101
|
Zhuang L, Ma W, Yan J, Zhong H. Evaluation of the effects of IL‑22 on the proliferation and differentiation of keratinocytes in vitro. Mol Med Rep 2020; 22:2715-2722. [PMID: 32945375 PMCID: PMC7453634 DOI: 10.3892/mmr.2020.11348] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 06/12/2020] [Indexed: 12/28/2022] Open
Abstract
Psoriasis is one of the most common chronic inflammatory skin diseases, it is characterized by hyperproliferation of keratinocytes and infiltration of inflammatory cells. Several in vitro studies have reported that interleukin (IL)‑22 is involved in excessive proliferation and abnormal differentiation of human keratinocytes. However, the association between IL‑22 and CCAAT enhancer binding protein α (C/EBPα) in the pathogenesis of psoriasis remains unclear. Therefore, the present study aimed to investigate the association between IL‑22 and C/EBPα, and the effects of IL‑22 on the proliferation and differentiation of keratinocytes. Keratinocytes were treated with different concentrations of IL‑22 (30, 60 and 90 ng/ml) and subsequently cells were collected at different time intervals. The expression levels of the key molecules of the mitogen‑activated protein kinase (MAPK) signaling pathway were detected using western blot analysis. In addition, the effect of IL‑22 on the proliferation rate of keratinocytes and the mRNA expression levels of C/EBPα were determined using a Cell Counting Kit‑8 assay and reverse transcription‑quantitative PCR, respectively. Furthermore, keratinocytes were transfected with C/EBPα small interfering (si)RNA or control using Lipofectamine® 2000. The results revealed that IL‑22 significantly induced the proliferation of keratinocytes and the expression of phosphorylated (p)‑JNK, p‑ERK and p‑p38 (P<0.05). Additionally, IL‑22 significantly inhibited the differentiation of keratinocytes, and the mRNA and protein expression of C/EBPα (P<0.05). Furthermore, downregulation of C/EBPα increased the proliferation rate of keratinocytes and reduced the expression levels of cytokeratin 10 and involucrin. Therefore, these results suggested that the effect of IL‑22 on the proliferation and differentiation of keratinocytes may be mediated via the regulation of the MAPK signaling pathway and the expression of C/EBPα.
Collapse
Affiliation(s)
- Le Zhuang
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Weiyuan Ma
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jianjun Yan
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Hua Zhong
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
102
|
Sabihi M, Böttcher M, Pelczar P, Huber S. Microbiota-Dependent Effects of IL-22. Cells 2020; 9:E2205. [PMID: 33003458 PMCID: PMC7599675 DOI: 10.3390/cells9102205] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Cytokines are important contributors to immune responses against microbial and environmental threats and are of particular importance at epithelial barriers. These interfaces are continuously exposed to external factors and thus require immune components to both protect the host from pathogen invasion and to regulate overt inflammation. Recently, substantial efforts have been devoted to understanding how cytokines act on certain cells at barrier sites, and why the dysregulation of immune responses may lead to pathogenesis. In particular, the cytokine IL-22 is involved in preserving an intact epithelium, maintaining a balanced microbiota and a functioning defense system against external threats. However, a tight regulation of IL-22 is generally needed, since uncontrolled IL-22 production can lead to the progression of autoimmunity and cancer. Our aim in this review is to summarize novel findings on IL-22 and its interactions with specific microbial stimuli, and subsequently, to understand their contributions to the function of IL-22 and the clinical outcome. We particularly focus on understanding the detrimental effects of dysregulated control of IL-22 in certain disease contexts.
Collapse
Affiliation(s)
| | | | | | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; (M.S.); (M.B.); (P.P.)
| |
Collapse
|
103
|
Arshad T, Mansur F, Palek R, Manzoor S, Liska V. A Double Edged Sword Role of Interleukin-22 in Wound Healing and Tissue Regeneration. Front Immunol 2020; 11:2148. [PMID: 33042126 PMCID: PMC7527413 DOI: 10.3389/fimmu.2020.02148] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
Wound healing and tissue regeneration is an intricate biological process that involves repair of cellular damage and maintenance of tissue integrity. Cascades involved in wound healing and tissue regeneration highly overlap with cancer causing pathways. Usually, subsequent tissue damage events include release of a number of cytokines to accomplish post-trauma restoration. IL-22 is one of the cytokines that are immediately produced to initiate immune response against several tissue impairments. IL-22 is a fundamental mediator in inflammation, mucous production, protective role against pathogens, wound healing, and tissue regeneration. However, accumulating evidence suggests pivotal role of IL-22 in instigation of various cancers due to its pro-inflammatory and tissue repairing activity. In this review, we summarize how healing effects of IL-22, when executed in an uncontrollable fashion can lead to carcinogenesis.
Collapse
Affiliation(s)
- Tanzeela Arshad
- Molecular Virology and Immunology Research Group, Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Fizzah Mansur
- Molecular Virology and Immunology Research Group, Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Richard Palek
- Department of Surgery, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
- Laboratory of Cancer Treatment and Tissue Regeneration, Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Sobia Manzoor
- Molecular Virology and Immunology Research Group, Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology, Islamabad, Pakistan
- Laboratory of Cancer Treatment and Tissue Regeneration, Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Vaclav Liska
- Department of Surgery, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
- Laboratory of Cancer Treatment and Tissue Regeneration, Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| |
Collapse
|
104
|
Sasidharan Nair V, Saleh R, Toor SM, Alajez NM, Elkord E. Transcriptomic Analyses of Myeloid-Derived Suppressor Cell Subsets in the Circulation of Colorectal Cancer Patients. Front Oncol 2020; 10:1530. [PMID: 32984004 PMCID: PMC7492613 DOI: 10.3389/fonc.2020.01530] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/16/2020] [Indexed: 12/24/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) promote tumor immune evasion and favor tumorigenesis by activating various tumor-promoting downstream signals. MDSC expansion is evident in the circulation and tumor microenvironment of many solid tumors including colorectal cancer (CRC). We have recently reported the transcriptomic profiles of tumor-infiltrating MDSCs in CRC patients and uncovered pathways, which could potentially assist tumor progression. In this study, we sorted different subsets of circulating MDSCs in CRC patients and investigated their transcriptomic profiles in order to disclose pathways, which could potentially contribute to disease progression. The sorted subsets included polymorphonuclear/granulocytic MDSCs (PMN-MDSCs), immature MDSCs (I-MDSCs), and monocytic MDSCs (M-MDSCs). Our functional annotation analyses revealed that multiple pathways including DNA damage-, chemotaxis-, apoptosis-, mitogen-activated protein kinase-, transforming growth factor β-, and myeloid differentiation–related transcripts were higher in PMN-MDSCs, compared with monocytic antigen-presenting cells (APCs) or I-MDSCs. Furthermore, genes related to Janus kinase (JAK)–signal transducer and activator of transcription (STAT) were also elevated in PMN-MDSCs. These data suggest that upregulation of JAK-STAT pathway could trigger multiple downstream targets in PMN-MDSCs, which favor tumor progression. Additionally, we found that pathways including phosphatidyl inositol 3-kinase (PI3K), interleukin 6, and TGF-β in M-MDSCs and cell cycle–related pathways in I-MDSCs were upregulated, compared with monocytic APCs. Moreover, acetylation-related genes were upregulated in both PMN-MDSCs and M-MDSCs. This latter finding implicates that epigenetic modifications could also play a role in the regulation of multiple tumor-promoting genes in PMN-MDSCs and M-MDSCs. Taken together, this study reveals various signaling pathways, which regulate the function of MDSC subsets in the circulation of CRC patients. However, functional studies are warranted to support these findings.
Collapse
Affiliation(s)
- Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Nehad M Alajez
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
105
|
Mossner S, Kuchner M, Fazel Modares N, Knebel B, Al-Hasani H, Floss DM, Scheller J. Synthetic interleukin 22 (IL-22) signaling reveals biological activity of homodimeric IL-10 receptor 2 and functional cross-talk with the IL-6 receptor gp130. J Biol Chem 2020; 295:12378-12397. [PMID: 32611765 PMCID: PMC7458808 DOI: 10.1074/jbc.ra120.013927] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/22/2020] [Indexed: 12/22/2022] Open
Abstract
Cytokine signaling is transmitted by cell-surface receptors that function as biological switches controlling mainly immune-related processes. Recently, we have designed synthetic cytokine receptors (SyCyRs) consisting of GFP and mCherry nanobodies fused to transmembrane and intracellular domains of cytokine receptors that phenocopy cytokine signaling induced by nonphysiological homo- and heterodimeric GFP-mCherry ligands. Interleukin 22 (IL-22) signals via both IL-22 receptor α1 (IL-22Rα1) and the common IL-10R2, belongs to the IL-10 cytokine family, and is critically involved in tissue regeneration. Here, IL-22 SyCyRs phenocopied native IL-22 signal transduction, indicated by induction of cytokine-dependent cellular proliferation, signal transduction, and transcriptome analysis. Whereas homodimeric IL-22Rα1 SyCyRs failed to activate signaling, homodimerization of the second IL-22 signaling chain, SyCyR(IL-10R2), which previously was considered not to induce signal transduction, led to induction of signal transduction. Interestingly, the SyCyR(IL-10R2) and SyCyR(IL-22Rα1) constructs could form functional heterodimeric receptor signaling complexes with the synthetic IL-6 receptor chain SyCyR(gp130). In summary, we have demonstrated that IL-22 signaling can be phenocopied by synthetic cytokine receptors, identified a functional IL-10R2 homodimeric receptor complex, and uncovered broad receptor cross-talk of IL-22Rα1 and IL-20R2 with gp130.
Collapse
Affiliation(s)
- Sofie Mossner
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Marcus Kuchner
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Nastaran Fazel Modares
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Birgit Knebel
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Doreen M Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
106
|
Kinsella S, Dudakov JA. When the Damage Is Done: Injury and Repair in Thymus Function. Front Immunol 2020; 11:1745. [PMID: 32903477 PMCID: PMC7435010 DOI: 10.3389/fimmu.2020.01745] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/30/2020] [Indexed: 01/02/2023] Open
Abstract
Even though the thymus is exquisitely sensitive to acute insults like infection, shock, or common cancer therapies such as cytoreductive chemo- or radiation-therapy, it also has a remarkable capacity for repair. This phenomenon of endogenous thymic regeneration has been known for longer even than its primary function to generate T cells, however, the underlying mechanisms controlling the process have been largely unstudied. Although there is likely continual thymic involution and regeneration in response to stress and infection in otherwise healthy people, acute and profound thymic damage such as that caused by common cancer cytoreductive therapies or the conditioning regimes as part of hematopoietic cell transplantation (HCT), leads to prolonged T cell deficiency; precipitating high morbidity and mortality from opportunistic infections and may even facilitate cancer relapse. Furthermore, this capacity for regeneration declines with age as a function of thymic involution; which even at steady state leads to reduced capacity to respond to new pathogens, vaccines, and immunotherapy. Consequently, there is a real clinical need for strategies that can boost thymic function and enhance T cell immunity. One approach to the development of such therapies is to exploit the processes of endogenous thymic regeneration into novel pharmacologic strategies to boost T cell reconstitution in clinical settings of immune depletion such as HCT. In this review, we will highlight recent work that has revealed the mechanisms by which the thymus is capable of repairing itself and how this knowledge is being used to develop novel therapies to boost immune function.
Collapse
Affiliation(s)
- Sinéad Kinsella
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Jarrod A. Dudakov
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Immunotherapy Integrated Research Center, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Immunology, University of Washington, Seattle, WA, United States
| |
Collapse
|
107
|
Shohan M, Dehghani R, Khodadadi A, Dehnavi S, Ahmadi R, Joudaki N, Houshmandfar S, Shamshiri M, Shojapourian S, Bagheri N. Interleukin-22 and intestinal homeostasis: Protective or destructive? IUBMB Life 2020; 72:1585-1602. [PMID: 32365282 DOI: 10.1002/iub.2295] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/08/2020] [Accepted: 04/11/2020] [Indexed: 12/16/2022]
Abstract
Interleukin (IL)-22 is a member of IL-10 family cytokines with various immunologic functions. As its name implies, IL-22 is known to be secreted mainly by Th22 cells, a recently discovered lineage of CD4+ T cells. Also, Th17, Th1, natural killer cells, γδT cells, and innate immune cells along with some nonlymphoid cells have been confirmed as secondary cellular sources of IL-22. Different cell types such as bronchial and intestinal epithelial cells, keratinocytes, hepatocytes, dermal fibroblasts, and tubular epithelial cells are affected by IL-22. Both pathologic and protective roles have been attributed to IL-22 in maintaining gut homeostasis and inflammation. According to the latest fast-growing investigations, IL-22 is significantly involved in various pathologies including allergic diseases, infection, autoimmunity, and cancer development. Regulating gut immune responses, barrier integrity, and inflammation is dependent on a diverse complex of cytokines and mediators which are secreted by mucosal immune cells. Several investigations have been designed to recognize the role of IL-22 in gastrointestinal immunity. This article tries to discuss the latest knowledge on this issue and clarify the potential of IL-22 to be used in the future therapeutic approaches of intestinal disorders including inflammatory bowel diseases and colon cancer.
