1
|
Raavi, Koehler AN, Vegas AJ. At The Interface: Small-Molecule Inhibitors of Soluble Cytokines. Chem Rev 2025. [PMID: 40233276 DOI: 10.1021/acs.chemrev.4c00469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cytokines are crucial regulators of the immune system that orchestrate interactions between cells and, when dysregulated, contribute to the progression of chronic inflammation, cancer, and autoimmunity. Numerous biologic-based clinical agents, mostly monoclonal antibodies, have validated cytokines as important clinical targets and are now part of the standard of care for a number of diseases. These agents, while impactful, still suffer from limitations including a lack of oral bioavailability, high cost of production, and immunogenicity. Small-molecule cytokine inhibitors are attractive alternatives that can address these limitations. Although targeting cytokine-cytokine receptor complexes with small molecules has been a challenging research endeavor, multiple small-molecule inhibitors have now been identified, with a number of them undergoing clinical evaluation. In this review, we highlight the recent advancements in the discovery and development of small-molecule inhibitors targeting soluble cytokines. The strategies for identifying these novel ligands as well as the structural and mechanistic insights into their activity represent important milestones in tackling these challenging and clinically important protein-protein interactions.
Collapse
Affiliation(s)
- Raavi
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Angela N Koehler
- Koch Institute for Integrative Cancer Research, and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Arturo J Vegas
- Department of Chemistry, Boston University, Boston, Massachusetts 02115, United States
| |
Collapse
|
2
|
He F, Yu J, Ma S, Zhao W, Zhang M, Wang J, Zhang C, Wu J, Zhu L. γδT Cells Induce the Inflammatory Response of Human Fibroblast-Like Synoviocytes Directly or by Stimulating B Cells to Activate IL-17/STAT3 Signaling Pathway. Int Arch Allergy Immunol 2024; 185:1154-1165. [PMID: 38991517 DOI: 10.1159/000539703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) combined with hashimoto thyroiditis (HT) is an important cause of various fatal comorbidities of RA. There is no precise conclusion about the cause of this disease. METHODS Peripheral blood and synovial tissue were collected from healthy participants, patients with RA, and patients with both RA and HT. Immunofluorescence staining and Pearson correlation analysis were used to detect the levels of γδTCR and the correlation between IL-17 and p-STAT3, respectively. ELISA, chemiluminescence assays, qRT-PCR and Western blot were performed to detect the levels of IgG, IgM, IFN-γ, IL-1β, TNF-α, Tg-Ab, Tpo-Ab, IL-17, IL-2, p-SATA3, and STAT3, respectively. RESULTS There was increased proportion of γδT cells, IL-17, and p-STAT3 levels in RA and HT patients. IL-17 was positively correlated with p-STAT3. γδT cells significantly promoted the expression of IgG, Tg-Ab, Tpo-Ab, and IL-17. When γδT and human fibroblast-like synoviocytes (FLSs) were co-cultured, the levels of IL-2, IFN-γ, IL-1β, TNF-α, and IL-17 were increased, and the IL-17/STAT3 signaling pathway was activated. When IL-17-silenced γδT cells and STAT3-silenced FLSs were co-cultured, the levels of IL-1β and TNF-α in FLSs were significantly decreased. Furthermore, when STAT3-silenced FLSs were added to the co-culture medium of B cells and γδT cells, the levels of IL-1β and TNF-α were also decreased significantly. CONCLUSION γδT cells induced RA directly or by stimulating B cells to activate STAT3 through IL-17.
Collapse
Affiliation(s)
- Fang He
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Juan Yu
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Sha Ma
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Weiqing Zhao
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Mingxing Zhang
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Juan Wang
- Department of Rheumatology and Immunology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Chunpan Zhang
- Department of Pain, The First People's Hospital of Yunnan Province, Kunming, China
| | - Jiangping Wu
- Department of Pain, The First People's Hospital of Yunnan Province, Kunming, China
| | - Lixuan Zhu
- Department of Pain, The First People's Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
3
|
Thorenoor N, Floros J. The Lung Alveolar Cell (LAC) miRNome and Gene Expression Profile of the SP-A-KO Mice After Infection With and Without Rescue With Human Surfactant Protein-A2 (1A0). Front Immunol 2022; 13:854434. [PMID: 35844510 PMCID: PMC9283764 DOI: 10.3389/fimmu.2022.854434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Human surfactant protein (SP)-A1 and SP-A2 exhibit differential qualitative and quantitative effects on the alveolar macrophage (AM), including a differential impact on the AM miRNome. Moreover, SP-A rescue (treatment) of SP-A-knockout (KO) infected mice impoves survival. Here, we studied for the first time the role of exogenous SP-A protein treatment on the regulation of lung alveolar cell (LAC) miRNome, the miRNA-RNA targets, and gene expression of SP-A-KO infected mice of both sexes. Toward this, SP-A-KO mice of both sexes were infected with Klebsiella pneumoniae, and half of them were also treated with SP-A2 (1A0). After 6 h of infection/SP-A treatment, the expression levels and pathways of LAC miRNAs, genes, and target miRNA-mRNAs were studied in both groups. We found 1) significant differences in the LAC miRNome, genes, and miRNA-mRNA targets in terms of sex, infection, and infection plus SP-A2 (1A0) protein rescue; 2) an increase in the majority of miRNA-mRNA targets in both study groups in KO male vs. female mice and involvement of the miRNA-mRNA targets in pathways of inflammation, antiapoptosis, and cell cycle; 3) genes with significant changes to be involved in TP-53, tumor necrosis factor (TNF), and cell cycle signaling nodes; 4) when significant changes in the expression of molecules from all analyses (miRNAs, miRNA-mRNA targets, and genes) were considered, two signaling pathways, the TNF and cell cycle, referred to as “integrated pathways” were shown to be significant; 5) the cell cycle pathway to be present in all comparisons made. Because SP-A could be used therapeutically in pulmonary diseases, it is important to understand the molecules and pathways involved in response to an SP-A acute treatment. The information obtained contributes to this end and may help to gain insight especially in the case of infection.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
- Department of Biochemistry and Molecular Biology, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
| | - Joanna Floros
- Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
- Department of Obstetrics and Gynecology, College of Medicine, The Pennsylvania State University, Hershey, PA, United States
| |
Collapse
|
4
|
Akhter MS, Uddin MA, Kubra KT, Barabutis N. Elucidation of the Molecular Pathways Involved in the Protective Effects of AUY-922 in LPS-Induced Inflammation in Mouse Lungs. Pharmaceuticals (Basel) 2021; 14:ph14060522. [PMID: 34072430 PMCID: PMC8226636 DOI: 10.3390/ph14060522] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/20/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) cause thousands of deaths every year and are associated with high mortality rates (~40%) due to the lack of efficient therapies. Understanding the molecular mechanisms associated with those diseases will most probably lead to novel therapeutics. In the present study, we investigated the effects of the Hsp90 inhibitor AUY-922 in the major inflammatory pathways of mouse lungs. Mice were treated with LPS (1.6 mg/kg) via intratracheal instillation for 24 h and were then post-treated intraperitoneally with AUY-922 (10 mg/kg). The animals were examined 48 h after AUY-922 injection. LPS activated the TLR4-mediated signaling pathways, which in turn induced the release of different inflammatory cytokines and chemokines. AUY-922 suppressed the LPS-induced inflammation by inhibiting major pro-inflammatory pathways (e.g., JAK2/STAT3, MAPKs), and downregulated the IL-1β, IL-6, MCP-1 and TNFα. The expression levels of the redox regulator APE1/Ref1, as well as the DNA-damage inducible kinases ATM and ATR, were also increased after LPS treatment. Those effects were counteracted by AUY-922. Interestingly, this Hsp90 inhibitor abolished the LPS-induced pIRE1α suppression, a major component of the unfolded protein response. Our study elucidates the molecular pathways involved in the progression of murine inflammation and supports our efforts on the development of new therapeutics against lung inflammatory diseases and sepsis.
Collapse
|
5
|
Shi M, Sekulovski N, Whorton AE, MacLean JA, Greaves E, Hayashi K. Efficacy of niclosamide on the intra-abdominal inflammatory environment in endometriosis. FASEB J 2021; 35:e21584. [PMID: 33860549 PMCID: PMC10777336 DOI: 10.1096/fj.202002541rrr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 12/24/2022]
Abstract
Endometriosis, a common gynecological disease, causes chronic pelvic pain and infertility in women of reproductive age. Due to the limited efficacy of current therapies, a critical need exists to develop new treatments for endometriosis. Inflammatory dysfunction, instigated by abnormal macrophage (MΦ) function, contributes to disease development and progression. However, the fundamental role of the heterogeneous population of peritoneal MΦ and their potential druggable functions is uncertain. Here we report that GATA6-expressing large peritoneal MΦ (LPM) were increased in the peritoneal cavity following lesion induction. This was associated with increased cytokine and chemokine secretion in the peritoneal fluid (PF), as well as MΦ infiltration, vascularization and innervation in endometriosis-like lesions (ELL). Niclosamide, an FDA-approved anti-helminthic drug, was effective in reducing LPM number, but not small peritoneal MΦ (SPM), in the PF. Niclosamide also inhibits aberrant inflammation in the PF, ELL, pelvic organs (uterus and vagina) and dorsal root ganglion (DRG), as well as MΦ infiltration, vascularization and innervation in the ELL. PF from ELL mice stimulated DRG outgrowth in vitro, whereas the PF from niclosamide-treated ELL mice lacked the strong stimulatory nerve growth response. These results suggest LPM induce aberrant inflammation in endometriosis promoting lesion progression and establishment of the inflammatory environment that sensitizes peripheral nociceptors in the lesions and other pelvic organs, leading to increased hyperalgesia. Our findings provide the rationale for targeting LPM and their functions with niclosamide and its efficacy in endometriosis as a new non-hormonal therapy to reduce aberrant inflammation which may ultimately diminish associated pain.
