1
|
Ji H, Jiang W, Zhang J, Liu M, Su D, Lei J, Li L, Zheng M, Liu T, Liu Z, Cao Q, Xu L, Xiong S, Wen Z. ENPP1 governs the metabolic regulation of effector T cells in autoimmunity by detecting cytosolic mitochondrial DNA. Cell Rep 2025; 44:115851. [PMID: 40516055 DOI: 10.1016/j.celrep.2025.115851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 04/25/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025] Open
Abstract
T cells play a pivotal role in the pathogenesis of systemic lupus erythematosus (SLE), yet the underlying molecular mechanisms governing their fate remain elusive. Here, we identify cytosolic mitochondrial DNA (mtDNA) as an intrinsic trigger for driving effector T cell differentiation in patients with SLE. Specifically, accumulated cytosolic mtDNA is sensed by ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), which enhances the transcription of GLUT1 and glycolysis in SLE T cells. This metabolic shift reduces lipogenesis and depletes free fatty acids (FFAs), impairing the N-myristylation and lysosomal localization of AMP-activated protein kinase (AMPK). Inactive AMPK fails to restrain mammalian target of rapamycin complex 1 (mTORC1), leading to its hyperactivation and driving the mal-differentiation of effector T cells. Consequently, interventions targeting ENPP1, glycolysis, AMPK, and mTORC1 effectively inhibit the generation of immunoglobulin (Ig)G anti-double-stranded DNA (dsDNA) and the progression of lupus nephritis in humanized SLE chimeras. Overall, our findings uncover an mtDNA-ENPP1-metabolic axis that governs effector T cell fate in autoimmunity.
Collapse
Affiliation(s)
- Huiyan Ji
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| | - Wanwan Jiang
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| | - Juan Zhang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| | - Mengdi Liu
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| | - Danhua Su
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| | - Jiaxin Lei
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| | - Lingyi Li
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| | - Ming Zheng
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| | - Ting Liu
- Department of Rheumatology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Zhichun Liu
- Department of Rheumatology, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, China
| | - Qinghua Cao
- Renal Medicine, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, St Leonards, NSW, Australia; Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Lin Xu
- Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Sidong Xiong
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China
| | - Zhenke Wen
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, MOE Key Laboratory of Geriatric Diseases and Immunology, Soochow University, Suzhou, China.
| |
Collapse
|
2
|
Zheng Y, Peng X, Zhang Y, Liu R, Long J. ELF4 improves sepsis-induced myocardial injury by regulating STING signaling-mediated T cells differentiation. Cell Biol Toxicol 2025; 41:82. [PMID: 40335763 PMCID: PMC12058945 DOI: 10.1007/s10565-025-10029-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 04/13/2025] [Indexed: 05/09/2025]
Abstract
Septic cardiomyopathy (SCM) is a common complication caused by sepsis. T cells differentiation is involved in SCM progression. However, the role and underlying mechanisms of T cells-mediated immunity in SCM remain unclear. This study aimed to investigate the role of STING-mediated T cells differentiation in SCM. Cecal ligation and puncture (CLP) surgery was conducted in mice to establish SCM model. The mice were injected intraperitoneally with STING agonist ADU-S100 and C-176 after modeling. Wild type (WT) mice and CD4-STING-/- mice were employed. Besides, overexpressing vectors of ELF4 (oe-ELF4), short hairpin RNA targeting ELF4 (sh-ELF4) were transfected into 293T cells. STING signaling was found to be activated in sepsis-induced myocardial immune injury in mice. The administration of ADU-S100 exacerbated myocardial injury and inflammation, while C-176 alleviated these effects. Additionally, STING activation influenced T cells differentiation, with an increase in Th1 and Th17 cells and a decrease in Treg cells. Conditional knockout of STING in CD4+ T cells reduced Th1 and Th17 populations and improved myocardial function and histology. Furthermore, ELF4 was found to inhibit STING activation, reducing T cells differentiation into pro-inflammatory subsets. Overexpression of ELF4 in CD4+ T cells ameliorated myocardial damage and improved cardiac function in CLP mice, suggesting that the ELF4-STING signaling axis plays a protective role in sepsis-induced myocardial injury by regulating T cells differentiation.
Collapse
Affiliation(s)
- Yawen Zheng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Xiongjun Peng
- Department of Social Affairs, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Yusha Zhang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Furong District, Changsha City, 410011, Hunan Province, China
| | - Ruilin Liu
- Department of Cardiac Surgery, The Second Xiangya Hospital of Central South University, Changsha City, Hunan Province, China
| | - Junke Long
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139, Middle Renmin Road, Furong District, Changsha City, 410011, Hunan Province, China.
| |
Collapse
|
3
|
Sharma R, Wu K, Han K, Russo AC, Dagur PK, Combs CA, Yao X, Levine SJ, Sack MN. BLOC1S1 Control of Vacuolar Organelle Fidelity Modulates Murine T H2 Cell Immunity and Allergy Susceptibility. Allergy 2024. [PMID: 39737471 DOI: 10.1111/all.16461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/15/2024] [Accepted: 12/09/2024] [Indexed: 01/01/2025]
Abstract
BACKGROUND The levels of biogenesis of lysosome organelles complex 1 subunit 1 (BLOC1S1) control mitochondrial and endolysosome organelle homeostasis and function. Reduced fidelity of these vacuolar organelles is increasingly being recognized as important in instigating cell-autonomous immune cell activation. We reasoned that exploring the role of BLOC1S1 in CD4+ T cells may further advance our understanding of regulatory events linked to mitochondrial and/or endolysosomal function in adaptive immunity. METHODS CD4+ T cells were analyzed from control and CD4+ T-cell-specific BLOC1S1 knockout mice. Polarization profiles were assayed using biochemical and molecular signatures, and signaling pathways were disrupted pharmacologically or via siRNA. Mouse models of airway and skin inflammation were generated by Ovalbumin and MC903 exposure, respectively. RESULTS TH2 regulator GATA3 and phosphorylated STAT6 were preferentially induced in BLOC1S1-depleted primary CD4+ T (TKO) cells. The levels of IL-4, IL-5, and IL-13 were markedly induced in the absence of BLOC1S1. At the organelle level, mitochondrial DNA leakage evoked cGAS-STING and NF-κB pathway activation with subsequent TH2 polarization. The induction of autophagy with rapamycin reduced cytosolic mtDNA and reversed these TH2 signatures. Furthermore, genetic knockdown of STING and NF-κB inhibition ameliorated this immune regulatory cascade in TKO cells. Finally, at a functional level, TKO mice displayed an increased susceptibility to allergic conditions, including dermatitis and allergic asthma. CONCLUSIONS BLOC1S1 depletion in mouse CD4+ T cells mediated disruption of mitochondrial integrity to initiate a predominant TH2-responsive phenotype via STING-NF-κB-driven signaling of the canonical TH2 regulatory program.
Collapse
Affiliation(s)
- Rahul Sharma
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
| | - Kaiyuan Wu
- Cardiovascular Branch, NHLBI, NIH, Bethesda, Maryland, USA
| | - Kim Han
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
| | - Anna Chiara Russo
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
| | | | | | - Xianglan Yao
- Critical Care Medicine and Pulmonary Branch, Bethesda, Maryland, USA
| | - Stewart J Levine
- Critical Care Medicine and Pulmonary Branch, Bethesda, Maryland, USA
| | - Michael N Sack
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
- Cardiovascular Branch, NHLBI, NIH, Bethesda, Maryland, USA
| |
Collapse
|
4
|
Sharma R, Wu K, Han K, Russo AC, Dagur PK, Combs CA, Sack MN. BLOC1S1 control of vacuolar organelle fidelity modulates T H2 cell immunity and allergy susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586144. [PMID: 39803487 PMCID: PMC11722528 DOI: 10.1101/2024.03.21.586144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The levels of biogenesis of lysosome organelles complex 1 subunit 1 (BLOC1S1) control mitochondrial and endolysosome organelle homeostasis and function. Reduced fidelity of these vacuolar organelles is increasingly being recognized as important in instigating cell-autonomous immune cell activation. We reasoned that exploring the role of BLOC1S1 in CD4+ T cells, may further advance our understanding of regulatory events linked to mitochondrial and/or endolysosomal function in adaptive immunity. Transcript levels of the canonical transcription factors driving CD4+T cell polarization in response to activation showed that, the TH2 regulator GATA3 and phosphorylated STAT6 were preferentially induced in BLOC1S1 depleted primary CD4+ T (TKO) cells. In parallel, in response to both T cell receptor activation and in response to TH2 polarization the levels of IL-4, IL-5 and IL-13 were markedly induced in the absence of BLOC1S1. At the organelle level, mitochondrial DNA leakage evoked cGAS-STING and NF-kB pathway activation with subsequent TH2 polarization. The induction of autophagy with rapamycin reduced cytosolic mtDNA and reverses these TH2 signatures. Furthermore, genetic knockdown of STING and STING and NF-κB inhibition ameliorated this immune regulatory cascade in TKO cells. Finally, at a functional level, TKO mice displayed increased susceptible to allergic conditions including atopic dermatitis and allergic asthma. In conclusion, BLOC1S1 depletion mediated disruption of mitochondrial integrity to initiate a predominant TH2 responsive phenotype via STING-NF-κB driven signaling of the canonical TH2 regulatory program.
