023; 102:e35438. [PMID: 37904411 PMCID: PMC10615469 DOI: 10.1097/md.0000000000035438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/08/2023] [Indexed: 11/01/2023] Open
Abstract
Using network pharmacology, molecular docking, and microRNA recognition, we have elucidated the mechanisms underlying the treatment of asthma by Jinxin oral liquid (JXOL). We began by identifying and normalizing the active compounds in JXOL through searches in the traditional Chinese medicine systems pharmacology database, SwissADME database, encyclopedia of traditional Chinese medicine database, HERB database, and PubChem. Subsequently, we gathered and standardized the targets of these active compounds from sources including the encyclopedia of traditional Chinese medicine database, similarity ensemble approach dataset, UniProt, and other databases. Disease targets were extracted from GeneCards, PharmGKB, OMIM, comparative toxicogenomics database, and DisGeNET. The intersection of targets between JXOL and asthma was determined using a Venn diagram. We visualized a Formula-Herb-Compound-Target-Disease network and a protein-protein interaction network using Cytoscape 3.9.0. Molecular docking studies were performed using Schrodinger software. To identify pathways related to asthma, we conducted gene ontology functional analysis and Kyoto encyclopedia of genes and genomes pathway enrichment analysis using Metascape. MicroRNAs regulating the hub genes were obtained from the miRTarBase database, and a network linking these targets and miRNAs was constructed. Finally, we found 88 bioactive components in JXOL and 218 common targets with asthma. Molecular docking showed JXOL key compounds strongly bind to HUB targets. According to gene ontology biological process analysis and Kyoto encyclopedia of genes and genomes pathway enrichment analysis, the PI3K-Akt signaling pathway, the MAPK signaling pathway, or the cAMP signaling pathway play a key role in treating of asthma by JXOL. The HUB target-miRNA network showed that 6 miRNAs were recognized. In our study, we have revealed for the first time the unique components, multiple targets, and diverse pathways in JXOL that underlie its mechanism of action in treating asthma through miRNAs.
Collapse
Affiliation(s)
- Jing Chen
- Shanghai municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Qiaozhen Zhu
- Clinical Medical School, Henan University, Kaifeng, People’s Republic of China
| | - Yanling Mo
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Hao Ling
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Yan Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Huihui Xie
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Lan Li
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| |
Collapse
|
9
|
Fassett MS, Braz JM, Castellanos CA, Salvatierra JJ, Sadeghi M, Yu X, Schroeder AW, Caston J, Munoz-Sandoval P, Roy S, Lazarevsky S, Mar DJ, Zhou CJ, Shin JS, Basbaum AI, Ansel KM. IL-31-dependent neurogenic inflammation restrains cutaneous type 2 immune response in allergic dermatitis. Sci Immunol 2023; 8:eabi6887. [PMID: 37831760 PMCID: PMC10890830 DOI: 10.1126/sciimmunol.abi6887] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/18/2023] [Indexed: 10/15/2023]
Abstract
Despite robust literature associating IL-31 with pruritic inflammatory skin diseases, its influence on cutaneous inflammation and the interplay between inflammatory and neurosensory pathways remain unmapped. Here, we examined the consequences of disrupting Il31 and its receptor Il31ra in a mouse model of house dust mite (HDM)-induced allergic dermatitis. Il31-deficient mice displayed a deficit in HDM dermatitis-associated scratching, consistent with its well-established role as a pruritogen. In contrast, Il31 deficiency increased the number and proportion of cutaneous type 2 cytokine-producing CD4+ T cells and serum IgE in response to HDM. Furthermore, Il4ra+ monocytes and macrophages capable of fueling a feedforward type 2 inflammatory loop were selectively enriched in Il31ra-deficient HDM dermatitis skin. Thus, IL-31 is not strictly a proinflammatory cytokine but rather an immunoregulatory factor that limits the magnitude of type 2 inflammatory responses in skin. Our data support a model wherein IL-31 activation of IL31RA+ pruritoceptors triggers release of calcitonin gene-related protein (CGRP), which can mediate neurogenic inflammation, inhibit CD4+ T cell proliferation, and reduce T cell production of the type 2 cytokine IL-13. Together, these results illustrate a previously unrecognized neuroimmune pathway that constrains type 2 tissue inflammation in the setting of chronic cutaneous allergen exposure and may explain paradoxical dermatitis flares in atopic patients treated with anti-IL31RA therapy.
Collapse
Affiliation(s)
- Marlys S Fassett
- Department of Dermatology, University of California, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
- Sandler Asthma Basic Research Center (SABRe), San Francisco, CA, USA
| | - Joao M Braz
- Department of Anatomy, University of California, San Francisco, CA, USA
| | - Carlos A Castellanos
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
- Sandler Asthma Basic Research Center (SABRe), San Francisco, CA, USA
| | | | - Mahsa Sadeghi
- Department of Anatomy, University of California, San Francisco, CA, USA
| | - Xiaobing Yu
- Department of Anatomy, University of California, San Francisco, CA, USA
- Department of Anesthesiology, University of California, San Francisco, CA, USA
| | | | - Jaela Caston
- Department of Dermatology, University of California, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Priscila Munoz-Sandoval
- Department of Dermatology, University of California, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
- Sandler Asthma Basic Research Center (SABRe), San Francisco, CA, USA
- Howard Hughes Medical Institute, San Francisco, CA 94143, USA
| | - Suparna Roy
- Department of Dermatology, University of California, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
- Sandler Asthma Basic Research Center (SABRe), San Francisco, CA, USA
| | - Steven Lazarevsky
- Department of Dermatology, University of California, San Francisco, CA, USA
| | - Darryl J Mar
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Connie J Zhou
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
| | - Jeoung-Sook Shin
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
- Sandler Asthma Basic Research Center (SABRe), San Francisco, CA, USA
| | - Allan I Basbaum
- Department of Anatomy, University of California, San Francisco, CA, USA
| | - K Mark Ansel
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA
- Sandler Asthma Basic Research Center (SABRe), San Francisco, CA, USA
| |
Collapse
|
10
|
Li Z, Lei Z, Cai Y, Cheng DB, Sun T. MicroRNA therapeutics and nucleic acid nano-delivery systems in bacterial infection: a review. J Mater Chem B 2023; 11:7804-7833. [PMID: 37539650 DOI: 10.1039/d3tb00694h] [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: 08/05/2023]
Abstract
Bacteria that have worked with humans for thousands of years pose a major threat to human health even today, as drug resistance has become a prominent problem. Compared to conventional drug therapy, nucleic acid-based therapies are a promising and potential therapeutic strategy for diseases in which nucleic acids are delivered through a nucleic acid delivery system to regulate gene expression in specific cells, offering the possibility of curing intractable diseases that are difficult to treat at this stage. Among the many nucleic acid therapeutic ideas, microRNA, a class of small nucleic acids with special properties, has made great strides in biology and medicine in just over two decades, showing promise in preclinical drug development. In this review, we introduce recent advances in nucleic acid delivery systems and their clinical applications, highlighting the potential of nucleic acid therapies, especially miRNAs extracted from traditional herbs, in combination with the existing set of nucleic acid therapeutic systems, to potentially open up a new line of thought in the treatment of cancer, viruses, and especially bacterial infectious diseases.
Collapse
Affiliation(s)
- Ze Li
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Yilun Cai
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Hospital of Wuhan University of Technology, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| |
Collapse
|
11
|
Bao K, Isik Can U, Miller MM, Brown IK, Dell'Aringa M, Dooms H, Seibold MA, Scott-Browne J, Lee Reinhardt R. A bifurcated role for c-Maf in Th2 and Tfh2 cells during helminth infection. Mucosal Immunol 2023; 16:357-372. [PMID: 37088263 PMCID: PMC10290510 DOI: 10.1016/j.mucimm.2023.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/03/2023] [Accepted: 04/13/2023] [Indexed: 04/25/2023]
Abstract
Differences in transcriptomes, transcription factor usage, and function have identified T follicular helper 2 (Tfh2) cells and T helper 2 (Th2) cells as distinct clusters of differentiation 4+",(CD4) T-cell subsets in settings of type-2 inflammation. Although the transcriptional programs driving Th2 cell differentiation and cytokine production are well defined, dependence on these classical Th2 programs by Tfh2 cells is less clear. Using cytokine reporter mice in combination with transcription factor inference analysis, the b-Zip transcription factor c-Maf and its targets were identified as an important regulon in both Th2 and Tfh2 cells. Conditional deletion of c-Maf in T cells confirmed its importance in type-2 cytokine expression by Th2 and Tfh2 cells. However, while c-Maf was not required for Th2-driven helminth clearance or lung eosinophilia, it was required for Tfh2-driven Immunoglobulin E production and germinal center formation. This differential regulation of cell-mediated and humoral immunity by c-Maf was a result of redundant pathways in Th2 cells that were absent in Tfh2 cells, and c-Maf-specific mechanisms in Tfh2 cells that were absent in Th2 cells. Thus, despite shared expression by Tfh2 and Th2 cells, c-Maf serves as a unique regulator of Tfh2-driven humoral hallmarks during type-2 immunity.
Collapse
Affiliation(s)
- Katherine Bao
- Department of Immunology, Duke University Medical Center, Durham, USA
| | - Uryan Isik Can
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, USA
| | - Mindy M Miller
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, USA
| | - Ivy K Brown
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, USA
| | - Mark Dell'Aringa
- Department of Immunology, Duke University Medical Center, Durham, USA; Department of Immunology and Genomic Medicine, National Jewish Health, Denver, USA
| | - Hans Dooms
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Max A Seibold
- Center for Genes, Environment, and Health, National Jewish Health, Denver, USA; Department of Pediatrics, National Jewish Health, Denver, USA; Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, USA
| | - James Scott-Browne
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Richard Lee Reinhardt
- Department of Immunology, Duke University Medical Center, Durham, USA; Department of Immunology and Genomic Medicine, National Jewish Health, Denver, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, USA.
| |
Collapse
|
12
|
Johansson K, Gagnon JD, Zhou S, Fassett MS, Schroeder AW, Kageyama R, Bautista RA, Pham H, Woodruff PG, Ansel KM. An essential role for miR-15/16 in Treg suppression and restriction of proliferation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.26.533356. [PMID: 36993421 PMCID: PMC10055372 DOI: 10.1101/2023.03.26.533356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
The miR-15/16 family is a highly expressed group of tumor suppressor miRNAs that target a large network of genes in T cells to restrict their cell cycle, memory formation and survival. Upon T cell activation, miR-15/16 are downregulated, allowing rapid expansion of differentiated effector T cells to mediate a sustained immune response. Here, using conditional deletion of miR-15/16 in immunosuppressive regulatory T cells (Tregs) that express FOXP3, we identify new functions of the miR-15/16 family in T cell immunity. miR-15/16 are indispensable to maintain peripheral tolerance by securing efficient suppression by a limited number of Tregs. miR-15/16-deficiency alters Treg expression of critical functional proteins including FOXP3, IL2Rα/CD25, CTLA4, PD-1 and IL7Rα/CD127, and results in accumulation of functionally impaired FOXP3loCD25loCD127hi Tregs. Excessive proliferation in the absence of miR-15/16 inhibition of cell cycle programs shifts Treg diversity and produces an effector Treg phenotype characterized by low expression of TCF1, CD25 and CD62L, and high expression of CD44. These Tregs fail to control immune activation of CD4+ effector T cells, leading to spontaneous multi-organ inflammation and increased allergic airway inflammation in a mouse model of asthma. Together, our results demonstrate that miR-15/16 expression in Tregs is essential to maintain immune tolerance.