Collapse
Affiliation(s)
- Mingxin Shi
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
| | - Nikola Sekulovski
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - Allison E. Whorton
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA
| | - James A. MacLean
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
| | - Erin Greaves
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Kanako Hayashi
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA
- Center for Reproductive Biology, Washington State University, Pullman, WA, USA
| |
Collapse
|
6
|
Song MK, Kim DI, Lee K. Causal relationship between humidifier disinfectant exposure and Th17-mediated airway inflammation and hyperresponsiveness. Toxicology 2021; 454:152739. [PMID: 33640443 DOI: 10.1016/j.tox.2021.152739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 12/15/2022]
Abstract
In this study, we investigated whether humidifier disinfectants (HDs) induce asthmatic airway inflammation in an animal model and compared the features of HD-induced inflammatory symptoms with ovalbumin (OVA)-induced allergic asthma. Mice were intratracheally instilled three times with either the control or 0.1, 0.3, or 0.5 mg/kg of polyhexamethylene guanidine phosphate (PHMG-P). To characterize asthmatic features, the following parameters were analyzed: (i) differential cell counts and cytokine expression in the bronchoalveolar lavage fluid (BALF); (ii) presence of mucus-producing goblet cells and pulmonary eosinophilic infiltration in the lungs; (iii) serum immunoglobulin levels; and (iv) airway hyperresponsiveness (AHR). RNA-Seq and bioinformatics tools were used to investigate whether PHMG-P altered asthma-related gene expression in lung tissues. The PHMG-P exposure groups showed higher peribronchial/perivascular inflammation, elevated goblet cell hyperplasia, and inhaled methacholine-induced airway resistance. Additionally, IL-13 and IL-17 in BALF were significantly increased in the PHMG-P exposure groups. However, there were no significant differences in total serum IgE and BALF IL-4 and IL-5 levels in the PHMG-P exposure groups compared to the control group. PHMG-P exposure modulated the expression of genes related to Th17 signaling pathways including the IL-17A, IL-23, and STAT3 signaling pathways, but not the Th2 signaling pathway. Altogether, our results suggest that repeated exposure to low does PHMG-P induces asthma-like symptoms and is thus a possible risk factor for developing asthma. The PHMG-P-induced asthmatic airway inflammation showed a different pattern from that found in typical allergic asthma and may be related to irritant-induced airway inflammation and hyperresponsiveness characterized by Th2-low, Th17-related, IgE-independent, and mixed granulocytic features.
Collapse
Affiliation(s)
- Mi-Kyung Song
- National Center for Efficacy Evaluation for Respiratory Disease Products, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea; Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Dong Im Kim
- National Center for Efficacy Evaluation for Respiratory Disease Products, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea
| | - Kyuhong Lee
- National Center for Efficacy Evaluation for Respiratory Disease Products, Korea Institute of Toxicology, 30 Baehak1-gil, Jeongeup, Jeollabuk-do, 56212, Republic of Korea; Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
7
|
Thorenoor N, Phelps DS, Floros J. Differential Sex-Dependent Regulation of the Alveolar Macrophage miRNome of SP-A2 and co-ex (SP-A1/SP-A2) and Sex Differences Attenuation after 18 h of Ozone Exposure. Antioxidants (Basel) 2020; 9:antiox9121190. [PMID: 33260937 PMCID: PMC7768498 DOI: 10.3390/antiox9121190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/19/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Human SP-A1 and SP-A2, encoded by SFTPA1 and SFTPA2, and their genetic variants differentially impact alveolar macrophage (AM) functions and regulation, including the miRNome. We investigated whether miRNome differences previously observed between AM from SP-A2 and SP-A1/SP-A2 mice are due to continued qualitative differences or a delayed response of mice carrying a single gene. Methods: Human transgenic (hTG) mice, carrying SP-A2 or both SP-A genes, and SP-A-KO mice were exposed to filtered air (FA) or ozone (O3). AM miRNA levels, target gene expression, and pathways determined 18 h after O3 exposure. RESULTS: We found (a) differences in miRNome due to sex, SP-A genotype, and exposure; (b) miRNome of both sexes was largely downregulated by O3, and co-ex had fewer changed (≥2-fold) miRNAs than either group; (c) the number and direction of the expression of genes with significant changes in males and females in co-ex are almost the opposite of those in SP-A2; (d) the same pathways were found in the studied groups; and (e) O3 exposure attenuated sex differences with a higher number of genotype-dependent and genotype-independent miRNAs common in both sexes after O3 exposure. Conclusion: Qualitative differences between SP-A2 and co-ex persist 18 h post-O3, and O3 attenuates sex differences.
Collapse
Affiliation(s)
- Nithyananda Thorenoor
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (N.T.); (D.S.P.)
- Department of Biochemistry & Molecular Biology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - David S. Phelps
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (N.T.); (D.S.P.)
| | - Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (N.T.); (D.S.P.)
- Department of Obstetrics & Gynecology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
- Correspondence:
| |
Collapse
|
8
|
Faustino LD, Griffith JW, Rahimi RA, Nepal K, Hamilos DL, Cho JL, Medoff BD, Moon JJ, Vignali DAA, Luster AD. Interleukin-33 activates regulatory T cells to suppress innate γδ T cell responses in the lung. Nat Immunol 2020; 21:1371-1383. [PMID: 32989331 PMCID: PMC7578082 DOI: 10.1038/s41590-020-0785-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/13/2020] [Indexed: 12/20/2022]
Abstract
Foxp3+ regulatory T (Treg) cells expressing the interleukin (IL)-33 receptor ST2 mediate tissue repair in response to IL-33. Whether Treg cells also respond to the alarmin IL-33 to regulate specific aspects of the immune response is not known. Here we describe an unexpected function of ST2+ Treg cells in suppressing the innate immune response in the lung to environmental allergens without altering the adaptive immune response. Following allergen exposure, ST2+ Treg cells were activated by IL-33 to suppress IL-17-producing γδ T cells. ST2 signaling in Treg cells induced Ebi3, a component of the heterodimeric cytokine IL-35 that was required for Treg cell-mediated suppression of γδ T cells. This response resulted in fewer eosinophil-attracting chemokines and reduced eosinophil recruitment into the lung, which was beneficial to the host in reducing allergen-induced inflammation. Thus, we define a fundamental role for ST2+ Treg cells in the lung as a negative regulator of the early innate γδ T cell response to mucosal injury.
Collapse
Affiliation(s)
- Lucas D Faustino
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jason W Griffith
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rod A Rahimi
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keshav Nepal
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel L Hamilos
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Josalyn L Cho
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin D Medoff
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - James J Moon
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine and Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Wang WW, Yu HW, Zhang B, Pan YL, Shao SW. Interleukin-17A up-regulates thymic stromal lymphopoietin production by nasal fibroblasts from patients with allergic rhinitis. Eur Arch Otorhinolaryngol 2020; 278:127-133. [PMID: 32783069 DOI: 10.1007/s00405-020-06274-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 08/04/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE Emerging evidence has shown that interleukin (IL)-17A is implicated in the pathogenesis of allergic rhinitis (AR). Thymic stromal lymphopoietin (TSLP) orchestrates the immune response toward a Th2 phenotype. Although increased TSLP is found in AR, the contribution of IL-17A in TSLP production by nasal fibroblasts is not well understood. We aimed to investigate the effect and mechanism of IL-17A on TSLP production by human nasal fibroblasts (HNFs) from AR patients. METHODS HNFs from AR patients were cultured and stimulated with IL-17A in the absence or presence of a Janus kinase (JAK) 2 or JAK1/3 inhibitor. Western blotting was used to assay phosphorylated signal transducer and activator of transcription 3 (p-STAT3) and nuclear factor-kappa B (NF-κB) p65 in HNFs. The TSLP expression in the cells and culture supernatants was evaluated by real-time polymerase chain reaction and enzyme-linked immunoassay. RESULTS Stimulation with IL-17A induced STAT3 phosphorylation, which was inhibited by the pretreatment with JAK2 inhibitor AZD1480 or JAK1/3 inhibitor tofacitinib. IL-17A promoted the nuclear translocation of NF-κBp65 protein, leading to increased TSLP production, while the pre-incubation with AZD1480 prior to IL-17A attenuated these effects. However, the pre-incubation with tofacitinib before IL-17A stimulation had no impact on the expression of NF-κBp65 and TSLP. CONCLUSIONS IL-17A up-regulated TSLP production by HNFs through JAK2/NF-κB pathway. Although IL-17A induced STAT3 activation through JAK1/2/3, IL-17A-mediated TSLP expression was not dependent on STAT3 signaling. These observations would provide mechanistic insight into therapeutic strategies to improve the immune and inflammation associated with Th17A in the management of AR.
Collapse
Affiliation(s)
- Wei Wei Wang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou Central Hospital, School of Medicine, Huzhou University, No. 759, East Second Ring Road, Huzhou, 313000, Zhejiang, China
| | - Hong Wei Yu
- School of Medicine, Huzhou University, No. 759, East Second Ring Road, Huzhou, 313000, Zhejiang, China
| | - Bo Zhang
- School of Medicine, Huzhou University, No. 759, East Second Ring Road, Huzhou, 313000, Zhejiang, China
| | - Yong Liang Pan
- School of Medicine, Huzhou University, No. 759, East Second Ring Road, Huzhou, 313000, Zhejiang, China
| | - Sheng Wen Shao
- School of Medicine, Huzhou University, No. 759, East Second Ring Road, Huzhou, 313000, Zhejiang, China.
| |
Collapse
|
10
|
Muscarinic Toxin 7 Signals Via Ca 2+/Calmodulin-Dependent Protein Kinase Kinase β to Augment Mitochondrial Function and Prevent Neurodegeneration. Mol Neurobiol 2020; 57:2521-2538. [PMID: 32198698 PMCID: PMC7253379 DOI: 10.1007/s12035-020-01900-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/09/2020] [Indexed: 12/29/2022]
Abstract
Mitochondrial dysfunction is implicated in a variety of neurodegenerative diseases of the nervous system. Peroxisome proliferator–activated receptor-γ coactivator-1α (PGC-1α) is a regulator of mitochondrial function in multiple cell types. In sensory neurons, AMP-activated protein kinase (AMPK) augments PGC-1α activity and this pathway is depressed in diabetes leading to mitochondrial dysfunction and neurodegeneration. Antimuscarinic drugs targeting the muscarinic acetylcholine type 1 receptor (M1R) prevent/reverse neurodegeneration by inducing nerve regeneration in rodent models of diabetes and chemotherapy-induced peripheral neuropathy (CIPN). Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ) is an upstream regulator of AMPK activity. We hypothesized that antimuscarinic drugs modulate CaMKKβ to enhance activity of AMPK, and PGC-1α, increase mitochondrial function and thus protect from neurodegeneration. We used the specific M1R antagonist muscarinic toxin 7 (MT7) to manipulate muscarinic signaling in the dorsal root ganglia (DRG) neurons of normal rats or rats with streptozotocin-induced diabetes. DRG neurons treated with MT7 (100 nM) or a selective muscarinic antagonist, pirenzepine (1 μM), for 24 h showed increased neurite outgrowth that was blocked by the CaMKK inhibitor STO-609 (1 μM) or short hairpin RNA to CaMKKβ. MT7 enhanced AMPK phosphorylation which was blocked by STO-609 (1 μM). PGC-1α reporter activity was augmented up to 2-fold (p < 0.05) by MT7 and blocked by STO-609. Mitochondrial maximal respiration and spare respiratory capacity were elevated after 3 h of exposure to MT7 (p < 0.05). Diabetes and CIPN induced a significant (p < 0.05) decrease in corneal nerve density which was corrected by topical delivery of MT7. We reveal a novel M1R-modulated, CaMKKβ-dependent pathway in neurons that represents a therapeutic target to enhance nerve repair in two of the most common forms of peripheral neuropathy.