Collapse
Affiliation(s)
- Rahul Sharma
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Maryland, USA
| | - Kaiyuan Wu
- Cardiovascular Branch, NHLBI, NIH, Maryland, USA
| | - Kim Han
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Maryland, USA
| | - Anna Chiara Russo
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Maryland, USA
| | | | | | - Michael N. Sack
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Maryland, USA
| |
Collapse
|
5
|
Leite JA, Menezes L, Martins E, Rodrigues TS, Tavares L, Ebering A, Schelmbauer C, Martelossi Cebinelli GC, Zinina V, Golden A, Soshnikova N, Zamboni DS, Cunha FQ, Huber M, Silva JS, Waisman A, Carlos D, Saraiva Câmara NO. AIM2 promotes T H17 cells differentiation by regulating RORγt transcription activity. iScience 2023; 26:108134. [PMID: 37867943 PMCID: PMC10585393 DOI: 10.1016/j.isci.2023.108134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/15/2023] [Accepted: 10/02/2023] [Indexed: 10/24/2023] Open
Abstract
AIM2 is an interferon-inducible HIN-200 protein family member and is well-documented for its roles in innate immune responses as a DNA sensor. Recent studies have highlighted AIM2's function on regulatory T cells (Treg) and follicular T cells (Tfh). However, its involvement in Th17 cell differentiation remains unclear. This study reveals that AIM2 promotes Th17 cell differentiation. AIM2 deficiency decreases IL-17A production and downregulates key Th17 associated proteins (RORγt, IL-1R1, IL-23R). AIM2 is located in the nucleus of Th17 cells, where it interacts with RORγt, enhancing its binding to the Il17a promoter. The absence of AIM2 hinders naive CD4 T cells from differentiating into functional Th17 cells and from inducing colitis in Rag1-/- mice. This study uncovers AIM2's role as a regulator of Th17 cell transcriptional programming, highlighting its potential as a therapeutic target for Th17 cell-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Jefferson Antônio Leite
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Luísa Menezes
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Eloisa Martins
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
- Division of Nephrology, School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Tamara Silva Rodrigues
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Lucas Tavares
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Anna Ebering
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Carsten Schelmbauer
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Guilherme C. Martelossi Cebinelli
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Pharmacology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Valeriya Zinina
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Artemiy Golden
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Natalia Soshnikova
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Dario S. Zamboni
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando Q. Cunha
- Department of Pharmacology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Magdalena Huber
- Institute of Systems Immunology, Center for Tumor and Immunology, University of Marburg, Marburg, Germany
| | - João Santana Silva
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Fiocruz-Bi-Institutional Translational Medicine Project, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
- Division of Nephrology, School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Liu S, Yu CY, Wei H. Spherical nucleic acids-based nanoplatforms for tumor precision medicine and immunotherapy. Mater Today Bio 2023; 22:100750. [PMID: 37545568 PMCID: PMC10400933 DOI: 10.1016/j.mtbio.2023.100750] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/08/2023] Open
Abstract
Precise diagnosis and treatment of tumors currently still face considerable challenges due to the development of highly degreed heterogeneity in the dynamic evolution of tumors. With the rapid development of genomics, personalized diagnosis and treatment using specific genes may be a robust strategy to break through the bottleneck of traditional tumor treatment. Nevertheless, efficient in vivo gene delivery has been frequently hampered by the inherent defects of vectors and various biological barriers. Encouragingly, spherical nucleic acids (SNAs) with good modularity and programmability are excellent candidates capable of addressing traditional gene transfer-associated issues, which enables SNAs a precision nanoplatform with great potential for diverse biomedical applications. In this regard, there have been detailed reviews of SNA in drug delivery, gene regulation, and dermatology treatment. Still, to the best of our knowledge, there is no published systematic review summarizing the use of SNAs in oncology precision medicine and immunotherapy, which are considered new guidelines for oncology treatment. To this end, we summarized the notable advances in SNAs-based precision therapy and immunotherapy for tumors following a classification standard of different types of precise spatiotemporal control on active species by SNAs. Specifically, we focus on the structural diversity and programmability of SNAs. Finally, the challenges and possible solutions were discussed in the concluding remarks. This review will promote the rational design and development of SNAs for tumor-precise medicine and immunotherapy.
Collapse
Affiliation(s)
- Songbin Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
7
|
He X, Sun Y, Lu J, Naz F, Ma S, Liu J. Cytoplasmic DNAs: Sources, sensing, and roles in the development of lung inflammatory diseases and cancer. Front Immunol 2023; 14:1117760. [PMID: 37122745 PMCID: PMC10130589 DOI: 10.3389/fimmu.2023.1117760] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Cytoplasmic DNA is emerging as a pivotal contributor to the pathogenesis of inflammatory diseases and cancer, such as COVID-19 and lung carcinoma. However, the complexity of various cytoplasmic DNA-related pathways and their crosstalk remains challenging to distinguish their specific roles in many distinct inflammatory diseases, especially for the underlying mechanisms. Here, we reviewed the latest findings on cytoplasmic DNA and its signaling pathways in inflammatory lung conditions and lung cancer progression. We found that sustained activation of cytoplasmic DNA sensing pathways contributes to the development of common lung diseases, which may result from external factors or mutations of key genes in the organism. We further discussed the interplays between cytoplasmic DNA and anti-inflammatory or anti-tumor effects for potential immunotherapy. In sum, this review aids in understanding the roles of cytoplasmic DNAs and exploring more therapeutic strategies.
Collapse
Affiliation(s)
- Xintong He
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Ye Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jianzhang Lu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Faiza Naz
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Shenglin Ma
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jian Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
- Cancer Center, Zhejiang University, Hangzhou, China
- Biomedical and Heath Translational Research Center of Zhejiang Province, Haining, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Sharma RK, Sharma J, Kumar R, Badal D, Pattekar A, Sehgal S, Gupta A, Jain P, Sachdeva N. TLR9 signalling activation via direct ligation and its functional consequences in CD4 + T cells. Scand J Immunol 2022; 96:e13214. [PMID: 37406035 PMCID: PMC9788197 DOI: 10.1111/sji.13214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 11/29/2022]
Abstract
CpG Oligodeoxynucleotides (ODNs) are established TLR9 ligands; however, their functional responses in CD4+ T cells are believed to be independent of TLR9 and MyD88. We studied ligand-receptor interactions of ODN 2216 and TLR9 in human CD4+ T cells and assessed their consequences in terms of TLR9 signalling and cell phenotype. We demonstrated that the uptake of ODN 2216, a synthetic TLR9 agonist, is controlled by TLR9 signalling molecules and results in an increase in the expression of TLR9 signalling molecules, regulated via a feedback mechanism. Next, the uptake of ODN 2216 resulted in TLR9 signalling dependent but MyD88 independent increase in expression of TGF-β. Finally, ODN 2216 treated CD4+ T cells showed an anti-inflammatory phenotype that was similar to Th3 type of regulatory T cells. These Th3-like cells were able to suppress the proliferation of untreated CD4+ T cells. Collectively, our results demonstrate a direct and interdependent relationship between ODN 2216 uptake and TLR9 signalling in CD4+ T cells. Our findings thus pave the way for future research to explore direct modulation of adaptive immune cells, using innate immune ligands, to subvert exaggerated inflammatory responses.
