1
|
Wu L, Zhu X, Luo C, Zhao Y, Pan S, Shi K, Chen Y, Qiu J, Shen Z, Guo J, Jie W. Mechanistic role of RND3-regulated IL33/ST2 signaling on cardiomyocyte senescence. Life Sci 2024; 348:122701. [PMID: 38724005 DOI: 10.1016/j.lfs.2024.122701] [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: 03/20/2024] [Revised: 04/23/2024] [Accepted: 05/05/2024] [Indexed: 05/12/2024]
Abstract
Hyperinflammatory responses are pivotal in the cardiomyocyte senescence pathophysiology, with IL33 serving as a crucial pro-inflammatory mediator. Our previous findings highlighted RND3's suppressive effect on IL33 expression. This study aims to explore the role of RND3 in IL33/ST2 signaling activation and in cardiomyocyte senescence. Intramyocardial injection of exogenous IL33 reduces the ejection fraction and fractional shortening of rats, inducing the appearance of senescence-associated secretory phenotype (SASP) in myocardial tissues. Recombinant IL33 treatment of AC16 cardiomyocytes significantly upregulated expression of SASP factors like IL1α, IL6, and MCP1, and increased the p-p65/p65 ratio and proportions of SA-β-gal and γH2AX-positive cells. NF-κB inhibitor pyrrolidinedithiocarbamate ammonium (PDTC) and ST2 antibody astegolimab treatments mitigated above effects. RND3 gene knockout H9C2 cardiomyocytes using CRISPR/Cas9 technology upregulated IL33, ST2L, IL1α, IL6, and MCP1 levels, decreased sST2 levels, and increased SA-β-gal and γH2AX-positive cells. A highly possibility of binding between RND3 and IL33 proteins was showed by molecular docking and co-immunoprecipitation, and loss of RND3 attenuated ubiquitination mediated degradation of IL33; what's more, a panel of ubiquitination regulatory genes closely related to RND3 were screened using qPCR array. In contrast, RND3 overexpression in rats by injection of AAV9-CMV-RND3 particles inhibited IL33, ST2L, IL1α, IL6, and MCP1 expression in cardiac tissues, decreased serum IL33 levels, and increased sST2 levels. These results suggest that RND3 expression in cardiomyocytes modulates cell senescence by inhibiting the IL33/ST2/NF-κB signaling pathway, underscoring its potential as a therapeutic target in cardiovascular senescence.
Collapse
Affiliation(s)
- Linxu Wu
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China; Public Research Center of Hainan Medical University, Haikou 571199, P.R. China
| | - Xinglin Zhu
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Cai Luo
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Yangyang Zhao
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Shanshan Pan
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Kaijia Shi
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Yan Chen
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Jianmin Qiu
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China
| | - Zhihua Shen
- Department of Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang 524023, P.R. China.
| | - Junli Guo
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China.
| | - Wei Jie
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, the First Affiliated Hospital, Hainan Medical University, Haikou 571199, P.R. China.