Collapse
|
11
|
Abstract
There are multiple proinflammatory pathways in the pathogenesis of asthma. These include both innate and adaptive inflammation, in addition to inflammatory and physiologic responses mediated by eicosanoids. An important component of the innate allergic immune response is ILC2 activated by interleukin (IL)-33, thymic stromal lymphopoietin, and IL-25 to produce IL-5 and IL-13. In terms of the adaptive T-lymphocyte immunity, CD4+ Th2 and IL-17-producing cells are critical in the inflammatory responses in asthma. Last, eicosanoids involved in asthma pathogenesis include prostaglandin D2 and the cysteinyl leukotrienes that promote smooth muscle constriction and inflammation that propagate allergic responses.
Collapse
Affiliation(s)
- R Stokes Peebles
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, VUMC, T-1218 MCN, 1161 21st Avenue South, Nashville, TN 37232-2650, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, VUMC, T-1218 MCN, 1161 21st Avenue South, Nashville, TN 37232-2650, USA.
| | - Mark A Aronica
- Department of Pathobiology, Respiratory Institute, Cleveland Clinic Lerner College of Medicine, CWRU, 9500 Euclid Avenue, NB2-85, Cleveland, OH 44195, USA
| |
Collapse
|
12
|
IL-17A secreted from lymphatic endothelial cells promotes tumorigenesis by upregulation of PD-L1 in hepatoma stem cells. J Hepatol 2019; 71:1206-1215. [PMID: 31499129 DOI: 10.1016/j.jhep.2019.08.034] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS The microenvironment regulates hepatoma stem cell behavior. However, the contributions of lymphatic endothelial cells to the hepatoma stem cell niche remain largely unknown; we aimed to analyze this contribution and elucidate the mechanisms behind it. METHODS Associations between lymphatic endothelial cells and CD133+ hepatoma stem cells were analyzed by immunofluorescence and adhesion assays; with the effects of their association on IL-17A expression examined using western blot, quantitative reverse transcription PCR and luciferase reporter assay. The effects of IL-17A on the self-renewal and tumorigenesis of hepatoma stem cells were examined using sphere and tumor formation assays. The role of IL-17A in immune escape by hepatoma stem cells was examined using flow cytometry. The expression of IL-17A in hepatoma tissues was examined using immunohistochemistry. RESULTS CD133+ hepatoma stem cells preferentially interact with lymphatic endothelial cells. The interaction between the mannose receptor and high-mannose type N-glycans mediates the interaction between CD133+ hepatoma stem cells and lymphatic endothelial cells. This interaction activates cytokine IL-17A expression in lymphatic endothelial cells. IL-17A promotes the self-renewal of hepatoma stem cells. It also promotes their immune escape, partly through upregulation of PD-L1. CONCLUSION Interactions between lymphatic endothelial cells and hepatoma stem cells promote the self-renewal and immune escape of hepatoma stem cells, by activating IL-17A signaling. Thus, inhibiting IL-17A signaling may be a promising approach for hepatoma treatment. LAY SUMMARY The microenvironment is crucial for the self-renewal and development of hepatoma stem cells, which lead to the development of liver cancer. Lymphatic endothelial cells are an important component of this niche microenvironment, helping hepatoma stem cells to self-renew and escape immune attack, by upregulating IL-17A signaling. Thus, targeting IL-17A signaling is a potential strategy for the treatment of hepatoma.
Collapse
|
13
|
Bourgonje AR, von Martels JZH, Gabriëls RY, Blokzijl T, Buist-Homan M, Heegsma J, Jansen BH, van Dullemen HM, Festen EAM, Ter Steege RWF, Visschedijk MC, Weersma RK, de Vos P, Faber KN, Dijkstra G. A Combined Set of Four Serum Inflammatory Biomarkers Reliably Predicts Endoscopic Disease Activity in Inflammatory Bowel Disease. Front Med (Lausanne) 2019; 6:251. [PMID: 31772929 PMCID: PMC6849495 DOI: 10.3389/fmed.2019.00251] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/18/2019] [Indexed: 12/20/2022] Open
Abstract
Introduction: Blood C-reactive protein (CRP) and fecal calprotectin levels are routinely measured as surrogate markers of disease activity in Inflammatory Bowel Disease (IBD), but often do not correlate well with the degree of mucosal inflammation in the intestine as established by endoscopy. Therefore, novel predictive biomarkers are urgently needed that better reflect mucosal disease activity in IBD. The aim of this study was to identify a combination of serum inflammatory biomarkers predictive for endoscopic disease activity. Methods: Serum concentrations of 10 inflammatory biomarkers were analyzed in 118 IBD patients [64 Crohn's disease (CD), 54 ulcerative colitis (UC)] and 20 healthy controls. In a subset of 71 IBD patients, endoscopic disease activity was established. Non-parametric ROC estimation with bootstrap inference was used to establish the best combination of inflammatory biomarkers predicting endoscopic disease activity. Results: Six (6) inflammatory biomarkers (serum amyloid A (SAA), Eotaxin-1, IL-6, IL-8, IL-17A, and TNF-α) showed better prediction of IBD disease activity than routine measures (CRP, fecal calprotectin and HBI/SCCAI scores). The best combination of predictive inflammatory biomarkers consisted of serum SAA, IL-6, IL-8, and Eotaxin-1, showing an optimism-adjusted area under the ROC (AuROC) curve of 0.84 (95% CI: 0.73-0.94, P < 0.0001), which predicted significantly better (P = 0.002) than serum CRP levels with an AuROC of 0.57 (95% CI: 0.43-0.72, P = 0.32). Conclusion: The combination of SAA, IL-6, IL-8, and Eotaxin-1 reliably predicts endoscopic disease activity in IBD and might be valuable for monitoring disease activity and management of the disease.
Collapse
Affiliation(s)
- Arno R Bourgonje
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Julius Z H von Martels
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ruben Y Gabriëls
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Tjasso Blokzijl
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Manon Buist-Homan
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Janette Heegsma
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bernadien H Jansen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hendrik M van Dullemen
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Eleonora A M Festen
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rinze W F Ter Steege
- Department of Gastroenterology and Hepatology, Martini Hospital, Groningen, Netherlands
| | - Marijn C Visschedijk
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Paul de Vos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
14
|
Yap HM, Israf DA, Harith HH, Tham CL, Sulaiman MR. Crosstalk Between Signaling Pathways Involved in the Regulation of Airway Smooth Muscle Cell Hyperplasia. Front Pharmacol 2019; 10:1148. [PMID: 31649532 PMCID: PMC6794426 DOI: 10.3389/fphar.2019.01148] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/06/2019] [Indexed: 12/14/2022] Open
Abstract
Increased ASM mass, primarily due to ASM hyperplasia, has been recognized as a hallmark of airway remodeling in asthma. Increased ASM mass is the major contributor to the airway narrowing, thus worsening the bronchoconstriction in response to stimuli. Inflammatory mediators and growth factors released during inflammation induce increased ASM mass surrounding airway wall via increased ASM proliferation, diminished ASM apoptosis and increased ASM migration. Several major pathways, such as MAPKs, PI3K/AKT, JAK2/STAT3 and Rho kinase, have been reported to regulate these cellular activities in ASM and were reported to be interrelated at certain points. This article aims to provide an overview of the signaling pathways/molecules involved in ASM hyperplasia as well as the mapping of the interplay/crosstalk between these major pathways in mediating ASM hyperplasia. A more comprehensive understanding of the complexity of cellular signaling in ASM cells will enable more specific and safer drug development in the control of asthma.
Collapse
Affiliation(s)
- Hui Min Yap
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Daud Ahmad Israf
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Hanis Hazeera Harith
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Mohd Roslan Sulaiman
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
15
|
Interleukin-Mediated Pendrin Transcriptional Regulation in Airway and Esophageal Epithelia. Int J Mol Sci 2019; 20:ijms20030731. [PMID: 30744098 PMCID: PMC6386862 DOI: 10.3390/ijms20030731] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 12/28/2022] Open
Abstract
Pendrin (SLC26A4), a Cl−/anion exchanger, is expressed at high levels in kidney, thyroid, and inner ear epithelia, where it has an essential role in bicarbonate secretion/chloride reabsorption, iodide accumulation, and endolymph ion balance, respectively. Pendrin is expressed at lower levels in other tissues, such as airways and esophageal epithelia, where it is transcriptionally regulated by the inflammatory cytokines interleukin (IL)-4 and IL-13 through a signal transducer and activator of transcription 6 (STAT6)-mediated pathway. In the airway epithelium, increased pendrin expression during inflammatory diseases leads to imbalances in airway surface liquid thickness and mucin release, while, in the esophageal epithelium, dysregulated pendrin expression is supposed to impact the intracellular pH regulation system. In this review, we discuss some of the recent findings on interleukin-mediated transcriptional regulation of pendrin and how this dysregulation impacts airway and esophagus epithelial homeostasis during inflammatory diseases.
Collapse
|
16
|
Ardain A, Porterfield JZ, Kløverpris HN, Leslie A. Type 3 ILCs in Lung Disease. Front Immunol 2019; 10:92. [PMID: 30761149 PMCID: PMC6361816 DOI: 10.3389/fimmu.2019.00092] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
The lungs represent a complex immune setting, balancing external environmental signals with a poised immune response that must protect from infection, mediate tissue repair, and maintain lung function. Innate lymphoid cells (ILCs) play a central role in tissue repair and homeostasis, and mediate protective immunity in a variety of mucosal tissues, including the lung. All three ILC subsets are present in the airways of both mice and humans; and ILC2s shown to have pivotal roles in asthma, airway hyper-responsiveness, and parasitic worm infection. The involvement of ILC3s in respiratory diseases is less well-defined, but they are known to be critical in homeostasis, infection and inflammation at other mucosal barriers, such as the gut. Moreover, they are important players in the IL17/IL22 axis, which is key to lung health. In this review, we discuss the emerging role of ILC3s in the context of infectious and inflammatory lung diseases, with a focus on data from human subjects.
Collapse
Affiliation(s)
- Amanda Ardain
- Africa Health Research Institute, Durban, South Africa
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - James Zachary Porterfield
- Africa Health Research Institute, Durban, South Africa
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
- Yale School of Public Health, Yale University, New Haven, CT, United States
| | - Henrik N. Kløverpris
- Africa Health Research Institute, Durban, South Africa
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infection and Immunity, University College London, London, United Kingdom
| | - Alasdair Leslie
- Africa Health Research Institute, Durban, South Africa
- Department of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
17
|
Schmidt T, Luebbe J, Paust HJ, Panzer U. Mechanisms and functions of IL-17 signaling in renal autoimmune diseases. Mol Immunol 2018; 104:90-99. [PMID: 30448610 DOI: 10.1016/j.molimm.2018.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/31/2018] [Accepted: 09/07/2018] [Indexed: 12/27/2022]
Abstract
Immune-mediated glomerular diseases (glomerulonephritis) encompass a heterogeneous collection of diseases that cause inflammation within the glomerulus and other renal compartments with significant morbidity and mortality. In general, CD4+ T cells orchestrate the immune response and play a unique role in autoimmune and chronic inflammatory diseases. In particular, the characterization of a distinct, IL-17 cytokines producing CD4+ T cell subset named TH17 cells has significantly advanced the current understanding of the pathogenic mechanisms of organ-specific immunity. Our group and others have shown that the recruitment of TH17 cells to the inflamed kidney drives renal tissue injury in experimental and possibly human crescentic glomerulonephritis (GN), but much remains to be understood about the biological functions, regulation, and signaling pathways of the TH17/IL-17 axis leading to organ damage. Here we review our current knowledge about the mechanisms and functions of IL-17 signaling in renal autoimmune diseases, with a special focus on experimental and human crescentic GN.