Collapse
Affiliation(s)
- Ravi Kumar Sharma
- Advanced Eye CentrePost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
- Department of Microbiology and Immunology and the Institute for Molecular Medicine and Infectious DiseaseDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
- Division of Rheumatology, Department of MedicineKarolinska InstitutetSolnaSweden
| | - Jyoti Sharma
- Advanced Eye CentrePost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Rajendra Kumar
- Division of Biological SciencesIndian Institute of Science Education and ResearchMohaliPunjabIndia
- Department of OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Darshan Badal
- Department of EndocrinologyPost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Ajinkya Pattekar
- Department of Microbiology and Immunology and the Institute for Molecular Medicine and Infectious DiseaseDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - Shobha Sehgal
- Department of ImmunopathologyPost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Amod Gupta
- Advanced Eye CentrePost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| | - Pooja Jain
- Department of Microbiology and Immunology and the Institute for Molecular Medicine and Infectious DiseaseDrexel University College of MedicinePhiladelphiaPennsylvaniaUSA
| | - Naresh Sachdeva
- Department of EndocrinologyPost Graduate Institute of Medical Education and Research (PGIMER)ChandigarhIndia
| |
Collapse
|
9
|
Contribution of T- and B-cell intrinsic toll-like receptors to the adaptive immune response in viral infectious diseases. Cell Mol Life Sci 2022; 79:547. [PMID: 36224474 PMCID: PMC9555683 DOI: 10.1007/s00018-022-04582-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 11/03/2022]
Abstract
Toll-like receptors (TLRs) comprise a class of highly conserved molecules that recognize pathogen-associated molecular patterns and play a vital role in host defense against multiple viral infectious diseases. Although TLRs are highly expressed on innate immune cells and play indirect roles in regulating antiviral adaptive immune responses, intrinsic expression of TLRs in adaptive immune cells, including T cells and B cells, cannot be ignored. TLRs expressed in CD4 + and CD8 + T cells play roles in enhancing TCR signal-induced T-cell activation, proliferation, function, and survival, serving as costimulatory molecules. Gene knockout of TLR signaling molecules has been shown to diminish antiviral adaptive immune responses and affect viral clearance in multiple viral infectious animal models. These results have highlighted the critical role of TLRs in the long-term immunological control of viral infection. This review summarizes the expression and function of TLR signaling pathways in T and B cells, focusing on the in vitro and vivo mechanisms and effects of intrinsic TLR signaling in regulating T- and B-cell responses during viral infection. The potential clinical use of TLR-based immune regulatory drugs for viral infectious diseases is also explored.
Collapse
|
10
|
Record Ritchie RD, Salmon SL, Hiles MC, Metzger DW. Lack of immunogenicity of xenogeneic DNA from porcine biomaterials. Surg Open Sci 2022; 10:83-90. [PMID: 36039075 PMCID: PMC9418979 DOI: 10.1016/j.sopen.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/27/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Affiliation(s)
- Rae D. Record Ritchie
- Cook Biotech, Inc, 1425 Innovation Place, West Lafayette, IN, USA 47906
- Corresponding author at: 1425 Innovation Place, West Lafayette, IN 47906. Tel.: + 1-765-497-3355; fax: + 1-765-497-2361.
| | - Sharon L. Salmon
- Department of Immunology and Microbial Disease, Albany Medical College, 47 New Scotland Ave, MC-151, Albany, NY, USA 12208-3479
| | - Michael C. Hiles
- Cook Biotech, Inc, 1425 Innovation Place, West Lafayette, IN, USA 47906
| | - Dennis W. Metzger
- Department of Immunology and Microbial Disease, Albany Medical College, 47 New Scotland Ave, MC-151, Albany, NY, USA 12208-3479
| |
Collapse
|
11
|
Kim JY, Stevens P, Karpurapu M, Lee H, Englert JA, Yan P, Lee TJ, Pabla N, Pietrzak M, Park GY, Christman JW, Chung S. Targeting ETosis by miR-155 inhibition mitigates mixed granulocytic asthmatic lung inflammation. Front Immunol 2022; 13:943554. [PMID: 35958610 PMCID: PMC9360579 DOI: 10.3389/fimmu.2022.943554] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Asthma is phenotypically heterogeneous with several distinctive pathological mechanistic pathways. Previous studies indicate that neutrophilic asthma has a poor response to standard asthma treatments comprising inhaled corticosteroids. Therefore, it is important to identify critical factors that contribute to increased numbers of neutrophils in asthma patients whose symptoms are poorly controlled by conventional therapy. Leukocytes release chromatin fibers, referred to as extracellular traps (ETs) consisting of double-stranded (ds) DNA, histones, and granule contents. Excessive components of ETs contribute to the pathophysiology of asthma; however, it is unclear how ETs drive asthma phenotypes and whether they could be a potential therapeutic target. We employed a mouse model of severe asthma that recapitulates the intricate immune responses of neutrophilic and eosinophilic airway inflammation identified in patients with severe asthma. We used both a pharmacologic approach using miR-155 inhibitor-laden exosomes and genetic approaches using miR-155 knockout mice. Our data show that ETs are present in the bronchoalveolar lavage fluid of patients with mild asthma subjected to experimental subsegmental bronchoprovocation to an allergen and a severe asthma mouse model, which resembles the complex immune responses identified in severe human asthma. Furthermore, we show that miR-155 contributes to the extracellular release of dsDNA, which exacerbates allergic lung inflammation, and the inhibition of miR-155 results in therapeutic benefit in severe asthma mice. Our findings show that targeting dsDNA release represents an attractive therapeutic target for mitigating neutrophilic asthma phenotype, which is clinically refractory to standard care.
Collapse
Affiliation(s)
- Ji Young Kim
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Patrick Stevens
- Comprehensive Cancer Center, Biomedical Informatics Shared Resources, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Manjula Karpurapu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Hyunwook Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Joshua A. Englert
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Pearlly Yan
- Comprehensive Cancer Center, Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Tae Jin Lee
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Navjot Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Maciej Pietrzak
- Comprehensive Cancer Center, Biomedical Informatics Shared Resources, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Gye Young Park
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - John W. Christman
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Sangwoon Chung
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| |
Collapse
|
12
|
Challagundla N, Saha B, Agrawal-Rajput R. Insights into inflammasome regulation: cellular, molecular, and pathogenic control of inflammasome activation. Immunol Res 2022; 70:578-606. [PMID: 35610534 DOI: 10.1007/s12026-022-09286-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
Abstract
Maintenance of immune homeostasis is an intricate process wherein inflammasomes play a pivotal role by contributing to innate and adaptive immune responses. Inflammasomes are ensembles of adaptor proteins that can trigger a signal following innate sensing of pathogens or non-pathogens eventuating in the inductions of IL-1β and IL-18. These inflammatory cytokines substantially influence the antigen-presenting cell's costimulatory functions and T helper cell differentiation, contributing to adaptive immunity. As acute and chronic disease conditions may accompany parallel tissue damage, we analyze the critical role of extracellular factors such as cytokines, amyloids, cholesterol crystals, etc., intracellular metabolites, and signaling molecules regulating inflammasome activation/inhibition. We develop an operative framework for inflammasome function and regulation by host cell factors and pathogens. While inflammasomes influence the innate and adaptive immune components' interplay modulating the anti-pathogen adaptive immune response, pathogens may target inflammasome inhibition as a survival strategy. As trapped between health and diseases, inflammasomes serve as promising therapeutic targets and their modus operandi serves as a scientific rationale for devising better therapeutic strategies.