Collapse
|
13
|
Crosstalk of Transcriptional Regulators of Adaptive Immune System and microRNAs: An Insight into Differentiation and Development. Cells 2023; 12:cells12040635. [PMID: 36831302 PMCID: PMC9953855 DOI: 10.3390/cells12040635] [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: 12/14/2022] [Revised: 01/27/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
MicroRNAs (miRNAs), as small regulatory RNA molecules, are involved in gene expression at the post-transcriptional level. Hence, miRNAs contribute to gene regulation of various steps of different cell subsets' differentiation, maturation, and activation. The adaptive immune system arm, which exhibits the most specific immune responses, is also modulated by miRNAs. The generation and maturation of various T-cell subsets concomitant with B-cells is under precise regulation of miRNAs which function directly on the hallmark genes of each cell subset or indirectly through regulation of signaling pathway mediators and/or transcription factors involved in this maturation journey. In this review, we first discussed the origination process of common lymphocyte progenitors from hematopoietic stem cells, which further differentiate into various T-cell subsets under strict regulation of miRNAs and transcription factors. Subsequently, the differentiation of B-cells from common lymphocyte progenitors in bone marrow and periphery were discussed in association with a network of miRNAs and transcription factors.
Collapse
|
14
|
Studies on the role of non-coding RNAs in controlling the activity of T cells in asthma. Noncoding RNA Res 2023; 8:211-217. [PMID: 36865391 PMCID: PMC9972402 DOI: 10.1016/j.ncrna.2023.02.004] [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: 01/20/2023] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Bronchial asthma, commonly known as asthma, is a chronic inflammatory disease characterized by airway inflammation, increased responsiveness and changes in airway structure. T cells, particularly T helper cells, play a crucial role in the disease. Non-coding RNAs, which are RNAs that do not code for proteins, mainly include microRNAs, long non-coding RNAs, and circular RNAs, play a role in regulating various biological processes. Studies have shown that non-coding RNAs have an important role in the activation and transformation of T cells and other biological processes in asthma. The specific mechanisms and clinical applications are worth further examination. This article reviews the recent research on the role of microRNAs, long non-coding RNAs and circular RNAs in T cells in asthma.
Collapse
|
15
|
Airway epithelial ITGB4 deficiency induces airway remodeling in a mouse model. J Allergy Clin Immunol 2023; 151:431-446.e16. [PMID: 36243221 DOI: 10.1016/j.jaci.2022.09.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 08/25/2022] [Accepted: 09/16/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Airway epithelial cells (AECs) with impaired barrier function contribute to airway remodeling through the activation of epithelial-mesenchymal trophic units (EMTUs). Although the decreased expression of ITGB4 in AECs is implicated in the pathogenesis of asthma, how ITGB4 deficiency impacts airway remodeling remains obscure. OBJECTIVE This study aims to determine the effect of epithelial ITGB4 deficiency on the barrier function of AECs, asthma susceptibility, airway remodeling, and EMTU activation. METHODS AEC-specific ITGB4 conditional knockout mice (ITGB4-/-) were generated and an asthma model was employed by the sensitization and challenge of house dust mite (HDM). EMTU activation-related growth factors were examined in ITGB4-silenced primary human bronchial epithelial cells of healthy subjects after HDM stimulation. Dexamethasone, the inhibitors of JNK phosphorylation or FGF2 were administered for the identification of the molecular mechanisms of airway remodeling in HDM-exposed ITGB4-/- mice. RESULTS ITGB4 deficiency in AECs enhanced asthma susceptibility and airway remodeling by disrupting airway epithelial barrier function. Aggravated airway remodeling in HDM-exposed ITGB4-/- mice was induced through the enhanced activation of EMTU mediated by Src homology domain 2-containing protein tyrosine phosphatase 2/c-Jun N-terminal kinase/Jun N-terminal kinase-dependent transcription factor/FGF2 (SHP2/JNK/c-Jun/FGF2) signaling pathway, which was partially independent of airway inflammation. Both JNK and FGF2 inhibitors significantly inhibited the aggravated airway remodeling and EMTU activation in HDM-exposed ITGB4-/- mice. CONCLUSIONS Airway epithelial ITGB4 deficiency induces airway remodeling in a mouse model of asthma through enhanced EMTU activation that is regulated by the SHP2/JNK/c-Jun/FGF2 pathway.
Collapse
|
16
|
He S, Zhou J, Ma Y, Wang W, Yang J. MicroRNA-19a Inhibition Directly and Indirectly Ameliorates Th2 Airway Inflammation in Asthma by Targeting RUNX3. Inflammation 2023; 46:370-387. [PMID: 36112239 DOI: 10.1007/s10753-022-01739-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 12/07/2022]
Abstract
Disruption of T-cell differentiation is characteristic of airway inflammation in allergic asthma. How miR-19a works in asthma has not been completely elucidated. This study aimed to examine whether microRNA-19a regulates helper T-cell proliferation and to identify the factors involved and the underlying mechanisms. Our results showed that miR-19a levels were upregulated in parallel with a reduction in RUNX3 expression in a house dust mite (HDM)-induced murine model of asthma. A dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay showed that RUNX3 was a direct target of miR-19a. Inhibiting the expression of miR-19a attenuated inflammation and mucus production, induced Th1 cells, suppressed the Th2 inflammatory response, and repressed dendritic cell (DC) maturation by increasing RUNX3 expression in WT asthmatic mice but not RUNX3+/- mice. In vitro experiments revealed that miR-19a inhibition could target RUNX3 to induce Th1 polarization and inhibit Th2 polarization by directly acting on naïve CD4+ T cells or indirectly mediating the maturation and antigen-presenting abilities of DCs. These findings indicate that miR-19a directly and indirectly regulates immunoinflammatory responses in asthma by targeting RUNX3.
Collapse
Affiliation(s)
- Shaojun He
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Jingrun Zhou
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Ying Ma
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China
| | - Wei Wang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China.
| | - Jiong Yang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Donghu Road 169, Wuhan, 430071, Hubei, China.
| |
Collapse
|
17
|
Allegra A, Murdaca G, Gammeri L, Ettari R, Gangemi S. Alarmins and MicroRNAs, a New Axis in the Genesis of Respiratory Diseases: Possible Therapeutic Implications. Int J Mol Sci 2023; 24:ijms24021783. [PMID: 36675299 PMCID: PMC9861898 DOI: 10.3390/ijms24021783] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 01/18/2023] Open
Abstract
It is well ascertained that airway inflammation has a key role in the genesis of numerous respiratory pathologies, including asthma, chronic obstructive pulmonary disease, and acute respiratory distress syndrome. Pulmonary tissue inflammation and anti-inflammatory responses implicate an intricate relationship between local and infiltrating immune cells and structural pulmonary cells. Alarmins are endogenic proteins discharged after cell injury in the extracellular microenvironment. The purpose of our review is to highlight the alterations in respiratory diseases involving some alarmins, such as high mobility group box 1 (HMGB1) and interleukin (IL)-33, and their inter-relationships and relationships with genetic non-coding material, such as microRNAs. The role played by these alarmins in some pathophysiological processes confirms the existence of an axis composed of HMGB1 and IL-33. These alarmins have been implicated in ferroptosis, the onset of type 2 inflammation and airway alterations. Moreover, both factors can act on non-coding genetic material capable of modifying respiratory function. Finally, we present an outline of alarmins and RNA-based therapeutics that have been proposed to treat respiratory pathologies.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
| | - Giuseppe Murdaca
- Department of Internal Medicine, Ospedale Policlinico San Martino, 16132 Genoa, Italy
- Correspondence:
| | - Luca Gammeri
- Department of Clinical and Experimental Medicine, Unit and School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, Unit and School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
18
|
Advances and Highlights of miRNAs in Asthma: Biomarkers for Diagnosis and Treatment. Int J Mol Sci 2023; 24:ijms24021628. [PMID: 36675145 PMCID: PMC9862966 DOI: 10.3390/ijms24021628] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Asthma is a heterogeneous inflammatory disease of the airways that causes breathing difficulties, episodes of cough and wheezing, and in more severe cases can greatly diminish quality of life. Epigenetic regulation, including post-transcriptional mediation of microRNAs (miRNAs), is one of the mechanisms behind the development of the range of asthma phenotypes and endotypes. As in every other immune-mediated disease, miRNAs regulate the behavior of cells that shape the airway structure as well as those in charge of the defense mechanisms in the bronchi and lungs, controlling cell survival, growth, proliferation, and the ability of cells to synthesize and secrete chemokines and immune mediators. More importantly, miRNAs are molecules with chemical and biological properties that make them appropriate biomarkers for disease, enabling stratification of patients for optimal drug selection and thereby simplifying clinical management and reducing both the economic burden and need for critical care associated with the disease. In this review, we summarize the roles of miRNAs in asthma and describe how they regulate the mechanisms of the disease. We further describe the current state of miRNAs as biomarkers for asthma phenotyping, endotyping, and treatment selection.
Collapse
|
19
|
Putting the "mi" in omics: discovering miRNA biomarkers for pediatric precision care. Pediatr Res 2023; 93:316-323. [PMID: 35906312 PMCID: PMC9884316 DOI: 10.1038/s41390-022-02206-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 06/27/2022] [Indexed: 01/31/2023]
Abstract
In the past decade, growing interest in micro-ribonucleic acids (miRNAs) has catapulted these small, non-coding nucleic acids to the forefront of biomarker research. Advances in scientific knowledge have made it clear that miRNAs play a vital role in regulating cellular physiology throughout the human body. Perturbations in miRNA signaling have also been described in a variety of pediatric conditions-from cancer, to renal failure, to traumatic brain injury. Likewise, the number of studies across pediatric disciplines that pair patient miRNA-omics with longitudinal clinical data are growing. Analyses of these voluminous, multivariate data sets require understanding of pediatric phenotypic data, data science, and genomics. Use of machine learning techniques to aid in biomarker detection have helped decipher background noise from biologically meaningful changes in the data. Further, emerging research suggests that miRNAs may have potential as therapeutic targets for pediatric precision care. Here, we review current miRNA biomarkers of pediatric diseases and studies that have combined machine learning techniques, miRNA-omics, and patient health data to identify novel biomarkers and potential therapeutics for pediatric diseases. IMPACT: In the following review article, we summarized how recent developments in microRNA research may be coupled with machine learning techniques to advance pediatric precision care.