Collapse
Affiliation(s)
- Tilman Schmidt
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jonas Luebbe
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Section of Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Joachim Paust
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Section of Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Section of Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
18
|
Alves JJP, De Medeiros Fernandes TAA, De Araújo JMG, Cobucci RNO, Lanza DCF, Bezerra FL, Andrade VS, Fernandes JV. Th17 response in patients with cervical cancer. Oncol Lett 2018; 16:6215-6227. [PMID: 30405758 PMCID: PMC6202464 DOI: 10.3892/ol.2018.9481] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 08/07/2018] [Indexed: 12/21/2022] Open
Abstract
Persistent infection by high-risk human papillomavirus (HR-HPV) is the main risk factor for uterine cervical cancer (UCC). However, viral infection alone is not sufficient for the development and progression of premalignant cervical lesions for cancer. In previous years it has been suggested that the adaptive immune response triggered by the differentiation of naïve helper T cells in Th17 cells may serve an important role in disease development. It has been hypothesized that Th17 cells may be involved in the promotion of UCC, as high levels of interleukin 17 (IL17) expression have been detected in the mucosa of the uterine cervix of patients affected by the disease. However, the role of Th17 cells in the tumor development and progression remains unclear. It is believed that the immune response of the Th17 type during persistent infection of the genital tract with HR-HPV triggers chronic inflammation with a long duration with the production of IL17 and other pro-inflammatory cytokines, creating a favorable environment for tumor development. These cytokines are produced by immune system cells in addition to tumor cells and appear to function by modulating the host immune system, resulting in an immunosuppressive response as opposed to inducing an effective protective immune response, thus contributing to the growth and progression of the tumor. In the present review, the latest advances are presented about the function of Th17 cells and the cytokines produced by them in the development and progression of UCC.
Collapse
Affiliation(s)
- Jayra Juliana Paiva Alves
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil
| | | | | | | | | | - Fabiana Lima Bezerra
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil
| | - Vânia Sousa Andrade
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil
| | - José Veríssimo Fernandes
- Department of Microbiology and Parasitology, Federal University of Rio Grande do Norte, Natal, RN 59072-970, Brazil
| |
Collapse
|
19
|
Shi S, Jin L, Zhang S, Li H, Zhang B, Sun M. MicroRNA-590-5p represses proliferation of human fetal airway smooth muscle cells by targeting signal transducer and activator of transcription 3. Arch Med Sci 2018; 14:1093-1101. [PMID: 30154893 PMCID: PMC6111361 DOI: 10.5114/aoms.2018.74538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/07/2018] [Indexed: 01/18/2023] Open
Abstract
INTRODUCTION Pediatric asthma has remained a health threat to children in recent years. The abnormal proliferation of airway smooth muscle (ASM) cells contributes to airway remodeling during development of asthma. MicroRNAs (miRNAs) are critical regulators of ASM cell proliferation during airway remodeling. miR-590-5p has been reported to regulate cell proliferation in various cell types. However, it remains unclear whether miR-590-5p regulates ASM cell proliferation. In this study, we aimed to investigate the potential role of miR-590-5p in regulating fetal ASM cell proliferation in vitro stimulated by platelet-derived growth factor (PDGF). MATERIAL AND METHODS miRNA, mRNA, and protein expression was detected by real-time quantitative polymerase chain reaction and western blot. Cell proliferation was detected by CCK-8 and BrdU assays. The target of miR-590-5p was confirmed by dual-luciferase reporter assay. RESULTS MiR-590-5p expression was significantly down-regulated in fetal ASM cells stimulated with PDGF (p < 0.05). Overexpression of miR-590-5p inhibited cell proliferation (p < 0.05), whereas the suppression of miR-590-5p promoted cell proliferation of fetal ASM cells stimulated with PDGF (p < 0.05). Signal transducer and activator of transcription 3 (STAT3) was identified as a target gene of miR-590-5p. In addition, miR-590-5p negatively regulated STAT3 expression (p < 0.05). Moreover, miR-590-5p also modulated downstream genes of STAT3 including cyclin D3 and p27 (p < 0.05). The restoration of STAT3 significantly reversed the inhibitory effect of miR-590-5p on fetal ASM cell proliferation. CONCLUSIONS MiR-590-5p inhibits proliferation of fetal ASM cells by down-regulating STAT3, thereby suggesting a novel therapeutic target for the treatment of pediatric asthma.
Collapse
Affiliation(s)
- Shan Shi
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lianhua Jin
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Sai Zhang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Haibo Li
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Bo Zhang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Meihua Sun
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
20
|
Chen X, Xie ZH, Lv YX, Tang QP, Zhang H, Zhang JY, Wu B, Jiang WH. A proteomics analysis reveals that A2M might be regulated by STAT3 in persistent allergic rhinitis. Clin Exp Allergy 2017; 46:813-24. [PMID: 27228572 DOI: 10.1111/cea.12711] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 12/11/2015] [Accepted: 01/05/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND Proteomics tools can be used to identify the differentially expressed proteins related to allergic rhinitis (AR). However, the large numbers of proteins related to AR have not yet been explored using an advanced quantitative proteomics approach, known as isobaric tags for relative and absolute quantitation (iTRAQ). OBJECTIVES To identify differentially expressed proteins in persistent AR patients and to explore the regulatory signalling pathways involving the identified proteins. METHODS Forty-five persistent AR patients and 20 healthy controls were recruited for this study. iTRAQ was used to identify the proteins that were differentially expressed between these two groups, and a bioinformatics analysis was then conducted to identify the signalling pathways associated with the identified proteins. Immunofluorescence labelling was performed to detect alpha-2-macroglobulin (A2M), STAT3, p-STAT3 and IL17 in the nasal mucosa. RESULTS A total of 133 differentially expressed proteins were identified. We then determined the top 10 regulatory pathways associated with these proteins and found that the blood coagulation pathway had the most significant association. A2M, a protein involved in the blood coagulation pathway, was found to be differentially expressed in the serum of AR patients. The bioinformatics analysis indicated that STAT3 is an upstream transcription factor that might regulate A2M expression. An immunofluorescence study further confirmed that STAT3 and A2M are co-localized in nasal mucosa cells. Additionally, A2M, STAT3, p-STAT3, and IL17 are elevated in AR patients. The expressional level of A2M is positively related to IL17 and the symptom of the congestion in AR subjects. CONCLUSIONS The blood coagulation pathway may be a key regulatory network pathway contributing to the allergic inflammatory response in AR patients. A2M, which is regulated by STAT3, may be an important protein in the pathogenesis of allergic rhinitis in AR patients.
Collapse
Affiliation(s)
- X Chen
- Department of Otolaryngology-Skull Base Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Z H Xie
- Department of Otolaryngology-Skull Base Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Y X Lv
- Department of Otolaryngology-Skull Base Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Q P Tang
- Department of Rehabilitation, Brain Hospital of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - H Zhang
- Department of Otolaryngology-Skull Base Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - J Y Zhang
- Department of Otolaryngology-Skull Base Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - B Wu
- Department of Otolaryngology-Skull Base Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - W H Jiang
- Department of Otolaryngology-Skull Base Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
21
|
Huang Z, Zhong L, Lee JTH, Zhang J, Wu D, Geng L, Wang Y, Wong CM, Xu A. The FGF21-CCL11 Axis Mediates Beiging of White Adipose Tissues by Coupling Sympathetic Nervous System to Type 2 Immunity. Cell Metab 2017; 26:493-508.e4. [PMID: 28844880 DOI: 10.1016/j.cmet.2017.08.003] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 05/24/2017] [Accepted: 08/01/2017] [Indexed: 02/08/2023]
Abstract
Type 2 cytokines are important signals triggering biogenesis of thermogenic beige adipocytes in white adipose tissue (WAT) during cold acclimation. However, how cold activates type 2 immunity in WAT remains obscure. Here we show that cold-induced type 2 immune responses and beiging in subcutaneous WAT (scWAT) are abrogated in mice with adipose-selective ablation of FGF21 or its co-receptor β-Klotho, whereas such impairments are reversed by replenishment with chemokine CCL11. Mechanistically, FGF21 acts on adipocytes in an autocrine manner to promote the expression and secretion of CCL11 via activation of ERK1/2, which drives recruitment of eosinophils into scWAT, leading to increases in accumulation of M2 macrophages, and proliferation and commitment of adipocyte precursors into beige adipocytes. These FGF21-elicited type 2 immune responses and beiging are blocked by CCL11 neutralization. Thus, the adipose-derived FGF21-CCL11 axis triggers cold-induced beiging and thermogenesis by coupling sympathetic nervous system to activation of type 2 immunity in scWAT.
Collapse
Affiliation(s)
- Zhe Huang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ling Zhong
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jimmy Tsz Hang Lee
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jialiang Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Donghai Wu
- The Key Laboratory of Regenerative Biology, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Leiluo Geng
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Chi-Ming Wong
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Laboratory Block, Pokfulam, Hong Kong, China; Department of Medicine, The University of Hong Kong, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
22
|
Che KF, Kaarteenaho R, Lappi-Blanco E, Levänen B, Sun J, Wheelock Å, Palmberg L, Sköld CM, Lindén A. Interleukin-26 Production in Human Primary Bronchial Epithelial Cells in Response to Viral Stimulation: Modulation by Th17 cytokines. Mol Med 2017; 23:247-257. [PMID: 28853490 DOI: 10.2119/molmed.2016.00064] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 08/21/2017] [Indexed: 12/22/2022] Open
Abstract
Interleukin (IL)-26 is abundant in human airways and this cytokine is involved in the local immune response to a bacterial stimulus in vivo. Specifically, local exposure to the toll-like receptor (TLR) 4 agonist endotoxin does increase IL-26 in human airways and this cytokine potentiates chemotactic responses in human neutrophils. In addition to T-helper (Th) 17 cells, alveolar macrophages can produce IL-26, but it remains unknown whether this cytokine can also be produced in the airway mucosa per se in response to a viral stimulus. Here, we evaluated whether this is the case using primary bronchial epithelial cells from the airway epithelium in vitro, and exploring the signaling mechanisms involved, including the modulatory effects of additional Th17 cytokines. Finally, we assessed IL-26 and its archetype signaling responses in healthy human airways in vivo. We found increased transcription and release of IL-26 protein after stimulation with the viral-related double stranded (ds) RNA polyinosinic-polycytidylic acid (poly-IC) and showed that this IL-26 release involved mitogen-activated protein (MAP) kinases and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). The release of IL-26 in response to a viral stimulus was modulated by additional Th17 cytokines. Moreover, there was transcription of IL26 mRNA and expression of the protein in epithelial cells of bronchial brush and tissue biopsies respectively after harvest in vivo. In addition, the extracellular IL-26 protein concentrations in bronchoalveolar lavage (BAL) samples did correlate with increased epithelial cell transcription of an archetype intracellular signaling molecule downstream of the IL-26-receptor complex, STAT1, in the bronchial brush biopsies. Thus, our study suggests that viral stimulation causes the production of IL-26 in lining epithelial cells of human airway structural cells that constitute a critical immune barrier and that this production is modulated by Th17 cytokines.
