501
|
Blocking of the IL-33/ST2 Signaling Axis by a Single-Chain Antibody Variable Fragment (scFv) Specific to IL-33 with a Defined Epitope. Int J Mol Sci 2020; 21:ijms21186953. [PMID: 32971846 PMCID: PMC7554688 DOI: 10.3390/ijms21186953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 12/03/2022] Open
Abstract
Interleukin 33 (IL-33) is an IL-1 family cytokine that plays a central role in immune system by regulating and initiating inflammatory responses. The binding of IL-33 to the suppressor of tumorigenicity 2 (ST2) receptor induces mitogen-activated protein kinases (MAPK) and nuclear factor κB (NF-κB) pathways, thereby leading to inflammatory cytokines production in type 2 helper T cells and type 2 innate lymphoid cells. To develop an antibody specific to IL-33 with a defined epitope, we characterized a single-chain antibody variable fragments (scFvs) clone specific to IL-33, C2_2E12, which was selected from a human synthetic library of scFvs using phage display. Affinity (Kd) of C2_2E12 was determined to be 38 nM using enzyme-linked immunosorbent assay. C2_2E12 did not show cross-reactivity toward other interleukin cytokines, including closely related IL-1 family cytokines and unrelated proteins. Mutational scanning analysis revealed that the epitope of IL-33 consisted of residues 149–158 with key residues being L150 and K151 of IL-33. Structural modeling suggested that L150 and K151 residues are important for the interaction of IL-33 with C2_2E12, implicating that C2_2E12 could block the binding of ST2 to IL-33. Pull-down and in-cell assays supported that C2_2E12 can inhibit the IL-33/ST2 signaling axis. These results suggest that the scFv clone characterized here can function as a neutralizing antibody.
Collapse
|
502
|
Oshimori N. Cancer stem cells and their niche in the progression of squamous cell carcinoma. Cancer Sci 2020; 111:3985-3992. [PMID: 32888236 PMCID: PMC7648029 DOI: 10.1111/cas.14639] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/27/2022] Open
Abstract
Most cancers harbor a small population of highly tumorigenic cells known as cancer stem cells (CSCs). Because of their stem cell-like properties and resistance to conventional therapies, CSCs are considered to be a rational target for curable cancer treatment. However, despite recent advances in CSC research, CSC-targeted therapies are not as successful as was initially hoped. The proliferative, invasive, and drug-resistant properties of CSCs are regulated by the tumor microenvironment associated with them, the so-called CSC niche. Thus, targeting tumor-promoting cellular crosstalk between CSCs and their niches is an attractive avenue for developing durable therapies. Using mouse models of squamous cell carcinoma (SCC), we have demonstrated that tumor cells responding to transforming growth factor β (TGF-β) function as drug-resistant CSCs. The gene expression signature of TGF-β-responding tumor cells has accelerated the identification of novel pathways that drive invasive tumor progression. Moreover, by focusing on the cytokine milieu and macrophages in the proximity of TGF-β-responding tumor cells, we recently uncovered the molecular basis of a CSC-niche interaction that emerges during early tumor development. This review article summarizes the specialized tumor microenvironment associated with CSCs and discusses mechanisms by which malignant properties of CSCs are maintained and promoted.
Collapse
Affiliation(s)
- Naoki Oshimori
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR, USA.,Department of Dermatology, Oregon Health and Science University, Portland, OR, USA.,Department of Otolaryngology, Head & Neck Surgery, Oregon Health and Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
503
|
Cuffaro B, Assohoun ALW, Boutillier D, Súkeníková L, Desramaut J, Boudebbouze S, Salomé-Desnoulez S, Hrdý J, Waligora-Dupriet AJ, Maguin E, Grangette C. In Vitro Characterization of Gut Microbiota-Derived Commensal Strains: Selection of Parabacteroides distasonis Strains Alleviating TNBS-Induced Colitis in Mice. Cells 2020; 9:cells9092104. [PMID: 32947881 PMCID: PMC7565435 DOI: 10.3390/cells9092104] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022] Open
Abstract
Alterations in the gut microbiota composition and diversity seem to play a role in the development of chronic diseases, including inflammatory bowel disease (IBD), leading to gut barrier disruption and induction of proinflammatory immune responses. This opens the door for the use of novel health-promoting bacteria. We selected five Parabacteroides distasonis strains isolated from human adult and neonates gut microbiota. We evaluated in vitro their immunomodulation capacities and their ability to reinforce the gut barrier and characterized in vivo their protective effects in an acute murine model of colitis. The in vitro beneficial activities were highly strain dependent: two strains exhibited a potent anti-inflammatory potential and restored the gut barrier while a third strain reinstated the epithelial barrier. While their survival to in vitro gastric conditions was variable, the levels of P. distasonis DNA were higher in the stools of bacteria-treated animals. The strains that were positively scored in vitro displayed a strong ability to rescue mice from colitis. We further showed that two strains primed dendritic cells to induce regulatory T lymphocytes from naïve CD4+ T cells. This study provides better insights on the functionality of commensal bacteria and crucial clues to design live biotherapeutics able to target inflammatory chronic diseases such as IBD.
Collapse
Affiliation(s)
- Bernardo Cuffaro
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Centre d’Infection et d’Immunité de Lille, 59000 Lille, France; (B.C.); (D.B.); (J.D.)
- Institut Micalis, MIHA Team, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France; (A.L.W.A.); (S.B.)
| | - Aka L. W. Assohoun
- Institut Micalis, MIHA Team, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France; (A.L.W.A.); (S.B.)
- Laboratoire de Biotechnologie et Microbiologie des Aliments, UFR en Sciences et Technologies des Aliments, Université Nangui Abrogoua, Abidjan 00225, Côte d’Ivoire
| | - Denise Boutillier
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Centre d’Infection et d’Immunité de Lille, 59000 Lille, France; (B.C.); (D.B.); (J.D.)
| | - Lenka Súkeníková
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, 121 08 Prague, Czech Republic; (L.S.); (J.H.)
| | - Jérémy Desramaut
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Centre d’Infection et d’Immunité de Lille, 59000 Lille, France; (B.C.); (D.B.); (J.D.)
| | - Samira Boudebbouze
- Institut Micalis, MIHA Team, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France; (A.L.W.A.); (S.B.)
| | - Sophie Salomé-Desnoulez
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41-UMS 2014-PLBS, 59000 Lille, France;
| | - Jiří Hrdý
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University and General University Hospital, 121 08 Prague, Czech Republic; (L.S.); (J.H.)
| | | | - Emmanuelle Maguin
- Institut Micalis, MIHA Team, Université Paris-Saclay, INRAE, AgroParisTech, 78350 Jouy-en-Josas, France; (A.L.W.A.); (S.B.)
- Correspondence: (E.M.); (C.G.); Tel.: +33-681-151-925 (E.M.); +33-320-877-392 (C.G.)
| | - Corinne Grangette
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Centre d’Infection et d’Immunité de Lille, 59000 Lille, France; (B.C.); (D.B.); (J.D.)
- Correspondence: (E.M.); (C.G.); Tel.: +33-681-151-925 (E.M.); +33-320-877-392 (C.G.)
| |
Collapse
|
504
|
Characterisation of the secreted apyrase family of Heligmosomoides polygyrus. Int J Parasitol 2020; 51:39-48. [PMID: 32931780 DOI: 10.1016/j.ijpara.2020.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022]
Abstract
Apyrases are a recurrent feature of secretomes from numerous species of parasitic nematodes. Here we characterise the five apyrases secreted by Heligmosomoides polygyrus, a natural parasite of mice and a widely used laboratory model for intestinal nematode infection. All five enzymes are closely related to soluble calcium-activated nucleotidases described in a variety of organisms, and distinct from the CD39 family of ecto-nucleotidases. Expression is maximal in adult worms and restricted to adults and L4s. Recombinant apyrases were produced and purified from Pichia pastoris. The five enzymes showed very similar biochemical properties, with strict calcium dependence and a broad substrate specificity, catalysing the hydrolysis of all nucleoside tri- and diphosphates, with no activity against nucleoside monophosphates. Natural infection of mice provoked very low antibodies to any enzyme, but immunisation with an apyrase cocktail showed partial protection against reinfection, with reduced egg output and parasite recovery. The most likely role for nematode secreted apyrases is hydrolysis of extracellular ATP, which acts as an alarmin for cellular release of IL-33 and initiation of type 2 immunity.
Collapse
|
505
|
Reverchon F, de Concini V, Larrigaldie V, Benmerzoug S, Briault S, Togbé D, Ryffel B, Quesniaux VFJ, Menuet A. Hippocampal interleukin-33 mediates neuroinflammation-induced cognitive impairments. J Neuroinflammation 2020; 17:268. [PMID: 32917228 PMCID: PMC7488545 DOI: 10.1186/s12974-020-01939-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/24/2020] [Indexed: 12/23/2022] Open
Abstract
Background Interleukin (IL)-33 is expressed in a healthy brain and plays a pivotal role in several neuropathologies, as protective or contributing to the development of cerebral diseases associated with cognitive impairments. However, the role of IL-33 in the brain is poorly understood, raising the question of its involvement in immunoregulatory mechanisms. Methods We administered recombinant IL-33 (rmIL-33) by intra-hippocampal injection to C57BL/6 J (WT) and IL-1αβ deficient mice. Chronic minocycline administration was performed and cognitive functions were examined trough spatial habituation test. Hippocampal inflammatory responses were investigated by RT-qPCR. The microglia activation was assessed using immunohistological staining and fluorescence-activated cell sorting (FACS). Results We showed that IL-33 administration in mice led to a spatial memory performance defect associated with an increase of inflammatory markers in the hippocampus while minocycline administration limited the inflammatory response. Quantitative assessment of glial cell activation in situ demonstrated an increase of proximal intersections per radius in each part of the hippocampus. Moreover, rmIL-33 significantly promoted the outgrowth of microglial processes. Fluorescence-activated cell sorting analysis on isolated microglia, revealed overexpression of IL-1β, 48 h post-rmIL-33 administration. This microglial reactivity was closely related to the onset of cognitive disturbance. Finally, we demonstrated that IL-1αβ deficient mice were resistant to cognitive disorders after intra-hippocampal IL-33 injection. Conclusion Thus, hippocampal IL-33 induced an inflammatory state, including IL-1β overexpression by microglia cells, being causative of the cognitive impairment. These results highlight the pathological role for IL-33 in the central nervous system, independently of a specific neuropathological model.