Collapse
|
20
|
Weidner J, Malmhäll C, Arabkari V, Barrett A, Boberg E, Ekerljung L, Rådinger M. The Serum/Glucocorticoid-Regulated Kinase 1 Is Targeted by miR-19a in CD4+ T Cells. Cells 2022; 12:cells12010133. [PMID: 36611927 PMCID: PMC9818172 DOI: 10.3390/cells12010133] [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: 10/08/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
The polarization of CD4+ T cells into different T helper subsets is an important process in many diseases, including asthma. Part of the adaptive immune system, T cells are responsible for propagating signals to alert and prime the immune system. MicroRNAs (miRNAs) are small non-coding RNAs that act on numerous targets in the cell to regulate a variety of cellular processes, including roles in T cell polarization. In this study, we aimed to identify genes dysregulated in peripheral blood mononuclear cells from individuals with asthma. Moreover, we sought to examine miRNAs that may regulate the candidate genes and explore their functional relationship. Utilizing a focused gene array, we identified the serum/glucocorticoid-regulated kinase 1 (SGK1) gene to be upregulated in circulating peripheral blood mononuclear cells, which included T cells, from individuals with asthma. Several miRNAs were bioinformatically identified to target SGK1, but miR-19a was the only screened candidate that negatively correlated to SGK1 expression. Further analysis of the miR-19a-SGK1 relationship showed a negative correlation in CD4+ T cells in situ and direct binding in vitro during T cell activation. Moreover, we observed a negative correlation of miR-19a and SGK1 during early type 2 polarization of CD4+ naïve human T cells. Thus, we suggest that miR-19a has a role in binding and regulating SGK1 transcript levels during T cell development.
Collapse
|
21
|
MicroRNAs in T Cell-Immunotherapy. Int J Mol Sci 2022; 24:ijms24010250. [PMID: 36613706 PMCID: PMC9820302 DOI: 10.3390/ijms24010250] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/06/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
MicroRNAs (miRNAs) act as master regulators of gene expression in homeostasis and disease. Despite the rapidly growing body of evidence on the theranostic potential of restoring miRNA levels in pre-clinical models, the translation into clinics remains limited. Here, we review the current knowledge of miRNAs as T-cell targeting immunotherapeutic tools, and we offer an overview of the recent advances in miRNA delivery strategies, clinical trials and future perspectives in RNA interference technologies.
Collapse
|
22
|
Zhipu N, Zitao H, Jichao S, Cuida M. Research advances in roles of microRNAs in nasal polyp. Front Genet 2022; 13:1043888. [PMID: 36506304 PMCID: PMC9732428 DOI: 10.3389/fgene.2022.1043888] [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: 09/14/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
MicroRNAs (miRNAs), a subset of endogenous RNAs highly conservative with short chains, play key regulatory role in the biological relevant events of the cells. Exosomes are extracellular vesicles like the plasma membrane components being able to deliver information molecules such as miRNA between cells and to regulate the fate of the target cells. The progression of chronic rhinosinusitis with nasal polyps (CRSwNP) is closely associated with significant alterations of miRNA levels in both cells and exosomes. RNA-binding proteins (RBPs) have been acknowledged to play important roles in intracellular miRNA transport to exosomes, and specific membrane proteins such as caveolin-1 critically involved in HNRNPA1 -mediated transport of miRNA to exosomes. Aberrant alteration in endogenous miRNA levels significantly contributes to the process of airway remodeling in the nasal tissue and to the occurrence and progression of inflammatory responses in CRSwNP. Exogenous miRNAs delivered via exosomes has also been shown to play an important role in activating macrophages or in regulating vascular permeability in the CRSwNP.This paper highlights the mechanism of RBP-mediated delivery of miRNAs to exosomes and the important contribution of endogenous miRNAs to the development of CRSwNP in response to inflammation and airway remodeling. Finally, we discuss the future research directions for regulation of the miRNAs to CRSwNP.Delivery of exogenous miRNAs by exosomes alters the endogenous miRNAs content in nasal mucosal epithelial cells or in associated inflammatory cells in the CRSwNP, and altered endogenous miRNAs affects the inflammatory response and airway remodeling, which then regulates the occurrence and progression of CRSwNP.RBPs and associated membrane proteins such as caveolin-1 may play a crucial role in the entry of exogenous miRNA into exosomes.
Collapse
Affiliation(s)
- Niu Zhipu
- Clinical Medicine, China-Japan Union Hospital of Jilin University Norman Bethune Third School of Jilin University, Changchun, China
| | - Huo Zitao
- Clinical Medicine, China-Japan Union Hospital of Jilin University Norman Bethune Third School of Jilin University, Changchun, China
| | - Sha Jichao
- Department of Otorhinolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University Norman Bethune Third School of Jilin University, Changchun, China,*Correspondence: Sha Jichao, ; Meng Cuida,
| | - Meng Cuida
- Department of Otorhinolaryngology Head and Neck Surgery, China-Japan Union Hospital of Jilin University Norman Bethune Third School of Jilin University, Changchun, China,*Correspondence: Sha Jichao, ; Meng Cuida,
| |
Collapse
|
23
|
Dölz M, Hasiuk M, Gagnon JD, Kornete M, Marone R, Bantug G, Kageyama R, Hess C, Ansel KM, Seyres D, Roux J, Jeker LT. Forced expression of the non-coding RNA miR-17∼92 restores activation and function in CD28-deficient CD4 + T cells. iScience 2022; 25:105372. [PMID: 36388982 PMCID: PMC9646923 DOI: 10.1016/j.isci.2022.105372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 08/12/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
CD28 provides the prototypical costimulatory signal required for productive T-cell activation. Known molecular consequences of CD28 costimulation are mostly based on studies of protein signaling molecules. The microRNA cluster miR-17∼92 is induced by T cell receptor stimulation and further enhanced by combined CD28 costimulation. We demonstrate that transgenic miR-17∼92 cell-intrinsically largely overcomes defects caused by CD28 deficiency. Combining genetics, transcriptomics, bioinformatics, and biochemical miRNA:mRNA interaction maps we empirically validate miR-17∼92 target genes that include several negative regulators of T cell activation. CD28-deficient T cells exhibit derepressed miR-17∼92 target genes during activation. CRISPR/Cas9-mediated ablation of the miR-17∼92 targets Pten and Nrbp1 in naive CD28-/- CD4+ T cells differentially increases proliferation and expression of the activation markers CD25 and CD44, respectively. Thus, we propose that miR-17∼92 constitutes a central mediator for T cell activation, integrating signals by the TCR and CD28 costimulation by dampening multiple brakes that prevent T cell activation.
Collapse
Affiliation(s)
- Marianne Dölz
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology & Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Marko Hasiuk
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology & Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| | - John D. Gagnon
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mara Kornete
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Romina Marone
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology & Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Glenn Bantug
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Robin Kageyama
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christoph Hess
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Department of Medicine – CITIID, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - K. Mark Ansel
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Denis Seyres
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology & Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Julien Roux
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Lukas T. Jeker
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology & Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| |
Collapse
|
24
|
The Role of Noncoding RNA in Airway Allergic Diseases through Regulation of T Cell Subsets. Mediators Inflamm 2022; 2022:6125698. [PMID: 36248190 PMCID: PMC9553461 DOI: 10.1155/2022/6125698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/31/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
Allergic rhinitis and asthma are common airway allergic diseases, the incidence of which has increased annually in recent years. The human body is frequently exposed to allergens and environmental irritants that trigger immune and inflammatory responses, resulting in altered gene expression. Mounting evidence suggested that epigenetic alterations were strongly associated with the progression and severity of allergic diseases. Noncoding RNAs (ncRNAs) are a class of transcribed RNA molecules that cannot be translated into polypeptides and consist of three major categories, microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs). Previous studies showed that ncRNAs were involved in the physiopathological mechanisms of airway allergic diseases and contributed to their occurrence and development. This article reviews the current state of understanding of the role of noncoding RNAs in airway allergic diseases, highlights the limitations of recent studies, and outlines the prospects for further research to facilitate the clinical translation of noncoding RNAs as therapeutic targets and biomarkers.
Collapse
|
25
|
Tang WW, Bauer KM, Barba C, Ekiz HA, O’Connell RM. miR-aculous new avenues for cancer immunotherapy. Front Immunol 2022; 13:929677. [PMID: 36248881 PMCID: PMC9554277 DOI: 10.3389/fimmu.2022.929677] [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: 04/27/2022] [Accepted: 08/18/2022] [Indexed: 01/25/2023] Open
Abstract
The rising toll of cancer globally necessitates ingenuity in early detection and therapy. In the last decade, the utilization of immune signatures and immune-based therapies has made significant progress in the clinic; however, clinical standards leave many current and future patients without options. Non-coding RNAs, specifically microRNAs, have been explored in pre-clinical contexts with tremendous success. MicroRNAs play indispensable roles in programming the interactions between immune and cancer cells, many of which are current or potential immunotherapy targets. MicroRNAs mechanistically control a network of target genes that can alter immune and cancer cell biology. These insights provide us with opportunities and tools that may complement and improve immunotherapies. In this review, we discuss immune and cancer cell-derived miRNAs that regulate cancer immunity and examine miRNAs as an integral part of cancer diagnosis, classification, and therapy.
Collapse
Affiliation(s)
- William W. Tang
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Kaylyn M. Bauer
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Cindy Barba
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Huseyin Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, İzmir, Turkey
| | - Ryan M. O’Connell
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
26
|
Jakwerth CA, Kitzberger H, Pogorelov D, Müller A, Blank S, Schmidt-Weber CB, Zissler UM. Role of microRNAs in type 2 diseases and allergen-specific immunotherapy. FRONTIERS IN ALLERGY 2022; 3:993937. [PMID: 36172292 PMCID: PMC9512106 DOI: 10.3389/falgy.2022.993937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/11/2022] [Indexed: 12/07/2022] Open
Abstract
MicroRNAs (miRs) have gained scientific attention due to their importance in the pathophysiology of allergic diseases as well as their potential as biomarkers in allergen-specific treatment options. Their function as post-transcriptional regulators, controlling various cellular processes, is of high importance since any single miR can target multiple mRNAs, often within the same signalling pathway. MiRs can alter dysregulated expression of certain cellular responses and contribute to or cause, but in some cases prevent or repress, the development of various diseases. In this review article, we describe current research on the role of specific miRs in regulating immune responses in epithelial cells and specialized immune cells in response to various stimuli, in allergic diseases, and regulation in the therapeutic approach of allergen-specific immunotherapy (AIT). Despite the fact that AIT has been used successfully as a causative treatment option since more than a century, very little is known about the mechanisms of regulation and its connections with microRNAs. In order to fill this gap, this review aims to provide an overview of the current knowledge.