Collapse
Affiliation(s)
- Mojtaba Shohan
- 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
| | - Razieh Dehghani
- Department of Pediatrics, Abuzar Children's Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ali Khodadadi
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sajad Dehnavi
- 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
| | - Reza Ahmadi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nazanin Joudaki
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sheyda Houshmandfar
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Marziye Shamshiri
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Samira Shojapourian
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
108
|
Furue M. Regulation of Filaggrin, Loricrin, and Involucrin by IL-4, IL-13, IL-17A, IL-22, AHR, and NRF2: Pathogenic Implications in Atopic Dermatitis. Int J Mol Sci 2020; 21:E5382. [PMID: 32751111 PMCID: PMC7432778 DOI: 10.3390/ijms21155382] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 12/16/2022] Open
Abstract
Atopic dermatitis (AD) is an eczematous, pruritic skin disorder with extensive barrier dysfunction and elevated interleukin (IL)-4 and IL-13 signatures. The barrier dysfunction correlates with the downregulation of barrier-related molecules such as filaggrin (FLG), loricrin (LOR), and involucrin (IVL). IL-4 and IL-13 potently inhibit the expression of these molecules by activating signal transducer and activator of transcription (STAT)6 and STAT3. In addition to IL-4 and IL-13, IL-22 and IL-17A are probably involved in the barrier dysfunction by inhibiting the expression of these barrier-related molecules. In contrast, natural or medicinal ligands for aryl hydrocarbon receptor (AHR) are potent upregulators of FLG, LOR, and IVL expression. As IL-4, IL-13, IL-22, and IL-17A are all capable of inducing oxidative stress, antioxidative AHR agonists such as coal tar, glyteer, and tapinarof exert particular therapeutic efficacy for AD. These antioxidative AHR ligands are known to activate an antioxidative transcription factor, nuclear factor E2-related factor 2 (NRF2). This article focuses on the mechanisms by which FLG, LOR, and IVL expression is regulated by IL-4, IL-13, IL-22, and IL-17A. The author also summarizes how AHR and NRF2 dual activators exert their beneficial effects in the treatment of AD.
Collapse
Affiliation(s)
- Masutaka Furue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka 812-8582, Japan; ; Tel.: +81-92-642-5581; Fax: +81-92-642-5600
- Research and Clinical Center for Yusho and Dioxin, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka 812-8582, Japan
- Division of Skin Surface Sensing, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka 812-8582, Japan
| |
Collapse
|
109
|
Wei HX, Wang B, Li B. IL-10 and IL-22 in Mucosal Immunity: Driving Protection and Pathology. Front Immunol 2020; 11:1315. [PMID: 32670290 PMCID: PMC7332769 DOI: 10.3389/fimmu.2020.01315] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
The barrier surfaces of the gastrointestinal tract are in constant contact with various microorganisms. Cytokines orchestrate the mucosal adaptive and innate immune cells in the defense against pathogens. IL-10 and IL-22 are the best studied members of the IL-10 family and play essential roles in maintaining mucosal homeostasis. IL-10 serves as an important regulator in preventing pro-inflammatory responses while IL-22 plays a protective role in tissue damage and contributes to pathology in certain settings. In this review, we focus on these two cytokines in the development of gastrointestinal diseases, including inflammatory bowel diseases (IBD) and colitis-associated cancer (CAC). We summarize the recent studies and try to gain a better understanding on how they regulate immune responses to maintain equilibrium under inflammatory conditions.
Collapse
Affiliation(s)
- Hua-Xing Wei
- Division of Life Sciences and Medicine, Department of Laboratory Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Baolong Wang
- Division of Life Sciences and Medicine, Department of Laboratory Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Bofeng Li
- Division of Life Sciences and Medicine, Department of Medical Oncology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| |
Collapse
|
110
|
Interleukin-22 Inhibits Respiratory Syncytial Virus Production by Blocking Virus-Mediated Subversion of Cellular Autophagy. iScience 2020; 23:101256. [PMID: 32580124 PMCID: PMC7317237 DOI: 10.1016/j.isci.2020.101256] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/28/2020] [Accepted: 06/05/2020] [Indexed: 12/18/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection can cause severe bronchiolitis in infants requiring hospitalization, whereas the elderly and immunocompromised are prone to RSV-induced pneumonia. RSV primarily infects lung epithelial cells. Given that no vaccine against RSV is currently available, we tested the ability of the epithelial-barrier protective cytokine interleukin-22 (IL-22) to control RSV production. When used in a therapeutic modality, IL-22 efficiently blunted RSV production from infected human airway and alveolar epithelial cells and IL-22 administration drastically reduced virus titer in the lungs of infected newborn mice. RSV infection resulted in increased expression of LC3B, a key component of the cellular autophagic machinery, and knockdown of LC3B ablated virus production. RSV subverted LC3B with evidence of co-localization and caused a significant reduction in autophagic flux, both reversed by IL-22 treatment. Our findings inform a previously unrecognized anti-viral effect of IL-22 that can be harnessed to prevent RSV-induced severe respiratory disease. RSV infection of lung epithelial cells subverts the cellular autophagic machinery RSV infection inhibits autophagic flux in infected cells IL-22 inhibits RSV production from human lung epithelial cells and in neonatal mice IL-22 blocks RSV-LC3B co-localization and restores cellular autophagic flux
Collapse
|
111
|
Paternal restraint stress affects offspring metabolism via ATF-2 dependent mechanisms in Drosophila melanogaster germ cells. Commun Biol 2020; 3:208. [PMID: 32367035 PMCID: PMC7198565 DOI: 10.1038/s42003-020-0935-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 04/07/2020] [Indexed: 12/22/2022] Open
Abstract
Paternal environmental factors can epigenetically influence gene expressions in offspring. We demonstrate that restraint stress, an experimental model for strong psychological stress, to fathers affects the epigenome, transcriptome, and metabolome of offspring in a MEKK1-dATF2 pathway-dependent manner in Drosophila melanogaster. Genes involved in amino acid metabolism are upregulated by paternal restraint stress, while genes involved in glycolysis and the tricarboxylic acid (TCA) cycle are downregulated. The effects of paternal restraint stress are also confirmed by metabolome analysis. dATF-2 is highly expressed in testicular germ cells, and restraint stress also induces p38 activation in the testes. Restraint stress induces Unpaired 3 (Upd3), a Drosophila homolog of Interleukin 6 (IL-6). Moreover, paternal overexpression of upd3 in somatic cells disrupts heterochromatin in offspring but not in offspring from dATF-2 mutant fathers. These results indicate that paternal restraint stress affects metabolism in offspring via inheritance of dATF-2-dependent epigenetic changes. Ki-Hyeon Seong et al. report that paternal environmental stress affects the metabolism of their offspring in Drosophila melanogaster. They exposed male flies to stress by preventing them from moving for 10 hours at a time and then measured gene expression and metabolite levels in their offspring, who showed increased expression of amino acid and one-carbon metabolism-related genes and downregulation of glycolysis and the TCA cycle.
Collapse
|
112
|
Interleukin-22 regulates gastric cancer cell proliferation through regulation of the JNK signaling pathway. Exp Ther Med 2020; 20:205-210. [PMID: 32536992 PMCID: PMC7282062 DOI: 10.3892/etm.2020.8707] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/05/2019] [Indexed: 01/01/2023] Open
Abstract
Inflammation is considered as one of the major hallmarks of cancer and is associated with gastric cancer. Interleukin-22 (IL-22), a member of the IL-10 family, serves an important role in inflammatory diseases and tumors. The aim of the present study was to examine the effects of IL-22 on the proliferation of gastric cancer cells (AGS cells) in vitro and explore the associated molecular mechanism. The results of a Cell Counting kit-8 assay using AGS cells transfected with an IL-22-plasmid indicated that IL-22 could promote AGS cell viability. However, when IL-22 was knocked down by IL-22-short hairpin (sh)RNA, the viability of AGS cells was significantly impaired. Western blotting results indicated that IL-22 decreased the activation of the mitogen-activated protein kinase (MAPK) signaling pathway. Furthermore, IL-22-shRNA transfection increased the activation of MAPK, as evidenced by the upregulated phosphorylation of ERK and JNK. Taken together, the results of the present study suggest that IL-22 regulated the viability of gastric cancer cells through the JNK signaling pathway, suggesting a therapeutic approach for gastric cancer via targeting IL-22.
Collapse
|
113
|
Samotij D, Nedoszytko B, Bartosińska J, Batycka-Baran A, Czajkowski R, Dobrucki IT, Dobrucki LW, Górecka-Sokołowska M, Janaszak-Jasienicka A, Krasowska D, Kalinowski L, Macieja-Stawczyk M, Nowicki RJ, Owczarczyk-Saczonek A, Płoska A, Purzycka-Bohdan D, Radulska A, Reszka E, Siekierzycka A, Słomiński A, Słomiński R, Sobalska-Kwapis M, Strapagiel D, Szczerkowska-Dobosz A, Szczęch J, Żmijewski M, Reich A. Pathogenesis of psoriasis in the "omic" era. Part I. Epidemiology, clinical manifestation, immunological and neuroendocrine disturbances. Postepy Dermatol Alergol 2020; 37:135-153. [PMID: 32489346 PMCID: PMC7262814 DOI: 10.5114/ada.2020.94832] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Psoriasis is a common, chronic, inflammatory, immune-mediated skin disease affecting about 2% of the world's population. According to current knowledge, psoriasis is a complex disease that involves various genes and environmental factors, such as stress, injuries, infections and certain medications. The chronic inflammation of psoriasis lesions develops upon epidermal infiltration, activation, and expansion of type 1 and type 17 Th cells. Despite the enormous progress in understanding the mechanisms that cause psoriasis, the target cells and antigens that drive pathogenic T cell responses in psoriatic lesions are still unproven and the autoimmune basis of psoriasis still remains hypothetical. However, since the identification of the Th17 cell subset, the IL-23/Th17 immune axis has been considered a key driver of psoriatic inflammation, which has led to the development of biologic agents that target crucial elements of this pathway. Here we present the current understanding of various aspects in psoriasis pathogenesis.
Collapse
Affiliation(s)
- Dominik Samotij
- Department of Dermatology, University of Rzeszow, Rzeszow, Poland
| | - Bogusław Nedoszytko
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Joanna Bartosińska
- Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Aleksandra Batycka-Baran
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - Rafał Czajkowski
- Department of Dermatology and Venereology, Faculty of Medicine, Ludwik Rydygier Medical College in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Iwona T. Dobrucki
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
| | - Lawrence W. Dobrucki
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Magdalena Górecka-Sokołowska
- Department of Dermatology, Sexually Transmitted Disorders and Immunodermatology, Jurasz University Hospital No. 1, Bydgoszcz, Poland
| | - Anna Janaszak-Jasienicka
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Dorota Krasowska
- Department of Dermatology, Venereology and Pediatric Dermatology, Medical University of Lublin, Lublin, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Marta Macieja-Stawczyk
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Roman J. Nowicki
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, University of Warmia and Mazury, Olsztyn, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Dorota Purzycka-Bohdan
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Adrianna Radulska
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Edyta Reszka
- Department of Molecular Genetics and Epigenetics, Nofer Institute of Occupational Medicine, Lodz, Poland
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure, (BBMRI.PL), Gdansk, Poland
| | - Andrzej Słomiński
- Department of Dermatology, Birmingham, AL, USA
- Comprehensive Cancer Center, Cancer Chemoprevention Program, Birmingham, AL, USA
- VA Medical Center, Birmingham, AL, USA
| | - Radomir Słomiński
- Department of Medicine, Division of Rheumatology, University of Alabama, Birmingham, AL, USA
| | - Marta Sobalska-Kwapis
- Biobank Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Dominik Strapagiel
- Biobank Lab, Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Aneta Szczerkowska-Dobosz
- Department of Dermatology, Venereology and Allergology, Medical University of Gdansk, Gdansk, Poland
| | - Justyna Szczęch
- Department of Dermatology, University of Rzeszow, Rzeszow, Poland
| | - Michał Żmijewski
- Department of Histology, Medical University of Gdansk, Gdansk, Poland
| | - Adam Reich
- Department of Dermatology, University of Rzeszow, Rzeszow, Poland
| |
Collapse
|
114
|
Che Y, Su Z, Xia L. Effects of IL-22 on cardiovascular diseases. Int Immunopharmacol 2020; 81:106277. [PMID: 32062077 DOI: 10.1016/j.intimp.2020.106277] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/29/2020] [Accepted: 02/01/2020] [Indexed: 12/11/2022]
Abstract
Interleukin-22 (IL-22), which belongs to the IL-10 family, is an alpha helix cytokine specifically produced by many lymphocytes, such as Th1, Th17, Th22, ILCs, CD4+ and CD8+ T cells. In recent years, more and more studies have demonstrated that IL-22 has an interesting relationship with various cardiovascular diseases, including myocarditis, myocardial infarction, atherosclerosis, and other cardiovascular diseases, and IL-22 signal may play a dual role in cardiovascular diseases. Here, we summarize the recent progress on the source, function, regulation of IL-22 and the effects of IL-22 signal in cardiovascular diseases. The study of IL-22 will suggest more specific strategies to maneuver these functions for the effective treatment of cardiovascular diseases and future clinical treatment.