| |
Collapse
|
2
|
Tian C, Liu Q, Zhang X, Li Z. Blocking group 2 innate lymphoid cell activation and macrophage M2 polarization: potential therapeutic mechanisms in ovalbumin-induced allergic asthma by calycosin. BMC Pharmacol Toxicol 2024; 25:30. [PMID: 38650035 PMCID: PMC11036756 DOI: 10.1186/s40360-024-00751-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Calycosin, a flavonoid compound extracted from Astragalus membranaceus, has shown anti-asthma benefits in house dust mite-induced asthma. Recent studies have suggested that innate-type cells, including group 2 innate lymphoid cells (ILC2s) and macrophages, serve as incentives for type 2 immunity and targets for drug development in asthma. This work focuses on the effects of calycosin on the dysregulated ILC2s and macrophages in allergic asthma. METHODS In vivo, the asthmatic mouse model was established with ovalbumin (OVA) sensitization and challenge, and calycosin was intraperitoneally administered at doses of 20 and 40 mg/kg. In vivo, mouse primary ILC2s were stimulated with interleukin (IL)-33 and mouse RAW264.7 macrophages were stimulated with IL-4 and IL-13 to establish the cell models. Cells were treated with calycosin at doses of 5 and 10 µM. RESULTS In vivo, we observed significantly reduced numbers of eosinophils, neutrophils, monocyte macrophages and lymphocytes in the bronchoalveolar lavage fluid (BALF) of OVA-exposed mice with 40 mg/kg calycosin. Histopathological assessment showed that calycosin inhibited the airway inflammation and remodeling caused by OVA. Calycosin markedly decreased the up-regulated IL-4, IL-5, IL-13, IL-33, and suppression tumorigenicity 2 (ST2) induced by OVA in BALF and/or lung tissues of asthmatic mice. Calycosin repressed the augment of arginase 1 (ARG1), IL-10, chitinase-like 3 (YM1) and mannose receptor C-type 1 (MRC1) levels in the lung tissues of asthmatic mice. In vivo, calycosin inhibited the IL-33-induced activation as well as the increase of IL-4, IL-5, IL-13 and ST2 in ILC2s. Calycosin also repressed the increase of ARG1, IL-10, YM1 and MRC1 induced by IL-4 and IL-13 in RAW264.7 macrophages. In addition, we found that these changes were more significant in 40 mg/kg calycosin treatment than 20 mg/kg calycosin. CONCLUSIONS Collectively, this study showed that calycosin might attenuate OVA-induced airway inflammation and remodeling in asthmatic mice via preventing ILC2 activation and macrophage M2 polarization. Our study might contribute to further study of asthmatic therapy.
Collapse
Affiliation(s)
- Chunyan Tian
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Graduate, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qi Liu
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaoyu Zhang
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhuying Li
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
3
|
Mocking TR, Duetz C, van Kuijk BJ, Westers TM, Cloos J, Bachas C. Merging and imputation of flow cytometry data: A critical assessment. Cytometry A 2023; 103:818-829. [PMID: 37338802 DOI: 10.1002/cyto.a.24774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/16/2023] [Accepted: 06/17/2023] [Indexed: 06/21/2023]
Abstract
Although most modern techniques and analysis methods in multiparameter flow cytometry (MFC) allow for increased dimensionality for the characterization and quantification of cell populations, most MFC applications depend on flow cytometers measuring relatively small (<16) numbers of parameters. When more markers than the available parameters need to be acquired, these are commonly distributed over multiple independent measurements that include a backbone of common markers. Several methods have been proposed to impute values for combinations of markers that were not measured simultaneously. These imputation methods are frequently used without proper validation and knowledge of their effects on data analysis. We evaluated the performance of existing imputation software (Infinicyt, CyTOFmerge, CytoBackBone, and cyCombine) in approximating known measured expression data in terms of similarity in visual appearance, cell expression, and gating in different datasets by splitting MFC samples into separate measurements with partially overlapping markers and re-calculating missing marker expression. Out of the assessed packages, CyTOFmerge showed the most accurate approximation of the known expression in terms of similar expression values and concordance with manual gating, with a mean F-score between 0.53 and 0.87 when retrieving cell populations in different datasets. Performance remained inadequate for all methods, with only limited similarity at the cell level. In conclusion, the use of imputed MFC data should take such limitations into account and include independent validation of results to justify conclusions.