Collapse
Affiliation(s)
- Naveen Challagundla
- Immunology lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382007, India
| | - Bhaskar Saha
- National Centre for Cell Science, Lab-5, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Reena Agrawal-Rajput
- Immunology lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382007, India.
| |
Collapse
|
13
|
Damasceno LEA, Cebinelli GCM, Fernandes MF, Nascimento DC, Públio GA, Vinolo MAR, Oliveira SC, Sparwasser T, Cunha TM, Cunha FQ, Alves-Filho JC. STING is an intrinsic checkpoint inhibitor that restrains the T H17 cell pathogenic program. Cell Rep 2022; 39:110838. [PMID: 35613599 PMCID: PMC9188824 DOI: 10.1016/j.celrep.2022.110838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 03/02/2022] [Accepted: 04/28/2022] [Indexed: 12/01/2022] Open
Abstract
External and intrinsic factors regulate the transcriptional profile of T helper 17 (TH17) cells, thereby affecting their pathogenic potential and revealing their context-dependent plasticity. The stimulator of interferon genes (STING), a component of the intracellular DNA-sensing pathway, triggers immune responses but remains largely unexplored in T cells. Here, we describe an intrinsic role of STING in limiting the TH17 cell pathogenic program. We demonstrate that non-pathogenic TH17 cells express higher levels of STING than those activated under pathogenic conditions. Activation of STING induces interleukin-10 (IL-10) production in TH17 cells, decreasing IL-17A and IL-23R expression in a type I interferon (IFN)-independent manner. Mechanistically, STING-induced IL-10 production partially requires aryl hydrocarbon receptor (AhR) signaling, while the decrease of IL-17A expression occurs due to a reduction of Rorγt transcriptional activity. Our findings reveal a regulatory function of STING in the TH17 cell activation program, proposing it as a valuable target to limit TH17-cell-mediated inflammation. TH17 cells display a spectrum of pathogenic states depending on environmental and intrinsic cues. Damasceno et al. demonstrate that STING activation induces a non-pathogenic TH17 profile. Mechanistically, STING impairs Rorγt-mediated Il17a transcription, thereby reducing IL-17A production. Besides that, STING activation promotes IL-10 expression through AhR signaling pathway.
Collapse
Affiliation(s)
- Luis Eduardo Alves Damasceno
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Guilherme Cesar Martelossi Cebinelli
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Mariane Font Fernandes
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Daniele Carvalho Nascimento
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Gabriel Azevedo Públio
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Marco Aurélio Ramirez Vinolo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Sergio Costa Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University, Mainz 55131, Germany
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Fernando Queiroz Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - José Carlos Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil.
| |
Collapse
|
14
|
New-Onset Acute Kidney Disease Post COVID-19 Vaccination. Vaccines (Basel) 2022; 10:vaccines10050742. [PMID: 35632497 PMCID: PMC9147880 DOI: 10.3390/vaccines10050742] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 02/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused an exceptional setback to the global economy and health. Vaccination is one of the most effective interventions to markedly decrease severe illness and death from COVID-19. In recent years, there have been increasingly more reports of new acute kidney injury (AKI) after COVID-19 vaccination. Podocyte injury, IgA nephropathy, vasculitis, tubulointerstitial injury, and thrombotic microangiopathy appear to be the main pathological phenotypes. Nonetheless, whether the link between the COVID-19 vaccine and acute kidney disease (AKD) is causal or coincidental remains to be verified. Here, we generalize some hypotheses for the emergence of AKD and its pathogenesis in response to certain COVID-19 vaccines. In fact, the enormous benefits of mass vaccination against COVID-19 in preventing COVID-19 morbidity and mortality cannot be denied. The purpose of this review is to assist in the clinical assessment and management of AKD following COVID-19 vaccination.
Collapse
|
15
|
Virtanen T. Inhalant Mammal-Derived Lipocalin Allergens and the Innate Immunity. FRONTIERS IN ALLERGY 2022; 2:824736. [PMID: 35387007 PMCID: PMC8974866 DOI: 10.3389/falgy.2021.824736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/30/2021] [Indexed: 12/03/2022] Open
Abstract
A major part of important mammalian respiratory allergens belongs to the lipocalin family of proteins. By this time, 19 respiratory mammalian lipocalin allergens have been registered in the WHO/IUIS Allergen Nomenclature Database. Originally, lipocalins, small extracellular proteins (molecular mass ca. 20 kDa), were characterized as transport proteins but they are currently known to exert a variety of biological functions. The three-dimensional structure of lipocalins is well-preserved, and lipocalin allergens can exhibit high amino acid identities, in several cases more than 50%. Lipocalins contain an internal ligand-binding site where they can harbor small principally hydrophobic molecules. Another characteristic feature is their capacity to bind to specific cell-surface receptors. In all, the physicochemical properties of lipocalin allergens do not offer any straightforward explanations for their allergenicity. Allergic sensitization begins at epithelial barriers where diverse insults through pattern recognition receptors awaken innate immunity. This front-line response is manifested by epithelial barrier-associated cytokines which together with other components of immunity can initiate the sensitization process. In the following, the crucial factor in allergic sensitization is interleukin (IL)-4 which is needed for stabilizing and promoting the type 2 immune response. The source for IL-4 has been searched widely. Candidates for it may be non-professional antigen-presenting cells, such as basophils or mast cells, as well as CD4+ T cells. The synthesis of IL-4 by CD4+ T cells requires T cell receptor engagement, i.e., the recognition of allergen peptides, which also provides the specificity for sensitization. Lipocalin and innate immunity-associated cell-surface receptors are implicated in facilitating the access of lipocalin allergens into the immune system. However, the significance of this for allergic sensitization is unclear, as the recognition by these receptors has been found to produce conflicting results. As to potential adjuvants associated with mammalian lipocalin allergens, the hydrophobic ligands transported by lipocalins have not been reported to enhance sensitization while it is justified to suppose that lipopolysaccharide plays a role in it. Taken together, type 2 immunity to lipocalin allergens appears to be a harmful immune response resulting from a combination of signals involving both the innate and adaptive immunities.
Collapse
Affiliation(s)
- Tuomas Virtanen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
16
|
Kobayashi S, Fugo K, Yamazaki K, Terawaki H. Minimal change disease soon after Pfizer-BioNTech COVID-19 vaccination. Clin Kidney J 2021; 14:2606-2607. [PMID: 34938534 PMCID: PMC8499758 DOI: 10.1093/ckj/sfab156] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 08/09/2021] [Indexed: 11/14/2022] Open
Abstract
ABSTRACT
We report on the onset of minimal change disease (MCD) presenting with anasarca after a second dose of the messenger RNA (mRNA)-based Pfizer-BioNTech vaccine against coronavirus disease 2019 (COVID-19). A 75-year-old previously healthy male was admitted with rapidly progressive anasarca and proteinuria of 7.7 g/day following the second dose. A kidney biopsy revealed MCD with nephrotic syndrome. He was treated with intravenous methylprednisolone followed by prednisolone, leading to complete remission after 35 days in the hospital. Since definite causality between the vaccine and MCD remains unclear, awareness of this potential adverse effect of mRNA vaccines is important to determine its true incidence and frequency.