Collapse
|
27
|
Wang J, Cheng Y. The interaction of hsa_circ_0002594 and eIF4A3 promotes T-helper 2 cell differentiation by the regulation of PTEN. Clin Exp Med 2022:10.1007/s10238-022-00862-9. [DOI: 10.1007/s10238-022-00862-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/06/2022] [Indexed: 11/28/2022]
|
28
|
Zheng M, Fan P, Yang P, Zheng J, Zhao D. Respiratory Syncytial Virus Nonstructural Protein 1 Promotes 5-Lipoxygenase via miR-19a-3p. J Immunol Res 2022; 2022:4086710. [PMID: 35637792 PMCID: PMC9146443 DOI: 10.1155/2022/4086710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/23/2021] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) infection can regulate the expression of a wide range of noncoding microRNAs (miRNAs), in which mir-19a-3p can participate in airway inflammatory response by regulating 5-lipoxygenase (5-LO) pathway. RSV nonstructural protein (NS) 1 is involved in the airway hyperresponsiveness during RSV infection. Methods The expression levels of miR-19a-3p and inflammatory signaling-related indicators were detected using quantitative real-time PCR and western blot analyses on the A549 cells transfected with NS1 expression plasmids (pNS1). The 5-LO-mediated inflammatory signaling pathway was assessed when the miR-19a-3p or 5-LO was inhibited. Results The immunofluorescence analysis showed that the plasmid-mediated NS1 protein was observed in both the cytoplasm and nucleus. The expression level of miR-19a-3p was significantly upregulated in the pNS1 or RSV-treated cells, which was reversed by the NS1 small interfering RNA. In addition, pNS1 also upregulated the expression of 5-LO, interleukin-5 (IL-5), and leukotriene B4 (LTB4), which was also significantly inhibited by the miR-19a-3p antagonists. The 5-LO inhibitor MK886 prevented the increase in the expression level of IL-5 induced by pNS1. Conclusions These results suggested that the RSV NS1 might play an important role in the pathogenesis of RSV by activating the 5-LO and subsequent inflammatory cytokines through miR-19a-3p.
Collapse
Affiliation(s)
- Mei Zheng
- Department of Pediatrics, Children's Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Panpan Fan
- Department of Pediatrics, Children's Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Pu Yang
- Department of Pediatrics, Children's Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Junwen Zheng
- Department of Pediatrics, Children's Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dongchi Zhao
- Department of Pediatrics, Children's Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
29
|
Jakwerth CA, Ordovas-Montanes J, Blank S, Schmidt-Weber CB, Zissler UM. Role of Respiratory Epithelial Cells in Allergic Diseases. Cells 2022; 11:1387. [PMID: 35563693 PMCID: PMC9105716 DOI: 10.3390/cells11091387] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/11/2022] [Accepted: 04/19/2022] [Indexed: 02/07/2023] Open
Abstract
The airway epithelium provides the first line of defense to the surrounding environment. However, dysfunctions of this physical barrier are frequently observed in allergic diseases, which are tightly connected with pro- or anti-inflammatory processes. When the epithelial cells are confronted with allergens or pathogens, specific response mechanisms are set in motion, which in homeostasis, lead to the elimination of the invaders and leave permanent traces on the respiratory epithelium. However, allergens can also cause damage in the sensitized organism, which can be ascribed to the excessive immune reactions. The tight interaction of epithelial cells of the upper and lower airways with local and systemic immune cells can leave an imprint that may mirror the pathophysiology. The interaction with effector T cells, along with the macrophages, play an important role in this response, as reflected in the gene expression profiles (transcriptomes) of the epithelial cells, as well as in the secretory pattern (secretomes). Further, the storage of information from past exposures as memories within discrete cell types may allow a tissue to inform and fundamentally alter its future responses. Recently, several lines of evidence have highlighted the contributions from myeloid cells, lymphoid cells, stromal cells, mast cells, and epithelial cells to the emerging concepts of inflammatory memory and trained immunity.
Collapse
Affiliation(s)
- Constanze A. Jakwerth
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA 02115, USA;
- Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| | - Carsten B. Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| | - Ulrich M. Zissler
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Member of the Immunology and Inflammation Initiative of the Helmholtz Association, 80802 Munich, Germany; (C.A.J.); (S.B.); (C.B.S.-W.)
| |
Collapse
|
30
|
Kim HJ, Park SO, Byeon HW, Eo JC, Choi JY, Tanveer M, Uyangaa E, Kim K, Eo SK. T cell-intrinsic miR-155 is required for Th2 and Th17-biased responses in acute and chronic airway inflammation by targeting several different transcription factors. Immunology 2022; 166:357-379. [PMID: 35404476 DOI: 10.1111/imm.13477] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/30/2022] [Accepted: 03/02/2022] [Indexed: 11/29/2022] Open
Abstract
Asthmatic airway inflammation is divided into two typical endotypes: Th2-mediated eosinophilic and Th1- or Th17-mediated neutrophilic airway inflammation. The miRNA miR-155 has well-documented roles in the regulation of adaptive T-cell responses and innate immunity. However, no specific cell-intrinsic role has yet been elucidated for miR-155 in T cells in the course of Th2-eosinophilic and Th17-neutrophilic airway inflammation using actual in vivo asthma models. Here, using conditional KO (miR155ΔCD4 cKO) mice that have the specific deficiency of miR-155 in T cells, we found that the specific deficiency of miR-155 in T cells resulted in fully suppressed Th2-type eosinophilic airway inflammation following acute allergen exposure, as well as greatly attenuated the Th17-type neutrophilic airway inflammation induced by repeated allergen exposure. Furthermore, miR-155 in T cells appeared to regulate the expression of several different target genes in the functional activation of CD4+ Th2 and Th17 cells. To be more precise, the deficiency of miR-155 in T cells enhanced the expression of c-Maf, SOCS1, Fosl2, and Jarid2 in the course of CD4+ Th2 cell activation, while C/EBPβ was highly enhanced in CD4+ Th17 cell activation in the absence of miR-155 expression. Conclusively, our data revealed that miR-155 could promote Th2 and Th17-mediated airway inflammation via the regulation of several different target genes, depending on the context of asthmatic diseases. Therefore, these results provide valuable insights in actual understanding of specific cell-intrinsic role of miR-155 in eosinophilic and neutrophilic airway inflammation for the development of fine-tune therapeutic strategies.
Collapse
Affiliation(s)
- Hyo Jin Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Seong Ok Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Hee Won Byeon
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Jun Cheol Eo
- Division of Biotechnology, College of Environmental & Biosource Science, Jeonbuk National University, Iksan, South Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Maryum Tanveer
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Erdenebelig Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| |
Collapse
|
31
|
Di Gioacchino M, Della Valle L, Allegra A, Pioggia G, Gangemi S. AllergoOncology: Role of immune cells and immune proteins. Clin Transl Allergy 2022; 12:e12133. [PMID: 35344301 PMCID: PMC8967267 DOI: 10.1002/clt2.12133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 12/21/2021] [Accepted: 02/18/2022] [Indexed: 12/13/2022] Open
Abstract
Background Immune cells and immune proteins play a pivotal role in host responses to pathogens, allergens and cancer. Understanding the crosstalk between allergic response and cancer, immune surveillance, immunomodulation, role of immunoglobulin E (IgE)‐mediated functions and help to develop novel therapeutic strategies. Allergy and oncology show two opposite scenarios: whereas immune tolerance is desired in allergy, it is detrimental in cancer. Aim The current review provides an update on the role of immune cells and immune proteins in allergy and cancer fields. Methods Authors investigated the role of relevant immunological markers and the correlation with cancer progression or cancer suppression. Results Activated immune cells such as macrophages ‘M1’, dendritic cells (DCs), innate lymphoid cells (ILC2), NK cells, Th1, follicular T helper cells (TFH), TCD8+, B lymphocytes and eosinophils have inhibitory effects on tumourigenesis, while tolerogenic cells such as macrophages ‘M2,’ tolerogenic DCs, ILC3, T and B regulatory lymphocytes appear to favour carcinogenesis. Mastocytes and alarmins can have both effects. RIgE antibodies and CCCL5 chemokine have an anticancer role, whereas IgG4, free light chains, Il‐10, TGF‐β, lipocalin‐2, CCL1 chemokine promote cancer progression. Fundamental is also the contribution of epigenetic changes regulated by the microRNA in cancer progression. Conclusion This knowledge represents the key to developing new anticancer therapies.
Collapse
Affiliation(s)
- Mario Di Gioacchino
- Center for Advanced Science and Technology, G. d'Annunzio University, Chieti, Italy.,IDA - Institute of Clinical Immunotherapy and Advanced Biological Treatments, Pescara, Italy
| | - Loredana Della Valle
- Center for Advanced Science and Technology, G. d'Annunzio University, Chieti, Italy.,IDA - Institute of Clinical Immunotherapy and Advanced Biological Treatments, Pescara, Italy
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood 'Gaetano Barresi', University of Messina, Messina, Italy
| | - Giovanni Pioggia
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), Messina, Italy
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School of Allergy and Clinical Immunology, and Operative Unit of Allergy and Clinical Immunology, University of Messina, Messina, Italy
| |
Collapse
|
32
|
Zhang X, Xu Z, Wen X, Huang G, Nian S, Li L, Guo X, Ye Y, Yuan Q. The onset, development and pathogenesis of severe neutrophilic asthma. Immunol Cell Biol 2022; 100:144-159. [PMID: 35080788 DOI: 10.1111/imcb.12522] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 12/02/2021] [Accepted: 01/23/2022] [Indexed: 12/12/2022]
Abstract
Bronchial asthma is divided into Th2 high, Th2 low and mixed types. The Th2 high type is dominated by eosinophils while the Th2 low type is divided into neutrophilic and paucigranulocytic types. Eosinophilic asthma has gained increased attention recently, and its pathogenesis and treatment are well understood. However, severe neutrophilic asthma requires more in-depth research because its pathogenesis is not well understood, and no effective treatment exists. This review looks at the advances made in asthma research, the pathogenesis of neutrophilic asthma, the mechanisms of progression to severe asthma, risk factors for asthma exacerbations, and biomarkers and treatment of neutrophilic asthma. The pathogenesis of neutrophilic asthma is further discussed from four aspects: Th17-type inflammatory response, inflammasomes, exosomes and microRNAs. This review provides direction for the mechanistic study, diagnosis and treatment of neutrophilic asthma. The treatment of neutrophilic asthma remains a significant challenge for clinical therapists and is an important area of future clinical research.