Collapse
Affiliation(s)
- Flora Reverchon
- UMR7355, Experimental and Molecular Immunology and Neurogenetics, CNRS and University of Orléans, 3B rue de la Ferollerie, 45071, Orléans, France.,Current address: Center for Molecular Biophysics, CNRS UPR4301, 45071, Orléans, France
| | - Vidian de Concini
- UMR7355, Experimental and Molecular Immunology and Neurogenetics, CNRS and University of Orléans, 3B rue de la Ferollerie, 45071, Orléans, France
| | - Vanessa Larrigaldie
- UMR7355, Experimental and Molecular Immunology and Neurogenetics, CNRS and University of Orléans, 3B rue de la Ferollerie, 45071, Orléans, France
| | - Sulayman Benmerzoug
- UMR7355, Experimental and Molecular Immunology and Neurogenetics, CNRS and University of Orléans, 3B rue de la Ferollerie, 45071, Orléans, France.,Current address:Department of Urology, Urology Research Unit, CHUV, Lausanne, Switzerland
| | - Sylvain Briault
- UMR7355, Experimental and Molecular Immunology and Neurogenetics, CNRS and University of Orléans, 3B rue de la Ferollerie, 45071, Orléans, France.,Department of Genetics, Regional Hospital, Orléans, France
| | | | - Bernhard Ryffel
- UMR7355, Experimental and Molecular Immunology and Neurogenetics, CNRS and University of Orléans, 3B rue de la Ferollerie, 45071, Orléans, France
| | - Valérie F J Quesniaux
- UMR7355, Experimental and Molecular Immunology and Neurogenetics, CNRS and University of Orléans, 3B rue de la Ferollerie, 45071, Orléans, France
| | - Arnaud Menuet
- UMR7355, Experimental and Molecular Immunology and Neurogenetics, CNRS and University of Orléans, 3B rue de la Ferollerie, 45071, Orléans, France.
| |
Collapse
|
506
|
Ennerfelt HE, Lukens JR. The role of innate immunity in Alzheimer's disease. Immunol Rev 2020; 297:225-246. [PMID: 32588460 PMCID: PMC7783860 DOI: 10.1111/imr.12896] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/23/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022]
Abstract
The amyloid hypothesis has dominated Alzheimer's disease (AD) research for almost 30 years. This hypothesis hinges on the predominant clinical role of the amyloid beta (Aβ) peptide in propagating neurofibrillary tangles (NFTs) and eventual cognitive impairment in AD. Recent research in the AD field has identified the brain-resident macrophages, known as microglia, and their receptors as integral regulators of both the initiation and propagation of inflammation, Aβ accumulation, neuronal loss, and memory decline in AD. Emerging studies have also begun to reveal critical roles for distinct innate immune pathways in AD pathogenesis, which has led to great interest in harnessing the innate immune response as a therapeutic strategy to treat AD. In this review, we will highlight recent advancements in our understanding of innate immunity and inflammation in AD onset and progression. Additionally, there has been mounting evidence suggesting pivotal contributions of environmental factors and lifestyle choices in AD pathogenesis. Therefore, we will also discuss recent findings, suggesting that many of these AD risk factors influence AD progression via modulation of microglia and immune responses.
Collapse
Affiliation(s)
- Hannah E. Ennerfelt
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
- Cell and Molecular Biology Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
- Cell and Molecular Biology Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
507
|
Jiao M, Li X, Chen L, Wang X, Yuan B, Liu T, Dong Q, Mei H, Yin H. Neuroprotective effect of astrocyte-derived IL-33 in neonatal hypoxic-ischemic brain injury. J Neuroinflammation 2020; 17:251. [PMID: 32859229 PMCID: PMC7455908 DOI: 10.1186/s12974-020-01932-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 08/18/2020] [Indexed: 02/07/2023] Open
Abstract
Background Interleukin-33 (IL-33) is a well-recognized pleiotropic cytokine which plays crucial roles in immune regulation and inflammatory responses. Recent studies suggest that IL-33 and its receptor ST2 are involved in the pathogenesis of neurological diseases. Here, we explore the effect of IL-33/ST2 signaling in neonatal hypoxic-ischemic (HI) brain injury and elucidate the underlying mechanisms of action. Methods The brain HI model was established in neonatal C57BL/6 mice by left common carotid artery occlusion with 90 min hypoxia and treated with IL-33 at a dose of 0.2 μg/day i.p. for 3 days. TTC staining and neurobehavioral observation were used to evaluate the HI brain injury. Immunofluorescence and flow cytometry were applied to determine the expression of IL-33 and its receptor ST2 on brain CNS cells and cell proliferation and apoptosis. OGD experiment was used to assay the viability of astrocytes and neurons. RT-qPCR was used to measure the expression of neurotrophic factor-associated genes. Results The expression level of IL-33 was markedly enhanced in astrocytes 24 h after cerebral HI in neonatal mice. Exogenous delivery of IL-33 significantly alleviated brain injury 7 days after HI, whereas ST2 deficiency exacerbated brain infarction and neurological deficits post HI. Flow cytometry analyses demonstrated high levels of ST2 expression on astrocytes, and the expression of ST2 was further elevated after HI. Intriguingly, IL-33 treatment apparently improved astrocyte response and attenuated HI-induced astrocyte apoptosis through ST2 signaling pathways. Further in vitro studies revealed that IL-33-activated astrocytes released a series of neurotrophic factors, which are critical for raising neuronal survival against oxygen glucose deprivation. Conclusions The activation of IL-33/ST2 signaling in the ischemic brain improves astrocyte response, which in turn affords protection to ischemic neurons in a glial-derived neurotrophic factor-dependent manner.
Collapse
Affiliation(s)
- Mengya Jiao
- Department of Biochemistry and Molecular Biology, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xiangyong Li
- Institute of Biochemistry and Molecular Biology, Guangdong Medical University, Zhanjiang, 524023, China
| | - Liying Chen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China.,Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xiaodi Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China.,Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Baohong Yuan
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Tao Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Qun Dong
- Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Hanfang Mei
- Department of Biochemistry and Molecular Biology, Guangdong Pharmaceutical University, Guangzhou, 510006, China. .,Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Hui Yin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China. .,Department of Microbiology and Immunology, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
508
|
Finlay CM, Cunningham KT, Doyle B, Mills KHG. IL-33–Stimulated Murine Mast Cells Polarize Alternatively Activated Macrophages, Which Suppress T Cells That Mediate Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2020; 205:1909-1919. [DOI: 10.4049/jimmunol.1901321] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 07/31/2020] [Indexed: 01/06/2023]
|
509
|
Piyadasa H, Lloyd D, Lee AHY, Altieri A, Hemshekhar M, Osawa N, Basu S, Blimkie T, Falsafi R, Halayko AJ, Hancock REW, Mookherjee N. Characterization of immune responses and the lung transcriptome in a murine model of IL-33 challenge. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165950. [PMID: 32841733 DOI: 10.1016/j.bbadis.2020.165950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/26/2020] [Accepted: 08/20/2020] [Indexed: 12/25/2022]
Abstract
IL-33 induces airway inflammation and hyper-responsiveness in respiratory diseases. Although defined as a therapeutic target, there are limited studies that have comprehensively investigated IL-33-mediated responses in the lungs in vivo. In this study, we characterized immunological and physiological responses induced by intranasal IL-33 challenge, in a mouse model. We identified specific cytokines, IL-4, IL-5, IL-6, IL-10, IP-10 and MIP1-α, that are increased in bronchoalveolar lavage and lung tissues by IL-33. Using transcriptomics (RNA-Seq) we demonstrated that 2279 transcripts were up-regulated and 1378 downregulated (≥ 2-fold, p < 0.01) in lung tissues, in response to IL-33. Bioinformatic interrogation of the RNA-Seq data was used to predict biological pathways and upstream regulators involved in IL-33-mediated responses. We showed that the mRNA and protein of STAT4, a predicted upstream regulator of IL-33-induced transcripts, was significantly enhanced in the lungs following IL-33 challenge. Overall, this study provides specific IL-33-induced molecular targets and endpoints that can be used as a resource for in vivo studies, e.g. in preclinical murine models examining novel interventions to target downstream effects of IL-33.
Collapse
Affiliation(s)
- Hadeesha Piyadasa
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada; Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Dylan Lloyd
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Amy H Y Lee
- Centre for Microbial Disease and Immunity Research, University of British Columbia, Vancouver, Canada; Department of Molecular Biology & Biochemistry, Simon Fraser University, Burnaby, Canada
| | - Anthony Altieri
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada; Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Mahadevappa Hemshekhar
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Natasha Osawa
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Sujata Basu
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada; Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Travis Blimkie
- Centre for Microbial Disease and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Reza Falsafi
- Centre for Microbial Disease and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Andrew J Halayko
- Children's Hospital Research Institute of Manitoba, Winnipeg, Canada; Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada
| | - Robert E W Hancock
- Centre for Microbial Disease and Immunity Research, University of British Columbia, Vancouver, Canada
| | - Neeloffer Mookherjee
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, Winnipeg, Canada; Department of Immunology, University of Manitoba, Winnipeg, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg, Canada.
| |
Collapse
|
510
|
Bechter K. The Challenge of Assessing Mild Neuroinflammation in Severe Mental Disorders. Front Psychiatry 2020; 11:773. [PMID: 32973573 PMCID: PMC7469926 DOI: 10.3389/fpsyt.2020.00773] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 07/21/2020] [Indexed: 12/13/2022] Open
Abstract
Recent psychoneuroimmunology research has provided new insight into the etiology and pathogenesis of severe mental disorders (SMDs). The mild encephalitis (ME) hypothesis was developed with the example of human Borna disease virus infection years ago and proposed, that a subgroup SMD patients, mainly from the broad schizophrenic and affective spectrum, could suffer from mild neuroinflammation, which remained undetected because hard to diagnose with available diagnostic methods. Recently, in neurology an emerging new subgroup of autoimmune encephalitis (AE) cases suffering from various neurological syndromes was described in context with the discovery of an emerging list of Central Nervous System (CNS) autoantibodies. Similarly in psychiatry, consensus criteria of autoimmune psychosis (AP) were developed for patients presenting with CNS autoantibodies together with isolated psychiatric symptoms and paraclinical findings of (mild) neuroinflammation, which in fact match also the previously proposed ME criteria. Nevertheless, identifying mild neuroinflammation in vivo in the individual SMD case remains still a major clinical challenge and the possibility that further cases of ME remain still under diagnosed appears an plausible possibility. In this paper a critical review of recent developments and remaining challenges in the research and clinical diagnosis of mild neuroinflammation in SMDs and in general and in transdisciplinary perspective to psycho-neuro-immunology and neuropsychiatry is given. Present nosological classifications of neuroinflammatory disorders are reconsidered with regard to findings from experimental and clinical research. A refined grading list of clinical states including "classical" encephalitis, AE, AP/ME,and newly proposed terms like parainflammation, stress-induced parainflammation and neuroprogression, and their respective relation to neurodegeneration is presented, which may be useful for further research on the possible causative role of mild neuroinflammation in SMDs. Beyond, an etiology-focused subclassification of ME subtypes, like autoimmune ME or infectious ME, appears to be required for differential diagnosis and individualized treatment. The present status of the clinical diagnosis of mild neuroinflammatory mechanisms involved in SMDs is outlined with the example of actual diagnosis and therapy in AP. Ideas for future research to unravel the contribution of mild neuroinflammation in the causality of SMDs and the difficulties expected to come to novel immune modulatory, anti-infectious or anti-inflammatory therapeutic principles in the sense of precision medicine are discussed.
Collapse
Affiliation(s)
- Karl Bechter
- Department for Psychiatry and Psychotherapy II, Ulm University, Bezirkskrankenhaus Günzburg, Günzburg, Germany
| |
Collapse
|
511
|
Aihara R, Ookawara T, Morimoto A, Iwashita N, Takagi Y, Miyasaka A, Kushiro M, Miyake S, Fukuyama T. Acute and subacute oral administration of mycotoxin deoxynivalenol exacerbates the pro-inflammatory and pro-pruritic responses in a mouse model of allergic dermatitis. Arch Toxicol 2020; 94:4197-4207. [DOI: 10.1007/s00204-020-02875-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 08/12/2020] [Indexed: 12/24/2022]
|
512
|
The Functional Roles of IL-33/ST2 Axis in Ocular Diseases. Mediators Inflamm 2020; 2020:5230716. [PMID: 32908451 PMCID: PMC7450335 DOI: 10.1155/2020/5230716] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/25/2020] [Accepted: 07/31/2020] [Indexed: 01/10/2023] Open
Abstract
Interleukin-33 (IL-33), an important member of the IL-1 family, plays a pivotal role in regulating immune responses via combining with its receptor suppression of tumorigenicity 2 (ST2). We have already known IL-33/ST2 axis participates in the pathogenesis of various diseases, including liver diseases, renal diseases, and neurological diseases. Recently, emerging studies are indicating that IL-33/ST2 is also involved in a wide range of ocular diseases, such as allergic eye disease, keratitis and corneal regeneration, dry eye disease, uveitis, vitreoretinal diseases, and neuromyelitis optica spectrum disorder. In this review, we will summarize and discuss the current understanding about the functional roles of IL-33/ST2 in eyes, with an attempt to explore the possible study perspectives and therapeutic alternatives in the future.