Collapse
Affiliation(s)
| | - Kazunori Fugo
- Department of Pathology, Teikyo University Chiba Medical Center, Chiba, Japan
| | - Kazuto Yamazaki
- Department of Pathology, Teikyo University Chiba Medical Center, Chiba, Japan
| | - Hiroyuki Terawaki
- Division of Nephrology, Department of Internal Medicine, Teikyo University Chiba Medical Center, Chiba, Japan
| |
Collapse
|
17
|
Wen Z, Xu L, Xu W, Xiong S. Retinoic Acid Receptor-Related Orphan Nuclear Receptor γt Licenses the Differentiation and Function of a Unique Subset of Follicular Helper T Cells in Response to Immunogenic Self-DNA in Systemic Lupus Erythematosus. Arthritis Rheumatol 2021; 73:1489-1500. [PMID: 33559400 DOI: 10.1002/art.41687] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Accumulating studies have identified self-DNA as driving IgG anti-double-stranded DNA (anti-dsDNA) in lupus, though the underpinning mechanisms of this process remain largely undefined. Here, we explored the activity of transcription factor retinoic acid receptor-related orphan nuclear receptor γt (RORγt) in the differentiation and function of self-DNA-specific follicular helper T (Tfh) cells in lupus. METHODS B6, TCRα-/- , CD4-/- , RORγtfl/fl CD4Cre, RORγt+/+ CD4Cre, Bcl-6fl/fl CD4Cre, Bcl-6+/+ CD4Cre, IL-17-/- , and ICOS-/- mice were immunized with normal self-DNA, immunogenic self-DNA, and pathogen DNA to induce the production of Tfh cells and IgG anti-dsDNA. Tfh cells with or without interleukin-17 (IL-17) were evaluated for their role in supporting the generation of IgG. NSG mice were reconstituted with immune cells and circulating DNA from human subjects for translational studies. IL-17-positive Tfh cells were analyzed for their correlation with IgG anti-dsDNA levels as well as their response to circulating self-DNA in lupus patients. RESULTS Unlike normal self-DNA, immunogenic self-DNA and pathogen DNA efficiently induced IgG responses. Immunogenic self-DNA induced IgG in a CD4+ T cell-dependent manner, which was abrogated by RORγt deficiency. In contrast, RORγt was not required for the generation of pathogen DNA-induced IgG. Further analyses identified RORγt as essential for the differentiation and function of Tfh cells in response to immunogenic self-DNA, assigning IL-17 as a feature cytokine. These IL-17-positive Tfh cells functioned independent of inducible costimulator (ICOS), critically supporting IgG generation. Targeting immunogenic self-DNA-specific Tfh cells by RORγ knockdown and IL-17 blockade ameliorated IgG response and lupus nephritis in a humanized mouse model. The presence of IL-17-positive Tfh cells was associated with IgG anti-dsDNA levels and were expanded by circulating immunogenic self-DNA in lupus patients. CONCLUSION Immunogenic self-DNA instructs ICOS-dispensable IL-17-positive Tfh cells via RORγt to produce an IgG anti-dsDNA response. As such, IL-17-positive Tfh cells are a promising therapeutic target for lupus patients.
Collapse
Affiliation(s)
| | - Lin Xu
- Soochow University, Suzhou, China
| | - Wei Xu
- Soochow University, Suzhou, China
| | | |
Collapse
|
18
|
STING Gain-of-Function Disrupts Lymph Node Organogenesis and Innate Lymphoid Cell Development in Mice. Cell Rep 2021; 31:107771. [PMID: 32553167 DOI: 10.1016/j.celrep.2020.107771] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 01/31/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022] Open
Abstract
STING gain-of-function causes autoimmunity and immunodeficiency in mice and STING-associated vasculopathy with onset in infancy (SAVI) in humans. Here, we report that STING gain-of-function in mice prevents development of lymph nodes and Peyer's patches. We show that the absence of secondary lymphoid organs is associated with diminished numbers of innate lymphoid cells (ILCs), including lymphoid tissue inducer (LTi) cells. Although wild-type (WT) α4β7+ progenitors differentiate efficiently into LTi cells, STING gain-of-function progenitors do not. Furthermore, STING gain-of-function impairs development of all types of ILCs. Patients with STING gain-of-function mutations have fewer ILCs, although they still have lymph nodes. In mice, expression of the STING mutant in RORγT-positive lineages prevents development of lymph nodes and reduces numbers of LTi cells. RORγT lineage-specific expression of STING gain-of-function also causes lung disease. Since RORγT is expressed exclusively in LTi cells during fetal development, our findings suggest that STING gain-of-function prevents lymph node organogenesis by reducing LTi cell numbers in mice.
Collapse
|
19
|
Imanishi T, Unno M, Kobayashi W, Yoneda N, Akira S, Saito T. mTORC1 Signaling Controls TLR2-Mediated T-Cell Activation by Inducing TIRAP Expression. Cell Rep 2021; 32:107911. [PMID: 32698010 DOI: 10.1016/j.celrep.2020.107911] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/07/2020] [Accepted: 06/24/2020] [Indexed: 01/21/2023] Open
Abstract
Effector, but not naïve, T cells are activated by toll-like receptor-2 (TLR2) stimulation, leading to cytokine production and proliferation. We found that the differential response is attributable to the lack of expression of the adaptor protein TIRAP in naive T cells. TIRAP expression is induced upon T-cell receptor (TCR) stimulation and sustained by strong interleukin-2 (IL-2) signals. Expression of TIRAP requires TCR- and IL-2-induced mTORC1 activation. TLR2 stimulation induced the activation of nuclear factor κB (NF-κB) and ERK, leading to much higher production of interferon-γ (IFN-γ) by T helper 1 (Th1) cells cultured in a high concentration of IL-2 than by those cultured in a low concentration of IL-2. In contrast, TLR2 stimulation induces mTORC1 activation through TIRAP, which is essential for TLR2-mediated IFN-γ production. These data demonstrate that the mTORC1 signal confers the response to TLR2 signaling by inducing TIRAP expression and that the TIRAP-mTORC1 axis is critical for TLR2-mediated IFN-γ production by effector T cells.
Collapse
Affiliation(s)
- Takayuki Imanishi
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan.
| | - Midori Unno
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan
| | - Wakana Kobayashi
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan
| | - Natsumi Yoneda
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Takashi Saito
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan; Laboratory for Cell Signaling, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
20
|
Induction of Chicken Host Defense Peptides within Disease-Resistant and -Susceptible Lines. Genes (Basel) 2020; 11:genes11101195. [PMID: 33066561 PMCID: PMC7602260 DOI: 10.3390/genes11101195] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 11/17/2022] Open
Abstract
Host defense peptides (HDPs) are multifunctional immune molecules that respond to bacterial and viral pathogens. In the present study, bone marrow-derived cells (BMCs) and chicken embryonic fibroblasts (CEFs) were cultured from a Leghorn line (Ghs6) and Fayoumi line (M15.2), which are inbred chicken lines relatively susceptible and resistant to various diseases, respectively. The cells were treated by lipopolysaccharide (LPS) or polyinosinic-polycytidylic acid (poly(I:C)) and, subsequently, mRNA expression of 20 chicken HDPs was analyzed before and after the stimulation. At homeostasis, many genes differed between the chicken lines, with the Fayoumi line having significantly higher expression (p < 0.05) than the Leghorn line: AvBD1, 2, 3, 4, 6, and 7 in BMCs; CATH1, CATH3, and GNLY in CEFs; and AvDB5, 8, 9, 10, 11, 12, 13 in both BMCs and CEFs. After LPS treatment, the expression of AvBD1, 2, 3, 4, 5, 9, 12, CATH1, and CATHB1 was significantly upregulated in BMCs, but no genes changed expression in CEFs. After poly(I:C) treatment, AvBD2, 11, 12, 13, CATHB1 and LEAP2 increased in both cell types; CATH2 only increased in BMCs; and AvBD3, 6, 9, 14, CATH1, CATH3, and GNLY only increased in CEFs. In addition, AvBD7, AvBD14, CATH1, CATH2, GNLY, and LEAP2 showed line-specific expression dependent upon cell type (BMC and CEF) and stimulant (LPS and poly(I:C)). The characterization of mRNA expression patterns of chicken HDPs in the present study suggests that their functions may be associated with multiple types of disease resistance in chickens.
Collapse
|
21
|
Miki H, Pei H, Gracias DT, Linden J, Croft M. Clearance of apoptotic cells by lung alveolar macrophages prevents development of house dust mite-induced asthmatic lung inflammation. J Allergy Clin Immunol 2020; 147:1087-1092.e3. [PMID: 33065121 DOI: 10.1016/j.jaci.2020.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 09/25/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Poor clearance of apoptotic cells has been suggested to contribute to severe asthma, but whether uptake of apoptotic cells by lung phagocytes might dampen house dust mite (HDM)-induced lung inflammation has not been shown. OBJECTIVES This study investigated whether apoptotic cell engulfment in the murine lung impacts the development of allergen-induced asthmatic airway inflammation and which immune modulating mechanisms were activated. METHODS Apoptotic cells were infused into the lungs of mice challenged with HDM allergen and lung inflammation, expression of suppressive molecules, and induction of regulatory T cells were monitored. Additionally, an adenosine receptor agonist was tested to study the mechanism of suppression elicited by apoptotic cells. RESULTS Apoptotic cell uptake by lung alveolar macrophages suppressed HDM-driven allergic asthma. This was associated with promoting the regulatory T cell-inducing molecule retinoic acid, inhibiting inflammatory cytokine production, and making macrophages more susceptible to receiving suppressive signals from adenosine. Correspondingly, adenosine receptor agonist treatment also limited HDM-driven allergic airway inflammation through an action on alveolar macrophages. CONCLUSIONS These data provide insight into the mechanisms by which lung macrophages dampen allergen-induced airway inflammation. They suggest that targeting lung macrophages to increase their phagocytic capacity, enhance their ability to make retinoic acid, dampen their capacity to make inflammatory cytokines, and increase their responsiveness to adenosine, could be useful to suppress allergic responses.