Collapse
Affiliation(s)
- Xingli Zhang
- Public Center of Experimental Technology, Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, School of Basic Medical Science of Southwest Medical University, Luzhou, Sichuan, China
| | - Zixi Xu
- Public Center of Experimental Technology, Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, School of Basic Medical Science of Southwest Medical University, Luzhou, Sichuan, China
| | - Xue Wen
- Department of Laboratory Medicine, The Affiliated Hospital of Southwest Medical University, Sichuan, China
| | - Guoping Huang
- Zigong Hospital of Woman and Children Healthcare, Sichuan, China
| | - Siji Nian
- Public Center of Experimental Technology, Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, School of Basic Medical Science of Southwest Medical University, Luzhou, Sichuan, China
| | - Lin Li
- Public Center of Experimental Technology, Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, School of Basic Medical Science of Southwest Medical University, Luzhou, Sichuan, China
| | - Xiyuan Guo
- Public Center of Experimental Technology, Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, School of Basic Medical Science of Southwest Medical University, Luzhou, Sichuan, China
| | - Yingchun Ye
- Public Center of Experimental Technology, Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, School of Basic Medical Science of Southwest Medical University, Luzhou, Sichuan, China
| | - Qing Yuan
- Public Center of Experimental Technology, Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, School of Basic Medical Science of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
33
|
Adamič N, Prpar Mihevc S, Blagus R, Kramarič P, Krapež U, Majdič G, Viel L, Hoffman AM, Bienzle D, Vengust M. Effect of intrabronchial administration of autologous adipose-derived mesenchymal stem cells on severe equine asthma. Stem Cell Res Ther 2022; 13:23. [PMID: 35063028 PMCID: PMC8777441 DOI: 10.1186/s13287-022-02704-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/04/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Severe equine asthma (SEA) is a common chronic respiratory disease and a significant health and well-being problem in horses. Current therapeutic strategies improve pulmonary function and clinical signs in some horses, but in the long-term, return to full athletic function appears to be rare. The aim of this study was to assess the safety and the effect of intrabronchial administration of adipose-derived mesenchymal stem cells (AD-MSC) on pulmonary inflammatory and clinical parameters in horses with SEA. METHODS This was a randomized controlled trial. Twenty adult horses diagnosed with SEA were randomly divided into two groups (n = 10), and treated either with a single intrabronchial application of autologous AD-MSC or oral dexamethasone for three weeks. A targeted clinical examination with determination of clinical score, maximal change in pleural pressure during the breathing cycle, and an endoscopic examination of the airways were performed at baseline and three weeks after treatment. Bronchoalveolar lavage fluid was analyzed cytologically, and IL-1β, IL-4, IL-8, IL-17, TNFα and IFNγ mRNA and protein concentrations were measured at baseline and three weeks. The horses were then monitored over one year for recurrence of SEA. A non-inferiority analysis and a linear mixed-effects model were performed to assess differences between treatments. RESULTS The non-inferiority of AD-MSC treatment was not established. However, AD-MSC administration significantly ameliorated the clinical score (P = 0.01), decreased the expression of IL-17 mRNA (P = 0.05) and IL-1β (P ≤ 0.001), IL-4 (P ≤ 0.001), TNFα (P = 0.02) protein levels, and had a positive long-term effect on SEA-associated clinical signs (P = 0.02). CONCLUSIONS Intrabronchial administration of AD-MSC had limited short-term anti-inflammatory effects but improved the clinical signs of SEA at one year.
Collapse
Affiliation(s)
- Neža Adamič
- Veterinary Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | | | - Rok Blagus
- Institute for Biostatistics and Medical Informatics, Faculty of Medicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Petra Kramarič
- Veterinary Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Uroš Krapež
- Veterinary Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Gregor Majdič
- Veterinary Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Laurent Viel
- Clinical Studies, University of Guelph, Guelph, ON, Canada
| | - Andrew M Hoffman
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dorothee Bienzle
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Modest Vengust
- Veterinary Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia.
| |
Collapse
|
34
|
Engeroff P, Vogel M. The Potential of Exosomes in Allergy Immunotherapy. Vaccines (Basel) 2022; 10:vaccines10010133. [PMID: 35062793 PMCID: PMC8780385 DOI: 10.3390/vaccines10010133] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 02/01/2023] Open
Abstract
Allergic diseases represent a global health and economic burden of increasing significance. The lack of disease-modifying therapies besides specific allergen immunotherapy (AIT) which is not available for all types of allergies, necessitates the study of novel therapeutic approaches. Exosomes are small endosome-derived vesicles delivering cargo between cells and thus allowing inter-cellular communication. Since immune cells make use of exosomes to boost, deviate, or suppress immune responses, exosomes are intriguing candidates for immunotherapy. Here, we review the role of exosomes in allergic sensitization and inflammation, and we discuss the mechanisms by which exosomes could potentially be used in immunotherapeutic approaches for the treatment of allergic diseases. We propose the following approaches: (a) Mast cell-derived exosomes expressing IgE receptor FcεRI could absorb IgE and down-regulate systemic IgE levels. (b) Tolerogenic exosomes could suppress allergic immune responses via induction of regulatory T cells. (c) Exosomes could promote TH1-like responses towards an allergen. (d) Exosomes could modulate IgE-facilitated antigen presentation.
Collapse
Affiliation(s)
- Paul Engeroff
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), F-75005 Paris, France;
| | - Monique Vogel
- Department of Immunology, University Hospital for Rheumatology, Immunology, and Allergology, 3010 Bern, Switzerland
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Correspondence:
| |
Collapse
|
35
|
Headland SE, Dengler HS, Xu D, Teng G, Everett C, Ratsimandresy RA, Yan D, Kang J, Ganeshan K, Nazarova EV, Gierke S, Wedeles CJ, Guidi R, DePianto DJ, Morshead KB, Huynh A, Mills J, Flanagan S, Hambro S, Nunez V, Klementowicz JE, Shi Y, Wang J, Bevers J, Ramirez-Carrozzi V, Pappu R, Abbas A, Vander Heiden J, Choy DF, Yadav R, Modrusan Z, Panettieri RA, Koziol-White C, Jester WF, Jenkins BJ, Cao Y, Clarke C, Austin C, Lafkas D, Xu M, Wolters PJ, Arron JR, West NR, Wilson MS. Oncostatin M expression induced by bacterial triggers drives airway inflammatory and mucus secretion in severe asthma. Sci Transl Med 2022; 14:eabf8188. [PMID: 35020406 DOI: 10.1126/scitranslmed.abf8188] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Sarah E Headland
- Immunology Discovery,Genentech Inc., South San Francisco, CA 94080, USA
| | - Hart S Dengler
- Immunology Discovery,Genentech Inc., South San Francisco, CA 94080, USA
| | - Daqi Xu
- Immunology Discovery,Genentech Inc., South San Francisco, CA 94080, USA
| | - Grace Teng
- Immunology Discovery,Genentech Inc., South San Francisco, CA 94080, USA
| | - Christine Everett
- Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA 94080, USA
| | | | - Donghong Yan
- Translational Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Jing Kang
- Translational Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Kirthana Ganeshan
- Immunology Discovery,Genentech Inc., South San Francisco, CA 94080, USA
| | | | - Sarah Gierke
- Center for Advanced Light Microscopy, Genentech Inc., South San Francisco, CA 94080, USA.,Pathology, Genentech Inc., South San Francisco, CA 94080, USA
| | | | - Riccardo Guidi
- Immunology Discovery,Genentech Inc., South San Francisco, CA 94080, USA
| | - Daryle J DePianto
- Immunology Discovery,Genentech Inc., South San Francisco, CA 94080, USA
| | | | - Alison Huynh
- Necropsy, Genentech Inc., South San Francisco, CA 94080, USA
| | - Jessica Mills
- Necropsy, Genentech Inc., South San Francisco, CA 94080, USA
| | - Sean Flanagan
- Necropsy, Genentech Inc., South San Francisco, CA 94080, USA
| | - Shannon Hambro
- Necropsy, Genentech Inc., South San Francisco, CA 94080, USA
| | - Victor Nunez
- Necropsy, Genentech Inc., South San Francisco, CA 94080, USA
| | | | - Yongchang Shi
- Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Jianyong Wang
- Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Jack Bevers
- Antibody Discovery, Genentech Inc., South San Francisco, CA 94080, USA
| | | | - Rajita Pappu
- Immunology Discovery,Genentech Inc., South San Francisco, CA 94080, USA
| | - Alex Abbas
- OMNI Bioinformatics, Genentech Inc., South San Francisco, CA 94080, USA
| | | | - David F Choy
- Biomarker Discovery OMNI, Genentech Inc., South San Francisco, CA 94080, USA
| | - Rajbharan Yadav
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc., South San Francisco, CA 94080, USA
| | - Zora Modrusan
- Molecular Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, State University of New Jersey, New Brunswick, NJ 08901, USA
| | - William F Jester
- Rutgers Institute for Translational Medicine and Science, State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular Translational Science, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Yi Cao
- OMNI Bioinformatics, Genentech Inc., South San Francisco, CA 94080, USA
| | - Christine Clarke
- OMNI Bioinformatics, Genentech Inc., South San Francisco, CA 94080, USA
| | - Cary Austin
- Pathology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Daniel Lafkas
- Immunology Discovery,Genentech Inc., South San Francisco, CA 94080, USA
| | - Min Xu
- Translational Immunology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Paul J Wolters
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Joseph R Arron
- Immunology Discovery,Genentech Inc., South San Francisco, CA 94080, USA
| | - Nathaniel R West
- Cancer Immunology Discovery, Genentech Inc., South San Francisco, CA 94080, USA
| | - Mark S Wilson
- Immunology Discovery,Genentech Inc., South San Francisco, CA 94080, USA
| |
Collapse
|
36
|
Liang J, Liu XH, Chen XM, Song XL, Li W, Huang Y. Emerging Roles of Non-Coding RNAs in Childhood Asthma. Front Pharmacol 2022; 13:856104. [PMID: 35656293 PMCID: PMC9152219 DOI: 10.3389/fphar.2022.856104] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Asthma is a chronic airway inflammatory disease in children characterized by airway inflammation, airway hyperresponsiveness and airway remodeling. Childhood asthma is usually associated with allergy and atopy, unlike adult asthma, which is commonly associated with obesity, smoking, etc. The pathogenesis and diagnosis of childhood asthma also remains more challenging than adult asthma, such as many diseases showing similar symptoms may coexist and be confused with asthma. In terms of the treatment, although most childhood asthma can potentially be self-managed and controlled with drugs, approximately 5-10% of children suffer from severe uncontrolled asthma, which carries significant health and socioeconomic burdens. Therefore, it is necessary to explore the pathogenesis of childhood asthma from a new perspective. Studies have revealed that non-coding RNAs (ncRNAs) are involved in the regulation of respiratory diseases. In addition, altered expression of ncRNAs in blood, and in condensate of sputum or exhalation affects the progression of asthma via regulating immune response. In this review, we outline the regulation and pathogenesis of asthma and summarize the role of ncRNAs in childhood asthma. We also hold promise that ncRNAs may be used for the development of biomarkers and support a new therapeutic strategy for childhood asthma.
