51
|
Ihim SA, Abubakar SD, Zian Z, Sasaki T, Saffarioun M, Maleknia S, Azizi G. Interleukin-18 cytokine in immunity, inflammation, and autoimmunity: Biological role in induction, regulation, and treatment. Front Immunol 2022; 13:919973. [PMID: 36032110 PMCID: PMC9410767 DOI: 10.3389/fimmu.2022.919973] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/25/2022] [Indexed: 12/13/2022] Open
Abstract
Interleukin-18 (IL-18) is a potent pro-inflammatory cytokine involved in host defense against infections and regulates the innate and acquired immune response. IL-18 is produced by both hematopoietic and non-hematopoietic cells, including monocytes, macrophages, keratinocytes and mesenchymal cell. IL-18 could potentially induce inflammatory and cytotoxic immune cell activities leading to autoimmunity. Its elevated levels have been reported in the blood of patients with some immune-related diseases, including rheumatoid arthritis, systemic lupus erythematosus, type I diabetes mellitus, atopic dermatitis, psoriasis, and inflammatory bowel disease. In the present review, we aimed to summarize the biological properties of IL-18 and its pathological role in different autoimmune diseases. We also reported some monoclonal antibodies and drugs targeting IL-18. Most of these monoclonal antibodies and drugs have only produced partial effectiveness or complete ineffectiveness in vitro, in vivo and human studies. The ineffectiveness of these drugs targeting IL-18 may be largely due to the loophole caused by the involvement of other cytokines and proteins in the signaling pathway of many inflammatory diseases besides the involvement of IL-18. Combination drug therapies, that focus on IL-18 inhibition, in addition to other cytokines, are highly recommended to be considered as an important area of research that needs to be explored.
Collapse
Affiliation(s)
- Stella Amarachi Ihim
- Department of Molecular and Cellular Pharmacology, University of Shizuoka, Shizuoka, Japan
- Department of Pharmacology and Toxicology, University of Nigeria, Nsukka, Nigeria
- Department of Science Laboratory Technology, University of Nigeria, Nsukka, Nigeria
| | - Sharafudeen Dahiru Abubakar
- Division of Molecular Pathology, Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan
- Department of Medical Laboratory Science, College of Medical Science, Ahmadu Bello University, Zaria, Nigeria
| | - Zeineb Zian
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Takanori Sasaki
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Mohammad Saffarioun
- Biopharmaceutical Research Center, AryoGen Pharmed Inc., Alborz University of Medical Sciences, Karaj, Iran
| | - Shayan Maleknia
- Biopharmaceutical Research Center, AryoGen Pharmed Inc., Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
- *Correspondence: Gholamreza Azizi,
| |
Collapse
|
52
|
Cao MC, Scotter EL. Novel and known transcriptional targets of ALS/FTD protein TDP-43: Meta-analysis and interactive graphical database. Dis Model Mech 2022; 15:276263. [PMID: 35946434 PMCID: PMC9509890 DOI: 10.1242/dmm.049418] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 07/26/2022] [Indexed: 11/20/2022] Open
Abstract
TDP-43 proteinopathy is the major pathology in amyotrophic lateral sclerosis (ALS) and tau-negative frontotemporal dementia (FTD). Mounting evidence implicates loss of normal TDP-43 RNA processing function as a key pathomechanism. However, the RNA targets of TDP-43 differ by report, and have never been formally collated or compared between models and disease, hampering understanding of TDP-43 function. Here, we conducted re-analysis and meta-analysis of publicly available RNA-sequencing datasets from six TDP-43-knockdown models, and TDP-43-immunonegative neuronal nuclei from ALS/ FTD brain, to identify differentially expressed genes (DEGs) and exon usage (DEU) events. There was little overlap in DEGs between knockdown models, but PFKP, STMN2, CFP, KIAA1324 and TRHDE were common targets and were also differentially expressed in TDP-43-immunonegative neurons. DEG enrichment analysis revealed diverse biological pathways including immune and synaptic functions. Common DEU events in human datasets included well-known targets POLDIP3 and STMN2, and novel targets EXD3, MMAB, DLG5 and GOSR2. Our interactive database https://phpstack-449938-2576646.cloudwaysapps.com/ allows further exploration of TDP-43 DEG and DEU targets. Together, these data identify TDP-43 targets that can be exploited therapeutically or to validate loss-of-function processes.