Collapse
Affiliation(s)
- Haruka Miki
- Center of Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, Calif
| | - Hong Pei
- Center of Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, Calif
| | - Donald Tom Gracias
- Center of Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, Calif
| | - Joel Linden
- Center of Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, Calif
| | - Michael Croft
- Center of Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, Calif; Department of Medicine, University of California San Diego, La Jolla, Calif.
| |
Collapse
|
22
|
Harris E, Elmer JJ. Optimization of electroporation and other non-viral gene delivery strategies for T cells. Biotechnol Prog 2020; 37:e3066. [PMID: 32808434 DOI: 10.1002/btpr.3066] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 12/21/2022]
Abstract
CAR-T therapy is a particularly effective treatment for some types of cancer that uses retroviruses to deliver the gene for a chimeric antigen receptor (CAR) to a patient's T cells ex vivo. The CAR enables the T cells to bind and eradicate cells with a specific surface marker (e.g., CD19+ B cells) after they are transfused back into the patient. This treatment was proven to be particularly effective in treating non-Hodgkin's lymphoma (NHL) and acute lymphoblastic leukemia (ALL), but the current CAR-T cell manufacturing process has a few significant drawbacks. For example, while lentiviral and gammaretroviral transduction are both relatively effective, the process of producing viral vectors is time-consuming and costly. Additionally, patients must undergo follow up appointments for several years to monitor them for any unanticipated side effects associated with the virus. Therefore, several studies have endeavored to find alternative non-viral gene delivery methods that are less expensive, more precise, simple, and safe. This review focuses on the current state of the most promising non-viral gene delivery techniques, including electroporation and transfection with cationic polymers or lipids.
Collapse
Affiliation(s)
- Emily Harris
- Villanova University, Department of Chemical & Biological Engineering, Villanova, Pennsylvania, USA
| | - Jacob J Elmer
- Villanova University, Department of Chemical & Biological Engineering, Villanova, Pennsylvania, USA
| |
Collapse
|
23
|
Stögerer T, Stäger S. Innate Immune Sensing by Cells of the Adaptive Immune System. Front Immunol 2020; 11:1081. [PMID: 32547564 PMCID: PMC7274159 DOI: 10.3389/fimmu.2020.01081] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/05/2020] [Indexed: 01/05/2023] Open
Abstract
Sensing of microbes or of danger signals has mainly been attributed to myeloid innate immune cells. However, T and B cells also express functional pattern recognition receptors (PRRs). In these cells, PRRs mediate signaling cascades that result in different functions depending on the cell's activation and/or differentiation status, on the environment, and on the ligand/agonist. Some of these functions are beneficial for the host; however, some are detrimental and are exploited by pathogens to establish persistent infections. In this review, we summarize the available literature on innate immune sensing by cells of the adaptive immune system and discuss possible implications for chronic infections.
Collapse
Affiliation(s)
- Tanja Stögerer
- INRS Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| | - Simona Stäger
- INRS Centre Armand-Frappier Santé Biotechnologie, Laval, QC, Canada
| |
Collapse
|
24
|
Wan D, Jiang W, Hao J. Research Advances in How the cGAS-STING Pathway Controls the Cellular Inflammatory Response. Front Immunol 2020; 11:615. [PMID: 32411126 PMCID: PMC7198750 DOI: 10.3389/fimmu.2020.00615] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/17/2020] [Indexed: 12/19/2022] Open
Abstract
Double-stranded DNA (dsDNA) sensor cyclic-GMP-AMP synthase (cGAS) along with the downstream stimulator of interferon genes (STING) acting as essential immune-surveillance mediators have become hot topics of research. The intrinsic function of the cGAS-STING pathway facilitates type-I interferon (IFN) inflammatory signaling responses and other cellular processes such as autophagy, cell survival, senescence. cGAS-STING pathway interplays with other innate immune pathways, by which it participates in regulating infection, inflammatory disease, and cancer. The therapeutic approaches targeting this pathway show promise for future translation into clinical applications. Here, we present a review of the important previous works and recent advances regarding the cGAS-STING pathway, and provide a comprehensive understanding of the modulatory pattern of the cGAS-STING pathway under multifarious pathologic states.
Collapse
Affiliation(s)
- Dongshan Wan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Jiang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Junwei Hao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
25
|
Imanishi T, Saito T. T Cell Co-stimulation and Functional Modulation by Innate Signals. Trends Immunol 2020; 41:200-212. [PMID: 32035763 DOI: 10.1016/j.it.2020.01.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 01/10/2020] [Accepted: 01/10/2020] [Indexed: 12/22/2022]
Abstract
Pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs), NOD-like receptors (NLRs), and RIG-I-like receptors (RLRs), play a pivotal role in the initiation of innate immune responses. Certain PRRs are also expressed by CD4+ and CD8+ T cells, where they function to provide co-stimulatory signals for their activation and differentiation. Recently, stimulator of interferon genes (STING) was found to be highly expressed in CD4+ and CD8+ T cells and to modulate T cell function. STING signaling inhibits cell growth and stimulates type I interferon (IFN-I) responses in T cells through reciprocal regulation between T cell receptor (TCR) and STING signals. Here, we propose a model whereby innate signals by TLRs and STING regulate TCR signals and T cell functions.
Collapse
Affiliation(s)
- Takayuki Imanishi
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan.
| | - Takashi Saito
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan; Laboratory for Cell Signaling, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
26
|
Ozasa K, Temizoz B, Kusakabe T, Kobari S, Momota M, Coban C, Ito S, Kobiyama K, Kuroda E, Ishii KJ. Cyclic GMP-AMP Triggers Asthma in an IL-33-Dependent Manner That Is Blocked by Amlexanox, a TBK1 Inhibitor. Front Immunol 2019; 10:2212. [PMID: 31616416 PMCID: PMC6775192 DOI: 10.3389/fimmu.2019.02212] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/02/2019] [Indexed: 01/01/2023] Open
Abstract
Extracellular host-derived DNA, as one of damage associated molecular patterns (DAMPs), is associated with allergic type 2 immune responses. Immune recognition of such DNA generates the second messenger cyclic GMP-AMP (cGAMP) and induces type-2 immune responses; however, its role in allergic diseases, such as asthma, has not been fully elucidated. This study aimed to determine whether cGAMP could induce asthma when used as an adjuvant. We intranasally sensitized mice with cGAMP together with house dust mite antigen (HDM), followed by airway challenge with HDM. We then assessed the levels of eosinophils in the broncho-alveolar lavage fluid (BALF) and serum HDM-specific antibodies. cGAMP promoted HDM specific allergic asthma, characterized by significantly increased HDM specific IgG1 and total IgE in the serum and infiltration of eosinophils in the BALF. cGAMP stimulated lung fibroblast cells to produce IL-33 in vitro, and mice deficient for IL-33 or IL-33 receptor (ST2) failed to develop asthma enhancement by cGAMP. Not only Il-33 -/- mice, but also Sting -/-, Tbk1 -/-, and Irf3 -/- Irf7 -/- mice which lack the cGAMP-mediated innate immune activation failed to increase eosinophils in the BALF than that from wild type mice. Consistently, intranasal and oral administration of amlexanox, a TBK1 inhibitor, decreased cGAMP-induced lung allergic inflammation. Thus, cGAMP functions as a type 2 adjuvant in the lung and can promote allergic asthma in manners that dependent on the intracellular STING/TBK1/IRF3/7 signaling pathway and the resultant intercellular signaling pathway via IL-33 and ST2 might be a novel therapeutic target for allergic asthma.