Collapse
Affiliation(s)
- Karlhans Fru Che
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - Riitta Kaarteenaho
- Unit of Medicine and Clinical Research, Pulmonary Division, University of Eastern Finland and Center of Medicine and Clinical Research, Division of Respiratory Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Elisa Lappi-Blanco
- Department of Pathology, Center for Cancer Research and Translational Medicine, University of Oulu, Oulu, Finland
| | - Bettina Levänen
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - Jitong Sun
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - Åsa Wheelock
- Respiratory Medicine Unit. Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, SE-171 76 Stockholm
| | - Lena Palmberg
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden
| | - C Magnus Sköld
- Respiratory Medicine Unit. Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, SE-171 76 Stockholm.,Lung Allergy Clinic, Karolinska University Hospital Solna, Stockholm, SE-171 76 Stockholm, Sweden
| | - Anders Lindén
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, SE-171 77 Stockholm, Sweden.,Lung Allergy Clinic, Karolinska University Hospital Solna, Stockholm, SE-171 76 Stockholm, Sweden
| |
Collapse
|
23
|
Fatima N, Faisal SM, Zubair S, Siddiqui SS, Moin S, Owais M. Emerging role of Interleukins IL-23/IL-17 axis and biochemical markers in the pathogenesis of Type 2 Diabetes: Association with age and gender in human subjects. Int J Biol Macromol 2017; 105:1279-1288. [PMID: 28757426 DOI: 10.1016/j.ijbiomac.2017.07.155] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 01/10/2023]
Abstract
Chronic hyperglycaemia in type 2 diabetes (T2D) is associated with increased oxidative stress and inflammation. Keeping the above fact into consideration we analyse the effect of age and gender on oxidative stress biomarkers and pro-inflammatory cytokines in T2D patients. The study included 148 diabetic and 110 healthy subjects, grouped on the basis of age and gender. Plasma malondialdehyde, protein carbonyl content and nitric oxide levels were elevated significantly in diabetic patients, with significant decrease in Ferric reducing ability of plasma, vitamin C, reduced glutathione, erythrocyte thiol groups and erythrocyte antioxidant enzyme activity and these changes were even more pronounced as age progressed. Serum IL-1β, IL-6, IL-17A, IL-22 levels and TNF-α mRNA expression was significantly upregulated in all the age groups whereas IL-23 mRNA was upregulated only in the higher age group. Female diabetic patients experienced higher oxidative stress and greater serum IL-6 levels and TNF-α mRNA expression as compared to their male counterparts. This study suggested that diabetes onset is accompanied with increased oxidative stress and elevated levels of inflammatory mediators. The effect was more prominent in aged patients. Female patients experienced greater oxidative stress as compared to males of those age groups with slightly higher levels of inflammatory cytokines.
Collapse
Affiliation(s)
- Naureen Fatima
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Syed Mohd Faisal
- Molecular Immunology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Swaleha Zubair
- Women's College, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Sheelu Shafiq Siddiqui
- Rajiv Gandhi Centre for Diabetes and Endocrinology, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Shagufta Moin
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Mohammad Owais
- Molecular Immunology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India.
| |
Collapse
|
24
|
The Role of IL-17 and Related Cytokines in Inflammatory Autoimmune Diseases. Mediators Inflamm 2017; 2017:3908061. [PMID: 28316374 PMCID: PMC5337858 DOI: 10.1155/2017/3908061] [Citation(s) in RCA: 335] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/01/2017] [Indexed: 12/12/2022] Open
Abstract
Interleukin-17 (IL-17) induces the production of granulocyte colony-stimulating factor (G-CSF) and chemokines such as CXCL1 and CXCL2 and is a cytokine that acts as an inflammation mediator. During infection, IL-17 is needed to eliminate extracellular bacteria and fungi, by inducing antimicrobial peptides such as defensin. This cytokine also plays an important role in chronic inflammation that occurs during the pathogenesis of autoimmune diseases and allergies such as human rheumatoid arthritis (RA) for which a mouse model of collagen-induced arthritis (CIA) is available. In autoimmune diseases such as RA and multiple sclerosis (MS), IL-17 is produced by helper T (Th) cells that are stimulated by IL-1β and IL-6 derived from phagocytes such as macrophages and from tissue cells. IL-17 contributes to various lesions that are produced by Th17 cells, one subset of helper T cells, and by γδ T cells and innate lymphoid cells. It strongly contributes to autoimmune diseases that are accompanied by chronic inflammation. Thus, a functional understanding of Th17 cells is extremely important. In this review, we highlight the roles of cytokines that promote the development and maintenance of pathogenic Th17 cells in autoimmune diseases.
Collapse
|
25
|
Combination of Interleukin-27 and MicroRNA for Enhancing Expression of Anti-Inflammatory and Proosteogenic Genes. ARTHRITIS 2017; 2017:6365857. [PMID: 28265470 PMCID: PMC5318630 DOI: 10.1155/2017/6365857] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/28/2016] [Accepted: 12/18/2016] [Indexed: 12/27/2022]
Abstract
Remission of inflammation has become an achievable goal in inflammatory or rheumatoid arthritis (RA); however, bone erosion continues in many patients. Interleukin- (IL-) 27 regulates immune and bone cell balance and also suppresses activities of several inflammatory cell types in RA. Despite its promise, challenges to clinical translation of IL-27 have been its partial effects in vivo. Due to their ability to modulate plasticity of bone and immune cell differentiation, we examined the potential for several microRNA (miR) candidates in enhancing the effects of IL-27. Using differentiation, luciferase, and real time quantitative PCR assays, we show that IL-27 promotes osteoblast differentiation, reduces expression of osteoblast inhibitory genes, and reduces osteoclast differentiation, and results suggest a potential coordination with TGFβ/BMP/SMAD and JAK/STAT pathways. We selected miRNA regulators of these and related pathways to examine whether the effects of IL-27 could be augmented for therapeutic applications. miR-29b and miR-21 augmented IL-27 proosteogenic while downregulating osteoclastogenic signals and also worked to reduce inflammatory signaling in activated macrophages, while miR-21 and miR-20b worked with IL-27 to reduce inflammatory gene expression in fibroblasts and T cells. It appears that several miRNAs can be utilized to enhance IL-27's impact on modulating osteogenesis and reducing proinflammatory signaling.
Collapse
|
26
|
Richards CD. Innate Immune Cytokines, Fibroblast Phenotypes, and Regulation of Extracellular Matrix in Lung. J Interferon Cytokine Res 2017; 37:52-61. [PMID: 28117653 DOI: 10.1089/jir.2016.0112] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation can be caused by adaptive immune responses in autoimmune and allergic conditions, driven by a T lymphocyte subset balance (TH1, TH2, Th17, Th22, and/or Treg) and skewed cellular profiles in an antigen-specific manner. However, several chronic inflammatory diseases have no clearly defined adaptive immune mechanisms that drive chronicity. These conditions include those that affect the lung such as nonatopic asthma or idiopathic pulmonary fibrosis comprising significant health problems. The remodeling of extracellular matrix (ECM) causes organ dysfunction, and it is largely generated by fibroblasts as the major cell controlling net ECM. As such, these are potential targets of treatment approaches in the context of ECM pathology. Fibroblast phenotypes contribute to ECM and inflammatory cell accumulation, and they are integrated into chronic disease mechanisms including cancer. Evidence suggests that innate cytokine responses may be critical in nonallergic/nonautoimmune disease, and they enable environmental agent exposure mechanisms that are independent of adaptive immunity. Innate immune cytokines derived from macrophage subsets (M1/M2) and innate lymphoid cell (ILC) subsets can directly regulate fibroblast function. We also suggest that STAT3-activating gp130 cytokines can sensitize fibroblasts to the innate cytokine milieu to drive phenotypes and exacerbate existing adaptive responses. Here, we review evidence exploring innate cytokine regulation of fibroblast behavior.
Collapse
Affiliation(s)
- Carl D Richards
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University , Hamilton, Canada
| |
Collapse
|
27
|
Kanai K, Park AM, Yoshida H, Tsunoda I, Yoshie O. IL-35 Suppresses Lipopolysaccharide-Induced Airway Eosinophilia in EBI3-Deficient Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:119-127. [PMID: 27881708 DOI: 10.4049/jimmunol.1600506] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 10/19/2016] [Indexed: 02/05/2023]
Abstract
EBI3 functions as the subunit of immune-regulatory cytokines, such as IL-27 and IL-35, by pairing with p28 and p35, respectively. We treated wild-type and EBI3-deficient mice with intratracheal administration of LPS and obtained bronchoalveolar lavage fluid (BALF) 24 h later. Although neutrophils were the predominant cells in BALF from both groups of mice, eosinophils were highly enriched and there was increased production of eosinophil-attracting chemokines CCL11 and CCL24 in BALF of EBI3-deficient mice. The bronchial epithelial cells and alveolar macrophages were the major producers of CCL11 and CCL24. Because no such increases in eosinophils were seen in BALF of p28/IL-27-deficient mice or WSX-1/IL-27Rα subunit-deficient mice upon intratracheal stimulation with LPS, we considered that the lack of IL-35 was responsible for the enhanced airway eosinophilia in EBI3-deficient mice. In vitro, IL-35 potently suppressed production of CCL11 and CCL24 by human lung epithelial cell lines treated with TNF-α and IL-1β. IL-35 also suppressed phosphorylation of STAT1 and STAT3 and induced suppressor of cytokine signaling 3. In vivo, rIL-35 dramatically reduced LPS-induced airway eosinophilia in EBI3-deficient mice, with concomitant reduction of CCL11 and CCL24, whereas neutralization of IL-35 significantly increased airway eosinophils in LPS-treated wild-type mice. Collectively, our results suggest that IL-35 negatively regulates airway eosinophilia, at least in part by reducing the production of CCL11 and CCL24.