Collapse
Affiliation(s)
- Yang Che
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; International Genome Center, Jiangsu University, Zhenjiang 212013, China
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang 212013, China; Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; International Genome Center, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
115
|
Grän F, Kerstan A, Serfling E, Goebeler M, Muhammad K. Current Developments in the Immunology of Psoriasis. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:97-110. [PMID: 32226340 PMCID: PMC7087066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Psoriasis is a frequent inflammatory skin disease. Fundamental research on the pathogenesis of psoriasis has substantially increased our understanding of skin immunology, which has helped to introduce innovative and highly effective therapies. Psoriasis is a largely T lymphocyte-mediated disease in which activation of innate immune cells and pathogenic T cells result in skin inflammation and hyperproliferation of keratinocytes. B cells have thus far largely been neglected regarding their role for the pathogenesis of psoriasis. However, recent data shed light on their role in inflammatory skin diseases. Interestingly, interleukin (IL)-10-producing regulatory B cells have been assumed to ameliorate psoriasis. In this review, we will discuss the development of disease, pathogenicity, and current developments in therapeutic options. We describe different roles of T cells, B cells, and cytokines for the immunopathology and disease course of psoriasis.
Collapse
Affiliation(s)
- Franziska Grän
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Kerstan
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Edgar Serfling
- Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Matthias Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Khalid Muhammad
- Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany,Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates,To whom all correspondence should be addressed: K. Muhammad, Tel: +971 3 713 6517, Fax: +971 3 713 4927;
| |
Collapse
|
116
|
Powell N, Pantazi E, Pavlidis P, Tsakmaki A, Li K, Yang F, Parker A, Pin C, Cozzetto D, Minns D, Stolarczyk E, Saveljeva S, Mohamed R, Lavender P, Afzali B, Digby-Bell J, Tjir-Li T, Kaser A, Friedman J, MacDonald TT, Bewick GA, Lord GM. Interleukin-22 orchestrates a pathological endoplasmic reticulum stress response transcriptional programme in colonic epithelial cells. Gut 2020; 69:578-590. [PMID: 31792136 PMCID: PMC7034350 DOI: 10.1136/gutjnl-2019-318483] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 10/15/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The functional role of interleukin-22 (IL22) in chronic inflammation is controversial, and mechanistic insights into how it regulates target tissue are lacking. In this study, we evaluated the functional role of IL22 in chronic colitis and probed mechanisms of IL22-mediated regulation of colonic epithelial cells. DESIGN To investigate the functional role of IL22 in chronic colitis and how it regulates colonic epithelial cells, we employed a three-dimentional mini-gut epithelial organoid system, in vivo disease models and transcriptomic datasets in human IBD. RESULTS As well as inducing transcriptional modules implicated in antimicrobial responses, IL22 also coordinated an endoplasmic reticulum (ER) stress response transcriptional programme in colonic epithelial cells. In the colon of patients with active colonic Crohn's disease (CD), there was enrichment of IL22-responsive transcriptional modules and ER stress response modules. Strikingly, in an IL22-dependent model of chronic colitis, targeting IL22 alleviated colonic epithelial ER stress and attenuated colitis. Pharmacological modulation of the ER stress response similarly impacted the severity of colitis. In patients with colonic CD, antibody blockade of IL12p40, which simultaneously blocks IL12 and IL23, the key upstream regulator of IL22 production, alleviated the colonic epithelial ER stress response. CONCLUSIONS Our data challenge perceptions of IL22 as a predominantly beneficial cytokine in IBD and provide novel insights into the molecular mechanisms of IL22-mediated pathogenicity in chronic colitis. Targeting IL22-regulated pathways and alleviating colonic epithelial ER stress may represent promising therapeutic strategies in patients with colitis. TRIAL REGISTRATION NUMBER NCT02749630.
Collapse
Affiliation(s)
- Nick Powell
- School of Immunology and Microbial Sciences, King's College London, London, UK
- National Institute for Health Research Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK
| | - Eirini Pantazi
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | | | - Anastasia Tsakmaki
- Diabetes Research Group, School of Life Course Sciences, Faculty of Life Sciences and Medicine, Kings College London, London, UK
| | - Katherine Li
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Feifei Yang
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Aimee Parker
- Quadram Institute Bioscience, Norwich, Norfolk, UK
| | - Carmen Pin
- Quadram Institute Bioscience, Norwich, Norfolk, UK
| | - Domenico Cozzetto
- National Institute for Health Research Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
- Department of Translational Bioinformatics, National Institute for Health Research Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Danielle Minns
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Emilie Stolarczyk
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Svetlana Saveljeva
- Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Rami Mohamed
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Paul Lavender
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Behdad Afzali
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Jonathan Digby-Bell
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Tsui Tjir-Li
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Joshua Friedman
- Janssen Research & Development, Spring House, Pennsylvania, USA
| | - Thomas T MacDonald
- Centre for Immunology and Infectious Disease, Bart's & the London School of Medicine and Dentistry, Blizard Institute of Cell and Molecular Science, London, UK
| | - Gavin A Bewick
- Diabetes Research Group, School of Life Course Sciences, Faculty of Life Sciences and Medicine, Kings College London, London, UK
| | - Graham M Lord
- School of Immunology and Microbial Sciences, King's College London, London, UK
- National Institute for Health Research Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
117
|
He Y, Yang Y, Xu J, Liao Y, Liu L, Deng L, Xiong X. IL22 drives cutaneous melanoma cell proliferation, migration and invasion through activation of miR-181/STAT3/AKT axis. J Cancer 2020; 11:2679-2687. [PMID: 32201538 PMCID: PMC7066011 DOI: 10.7150/jca.40974] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/31/2020] [Indexed: 12/13/2022] Open
Abstract
Cutaneous melanoma (CM) is neoplastic growth of melanocytes with strong potential to proliferate and invade, prone to a fatal disease soon which is beyond surgical clearance. The use of regulator involving in antitumor immune responses has been identified as a potential therapeutic option for CM, but still need fully understood at present. Recently, interleukin 22 (IL22), an immune molecule secreted mostly by CD4+ T cells, was reported having functions in a variety of human diseases including encouragement of lung cancer progression, yet, its role in CM is lacking. Here, we first found elevated expression of IL22 in both serum of CM patients and tissues. Up-regulated IL22 significantly promoted cell proliferation, migration and invasion in CM cells deriving from different original culture history. Moreover, in vivo CM model, IL22 treatment caused a significant increase in tumor size. Additionally, we found these effects accompanied by obvious increased miR-181 expression in CM. Importantly, both in vivo and in vitro results revealed that miR-181 downregulation reversed the effects of IL22 on CM cell proliferation, migration, invasion, and CM tumor size as well. Finally, in CM cells deriving from different culture history, we identified STAT3 to be a target gene of miR-181. Higher expression level of IL22 suppressed STAT3 expression, while enhanced expression of p-AKT, p-β-catenin and MMP4; however, down-regulation of miR-181 reversed these situations. Thus, we conclude that IL22 promotes CM progression by driving miR-181/STAT3/AKT axis.
Collapse
Affiliation(s)
- Yuanmin He
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Yang
- Department of Public Health, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jixiang Xu
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yongmei Liao
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Li Liu
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Li Deng
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xia Xiong
- Department of Dermatology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
118
|
Deng S, Clowers MJ, Velasco WV, Ramos-Castaneda M, Moghaddam SJ. Understanding the Complexity of the Tumor Microenvironment in K-ras Mutant Lung Cancer: Finding an Alternative Path to Prevention and Treatment. Front Oncol 2020; 9:1556. [PMID: 32039025 PMCID: PMC6987304 DOI: 10.3389/fonc.2019.01556] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/23/2019] [Indexed: 12/16/2022] Open
Abstract
Kirsten rat sarcoma viral oncogene (K-ras) is a well-documented, frequently mutated gene in lung cancer. Since K-ras regulates numerous signaling pathways related to cell survival and proliferation, mutations in this gene are powerful drivers of tumorigenesis and confer prodigious survival advantages to developing tumors. These malignant cells dramatically alter their local tissue environment and in the process recruit a powerful ally: inflammation. Inflammation in the context of the tumor microenvironment can be described as either antitumor or protumor (i.e., aiding or restricting tumor progression, respectively). Many current treatments, like immune checkpoint blockade, seek to augment antitumor inflammation by alleviating inhibitory signaling in cytotoxic T cells; however, a burgeoning area of research is now focusing on ways to modulate and mitigate protumor inflammation. Here, we summarize the interplay of tumor-promoting inflammation and K-ras mutant lung cancer pathogenesis by exploring the cytokines, signaling pathways, and immune cells that mediate this process.
Collapse
Affiliation(s)
- Shanshan Deng
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Michael J Clowers
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Walter V Velasco
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marco Ramos-Castaneda
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
| |
Collapse
|
119
|
Boldeanu MV, Siloşi I, Bărbulescu AL, Sandu RE, Geormăneanu C, Pădureanu V, Popescu-Drigă MV, Poenariu IS, Siloşi CA, Ungureanu AM, Dijmărescu AL, Boldeanu L. Host immune response in chronic hepatitis C infection: involvement of cytokines and inflammasomes. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2020; 61:33-43. [PMID: 32747893 PMCID: PMC7728117 DOI: 10.47162/rjme.61.1.04] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/19/2020] [Indexed: 12/13/2022]
Abstract
Chronic liver disease is a major health issue worldwide and chronic hepatitis C (CHC) is associated with an increased risk of cirrhosis and hepatocellular carcinoma (HCC). There is evidence that the hepatitis C virus (HCV) infection is correlated with immune senescence by way of immune activation and chronic inflammation, which lead to increased metabolic and cardiovascular risk, as well as progressive liver damage. Both the innate and adaptive immunity are firmly tied to the prognosis of an infection with HCV and its response to antiviral therapy. HCV is therefore associated with increased pro-inflammatory status, heightened production of cytokines, prolonged systemic inflammation, as well as increased morbidity and mortality, mainly due to the progression of hepatic fibrosis and HCC, but also secondary to cardiovascular diseases. Viral hepatic pathology is increasingly considered a disease that is no longer merely limited to the liver, but one with multiple metabolic consequences. Numerous in vitro studies, using experimental models of acute or chronic inflammation of the liver, has brought new information on immunopathological mechanisms resulting from viral infections and have highlighted the importance of involving complex structures, inflammasomes complex, in these mechanisms, in addition to the involvement of numerous proinflammatory cytokines. Beyond obtaining a sustained viral response and halting the aforementioned hepatic fibrosis, the current therapeutic "treat-to-target" strategies are presently focused on immune-mediated and metabolic disorders, to improve the quality of life and long-term prognosis of CHC patients.
Collapse
Affiliation(s)
- Mihail Virgil Boldeanu
- Department of Immunology, University of Medicine and Pharmacy of Craiova, Romania
- Medico Science SRL – Stem Cell Bank Unit, Craiova, Romania
| | - Isabela Siloşi
- Department of Immunology, University of Medicine and Pharmacy of Craiova, Romania
| | | | - Raluca Elena Sandu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova, Romania
| | - Cristiana Geormăneanu
- Department of Emergency Medicine and First Aid, University of Medicine and Pharmacy of Craiova, Romania
| | - Vlad Pădureanu
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, Romania
| | | | | | | | | | - Anda Lorena Dijmărescu
- Department of Obstetrics and Gynecology, University of Medicine and Pharmacy of Craiova, Romania
| | - Lidia Boldeanu
- Department of Microbiology, University of Medicine and Pharmacy of Craiova, Romania
| |
Collapse
|
120
|
Moré DD, Cardoso FF, Mudadu MA, Malagó-Jr W, Gulias-Gomes CC, Sollero BP, Ibelli AMG, Coutinho LL, Regitano LCA. Network analysis uncovers putative genes affecting resistance to tick infestation in Braford cattle skin. BMC Genomics 2019; 20:998. [PMID: 31856720 PMCID: PMC6923859 DOI: 10.1186/s12864-019-6360-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 12/01/2019] [Indexed: 12/13/2022] Open
Abstract
Background Genetic resistance in cattle is considered a suitable way to control tick burden and its consequent losses for livestock production. Exploring tick-resistant (R) and tick-susceptible (S) hosts, we investigated the genetic mechanisms underlying the variation of Braford resistance to tick infestation. Skin biopsies from four-times-artificially infested R (n = 20) and S (n = 19) hosts, obtained before the first and 24 h after the fourth tick infestation were submitted to RNA-Sequencing. Differential gene expression, functional enrichment, and network analysis were performed to identify genetic pathways and transcription factors (TFs) affecting host resistance. Results Intergroup comparisons of hosts before (Rpre vs. Spre) and after (Rpost vs. Spost) tick infestation found 51 differentially expressed genes (DEGs), of which almost all presented high variation (TopDEGs), and 38 were redundant genes. Gene expression was consistently different between R and S hosts, suggesting the existence of specific anti-tick mechanisms. In the intragroup comparisons, Rpost vs. Rpre and Spost vs. Spre, we found more than two thousand DEGs in response to tick infestation in both resistance groups. Redundant and non-redundant TopDEGs with potential anti-tick functions suggested a role in the development of different levels of resistance within the same breed. Leukocyte chemotaxis was over-represented in both hosts, whereas skin degradation and remodeling were only found in TopDEGs from R hosts. Also, these genes indicated the participation of cytokines, such as IL6 and IL22, and the activation of Wingless (WNT)-signaling pathway. A central gene of this pathway, WNT7A, was consistently modulated when hosts were compared. Moreover, the findings based on a genome-wide association study (GWAS) corroborate the prediction of the WNT-signaling pathway as a candidate mechanism of resistance. The regulation of immune response was the most relevant pathway predicted for S hosts. Members of Ap1 and NF-kB families were the most relevant TFs predicted for R and S, respectively. Conclusion This work provides indications of genetic mechanisms presented by Braford cattle with different levels of resistance in response to tick infestation, contributing to the search of candidate genes for tick resistance in bovine.