Collapse
Affiliation(s)
- T R Mocking
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - C Duetz
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - B J van Kuijk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - T M Westers
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J Cloos
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - C Bachas
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Xiao J, Chen X, Liu W, Qian W, Bulek K, Hong L, Miller-Little W, Li X, Liu C. TRAF4 is crucial for ST2+ memory Th2 cell expansion in IL-33-driven airway inflammation. JCI Insight 2023; 8:e169736. [PMID: 37607012 PMCID: PMC10561728 DOI: 10.1172/jci.insight.169736] [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: 02/14/2023] [Accepted: 08/17/2023] [Indexed: 08/23/2023] Open
Abstract
Tumor necrosis factor receptor-associated factor 4 (TRAF4) is an important regulator of type 2 responses in the airway; however, the underlying cellular and molecular mechanisms remain elusive. Herein, we generated T cell-specific TRAF4-deficient (CD4-cre Traf4fl/fl) mice and investigated the role of TRAF4 in memory Th2 cells expressing IL-33 receptor (ST2, suppression of tumorigenicity 2) (ST2+ mTh2 cells) in IL-33-mediated type 2 airway inflammation. We found that in vitro-polarized TRAF4-deficient (CD4-cre Traf4fl/fl) ST2+ mTh2 cells exhibited decreased IL-33-induced proliferation as compared with TRAF4-sufficient (Traf4fl/fl) cells. Moreover, CD4-cre Traf4fl/fl mice showed less ST2+ mTh2 cell proliferation and eosinophilic infiltration in the lungs than Traf4fl/fl mice in the preclinical models of IL-33-mediated type 2 airway inflammation. Mechanistically, we discovered that TRAF4 was required for the activation of AKT/mTOR and ERK1/2 signaling pathways as well as the expression of transcription factor Myc and nutrient transporters (Slc2a1, Slc7a1, and Slc7a5), signature genes involved in T cell growth and proliferation, in ST2+ mTh2 cells stimulated by IL-33. Taken together, the current study reveals a role of TRAF4 in ST2+ mTh2 cells in IL-33-mediated type 2 pulmonary inflammation, opening up avenues for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Jianxin Xiao
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Xing Chen
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Weiwei Liu
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Wen Qian
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Katarzyna Bulek
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Lingzi Hong
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - William Miller-Little
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
- Medical Scientist Training Program
- Department of Pathology, and
| | - Xiaoxia Li
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
| | - Caini Liu
- Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, USA
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
5
|
Topczewska PM, Rompe ZA, Jakob MO, Stamm A, Leclère PS, Preußer A, Duerr CU, Thole LML, Kotsch K, Artis D, Klose CSN. ILC2 require cell-intrinsic ST2 signals to promote type 2 immune responses. Front Immunol 2023; 14:1130933. [PMID: 37063913 PMCID: PMC10104602 DOI: 10.3389/fimmu.2023.1130933] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/08/2023] [Indexed: 04/03/2023] Open
Abstract
The initiation of type 2 immune responses at mucosal barriers is regulated by rapidly secreted cytokines called alarmins. The alarmins IL-33, IL-25 and TSLP are mainly secreted by stromal and epithelial cells in tissues and were linked to chronic inflammatory diseases, such as allergic lung inflammation, or to resistance against worm infections. Receptors for alarmins are expressed by a variety of immune cells, including group 2 innate lymphoid cells (ILC2s), an early source of the type 2 cytokines, such as IL-5 and IL-13, which have been linked to atopic diseases and anti-worm immunity as well. However, the precise contribution of the IL-33 receptor signals for ILC2 activation still needs to be completed due to limitations in targeting genes in ILC2. Using the newly established Nmur1 iCre-eGFP mouse model, we obtained specific conditional genetic ablation of the IL-33 receptor subunit ST2 in ILC2s. ST2-deficient ILC2s were unresponsive to IL-33 but not to stimulation with the alarmin IL-25. As a result of defective ST2 signals, ILC2s produced limited amounts of IL-5 and IL-13 and failed to support eosinophil homeostasis. Further, ST2-deficient ILC2s were unable to expand and promote the recruitment of eosinophils during allergic lung inflammation provoked by papain administration. During infection with Nippostrongylus brasiliensis, ILC2-intrinsic ST2 signals were required to mount an effective type 2 immune response against the parasite leading to higher susceptibility against worm infection in conditional knockout mice. Therefore, this study argues for a non-redundant role of cell-intrinsic ST2 signals triggering proper activation of ILC2 for initiation of type 2 immunity.