Collapse
Affiliation(s)
- Kyosuke Kanai
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan; and
| | - Ah-Mee Park
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan; and
| | - Hiroki Yoshida
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Saga University Faculty of Medicine, Saga 840-8502, Japan
| | - Ikuo Tsunoda
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan; and
| | - Osamu Yoshie
- Department of Microbiology, Kindai University Faculty of Medicine, Osaka 589-8511, Japan; and
| |
Collapse
|
28
|
Lee H, Ryu WI, Kim HJ, Bae HC, Ryu HJ, Shin JJ, Song KH, Kim TW, Son SW. TSLP Down-Regulates S100A7 and ß-Defensin 2 Via the JAK2/STAT3-Dependent Mechanism. J Invest Dermatol 2016; 136:2427-2435. [DOI: 10.1016/j.jid.2016.07.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 07/07/2016] [Accepted: 07/25/2016] [Indexed: 12/21/2022]
|
29
|
Redhu NS, Gounni AS. IgE regulates airway smooth muscle phenotype: Future perspectives in allergic asthma. World J Immunol 2016; 6:126-130. [DOI: 10.5411/wji.v6.i3.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 08/15/2016] [Accepted: 10/27/2016] [Indexed: 02/05/2023] Open
Abstract
The purpose of this commentary is to highlight the emerging role of IgE on airway smooth muscle (ASM) cells function through activation of the high-affinity Fc receptor for IgE. We discuss the potential implications of IgE-mediated ASM sensitization in airway inflammation and remodeling, the hallmark features of allergic asthma.
Collapse
|
30
|
Massoud AH, Charbonnier LM, Lopez D, Pellegrini M, Phipatanakul W, Chatila TA. An asthma-associated IL4R variant exacerbates airway inflammation by promoting conversion of regulatory T cells to TH17-like cells. Nat Med 2016; 22:1013-22. [PMID: 27479084 PMCID: PMC5014738 DOI: 10.1038/nm.4147] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 06/15/2016] [Indexed: 12/14/2022]
Abstract
Mechanisms by which regulatory T (Treg) cells fail to control inflammation in asthma remain poorly understood. We show that a severe asthma-associated polymorphism in the gene encoding the interleukin (IL)-4 receptor alpha chain (Il4ra(R576)) promotes conversion of induced Treg (iTreg) cells toward a T helper 17 (TH17) cell fate. This skewing is mediated by the recruitment by IL-4Rα(R576) of the growth-factor-receptor-bound protein 2 (GRB2) adaptor protein, which drives IL-17 expression by activating a pathway that involves extracellular-signal-regulated kinase, IL-6 and the transcription factor STAT3. Treg cell-specific deletion of genes that regulate TH17 cell differentiation, including Il6ra and RAR-related orphan receptor gamma (Rorc), but not of Il4 or Il13, prevented exacerbated airway inflammation in mice expressing Il4ra(R576) (hereafter referred to as Il4ra(R576) mice). Furthermore, treatment of Il4ra(R576) mice with a neutralizing IL-6-specific antibody prevented iTreg cell reprogramming into TH17-like cells and protected against severe airway inflammation. These findings identify a previously unknown mechanism for the development of mixed TH2-TH17 cell inflammation in genetically prone individuals and point to interventions that stabilize iTreg cells as potentially effective therapeutic strategies.
Collapse
MESH Headings
- Adolescent
- Adult
- Animals
- Antibodies, Neutralizing/pharmacology
- Asthma/genetics
- Asthma/immunology
- Case-Control Studies
- Cell Differentiation/drug effects
- Cell Differentiation/immunology
- Child
- Disease Models, Animal
- Female
- Flow Cytometry
- GRB2 Adaptor Protein/immunology
- Gene Expression Profiling
- Humans
- Immunoblotting
- Immunoprecipitation
- Inflammation/immunology
- Interleukin-13/immunology
- Interleukin-17/immunology
- Interleukin-4 Receptor alpha Subunit/genetics
- Interleukin-4 Receptor alpha Subunit/immunology
- Interleukin-6/immunology
- Lung/immunology
- Male
- Mice
- Middle Aged
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Polymorphism, Genetic
- Real-Time Polymerase Chain Reaction
- Receptors, Cell Surface/genetics
- Receptors, Interleukin-6/genetics
- Receptors, Interleukin-6/immunology
- Respiratory Hypersensitivity/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- STAT3 Transcription Factor/immunology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Young Adult
Collapse
Affiliation(s)
- Amir Hossein Massoud
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Louis-Marie Charbonnier
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - David Lopez
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California, USA
| | - Wanda Phipatanakul
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Zhang Y, Li AL, Sun YQ, Du P, Liu LB, Li S, Zhang C. Lactobacillus acidophilus regulates STAT3 and STAT5 signaling in bovine β-lg-sensitized mice model. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s13594-016-0284-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
32
|
Regulation of Interleukin-17 Production. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 941:139-166. [DOI: 10.1007/978-94-024-0921-5_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Kwofie K, Scott M, Rodrigues R, Guerette J, Radford K, Nair P, Richards CD. Regulation of IL-17A responses in human airway smooth muscle cells by Oncostatin M. Respir Res 2015; 16:14. [PMID: 25849622 PMCID: PMC4332894 DOI: 10.1186/s12931-014-0164-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 12/30/2014] [Indexed: 11/25/2022] Open
Abstract
Background Regulation of human airway smooth muscle cells (HASMC) by cytokines contributes to chemotactic factor levels and thus to inflammatory cell accumulation in lung diseases. Cytokines such as the gp130 family member Oncostatin M (OSM) can act synergistically with Th2 cytokines (IL-4 and IL-13) to modulate lung cells, however whether IL-17A responses by HASMC can be altered is not known. Objective To determine the effects of recombinant OSM, or other gp130 cytokines (LIF, IL-31, and IL-6) in regulating HASMC responses to IL-17A, assessing MCP-1/CCL2 and IL-6 expression and cell signaling pathways. Methods Cell responses of primary HASMC cultures were measured by the assessment of protein levels in supernatants (ELISA) and mRNA levels (qRT-PCR) in cell extracts. Activation of STAT, MAPK (p38) and Akt pathways were measured by immunoblot. Pharmacological agents were used to assess the effects of inhibition of these pathways. Results OSM but not LIF, IL-31 or IL-6 could induce detectable responses in HASMC, elevating MCP-1/CCL2, IL-6 levels and activation of STAT-1, 3, 5, p38 and Akt cell signaling pathways. OSM induced synergistic action with IL-17A enhancing MCP-1/CCL-2 and IL-6 mRNA and protein expression, but not eotaxin-1 expression, while OSM in combination with IL-4 or IL-13 synergistically induced eotaxin-1 and MCP-1/CCL2. OSM elevated steady state mRNA levels of IL-4Rα, OSMRβ and gp130, but not IL-17RA or IL-17RC. Pharmacologic inhibition of STAT3 activation using Stattic down-regulated OSM, OSM/IL-4 or OSM/IL-13, and OSM/IL-17A synergistic responses of MCP-1/CCL-2 induction, whereas, inhibitors of Akt and p38 MAPK resulted in less reduction in MCP-1/CCL2 levels. IL-6 expression was more sensitive to inhibition of p38 (using SB203580) and was affected by Stattic in response to IL-17A/OSM stimulation. Conclusions Oncostatin M can regulate HASMC responses alone or in synergy with IL-17A. OSM/IL-17A combinations enhance MCP-1/CCL2 and IL-6 but not eotaxin-1. Thus, OSM through STAT3 activation of HASMC may participate in inflammatory cell recruitment in inflammatory airway disease. Electronic supplementary material The online version of this article (doi:10.1186/s12931-014-0164-4) contains supplementary material, which is available to authorized users.
Collapse
|
34
|
Th17 differentiation and their pro-inflammation function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 841:99-151. [PMID: 25261206 DOI: 10.1007/978-94-017-9487-9_5] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CD4(+) T helper cells are classical but constantly reinterpreted T-cell subset, playing critical roles in a diverse range of inflammatory responses or diseases. Depending on the cytokines they release and the immune responses they mediate, CD4(+) T cells are classically divided into two major cell populations: Th1 and Th2 cells. However, recent studies challenged this Th1/Th2 paradigm by discovering several T-helper cell subsets with specific differentiation program and functions, including Th17 cells, Treg cells, and Tfh cells. In this chapter, we summarize the current understanding and recent progresses on the Th17 lineage differentiation and its effector impacts on variety of inflammatory responses or disease pathogenesis.
Collapse
|
35
|
Abstract
: Interleukin-17 receptor A (IL-17RA) is responsible for both IL-17A and IL-25 (IL-17E) signaling pathways. Current evidences suggest distinct but interactive responses between IL-17A and IL-25 signaling, both of which are critical for intestinal immune homeostasis. IL-17RA is assumed to regulate this counterbalance and therefore becomes a crucial molecule in mucosal immunology. In this review, we will describe the structure of IL-17RA, compare IL-17A and IL-25 signaling pathways, and emphasize on the function of IL-17RA in intestinal inflammation and discuss current evidences of accomplished and ongoing clinical trials with monoclonal antibodies targeting Th17 pathway, especially IL-17RA.
Collapse
|
36
|
Yousefidaredor H, Zare-Bidaki M, Hakimi H, Assar S, Bagheri V, Arababadi MK. IL-17A plays an important role in induction of type 2 diabetes and its complications. ASIAN PACIFIC JOURNAL OF TROPICAL DISEASE 2014. [DOI: 10.1016/s2222-1808(14)60598-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
37
|
Huber AK, Wang L, Han P, Zhang X, Ekholm S, Srinivasan A, Irani DN, Segal BM. Dysregulation of the IL-23/IL-17 axis and myeloid factors in secondary progressive MS. Neurology 2014; 83:1500-7. [PMID: 25253754 DOI: 10.1212/wnl.0000000000000908] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE In the current exploratory study, we longitudinally measured immune parameters in the blood of individuals with relapsing-remitting multiple sclerosis (RRMS) and secondary progressive multiple sclerosis (SPMS), and investigated their relationship to disease duration and clinical and radiologic measures of CNS injury. METHODS Peripheral blood mononuclear cells (PBMCs) and plasma were obtained from subjects with RRMS, SPMS, and from healthy controls on a monthly basis over the course of 1 year. MRI and Expanded Disability Status Scale evaluations were performed serially. PBMCs were analyzed by enzyme-linked immunosorbent spot assay to enumerate myelin basic protein-specific interleukin (IL)-17- and interferon (IFN)-γ-producing cells. Plasma concentrations of proinflammatory factors were measured using customized Luminex panels. RESULTS Frequencies of myelin basic protein-specific IL-17- and IFN-γ-producing PBMCs were higher in individuals with RRMS and SPMS compared to healthy controls. Patients with SPMS expressed elevated levels of IL-17-inducible chemokines that activate and recruit myeloid cells. In the cohort of patients with SPMS without inflammatory activity, upregulation of myeloid-related factors correlated directly with MRI T2 lesion burden and inversely with brain parenchymal tissue volume. CONCLUSIONS The results of this exploratory study raise the possibility that Th17 responses and IL-17-inducible myeloid factors are elevated during SPMS compared with RRMS, and correlate with lesion burden. Our data endorse further investigation of Th17- and myeloid-related factors as candidate therapeutic targets in SPMS.