Collapse
Affiliation(s)
| | - Fernando F Cardoso
- EMBRAPA Pecuária Sul, Bagé, Rio Grande do Sul, Brazil.,Federal University of Pelotas, Capão do Leão, Rio Grande do Sul, Brazil
| | | | | | | | | | | | - Luiz L Coutinho
- Luiz de Queiroz College of Agriculture, University of São Paulo, Piracicaba, São Paulo, Brazil
| | | |
Collapse
|
121
|
Abdel-Dayem MA, Shaker ME, Gameil NM. Impact of interferon β-1b, interferon β-1a and fingolimod therapies on serum interleukins-22, 32α and 34 concentrations in patients with relapsing-remitting multiple sclerosis. J Neuroimmunol 2019; 337:577062. [PMID: 31521828 DOI: 10.1016/j.jneuroim.2019.577062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 08/29/2019] [Accepted: 09/05/2019] [Indexed: 10/26/2022]
Abstract
Interleukins (ILs)-22, 32α and 34 were monitored in the sera of relapsing-remitting multiple sclerosis (RRMS) patients at different time intervals with or without interferon β-1b, interferon β-1a and fingolimod treatments. The results showed that sera of untreated RRMS patients were statistically higher in concentration of IL-22 (P < .001), but not IL-32α and IL-34, than those of healthy individuals. Interestingly, interferon β-1b, interferon β-1a and fingolimod treatments led to a significant decrease of serum concentrations of ILs-22 and 32α, but not 34, at 6 and 12 months of treatment, compared to their initial concentrations before initiating therapy. The correlation analysis revealed that the changes of serum IL-22 (r = 0.814) and, to a lesser extent, IL-32α (r = 0.381) concentrations were positively correlated with those of expanded disability status score. In conclusion, serum IL-22 concentration may be a potential marker for MS disease severity and efficacy of treatment.
Collapse
Affiliation(s)
- Marwa A Abdel-Dayem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Mohamed E Shaker
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacology, Faculty of Pharmacy, Jouf University, Sakaka 2014, Saudi Arabia.
| | - Nariman M Gameil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
122
|
Abstract
Helicobacter pylori is a Gram-negative bacterium that infects the gastric epithelia of its human host. Everyone who is colonized with these pathogenic bacteria can develop gastric inflammation, termed gastritis. Additionally, a small proportion of colonized people develop more adverse outcomes, including gastric ulcer disease, gastric adenocarcinoma, or gastric mucosa-associated lymphoid tissue lymphoma. The development of these adverse outcomes is dependent on the establishment of a chronic inflammatory response. The development and control of this chronic inflammatory response are significantly impacted by CD4+ T helper cell activity. Noteworthy, T helper 17 (Th17) cells, a proinflammatory subset of CD4+ T cells, produce several proinflammatory cytokines that activate innate immune cell antimicrobial activity, drive a pathogenic immune response, regulate B cell responses, and participate in wound healing. Therefore, this review was written to take an intricate look at the involvement of Th17 cells and their affiliated cytokines (interleukin-17A [IL-17A], IL-17F, IL-21, IL-22, and IL-26) in regulating the immune response to H. pylori colonization and carcinogenesis.
Collapse
|
123
|
Howell MD, Kuo FI, Smith PA. Targeting the Janus Kinase Family in Autoimmune Skin Diseases. Front Immunol 2019; 10:2342. [PMID: 31649667 PMCID: PMC6794457 DOI: 10.3389/fimmu.2019.02342] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022] Open
Abstract
Autoimmune skin diseases are characterized by significant local and systemic inflammation that is largely mediated by the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway. Advanced understanding of this pathway has led to the development of targeted inhibitors of Janus kinases (JAKinibs). As a class, JAK inhibitors effectively treat a multitude of hematologic and inflammatory diseases. Growing evidence suggests that JAK inhibitors are efficacious in atopic dermatitis, alopecia areata, psoriasis, and vitiligo. Additional evidence suggests that JAK inhibition might be broadly useful in dermatology, with early reports of efficacy in several other conditions. JAK inhibitors can be administered orally or used topically and represent a promising new class of medications. Here we review the evolving data on the role of the JAK-STAT pathway in inflammatory dermatoses and the potential therapeutic benefit of JAK-STAT antagonism.
Collapse
Affiliation(s)
| | - Fiona I Kuo
- Incyte Corporation, Wilmington, DE, United States
| | - Paul A Smith
- Incyte Corporation, Wilmington, DE, United States
| |
Collapse
|
124
|
lncRNA Mirt2 Is Downregulated in Ulcerative Colitis and Regulates IL-22 Expression and Apoptosis in Colonic Epithelial Cells. Gastroenterol Res Pract 2019; 2019:8154692. [PMID: 31687015 PMCID: PMC6800894 DOI: 10.1155/2019/8154692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/05/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
lncRNA Mirt2 is a lipopolysaccharide- (LPS-) inducible inflammation inhibitor. We found that Mirt2 was downregulated in plasma of ulcerative colitis (UC) patients and the downregulation of Mirt2 distinguished UC patients from healthy controls. IL-22 was also downregulated in UC patients and positively correlated with Mirt2. Mirt2 overexpression led to upregulated, while Mirt2 siRNA silencing led to inhibited secretion of IL-22 from colonic epithelial cells treated with LPS. In addition, under LPS treatment, Mirt2 overexpression led to decreased, while Mirt2 siRNA silencing led to increased apoptotic rate of colonic epithelial cells. Therefore, Mirt2 is downregulated in ulcerative colitis and regulates IL-22 expression in colonic epithelial cells.
Collapse
|
125
|
Abood RN, McHugh KJ, Rich HE, Ortiz MA, Tobin JM, Ramanan K, Robinson KM, Bomberger JM, Kolls JK, Manni ML, Pociask DA, Alcorn JF. IL-22-binding protein exacerbates influenza, bacterial super-infection. Mucosal Immunol 2019; 12:1231-1243. [PMID: 31296910 PMCID: PMC6717528 DOI: 10.1038/s41385-019-0188-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 06/15/2019] [Accepted: 06/24/2019] [Indexed: 02/04/2023]
Abstract
Secondary bacterial pneumonia is a significant complication of severe influenza infection and Staphylococcus aureus and Streptococcus pneumoniae are the primary pathogens of interest. IL-22 promotes S. aureus and S. pneumoniae host defense in the lung through epithelial integrity and induction of antimicrobial peptides and is inhibited by the soluble decoy receptor IL-22-binding protein (IL-22BP). Little is known about the effect of the IL-22/IL-22BP regulatory pathway on lung infection, and it has not been studied in the setting of super-infection. We exposed wild-type and IL-22BP-/- mice to influenza A/PR/8/34 for 6 days prior to infection with S. aureus (USA300) S. pneumoniae. Super-infected IL-22BP-/- mice had decreased bacterial burden and improved survival compared to controls. IL-22BP-/- mice exhibited decreased inflammation, increased lipocalin 2 expression, and deletion of IL-22BP was associated with preserved epithelial barrier function with evidence of improved tight junction stability. Human bronchial epithelial cells treated with IL-22Fc showed evidence of improved tight junctions compared to untreated cells. This study revealed that IL-22BP-/- mice are protected during influenza, bacterial super-infection, suggesting that IL-22BP has a pro-inflammatory role and impairs epithelial barrier function likely through interaction with IL-22.
Collapse
Affiliation(s)
- Robert N Abood
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin J McHugh
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Helen E Rich
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Marianna A Ortiz
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Joshua M Tobin
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Krishnaveni Ramanan
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Keven M Robinson
- Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jennifer M Bomberger
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Michelle L Manni
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Derek A Pociask
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - John F Alcorn
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
126
|
Codenotti S, Mansoury W, Pinardi L, Monti E, Marampon F, Fanzani A. Animal models of well-differentiated/dedifferentiated liposarcoma: utility and limitations. Onco Targets Ther 2019; 12:5257-5268. [PMID: 31308696 PMCID: PMC6613351 DOI: 10.2147/ott.s175710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 06/04/2019] [Indexed: 12/31/2022] Open
Abstract
Liposarcoma is a malignant neoplasm of fat tissue. Well-differentiated and dedifferentiated liposarcoma (WDL/DDL) represent the two most clinically observed histotypes occurring in middle-aged to older adults, particularly within the retroperitoneum or extremities. WDL/DDL are thought to represent the broad spectrum of one disease, as they are both associated with the amplification in the chromosomal 12q13-15 region that causes MDM2 and CDK4 overexpression, the most useful predictor for liposarcoma diagnosis. In comparison to WDL, DDL contains additional genetic abnormalities, principally coamplifications of 1p32 and 6q23, that increase recurrence and metastatic rate. In this review, we discuss the xenograft and transgenic animal models generated for studying progression of WDL/DDL, highlighting utilities and pitfalls in such approaches that can facilitate or impede the development of new therapies.
Collapse
Affiliation(s)
- Silvia Codenotti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Walaa Mansoury
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luca Pinardi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Eugenio Monti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesco Marampon
- Department of Radiotherapy, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Alessandro Fanzani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
127
|
Evasovic JM, Singer CA. Regulation of IL-17A and implications for TGF-β1 comodulation of airway smooth muscle remodeling in severe asthma. Am J Physiol Lung Cell Mol Physiol 2019; 316:L843-L868. [PMID: 30810068 PMCID: PMC6589583 DOI: 10.1152/ajplung.00416.2018] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 02/04/2019] [Accepted: 02/19/2019] [Indexed: 12/14/2022] Open
Abstract
Severe asthma develops as a result of heightened, persistent symptoms that generally coincide with pronounced neutrophilic airway inflammation. In individuals with severe asthma, symptoms are poorly controlled by high-dose inhaled glucocorticoids and often lead to elevated morbidity and mortality rates that underscore the necessity for novel drug target identification that overcomes limitations in disease management. Many incidences of severe asthma are mechanistically associated with T helper 17 (TH17) cell-derived cytokines and immune factors that mediate neutrophilic influx to the airways. TH17-secreted interleukin-17A (IL-17A) is an independent risk factor for severe asthma that impacts airway smooth muscle (ASM) remodeling. TH17-derived cytokines and diverse immune mediators further interact with structural cells of the airway to induce pathophysiological processes that impact ASM functionality. Transforming growth factor-β1 (TGF-β1) is a pivotal mediator involved in airway remodeling that correlates with enhanced TH17 activity in individuals with severe asthma and is essential to TH17 differentiation and IL-17A production. IL-17A can also reciprocally enhance activation of TGF-β1 signaling pathways, whereas combined TH1/TH17 or TH2/TH17 immune responses may additively impact asthma severity. This review seeks to provide a comprehensive summary of cytokine-driven T cell fate determination and TH17-mediated airway inflammation. It will further review the evidence demonstrating the extent to which IL-17A interacts with various immune factors, specifically TGF-β1, to contribute to ASM remodeling and altered function in TH17-driven endotypes of severe asthma.
Collapse
Affiliation(s)
- Jon M Evasovic
- Department of Pharmacology, School of Medicine, University of Nevada , Reno, Nevada
| | - Cherie A Singer
- Department of Pharmacology, School of Medicine, University of Nevada , Reno, Nevada
| |
Collapse
|
128
|
Tang KY, Lickliter J, Huang ZH, Xian ZS, Chen HY, Huang C, Xiao C, Wang YP, Tan Y, Xu LF, Huang YL, Yan XQ. Safety, pharmacokinetics, and biomarkers of F-652, a recombinant human interleukin-22 dimer, in healthy subjects. Cell Mol Immunol 2019; 16:473-482. [PMID: 29670279 PMCID: PMC6474205 DOI: 10.1038/s41423-018-0029-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/26/2018] [Indexed: 12/13/2022] Open
Abstract
F-652 is a recombinant fusion protein consisting of two human interleukin-22 (IL-22) molecules linked to an immunoglobulin constant region (IgG2-Fc). IL-22 plays critical roles in promoting tissue repair and suppressing bacterial infection. The safety, pharmacokinetics (PK), tolerability, and biomarkers of F-652 were evaluated following a single dose in healthy male volunteers in a randomized, double-blind, placebo-controlled study. Following single-dose subcutaneous (SC) injection of F-652 at 2.0 µg/kg into healthy subjects, six out of six subjects experienced delayed injection site reactions, which presented as erythematous and/or discoid eczematous lesions 10 to 17 days post-dosing. F-652 was then administered to the healthy subjects via an intravenous (IV) infusion at 2.0, 10, 30, and 45 µg/kg. No severe adverse event (SAE) was observed during the study. Among the IV-dosed cohorts, eye and skin treatment emergent adverse events (TEAEs) were observed in the 30 and 45 µg/kg cohorts. F-652 IV dosing resulted in linear increases in Cmax and AUC(0-t), and the T1/2 ranged from 39.4 to 206 h in the cohorts. An IV injection of F-652 induced dose-dependent increases in serum marker serum amyloid A, C-reactive protein, and FIB, and decreased serum triglycerides. The serum levels of 36 common pro-inflammatory cytokines/chemokines were not altered by the treatment of F-652 at 45 μg/kg. In conclusion, IV administration of F-652 to healthy male volunteers is safe and well-tolerated and demonstrates favorable PK and pharmacodynamic properties. These results warrant further clinical development of F-652 to treat inflammatory diseases.