Collapse
Affiliation(s)
- Juan Liang
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Xiao-Hua Liu
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Xue-Mei Chen
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Xiu-Ling Song
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Wen Li
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yuge Huang
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
37
|
Johansson K, Woodruff PG, Ansel KM. Regulation of airway immunity by epithelial miRNAs. Immunol Rev 2021; 304:141-153. [PMID: 34549450 PMCID: PMC9135676 DOI: 10.1111/imr.13028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023]
Abstract
The airway epithelium is essential to protect the host from inhaled pathogens and particles. It maintains immune homeostasis and mediates tissue repair after injury. Inflammatory diseases of the airways are associated with failure of epithelial functions, including loss of barrier integrity that results in increased tissue permeability and immune activation; excessive mucus secretion and impaired mucociliary clearance that leads to airflow obstruction and microbial overgrowth; and dysregulation of cellular signals that promotes inflammation and alters tissue structure and airway reactivity. MicroRNAs play crucial roles in mounting appropriate cellular responses to environmental stimuli and preventing disease, using a common machinery and mechanism to regulate gene expression in epithelial cells, immune cells of hematopoietic origin, and other cellular components of the airways. Respiratory diseases are accompanied by dramatic changes in epithelial miRNA expression that drive persistent immune dysregulation. In this review, we discuss responses of the epithelium that promote airway immunopathology, with a focus on miRNAs that contribute to the breakdown of essential epithelial functions. We emphasize the emerging role of miRNAs in regulation of epithelial responses in respiratory health and their value as diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Kristina Johansson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of California, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| | - Prescott G. Woodruff
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of California, San Francisco, California, USA
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - K. Mark Ansel
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| |
Collapse
|
38
|
Borbet TC, Hines MJ, Koralov SB. MicroRNA regulation of B cell receptor signaling. Immunol Rev 2021; 304:111-125. [PMID: 34523719 PMCID: PMC8616848 DOI: 10.1111/imr.13024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 12/19/2022]
Abstract
B lymphocytes play a central role in host immune defense. B cell receptor (BCR) signaling regulates survival, proliferation, and differentiation of B lymphocytes. Signaling through the BCR signalosome is a multi-component cascade that is tightly regulated and is important in the coordination of B cell differentiation and function. At different stages of development, B cells that have BCRs recognizing self are eliminated to prevent autoimmunity. microRNAs (miRNAs) are small single-stranded non-coding RNAs that contribute to post-transcriptional regulation of gene expression and have been shown to orchestrate cell fate decisions through the regulation of lineage-specific transcriptional profiles. Studies have identified miRNAs to be crucial for B cell development in the bone marrow and their subsequent population of the peripheral immune system. In this review, we focus on the role of miRNAs in the regulation of BCR signaling as it pertains to B lymphocyte development and function. In particular, we discuss the most recent studies describing the role of miRNAs in the regulation of both early B cell development and peripheral B cell responses and examine the ways by which miRNAs regulate signal downstream of B cell antigen receptor to prevent aberrant activation and autoimmunity.
Collapse
Affiliation(s)
- Timothy C. Borbet
- New York University School of Medicine, Department of Pathology, New York, NY 10016
| | - Marcus J. Hines
- New York University School of Medicine, Department of Pathology, New York, NY 10016
| | - Sergei B. Koralov
- New York University School of Medicine, Department of Pathology, New York, NY 10016
| |
Collapse
|
39
|
Peng M, Ren J, Jing Y, Jiang X, Xiao Q, Huang J, Tao Y, Lei L, Wang X, Yang Z, Yang Z, Zhan Q, Lin C, Jin G, Zhang X, Zhang L. Tumour-derived small extracellular vesicles suppress CD8+ T cell immune function by inhibiting SLC6A8-mediated creatine import in NPM1-mutated acute myeloid leukaemia. J Extracell Vesicles 2021; 10:e12168. [PMID: 34807526 PMCID: PMC8607980 DOI: 10.1002/jev2.12168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/11/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukaemia (AML) carrying nucleophosmin (NPM1) mutations has been defined as a distinct entity of acute leukaemia. Despite remarkable improvements in diagnosis and treatment, the long-term outcomes for this entity remain unsatisfactory. Emerging evidence suggests that leukaemia, similar to other malignant diseases, employs various mechanisms to evade killing by immune cells. However, the mechanism of immune escape in NPM1-mutated AML remains unknown. In this study, both serum and leukemic cells from patients with NPM1-mutated AML impaired the immune function of CD8+ T cells in a co-culture system. Mechanistically, leukemic cells secreted miR-19a-3p into the tumour microenvironment (TME) via small extracellular vesicles (sEVs), which was controlled by the NPM1-mutated protein/CCCTC-binding factor (CTCF)/poly (A)-binding protein cytoplasmic 1 (PABPC1) signalling axis. sEV-related miR-19a-3p was internalized by CD8+ T cells and directly repressed the expression of solute-carrier family 6 member 8 (SLC6A8; a creatine-specific transporter) to inhibit creatine import. Decreased creatine levels can reduce ATP production and impair CD8+ T cell immune function, leading to immune escape by leukemic cells. In summary, leukemic cell-derived sEV-related miR-19a-3p confers immunosuppression to CD8+ T cells by targeting SLC6A8-mediated creatine import, indicating that sEV-related miR-19a-3p might be a promising therapeutic target for NPM1-mutated AML.
Collapse
Affiliation(s)
- Meixi Peng
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Jun Ren
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Yipei Jing
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Xueke Jiang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Qiaoling Xiao
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Junpeng Huang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Yonghong Tao
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Li Lei
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Xin Wang
- Department of HematologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zailin Yang
- Department of Clinical Laboratory The Third Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Chongqing University Cancer HospitalChongqingChina
| | - Zesong Yang
- Department of HematologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Qian Zhan
- The Center for Clinical Molecular Medical detectionThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Can Lin
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| | - Guoxiang Jin
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Xian Zhang
- Immunology ProgramMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Ling Zhang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of EducationSchool of Laboratory MedicineChongqing Medical UniversityChongqingChina
| |
Collapse
|
40
|
Fang SB, Zhou ZR, Peng YQ, Liu XQ, He BX, Chen DH, Chen D, Fu QL. Plasma EVs Display Antigen-Presenting Characteristics in Patients With Allergic Rhinitis and Promote Differentiation of Th2 Cells. Front Immunol 2021; 12:710372. [PMID: 34691024 PMCID: PMC8531542 DOI: 10.3389/fimmu.2021.710372] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 09/20/2021] [Indexed: 12/23/2022] Open
Abstract
Background Allergic rhinitis (AR) is characterized by IgE-mediated mucosa response after exposure to allergens. Extracellular vesicles (EVs) are nano-size vesicles containing biological cargos for intercellular communications. However, the role of plasma EVs in pathogenesis of AR remains largely unknown. Methods Plasma EVs from patients with AR were isolated, quantified, and characterized. The expression of Der p 1 and antigen-presenting molecules on EVs was determined by Western blot, flow cytometry, or ELISA. PKH26- and CFSE (carboxyfluorescein succinimidyl ester)-stained AR-EVs were used to determine the uptake of EVs by CD4+T cells and their effects on CD4+T cell proliferation, respectively. Results Plasma EVs in healthy control (HC) and AR patients were similar in the concentration of particles, expression for specific EV markers, and both had structural lipid bilayer. However, the levels of Der p 1 on plasma EVs from both mild and moderate-severe AR patients were significantly higher than that on HC. The levels of antigen-presenting molecules on plasma EVs were similar from three subjects. Moreover, levels of Der p 1 on EVs in plasma, but not nasal secretion, were significantly associated with the symptom score of AR patients and level of plasma IL-13. Additionally, plasma EVs from patients with AR promoted the development of Th2 cells, while no effect was found on CD4+ T-cell proliferation. Conclusions Plasma EVs derived from patients with AR exhibited antigen-presenting characteristics and promoted differentiation of Th2 cells, thus providing novel understanding of the pathogenesis of AR.
Collapse
Affiliation(s)
- Shu-Bin Fang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Rou Zhou
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ya-Qi Peng
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Qing Liu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bi-Xin He
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - De-Hua Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dong Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing-Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
41
|
Role of Epigenetics in the Pathogenesis, Treatment, Prediction, and Cellular Transformation of Asthma. Mediators Inflamm 2021; 2021:9412929. [PMID: 34566492 PMCID: PMC8457970 DOI: 10.1155/2021/9412929] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Asthma is a mysterious disease with heterogeneity in etiology, pathogenesis, and clinical phenotypes. Although ongoing studies have provided a better understanding of asthma, its natural history, progression, pathogenesis, diversified phenotypes, and even the exact epigenetic linkage between childhood asthma and adult-onset/old age asthma remain elusive in many aspects. Asthma heritability has been established through genetic studies, but genetics is not the only influencing factor in asthma. The increasing incidence and some unsolved queries suggest that there may be other elements related to asthma heredity. Epigenetic mechanisms link genetic and environmental factors with developmental trajectories in asthma. This review provides an overview of asthma epigenetics and its components, including several epigenetic studies on asthma, and discusses the epigenetic linkage between childhood asthma and adult-onset/old age asthma. Studies involving asthma epigenetics present valuable novel approaches to solve issues related to asthma. Asthma epigenetic research guides us towards gene therapy and personalized T cell therapy, directs the discovery of new therapeutic agents, predicts long-term outcomes in severe cases, and is also involved in the cellular transformation of childhood asthma to adult-onset/old age asthma.
Collapse
|
42
|
Cho S, Dong J, Lu LF. Cell-intrinsic and -extrinsic roles of miRNAs in regulating T cell immunity. Immunol Rev 2021; 304:126-140. [PMID: 34549446 DOI: 10.1111/imr.13029] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 12/14/2022]
Abstract
T cells are crucial to generate an effective response against numerous invading microbial pathogens and play a pivotal role in tumor surveillance and elimination. However, unwanted T cell activation can also lead to deleterious immune-mediated inflammation and tissue damage. To ensure that an optimal T cell response can be established, each step, beginning from T cell development in the thymus to their activation and function in the periphery, is tightly regulated by many transcription factors and epigenetic regulators including microRNAs (miRNAs). Here, we first summarize recent progress in identifying major immune regulatory miRNAs in controlling the differentiation and function of distinct T cell subsets. Moreover, as emerging evidence has demonstrated that miRNAs can impact T cell immunity through targeting both immune- and non-immune cell populations that T cells closely interact with, the T cell-extrinsic role of miRNAs in regulating different aspects of T cell biology is also addressed. Finally, we discuss the complex nature of miRNA-mediated control of T cell immunity and highlight important questions that remain to be further investigated.