Collapse
Affiliation(s)
- Maize C Cao
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand. 3A Symonds Street, Auckland 1010, New Zealand
| | - Emma L Scotter
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand. 3A Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
53
|
Caspase-1 and interleukin-18 in children with post infectious bronchiolitis obliterans: a case-control study. Eur J Pediatr 2022; 181:3093-3101. [PMID: 35705877 DOI: 10.1007/s00431-022-04528-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/18/2022] [Accepted: 06/09/2022] [Indexed: 11/27/2022]
Abstract
UNLABELLED The exact immunological mechanisms of post infectious bronchiolitis obliterans (PIBO) in childhood are not fully known. It has been shown that the inflammasome and IL-18 pathway play important roles in the pathogenesis of lung fibrosis. We aimed to investigate the role of caspase-1, IL-18, and IL-18 components in PIBO. From January to May 2020, children with PIBO, children with history of influenza infection without PIBO, and healthy children were asked to participate in the study in three pediatric pulmonology centers. Serum caspase-1, IL-18, IL-18BP, IL-18R, and INF-γ levels were measured by ELISA and compared between the 3 groups. There were 21 children in the PIBO group, 16 children in the influenza group, and 39 children in the healthy control group. No differences in terms of age and gender between the 3 groups were found. IL-18 and IL-18BP levels were higher in the healthy control group (p = 0.018, p = 0.005, respectively). IL-18R was higher in the PIBO group (p = 0.001) and caspase-1 was higher in the PIBO and influenza group than the healthy control group (p = 0.002). IFN-γ levels did not differ between the 3 groups. IL-18BP/IL-18 was higher in the influenza group than the PIBO group and the healthy control group (p = 0.003). CONCLUSIONS Caspase-1 level was increased in patients with PIBO which suggests that inflammasome activation may have a role in fibrosis; however, IL-18 level was found to be low. Mediators other than IL-18 may be involved in the inflammatory pathway in PIBO. Further immunological studies investigating inflammasome pathway are needed for PIBO with chronic inflammation. WHAT IS KNOWN • Post infectious bronchiolitis obliterans (PIBO) is a rare, severe chronic lung disease during childhood which is associated with inflammation and fibrosis which lead to partial or complete luminal obstruction especially in small airways. • The exact immunological mechanisms of PIBO in childhood are not fully known. WHAT IS NEW • Inflammasome activation persists even years after acute infection and may play a role in fibrosis in PIBO. • Mediators other than IL-18 may be involved in these inflammatory pathway.
Collapse
|
54
|
Park SY, Hisham Y, Shin HM, Yeom SC, Kim S. Interleukin-18 Binding Protein in Immune Regulation and Autoimmune Diseases. Biomedicines 2022; 10:biomedicines10071750. [PMID: 35885055 PMCID: PMC9313042 DOI: 10.3390/biomedicines10071750] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/13/2022] [Accepted: 07/16/2022] [Indexed: 12/28/2022] Open
Abstract
Natural soluble antagonist and decoy receptor on the surface of the cell membrane are evolving as crucial immune system regulators as these molecules are capable of recognizing, binding, and neutralizing (so-called inhibitors) their targeted ligands. Eventually, these soluble antagonists and decoy receptors terminate signaling by prohibiting ligands from connecting to their receptors on the surface of cell membrane. Interleukin-18 binding protein (IL-18BP) participates in regulating both Th1 and Th2 cytokines. IL-18BP is a soluble neutralizing protein belonging to the immunoglobulin (Ig) superfamily as it harbors a single Ig domain. The Ig domain is essential for its binding to the IL-18 ligand and holds partial homology to the IL-1 receptor 2 (IL-1R2) known as a decoy receptor of IL-1α and IL-1β. IL-18BP was defined as a unique soluble IL-18BP that is distinct from IL-18Rα and IL-18Rβ chain. IL-18BP is encoded by a separated gene, contains 8 exons, and is located at chr.11 q13.4 within the human genome. In this review, we address the difference in the biological activity of IL-18BP isoforms, in the immunity balancing Th1 and Th2 immune response, its critical role in autoimmune diseases, as well as current clinical trials of recombinant IL-18BP (rIL-18BP) or equivalent.