Collapse
|
513
|
Drake LY, Prakash YS. Contributions of IL-33 in Non-hematopoietic Lung Cells to Obstructive Lung Disease. Front Immunol 2020; 11:1798. [PMID: 32903501 PMCID: PMC7438562 DOI: 10.3389/fimmu.2020.01798] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-33 plays important roles in pulmonary immune responses and lung diseases including asthma and chronic obstructive pulmonary disease (COPD). There is substantial interest in identifying and characterizing cellular sources vs. targets of IL-33, and downstream signaling pathways involved in disease pathophysiology. While epithelial and immune cells have largely been the focus, in this review, we summarize current knowledge of expression, induction, and function of IL-33 and its receptor ST2 in non-hematopoietic lung cells in the context of health and disease. Under basal conditions, epithelial cells and endothelial cells are thought to be the primary resident cell types that express high levels of IL-33 and serve as ligand sources compared to mesenchymal cells (smooth muscle cells and fibroblasts). Under inflammatory conditions, IL-33 expression is increased in most non-hematopoietic lung cells, including epithelial, endothelial, and mesenchymal cells. In comparison to its ligand, the receptor ST2 shows low expression levels at baseline but similar to IL-33, ST2 expression is upregulated by inflammation in these non-hematopoietic lung cells which may then participate in chronic inflammation both as sources and autocrine/paracrine targets of IL-33. Downstream effects of IL-33 may occur via direct receptor activation or indirect interactions with the immune system, overall contributing to lung inflammation, airway hyper-responsiveness and remodeling (proliferation and fibrosis). Accordingly from a therapeutic perspective, targeting IL-33 and/or its receptor in non-hematopoietic lung cells becomes relevant.
Collapse
Affiliation(s)
- Li Y Drake
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
514
|
Stevens WW, Kato A. Group 2 innate lymphoid cells in nasal polyposis. Ann Allergy Asthma Immunol 2020; 126:110-117. [PMID: 32781240 DOI: 10.1016/j.anai.2020.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Chronic rhinosinusitis with nasal polyps (CRSwNP) is characterized by a chronic type 2 inflammatory response in the paranasal sinuses. Group 2 innate lymphoid cells (ILC2s) are potent innate immune cells that contribute to type 2 inflammation by producing cytokines such as interleukin (IL)-4, IL-5, and IL-13. There is increasing evidence suggesting that ILC2s play an important role in the CRSwNP pathogenesis. DATA SOURCES We reviewed published literature obtained through PubMed inquiries. STUDY SELECTIONS Studies relevant to the presence, function, and activation of ILC2s in CRSwNP were included. RESULTS Nasal polyps (NPs) are one of the first tissues in which human ILC2s were discovered, and many groups have since reported that these cells are highly elevated in NPs. ILC2s in NPs are also highly activated and produce type 2 cytokines in vivo. Mediators known to activate ILC2s, including receptor activator of nuclear factor kappa-Β ligand, thymic stromal lymphopoietin, various lipid mediators (including prostaglandin D2 and cysteinyl leukotrienes), IL-4, and IL-13 have also been shown to be elevated in NPs compared with healthy sinonasal tissue. Other well-known ILC2 activators, IL-25 and IL-33, are sometimes elevated in NPs in some countries. Furthermore, activation of ILC2s by means of 4 distinct transcriptional pathways (nuclear factor kappa-light-chain-enhancer of activated B cells, nuclear factor of activated T cells, signal transducer and activator of transcription 5, and signal transducer and activator of transcription 6) is needed for the most robust generation of type 2 cytokines. CONCLUSION ILC2-mediated type 2 inflammation plays a crucial role in the pathogenesis of CRSwNP. Targeting the upstream mediators responsible for activating ILC2s and the downstream products that these cells release may play an important role in modifying the inflammatory response and improving clinical outcomes in CRSwNP.
Collapse
Affiliation(s)
- Whitney W Stevens
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Atsushi Kato
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
515
|
Aparicio-Domingo P, Cannelle H, Buechler MB, Nguyen S, Kallert SM, Favre S, Alouche N, Papazian N, Ludewig B, Cupedo T, Pinschewer DD, Turley SJ, Luther SA. Fibroblast-derived IL-33 is dispensable for lymph node homeostasis but critical for CD8 T-cell responses to acute and chronic viral infection. Eur J Immunol 2020; 51:76-90. [PMID: 32700362 DOI: 10.1002/eji.201948413] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 06/02/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022]
Abstract
Upon viral infection, stressed or damaged cells can release alarmins like IL-33 that act as endogenous danger signals alerting innate and adaptive immune cells. IL-33 coming from nonhematopoietic cells has been identified as important factor triggering the expansion of antiviral CD8+ T cells. In LN the critical cellular source of IL-33 is unknown, as is its potential cell-intrinsic function as a chromatin-associated factor. Using IL-33-GFP reporter mice, we identify fibroblastic reticular cells (FRC) and lymphatic endothelial cells (LEC) as the main IL-33 source. In homeostasis, IL-33 is dispensable as a transcriptional regulator in FRC, indicating it functions mainly as released cytokine. Early during infection with lymphocytic choriomeningitis virus (LCMV) clone 13, both FRC and LEC lose IL-33 protein expression suggesting cytokine release, correlating timewise with IL-33 receptor expression by reactive CD8+ T cells and their greatly augmented expansion in WT versus ll33-/- mice. Using mice lacking IL-33 selectively in FRC versus LEC, we identify FRC as key IL-33 source driving acute and chronic antiviral T-cell responses. Collectively, these findings show that LN T-zone FRC not only regulate the homeostasis of naïve T cells but also their expansion and differentiation several days into an antiviral response.
Collapse
Affiliation(s)
| | - Hélène Cannelle
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Matthew B Buechler
- Department of Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Sylvain Nguyen
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Sandra M Kallert
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Basel, Switzerland
| | - Stéphanie Favre
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Nagham Alouche
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Natalie Papazian
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St.Gallen, St. Gallen, Switzerland
| | - Tom Cupedo
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daniel D Pinschewer
- Department of Biomedicine, Division of Experimental Virology, University of Basel, Basel, Switzerland
| | - Shannon J Turley
- Department of Cancer Immunology, Genentech, South San Francisco, CA, USA
| | - Sanjiv A Luther
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
516
|
Xu D, Huang SK. IL-33: a key player in the development of iron-recycling red pulp macrophages. Cell Mol Immunol 2020; 17:1218-1219. [PMID: 32737432 DOI: 10.1038/s41423-020-0509-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 11/09/2022] Open
Affiliation(s)
- Damo Xu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China. .,Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| | - Shau-Ku Huang
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China. .,National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, China. .,Department of Respirology & Allergy, Third Affiliated Hospital of Shenzhen University, Shenzhen, 518020, China. .,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, USA.
| |
Collapse
|
517
|
Vohralik EJ, Psaila AM, Knights AJ, Quinlan KGR. EoTHINophils: Eosinophils as key players in adipose tissue homeostasis. Clin Exp Pharmacol Physiol 2020; 47:1495-1505. [PMID: 32163614 DOI: 10.1111/1440-1681.13304] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/24/2020] [Accepted: 03/09/2020] [Indexed: 12/22/2022]
Abstract
Eosinophils are granular cells of the innate immune system that are found in almost all vertebrates and some invertebrates. Knowledge of their wide-ranging roles in health and disease has largely been attained through studies in mice and humans. Although eosinophils are typically associated with helminth infections and allergic diseases such as asthma, there is building evidence that beneficial homeostatic eosinophils residing in specific niches are important for tissue development, remodelling and metabolic control. In recent years, the importance of immune cells in the regulation of adipose tissue homeostasis has been a focal point of research efforts. There is an abundance of anti-inflammatory innate immune cells in lean white adipose tissue, including macrophages, eosinophils and group 2 innate lymphoid cells, which promote energy homeostasis and stimulate the development of thermogenic beige adipocytes. This review will evaluate evidence for the role of adipose-resident eosinophils in local tissue homeostasis, beiging and systemic metabolism, highlighting where more research is needed to establish the specific effector functions that adipose eosinophils perform in response to different internal and external cues.
Collapse
Affiliation(s)
- Emily J Vohralik
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Annalise M Psaila
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Alexander J Knights
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Kate G R Quinlan
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
518
|
Relja B, Land WG. Damage-associated molecular patterns in trauma. Eur J Trauma Emerg Surg 2020; 46:751-775. [PMID: 31612270 PMCID: PMC7427761 DOI: 10.1007/s00068-019-01235-w] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/27/2019] [Indexed: 12/13/2022]
Abstract
In 1994, the "danger model" argued that adaptive immune responses are driven rather by molecules released upon tissue damage than by the recognition of "strange" molecules. Thus, an alternative to the "self versus non-self recognition model" has been provided. The model, which suggests that the immune system discriminates dangerous from safe molecules, has established the basis for the future designation of damage-associated molecular patterns (DAMPs), a term that was coined by Walter G. Land, Seong, and Matzinger. The pathological importance of DAMPs is barely somewhere else evident as in the posttraumatic or post-surgical inflammation and regeneration. Since DAMPs have been identified to trigger specific immune responses and inflammation, which is not necessarily detrimental but also regenerative, it still remains difficult to describe their "friend or foe" role in the posttraumatic immunogenicity and healing process. DAMPs can be used as biomarkers to indicate and/or to monitor a disease or injury severity, but they also may serve as clinically applicable parameters for optimized indication of the timing for, i.e., secondary surgeries. While experimental studies allow the detection of these biomarkers on different levels including cellular, tissue, and circulatory milieu, this is not always easily transferable to the human situation. Thus, in this review, we focus on the recent literature dealing with the pathophysiological importance of DAMPs after traumatic injury. Since dysregulated inflammation in traumatized patients always implies disturbed resolution of inflammation, so-called model of suppressing/inhibiting inducible DAMPs (SAMPs) will be very briefly introduced. Thus, an update on this topic in the field of trauma will be provided.
Collapse
Affiliation(s)
- Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany.
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt, Germany.
| | - Walter Gottlieb Land
- Molecular ImmunoRheumatology, INSERM UMR_S1109, Laboratory of Excellence Transplantex, University of Strasbourg, Strasbourg, France
| |
Collapse
|
519
|
She L, Alanazi HH, Yan L, Brooks EG, Dube PH, Xiang Y, Zhang F, Sun Y, Liu Y, Zhang X, Li XD. Sensing and signaling of immunogenic extracellular RNAs restrain group 2 innate lymphoid cell-driven acute lung inflammation and airway hyperresponsiveness. PLoS One 2020; 15:e0236744. [PMID: 32730309 PMCID: PMC7392318 DOI: 10.1371/journal.pone.0236744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/13/2020] [Indexed: 01/02/2023] Open
Abstract
Repeated exposures to environmental allergens in susceptible individuals drive the development of type 2 inflammatory conditions such as asthma, which have been traditionally considered to be mainly mediated by Th2 cells. However, emerging evidence suggest that a new innate cell type, group 2 innate lymphoid cells (ILC2), plays a central role in initiating and amplifying a type 2 response, even in the absence of adaptive immunity. At present, the regulatory mechanisms for controlling ILC2 activation remain poorly understood. Here we report that respiratory delivery of immunogenic extracellular RNA (exRNAs) derived from RNA- and DNA-virus infected cells, was able to activate a protective response against acute type 2 lung immunopathology and airway hyperresponsiveness (AHR) induced by IL-33 and a fungal allergen, A. flavus, in mice. Mechanistically, we found that the innate immune responses triggered by exRNAs had a potent suppressive effect in vivo on the proliferation and function of ILC2 without the involvement of adaptive immunity. We further provided the loss-of-function genetic evidence that the TLR3- and MAVS-mediated signaling axis is essential for the inhibitory effects of exRNAs in mouse lungs. Thus, our results indicate that the host detection of extracellular immunostimulatory RNAs generated during respiratory viral infections have an important function in the regulation of ILC2-driven acute lung inflammation.