Collapse
Affiliation(s)
- Patrycja M. Topczewska
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Zoe A. Rompe
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Manuel O. Jakob
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Anton Stamm
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Pierre S. Leclère
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Alexandra Preußer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Claudia U. Duerr
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| | - Linda Marie Laura Thole
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department for General and Visceral Surgery, Hindenburgdamm, Berlin, Germany
| | - Katja Kotsch
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department for General and Visceral Surgery, Hindenburgdamm, Berlin, Germany
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Friedman Center for Nutrition and Inflammation, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Christoph S. N. Klose
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Hindenburgdamm, Berlin, Germany
| |
Collapse
|
6
|
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
|
7
|
Resolution Potential of Necrotic Cell Death Pathways. Int J Mol Sci 2022; 24:ijms24010016. [PMID: 36613458 PMCID: PMC9819908 DOI: 10.3390/ijms24010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
During tissue damage caused by infection or sterile inflammation, not only damage-associated molecular patterns (DAMPs), but also resolution-associated molecular patterns (RAMPs) can be activated. These dying cell-associated factors stimulate immune cells localized in the tissue environment and induce the production of inflammatory mediators or specialized proresolving mediators (SPMs). Within the current prospect of science, apoptotic cell death is considered the main initiator of resolution. However, more RAMPs are likely to be released during necrotic cell death than during apoptosis, similar to what has been observed for DAMPs. The inflammatory potential of many regulated forms of necrotic cell death modalities, such as pyroptosis, necroptosis, ferroptosis, netosis, and parthanatos, have been widely studied in necroinflammation, but their possible role in resolution is less considered. In this review, we aim to summarize the relationship between necrotic cell death and resolution, as well as present the current available data regarding the involvement of certain forms of regulated necrotic cell death in necroresolution.
Collapse
|
8
|
Yi XM, Lian H, Li S. Signaling and functions of interleukin-33 in immune regulation and diseases. CELL INSIGHT 2022; 1:100042. [PMID: 37192860 PMCID: PMC10120307 DOI: 10.1016/j.cellin.2022.100042] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 05/18/2023]
Abstract
Interleukin-33 (IL-33) which belongs to the interleukin-1 (IL-1) family is an alarmin cytokine with critical roles in tissue homeostasis, pathogenic infection, inflammation, allergy and type 2 immunity. IL-33 transmits signals through its receptor IL-33R (also called ST2) which is expressed on the surface of T helper 2 (Th2) cells and group 2 innate lymphoid cells (ILC2s), thus inducing transcription of Th2-associated cytokine genes and host defense against pathogens. Moreover, the IL-33/IL-33R axis is also involved in development of multiple types of immune-related diseases. In this review, we focus on current progress on IL-33-trigggered signaling events, the important functions of IL-33/IL-33R axis in health and diseases as well as the promising therapeutic implications of these findings.
Collapse
Affiliation(s)
- Xue-Mei Yi
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Huan Lian
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Shu Li
- Department of Infectious Diseases, Zhongnan Hospital of Wuhan University, Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Research Unit of Innate Immune and Inflammatory Diseases, Chinese Academy of Medical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
9
|
Deng P, Wang L, Zhang Q, Chen S, Zhang Y, Xu H, Chen H, Xu Y, He W, Zhang J, Sun H. Therapeutic Potential of a Combination of Electroacupuncture and Human iPSC-Derived Small Extracellular Vesicles for Ischemic Stroke. Cells 2022; 11:820. [PMID: 35269441 PMCID: PMC8909871 DOI: 10.3390/cells11050820] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 11/24/2022] Open
Abstract
This paper aimed to explore the roles of the combination of electroacupuncture (EA) and induced pluripotent stem cell-derived small extracellular vesicles (iPSC-EVs) on mice with ischemic stroke and the underlying mechanisms. A focal cerebral ischemia model was established in C57BL/6 mice through middle cerebral artery occlusion (MCAO). After 3 days, neurological impairment and motor function were examined by performing behavioral tests. The infarct volume and neuronal apoptosis were examined using TTC staining and TUNEL assays. Flow cytometry was performed to assess the proliferation of T lymphocytes. The changes in the interleukin (IL)-33/ST2 axis were evaluated by immunofluorescence and Western blotting. The combination of EA and iPSC-EVs treatment ameliorated neurological impairments and reduced the infarct volume and neuronal apoptosis in MCAO mice. EA plus iPSC-EVs suppressed T helper (Th1) and Th17 responses and promoted the regulatory T cell (Treg) response. In addition, EA plus iPSC-EVs exerted neuroprotective effects by regulating the IL-33/ST2 axis and inhibiting the microglia and astrocyte activation. Taken together, the study shows that EA and iPSC-EVs exerted a synergistic neuroprotective effect in MCAO mice, and this treatment may represent a novel potent therapy for ischemic stroke and damage to other tissues.