Collapse
Affiliation(s)
- Koji Ozasa
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Burcu Temizoz
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takato Kusakabe
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Laboratory of Mock-Up Vaccine Project, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Shingo Kobari
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masatoshi Momota
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Mock-Up Vaccine Project, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Cevayir Coban
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Malaria Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shuichi Ito
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kouji Kobiyama
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Etsushi Kuroda
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Mock-Up Vaccine Project, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| |
Collapse
|
27
|
Sterile Lung Inflammation Induced by Silica Exacerbates Mycobacterium tuberculosis Infection via STING-Dependent Type 2 Immunity. Cell Rep 2019; 27:2649-2664.e5. [DOI: 10.1016/j.celrep.2019.04.110] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/17/2019] [Accepted: 04/26/2019] [Indexed: 12/18/2022] Open
|
28
|
Imanishi T, Unno M, Kobayashi W, Yoneda N, Matsuda S, Ikeda K, Hoshii T, Hirao A, Miyake K, Barber GN, Arita M, Ishii KJ, Akira S, Saito T. Reciprocal regulation of STING and TCR signaling by mTORC1 for T-cell activation and function. Life Sci Alliance 2019; 2:2/1/e201800282. [PMID: 30683688 PMCID: PMC6348487 DOI: 10.26508/lsa.201800282] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 12/26/2018] [Accepted: 01/07/2019] [Indexed: 12/12/2022] Open
Abstract
Stimulator of interferon genes (STING) plays a key role in detecting cytosolic DNA and induces type I interferon (IFN-I) responses for host defense against pathogens. Although T cells highly express STING, its physiological role remains unknown. Here, we show that costimulation of T cells with the STING ligand cGAMP and TCR leads to IFN-I production and strongly inhibits T-cell growth. TCR-mediated mTORC1 activation and sustained activation of IRF3 are required for cGAMP-induced IFN-I production, and the mTORC1 activity is partially counteracted by cGAMP, thereby blocking proliferation. This mTORC1 inhibition in response to costimulation depends on IRF3 and IRF7. Effector T cells produce much higher IFN-I levels than innate cells in response to cGAMP. Finally, we demonstrated that STING stimulation in T cells is effective in inducing antitumor responses in vivo. Our studies demonstrate that the outputs of STING and TCR signaling pathways are mutually regulated through mTORC1 to modulate T-cell functions.
Collapse
Affiliation(s)
- Takayuki Imanishi
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Midori Unno
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Wakana Kobayashi
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Natsumi Yoneda
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Satoshi Matsuda
- Department of Cell Signaling, Institute of Biomedical Sciences, Kansai Medical University, Hirakata, Japan
| | - Kazutaka Ikeda
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Japan Agency for Medical Research and Development (AMED)-PRIME, Japan Agency for Medical Research and Development, Tokyo, Japan.,Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Takayuki Hoshii
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Glen N Barber
- Department of Cell Biology and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan.,Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Ken J Ishii
- Laboratory of Vaccine Science, World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Suita, Japan.,Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Takashi Saito
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan .,Laboratory for Cell Signaling, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| |
Collapse
|
29
|
Farahnak S, Chronopoulos J, Martin JG. Nucleic Acid Sensing in Allergic Disorders. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 345:1-33. [PMID: 30904191 DOI: 10.1016/bs.ircmb.2018.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Recent advances indicate that there is crosstalk between allergic disorders and nucleic acid sensing. Triggers that activate inflammatory mechanisms via nucleic acid sensors affect both allergic phenotypes and anti-viral responses, depending on the timing and the order of exposure. Viral respiratory infections, such as those caused by the rhinovirus, influenza, and respiratory syncytial virus, are the most frequent cause of significant asthma exacerbations through effects mediated predominantly by TLR3. However, agonists of other nucleic acid sensors, such as TLR7/8 and TLR9 agonists, may inhibit allergic inflammation and reduce clinical manifestations of disease. The allergic state can predispose the immune system to both exaggerated responses to viral infections or protection from anti-viral inflammatory responses. TH2 cytokines appear to alter the epithelium, leading to defective viral clearance or exaggerated responses to viral infections. However, a TH2 skewed allergic response may be protective against a TH1-dependent inflammatory anti-viral response. This review briefly introduces the receptors involved in nucleic acid sensing, addresses mechanisms by which nucleic acid sensing and allergic responses can counteract one another, and discusses the strategies in experimental settings, both in animal and human studies, to harness the nucleic acid sensing machinery for the intervention of allergic disorders.
Collapse
Affiliation(s)
- Soroor Farahnak
- Meakins Christie Laboratories, Research Institute of the McGill University Health Centre and McGill University, Montreal, QC, Canada
| | - Julia Chronopoulos
- Meakins Christie Laboratories, Research Institute of the McGill University Health Centre and McGill University, Montreal, QC, Canada
| | - James G Martin
- Meakins Christie Laboratories, Research Institute of the McGill University Health Centre and McGill University, Montreal, QC, Canada.
| |
Collapse
|
30
|
TLR Specific Immune Responses against Helminth Infections. J Parasitol Res 2017; 2017:6865789. [PMID: 29225962 PMCID: PMC5684585 DOI: 10.1155/2017/6865789] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/21/2017] [Accepted: 10/03/2017] [Indexed: 01/07/2023] Open
Abstract
Despite marked improvement in the quality of lives across the globe, more than 2 million individuals in socioeconomically disadvantaged environments remain infected by helminth (worm) parasites. Owing to the longevity of the worms and paucity of immunologic controls, these parasites survive for long periods within the bloodstream, lymphatics, and gastrointestinal tract resulting in pathologic conditions such as anemia, cirrhosis, and lymphatic filariasis. Despite infection, an asymptomatic state may be maintained by the host immunoregulatory environment, which involves multiple levels of regulatory cells and cytokines; a breakdown of this regulation is observed in pathological disease. The role of TLR expression and function in relation to intracellular parasites has been documented but limited studies are available for multicellular helminth parasites. In this review, we discuss the unique and shared host effector mechanisms elicited by systemic helminth parasites and their derived products, including the role of TLRs and sphingolipids. Understanding and exploiting the interactions between these parasites and the host regulatory network are likely to highlight new strategies to control both infectious and immunological diseases.
Collapse
|
31
|
Toussaint M, Jackson DJ, Swieboda D, Guedán A, Tsourouktsoglou TD, Ching YM, Radermecker C, Makrinioti H, Aniscenko J, Bartlett NW, Edwards MR, Solari R, Farnir F, Papayannopoulos V, Bureau F, Marichal T, Johnston SL. Host DNA released by NETosis promotes rhinovirus-induced type-2 allergic asthma exacerbation. Nat Med 2017; 23:681-691. [PMID: 28459437 PMCID: PMC5821220 DOI: 10.1038/nm.4332] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/04/2017] [Indexed: 02/06/2023]
Abstract
Respiratory viral infections represent the most common cause of allergic asthma exacerbations. Amplification of the type-2 immune response is strongly implicated in asthma exacerbation, but how virus infection boosts type-2 responses is poorly understood. We report a significant correlation between the release of host double-stranded DNA (dsDNA) following rhinovirus infection and the exacerbation of type-2 allergic inflammation in humans. In a mouse model of allergic airway hypersensitivity, we show that rhinovirus infection triggers dsDNA release associated with the formation of neutrophil extracellular traps (NETs), known as NETosis. We further demonstrate that inhibiting NETosis by blocking neutrophil elastase or by degrading NETs with DNase protects mice from type-2 immunopathology. Furthermore, the injection of mouse genomic DNA alone is sufficient to recapitulate many features of rhinovirus-induced type-2 immune responses and asthma pathology. Thus, NETosis and its associated extracellular dsDNA contribute to the pathogenesis and may represent potential therapeutic targets of rhinovirus-induced asthma exacerbations.