Collapse
Affiliation(s)
- Amanda K Huber
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Lu Wang
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Peisong Han
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Xu Zhang
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Sven Ekholm
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Ashok Srinivasan
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - David N Irani
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY
| | - Benjamin M Segal
- From the Holtom-Garrett Program in Neuroimmunology and Multiple Sclerosis Center, Departments of Neurology (A.K.H., D.N.I., B.M.S.), Biostatistics (L.W.), and Radiology (A.S.), University of Michigan, Ann Arbor; Neurology Service (B.M.S.), VA Ann Arbor Healthcare System, MI; Department of Statistics and Actuarial Science (P.H.), University of Waterloo, Canada; Department of Mathematics and Statistics (X.Z.), Bowling Green State University, OH; and Department of Imaging Sciences (S.E.), University of Rochester Medical Center, NY.
| |
Collapse
|
38
|
Glosson NL, Bruns HA, Kaplan MH. Wheezing and itching: The requirement for STAT proteins in allergic inflammation. JAKSTAT 2014; 1:3-12. [PMID: 24058746 PMCID: PMC3670132 DOI: 10.4161/jkst.19086] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 12/16/2011] [Indexed: 12/16/2022] Open
Abstract
The development of allergic inflammation requires the orchestration of gene expression from the inflamed tissue and from the infiltrating immune cells. Since many of the cytokines that promote allergic inflammation signal through hematopoietin family receptors, the Signal Transducer and Activator of Transcription (STAT) family have obligate roles in pro-allergic cytokine-induced gene regulation in multiple cell types. In this review, we summarize work defining the contribution of each of the STAT family members to the development of allergic inflammation, using data from mouse models of allergic inflammation, studies on patient samples and correlations with single nucleotide polymorphisms in STAT genes.
Collapse
Affiliation(s)
- Nicole L Glosson
- Department of Pediatrics; Herman B. Wells Center for Pediatric Research; Department of Microbiology and Immunology; Indiana University School of Medicine; Indianapolis, IN USA
| | | | | |
Collapse
|
39
|
Xu Y, Xu X, Gao X, Chen H, Geng L. Shikonin suppresses IL-17-induced VEGF expression via blockage of JAK2/STAT3 pathway. Int Immunopharmacol 2014; 19:327-33. [DOI: 10.1016/j.intimp.2014.01.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 01/02/2014] [Accepted: 01/20/2014] [Indexed: 12/25/2022]
|
40
|
Thymic stromal lymphopoietin induces migration in human airway smooth muscle cells. Sci Rep 2014; 3:2301. [PMID: 23892442 PMCID: PMC3725475 DOI: 10.1038/srep02301] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 07/10/2013] [Indexed: 02/06/2023] Open
Abstract
Airway remodeling due to increased airway smooth muscle (ASM) mass, likely due to enhanced migration and proliferation, has been shown to be highly associated with decline in lung function in asthma. Thymic stromal lymphopoietin (TSLP) is an IL-7-like, pro-allergic cytokine that has been shown to be necessary and sufficient for the development of allergic asthma. Human ASM (HASM) cells express TSLP receptor (TSLPR), the activation of which leads to enhanced release of proinflammatory mediators such as IL-6, CCL11/eotaxin-1, and CXCL8/IL-8. We show here that TSLP induces HASM cell migration through STAT3 activation since lentiviral-shRNA inhibition of STAT3 abrogated the TSLP-induced cell migration. Moreover, TSLP induced multiple cytoskeleton changes in HASM cells such as actin polymerization, cell polarization, and activation of small GTPase Rac1. Collectively, our data suggest a pro-migratory function of TSLP in ASM remodeling and provides better rationale for targeting TSLP/TSLPR pathway for therapeutic approaches in allergic asthma.
Collapse
|
41
|
Th17-associated cytokines as a therapeutic target for steroid-insensitive asthma. Clin Dev Immunol 2013; 2013:609395. [PMID: 24454477 PMCID: PMC3886588 DOI: 10.1155/2013/609395] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 12/05/2013] [Indexed: 02/04/2023]
Abstract
Steroid-insensitive asthma is an infrequent but problematic airway disease that presents with persistent symptoms, airflow limitation, or recurrent exacerbations even when treated with steroid-based therapies. Because of unsatisfactory results obtained from currently available therapies for steroid-insensitive asthma, a better understanding of its pathogenesis and the development of new targeted molecular therapies are warranted. Recent studies indicated that levels of interleukin (IL)-17 are increased and both eosinophils and neutrophils infiltrate the airways of severe asthmatics. IL-17 is a proinflammatory cytokine mainly secreted from helper T (Th) 17 cells and is important for the induction of neutrophil recruitment and migration at sites of inflammation. This review focuses on the pathogenetic role of Th17 cells and their associated cytokines in steroid-insensitive asthma and discusses the prospects of novel therapeutic options targeting the Th17 signaling pathway.
Collapse
|
42
|
Saleh A, Smith DR, Tessler L, Mateo AR, Martens C, Schartner E, Van der Ploeg R, Toth C, Zochodne DW, Fernyhough P. Receptor for advanced glycation end-products (RAGE) activates divergent signaling pathways to augment neurite outgrowth of adult sensory neurons. Exp Neurol 2013; 249:149-59. [DOI: 10.1016/j.expneurol.2013.08.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/27/2013] [Accepted: 08/30/2013] [Indexed: 11/26/2022]
|
43
|
Saleh A, Chowdhury SKR, Smith DR, Balakrishnan S, Tessler L, Schartner E, Bilodeau A, Van Der Ploeg R, Fernyhough P. Diabetes impairs an interleukin-1β-dependent pathway that enhances neurite outgrowth through JAK/STAT3 modulation of mitochondrial bioenergetics in adult sensory neurons. Mol Brain 2013; 6:45. [PMID: 24152426 PMCID: PMC4016027 DOI: 10.1186/1756-6606-6-45] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/21/2013] [Indexed: 01/07/2023] Open
Abstract
Background A luminex-based screen of cytokine expression in dorsal root ganglia (DRG) and nerve of type 1 diabetic rodents revealed interleukin-1 (IL-1α) and IL-1β to be significantly depressed. We, therefore, tested the hypothesis that impaired IL-1α and IL-1β expression in DRG may contribute to aberrant axon regeneration and plasticity seen in diabetic sensory neuropathy. In addition, we determined if these cytokines could optimize mitochondrial bioenergetics since mitochondrial dysfunction is a key etiological factor in diabetic neuropathy. Results Cytokines IL-1α and IL-1β were reduced 2-fold (p<0.05) in DRG and/or nerve of 2 and 5 month streptozotocin (STZ)-diabetic rats. IL-2 and IL-10 were unchanged. IL-1α and IL-1β induced similar 2 to 3-fold increases in neurite outgrowth in cultures derived from control or diabetic rats (p<0.05). STAT3 phosphorylation on Tyr705 or Ser727 was depressed in DRG from STZ-diabetic mice and treatment of cultures derived from STZ-diabetic rats with IL-1β for 30 min raised phosphorylation of STAT3 on Tyr705 and Ser727 by 1.5 to 2-fold (p<0.05). shRNA-based or AG490 inhibition of STAT3 activity or shRNA blockade of endogenous IL-1β expression completely blocked neurite outgrowth. Cultured neurons derived from STZ-diabetic mice were treated for 24 hr with IL-1β and maximal oxygen consumption rate and spare respiratory capacity, both key measures of bioenergetic fidelity that were depressed in diabetic compared with control neurons, were enhanced 2-fold. This effect was blocked by AG490. Conclusions Endogenous synthesis of IL-1β is diminished in nerve tissue in type 1 diabetes and we propose this defect triggers reduced STAT3 signaling and mitochondrial function leading to sup-optimal axonal regeneration and plasticity.
Collapse
Affiliation(s)
- Ali Saleh
- Division of Neurodegenerative Disorders, St, Boniface Hospital Research Centre, R4048 - 351 Tache Ave, Winnipeg, MB R2H 2A6, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Redhu NS, Shan L, Al-Subait D, Ashdown HL, Movassagh H, Lamkhioued B, Gounni AS. IgE induces proliferation in human airway smooth muscle cells: role of MAPK and STAT3 pathways. Allergy Asthma Clin Immunol 2013; 9:41. [PMID: 24499258 PMCID: PMC3842672 DOI: 10.1186/1710-1492-9-41] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 10/01/2013] [Indexed: 12/27/2022] Open
Abstract
Airway remodeling is not specifically targeted by current asthma medications, partly owing to the lack of understanding of remodeling mechanisms, altogether posing great challenges in asthma treatment. Increased airway smooth muscle (ASM) mass due to hyperplasia/hypertrophy contributes significantly to overall airway remodeling and correlates with decline in lung function. Recent evidence suggests that IgE sensitization can enhance the survival and mediator release in inflammatory cells. Human ASM (HASM) cells express both low affinity (FcεRII/CD23) and high affinity IgE Fc receptors (FcεRI), and IgE can modulate the contractile and synthetic function of HASM cells. IgE was recently shown to induce HASM cell proliferation but the detailed mechanisms remain unknown. We report here that IgE sensitization induces HASM cell proliferation, as measured by 3H-thymidine, EdU incorporation, and manual cell counting. As an upstream signature component of FcεRI signaling, inhibition of spleen tyrosine kinase (Syk) abrogated the IgE-induced HASM proliferation. Further analysis of IgE-induced signaling depicted an IgE-mediated activation of Erk 1/2, p38, JNK MAPK, and Akt kinases. Lastly, lentiviral-shRNA-mediated STAT3 silencing completely abolished the IgE-mediated HASM cell proliferation. Collectively, our data provide mechanisms of a novel function of IgE which may contribute, at least in part, to airway remodeling observed in allergic asthma by directly inducing HASM cell proliferation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Abdelilah S Gounni
- Department of Immunology, Faculty of Medicine, University of Manitoba, 419 Apotex Centre- 750 McDermot Ave, Winnipeg, MB R3E 0T5, Canada.