Collapse
Affiliation(s)
- Peiying Deng
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| | - Liang Wang
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; (L.W.); (H.C.); (Y.X.)
| | - Qiongqiong Zhang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| | - Suhui Chen
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| | - Yamin Zhang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| | - Hong Xu
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| | - Hui Chen
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; (L.W.); (H.C.); (Y.X.)
| | - Yi Xu
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; (L.W.); (H.C.); (Y.X.)
| | - Wei He
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; (L.W.); (H.C.); (Y.X.)
| | - Jianmin Zhang
- CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China; (L.W.); (H.C.); (Y.X.)
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou 213000, China
- Guidon Pharmaceutics, Beijing 100176, China
| | - Hua Sun
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China; (P.D.); (Q.Z.); (S.C.); (Y.Z.); (H.X.)
| |
Collapse
|
10
|
Xu J, Tang J. Critical roles of interleukin-33/suppression of tumorigenicity 2 (IL-33/ST2) in pulmonary disorders. Chin Med J (Engl) 2022; 135:1508-1510. [PMID: 35170515 PMCID: PMC9481433 DOI: 10.1097/cm9.0000000000002007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
11
|
Xiong W, Liu W, Nishida S, Komiyama D, Liu W, Hirakawa J, Kawashima H. Therapeutic Effects of an Anti-sialyl Lewis X Antibody in a Murine Model of Allergic Asthma. Int J Mol Sci 2021; 22:9961. [PMID: 34576124 PMCID: PMC8471066 DOI: 10.3390/ijms22189961] [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: 08/11/2021] [Revised: 09/02/2021] [Accepted: 09/13/2021] [Indexed: 11/28/2022] Open
Abstract
Asthma is an allergic disease that causes severe infiltration of leukocytes into the lungs. Leukocyte infiltration is mediated by the binding of sialyl Lewis X (sLex) glycans present on the leukocytes to E-and P-selectins present on the endothelial cells at the sites of inflammation. Here, we found that mouse eosinophils express sLex glycans, and their infiltration into the lungs and proliferation in the bone marrow were significantly suppressed by an anti-sLex monoclonal antibody (mAb) F2 in a murine model of ovalbumin-induced asthma. The percentage of eosinophils in the bronchoalveolar lavage fluid and bone marrow and serum IgE levels decreased significantly in the F2-administered mice. Levels of T helper type 2 (Th2) cytokines and chemokines, involved in IgE class switching and eosinophil proliferation and recruitment, were also decreased in the F2-administered mice. An ex vivo cell rolling assay revealed that sLex glycans mediate the rolling of mouse eosinophils on P-selectin-expressing cells. These results indicate that the mAb F2 exerts therapeutic effects in a murine model of allergen-induced asthma, suggesting that sLex carbohydrate antigen could serve as a novel therapeutic target for allergic asthma.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hiroto Kawashima
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 260-8675, Japan; (W.X.); (W.L.); (S.N.); (D.K.); (W.L.); (J.H.)
| |
Collapse
|
12
|
Sánchez-Marteles M, Rubio-Gracia J, Peña-Fresneda N, Garcés-Horna V, Gracia-Tello B, Martínez-Lostao L, Crespo-Aznárez S, Pérez-Calvo JI, Giménez-López I. Early Measurement of Blood sST2 Is a Good Predictor of Death and Poor Outcomes in Patients Admitted for COVID-19 Infection. J Clin Med 2021; 10:3534. [PMID: 34441830 PMCID: PMC8396994 DOI: 10.3390/jcm10163534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/08/2021] [Accepted: 08/08/2021] [Indexed: 01/08/2023] Open
Abstract
Although several biomarkers have shown correlation to prognosis in COVID-19 patients, their clinical value is limited because of lack of specificity, suboptimal sensibility or poor dynamic behavior. We hypothesized that circulating soluble ST2 (sST2) could be associated to a worse outcome in COVID-19. In total, 152 patients admitted for confirmed COVID-19 were included in a prospective non-interventional, observational study. Blood samples were drawn at admission, 48-72 h later and at discharge. sST2 concentrations and routine blood laboratory were analyzed. Primary endpoints were admission at intensive care unit (ICU) and mortality. Median age was 57.5 years [Standard Deviation (SD: 12.8)], 60.4% males. 10% of patients (n = 15) were derived to ICU and/or died during admission. Median (IQR) sST2 serum concentration (ng/mL) rose to 53.1 (30.9) at admission, peaked at 48-72 h (79.5(64)) and returned to admission levels at discharge (44.9[36.7]). A concentration of sST2 above 58.9 ng/mL was identified patients progressing to ICU admission or death. Results remained significant after multivariable analysis. The area under the receiver operating characteristics curve (AUC) of sST2 for endpoints was 0.776 (p = 0.001). In patients admitted for COVID-19 infection, early measurement of sST2 was able to identify patients at risk of severe complications or death.