Collapse
Affiliation(s)
- Li She
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hamad H. Alanazi
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Liping Yan
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Edward G. Brooks
- Division of Immunology and Infectious Disease, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States of America
| | - Peter H. Dube
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Yan Xiang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Fushun Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Yilun Sun
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
| | - Yong Liu
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Zhang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiao-Dong Li
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States of America
- * E-mail:
| |
Collapse
|
520
|
The soluble glycoprotein NMB (GPNMB) produced by macrophages induces cancer stemness and metastasis via CD44 and IL-33. Cell Mol Immunol 2020; 18:711-722. [PMID: 32728200 DOI: 10.1038/s41423-020-0501-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
In cancer, myeloid cells have tumor-supporting roles. We reported that the protein GPNMB (glycoprotein nonmetastatic B) was profoundly upregulated in macrophages interacting with tumor cells. Here, using mouse tumor models, we show that macrophage-derived soluble GPNMB increases tumor growth and metastasis in Gpnmb-mutant mice (DBA/2J). GPNMB triggers in the cancer cells the formation of self-renewing spheroids, which are characterized by the expression of cancer stem cell markers, prolonged cell survival and increased tumor-forming ability. Through the CD44 receptor, GPNMB mechanistically activates tumor cells to express the cytokine IL-33 and its receptor IL-1R1L. We also determined that recombinant IL-33 binding to IL-1R1L is sufficient to induce tumor spheroid formation with features of cancer stem cells. Overall, our results reveal a new paracrine axis, GPNMB and IL-33, which is activated during the cross talk of macrophages with tumor cells and eventually promotes cancer cell survival, the expansion of cancer stem cells and the acquisition of a metastatic phenotype.
Collapse
|
521
|
Medara N, Lenzo JC, Walsh KA, Darby IB, O'Brien-Simpson NM, Reynolds EC. T helper 17 cell-related cytokines in serum and saliva during management of periodontitis. Cytokine 2020; 134:155186. [PMID: 32717609 DOI: 10.1016/j.cyto.2020.155186] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/27/2022]
Abstract
AIM T helper (Th)17 cells are implicated in the pathogenesis of periodontitis. This study investigated the effect of periodontal management on fifteen Th17-related cytokines in serum and saliva in periodontitis patients. MATERIALS AND METHODS Periodontal parameters, serum and saliva were collected from 40 healthy controls and 54 periodontitis subjects before treatment, and 3-, 6- and 12-months post-treatment. Cytokine concentrations of IL-1β, IL-4, IL-6, IL-10, IL-17A, IL-17F, IL-21, IL-22, IL-23, IL-25, IL-31, IL-33, IFN-γ, sCD40L and TNF-α were determined by Luminex assay. RESULTS IL-1β, IL-6, sCD40L and TNF-α in serum, and IL-1β, IL-6, IL-25 and IL-31 in saliva were significantly higher at baseline compared to health and decreased with treatment. In contrast, serum IL-31 was significantly lower at baseline compared to health and increased with treatment. In addition, salivary IL-10, IL-17A, IL-17F, IL-23, IL-33, IFN-γ and TNF-α also displayed treatment-related reduction. Correlation networks showed that cytokines in saliva displayed a higher number of correlations compared to serum in periodontitis. CONCLUSION Treatment generally decreased cytokine concentrations except for serum IL-31 which showed a treatment-related increase. Serum cytokine concentrations may not be reflective of salivary cytokines. Saliva may be a better medium for cytokine detection compared to serum. Serum IL-31 and salivary IL-1β, IL-6, IL-10 and TNF-α were significant predictors for mean probing depth and may be potential biomarkers of interest in the pathogenesis of periodontitis.
Collapse
Affiliation(s)
- Nidhi Medara
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia.
| | - Jason C Lenzo
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; Centre for Oral Health Research, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia.
| | - Katrina A Walsh
- Austin Hospital, 145 Studley Rd, Heidelberg, VIC 3084, Australia.
| | - Ivan B Darby
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia.
| | - Neil M O'Brien-Simpson
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; Centre for Oral Health Research, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia.
| | - Eric C Reynolds
- Melbourne Dental School, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia; Centre for Oral Health Research, The University of Melbourne, 720 Swanston Street, Carlton, VIC 3053, Australia.
| |
Collapse
|
522
|
Hill GR, Koyama M. Cytokines and costimulation in acute graft-versus-host disease. Blood 2020; 136:418-428. [PMID: 32526028 PMCID: PMC7378458 DOI: 10.1182/blood.2019000952] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/18/2020] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (alloSCT) is an important curative therapy for high-risk hematological malignancies, but the development of severe and/or steroid-refractory acute graft-versus-host disease (aGVHD) remains a significant limitation to optimal outcomes. New approaches to prevent and treat aGVHD remain an unmet need that can be best addressed by understanding the complex disease pathophysiology. It is now clear that chemoradiotherapy used prior to alloSCT induces the release of endogenous alarmins (eg, HMGB-1, ATP, IL-1α, IL-33) from recipient tissue. Exogenous pathogen-derived molecules (eg, lipopolysaccharide, nucleic acids) also translocate from the gastrointestinal tract lumen. Together, these danger signals activate antigen-presenting cells (APCs) to efficiently present alloantigen to donor T cells while releasing cytokines (eg, interleukin-12 [IL-12], IL-23, IL-6, IL-27, IL-10, transforming growth factor-β) that expand and differentiate both pathogenic and regulatory donor T cells. Concurrent costimulatory signals at the APC-T-cell interface (eg, CD80/CD86-CD28, CD40-CD40L, OX40L-OX40, CD155/CD112-DNAM-1) and subsequent coinhibitory signals (eg, CD80/CD86-CTLA4, PDL1/2-PD1, CD155/CD112-TIGIT) are critical to the acquisition of effector T-cell function and ensuing secretion of pathogenic cytokines (eg, IL-17, interferon-γ, tissue necrosis factor, granulocyte-macrophage colony-stimulating factor) and cytolytic degranulation pathway effectors (eg, perforin/granzyme). This review focuses on the combination of cytokine and costimulatory networks at the T-cell surface that culminates in effector function and subsequent aGVHD in target tissue. Together, these pathways now represent robust and clinically tractable targets for preventing the initiation of deleterious immunity after alloSCT.
Collapse
Affiliation(s)
- Geoffrey R Hill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA; and
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA
| | - Motoko Koyama
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA; and
| |
Collapse
|
523
|
Yang Q, Kong L, Huang W, Mohammadtursun N, Li X, Wang G, Wang L. Osthole attenuates ovalbumin‑induced lung inflammation via the inhibition of IL‑33/ST2 signaling in asthmatic mice. Int J Mol Med 2020; 46:1389-1398. [PMID: 32700747 PMCID: PMC7447319 DOI: 10.3892/ijmm.2020.4682] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 07/03/2020] [Indexed: 01/12/2023] Open
Abstract
Asthma is a common chronic inflammatory airway disease. Recent studies have reported that interleukin (IL)-33 is a potential link between the airway epithelium and Th2-type inflammatory responses, which are closely related to the progression of asthma. The IL-33 receptor, ST2, is highly expressed in group 2 innate lymphoid cells (ILC2s), Th2 cells, mast cells, eosinophils and natural killer (NK) cells. Cnidii Fructus is a Chinese herb with a long history of use in the treatment of asthma in China. Osthole is one of the major components of Cnidii Fructus. The present study examined the anti-asthmatic effects of osthole in mice and aimed to elucidate the underlying mechanisms involving the IL-33/ST2 pathway. BALB/c mice were sensitized and challenged with ovalbumin and then treated with an intraperitoneal injection of osthole (25 and 50 mg/kg). Subsequently, the airway hyper-responsiveness (AHR) and inflammation of the lungs were evaluated. The amounts of IL-4, IL-5, IL-13, interferon (IFN)-γ and IL-33 in the bronchoalveolar lavage fluid (BALF) were measured by Luminex assay and their mRNA levels in the lungs were measured by reverse transcription-quantitative PCR. The histopathology of the lungs was performed with H&E, PAS and Masson's staining. The expression of ST2 in the lungs was evaluated by immunohistochemistry. The data demonstrated that osthole markedly reduced AHR and decreased the number of eosinophils and lymphocytes in BALF. It was also observed that osthole significantly inhibited the release of Th2-type cytokines (IL-4, IL-5 and IL-13) and upregulated the IFN-γ level in BALF. Moreover, osthole significantly attenuated the IL-33 and ST2 expression in the lungs of asthmatic mice. On the whole, osthole attenuated ovalbumin-induced lung inflammation through the inhibition of IL-33/ST2 signaling in an asthmatic mouse model. These results suggest that osthole is a promising target for the development of an asthma medication.
Collapse
Affiliation(s)
- Qingqing Yang
- Department of Respiratory Medicine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Lingwen Kong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Weiling Huang
- Department of Respiratory Medicine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, P.R. China
| | - Nabijan Mohammadtursun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Xiumin Li
- Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Guifang Wang
- Department of Respiratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Lixin Wang
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, P.R. China
| |
Collapse
|
524
|
Taniguchi S, Elhance A, Van Duzer A, Kumar S, Leitenberger JJ, Oshimori N. Tumor-initiating cells establish an IL-33-TGF-β niche signaling loop to promote cancer progression. Science 2020; 369:eaay1813. [PMID: 32675345 PMCID: PMC10870826 DOI: 10.1126/science.aay1813] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 05/29/2020] [Indexed: 12/11/2022]
Abstract
Targeting the cross-talk between tumor-initiating cells (TICs) and the niche microenvironment is an attractive avenue for cancer therapy. We show here, using a mouse model of squamous cell carcinoma, that TICs play a crucial role in creating a niche microenvironment that is required for tumor progression and drug resistance. Antioxidant activity in TICs, mediated by the transcription factor NRF2, facilitates the release of a nuclear cytokine, interleukin-33 (IL-33). This cytokine promotes differentiation of macrophages that express the high-affinity immunoglobulin E receptor FcεRIα and are in close proximity to TICs. In turn, these IL-33-responding FcεRIα+ macrophages send paracrine transforming growth factor β (TGF-β) signals to TICs, inducing invasive and drug-resistant properties and further upregulating IL-33 expression. This TIC-driven, IL-33-TGF-β feedforward loop could potentially be exploited for cancer treatment.
Collapse
Affiliation(s)
- Sachiko Taniguchi
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ajit Elhance
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Avery Van Duzer
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sushil Kumar
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Justin J Leitenberger
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Naoki Oshimori
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA.
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Otolaryngology - Head & Neck Surgery, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
525
|
Liang CS, Su KP, Tsai CL, Lee JT, Chu CS, Yeh TC, Su MW, Lin GY, Lin YK, Chu HT, Tsai CK, Yang FC. The role of interleukin-33 in patients with mild cognitive impairment and Alzheimer's disease. Alzheimers Res Ther 2020; 12:86. [PMID: 32678011 PMCID: PMC7367330 DOI: 10.1186/s13195-020-00652-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/07/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND The neuroprotective role of interleukin (IL)-33 is supported by numerous preclinical studies, but it remains uninvestigated in clinical studies of Alzheimer's disease (AD). We aimed to examine the association between human blood levels of IL-33 and cognitive preservation in amnestic mild cognitive impairment (aMCI) and AD. METHODS A total of 100 participants (26 controls, 35 aMCI patients, and 39 AD patients) completed two Mini-Mental State Examinations (MMSEs) over a 1-year interval. In all 100 participants at the second MMSE, we examined the plasma levels of IL-33, IL-β, IL-1 receptor agonist (IL-1RA), beta amyloid (Aβ), and tau and apolipoprotein E (ApoE) genotyping; we also performed Hopkins Verbal Learning Test, Trail Making Test, forward and backward digit span, and Clinical Dementia Rating. RESULTS IL-33 expression showed a positive trend among controls (1/26 = 3.8%), aMCI (9/35 = 25.7%), and AD (17/39 = 43.6%) (trend analysis: P < 0.001). Patients expressing IL-33 preserved their cognitive function compared with IL-33 non-expressing patients (1-year ΔMMSE, 0.16 ± 1.6 vs - 1.5 ± 2.6; P = 0.006). The cognitive preservation was not associated with the lower levels of Aβ, tau, and ApoE ε4, while higher levels of ApoE ε4 and phosphorylated tau were indeed associated with cognitive decline. The aMCI patients with AD conversion during study period had higher proportion of IL-33(-) than non-AD converters (90.9% vs 53.3%, P = 0.04). CONCLUSIONS IL-33 or its associated signaling pathways may represent a new treatment paradigm for aMCI and AD.