Collapse
Affiliation(s)
- Sunglim Cho
- Division of Biological Sciences, University of California, La Jolla, California, USA
| | - Jiayi Dong
- Division of Biological Sciences, University of California, La Jolla, California, USA
| | - Li-Fan Lu
- Division of Biological Sciences, University of California, La Jolla, California, USA.,Moores Cancer Center, University of California, La Jolla, California, USA.,Center for Microbiome Innovation, University of California, La Jolla, California, USA
| |
Collapse
|
43
|
Jakwerth CA, Chaker AM, Guerth F, Oelsner M, Pechtold L, Zur Bonsen LS, Ullmann JT, Krauss-Etschmann S, Erb A, Kau J, Plaschke M, Winkler M, Kurz A, Kloss A, Esser-von Bieren J, Schmidt-Weber CB, Zissler UM. Sputum microRNA-screening reveals Prostaglandin EP3 receptor as selective target in allergen-specific immunotherapy. Clin Exp Allergy 2021; 51:1577-1591. [PMID: 34514658 DOI: 10.1111/cea.14013] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/08/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Several microRNAs (miRs) have been described as potential biomarkers in liquid biopsies and in the context of allergic asthma, while therapeutic effects on the airway expression of miRs remain elusive. In this study, we investigated epigenetic miR-associated mechanisms in the sputum of grass pollen-allergic patients with and without allergen-specific immunotherapy (AIT). METHODS Induced sputum samples of healthy controls (HC), AIT-treated and -untreated grass pollen-allergic rhinitis patients with (AA) and without asthma (AR) were profiled using miR microarray and whole-transcriptome microarray analysis of the same samples. miR targets were predicted in silico and used to identify inverse regulation. Local PGE2 levels were measured using ELISA. RESULTS Two hundred and fifty nine miRs were upregulated in the sputum of AA patients compared with HC, while only one was downregulated. The inverse picture was observed in induced sputum of AIT-treated patients: while 21 miRs were downregulated, only 4 miRs were upregulated in asthmatics upon AIT. Of these 4 miRs, miR-3935 stood out, as its predicted target PTGER3, the prostaglandin EP3 receptor, was downregulated in treated AA patients compared with untreated. The levels of its ligand PGE2 in the sputum supernatants of these samples were increased in allergic patients, especially asthmatics, and downregulated after AIT. Finally, local PGE2 levels correlated with ILC2 frequencies, secreted sputum IL-13 levels, inflammatory cell load, sputum eosinophils and symptom burden. CONCLUSIONS While profiling the sputum of allergic patients for novel miR expression patterns, we uncovered an association between miR-3935 and its predicted target gene, the prostaglandin E3 receptor, which might mediate AIT effects through suppression of the PGE2 -PTGER3 axis.
Collapse
Affiliation(s)
- Constanze A Jakwerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Adam M Chaker
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Ferdinand Guerth
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Madlen Oelsner
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Lisa Pechtold
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Lynn S Zur Bonsen
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Julia T Ullmann
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Susanne Krauss-Etschmann
- Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts University Kiel, Kiel, Germany
| | - Anna Erb
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Josephine Kau
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Mirjam Plaschke
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Marlene Winkler
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Alexandra Kurz
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Antonia Kloss
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany.,Department of Otorhinolaryngology, TUM School of Medicine, Technical University Munich, Klinikum rechts der Isar, Munich, Germany
| | - Julia Esser-von Bieren
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Ulrich M Zissler
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM), Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| |
Collapse
|
44
|
Xueyuan H, Qianru C, Zhaoyi L, Dayong T, Yu W, Yimei C, Shu L. Transcriptome analysis reveals that hydrogen sulfide exposure suppresses cell proliferation and induces apoptosis through ciR-PTPN23/miR-15a/E2F3 signaling in broiler thymus. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 284:117466. [PMID: 34062439 DOI: 10.1016/j.envpol.2021.117466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 06/12/2023]
Abstract
The immune organs, like thymus, are one of the targets of hydrogen sulfide (H2S). Previously we reported that H2S induced the differential expression of mRNAs that implicating apoptosis in thymus, however, the roles of noncoding RNAs (ncRNAs) in H2S-induced thymus injury are still unknown. Pollution gases could alter the expression of ncRNAs, which have been shown to play important roles in many physiological and pathophysiological processes, including immune activity. This study revealed that H2S exposure induced 9 differentially expressed circRNAs and 15 differentially expressed miRNAs in chicken thymus. Furthermore, the circRNA - miRNA - mRNA network was constructed. We discovered that circR-PTPN23 - miR-15a - E2F3 was involved in the cell cycle and apoptosis. Further, an in vitro H2S exposure model was established using HD11 cell line and demonstrated that H2S suppressed cell proliferation and induced apoptosis. Moreover, ciR-PTPN23 and E2F3 were downregulated, but miR-15a was upregulated in both the thymus and HD11 cell line after H2S exposure. Bioinformatics analysis revealed that ciR-PTPN23 directly bound to miR-15a and that E2F3 was the target gene of miR-15a. Knocking down ciR-PTPN23 suppressed HD11 proliferation and caused G1 arrest and apoptosis, however, this phenomenon could be partially reversed by ciR-PTPN23 overexpression or miR-15a silencing. In summary, the ciR-PTPN23 - miR-15a - E2F3 axis was involved in H2S-induced cell proliferation suppression and apoptosis.
Collapse
Affiliation(s)
- Hu Xueyuan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chi Qianru
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Liu Zhaoyi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Tao Dayong
- College of Animal Science, Tarim University, Alar, 843300, China
| | - Wang Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Cong Yimei
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China
| | - Li Shu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
45
|
miRNomic Signature in Very Low Birth-Weight Neonates Discriminates Late-Onset Gram-Positive Sepsis from Controls. Diagnostics (Basel) 2021; 11:diagnostics11081389. [PMID: 34441323 PMCID: PMC8391178 DOI: 10.3390/diagnostics11081389] [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] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/02/2022] Open
Abstract
Background and Objectives. Neonatal sepsis is a serious condition with a high rate of mortality and morbidity. Currently, the gold standard for sepsis diagnosis is a positive blood culture, which takes 48–72 h to yield results. We hypothesized that identifying differentially expressed miRNA pattern in neonates with late-onset Gram-positive sepsis would help with an earlier diagnosis and therapy. Methods. This is a prospective observational study in newborn infants with late-onset Gram positive bacterial sepsis and non-septic controls. Complementary to blood culture, an aliquot of 0.5 mL of blood was used to determine small non-coding RNA expression profiling using the GeneChip miRNA 4.0 Array. Results. A total of 11 very low birth-weight neonates with late-onset Gram-positive sepsis and 16 controls were analyzed. Further, 217 differentially expressed miRNAs were obtained between both groups. Subsequently, a combined analysis was performed with these miRNAs and 4297 differentially expressed genes. We identified 33 miRNAs that regulate our mRNAs, and the most relevant biological processes are associated with the immune system and the inflammatory response. Conclusions. The miRNA profiling in very low birth-weight neonates distinguishes late-onset Gram-positive sepsis versus control neonates.
Collapse
|
46
|
Magnaye KM, Naughton KA, Huffman J, Hogarth DK, Naureckas ET, White SR, Ober C. A-to-I editing of miR-200b-3p in airway cells is associated with moderate-to-severe asthma. Eur Respir J 2021; 58:13993003.03862-2020. [PMID: 33446603 DOI: 10.1183/13993003.03862-2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/19/2020] [Indexed: 11/05/2022]
Abstract
BACKGROUND Asthma is a chronic lung disease characterised by persistent airway inflammation. Altered microRNA (miRNA)-mediated gene silencing in bronchial epithelial cells (BECs) has been reported in asthma, yet adenosine deaminase acting on RNA (ADAR)-mediated miRNA editing in asthma remains unexplored. METHODS We first identified adenosine to inosine (A-to-I) edited sites in miRNAs in BECs from 142 adult asthma cases and controls. A-to-I edited sites were tested for associations with asthma severity and clinical measures of asthma. Paired RNA sequencing data were used to perform pathway enrichments and test for associations with bioinformatically predicted target genes of the unedited and edited miRNAs. RESULTS Of 19 A-to-I edited sites detected in these miRNAs, one site at position 5 of miR-200b-3p was edited less frequently in cases compared with controls (pcorrected=0.013), and especially compared with cases with moderate (pcorrected=0.029) and severe (pcorrected=3.9×10-4), but not mild (pcorrected=0.38), asthma. Bioinformatic prediction revealed 232 target genes of the edited miR-200b-3p, which were enriched for both interleukin-4 and interferon-γ signalling pathways, and included the SOCS1 (suppressor of cytokine signalling 1) gene. SOCS1 was more highly expressed in moderate (pcorrected=0.017) and severe (pcorrected=5.4×10-3) asthma cases compared with controls. Moreover, both miR-200b-3p editing and SOCS1 were associated with bronchoalveolar lavage eosinophil levels. CONCLUSIONS Reduced A-to-I editing of position 5 of miR-200b-3p in lower airway cells from moderate-to-severe asthmatic subjects may lead to overexpression of SOCS1 and impaired cytokine signalling. We propose ADAR-mediated editing as an epigenetic mechanism contributing to features of moderate-to-severe asthma in adulthood.
Collapse
Affiliation(s)
- Kevin M Magnaye
- Dept of Human Genetics, University of Chicago, Chicago, IL, USA.,These two authors contributed equally to this article as lead authors and supervised the work
| | | | - Janel Huffman
- Dept of Human Genetics, University of Chicago, Chicago, IL, USA
| | - D Kyle Hogarth
- Dept of Medicine, University of Chicago, Chicago, IL, USA
| | | | - Steven R White
- Dept of Medicine, University of Chicago, Chicago, IL, USA
| | - Carole Ober
- Dept of Human Genetics, University of Chicago, Chicago, IL, USA .,These two authors contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
47
|
Brown IK, Dyjack N, Miller MM, Krovi H, Rios C, Woolaver R, Harmacek L, Tu TH, O’Connor BP, Danhorn T, Vestal B, Gapin L, Pinilla C, Seibold MA, Scott-Browne J, Santos RG, Reinhardt RL. Single cell analysis of host response to helminth infection reveals the clonal breadth, heterogeneity, and tissue-specific programming of the responding CD4+ T cell repertoire. PLoS Pathog 2021; 17:e1009602. [PMID: 34106992 PMCID: PMC8216541 DOI: 10.1371/journal.ppat.1009602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 06/21/2021] [Accepted: 05/01/2021] [Indexed: 12/30/2022] Open
Abstract
The CD4+ T cell response is critical to host protection against helminth infection. How this response varies across different hosts and tissues remains an important gap in our understanding. Using IL-4-reporter mice to identify responding CD4+ T cells to Nippostrongylus brasiliensis infection, T cell receptor sequencing paired with novel clustering algorithms revealed a broadly reactive and clonally diverse CD4+ T cell response. While the most prevalent clones and clonotypes exhibited some tissue selectivity, most were observed to reside in both the lung and lung-draining lymph nodes. Antigen-reactivity of the broader repertoires was predicted to be shared across both tissues and individual mice. Transcriptome, trajectory, and chromatin accessibility analysis of lung and lymph-node repertoires revealed three unique but related populations of responding IL-4+ CD4+ T cells consistent with T follicular helper, T helper 2, and a transitional population sharing similarity with both populations. The shared antigen reactivity of lymph node and lung repertoires combined with the adoption of tissue-specific gene programs allows for the pairing of cellular and humoral responses critical to the orchestration of anti-helminth immunity. Using various “omic” approaches, the CD4+ T cell receptor (TCR) repertoire was explored after primary helminth infection. Infection generated a broadly reactive and clonally diverse CD4+ T cell response with the most prevalent clonotypes and predicted antigen specificities residing in both the lung and lung-draining lymph nodes. Tissue-specific programming of responding CD4+ T cells directed the establishment of committed Tfh and Th2 cells, both critical for driving distinct hallmarks of type-2 inflammation. These datasets help to explore the diverse yet tissue-specific nature of anti-helminth immunity.