Collapse
Affiliation(s)
- Marta Sánchez-Marteles
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
| | - Jorge Rubio-Gracia
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
| | - Natacha Peña-Fresneda
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
- Facultad de Medicina, University of Zaragoza, 50009 Zaragoza, Spain
| | - Vanesa Garcés-Horna
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
| | - Borja Gracia-Tello
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
| | - Luis Martínez-Lostao
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
- Facultad de Medicina, University of Zaragoza, 50009 Zaragoza, Spain
- Department of Immunology, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain
| | - Silvia Crespo-Aznárez
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
| | - Juan Ignacio Pérez-Calvo
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
- Facultad de Medicina, University of Zaragoza, 50009 Zaragoza, Spain
| | - Ignacio Giménez-López
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
- Facultad de Medicina, University of Zaragoza, 50009 Zaragoza, Spain
- Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
| |
Collapse
|
13
|
Dustin CM, Habibovic A, Hristova M, Schiffers C, Morris CR, Lin MCJ, Bauer RA, Heppner DE, Daphtary N, Aliyeva M, van der Vliet A. Oxidation-Dependent Activation of Src Kinase Mediates Epithelial IL-33 Production and Signaling during Acute Airway Allergen Challenge. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:2989-2999. [PMID: 34088769 PMCID: PMC8642476 DOI: 10.4049/jimmunol.2000995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 04/06/2021] [Indexed: 11/19/2022]
Abstract
The respiratory epithelium forms the first line of defense against inhaled pathogens and acts as an important source of innate cytokine responses to environmental insults. One critical mediator of these responses is the IL-1 family cytokine IL-33, which is rapidly secreted upon acute epithelial injury as an alarmin and induces type 2 immune responses. Our recent work highlighted the importance of the NADPH oxidase dual oxidase 1 (DUOX1) in acute airway epithelial IL-33 secretion by various airborne allergens associated with H2O2 production and reduction-oxidation-dependent activation of Src kinases and epidermal growth factor receptor (EGFR) signaling. In this study, we show that IL-33 secretion in response to acute airway challenge with house dust mite (HDM) allergen critically depends on the activation of Src by a DUOX1-dependent oxidative mechanism. Intriguingly, HDM-induced epithelial IL-33 secretion was dramatically attenuated by small interfering RNA- or Ab-based approaches to block IL-33 signaling through its receptor IL1RL1 (ST2), indicating that HDM-induced IL-33 secretion includes a positive feed-forward mechanism involving ST2-dependent IL-33 signaling. Moreover, activation of type 2 cytokine responses by direct airway IL-33 administration was associated with ST2-dependent activation of DUOX1-mediated H2O2 production and reduction-oxidation-based activation of Src and EGFR and was attenuated in Duox1 -/- and Src +/- mice, indicating that IL-33-induced epithelial signaling and subsequent airway responses involve DUOX1/Src-dependent pathways. Collectively, our findings suggest an intricate relationship between DUOX1, Src, and IL-33 signaling in the activation of innate type 2 immune responses to allergens, involving DUOX1-dependent epithelial Src/EGFR activation in initial IL-33 secretion and in subsequent IL-33 signaling through ST2 activation.