Collapse
Affiliation(s)
- Chih-Sung Liang
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Kuan-Pin Su
- Department of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
- An-Nan Hospital, China Medical University, Tainan, Taiwan
| | - Chia-Lin Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, No.325, Section 2, Cheng-Kung Road, Neihu District, Taipei City 114, Taiwan
| | - Jiunn-Tay Lee
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, No.325, Section 2, Cheng-Kung Road, Neihu District, Taipei City 114, Taiwan
| | - Che-Sheng Chu
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Center for Geriatric and Gerontology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Ta-Chuan Yeh
- Department of Psychiatry, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Wei Su
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Guan-Yu Lin
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, No.325, Section 2, Cheng-Kung Road, Neihu District, Taipei City 114, Taiwan
| | - Yu-Kai Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, No.325, Section 2, Cheng-Kung Road, Neihu District, Taipei City 114, Taiwan
| | - Hsuan-Te Chu
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chia-Kuang Tsai
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, No.325, Section 2, Cheng-Kung Road, Neihu District, Taipei City 114, Taiwan
| | - Fu-Chi Yang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.
- Department of Neurology, Tri-Service General Hospital, National Defense Medical Center, No.325, Section 2, Cheng-Kung Road, Neihu District, Taipei City 114, Taiwan.
| |
Collapse
|
526
|
Li W, Li Y, Jin J. The essential function of IL-33 in metabolic regulation. Acta Biochim Biophys Sin (Shanghai) 2020; 52:768-775. [PMID: 32445465 DOI: 10.1093/abbs/gmaa045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 12/20/2019] [Accepted: 02/24/2020] [Indexed: 12/18/2022] Open
Abstract
Interleukin-33 (IL-33) is produced by various types of cells under physical or pathological conditions. As a multifunctional partner in health and disease, current evidence reveals that IL-33 also participates in several metabolic processes. IL-33 has been proven to contribute to regulating the activity of ST2+ group 2 innate lymphoid cells and regulatory T cells in adipose, which leads to the shift of insulin sensitivity and glucose clearance in glucose metabolism, thermogenesis, and adipocyte beiging in adipose metabolism. In this review, we briefly summarize the biological characteristics of Il-33 and discuss its regulatory function in glucose and adipose metabolism. By clarifying the underlying mechanism of IL-33, we highlight the crosstalk between immune response and metabolic processes mediated by IL-33.
Collapse
Affiliation(s)
- Wenping Li
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Yiyuan Li
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Jin Jin
- MOE Laboratory of Biosystem Homeostasis and Protection, and Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
- Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310016, China
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
527
|
Amruta N, Rahman AA, Pinteaux E, Bix G. Neuroinflammation and fibrosis in stroke: The good, the bad and the ugly. J Neuroimmunol 2020; 346:577318. [PMID: 32682140 PMCID: PMC7794086 DOI: 10.1016/j.jneuroim.2020.577318] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/01/2020] [Accepted: 07/03/2020] [Indexed: 02/08/2023]
Abstract
Stroke is the leading cause of death and the main cause of disability in surviving patients. The detrimental interaction between immune cells, glial cells, and matrix components in stroke pathology results in persistent inflammation that progresses to fibrosis. A substantial effort is being directed toward understanding the exact neuroinflammatory events that take place as a result of stroke. The initiation of a potent cytokine response, along with immune cell activation and infiltration in the ischemic core, has massive acute deleterious effects, generally exacerbated by comorbid inflammatory conditions. There is secondary neuroinflammation that promotes further injury, resulting in cell death, but conversely plays a beneficial role, by promoting recovery. This highlights the need for a better understanding of the neuroinflammatory and fibrotic processes, as well as the need to identify new mechanisms and potential modulators. In this review, we summarize several aspects of stroke-induced inflammation, fibrosis, and include a discussion of cytokine inhibitors/inducers, immune cells, and fibro-inflammation signaling inhibitors in order to identify new pharmacological means of intervention.
Collapse
Affiliation(s)
- Narayanappa Amruta
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Abir A Rahman
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA.
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom.
| | - Gregory Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA 70112, USA; Faculty of Biology, Medicine and Health, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, United Kingdom; Tulane Brain Institute, Tulane University, New Orleans, LA 70118, USA.
| |
Collapse
|
528
|
Morales Del Valle C, Maxwell JR, Xu MM, Menoret A, Mittal P, Tsurutani N, Adler AJ, Vella AT. Costimulation Induces CD4 T Cell Antitumor Immunity via an Innate-like Mechanism. Cell Rep 2020; 27:1434-1445.e3. [PMID: 31042471 DOI: 10.1016/j.celrep.2019.04.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 12/12/2018] [Accepted: 04/02/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic exposure to tumor-associated antigens inactivates cognate T cells, restricting the repertoire of tumor-specific effector T cells. This problem was studied here by transferring TCR transgenic CD4 T cells into recipient mice that constitutively express a cognate self-antigen linked to MHC II on CD11c-bearing cells. Immunotherapeutic agonists to CD134 plus CD137, "dual costimulation," induces specific CD4 T cell expansion and expression of the receptor for the Th2-associated IL-1 family cytokine IL-33. Rather than producing IL-4, however, they express the tumoricidal Th1 cytokine IFNγ when stimulated with IL-33 or IL-36 (a related IL-1 family member) plus IL-12 or IL-2. IL-36, which is induced within B16-F10 melanomas by dual costimulation, reduces tumor growth when injected intratumorally as a monotherapy and boosts the efficacy of tumor-nonspecific dual costimulated CD4 T cells. Dual costimulation thus enables chronic antigen-exposed CD4 T cells, regardless of tumor specificity, to elaborate tumoricidal function in response to tumor-associated cytokines.
Collapse
Affiliation(s)
| | - Joseph R Maxwell
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Maria M Xu
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Antoine Menoret
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Payal Mittal
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Naomi Tsurutani
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA
| | - Adam J Adler
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA.
| | - Anthony T Vella
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030, USA.
| |
Collapse
|
529
|
Eissmann MF, Buchert M, Ernst M. IL33 and Mast Cells-The Key Regulators of Immune Responses in Gastrointestinal Cancers? Front Immunol 2020; 11:1389. [PMID: 32719677 PMCID: PMC7350537 DOI: 10.3389/fimmu.2020.01389] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 05/29/2020] [Indexed: 12/13/2022] Open
Abstract
The Interleukin (IL-)1 family IL33 is best known for eliciting type 2 immune responses by stimulating mast cells (MCs), regulatory T-cells (Tregs), innate lymphoid cells (ILCs) and other immune cells. MCs and IL33 provide critical control of immunological and epithelial homeostasis in the gastrointestinal (GI) tract. Meanwhile, the role of MCs in solid malignancies appears tissue-specific with both pro and anti-tumorigenic activities. Likewise, IL33 signaling significantly shapes immune responses in the tumor microenvironment, but these effects remain often dichotomous when assessed in experimental models of cancer. Thus, the balance between tumor suppressing and tumor promoting activities of IL33 are highly context dependent, and most likely dictated by the mixture of cell types responding to IL33. Adding to this complexity is the promiscuous nature by which MCs respond to cytokines other than IL33 and release chemotactic factors that recruit immune cells into the tumor microenvironment. In this review, we integrate the outcomes of recent studies on the role of MCs and IL33 in cancer with our own observations in the GI tract. We propose a working model where the most abundant IL33 responsive immune cell type is likely to dictate an overall tumor-supporting or tumor suppressing outcome in vivo. We discuss how these opposing responses affect the therapeutic potential of targeting MC and IL33, and highlight the caveats and challenges facing our ability to effectively harness MCs and IL33 biology for anti-cancer immunotherapy.
Collapse
Affiliation(s)
- Moritz F Eissmann
- Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Michael Buchert
- Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute, and La Trobe University School of Cancer Medicine, Heidelberg, VIC, Australia
| |
Collapse
|
530
|
Wang Y, Luo H, Wei M, Becker M, Hyde RK, Gong Q. IL-33/IL1RL1 axis regulates cell survival through the p38 MAPK pathway in acute myeloid leukemia. Leuk Res 2020; 96:106409. [PMID: 32652328 DOI: 10.1016/j.leukres.2020.106409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/13/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023]
Abstract
Acute myeloid leukemia (AML) is often characterized by the presence of specific and recurrent chromosomal abnormalities. Current treatments have greatly increased remission rate, but relapse still occurs. Therefore, novel therapeutic approaches are required. Previously, using a conditional Cbfb-MYH11 knockin mouse model, we showed that Cbfb-MYH11 induces the expression of a cytokine receptor, IL1RL1. Treatment with IL-33, the only known ligand of IL1RL1, promotes leukemia cell survival in vitro. We further found that IL1RL1+ cells survive better with chemotherapy than IL1RL1- population. However, the mechanism is not clear. Here, we show that IL-33 treatment decreased drug sensitivity in the human inv(16) AML cell line ME-1. By RT-PCR, we found that IL-33 increased the expression of IL-4 and IL-6 and led to the activation of both p38 MAPK and NF-κB. We also showed that IL-33 decreased apoptosis with increased phosphorylation of p38 MAPK. Moreover, pre-treatment with MAPK inhibitor attenuated the phosphorylation of p38 enhanced by IL-33 and reversed the anti-apoptotic effect by IL-33. Taken together, our findings give news insights into the potential mechanism of the anti-apoptotic effect by IL-33/IL1RL1 axis in AML which will help in future drug development.
Collapse
Affiliation(s)
- Yiqian Wang
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, PR China
| | - Huanmin Luo
- Third Clinical School, Guangzhou Medical University, Guangzhou, PR China
| | - Mengyi Wei
- Nanshan School, Guangzhou Medical University, Guangzhou, PR China
| | - Michelle Becker
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - R Katherine Hyde
- Department of Biochemistry and Molecular Biology, and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Qing Gong
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, PR China.
| |
Collapse
|
531
|
Akdis CA, Arkwright PD, Brüggen MC, Busse W, Gadina M, Guttman‐Yassky E, Kabashima K, Mitamura Y, Vian L, Wu J, Palomares O. Type 2 immunity in the skin and lungs. Allergy 2020; 75:1582-1605. [PMID: 32319104 DOI: 10.1111/all.14318] [Citation(s) in RCA: 342] [Impact Index Per Article: 68.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022]
Abstract
There has been extensive progress in understanding the cellular and molecular mechanisms of inflammation and immune regulation in allergic diseases of the skin and lungs during the last few years. Asthma and atopic dermatitis (AD) are typical diseases of type 2 immune responses. interleukin (IL)-25, IL-33, and thymic stromal lymphopoietin are essential cytokines of epithelial cells that are activated by allergens, pollutants, viruses, bacteria, and toxins that derive type 2 responses. Th2 cells and innate lymphoid cells (ILC) produce and secrete type 2 cytokines such as IL-4, IL-5, IL-9, and IL-13. IL-4 and IL-13 activate B cells to class-switch to IgE and also play a role in T-cell and eosinophil migration to allergic inflammatory tissues. IL-13 contributes to maturation, activation, nitric oxide production and differentiation of epithelia, production of mucus as well as smooth muscle contraction, and extracellular matrix generation. IL-4 and IL-13 open tight junction barrier and cause barrier leakiness in the skin and lungs. IL-5 acts on activation, recruitment, and survival of eosinophils. IL-9 contributes to general allergic phenotype by enhancing all of the aspects, such as IgE and eosinophilia. Type 2 ILC contribute to inflammation in AD and asthma by enhancing the activity of Th2 cells, eosinophils, and their cytokines. Currently, five biologics are licensed to suppress type 2 inflammation via IgE, IL-5 and its receptor, and IL-4 receptor alpha. Some patients with severe atopic disease have little evidence of type 2 hyperactivity and do not respond to biologics which target this pathway. Studies in responder and nonresponder patients demonstrate the complexity of these diseases. In addition, primary immune deficiency diseases related to T-cell maturation, regulatory T-cell development, and T-cell signaling, such as Omenn syndrome, severe combined immune deficiencies, immunodysregulation, polyendocrinopathy, enteropathy, X-linked syndrome, and DOCK8, STAT3, and CARD11 deficiencies, help in our understanding of the importance and redundancy of various type 2 immune components. The present review aims to highlight recent advances in type 2 immunity and discuss the cellular sources, targets, and roles of type 2 mechanisms in asthma and AD.