Collapse
Affiliation(s)
- Ivy K. Brown
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Nathan Dyjack
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, United States of America
| | - Mindy M. Miller
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Harsha Krovi
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Cydney Rios
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, United States of America
| | - Rachel Woolaver
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Laura Harmacek
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, United States of America
| | - Ting-Hui Tu
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, United States of America
| | - Brian P. O’Connor
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, United States of America
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America
| | - Thomas Danhorn
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, United States of America
| | - Brian Vestal
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, United States of America
| | - Laurent Gapin
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Clemencia Pinilla
- Florida International University, Port Saint Lucie, Florida, United States of America
| | - Max A. Seibold
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, United States of America
- Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - James Scott-Browne
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, United States of America
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado, United States of America
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Radleigh G. Santos
- Department of Mathematics, Nova Southeastern University, Fort Lauderdale, Florida, United States of America
| | - R. Lee Reinhardt
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, United States of America
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
48
|
Bersimbaev R, Aripova A, Bulgakova O, Kussainova А, Akparova A, Izzotti A. The Plasma Levels of hsa-miR-19b-3p, hsa-miR-125b-5p and hsa-miR-320c in Patients with Asthma, COPD and Asthma-COPD Overlap Syndrome (ACOS). Microrna 2021; 10:130-138. [PMID: 34151771 DOI: 10.2174/2211536610666210609142859] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 03/04/2021] [Accepted: 03/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Bronchial Asthma (BA) and Chronic Obstructive Pulmonary Disease (COPD) are chronic airway inflammation diseases. In recent years, patients with signs of both BA and COPD have been assigned to a separate group as Asthma-COPD Overlap Syndrome (ACOS). Free-circulating plasma microRNAs are considered as potential biomarkers of pulmonary diseases, including BA, COPD and ACOS. OBJECTIVE This study aimed to investigate the expression level of free-circulating plasma microRNAs hsa-miR-19b-3p, hsa-miR-125b-5p and hsa-miR-320c in patients with BA, COPD and ACOS for the detection and validation of new microRNAs as biomarkers for chronic lung diseases. METHODS The relative expression levels of 720 microRNAs were evaluated by Real Time-Polymerase Chain Reaction (RT-PCR) in patients with COPD and BA. Three upregulated microRNAs (hsa-miR-19b-3p, hsa-miR-125b-5p and hsa-miR-320c) were selected for further study. The obtained data was analyzed using the microRNA PCR Array Data Analysis tool. The sensitivity and specificity were estimated using the area under the Receiver Operating Characteristics curve (ROC). RESULTS The expression level of free-circulating hsa-miR-19b-3p was decreased in the blood plasma of patients with BA and ACOS, and increased in patients with COPD. hsa-miR-125b-5p was downregulated in the blood plasma of patients with COPD, and upregulated in patients with BA and ACOS. hsa-miR-320c was downregulated in the blood plasma of patients with BA, and upregulated in patients with COPD and ACOS. The ROC curves of patients with BA for hsa-miR-19b-3p, patients with ACOS for hsa-miR-125b-5p and patients with COPD for hsa-miR-320c revealed the probability of them as valuable biomarkers with AUCs of 0.824, 0.825, and 0.855, respectively. CONCLUSION Our study revealed three promising biomarkers for the diagnosis of COPD, BA and ACOS.
Collapse
Affiliation(s)
- Rakhmetkazhy Bersimbaev
- Department of General Biology and Genomics, Faculty of Natural Sciences, L.N. Gumilyov Eurasian National University, Nur-Sultan, Kazakhstan
| | - Akmaral Aripova
- Department of General Biology and Genomics, Faculty of Natural Sciences, L.N. Gumilyov Eurasian National University, Nur-Sultan, Kazakhstan
| | - Olga Bulgakova
- Department of General Biology and Genomics, Faculty of Natural Sciences, L.N. Gumilyov Eurasian National University, Nur-Sultan, Kazakhstan
| | - Аssya Kussainova
- Department of General Biology and Genomics, Faculty of Natural Sciences, L.N. Gumilyov Eurasian National University, Nur-Sultan, Kazakhstan
| | - Almira Akparova
- Department of General Biology and Genomics, Faculty of Natural Sciences, L.N. Gumilyov Eurasian National University, Nur-Sultan, Kazakhstan
| | - Alberto Izzotti
- Department of Health Sciences, University of Genoa, I-16132 Genoa, Italy
| |
Collapse
|
49
|
Zhang K, Feng Y, Liang Y, Wu W, Chang C, Chen D, Chen S, Gao J, Chen G, Yi L, Cheng D, Zhen G. Epithelial miR-206 targets CD39/extracellular ATP to upregulate airway IL-25 and TSLP in type 2-high asthma. JCI Insight 2021; 6:148103. [PMID: 33945508 PMCID: PMC8262281 DOI: 10.1172/jci.insight.148103] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/28/2021] [Indexed: 02/05/2023] Open
Abstract
The epithelial cell–derived cytokines IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) initiate type 2 inflammation in allergic diseases, including asthma. However, the signaling pathway regulating these cytokines expression remains elusive. Since microRNAs are pivotal regulators of gene expression, we profiled microRNA expression in bronchial epithelial brushings from type 2–low and type 2–high asthma patients. miR-206 was the most highly expressed epithelial microRNA in type 2–high asthma relative to type 2–low asthma but was downregulated in both subsets compared with healthy controls. CD39, an ectonucleotidase degrading ATP, was a target of miR-206 and upregulated in asthma. Allergen-induced acute extracellular ATP accumulation led to miR-206 downregulation and CD39 upregulation in human bronchial epithelial cells, forming a feedback loop to eliminate excessive ATP. Airway ATP levels were markedly elevated and strongly correlated with IL-25 and TSLP expression in asthma patients. Intriguingly, airway miR-206 antagonism increased Cd39 expression; reduced ATP accumulation; suppressed IL-25, IL-33, and Tslp expression and group 2 innate lymphoid cell expansion; and alleviated type 2 inflammation in a mouse model of allergic airway inflammation. In contrast, airway miR-206 overexpression had opposite effects. Overall, epithelial miR-206 upregulates airway IL-25 and TSLP expression by targeting the CD39–extracellular ATP axis, which represents a potentially novel therapeutic target in type 2–high asthma.
Collapse
Affiliation(s)
- Kan Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, and National Clinical Research Center for Respiratory Diseases, Wuhan, China.,Department of Respiratory and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yuchen Feng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, and National Clinical Research Center for Respiratory Diseases, Wuhan, China
| | - Yuxia Liang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, and National Clinical Research Center for Respiratory Diseases, Wuhan, China
| | - Wenliang Wu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, and National Clinical Research Center for Respiratory Diseases, Wuhan, China
| | - Chenli Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, and National Clinical Research Center for Respiratory Diseases, Wuhan, China
| | - Dian Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, and National Clinical Research Center for Respiratory Diseases, Wuhan, China
| | - Shengchong Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, and National Clinical Research Center for Respiratory Diseases, Wuhan, China
| | - Jiali Gao
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, and National Clinical Research Center for Respiratory Diseases, Wuhan, China
| | - Gongqi Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, and National Clinical Research Center for Respiratory Diseases, Wuhan, China
| | - Lingling Yi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, and National Clinical Research Center for Respiratory Diseases, Wuhan, China
| | - Dan Cheng
- Department of Respiratory and Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guohua Zhen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Respiratory Diseases, National Health Commission of People's Republic of China, and National Clinical Research Center for Respiratory Diseases, Wuhan, China
| |
Collapse
|
50
|
Weidner J, Bartel S, Kılıç A, Zissler UM, Renz H, Schwarze J, Schmidt‐Weber CB, Maes T, Rebane A, Krauss‐Etschmann S, Rådinger M. Spotlight on microRNAs in allergy and asthma. Allergy 2021; 76:1661-1678. [PMID: 33128813 PMCID: PMC8246745 DOI: 10.1111/all.14646] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/16/2020] [Accepted: 10/25/2020] [Indexed: 12/14/2022]
Abstract
In past 10 years, microRNAs (miRNAs) have gained scientific attention due to their importance in the pathophysiology of allergic diseases and their potential as biomarkers in liquid biopsies. They act as master post‐transcriptional regulators that control most cellular processes. As one miRNA can target several mRNAs, often within the same pathway, dysregulated expression of miRNAs may alter particular cellular responses and contribute, or lead, to the development of various diseases. In this review, we give an overview of the current research on miRNAs in allergic diseases, including atopic dermatitis, allergic rhinitis, and asthma. Specifically, we discuss how individual miRNAs function in the regulation of immune responses in epithelial cells and specialized immune cells in response to different environmental factors and respiratory viruses. In addition, we review insights obtained from experiments with murine models of allergic airway and skin inflammation and offer an overview of studies focusing on miRNA discovery using profiling techniques and bioinformatic modeling of the network effect of multiple miRNAs. In conclusion, we highlight the importance of research into miRNA function in allergy and asthma to improve our knowledge of the molecular mechanisms involved in the pathogenesis of this heterogeneous group of diseases.
Collapse
Affiliation(s)
- Julie Weidner
- Department of Internal Medicine and Clinical Nutrition Krefting Research Centre Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Sabine Bartel
- Department of Pathology and Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Ayse Kılıç
- Channing Division of Network Medicine Brigham and Women's Hospital Boston MA USA
| | - Ulrich M. Zissler
- Center for Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
| | - Harald Renz
- Institut für Laboratoriumsmedizin und Pathobiochemie Philipps University of Marburg Marburg Germany
| | - Jürgen Schwarze
- Centre for Inflammation Research The University of Edinburgh Edinburgh UK
| | - Carsten B. Schmidt‐Weber
- Center for Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
| | - Tania Maes
- Department of Respiratory Medicine Ghent University Ghent Belgium
| | - Ana Rebane
- Institute of Biomedicine and Translational Medicine University of Tartu Tartu Estonia
| | - Susanne Krauss‐Etschmann
- Research Center Borstel Borstel Germany
- Institute of Experimental Medicine Christian‐Albrechts University Kiel Kiel Germany
| | - Madeleine Rådinger
- Department of Internal Medicine and Clinical Nutrition Krefting Research Centre Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| |
Collapse
|