Collapse
Affiliation(s)
- Christopher M Dustin
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Aida Habibovic
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Milena Hristova
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Caspar Schiffers
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Carolyn R Morris
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Miao-Chong Joy Lin
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Robert A Bauer
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - David E Heppner
- Department of Chemistry, University at Buffalo, State University of New York, Buffalo, NY; and
| | - Nirav Daphtary
- Department of Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Minara Aliyeva
- Department of Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT;
| |
Collapse
|
14
|
Zhang J, Feng X, Fan Y, Zhu G, Bai C. Molecular hydrogen alleviates asthma through inhibiting IL-33/ILC2 axis. Inflamm Res 2021; 70:569-579. [PMID: 33852061 DOI: 10.1007/s00011-021-01459-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 03/24/2021] [Accepted: 03/30/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Asthma is one of the most common noninfectious chronic diseases characterized by type II inflammation. This study aimed to investigate the effects of molecular hydrogen on the pathogenesis of asthma. METHODS OVA sensitized asthma mouse model and house dust mite treated 16HBE cellular model were established and hydrogen/oxygen mixture was used to treat asthmatic mice and 16HBE cells. Serum and BALF cytokines were measured with specific ELISA assays. E-cadherin and ZO-1 were detected by immunohistochemical staining and expression of caspase 3 and 9, NF-κB, IL-33 and ST2 was assessed by quantitative real-time PCR, western blot and/or immunofluorescence. IL-33 promoter activity was analyzed by dual-luciferase assay. ILC2 population was assayed by flow cytometry and differentially expressed miRNAs were detected using miRNA array. RESULTS Serum and BALF levels of IL-33 and other alarmin and type II cytokines were greatly increased by OVA and inhibited by H2 in asthmatic mice. The expression of NF-κB (p65) and ST2 was upregulated by OVA and suppressed by H2. ILC2 population was markedly increased in OVA-induced asthmatic mice, and such increase was inhibited by H2. E-cadherin and ZO-1 levels in airway tissues of asthmatic mice were significantly lower than that of control mice, and the reduction was recovered by H2 treatment. H2 alleviated HDM induced apoptosis of 16HBE cells, upregulation of IL-33 and ST2, and elevation of IL-33 promoter activity. A group of miRNAs differentially expressed in HDM and HDM + H2 treated 16HBE cells were identified. CONCLUSIONS These data demonstrated that H2 is efficient in suppressing allergen-induced asthma and could be developed as a therapeutics for asthma and other conditions of type II inflammation.
Collapse
Affiliation(s)
- Jingxi Zhang
- Department of Respiratory Medicine, Changhai Hospital, Navy Medical University, Shanghai, 200433, China.
| | - Xiumin Feng
- Department of Respiratory Medicine, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Yunxin Fan
- Department of Respiratory Medicine, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Guanglin Zhu
- Department of Respiratory Medicine, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Chong Bai
- Department of Respiratory Medicine, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| |
Collapse
|
15
|
Zhou X, Qu M, Tebon P, Jiang X, Wang C, Xue Y, Zhu J, Zhang S, Oklu R, Sengupta S, Sun W, Khademhosseini A. Screening Cancer Immunotherapy: When Engineering Approaches Meet Artificial Intelligence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001447. [PMID: 33042756 PMCID: PMC7539186 DOI: 10.1002/advs.202001447] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/16/2020] [Indexed: 02/05/2023]
Abstract
Immunotherapy is a class of promising anticancer treatments that has recently gained attention due to surging numbers of FDA approvals and extensive preclinical studies demonstrating efficacy. Nevertheless, further clinical implementation has been limited by high variability in patient response to different immunotherapeutic agents. These treatments currently do not have reliable predictors of efficacy and may lead to side effects. The future development of additional immunotherapy options and the prediction of patient-specific response to treatment require advanced screening platforms associated with accurate and rapid data interpretation. Advanced engineering approaches ranging from sequencing and gene editing, to tumor organoids engineering, bioprinted tissues, and organs-on-a-chip systems facilitate the screening of cancer immunotherapies by recreating the intrinsic and extrinsic features of a tumor and its microenvironment. High-throughput platform development and progress in artificial intelligence can also improve the efficiency and accuracy of screening methods. Here, these engineering approaches in screening cancer immunotherapies are highlighted, and a discussion of the future perspectives and challenges associated with these emerging fields to further advance the clinical use of state-of-the-art cancer immunotherapies are provided.