Collapse
Affiliation(s)
- Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
| | - Peter D. Arkwright
- Lydia Becker Institute of Immunology and Inflammation University of Manchester Manchester UK
| | - Marie-Charlotte Brüggen
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
- Department of Dermatology University Hospital Zurich Zurich Switzerland
- Faculty of Medicine University Zurich Zurich Switzerland
| | - William Busse
- Department of Medicine School of Medicine and Public Health University of Wisconsin Madison WI USA
| | - Massimo Gadina
- Translational Immunology Section Office of Science and Technology National Institute of Arthritis Musculoskeletal and Skin Disease NIH Bethesda MD USA
| | - Emma Guttman‐Yassky
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases Icahn School of Medicine at Mount Sinai New York NY USA
- Laboratory for Investigative Dermatology The Rockefeller University New York NY USA
| | - Kenji Kabashima
- Department of Dermatology Kyoto University Graduate School of Medicine Kyoto Japan
- Agency for Science, Technology and Research (A*STAR) Singapore Immunology Network (SIgN) and Skin Research Institute of Singapore (SRIS) Singapore Singapore
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Laura Vian
- Translational Immunology Section Office of Science and Technology National Institute of Arthritis Musculoskeletal and Skin Disease NIH Bethesda MD USA
| | - Jianni Wu
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases Icahn School of Medicine at Mount Sinai New York NY USA
- Laboratory for Investigative Dermatology The Rockefeller University New York NY USA
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology School of Chemistry Complutense University of Madrid Madrid Spain
| |
Collapse
|
532
|
Chen JL, Zhang CL. [Changes of serum interleukin-33 in preterm infants with bronchopulmonary dysplasia]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2020; 22:716-720. [PMID: 32669167 PMCID: PMC7389611 DOI: 10.7499/j.issn.1008-8830.2001063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/17/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE To study the role of interleukin-33 (IL-33) in the development and progression of bronchopulmonary dysplasia (BPD) in preterm infants. METHODS A prospective cohort study was performed on 128 preterm infants with a gestational age of ≤32 weeks and/or a birth weight of ≤1 500 g. They were classified to a non-BPD group with 50 infants, a mild BPD group with 32 infants, a moderate BPD group with 30 infants, and a severe BPD group with 16 infants. Related data were collected, including antepartum factors of mothers (antepartum hormone and chorioamnionitis), intrapartum factors of preterm infants (sex, gestational age, birth weight, mode of birth, and birth asphyxia), treatment after birth (pulmonary surfactant, duration of invasive ventilation, duration of noninvasive ventilation, duration of parenteral nutrition, and length of hospital stay). The high-risk factors for BPD were analyzed. ELISA was used to measure the serum level of IL-33 in preterm infants on days 1, 14, and 28 after birth. The serum level of IL-33 was compared between groups at different time points after birth. The preterm infants with moderate or severe BPD were treated with conventional corticosteroid therapy (DART regimen), and the serum level of IL-33 was measured before and after treatment. RESULTS There were significant differences between the preterm infants with BPD and those without BPD in the incidence of maternal chorioamnionitis, gestational age, birth weight, the incidence of birth asphyxia, duration of invasive ventilation, duration of noninvasive ventilation, duration of parenteral nutrition, and total length of hospital stay (P<0.05). There were significant differences in the above indices among the preterm infants with different severities of BPD (P<0.05). On days 1, 14, and 28 after birth, the infants with BPD had a significantly higher serum level of IL-33 than those without BPD, and the serum level of IL-33 tended to increase with the severity of BPD and over the time after birth (P<0.05). The preterm infants with moderate or severe BPD had a significant reduction in the serum level of IL-33 after the treatment with DART regimen (P<0.05). CONCLUSIONS Serum IL-33 is closely associated with the development and severity of BPD. Anti-inflammatory therapy with DART regimen can decrease the serum level of IL-33.
Collapse
Affiliation(s)
- Jun-Long Chen
- Department of Neonatology, Inner Mongolia People's Hospital, Huhhot 010017, China.
| | | |
Collapse
|
533
|
Noor-eldeen E, Hassan WA, Behiry EG, El-monem AEHA. Serum, synovial and mRNA expression of interleukin-33 in juvenile idiopathic arthritis patients: Potential role as a marker of disease activity and relation to musculoskeletal ultrasound. THE EGYPTIAN RHEUMATOLOGIST 2020; 42:225-230. [DOI: 10.1016/j.ejr.2020.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
534
|
The emerging roles of eosinophils in mucosal homeostasis. Mucosal Immunol 2020; 13:574-583. [PMID: 32157190 DOI: 10.1038/s41385-020-0281-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 02/04/2023]
Abstract
Eosinophils are granulocytes, typically implicated as end-stage effector cells in type-II immune responses. They are capable of producing a wide array of pre-formed molecules which render them with vast potential to influence a wide variety of processes. Nonetheless, eosinophil research has traditionally focused on their role in anti-helminthic responses and pathophysiological processes in type-II immune disorders, such as allergy and asthma, where eosinophilia is a hallmark phenotype. However, a number of key studies over the past decade have placed this restricted view of eosinophil function into question, presenting additional evidence for eosinophils as critical regulators of various homeostatic processes including immune maintenance, organ development, and tissue regeneration.
Collapse
|
535
|
Chauché C, Vacca F, Chia SL, Richards J, Gregory WF, Ogunkanbi A, Wear M, McSorley HJ. A Truncated Form of HpARI Stabilizes IL-33, Amplifying Responses to the Cytokine. Front Immunol 2020; 11:1363. [PMID: 32695116 PMCID: PMC7338556 DOI: 10.3389/fimmu.2020.01363] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/28/2020] [Indexed: 12/26/2022] Open
Abstract
The murine intestinal nematode Heligmosomoides polygyrus releases the H. polygyrus Alarmin Release Inhibitor (HpARI) - a protein which binds to IL-33 and to DNA, effectively tethering the cytokine in the nucleus of necrotic cells. Previous work showed that a non-natural truncation consisting of the first 2 domains of HpARI (HpARI_CCP1/2) retains binding to both DNA and IL-33, and inhibited IL-33 release in vivo. Here, we show that the affinity of HpARI_CCP1/2 for IL-33 is significantly lower than that of the full-length protein, and that HpARI_CCP1/2 lacks the ability to prevent interaction of IL-33 with its receptor. When HpARI_CCP1/2 was applied in vivo it potently amplified IL-33-dependent immune responses to Alternaria alternata allergen, Nippostrongylus brasiliensis infection and recombinant IL-33 injection, in direct contrast to the IL-33-suppressive effects of full-length HpARI. Mechanistically, we found that HpARI_CCP1/2 is able to bind to and stabilize IL-33, preventing its degradation and maintaining the cytokine in its active form. This study highlights the importance of IL-33 inactivation, the potential for IL-33 stabilization in vivo, and describes a new tool for IL-33 research.
Collapse
Affiliation(s)
- Caroline Chauché
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.,Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Francesco Vacca
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.,Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Shin Li Chia
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Josh Richards
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - William F Gregory
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Adefunke Ogunkanbi
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Martin Wear
- The Edinburgh Protein Production Facility (EPPF), Wellcome Trust Centre for Cell Biology (WTCCB), University of Edinburgh, Edinburgh, United Kingdom
| | - Henry J McSorley
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.,Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
536
|
Epithelium-derived IL-33 activates mast cells to initiate neutrophil recruitment following corneal injury. Ocul Surf 2020; 18:633-640. [PMID: 32615259 DOI: 10.1016/j.jtos.2020.06.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/26/2020] [Accepted: 06/10/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE Neutrophils play a critical role in defending against threats such as microbial infection, yet their activation during innate immune response incites collateral damage to healthy tissues. We have previously shown that corneal injury induces mast cells to express the neutrophil chemoattractant CXCL2. Here we delineate the mechanism of injury-induced, non-IgE-mediated mast cell activation at the ocular surface. METHODS Corneal injury was induced by mechanical removal of the epithelium and anterior stroma in mast cell deficient (cKitW-sh) and C57BL/6 mice using Algerbrush II. Corneas were analyzed for frequencies of total CD45+ inflammatory cells, CD11b+Ly6G+ neutrophils, and cKit+FcεR1+ mast cells using flow cytometry. Mast cells were stimulated with different inflammatory factors known to increase during corneal injury (IL-33, IL-1β, IL-36γ, IL-6, SDF1α and Substance P) and assessed for the secretion of β-hexosaminidase, tryptase and CXCL2 using ELISA. IL-33 neutralizing antibody (1 mg/ml) was administered locally for mast cell inhibition in vivo. RESULTS Mast cell deficient mice failed to recruit early neutrophils to the injured corneas. IL-33 stimulation upregulated CXCL2 secretion by mast cells. Corneal injury resulted in amplified expression of IL-33 at the cornea and epithelium was identified as its primary source. Topical neutralization of IL-33 at the ocular surface inhibited mast cell activation, limited neutrophil infiltration, and reduced corneal inflammatory haze, normalizing tissue architecture following ocular injury. CONCLUSIONS These data implicate IL-33 in mast cell activation and early neutrophil recruitment in non-allergic inflammation, suggesting IL-33 as a potential therapeutic target in inflammatory disorders of the ocular surface.
Collapse
|
537
|
Tajima H, Tajiki-Nishino R, Watanabe Y, Kurata K, Fukuyama T. Activation of aryl hydrocarbon receptor by benzo[a]pyrene increases interleukin 33 expression and eosinophil infiltration in a mouse model of allergic airway inflammation. J Appl Toxicol 2020; 40:1545-1553. [PMID: 32557721 DOI: 10.1002/jat.4017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/05/2020] [Accepted: 05/16/2020] [Indexed: 12/15/2022]
Abstract
We recently demonstrated that benzo[a]pyrene (BaP), the aryl hydrocarbon receptor (AhR) ligand, directly contributes to aggravation of cutaneous allergy in a mouse model of allergic dermatitis. The present study aimed to determine whether BaP-induced AhR activation results in development of airway inflammation. Initially, the potential for a direct relationship between BaP-induced AhR activation and airway inflammation was investigated in vivo, using a mouse model of type 2 helper T cell (Th2) hapten toluene-2,4-diisocyanate (TDI)-induced airway inflammation. Mice were orally administered BaP at 48, 24, and 4 h before the final allergen challenge. Oral administration of BaP showed a significant increase in lung inflammation and eosinophil infiltration. While expression of Th2 cytokines such as interleukin 4 (IL-4) and IL-13 was not affected by exposure to BaP, AhR activation significantly increased IL-33 expression. To confirm the in vivo results, in vitro experiments were performed using the human eosinophilic leukemia cell line (EOL-1), human bronchial epithelial cell line (BEAS-2B), and human lung adenocarcinoma epithelial cell line (A549). Results indicated that pre-treatment with BaP increased expression of IL-8 in house dust mite-activated EOL-1, BEAS-2B, and A549 cells. In addition, IL-33 levels in BEAS-2B cells were significantly increased after BaP exposure. Our findings indicated that BaP-induced AhR activation is involved in the pro-inflammatory response in respiratory allergy, and that this effect may be mediated by increased IL-33 expression and eosinophil infiltration.