Collapse
Affiliation(s)
- Seung Yong Park
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea;
| | - Yasmin Hisham
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea;
| | - Hyun Mu Shin
- System Immunology, Wide River Institute of Immunology, Collage of Medicine, Seoul National University, Hongcheon-gun 25159, Korea;
| | - Su Cheong Yeom
- Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang 25354, Korea;
| | - Soohyun Kim
- College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea;
- Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea;
- Correspondence: ; Tel.: +82-2-457-0868
| |
Collapse
|
55
|
Chen PK, Wey SJ, Chen DY. Interleukin-18: a biomarker with therapeutic potential in adult-onset Still's disease. Expert Rev Clin Immunol 2022; 18:823-833. [PMID: 35771972 DOI: 10.1080/1744666x.2022.2096592] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Adult-onset Still's disease (AOSD) is an autoinflammatory disease driven by the innate immune response. Given the ambiguity in clinical presentation and lack of specific diagnostic biomarkers, AOSD diagnosis is usually delayed in the early stage. Because AOSD is a rare disease with clinical heterogeneity, there is no consensus on its treatment currently. This review summarizes the current research evidence regarding the pathogenic role and the diagnostic or therapeutic potential of interleukin (IL)-18 in AOSD. AREAS COVERED We searched the MEDLINE database using the PubMed interface and reviewed English-language literature from 1971 to 2022. This review focusing on IL-18 discusses its pathogenic role and clinical implications in AOSD. EXPERT OPINION NLRP3-inflammasome activation with IL-18 overproduction plays a pathogenic role in AOSD. IL-18 is closely linked to the clinical manifestations and disease activity of AOSD and may be a diagnostic biomarker. Given its pathogenic role in AOSD, IL-18 could become a potential therapeutic target. IL-18 binding protein (IL-18BP) negatively regulates the biological activity of IL-18 by inhibiting IL-18 signaling, and a clinical trial revealed that IL-18BP (Tadekinig alfa) treatment was well-tolerated and effective for AOSD. Recently, monoclonal antibodies against IL-18 have been under evaluation in a phase 1b trial.
Collapse
Affiliation(s)
- Po-Ku Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan.,Translational Medicine Laboratory, Rheumatology and Immunology Center, Taichung, Taiwan
| | - Shiow-Jiuan Wey
- Division of Dermatology, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan.,Translational Medicine Laboratory, Rheumatology and Immunology Center, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,D. Program in Translational Medicine and Rong Hsing Research Center for Translational Medicine, National Chung Hsing UniversityPh., Taichung, Taiwan
| |
Collapse
|
56
|
Evavold CL, Kagan JC. Diverse Control Mechanisms of the Interleukin-1 Cytokine Family. Front Cell Dev Biol 2022; 10:910983. [PMID: 35832789 PMCID: PMC9272893 DOI: 10.3389/fcell.2022.910983] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/17/2022] [Indexed: 11/15/2022] Open
Abstract
The majority of interleukin-1 (IL-1) family cytokines lack amino terminal secretion signals or transmembrane domains for secretion along the conventional biosynthetic pathway. Yet, these factors must be translocated from the cytoplasm across the plasma membrane into the extracellular space in order to regulate inflammation. Recent work has identified an array of mechanisms by which IL-1 family cytokines can be released into the extracellular space, with supramolecular organizing centers known as inflammasomes serving as dominant drivers of this process. In this review, we discuss current knowledge of the mechanisms of IL-1 family cytokine synthesis, processing, and release from cells. Using this knowledge, we propose a model whereby host metabolic state dictates the route of IL-1β secretion, with implications for microbial infection and sterile inflammation.