Collapse
Affiliation(s)
- Kai-Yang Tang
- Generon (Shanghai) Corporation Ltd., Zhangjiang Hi-Tech Park, Shanghai, China
| | | | - Zhi-Hua Huang
- Generon (Shanghai) Corporation Ltd., Zhangjiang Hi-Tech Park, Shanghai, China
| | - Zong-Shu Xian
- Generon (Shanghai) Corporation Ltd., Zhangjiang Hi-Tech Park, Shanghai, China
| | - Han-Yang Chen
- Generon (Shanghai) Corporation Ltd., Zhangjiang Hi-Tech Park, Shanghai, China
| | - Cheng Huang
- Generon (Shanghai) Corporation Ltd., Zhangjiang Hi-Tech Park, Shanghai, China
| | - Chong Xiao
- Generon (Shanghai) Corporation Ltd., Zhangjiang Hi-Tech Park, Shanghai, China
| | - Yu-Peng Wang
- Generon (Shanghai) Corporation Ltd., Zhangjiang Hi-Tech Park, Shanghai, China
| | - Ying Tan
- Generon (Shanghai) Corporation Ltd., Zhangjiang Hi-Tech Park, Shanghai, China
| | - Lin-Feng Xu
- Generon (Shanghai) Corporation Ltd., Zhangjiang Hi-Tech Park, Shanghai, China
| | - Yu-Liang Huang
- Generon (Shanghai) Corporation Ltd., Zhangjiang Hi-Tech Park, Shanghai, China
| | - Xiao-Qiang Yan
- Generon (Shanghai) Corporation Ltd., Zhangjiang Hi-Tech Park, Shanghai, China.
| |
Collapse
|
129
|
Protopsaltis NJ, Liang W, Nudleman E, Ferrara N. Interleukin-22 promotes tumor angiogenesis. Angiogenesis 2019; 22:311-323. [PMID: 30539314 DOI: 10.1007/s10456-018-9658-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 12/07/2018] [Indexed: 01/08/2023]
Abstract
TH17 cells play important yet complex roles in cancer development and progression. We previously reported that TH17 cells and IL-17 mediate resistance to anti-VEGF therapy by inducing recruitment of immunosuppressive and proangiogenic myeloid cells to the tumor microenvironment. Here, we demonstrate that IL-22, a key effector cytokine expressed by TH17 cells, directly acts on endothelial cells to promote tumor angiogenesis. IL-22 induces endothelial cell proliferation, survival, and chemotaxis in vitro and neovascularization in an ex vivo mouse choroid explant model. Blockade of IL-22, with a neutralizing antibody, significantly inhibits tumor growth associated with reduced microvascular density. No synergistic effect of IL-22 with VEGF was observed. These results identify IL-22 as a potential therapeutic target for blocking tumor angiogenesis.
Collapse
Affiliation(s)
| | - Wei Liang
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Eric Nudleman
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, USA
| | - Napoleone Ferrara
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
130
|
Li Y, Wang J, Li Y, Wu H, Zhao S, Yu Q. Protecting intestinal epithelial cells against deoxynivalenol and E. coli damage by recombinant porcine IL-22. Vet Microbiol 2019; 231:154-159. [PMID: 30955803 PMCID: PMC7172643 DOI: 10.1016/j.vetmic.2019.02.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 06/01/2018] [Accepted: 02/19/2019] [Indexed: 12/29/2022]
Abstract
Pigs suffer enteritis induced by pathogenic bacteria infection and toxins in the moldy feed, which cause intestinal epithelial damage and diarrhea through the whole breeding cycle. Interleukin-22 (IL-22) plays a critical role in maintaining intestinal mucosal barrier function through repairing intestinal epithelial damage. However, little was known about the effects of IL-22 against apoptosis caused by toxins and infection of intestinal pathogens in the intestinal epithelium, especially in pigs. In this study, we had successfully used prokaryotic expression system to produce recombinant porcine interleukin-22. Meanwhile, purified rIL-22 could activate STAT3 signal pathway and have been demonstrated to be safe to IPEC-J2 cells by increasing E-cadherin expression, without proinflammatory cytokines changes. Furthermore, rIL-22 reversed apoptosis induced by deoxynivalenol (DON) and played a vital part in repairing the intestinal injury. We also found that rIL-22 stimulated epithelial cells to secrete pBD-1 against enterotoxigenic E. coli (ETEC) K88 infection, as well as alleviating apoptosis ratio. This study provided a theoretical basis for curing intestinal inflammation caused by ETEC infection and epithelial apoptosis induced by DON with rIL-22 in pigs.
Collapse
Affiliation(s)
- Yunyun Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Jinquan Wang
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, Xinjiang, 830052, PR China
| | - Yuchen Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Haiqin Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Shiyi Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Qinghua Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China.
| |
Collapse
|
131
|
Abdallah F, Lecellier G, Raharivelomanana P, Pichon C. R. nukuhivensis acts by reinforcing skin barrier function, boosting skin immunity and by inhibiting IL-22 induced keratinocyte hyperproliferation. Sci Rep 2019; 9:4132. [PMID: 30858525 PMCID: PMC6411885 DOI: 10.1038/s41598-019-39831-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/31/2019] [Indexed: 12/27/2022] Open
Abstract
Rauvolfia nukuhivensis is a well-known plant used for its wide range of beneficial effects in Marquesas islands. It is made up of diverse indole alkaloids and is used as traditional medicine for skin application. The actual mechanism behind the virtue of this plant is still unknown. Hence, in this study we aimed at deciphering the impact of R. nukuhivensis on skin immune system in context of (1) homeostasis, (2) pathogen infection and (3) inflammation. Here we show that R. nukuhivensis enhances cellular metabolic activity and wound healing without inducing cellular stress or disturbing cellular homeostasis. It reinforces the epithelial barrier by up-regulating hBD-1. Nevertheless, in pathogenic stress, R. nukuhivensis acts by preparing the immune system to be reactive and effective directly. Indeed, it enhances the innate immune response by increasing pathogens sensors such as TLR5. Finally, R. nukuhivensis blocks IL-22 induced hyperproliferation via PTEN and Filaggrin up-regulation as well as BCL-2 downregulation. In conclusion, this study provides evidence on the several cutaneous application potentials of R. nukuhivensis such as boosting the immune response or in restoring the integrity of the epithelial barrier.
Collapse
Affiliation(s)
- Florence Abdallah
- Centre de Biophysique Moléculaire, CNRS-UPR4301, 45071, Orléans, France
| | - Gaël Lecellier
- Université de Paris-Saclay UVSQ, 55 Avenue de Paris, 78000, Versailles, France
| | - Phila Raharivelomanana
- Université de la Polynésie Française, UMR 241 EIO, 6570 - 98702, Faa'a, Tahiti, Polynésie Française
| | - Chantal Pichon
- Centre de Biophysique Moléculaire, CNRS-UPR4301, 45071, Orléans, France.
- Université d'Orléans, Collegium Sciences et Techniques, 45100, Orléans, France.
| |
Collapse
|
132
|
Prutsch N, Gurnhofer E, Suske T, Liang HC, Schlederer M, Roos S, Wu LC, Simonitsch-Klupp I, Alvarez-Hernandez A, Kornauth C, Leone DA, Svinka J, Eferl R, Limberger T, Aufinger A, Shirsath N, Wolf P, Hielscher T, Sternberg C, Aberger F, Schmoellerl J, Stoiber D, Strobl B, Jäger U, Staber PB, Grebien F, Moriggl R, Müller M, Inghirami GG, Sanda T, Look AT, Turner SD, Kenner L, Merkel O. Dependency on the TYK2/STAT1/MCL1 axis in anaplastic large cell lymphoma. Leukemia 2019; 33:696-709. [PMID: 30131584 PMCID: PMC8076043 DOI: 10.1038/s41375-018-0239-1] [Citation(s) in RCA: 36] [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: 02/25/2018] [Revised: 07/02/2018] [Accepted: 07/27/2018] [Indexed: 12/11/2022]
Abstract
TYK2 is a member of the JAK family of tyrosine kinases that is involved in chromosomal translocation-induced fusion proteins found in anaplastic large cell lymphomas (ALCL) that lack rearrangements activating the anaplastic lymphoma kinase (ALK). Here we demonstrate that TYK2 is highly expressed in all cases of human ALCL, and that in a mouse model of NPM-ALK-induced lymphoma, genetic disruption of Tyk2 delays the onset of tumors and prolongs survival of the mice. Lymphomas in this model lacking Tyk2 have reduced STAT1 and STAT3 phosphorylation and reduced expression of Mcl1, a pro-survival member of the BCL2 family. These findings in mice are mirrored in human ALCL cell lines, in which TYK2 is activated by autocrine production of IL-10 and IL-22 and by interaction with specific receptors expressed by the cells. Activated TYK2 leads to STAT1 and STAT3 phosphorylation, activated expression of MCL1 and aberrant ALCL cell survival. Moreover, TYK2 inhibitors are able to induce apoptosis in ALCL cells, regardless of the presence or absence of an ALK-fusion. Thus, TYK2 is a dependency that is required for ALCL cell survival through activation of MCL1 expression. TYK2 represents an attractive drug target due to its essential enzymatic domain, and TYK2-specific inhibitors show promise as novel targeted inhibitors for ALCL.
Collapse
Affiliation(s)
- Nicole Prutsch
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Elisabeth Gurnhofer
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Tobias Suske
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Huan Chang Liang
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Michaela Schlederer
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Simone Roos
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Lawren C Wu
- Department of Oncology, Amgen Discovery Research, South San Francisco, CA, 94080, USA
| | | | | | - Christoph Kornauth
- Clinical Institute of Pathology, Medical University of Vienna, Vienna, Austria
| | - Dario A Leone
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Jasmin Svinka
- Institute of Cancer Research, Medical University of Vienna & Comprehensive Cancer Center (CCC), Vienna, Austria
| | - Robert Eferl
- Institute of Cancer Research, Medical University of Vienna & Comprehensive Cancer Center (CCC), Vienna, Austria
| | - Tanja Limberger
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Astrid Aufinger
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria
| | - Nitesh Shirsath
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| | - Peter Wolf
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christina Sternberg
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Molecular Biology, Cancer Cluster Salzburg, Faculty of Natural Sciences, Paris Lodron University, Salzburg, Austria
- Department of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Fritz Aberger
- Department of Molecular Biology, Cancer Cluster Salzburg, Faculty of Natural Sciences, Paris Lodron University, Salzburg, Austria
| | | | - Dagmar Stoiber
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ulrich Jäger
- Department of Medicine I, Clinical Division of Hematology and Hemostaseology and Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria
| | - Philipp B Staber
- Department of Medicine I, Clinical Division of Hematology and Hemostaseology and Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria
| | - Florian Grebien
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
- Medical University of Vienna, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Giorgio G Inghirami
- Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NYC, USA
- European Research Initiative for ALK related malignancies (www.erialcl.net), Vienna, Austria
| | - Takaomi Sanda
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore, Singapore
| | - A Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, USA
| | - Suzanne D Turner
- European Research Initiative for ALK related malignancies (www.erialcl.net), Vienna, Austria
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Lukas Kenner
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria.
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, Vienna, Austria.
- Ludwig Boltzmann Institute for Cancer Research (LBI-CR), Vienna, Austria.
- European Research Initiative for ALK related malignancies (www.erialcl.net), Vienna, Austria.
- CBMed Core Lab2, Medical University of Vienna, Vienna, Austria.
| | - Olaf Merkel
- Clinical Institute of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, Vienna, Austria.
- European Research Initiative for ALK related malignancies (www.erialcl.net), Vienna, Austria.
| |
Collapse
|
133
|
Ito T, Hirose K, Nakajima H. Bidirectional roles of IL-22 in the pathogenesis of allergic airway inflammation. Allergol Int 2019; 68:4-8. [PMID: 30424940 DOI: 10.1016/j.alit.2018.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/27/2018] [Accepted: 10/08/2018] [Indexed: 02/07/2023] Open
Abstract
Asthma is the most prevalent allergic disease of the airway, which is characterized by eosinophilic inflammation, mucus hyperproduction, and airway hyper-responsiveness. Although these pathognomonic features are mainly mediated by antigen-specific Th2 cells and their cytokines, such as IL-4, IL-5, and IL-13, recent studies have revealed that other inflammatory cells, including Th17 cells and innate lymphoid cells (ILCs), also play a critical role in the pathogenesis of asthma. IL-22, one of the cytokines produced by Th17 cells and type 3 ILCs, has distinct functional properties, as IL-22 exclusively acts on non-hematopoietic cells including epithelial cells of mucosal surface and exhibits a broad range of action in regeneration and host protection. In accordance with the fact that lung epithelial cells play a critical role in the pathogenesis of asthma, we and other groups have shown that IL-22 is involved in the regulation of allergic airway inflammation. In this review, we discuss recent advances in the biology of IL-22 and its involvement in the pathogenesis of allergic airway inflammation.