Collapse
Affiliation(s)
- Hitoshi Tajima
- Toxicology Division, Institute of Environmental Toxicology, Ibaraki, Japan
| | | | - Yuko Watanabe
- Toxicology Division, Institute of Environmental Toxicology, Ibaraki, Japan
| | - Keigo Kurata
- Institute of Tokyo Environmental Allergy, ITEA Inc., Tokyo, Japan
| | - Tomoki Fukuyama
- Toxicology Division, Institute of Environmental Toxicology, Ibaraki, Japan.,Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Azabu University, Sagamihara-shi, Kanagawa, Japan
| |
Collapse
|
538
|
Zhou Z, Yan F, Liu O. Interleukin (IL)-33: an orchestrator of immunity from host defence to tissue homeostasis. Clin Transl Immunology 2020; 9:e1146. [PMID: 32566227 PMCID: PMC7299676 DOI: 10.1002/cti2.1146] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
Interleukin (IL)-33, a member of the IL-1 superfamily, functions as an alarm signal, which is released upon cell injury or tissue damage to alert the immune system. It has emerged as a chief orchestrator in immunity and has a broad pleiotropic action that influences differentiation, maintenance and function of various immune cell types via the ST2 receptor. Although it has been strongly associated with immunopathology, critically, IL-33 is involved in host defence, tissue repair and homeostasis. In this review, we provide an overview of the signalling pathway of IL-33 and highlight its regulatory functions in immune cells. Furthermore, we attempt a broader discussion of the emerging functions of IL-33 in host defence, tissue repair, metabolism, inflammatory disease and cancer, suggesting potential avenues to manoeuvre IL-33/ST2 signalling as treatment options.
Collapse
Affiliation(s)
- Zekun Zhou
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology Central South University Changsha Hunan China
| | - Fei Yan
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology Central South University Changsha Hunan China
| | - Ousheng Liu
- Hunan Key Laboratory of Oral Health Research & Hunan 3D Printing Engineering Research Center of Oral Care & Hunan Clinical Research Center of Oral Major Diseases and Oral Health & Xiangya Stomatological Hospital & Xiangya School of Stomatology Central South University Changsha Hunan China
| |
Collapse
|
539
|
Kienzl M, Hasenoehrl C, Valadez-Cosmes P, Maitz K, Sarsembayeva A, Sturm E, Heinemann A, Kargl J, Schicho R. IL-33 reduces tumor growth in models of colorectal cancer with the help of eosinophils. Oncoimmunology 2020; 9:1776059. [PMID: 32923137 PMCID: PMC7458617 DOI: 10.1080/2162402x.2020.1776059] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In many types of cancer, presence of eosinophils in tumors correlate with an improved disease outcome. In line with this, activated eosinophils have been shown to reduce tumor growth in colorectal cancer (CRC). Interleukin (IL)-33 has recently emerged as a cytokine that is able to inhibit the development of tumors through eosinophils and other cells of the tumor microenvironment thereby positively influencing disease progress. Here, we asked whether eosinophils are involved in the effects of IL-33 on tumor growth in CRC. In models of CT26 cell engraftment and colitis-associated CRC, tumor growth was reduced after IL-33 treatment. The growth reduction was absent in eosinophil-deficient ΔdblGATA-1 mice but was restored by adoptive transfer of ex vivo-activated eosinophils indicating that the antitumor effect of IL-33 depends on the presence of eosinophils. In vitro, IL-33 increased the expression of markers of activation and homing in eosinophils, such as CD11b and Siglec-F, and the degranulation markers CD63 and CD107a. Increased expression of Siglec-F, CD11b and CD107a was also seen in vivo in eosinophils after IL-33 treatment. Viability and cytotoxic potential of eosinophils and their migration properties toward CCL24 were enhanced indicating direct effects of IL-33 on eosinophils. IL-33 treatment led to increased levels of IL-5 and CCL24 in tumors. Our data show that the presence of eosinophils is mandatory for IL-33-induced tumor reduction in models of CRC and that the mechanisms include eosinophil recruitment, activation and degranulation. Our findings also emphasize the potential use of IL-33 as an adjuvants in CRC immunotherapy. Abbreviations AOM: azoxymethane; bmRPMI: bone marrow RPMI; CRC: colorectal cancer; CFSE: carboxyfluorescein succinimidyl ester; DSS: dextran sulfate sodium; EPX: eosinophil peroxidase; INF-γ: interferon gamma; ILC: innate lymphoid cell; IL-33: interleukin-33; IL-5: interleukin-5; MDSC: myeloid derived suppressor cells; NK cells: natural killer cells; P/S: penicillin/streptomycin; rm: recombinant mouse; T regs: regulatory T cells; TATE: tumor associated tissue eosinophilia; TNF-α: tumor necrosis factor alpha
Collapse
Affiliation(s)
- Melanie Kienzl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria.,BioTechMed, Graz, Austria
| | - Carina Hasenoehrl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Paulina Valadez-Cosmes
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Kathrin Maitz
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Arailym Sarsembayeva
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Eva Sturm
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria.,BioTechMed, Graz, Austria
| | - Julia Kargl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria
| | - Rudolf Schicho
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Graz, Austria.,BioTechMed, Graz, Austria
| |
Collapse
|
540
|
Li YQ, Zhong Y, Xiao XP, Li DD, Zhou Z, Tian YY. IL-33/ST2 axis promotes the inflammatory response of nasal mucosal epithelial cells through inducing the ERK1/2 pathway. Innate Immun 2020; 26:505-513. [PMID: 32456598 PMCID: PMC7491240 DOI: 10.1177/1753425920918911] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Allergic rhinitis (AR) is a nasal mucosal inflammatory disease mediated by environmental allergens. At present, the relationship between the IL-33/ST2 axis, ERK1/2 pathway and AR progression needs further exploration. In our study, an AR model was constructed in vitro by treating HNEpC cells with Der p1. qRT-PCR was applied to assess the mRNA levels of IL-33, ST2, TNF-α, IL-6, and IL-8. Western blotting was used to measure the protein levels of IL-33, ST2, and the downstream proteins p-ERK1/2, ERK1/2, p-RSK, and RSK. IL-6, IL-8, IL-33, and TNF-α protein levels in cell supernatants were evaluated by ELISA. Flow cytometry was performed to check cell apoptosis of HNEpC in the presence or absence of Der p1. Our results indicate that the relative levels of IL-33, ST2, TNF-α, IL-6, and IL-8 were increased significantly in the AR model group. The above effects were notably reversed after transfection with shIL-33 or shST2. IL-33 stimulation further resulted in the increase in both ST2 and inflammation-associated cytokines, and these effects were restored after shST2 treatment. Also, the levels of inflammatory factors induced by IL-33 stimulation or ST2 overexpression were reversed after applying an ERK1/2 pathway blocker. In conclusion, IL-33/ST2 mediated inflammation of nasal mucosal epithelial cells by inducing the ERK1/2 pathway.
Collapse
Affiliation(s)
- Yun-Qiu Li
- Department of Otolaryngology Head and Neck Surgery, Hunan Provincial People's Hospital, P.R. China
| | - Yu Zhong
- Department of Otolaryngology Head and Neck Surgery, Hunan Provincial People's Hospital, P.R. China
| | - Xu-Ping Xiao
- Department of Otolaryngology Head and Neck Surgery, Hunan Provincial People's Hospital, P.R. China
| | - Dan-Dan Li
- Department of Otolaryngology Head and Neck Surgery, Hunan Provincial People's Hospital, P.R. China
| | - Zheng Zhou
- Department of Otolaryngology Head and Neck Surgery, Hunan Provincial People's Hospital, P.R. China
| | - Yan-Yan Tian
- Chronic Disease Management Center of Hunan Provincial People's Hospital, P.R. China
| |
Collapse
|
541
|
Boberg E, Johansson K, Malmhäll C, Weidner J, Rådinger M. House Dust Mite Induces Bone Marrow IL-33-Responsive ILC2s and T H Cells. Int J Mol Sci 2020; 21:E3751. [PMID: 32466530 PMCID: PMC7312993 DOI: 10.3390/ijms21113751] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 01/05/2023] Open
Abstract
Type 2 innate lymphoid cells (ILC2s) and their adaptive counterpart type 2 T helper (TH2) cells respond to interleukin-33 (IL-33) by producing IL-5, which is a crucial cytokine for eosinophil development in the bone marrow. The aim of this study was to determine if bone marrow ILC2s, TH cells, and eosinophils are locally regulated by IL-33 in terms of number and activation upon exposure to the common aeroallergen house dust mite (HDM). Mice that were sensitized and challenged with HDM by intranasal exposures induced eosinophil development in the bone marrow with an initial increase of IL5Rα+ eosinophil progenitors, following elevated numbers of mature eosinophils and the induction of airway eosinophilia. Bone marrow ILC2s, TH2, and eosinophils all responded to HDM challenge by increased IL-33 receptor (ST2) expression. However, only ILC2s, but not TH cells, revealed increased ST2 expression at the onset of eosinophil development, which significantly correlated with the number of eosinophil progenitors. In summary, our findings suggest that airway allergen challenges with HDM activates IL-33-responsive ILC2s, TH cells, and eosinophils locally in the bone marrow. Targeting the IL-33/ST2 axis in allergic diseases including asthma may be beneficial by decreasing eosinophil production in the bone marrow.
Collapse
Affiliation(s)
| | | | | | | | - Madeleine Rådinger
- Krefting Research Centre, Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-40530 Gothenburg, Sweden; (E.B.); (K.J.); (C.M.); (J.W.)
| |
Collapse
|
542
|
Chen Z, Hu Y, Gong Y, Zhang X, Cui L, Chen R, Yu Y, Yu Q, Chen Y, Diao H, Chen J, Wang Y, Shi Y. Interleukin-33 alleviates psoriatic inflammation by suppressing the T helper type 17 immune response. Immunology 2020; 160:382-392. [PMID: 32306382 DOI: 10.1111/imm.13203] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 03/04/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease with unclear pathogenesis. Interleukin-33 (IL-33) is highly expressed in patients with psoriasis, but its role in psoriasis is unknown. The aim of this study was to investigate the possible role of IL-33 in the pathogenesis and treatment of psoriasis. IL-33 expression was determined using enzyme-linked immunosorbent assay, real-time fluorescent quantitative polymerase chain reaction and immunohistochemical staining. CD4+ T cells were sorted using magnetic beads and treated with or without IL-33. Imiquimod (IMQ) was used to induce psoriatic inflammation in mice. The frequency of immune cells was determined using flow cytometry. The cytokine level in mouse skin was measured using cytometric bead array. Our results showed that IL-33 was highly expressed in the lesional skin and serum of patients with moderate-to-severe plaque psoriasis. IL-33 inhibited the expression of IL-17 in CD4+ T cells of psoriasis patients. Subcutaneous injection of IL-33 alleviated the IMQ-induced psoriatic inflammation in mice, reduced tumor necrosis factor-α and IL-23 expression, and decreased the proportion of T helper type 17 (Th17) cells in the skin-draining lymph nodes in the mice. Our results suggest that IL-33 plays a protective role in the pathogenesis of psoriasis by suppressing Th17 cell differentiation and function. The potential therapeutic effect of IL-33 in treating psoriasis warrants further investigation.