Collapse
Affiliation(s)
- Marie Toussaint
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - David J Jackson
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
- Imperial College Healthcare NHS Trust, London, UK
- Guy's and St Thomas' NHS Trust, London, UK
| | - Dawid Swieboda
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Anabel Guedán
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | | | - Yee Man Ching
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Coraline Radermecker
- Laboratory of Cellular and Molecular Immunology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Heidi Makrinioti
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Julia Aniscenko
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Nathan W Bartlett
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Michael R Edwards
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Roberto Solari
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Frédéric Farnir
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- Fundamental and Applied Research for Animals &Health, University of Liège, Liège, Belgium
| | | | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- WELBIO, Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium
| | - Thomas Marichal
- Laboratory of Cellular and Molecular Immunology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
32
|
Kuroda E, Ishii KJ. Particulates Induce Type-2 Immune Responses. Nihon Eiseigaku Zasshi 2017; 72:27-31. [PMID: 28154357 DOI: 10.1265/jjh.72.27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Particulates are considered to be one of the causative factors for allergic asthma and rhinitis in developed countries. Indeed, particulates and crystals are reported to activate type-2 immunity, which is associated with allergic responses. Thus far, many studies have been carried out to determine how particulates trigger immune responses; however, the detailed mechanisms by which immune responses are triggered are still unknown. One important clue is that particulates have an adjuvant activity and boost immune responses toward type-2 responses. Most adjuvants are considered to activate innate immunity, and then activated innate immune cells stimulate adaptive immunity, which involves T cells and B cells. That is why many scientists believe that particulate adjuvants directly stimulate innate cells through unique sensor proteins such as pattern-recognition receptors. In this review, we will introduce the proposed mechanisms of particulate-induced immune activation from the viewpoint of the activation of innate immune responses and related receptors and sensors.
Collapse
Affiliation(s)
- Etsushi Kuroda
- Laboratory of Vaccine Science, WPI Immunology Frontier Research Center (IFReC), Osaka University
| | | |
Collapse
|
33
|
Noges LE, White J, Cambier JC, Kappler JW, Marrack P. Contamination of DNase Preparations Confounds Analysis of the Role of DNA in Alum-Adjuvanted Vaccines. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:1221-30. [PMID: 27357147 PMCID: PMC4974487 DOI: 10.4049/jimmunol.1501565] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 06/02/2016] [Indexed: 11/19/2022]
Abstract
Aluminum salt (alum) adjuvants have been used for many years as adjuvants for human vaccines because they are safe and effective. Despite its widespread use, the means by which alum acts as an adjuvant remains poorly understood. Recently, it was shown that injected alum is rapidly coated with host chromatin within mice. Experiments suggested that the host DNA in the coating chromatin contributed to alum's adjuvant activity. Some of the experiments used commercially purchased DNase and showed that coinjection of these DNase preparations with alum and Ag reduced the host's immune response to the vaccine. In this study, we report that some commercial DNase preparations are contaminated with proteases. These proteases are responsible for most of the ability of DNase preparations to inhibit alum's adjuvant activity. Nevertheless, DNase somewhat reduces responses to some Ags with alum. The effect of DNase is independent of its ability to cleave DNA, suggesting that alum improves CD4 responses to Ag via a pathway other than host DNA sensing.
Collapse
Affiliation(s)
- Laura E Noges
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; and
| | - Janice White
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206
| | - John C Cambier
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; and
| | - John W Kappler
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; and Howard Hughes Medical Institute, National Jewish Health, Denver, CO 80206
| | - Philippa Marrack
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045; and Howard Hughes Medical Institute, National Jewish Health, Denver, CO 80206
| |
Collapse
|
34
|
Impact of Histone H1 on the Progression of Allergic Rhinitis and Its Suppression by Neutralizing Antibody in Mice. PLoS One 2016; 11:e0153630. [PMID: 27088594 PMCID: PMC4835108 DOI: 10.1371/journal.pone.0153630] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 04/02/2016] [Indexed: 01/10/2023] Open
Abstract
Nuclear antigens are known to trigger off innate and adaptive immune responses. Recent studies have found that the complex of nucleic acids and core histones that are derived from damaged cells may regulate allergic responses. However, no fundamental study has been performed concerning the role of linker histone H1 in mast cell-mediated type I hyperreactivity. In this study, we explored the impact of histone H1 on mast cell-mediated allergic responses both in vitro and in vivo. In the course of a bona-fide experimental allergen sensitization model upon co-injection with alum adjuvant, ovalbumin (OVA), but not PBS, induced elevated levels of circulating histone H1. Intranasal challenge with histone H1 to OVA/alum- (but not PBS/alum)-sensitized mice induced significantly severer symptoms of allergic rhinitis than those in mice sensitized and challenged with OVA. A monoclonal antibody against histone H1 not only suppressed mast cell degranulation, but also ameliorated OVA-induced nasal hyperreactivity and IgE-mediated passive cutaneous anaphylaxis. Our present data suggest that nuclear histone H1 represents an alarmin-like endogenous mediator acting on mast cells, and that its blockage has a therapeutic potential for mast cell-mediated type I hyperreactivity.
Collapse
|
35
|
Regulation of IL-4 Expression in Immunity and Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 941:31-77. [PMID: 27734408 DOI: 10.1007/978-94-024-0921-5_3] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IL-4 was first identified as a T cell-derived growth factor for B cells. Studies over the past several decades have markedly expanded our understanding of its cellular sources and function. In addition to T cells, IL-4 is produced by innate lymphocytes, such as NTK cells, and myeloid cells, such as basophils and mast cells. It is a signature cytokine of type 2 immune response but also has a nonimmune function. Its expression is tightly regulated at several levels, including signaling pathways, transcription factors, epigenetic modifications, microRNA, and long noncoding RNA. This chapter will review in detail the molecular mechanism regulating the cell type-specific expression of IL-4 in physiological and pathological type 2 immune responses.
Collapse
|
36
|
Abstract
In the two decades since their initial discovery, DNA vaccines technologies have come a long way. Unfortunately, when applied to human subjects inadequate immunogenicity is still the biggest challenge for practical DNA vaccine use. Many different strategies have been tested in preclinical models to address this problem, including novel plasmid vectors and codon optimization to enhance antigen expression, new gene transfection systems or electroporation to increase delivery efficiency, protein or live virus vector boosting regimens to maximise immune stimulation, and formulation of DNA vaccines with traditional or molecular adjuvants. Better understanding of the mechanisms of action of DNA vaccines has also enabled better use of the intrinsic host response to DNA to improve vaccine immunogenicity. This review summarizes recent advances in DNA vaccine technologies and related intracellular events and how these might impact on future directions of DNA vaccine development.
Collapse
Affiliation(s)
- Lei Li
- a Vaxine Pty Ltd, Bedford Park , Adelaide , Australia.,b Department of Diabetes and Endocrinology , Flinders University, Flinders Medical Centre , Adelaide , SA , Australia
| | - Nikolai Petrovsky
- a Vaxine Pty Ltd, Bedford Park , Adelaide , Australia.,b Department of Diabetes and Endocrinology , Flinders University, Flinders Medical Centre , Adelaide , SA , Australia
| |
Collapse
|
37
|
Tindemans I, Serafini N, Di Santo JP, Hendriks RW. GATA-3 function in innate and adaptive immunity. Immunity 2014; 41:191-206. [PMID: 25148023 DOI: 10.1016/j.immuni.2014.06.006] [Citation(s) in RCA: 199] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/19/2014] [Indexed: 02/07/2023]
Abstract
The zinc-finger transcription factor GATA-3 has received much attention as a master regulator of T helper 2 (Th2) cell differentiation, during which it controls interleukin-4 (IL-4), IL-5, and IL-13 expression. More recently, GATA-3 was shown to contribute to type 2 immunity through regulation of group 2 innate lymphoid cell (ILC2) development and function. Furthermore, during thymopoiesis, GATA-3 represses B cell potential in early T cell precursors, activates TCR signaling in pre-T cells, and promotes the CD4(+) T cell lineage after positive selection. GATA-3 also functions outside the thymus in hematopoietic stem cells, regulatory T cells, CD8(+) T cells, thymic natural killer cells, and ILC precursors. Here we discuss the varied functions of GATA-3 in innate and adaptive immune cells, with emphasis on its activity in T cells and ILCs, and examine the mechanistic basis for the dose-dependent, developmental-stage- and cell-lineage-specific activity of this transcription factor.
Collapse
Affiliation(s)
- Irma Tindemans
- Department of Pulmonary Medicine, Erasmus MC, 3000 CA Rotterdam, the Netherlands
| | - Nicolas Serafini
- Innate Immunity Unit, Institut Pasteur, 75724 Paris, France; INSERM U668, 75724 Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, 75724 Paris, France; INSERM U668, 75724 Paris, France
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, 3000 CA Rotterdam, the Netherlands.
| |
Collapse
|