Collapse
Affiliation(s)
- Takashi Ito
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan; Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, Japan
| | - Koichi Hirose
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan; Department of Rheumatology, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
134
|
Xiu D, Wang D, Wang J, Ji F, Zhang W. MicroRNA-543 suppresses liver cancer growth and induces apoptosis via the JAK2/STAT3 signaling pathway. Oncol Lett 2018; 17:2451-2456. [PMID: 30675310 DOI: 10.3892/ol.2018.9838] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/06/2018] [Indexed: 12/17/2022] Open
Abstract
The aim of the present study was to investigate the function of microRNA-543 on liver cancer cell proliferation and apoptosis, and also to identify whether the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway was a direct target of microRNA-543. When compared with paracarcinoma tissue, microRNA-543 expression in tissue samples from patients with liver cancer was identified to be decreased. Furthermore, overall survival of patients with high microRNA-543 expression was increased compared with patients with low microRNA-543 expression. Inhibition of microRNA-543 expression increased cell proliferation and decreased apoptosis of liver cancer cells. It also activated the protein expression of phosphorylated JAK2, phosphorylated STAT3, c-Myc and B-cell lymphoma 2 (Bcl-2) in liver cancer cells. However, activation of microRNA-543 expression in turn decreased cell proliferation and increased apoptosis of liver cancer cells. The results of the present study highlight the pivotal function of microRNA-543 as an inhibitor in liver cancer cell proliferation by observing JAK2/STAT3/c-Myc/Bcl-2 in liver cancer.
Collapse
Affiliation(s)
- Dianhui Xiu
- Department of Radiology, The China Japan Union Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dawei Wang
- Department of Emergency, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jing Wang
- Department of Gastrointestinal Medicine, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Fujian Ji
- Department of General Surgery, The China Japan Union Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wenlei Zhang
- Department of Interventional Therapy, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
135
|
Miyazaki Y, Nakayamada S, Kubo S, Nakano K, Iwata S, Miyagawa I, Ma X, Trimova G, Sakata K, Tanaka Y. Th22 Cells Promote Osteoclast Differentiation via Production of IL-22 in Rheumatoid Arthritis. Front Immunol 2018; 9:2901. [PMID: 30619268 PMCID: PMC6295478 DOI: 10.3389/fimmu.2018.02901] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/26/2018] [Indexed: 12/29/2022] Open
Abstract
T helper (Th) cells can differentiate into functionally distinct subsets and play a pivotal role in inflammatory and autoimmune diseases such as rheumatoid arthritis (RA). Th22 cells have been identified as a new subset secreting interleukin (IL)-22. Although elevated levels of IL-22 in the synovial fluids of RA patients were reported, its pathological roles remain unclear. Here, we demonstrated that IL-22 was characteristically produced from CD3+CD4+CC-chemokine receptor (CCR)4+CCR6+CCR10+ cells and their ability of the production of IL-22 markedly exceeded that of other Th subsets and the subset, thereby, designated Th22 cells. Th22 cells were efficiently induced by the stimulation with tumor necrosis factor-α, IL-6, and IL-1β. Th22 cells were markedly infiltrated in synovial tissue in patients with active RA, but not in patients with osteoarthritis (OA). CCL17, CCL20, and CCL28, which are chemokine ligands of CCR4, CCR6, and CCR10, respectively, were abundantly expressed in RA synovial tissue compared to OA. By in vitro Trans-well migration assay, Th22 cells efficiently migrated toward CCL28. Co-culture of Th22 cells, which were sorted from peripheral blood, with monocytes in the presence of macrophage colony-stimulating factor and receptor activator of nuclear factor (NF)-κB ligand induced osteoclasts formation more efficiently than that of either Th1 cells or Th17 cells. Furthermore, IL-22 markedly augmented osteoclast differentiation by promoting nuclear factor of activated T cells c1 expression in CD14+ monocytes. Contrarily, the addition of IFN-γ to the culture significantly decreased osteoclasts number, whereas IL-17 had marginal effects. IL-22 neutralizing antibody inhibited osteoclast formation in the co-culture of Th22 cells with CD14+ monocytes. Collectively, the results indicated that Th22 cells, which co-express chemokine receptors CCR4, CCR6, and CCR10, possess strong potency of tissue migration and accumulate into inflamed synovial tissues where the ligands such as CCL28 are highly expressed. Thus, Th22 cells have the capacity to promote osteoclast differentiation through production of IL-22 and thus play a pivotal role in bone destruction in patients with RA.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Arthritis, Rheumatoid/complications
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/surgery
- Arthroplasty, Replacement, Knee
- Cell Differentiation/immunology
- Cells, Cultured
- Chemokines, CC/immunology
- Chemokines, CC/metabolism
- Coculture Techniques
- Humans
- Interleukins/immunology
- Interleukins/metabolism
- Male
- Middle Aged
- Osteoarthritis/immunology
- Osteoarthritis/pathology
- Osteoarthritis/surgery
- Osteoclasts/physiology
- Primary Cell Culture
- Receptors, Chemokine/immunology
- Receptors, Chemokine/metabolism
- Synovial Membrane/cytology
- Synovial Membrane/immunology
- Synovial Membrane/pathology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- Interleukin-22
Collapse
Affiliation(s)
- Yusuke Miyazaki
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Satoshi Kubo
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Kazuhisa Nakano
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Shigeru Iwata
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Ippei Miyagawa
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Xiaoxue Ma
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Gulzhan Trimova
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| | - Kei Sakata
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
- Research Unit Immunology & Inflammation, Innovative Research Division, Mitsubishi Tanabe Pharma, Yokohama, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Kitakyushu, Japan
| |
Collapse
|
136
|
da Cunha Colombo Tiveron LR, da Silva IR, da Silva MV, Peixoto AB, Rodrigues DBR, Rodrigues V. High in situ mRNA levels of IL-22, TFG-β, and ARG-1 in keloid scars. Immunobiology 2018; 223:812-817. [PMID: 30146129 DOI: 10.1016/j.imbio.2018.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/18/2018] [Accepted: 08/19/2018] [Indexed: 12/21/2022]
Abstract
Keloid scars are currently considered a chronic inflammatory process and no longer a benign skin tumor. Keloids are defined as highly inflamed, hyperproliferative pathological scars. Growth factors and cytokines have important functions in the keloid inflammatory etiopathogenesis. The aim of this study was to analyze the in situ expression of cytokines and growth factors in keloid scars in comparison with that in normal scars. Among them, we specifically assessed TGF-β, FGF, IL-33, IL-22, ARG-1, ARG-2, iNOS, VIP, VIP-R1, TAC, and TAC-R1. A total of 98 biopsies were evaluated, including of 53 keloid and 45 normal scars. The age of patients with keloids ranged from 11 to 73 years, with a mean age of 28 years and predominance of the female gender (58.5% of the total patients). Around 64.15% of the patients belonged to the black ethnic group. Evaluated keloids were most commonly located in the earlobe because of ear piercing, representing 73.6% of the cases. We found significantly greater expression of TGF-β, IL-22, and ARG-1 in keloids when compared with that in normal scars. As for IL-33, ARG-2, and VIP-R1, despite the higher number of mRNA copies found in keloids, this difference was not significant. Furthermore, FGF, iNOS, VIP, TAC, and TAC-R1 mRNA levels were not detectable, and therefore these results were inconclusive in this study. Considering these results, understanding the cellular and molecular mechanisms that control the inflammatory response during cutaneous healing may promote the development of strategies to improve the treatment of patients with keloids.
Collapse
Affiliation(s)
| | - Isabela Rios da Silva
- Federal University of Triângulo Mineiro (UFTM) ICBN and CEFORES, Uberaba, MG, Brazil
| | | | - Alberto Borges Peixoto
- Laboratory of Biopathology and Molecular Biology, University of Uberaba (UNIUBE), Uberaba, MG, Brazil
| | - Denise Bertulucci Rocha Rodrigues
- Federal University of Triângulo Mineiro (UFTM) ICBN and CEFORES, Uberaba, MG, Brazil; Laboratory of Biopathology and Molecular Biology, University of Uberaba (UNIUBE), Uberaba, MG, Brazil
| | - Virmondes Rodrigues
- Federal University of Triângulo Mineiro (UFTM) ICBN and CEFORES, Uberaba, MG, Brazil.
| |
Collapse
|
137
|
Burmeister AR, Marriott I. The Interleukin-10 Family of Cytokines and Their Role in the CNS. Front Cell Neurosci 2018; 12:458. [PMID: 30542269 PMCID: PMC6277801 DOI: 10.3389/fncel.2018.00458] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022] Open
Abstract
Resident cells of the central nervous system (CNS) play an important role in detecting insults and initiating protective or sometimes detrimental host immunity. At peripheral sites, immune responses follow a biphasic course with the rapid, but transient, production of inflammatory mediators giving way to the delayed release of factors that promote resolution and repair. Within the CNS, it is well known that glial cells contribute to the onset and progression of neuroinflammation, but it is only now becoming apparent that microglia and astrocytes also play an important role in producing and responding to immunosuppressive factors that serve to limit the detrimental effects of such responses. Interleukin-10 (IL-10) is generally considered to be the quintessential immunosuppressive cytokine, and its ability to resolve inflammation and promote wound repair at peripheral sites is well documented. In the present review article, we discuss the evidence for the production of IL-10 by glia, and describe the ability of CNS cells, including microglia and astrocytes, to respond to this suppressive factor. Furthermore, we review the literature for the expression of other members of the IL-10 cytokine family, IL-19, IL-20, IL-22 and IL-24, within the brain, and discuss the evidence of a role for these poorly understood cytokines in the regulation of infectious and sterile neuroinflammation. In concert, the available data indicate that glia can produce IL-10 and the related cytokines IL-19 and IL-24 in a delayed manner, and these cytokines can limit glial inflammatory responses and/or provide protection against CNS insult. However, the roles of other IL-10 family members within the CNS remain unclear, with IL-20 appearing to act as a pro-inflammatory factor, while IL-22 may play a protective role in some instances and a detrimental role in others, perhaps reflecting the pleiotropic nature of this cytokine family. What is clear is that our current understanding of the role of IL-10 and related cytokines within the CNS is limited at best, and further research is required to define the actions of this understudied family in inflammatory brain disorders.
Collapse
Affiliation(s)
- Amanda R Burmeister
- Department of Biological Sciences, The University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Ian Marriott
- Department of Biological Sciences, The University of North Carolina at Charlotte, Charlotte, NC, United States
| |
Collapse
|
138
|
Li P, Shi X, Xu Y, Zhong B, Lu Y, Sun Y. Interleukin-22 Promotes Osteosarcoma Cell Proliferation and Invasion via STAT3 Activation. Med Sci Monit 2018; 24:7802-7808. [PMID: 30381753 PMCID: PMC6225732 DOI: 10.12659/msm.910962] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Interleukin-22 (IL-22) is one of the cytokines secreted by T-helper 17 (Th17) cells. It belongs to the IL-10 cytokine family and influences a variety of immune reactions. Studies have indicated that IL-22 can promote cancer progression and metastases. However, the function of IL-22 in osteosarcoma (OS) remains unclear. MATERIAL AND METHODS In this study, the expression of IL-22 in the OS cell line was detected by qRT-PCR. The role of IL-22 in proliferation and invasion in OS cells was tested by MTT and Transwell assays. The protein expression of STAT3, phospho-STAT3, AKT, and phospho-AKT was detected by Western blot analysis. RESULTS The results showed that IL-22 was upregulated in OS cells. IL-22 dose-independently promoted OS cells proliferation and invasion, which could be reversed by IL-22 antibody or STAT3 siRNA. Furthermore, IL-22 exposure of OS cells resulted in dose-independently increased levels of phosphorylated STAT3 protein kinases. Interestingly, IL-22 did not influence the expression of phosphorylated AKT. CONCLUSIONS These results suggest that IL-22 promotes OS cells proliferation and invasion and its effect is mediated by activation of the STAT3 pathway. These findings demonstrate that IL-22 may serve as a promising molecular biomarker for diagnosis and therapy for OS patients.
Collapse
Affiliation(s)
- Panpan Li
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Xin Shi
- Department of Traumatology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Yonghui Xu
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Binggang Zhong
- Department of Orthopedics, The First People’s Hospital of Zhao Tong, Zhaotong, Yunnan, P.R. China
| | - Yu Lu
- Department of Orthopedics, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Yong Sun
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| |
Collapse
|
139
|
Zheng Y, Li T. Interleukin-22, a potent target for treatment of non-autoimmune diseases. Hum Vaccin Immunother 2018; 14:2811-2819. [PMID: 30335564 DOI: 10.1080/21645515.2018.1509649] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Interleukin -22 (IL-22) is a member of interleukin-10 (IL-10) family cytokines that is produced by different types of lymphocytes included in both innate and adaptive immune systems. These lymphocytes include activated T cells, most notably Th17 and Th22 cells, as well as NK cells, γδ T cells, etc. IL-22 mediate its effects via the IL-22-IL-22R complex and subsequent Janus Kinase-signal transduces and activators transcription (JAK-STAT) signaling pathway. According to recent evidence, IL-22 played a critical role in the pathogenesis of many non-autoimmune diseases. In this review, we mainly discussed the recent findings and advancements of the role of IL-22 in several non-autoimmune diseases, such as acute lung injury, atherosclerosis and some bacterial infections, suggesting that IL-22 may have therapeutic potential for treating non-autoimmune diseases.