Collapse
Affiliation(s)
- Zeyu Chen
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Yifan Hu
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Yu Gong
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Xilin Zhang
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China.,Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lian Cui
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Rongfen Chen
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Yingyuan Yu
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Qian Yu
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Youdong Chen
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Hongyue Diao
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Jia Chen
- Physical Examination Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanyuan Wang
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Yuling Shi
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
543
|
Rao UK, Engelhardt BG. Predicting Immuno-Metabolic Complications After Allogeneic Hematopoietic Cell Transplant with the Cytokine Interleukin-33 (IL-33) and its Receptor Serum-Stimulation 2 (ST2). Clin Hematol Int 2020; 2:101-108. [PMID: 34595450 PMCID: PMC8432328 DOI: 10.2991/chi.d.200506.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/04/2020] [Indexed: 01/19/2023] Open
Abstract
Patients undergoing allogeneic hematopoietic cell transplantation (HCT) are at risk for numerous acute and long-term complications from this procedure. Post-transplant diabetes mellitus (PTDM) is a common but under-recognized problem. Similar to graft-versus-host disease (GVHD), new-onset diabetes is characterized by immune dysregulation that can negatively impact transplant outcomes. This review will discuss the biology of IL-33/ST2 in acute GVHD and PTDM development, and how this cytokine axis could be leveraged for predicting and treating immuno-metabolic complications after transplant.
Collapse
Affiliation(s)
- Uttam K Rao
- Department of Medicine, Vanderbilt University Medical Center, Medical Center Drive, Nashville, TN, USA
| | - Brian G Engelhardt
- Department of Medicine, Vanderbilt University Medical Center, Medical Center Drive, Nashville, TN, USA
| |
Collapse
|
544
|
Ryffel B, Alves-Filho JC. ILC2s and Basophils Team Up to Orchestrate IL-33-Induced Atopic Dermatitis. J Invest Dermatol 2020; 139:2077-2079. [PMID: 31543211 DOI: 10.1016/j.jid.2019.06.118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/06/2019] [Accepted: 06/10/2019] [Indexed: 01/14/2023]
Abstract
In this issue, Imai et al. (2019) provide new insights into the pathophysiology of AD-like inflammation using their model (Imai et al., 2013) and ask how ILC2s and basophils contribute to the IL-33-induced AD-like inflammation. Their findings show that continuous expression of IL-33 in keratinocytes is sufficient to cause AD-like inflammation in mice, and that this occurrence is largely independent of adaptive immune cells and is mediated by basophils and ILC2s.
Collapse
Affiliation(s)
- Bernhard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics(INEM), UMR 7355 CNRS-University of Orleans, Orleans, France.
| | - José Carlos Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
545
|
Li N, Yu Y, Chen X, Gao S, Zhang Q, Xu C. Bifidobacterium breve M-16V alters the gut microbiota to alleviate OVA-induced food allergy through IL-33/ST2 signal pathway. J Cell Physiol 2020; 235:9464-9473. [PMID: 32394447 DOI: 10.1002/jcp.29751] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/21/2020] [Indexed: 12/23/2022]
Abstract
There has been a marked increase in life-threatening food allergy (FA). One hypothesis is that changes in bacterial communities may be key to FA. To better understand how gut microbiota regulates FA in humans, we established a mouse model with FA induced by ovalbumin. We found that the mice with FA had abnormal bacterial composition, accompanied by increased immunoglobulin G, immunoglobulin E, and interleukin-4/interferon-γ, and there existed a certain coherence between them. Interestingly, Bifidobacterium breve M-16V may alter the gut microbiota to alleviate the allergy symptoms by IL-33/ST2 signaling. Our results indicate that gut microbiota is essential for regulating FA to dietary antigens and demonstrate that intervention in bacterial community regulation may be therapeutically related to FA.
Collapse
Affiliation(s)
- Na Li
- Department of Pediatric, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of tropical medicine, Hainan Medical University, HaiKou, China
| | - Yi Yu
- Department of Pediatric, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuehua Chen
- Department of Pediatric, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shenshen Gao
- Department of Pediatric, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingqing Zhang
- Department of Pediatric, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chundi Xu
- Department of Pediatric, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
546
|
Norlander AE, Peebles RS. Innate Type 2 Responses to Respiratory Syncytial Virus Infection. Viruses 2020; 12:E521. [PMID: 32397226 PMCID: PMC7290766 DOI: 10.3390/v12050521] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/07/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a common and contagious virus that results in acute respiratory tract infections in infants. In many cases, the symptoms of RSV remain mild, however, a subset of individuals develop severe RSV-associated bronchiolitis. As such, RSV is the chief cause of infant hospitalization within the United States. Typically, the immune response to RSV is a type 1 response that involves both the innate and adaptive immune systems. However, type 2 cytokines may also be produced as a result of infection of RSV and there is increasing evidence that children who develop severe RSV-associated bronchiolitis are at a greater risk of developing asthma later in life. This review summarizes the contribution of a newly described cell type, group 2 innate lymphoid cells (ILC2), and epithelial-derived alarmin proteins that activate ILC2, including IL-33, IL-25, thymic stromal lymphopoietin (TSLP), and high mobility group box 1 (HMGB1). ILC2 activation leads to the production of type 2 cytokines and the induction of a type 2 response during RSV infection. Intervening in this innate type 2 inflammatory pathway may have therapeutic implications for severe RSV-induced disease.
Collapse
Affiliation(s)
| | - R. Stokes Peebles
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN 37232-2650, USA;
| |
Collapse
|
547
|
Zhao XY, Zhou L, Chen Z, Ji Y, Peng X, Qi L, Li S, Lin JD. The obesity-induced adipokine sST2 exacerbates adipose T reg and ILC2 depletion and promotes insulin resistance. SCIENCE ADVANCES 2020; 6:eaay6191. [PMID: 32426492 PMCID: PMC7220368 DOI: 10.1126/sciadv.aay6191] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 03/02/2020] [Indexed: 05/02/2023]
Abstract
Depletion of fat-resident regulatory T cells (Tregs) and group 2 innate lymphoid cells (ILC2s) has been causally linked to obesity-associated insulin resistance. However, the molecular nature of the pathogenic signals suppress adipose Tregs and ILC2s in obesity remains unknown. Here, we identified the soluble isoform of interleukin (IL)-33 receptor ST2 (sST2) as an obesity-induced adipokine that attenuates IL-33 signaling and disrupts Treg/ILC2 homeostasis in adipose tissue, thereby exacerbates obesity-associated insulin resistance in mice. We demonstrated sST2 is a target of TNFα signaling in adipocytes that is countered by Zbtb7b. Fat-specific ablation of Zbtb7b augments adipose sST2 gene expression, leading to diminished fat-resident Tregs/ILC2s, more pronounced adipose tissue inflammation and fibrosis, and impaired glucose homeostasis in mice. Mechanistically, Zbtb7b suppresses NF-κB activation in response to TNFα through destabilizing IκBα. These findings uncover an adipokine-immune signaling pathway that is engaged in obesity to drive the pathological changes of the immunometabolic landscape.
Collapse
Affiliation(s)
- Xu-Yun Zhao
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- Corresponding author. (J.D.L.); (X.-Y.Z.)
| | - Linkang Zhou
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Zhimin Chen
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yewei Ji
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Xiaoling Peng
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Ling Qi
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Siming Li
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jiandie D. Lin
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
- Corresponding author. (J.D.L.); (X.-Y.Z.)
| |
Collapse
|
548
|
Liu T, Yang L, Han X, Ding X, Li J, Yang J. Local sympathetic innervations modulate the lung innate immune responses. SCIENCE ADVANCES 2020; 6:eaay1497. [PMID: 32426489 PMCID: PMC7220323 DOI: 10.1126/sciadv.aay1497] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 02/28/2020] [Indexed: 05/14/2023]
Abstract
Local immunity of the lung needs to be under tight control. However, how efferent neural signals influence lung immunity remains incompletely understood. Here, we report the development of a modified iDISCO-based protocol, iDISCO(ace), for whole-tissue 3D assessment of neural innervations and immune reactions in intact, unsectioned lung tissues. We observed that genetic, pharmacologic, or surgical removal of local sympathetic innervations promoted LPS-elicited innate immune response in the lung. Also, sympathetic ablation enhanced IL-33-elicited type 2 innate immunity. We further show that the sympathetic neurotransmitter norepinephrine, or specific agonists of the β2-adrenergic receptor, can inhibit the LPS- or IL-33-elicited immune response in a cell-intrinsic manner. Moreover, genetic deletion of the β2-adrenergic receptor produced immunomodulatory effects similar to those observed with sympathetic ablation. Together, this study elucidates the critical function of local sympathetic innervations in negatively modulating the lung innate immune responses.
Collapse
Affiliation(s)
- Tingting Liu
- Center for Life Sciences, Peking University, Beijing 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Lu Yang
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Xiangli Han
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Xiaofan Ding
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Jiali Li
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Jing Yang
- Center for Life Sciences, Peking University, Beijing 100871, China
- School of Life Sciences, Peking University, Beijing 100871, China
- IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
- State Key Laboratory of Membrane Biology, Peking University, Beijing 100871, China
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518055, China
- Corresponding author.
| |
Collapse
|
549
|
The Role of ST2 Receptor in the Regulation of Brucella abortus Oral Infection. Pathogens 2020; 9:pathogens9050328. [PMID: 32353980 PMCID: PMC7281115 DOI: 10.3390/pathogens9050328] [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: 01/27/2020] [Revised: 04/15/2020] [Accepted: 04/25/2020] [Indexed: 01/18/2023] Open
Abstract
The ST2 receptor plays an important role in the gut such as permeability regulation, epithelium regeneration, and promoting intestinal immune modulation. Here, we studied the role of ST2 receptor in a murine model of oral infection with Brucella abortus, its influence on gut homeostasis and control of bacterial replication. Balb/c (wild-type, WT) and ST2 deficient mice (ST2−/−) were infected by oral gavage and the results were obtained at 3 and 14 days post infection (dpi). Our results suggest that ST2−/− are more resistant to B. abortus infection, as a lower bacterial colony-forming unit (CFU) was detected in the livers and spleens of knockout mice, when compared to WT. Additionally, we observed an increase in intestinal permeability in WT-infected mice, compared to ST2−/− animals. Breakage of the intestinal epithelial barrier and bacterial dissemination might be associated with the presence of the ST2 receptor; since, in the knockout mice no change in intestinal permeability was observed after infection. Together with enhanced resistance to infection, ST2−/− produced greater levels of IFN-γ and TNF-α in the small intestine, compared to WT mice. Nevertheless, in the systemic model of infection ST2 plays no role in controlling Brucella replication in vivo. Our results suggest that the ST2 receptor is involved in the invasion process of B. abortus by the mucosa in the oral infection model.
Collapse
|
550
|
IL-33 Inhibits Hepatitis B Virus through Its Receptor ST2 in Hydrodynamic HBV Mouse Model. Mediators Inflamm 2020; 2020:1403163. [PMID: 32410845 PMCID: PMC7204199 DOI: 10.1155/2020/1403163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/08/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023] Open
Abstract
Interleukin-33 has been demonstrated to be associated with liver damage. However, its potential value in hepatitis B virus (HBV) infection remains unknown. This study was designed to investigate the role of IL-33 in hydrodynamic HBV mouse model. Different doses of IL-33 were used to treat HBV wild-type, ST2 knockout, CD8+ T depletion, NK depletion C57BL/6 mice and C.B-17 SCID and nod SCID mouse, respectively. The concentrations of HBV DNA, HBsAg, HBeAg, and molecules related to liver function were detected in the collected serum at different time points from model mice. Intrahepatic HBcAg was visualized by immunohistochemical staining of liver tissues. In vitro, hepG2 cells were transfected with pAAV-HBV 1.2, then treated with IL-33. The results showed that IL-33 significantly reduced HBV DNA and HBsAg in a dose-dependent manner in HBV wild-type mice. However, in the IL-33 specific receptor ST2 knockout mice, their antiviral effects could not be exerted. Through immunodeficient animal models and in vivo immune cell depletion mouse model, we found that IL-33 could not play antiviral effects without NK cells. Moreover, IL-33 could reduce the levels of HBsAg and HBeAg in the supernatant of HBV-transfected hepG2 cells in vitro. Our study revealed that IL-33 could inhibit HBV through ST2 receptor in the HBV mouse model, and this effect can be impaired without NK cell. Additionally, IL-33 had the direct anti-HBV effect in vitro, indicating that IL-33 could be a potent inducer of HBV clearance and a promising drug candidate.
Collapse
|