Collapse
Affiliation(s)
- Xingwu Zhou
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
- Department of Chemical and Biomolecular EngineeringHenry Samueli School of Engineering and Applied SciencesUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Moyuan Qu
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Peyton Tebon
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Xing Jiang
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
- School of NursingNanjing University of Chinese MedicineNanjing210023China
| | - Canran Wang
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Yumeng Xue
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Jixiang Zhu
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
- Department of Biomedical EngineeringSchool of Basic Medical SciencesGuangzhou Medical UniversityGuangzhou511436China
| | - Shiming Zhang
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Rahmi Oklu
- Minimally Invasive Therapeutics LaboratoryDivision of Vascular and Interventional RadiologyMayo ClinicPhoenixAZ85054USA
| | - Shiladitya Sengupta
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and TechnologyHarvard Medical SchoolBostonMA02115USA
| | - Wujin Sun
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
| | - Ali Khademhosseini
- Department of BioengineeringUniversity of California, Los AngelesLos AngelesCA90095USA
- Center for Minimally Invasive TherapeuticsCalifornia NanoSystems InstituteUniversity of California, Los AngelesLos AngelesCA90095USA
- Department of Chemical and Biomolecular EngineeringHenry Samueli School of Engineering and Applied SciencesUniversity of California, Los AngelesLos AngelesCA90095USA
- Jonsson Comprehensive Cancer CenterUniversity of California, Los AngelesLos AngelesCA90095USA
- Department of RadiologyDavid Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCA90095USA
- Terasaki Institute for Biomedical InnovationLos AngelesCA90064USA
| |
Collapse
|
16
|
What Makes the Lung Unique – Tissue-Specific Immunity in the Respiratory Tract. EUROPEAN MEDICAL JOURNAL 2020. [DOI: 10.33590/emj/20-00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The immune system constitutes a critical mechanism of the human body to preserve health and mitigate disease. In the lung, immunity is seen as a critical driver in many respiratory diseases, in particular in those characterised by aberrant inflammation, such as chronic obstructive pulmonary disease, fibrosis, and asthma. In this review, the specialised set of immune cells and lung tissue-specific regulators, including key cytokines such as granulocyte-macrophage colony-stimulating factor and transforming growth factor β, that control immune responses in the respiratory tract will be discussed. Furthermore, the current understanding of the impact of key environmental components such as the role of oxygen and lung microbiota on lung immunity will be highlighted. The goal is to identify the unique aspects of lung immune biology to facilitate insights into the aetiology of common lung inflammatory diseases and to provide the basis for a deeper mechanistic understanding of the underlying immune processes. Finally, key future avenues of research such as using more comprehensive quantitative approaches for elucidating molecular disease mechanisms as well as the potential to exploit tissue-specific regulators of immunity for therapy of lung inflammatory disorders will be discussed.
Collapse
|
17
|
Drake LY, Prakash YS. Contributions of IL-33 in Non-hematopoietic Lung Cells to Obstructive Lung Disease. Front Immunol 2020; 11:1798. [PMID: 32903501 PMCID: PMC7438562 DOI: 10.3389/fimmu.2020.01798] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-33 plays important roles in pulmonary immune responses and lung diseases including asthma and chronic obstructive pulmonary disease (COPD). There is substantial interest in identifying and characterizing cellular sources vs. targets of IL-33, and downstream signaling pathways involved in disease pathophysiology. While epithelial and immune cells have largely been the focus, in this review, we summarize current knowledge of expression, induction, and function of IL-33 and its receptor ST2 in non-hematopoietic lung cells in the context of health and disease. Under basal conditions, epithelial cells and endothelial cells are thought to be the primary resident cell types that express high levels of IL-33 and serve as ligand sources compared to mesenchymal cells (smooth muscle cells and fibroblasts). Under inflammatory conditions, IL-33 expression is increased in most non-hematopoietic lung cells, including epithelial, endothelial, and mesenchymal cells. In comparison to its ligand, the receptor ST2 shows low expression levels at baseline but similar to IL-33, ST2 expression is upregulated by inflammation in these non-hematopoietic lung cells which may then participate in chronic inflammation both as sources and autocrine/paracrine targets of IL-33. Downstream effects of IL-33 may occur via direct receptor activation or indirect interactions with the immune system, overall contributing to lung inflammation, airway hyper-responsiveness and remodeling (proliferation and fibrosis). Accordingly from a therapeutic perspective, targeting IL-33 and/or its receptor in non-hematopoietic lung cells becomes relevant.
Collapse
Affiliation(s)
- Li Y Drake
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|