Collapse
Affiliation(s)
- Yue Zheng
- a Cardiology , The Third Central Clinical College of Tianjin Medical University , Tianjin , China.,b Cardiology , Tianjin Key Laboratory of Artificial Cell.,c Artificial Cell Engineering Technology Research Center of Public Health Ministry , Tianjin , China.,d Tianjin Institute of Hepatobiliary Disease , Tianjin , China
| | - Tong Li
- b Cardiology , Tianjin Key Laboratory of Artificial Cell.,c Artificial Cell Engineering Technology Research Center of Public Health Ministry , Tianjin , China.,d Tianjin Institute of Hepatobiliary Disease , Tianjin , China.,e The Third Central Hospital of Tianjin , Tianjin , China
| |
Collapse
|
140
|
Wang S, Yao Y, Yao M, Fu P, Wang W. Interleukin-22 promotes triple negative breast cancer cells migration and paclitaxel resistance through JAK-STAT3/MAPKs/AKT signaling pathways. Biochem Biophys Res Commun 2018; 503:1605-1609. [PMID: 30072097 DOI: 10.1016/j.bbrc.2018.07.088] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 07/18/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE Owing to the absence of any targeted therapies, triple negative breast cancer (TNBC), which accounts for approximately 15% of all breast cancers, typically have a poorer outcome. In an attempt to find out the TNBCs' microenvironment, we ran into the endogenous expression of a newly discovered cytokine known as Interleukin (IL)-22. METHODS Thirty TNBC patients were recruited and the levels of IL-22 producing (Th22) cells in tumor, paratumor and normal breast tissues were measured. Then we studied the role and mechanism of IL-22 in migration and paclitaxel resistance in TNBC cells MDA-MB 468 and MDA-MB 231. RESULTS The prevalence of Th22 cells were gradually increased in normal, paratumor and tumor tissues. In vitro, IL-22 promotes TNBC cells migration and induces paclitaxel resistance. Importantly, IL-22 exposure of TNBC cells resulted in JAK-STAT3/MAPKs/AKT signaling pathways activated. CONCLUSION IL-22 may play an accelerating role in the development of TNBC and may open a new therapeutic approach for TNBC.
Collapse
Affiliation(s)
- Shuqian Wang
- Department of Breast Surgery, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, 310003, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, 310003, China.
| | - Yinan Yao
- Department of Respiratory Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China.
| | - Minya Yao
- Department of Breast Surgery, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, 310003, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, 310003, China.
| | - Peifen Fu
- Department of Breast Surgery, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou, Zhejiang, 310003, China.
| | - Weilin Wang
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Division of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
141
|
Jiang R, Zhang GR, Zhu DM, Shi ZC, Liao CL, Fan QX, Wei KJ, Ji W. Molecular characterization and expression analysis of IL-22 and its two receptors genes in yellow catfish (Pelteobagrus filvidraco) in response to Edwardsiella ictaluri challenge. FISH & SHELLFISH IMMUNOLOGY 2018; 80:250-263. [PMID: 29886141 DOI: 10.1016/j.fsi.2018.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/30/2018] [Accepted: 06/06/2018] [Indexed: 06/08/2023]
Abstract
Interleukin (IL)-22, as a member of the interleukin (IL)-10 family, is an important mediator between the immune cells and epithelial tissues during infection and inflammation. This study reported the characterization and mRNA expression patterns of Pf_IL-22 gene and its cell surface-associated receptors Pf_IL-22RA1 and soluble Pf_IL-22RA2 genes in yellow catfish (Pelteobagrus filvidraco). The open reading frames (ORFs) of the Pf_IL-22, Pf_IL-22RA1 and Pf_IL-22RA2 genes were 546 bp, 1740 bp and 690 bp in length, encoding 181, 579 and 229 amino acids, respectively. Alignments of the deduced amino acid sequences present that the Pf_IL-22 has a conserved IL-10 family signature motif, and the Pf_IL-22RA1 and Pf_IL-22RA2 have two conserved fibronectin type-III domains. Quantitative real-time PCR (qPCR) analyses showed that the Pf_IL-22 and Pf_IL-22RA1 mRNAs were highly expressed in mucosal tissues such as the fin, gill, intestine, skin mucus and stomach, and were weakly expressed in the kidney, liver and head kidney of adult yellow catfish, indicating that the Pf_IL-22 transcripts may be mainly produced by mucosal immune cells/tissues in healthy yellow catfish. The mRNA expression levels of the Pf_IL-22RA2 gene were high in the muscle and liver, and were relatively low in the spleen and kidney. The mRNA expression levels of the Pf_IL-22 and its two receptor genes were significantly up-regulated in both mucosal tissues (gill, hindgut, and skin mucus) and systemic immune tissues (spleen, head kidney and blood) after Edwardsiella ictaluri challenge. These results indicated that the Pf_IL-22 and its two receptors genes might play an important role in the innate immune defense against bacterial invasion.
Collapse
Affiliation(s)
- Rui Jiang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, PR China; Freshwater Aquaculture Collaborative Innovation Centre of Hubei Province, Wuhan, 430070, PR China
| | - Gui-Rong Zhang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, PR China; Freshwater Aquaculture Collaborative Innovation Centre of Hubei Province, Wuhan, 430070, PR China
| | - Dong-Mei Zhu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, PR China; Freshwater Aquaculture Collaborative Innovation Centre of Hubei Province, Wuhan, 430070, PR China
| | - Ze-Chao Shi
- Freshwater Aquaculture Collaborative Innovation Centre of Hubei Province, Wuhan, 430070, PR China; Key Laboratory of Freshwater Biodiversity Conservation, Ministry of Agriculture, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, 30223, PR China
| | - Chen-Lei Liao
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, PR China; Freshwater Aquaculture Collaborative Innovation Centre of Hubei Province, Wuhan, 430070, PR China
| | - Qi-Xue Fan
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, PR China; Freshwater Aquaculture Collaborative Innovation Centre of Hubei Province, Wuhan, 430070, PR China
| | - Kai-Jian Wei
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, PR China; Freshwater Aquaculture Collaborative Innovation Centre of Hubei Province, Wuhan, 430070, PR China.
| | - Wei Ji
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, PR China; Freshwater Aquaculture Collaborative Innovation Centre of Hubei Province, Wuhan, 430070, PR China.
| |
Collapse
|
142
|
Weinberg FD, Ramnath N. Targeting IL22: a potential therapeutic approach for Kras mutant lung cancer? Transl Lung Cancer Res 2018; 7:S243-S247. [PMID: 30393613 DOI: 10.21037/tlcr.2018.09.04] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Frank D Weinberg
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI 48109, USA
| | - Nithya Ramnath
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Rogel Cancer Center, Ann Arbor, MI 48109, USA
| |
Collapse
|
143
|
Hirose K, Ito T, Nakajima H. Roles of IL-22 in allergic airway inflammation in mice and humans. Int Immunol 2018; 30:413-418. [PMID: 29394345 DOI: 10.1093/intimm/dxy010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 01/28/2018] [Indexed: 12/17/2023] Open
Abstract
Asthma is a chronic inflammatory disease of the airways that is characterized by eosinophilic inflammation, mucus hypersecretion and airway remodeling that leads to airway obstruction. Although these pathognomonic features of asthma are primarily mediated by allergen-specific T helper type 2 cells (Th2 cells) and their cytokines, recent studies have revealed critical roles of lung epithelial cells in the pathogenesis of asthma. Lung epithelial cells not only form physical barriers by covering the surfaces of the airways but also sense inhaled allergens and initiate communication between the environment and the immune system. The causative involvement of lung epithelium in the pathogenesis of asthma suggests that some molecules that modulate epithelial function have a regulatory role in asthma. IL-22, an IL-10-family cytokine produced by IL-17A-producing T helper cells (Th17 cells), γδ T cells and group 3 innate lymphoid cells (ILC3s), primarily targets epithelial cells and promotes their proliferation. In addition, IL-22 has been shown to induce epithelial production of various molecules that regulate local immune responses. These findings indicate that IL-22 plays crucial roles in the pathogenesis of asthma by regulating epithelial function. Here, we review the current understanding of the molecular and cellular mechanisms underlying IL-22-mediated regulation of airway inflammation in asthma.
Collapse
Affiliation(s)
- Koichi Hirose
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
- Department of Rheumatology, School of Medicine, International University of Health and Welfare, Narita City, Chiba, Japan
| | - Takashi Ito
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba, Japan
| |
Collapse
|
144
|
Wang R, Zeng Y, Qin H, Lu Y, Huang H, Lei M, Tan T, Huang Y, Luo H, Lan Y, Wei Y. Association of interleukin 22 gene polymorphisms and serum IL-22 level with risk of systemic lupus erythematosus in a Chinese population. Clin Exp Immunol 2018; 193:143-151. [PMID: 29603203 PMCID: PMC6046499 DOI: 10.1111/cei.13133] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2018] [Indexed: 01/12/2023] Open
Abstract
The aim of this study was to investigate the association between the single-nucleotide polymorphisms (SNPs) of the interleukin 22 (IL-22) gene and systemic lupus erythematosus (SLE) in a Chinese population. Three IL-22 SNPs (rs2227485, rs2227513 and rs2227491) were genotyped using SNaPshot SNP genotyping assays and identified by sequencing in 314 SLE patients and 411 healthy controls. The IL-22 level of serum was assessed by enzyme-linked immunosorbent assay (ELISA) kits. Data were analysed by spss version 17.0 software. We found that rs2227513 was associated with an increased risk of SLE [AG versus AA: adjusted odds ratio (aOR) = 2·24, 95% confidence interval (CI) = 1·22-4·12, P = 0·010; G versus· A: adjusted OR = 2·18, 95% CI = 1·20-3·97, P = 0·011]. Further analysis in patients with SLE showed that the AG genotype and G allele were associated with an increased risk of renal disorder in SLE (G versus A: aOR = 3·09, 95% CI = 1·30-7·33, P = 0·011; AG versus· AA: aOR = 3·25, 95% CI = 1·35-7·85, P = 0·009). In addition, the concentration of IL-22 was significantly lower in the rs2227513 AG genotype compared with AA genotype (P = 0·028). These results suggest that rs2227513 polymorphism might contribute to SLE susceptibility, probably by decreasing the expression of IL-22.
Collapse
Affiliation(s)
- R. Wang
- Clinical Medical School, Youjiang Medical University for NationalitiesBaiseGuangxiChina
- Department of Laboratory Medicinethe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - Y.‐L. Zeng
- Department of Laboratory Medicinethe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - H.‐M. Qin
- Clinical Medical School, Youjiang Medical University for NationalitiesBaiseGuangxiChina
- Department of Laboratory Medicinethe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - Y.‐L. Lu
- Clinical Medical School, Youjiang Medical University for NationalitiesBaiseGuangxiChina
- Department of Laboratory Medicinethe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - H.‐T. Huang
- Clinical Medical School, Youjiang Medical University for NationalitiesBaiseGuangxiChina
- Department of Laboratory Medicinethe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - M. Lei
- Clinical Medical School, Youjiang Medical University for NationalitiesBaiseGuangxiChina
- Department of Laboratory Medicinethe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - T. Tan
- Clinical Medical School, Youjiang Medical University for NationalitiesBaiseGuangxiChina
- Department of Laboratory Medicinethe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - Y.‐Y. Huang
- Clinical Medical School, Youjiang Medical University for NationalitiesBaiseGuangxiChina
- Department of Laboratory Medicinethe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - H.‐C. Luo
- Department of Laboratory Medicinethe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - Y. Lan
- Department of Dermatologythe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - Y.‐S. Wei
- Department of Laboratory Medicinethe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| |
Collapse
|
145
|
Yin S, Feng D. WITHDRAWN: IL-22 in liver inflammation, injury and repair. LIVER RESEARCH 2018. [DOI: 10.1016/j.livres.2017.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
146
|
Zwarycz B, Gracz AD, Rivera KR, Williamson IA, Samsa LA, Starmer J, Daniele MA, Salter-Cid L, Zhao Q, Magness ST. IL22 Inhibits Epithelial Stem Cell Expansion in an Ileal Organoid Model. Cell Mol Gastroenterol Hepatol 2018; 7:1-17. [PMID: 30364840 PMCID: PMC6199238 DOI: 10.1016/j.jcmgh.2018.06.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 06/25/2018] [Indexed: 02/07/2023]
Abstract
Background & Aims Crohn's disease is an inflammatory bowel disease that affects the ileum and is associated with increased cytokines. Although interleukin (IL)6, IL17, IL21, and IL22 are increased in Crohn's disease and are associated with disrupted epithelial regeneration, little is known about their effects on the intestinal stem cells (ISCs) that mediate tissue repair. We hypothesized that ILs may target ISCs and reduce ISC-driven epithelial renewal. Methods A screen of IL6, IL17, IL21, or IL22 was performed on ileal mouse organoids. Computational modeling was used to predict microenvironment cytokine concentrations. Organoid size, survival, proliferation, and differentiation were characterized by morphometrics, quantitative reverse-transcription polymerase chain reaction, and immunostaining on whole organoids or isolated ISCs. ISC function was assayed using serial passaging to single cells followed by organoid quantification. Single-cell RNA sequencing was used to assess Il22ra1 expression patterns in ISCs and transit-amplifying (TA) progenitors. An IL22-transgenic mouse was used to confirm the impact of increased IL22 on proliferative cells in vivo. Results High IL22 levels caused decreased ileal organoid survival, however, resistant organoids grew larger and showed increased proliferation over controls. Il22ra1 was expressed on only a subset of ISCs and TA progenitors. IL22-treated ISCs did not show appreciable differentiation defects, but ISC biomarker expression and self-renewal-associated pathway activity was reduced and accompanied by an inhibition of ISC expansion. In vivo, chronically increased IL22 levels, similar to predicted microenvironment levels, showed increases in proliferative cells in the TA zone with no increase in ISCs. Conclusions Increased IL22 limits ISC expansion in favor of increased TA progenitor cell expansion.