| |
Collapse
|
45
|
Geraghty P, Wyman AE, Garcia-Arcos I, Dabo AJ, Gadhvi S, Foronjy R. STAT3 modulates cigarette smoke-induced inflammation and protease expression. Front Physiol 2013; 4:267. [PMID: 24101903 PMCID: PMC3787396 DOI: 10.3389/fphys.2013.00267] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 09/09/2013] [Indexed: 11/17/2022] Open
Abstract
Signal transducer and activator of transcription-3 (STAT3) regulates inflammation, apoptosis, and protease expression, which are critical processes associated with airway injury and lung tissue destruction. However, the precise role of STAT3 in the development of airway diseases such as chronic obstructive pulmonary disease (COPD) has not been established. This study shows that cigarette smoke activates STAT3 in the lungs of mice. Since cigarette smoke activated STAT3 in the lung, we then evaluated how the loss of STAT3 would impact on smoke-mediated lung inflammation, protease expression, and apoptosis. STAT3+/+ and STAT3−/− mice were exposed to 8 days of cigarette smoke. Compared to the STAT3+/+ mice bronchoalveolar lavage fluid (BALF) cellularity was significantly elevated in the STAT3−/− mice both before and after cigarette smoke exposure, with the increase in cells primarily macrophages. In addition, smoke exposure induced significantly higher BALF protein levels of Interleukin-1α (IL-1α), and monocyte chemotactic protein-1 (MCP-1) and higher tissue expression of keratinocyte chemoattractant (KC) in the STAT3−/− mice. Lung mRNA expression of MMP-12 was increased in STAT3−/− at baseline. However, the smoke-induced increase in MMP-10 expression seen in the STAT3+/+ mice was not observed in the STAT3−/− mice. Moreover, lung protein levels of the anti-inflammatory proteins SOCS3 and IL-10 were markedly lower in the STAT3−/− mice compared to the STAT3+/+ mice. Lastly, apoptosis, as determined by caspase 3/7 activity assay, was increased in the STAT3−/− at baseline to levels comparable to those observed in the smoke-exposed STAT3+/+ mice. Together, these results indicate that the smoke-mediated induction of lung STAT3 activity may play a critical role in maintaining normal lung homeostasis and function.
Collapse
Affiliation(s)
- Patrick Geraghty
- Division of Pulmonary and Critical Care Medicine, St. Luke's Roosevelt Health Sciences Center, New York NY, USA
| | | | | | | | | | | |
Collapse
|
46
|
Song X, Qian Y. IL-17 family cytokines mediated signaling in the pathogenesis of inflammatory diseases. Cell Signal 2013; 25:2335-47. [PMID: 23917206 DOI: 10.1016/j.cellsig.2013.07.021] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 07/26/2013] [Indexed: 12/26/2022]
Abstract
Inflammation is the immediate protective response of the body to pathogen invasions, allergen challenges, chemical exposures or physical injuries. Acute inflammation usually accompanies with transient infiltration of leukocytes, removal of danger signals and eventually tissue repair, while persistent and uncontrolled inflammation becomes a major stimulator in the progression of many chronic diseases in human, including autoimmune diseases, metabolic disorders and cancer. Interleukin (IL)-17 family is a recent classified subset of cytokines, playing critical roles in both acute and chronic inflammatory responses. In this review, we will summarize recent progresses on the signalings of this family cytokines and their impacts on the inflammatory responses or disorders.
Collapse
Affiliation(s)
- Xinyang Song
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | |
Collapse
|
47
|
Tan Z, Qian X, Jiang R, Liu Q, Wang Y, Chen C, Wang X, Ryffel B, Sun B. IL-17A plays a critical role in the pathogenesis of liver fibrosis through hepatic stellate cell activation. THE JOURNAL OF IMMUNOLOGY 2013; 191:1835-44. [PMID: 23842754 DOI: 10.4049/jimmunol.1203013] [Citation(s) in RCA: 247] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Liver fibrosis is a severe, life-threatening clinical condition resulting from nonresolving hepatitis of different origins. IL-17A is critical in inflammation, but its relation to liver fibrosis remains elusive. We find increased IL-17A expression in fibrotic livers from HBV-infected patients undergoing partial hepatectomy because of cirrhosis-related early-stage hepatocellular carcinoma in comparison with control nonfibrotic livers from uninfected patients with hepatic hemangioma. In fibrotic livers, IL-17A immunoreactivity localizes to the inflammatory infiltrate. In experimental carbon tetrachloride-induced liver fibrosis of IL-17RA-deficient mice, we observe reduced neutrophil influx, proinflammatory cytokines, hepatocellular necrosis, inflammation, and fibrosis as compared with control C57BL/6 mice. IL-17A is produced by neutrophils and T lymphocytes expressing the Th17 lineage-specific transcription factor Retinoic acid receptor-related orphan receptor γt. Furthermore, hepatic stellate cells (HSCs) isolated from naive C57BL/6 mice respond to IL-17A with increased IL-6, α-smooth muscle actin, collagen, and TGF-β mRNA expression, suggesting an IL-17A-driven fibrotic process. Pharmacologic ERK1/2 or p38 inhibition significantly attenuated IL-17A-induced HSC activation and collagen expression. In conclusion, IL-17A(+) Retinoic acid receptor-related orphan receptor γt(+) neutrophils and T cells are recruited into the injured liver driving a chronic, fibrotic hepatitis. IL-17A-dependent HSC activation may be critical for liver fibrosis. Thus, blockade of IL-17A could potentially benefit patients with chronic hepatitis and liver fibrosis.
Collapse
Affiliation(s)
- Zhongming Tan
- Liver Transplantation Center of First Affiliated Hospital and State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu Province 210029, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Waddell A, Ahrens R, Tsai YT, Sherrill JD, Denson LA, Steinbrecher KA, Hogan SP. Intestinal CCL11 and eosinophilic inflammation is regulated by myeloid cell-specific RelA/p65 in mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:4773-85. [PMID: 23562811 PMCID: PMC3969817 DOI: 10.4049/jimmunol.1200057] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In inflammatory bowel diseases (IBDs), particularly ulcerative colitis, intestinal macrophages (MΦs), eosinophils, and the eosinophil-selective chemokine CCL11, have been associated with disease pathogenesis. MΦs, a source of CCL11, have been reported to be of a mixed classical (NF-κB-mediated) and alternatively activated (STAT-6-mediated) phenotype. The importance of NF-κB and STAT-6 pathways to the intestinal MΦ/CCL11 response and eosinophilic inflammation in the histopathology of experimental colitis is not yet understood. Our gene array analyses demonstrated elevated STAT-6- and NF-κB-dependent genes in pediatric ulcerative colitis colonic biopsies. Dextran sodium sulfate (DSS) exposure induced STAT-6 and NF-κB activation in mouse intestinal F4/80(+)CD11b(+)Ly6C(hi) (inflammatory) MΦs. DSS-induced CCL11 expression, eosinophilic inflammation, and histopathology were attenuated in RelA/p65(Δmye) mice, but not in the absence of STAT-6. Deletion of p65 in myeloid cells did not affect inflammatory MΦ recruitment or alter apoptosis, but did attenuate LPS-induced cytokine production (IL-6) and Ccl11 expression in purified F4/80(+)CD11b(+)Ly6C(hi) inflammatory MΦs. Molecular and cellular analyses revealed a link between expression of calprotectin (S100a8/S100a9), Ccl11 expression, and eosinophil numbers in the DSS-treated colon. In vitro studies of bone marrow-derived MΦs showed calprotectin-induced CCL11 production via a p65-dependent mechanism. Our results indicate that myeloid cell-specific NF-κB-dependent pathways play an unexpected role in CCL11 expression and maintenance of eosinophilic inflammation in experimental colitis. These data indicate that targeting myeloid cells and NF-κB-dependent pathways may be of therapeutic benefit for the treatment of eosinophilic inflammation and histopathology in IBD.
Collapse
Affiliation(s)
- Amanda Waddell
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Richard Ahrens
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Yi Ting Tsai
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Joseph D. Sherrill
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Lee A. Denson
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Kris A. Steinbrecher
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| | - Simon P. Hogan
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229
| |
Collapse
|
49
|
Meephansan J, Komine M, Tsuda H, Karakawa M, Tominaga SI, Ohtsuki M. Expression of IL-33 in the epidermis: The mechanism of induction by IL-17. J Dermatol Sci 2013; 71:107-14. [PMID: 23764374 DOI: 10.1016/j.jdermsci.2013.04.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 04/08/2013] [Accepted: 04/11/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Interleukin (IL)-33 is a dual functional, IL-1 family member cytokine, whose exact roles in inflammatory skin diseases are still unknown. IL-17A is a key cytokine in the pathogenesis of psoriasis. OBJECTIVES We investigated if IL-17A could induce IL-33 in epidermal keratinocytes, and the signaling mechanisms involved. METHODS IL-33 levels were evaluated by RT-PCR and western blot in human keratinocytes following IL-17A simulation. IL-33 immunohistochemical staining of psoriatic skin samples was also performed and compared with that of control tissues. The role of signaling pathways downstream of IL-17A was investigated using small molecule inhibitors of EGFR, ERK, p38, and JAK. Adenovirus vector expressing dominant negative STAT1 was also utilized. RESULTS IL-33 and its receptor, ST2L, were expressed in the psoriatic epidermis, and the associated infiltrating cells. IL-17A induced IL-33 expression at mRNA and protein levels in a time- and concentration-dependent manner. IL-17A caused phosphorylation of EGFR, ERK, p38, and STAT1. IL-17A-induced IL-33 expression was blocked by the addition of EGFR, ERK, p38, and JAK inhibitors, and dominant negative STAT1-expressing adenovirus vector. CONCLUSION IL-17A induced IL-33 in NHEKs through EGFR, ERK, p38, and JAK/STAT1 pathways, which were necessary for the induction of IL-33. IL-33, induced by IL-17A in epidermal keratinocytes, may be involved in the pathophysiology of inflammatory skin diseases, including psoriasis.
Collapse
Affiliation(s)
- Jitlada Meephansan
- Department of Dermatology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Song X, Qian Y. The activation and regulation of IL-17 receptor mediated signaling. Cytokine 2013; 62:175-82. [PMID: 23557798 DOI: 10.1016/j.cyto.2013.03.014] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/27/2013] [Accepted: 03/08/2013] [Indexed: 12/19/2022]
Abstract
Interleukin-17 (IL-17), the signature cytokine produced by T helper 17 (Th17) cells, plays pivotal roles in host defense responses against microbial invasion, as well as in the pathogenesis of autoimmune diseases and allergic syndromes. IL-17 activates several downstream signaling pathways including NF-κB, MAPKs and C/EBPs to induce gene expression of antibacterial peptides, proinflammatory chemokines and cytokines and matrix metalloproteinases (MMPs). IL-17 can also stabilize mRNAs of genes induced by TNFα. Although the physiological and pathological functions of IL-17 have been studied for many years, the landscape of its signaling transduction has not been described until recently. The cytosolic adaptor molecule Act1 (also known as CIKS) is considered as the master mediator of IL-17 signaling. In this review, we will summarize recent progress on activation and regulation of IL-17 mediated signal transduction, especially on Act1 mediated regulation of the signaling.
Collapse
Affiliation(s)
- Xinyang Song
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | |
Collapse
|