Collapse
Key Words
- BSA, bovine serum albumin
- EGFP, enhanced green fluorescent protein
- FACS, fluorescence-activated cell sorter
- IBD, inflammatory bowel disease
- IL, interleukin
- IL22RA1, IL22 receptor A1
- IL22TG, IL22 transgenic
- ILC, innate lymphoid cell
- ILC3, IL22-secreting lymphocyte
- ISC, intestinal stem cell
- Inflammatory Bowel Disease
- Interleukin-22
- Intestinal Stem Cells
- OFE, organoid forming efficiency
- STAT3, signal transducer and activator of transcription 3
- TA, transit-amplifying
- TBS, Tris-buffered saline
- cDNA, complementary DNA
- mRNA, messenger RNA
Collapse
Affiliation(s)
- Bailey Zwarycz
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Adam D Gracz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kristina R Rivera
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill/North Carolina State University, Chapel Hill, North Carolina
| | - Ian A Williamson
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill/North Carolina State University, Chapel Hill, North Carolina
| | - Leigh A Samsa
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Josh Starmer
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michael A Daniele
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill/North Carolina State University, Chapel Hill, North Carolina; Department of Electrical and Computer Engineering, North Carolina State University, Raleigh, North Carolina
| | | | | | - Scott T Magness
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill/North Carolina State University, Chapel Hill, North Carolina; Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
147
|
Couturier-Maillard A, Froux N, Piotet-Morin J, Michaudel C, Brault L, Le Bérichel J, Sénéchal A, Robinet P, Chenuet P, Jejou S, Dumoutier L, Renauld JC, Iovanna J, Huber S, Chamaillard M, Quesniaux V, Sokol H, Chamaillard M, Ryffel B. Interleukin-22-deficiency and microbiota contribute to the exacerbation of Toxoplasma gondii-induced intestinal inflammation. Mucosal Immunol 2018; 11:1181-1190. [PMID: 29728643 DOI: 10.1038/s41385-018-0005-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 01/04/2018] [Accepted: 01/09/2018] [Indexed: 02/04/2023]
Abstract
Upon oral infection with Toxoplasma gondii cysts (76 K strain) tachyzoites are released into the intestinal lumen and cross the epithelial barrier causing damage and acute intestinal inflammation in C57BL/6 (B6) mice. Here we investigated the role of microbiota and IL-22 in T.gondii-induced small intestinal inflammation. Oral T.gondii infection in B6 mice causes inflammation with IFNγ and IL-22 production. In IL-22-deficient mice, T.gondii infection augments the Th1 driven inflammation. Deficiency in either IL-22bp, the soluble IL-22 receptor or Reg3γ, an IL-22-dependent antimicrobial lectin/peptide, did not reduce inflammation. Under germ-free conditions, T.gondii-induced inflammation was reduced in correlation with parasite load. But intestinal inflammation is still present in germ-free mice, at low level, in the lamina propria, independently of IL-22 expression. Exacerbated intestinal inflammation driven by absence of IL-22 appears to be independent of IL-22 deficiency associated-dysbiosis as similar inflammation was observed after fecal transplantation of IL-22-/- or WT microbiota to germ-free-WT mice. Our results suggest cooperation between parasite and intestinal microbiota in small intestine inflammation development and endogenous IL-22 seems to exert a protective role independently of its effect on the microbiota. In conclusion, IL-22 participates in T.gondii induced acute small intestinal inflammation independently of microbiota and Reg3γ.
Collapse
Affiliation(s)
- A Couturier-Maillard
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - N Froux
- CNRS UPS44 -TAAM, Orléans, France
| | - J Piotet-Morin
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - C Michaudel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - L Brault
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - J Le Bérichel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | | | - P Robinet
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - P Chenuet
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - S Jejou
- Sorbonne Universités, UPMC Univ. Paris 06, École Normale Supérieure, PSL Research University, CNRS, INSERM, APHP, Laboratoire des Biomolécules (LBM), 27 rue de Chaligny, 75005, Paris, France
| | - L Dumoutier
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, Brussels, Belgium
| | - J C Renauld
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, Brussels, Belgium
| | - J Iovanna
- INSERM U1068, Centre de Recherche en Cancérologie de Marseille (CRCM), Aix-Marseille Université and Institut Paoli-Calmette, Parc Scientifique et Technologique de Luminy, CNRS UMR 7258, Marseille, France
| | - S Huber
- Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, Hamburg, 20246, Germany
| | | | - Vfj Quesniaux
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France
| | - H Sokol
- Sorbonne Universités, UPMC Univ. Paris 06, École Normale Supérieure, PSL Research University, CNRS, INSERM, APHP, Laboratoire des Biomolécules (LBM), 27 rue de Chaligny, 75005, Paris, France
- Micalis Institute, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, 78352, France
- Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique-Hopitaux de Paris, UPMC, Paris, France
| | - M Chamaillard
- Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique-Hopitaux de Paris, UPMC, Paris, France
| | - B Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), CNRS and University of Orleans (UMR7355), Orléans, France.
| |
Collapse
|
148
|
Khosravi N, Caetano MS, Cumpian AM, Unver N, De la Garza Ramos C, Noble O, Daliri S, Hernandez BJ, Gutierrez BA, Evans SE, Hanash S, Alekseev AM, Yang Y, Chang SH, Nurieva R, Kadara H, Chen J, Ostrin EJ, Moghaddam SJ. IL22 Promotes Kras-Mutant Lung Cancer by Induction of a Protumor Immune Response and Protection of Stemness Properties. Cancer Immunol Res 2018; 6:788-797. [PMID: 29764837 PMCID: PMC6030457 DOI: 10.1158/2326-6066.cir-17-0655] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 03/22/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023]
Abstract
Somatic KRAS mutations are the most common oncogenic variants in lung cancer and are associated with poor prognosis. Using a Kras-induced lung cancer mouse model, CC-LR, we previously showed a role for inflammation in lung tumorigenesis through activation of the NF-κB pathway, along with induction of interleukin 6 (IL6) and an IL17-producing CD4+ T-helper cell response. IL22 is an effector molecule secreted by CD4+ and γδ T cells that we previously found to be expressed in CC-LR mice. IL22 mostly signals through the STAT3 pathway and is thought to act exclusively on nonhematopoietic cells with basal IL22 receptor (IL22R) expression on epithelial cells. Here, we found that higher expression of IL22R1 in patients with KRAS-mutant lung adenocarcinoma was an independent indicator of poor recurrence-free survival. We then showed that genetic ablation of Il22 in CC-LR mice (CC-LR/IL22KO mice) caused a significant reduction in tumor number and size. This was accompanied by significantly lower tumor cell proliferation, angiogenesis, and STAT3 activation. Il22 ablation was also associated with significant reduction in lung-infiltrating inflammatory cells and expression of protumor inflammatory cytokines. Conversely, this was accompanied with increased antitumor Th1 and cytotoxic CD8+ T-cell responses, while suppressing the protumor immunosuppressive T regulatory cell response. In CC-LR/IL22KO mice, we found significantly reduced expression of core stemness genes and the number of prototypical SPC+CCSP+ stem cells. Thus, we conclude that IL22 promotes Kras-mutant lung tumorigenesis by driving a protumor inflammatory microenvironment with proliferative, angiogenic, and stemness contextual cues in epithelial/tumor cells. Cancer Immunol Res; 6(7); 788-97. ©2018 AACR.
Collapse
Affiliation(s)
- Nasim Khosravi
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mauricio S Caetano
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amber M Cumpian
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nese Unver
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Oscar Noble
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, México
| | - Soudabeh Daliri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Belinda J Hernandez
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Berenice A Gutierrez
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott E Evans
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Andrei M Alekseev
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yi Yang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Radiation Oncology, The Second Hospital of Jilin University, China
| | - Seon Hee Chang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roza Nurieva
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jichao Chen
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Edwin J Ostrin
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
149
|
Li C, Zhao X, Yang Y, Liu S, Liu Y, Li X. Interleukin-22 (IL-22) Regulates Apoptosis of Paclitaxel-Resistant Non-Small Cell Lung Cancer Cells Through C-Jun N-Terminal Kinase Signaling Pathway. Med Sci Monit 2018; 24:2750-2757. [PMID: 29723165 PMCID: PMC5952719 DOI: 10.12659/msm.907336] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 10/17/2017] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Reducing drug resistance in tumor cells has become an important issue for cancer treatment. The purpose of this study was to investigate whether IL-22 was involved in lung cancer cell resistance to paclitaxel (PTX), and to explore the underlying molecular mechanism. MATERIAL AND METHODS Non-small cell lung cancer (NSCLC) cell line A549 and the drug resistant cell line A549/PTX were used in the present study. The inhibitory rate of PTX on A549 and A549/PTX cell proliferation was determined by MTT assay and the half-maximal inhibitory concentration (IC50) value was calculated. The expression level of IL-22 was detected using Western blot and qRT-PCR. To elucidate the mechanism by which IL-22 is involved in PTX resistance, a stable IL-22-silenced A549/PTX cell line was generated by using IL-22-siRNA. Cell apoptosis was analyzed by flow cytometry, and the c-Jun N-terminal kinase (JNK) signal pathway was determined using Western blot analysis. RESULTS We found that IL-22 expression level was markedly higher in A549/PTX cells than in A549 cells, and IL-22 gene knockdown significantly enhanced the cell proliferation inhibition rate of PTX to A549/PTX cells and decreased the IC50 value of PTX to A549/PTX cells, indicating IL-22 was involved in cell PTX resistance. Our findings also suggest that IL-22 knockdown notably increased PTX induced apoptosis in A549/PTX cells. Moreover, the results showed that p-JNK and Caspase 3 expression were significantly increased in IL-22 knockdown A549/PTX cells, while Bcl-2 expression was significantly decreased. CONCLUSIONS IL-22 is involved in A549 cell resistance to PTX through regulating cell apoptosis via the JNK signaling pathway.
Collapse
Affiliation(s)
- Chenchen Li
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, P.R. China
| | - Xia Zhao
- Department of Medical Oncology, First People’s Hospital of Yancheng, Fourth Affiliated Hospital of Nantong University, Yancheng, Jiangsu, P.R. China
| | - Yang Yang
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, P.R. China
| | - Siwen Liu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, P.R. China
| | - Yun Liu
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, P.R. China
| | - Xiaoyou Li
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, P.R. China
| |
Collapse
|
150
|
Granulocyte-macrophage colony-stimulating factor negatively regulates early IL-10-mediated responses. Future Sci OA 2018; 4:FSO288. [PMID: 29682323 PMCID: PMC5905582 DOI: 10.4155/fsoa-2017-0133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/09/2018] [Indexed: 01/10/2023] Open
Abstract
Aim: Treatment of inflammatory disorders relies on the intervention in immune responses thereby restoring homeostasis. IL-10 is a cytokine with therapeutic potential, but until now has not been as successful as previously anticipated. A reason for this may be that IL-10 responsiveness depends on the environment of the inflamed tissue. In this study we investigated whether GM-CSF is able to influence IL-10-mediated responses. Methodology: Dendritic cells and macrophages were differentiated from mouse bone marrow and treated or depleted from GM-CSF prior to analyze their response to IL-10. Activity was assessed by measuring cytokine expression upon lipopolysaccharide stimulation, IL-10-induced signaling and down-stream gene expression. Conclusion: This study describes that GM-CSF negatively regulates IL-10-mediated responses. Over the last couple of decades inflammatory disorders, like autoimmune disease and allergies, are becoming more prevalent in the western world. These inflammatory disorders are characterized by uncontrolled immune responses against harmless antigens or commensal bacteria. Treatment of these diseases relies on the intervention in inflammatory responses and thereby restoring the balance of the immune system. One approach used in the clinic to balance the immune system is by treating patients with molecules used by our own immune system to suppress inflammation, such as IL-10. However, treatment with IL-10 has not been as successful as previously anticipated. In this study we show that a particular signaling molecule of the immune system that contributes to inflammation negatively affects the response of immune cells toward IL-10 and thereby could contribute to the low efficacy of IL-10 treatment.
Collapse
|