Collapse
Affiliation(s)
- Charles L. Evavold
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
- *Correspondence: Charles L. Evavold, ; Jonathan C. Kagan,
| | - Jonathan C. Kagan
- Division of Gastroenterology, Boston Children’s Hospital and Harvard Medical School, Boston, MA, United States
- *Correspondence: Charles L. Evavold, ; Jonathan C. Kagan,
| |
Collapse
|
57
|
Makaremi S, Asgarzadeh A, Kianfar H, Mohammadnia A, Asghariazar V, Safarzadeh E. The role of IL-1 family of cytokines and receptors in pathogenesis of COVID-19. Inflamm Res 2022; 71:923-947. [PMID: 35751653 PMCID: PMC9243884 DOI: 10.1007/s00011-022-01596-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/29/2022] [Indexed: 12/12/2022] Open
Abstract
A global pandemic has erupted as a result of the new brand coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This pandemic has been consociated with widespread mortality worldwide. The antiviral immune response is an imperative factor in confronting the recent coronavirus disease 2019 (COVID-19) infections. Meantime, cytokines recognize as crucial components in guiding the appropriate immune pathways in the restraining and eradication of the virus. Moreover, SARS-CoV-2 can induce uncontrolled inflammatory responses characterized by hyper-inflammatory cytokine production, which causes cytokine storm and acute respiratory distress syndrome (ARDS). As excessive inflammatory responses are contributed to the severe stage of the COVID-19 disease, therefore, the pro-inflammatory cytokines are regarded as the Achilles heel during COVID-19 infection. Among these cytokines, interleukin (IL-) 1 family cytokines (IL-1, IL-18, IL-33, IL-36, IL-37, and IL-38) appear to have a strong inflammatory role in severe COVID-19. Hence, understanding the underlying inflammatory mechanism of these cytokines during infection is critical for reducing the symptoms and severity of the disease. Here, the possible mechanisms and pathways involved in inflammatory immune responses are discussed.
Collapse
Affiliation(s)
- Shima Makaremi
- School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran.,Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Asgarzadeh
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.,Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamed Kianfar
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.,Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Alireza Mohammadnia
- Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Vahid Asghariazar
- Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Department of Health Information Management, School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran. .,Department of Microbiology, Parasitology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
58
|
Generation of antagonistic monoclonal antibodies against the neoepitope of active mouse interleukin (IL)-18 cleaved by inflammatory caspases. Arch Biochem Biophys 2022; 727:109322. [PMID: 35709966 DOI: 10.1016/j.abb.2022.109322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 05/23/2022] [Accepted: 06/09/2022] [Indexed: 11/23/2022]
Abstract
Interleukin 18 (IL-18) is a member of the IL-1 family and plays an important role in both the innate and acquired immune systems. It is constitutively expressed as an inactive precursor (24 kDa) in various cell types, and the mature IL-18 (18 kDa) cleaved by inflammatory caspase-1/4 binds to the interleukin-18 receptor, thereby activating downstream signaling pathways. We previously generated anti-human IL-18 antibodies that specifically recognize the human IL-18 neoepitope cleaved by inflammatory caspase-1/4. Because the N-terminal amino acid sequences of the neoepitopes are different between human IL-18 and mouse IL-18, the anti-human IL-18 neoepitope antibodies do not recognize mouse mature IL-18. We have now generated novel anti-mouse IL-18 neoepitope antibodies. We also confirmed CXCL2 secretion from P-815 mouse cells by mouse IL-18 stimulation, and established a simple assay to evaluate the activity of mouse IL-18. Using this evaluation system, we confirmed that the anti-mouse IL-18 neoepitope antibodies could inhibit mouse IL-18. By demonstrating the therapeutic efficacy of the anti-mouse IL-18 neoepitope and function-blocking mAbs established in the present study in mouse models, corresponding to human inflammatory diseases in which IL-18 may be involved, such as inflammatory bowel diseases, we can provide the proof-of-concept that the previously established anti-human IL-18 neoepitope and function-blocking mAbs work in human inflammatory disorders corresponding to mouse models.
Collapse
|
59
|
Sun R, Gao DS, Shoush J, Lu B. The IL-1 family in tumorigenesis and antitumor immunity. Semin Cancer Biol 2022; 86:280-295. [DOI: 10.1016/j.semcancer.2022.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/24/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022]
|
60
|
Structural basis of human IL-18 sequestration by the decoy receptor IL-18 binding protein (IL-18BP) in inflammation and tumor immunity. J Biol Chem 2022; 298:101908. [PMID: 35398099 PMCID: PMC9111989 DOI: 10.1016/j.jbc.2022.101908] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 11/22/2022] Open
Abstract
Human Interleukin-18 (IL-18) is an omnipresent proinflammatory cytokine of the IL-1 family with central roles in autoimmune and inflammatory diseases and serves as a staple biomarker in the evaluation of inflammation in physiology and disease, including the inflammatory phase of COVID-19. The sequestration of IL-18 by its soluble decoy receptor IL-18-Binding Protein (IL-18BP) is critical to the regulation of IL-18 activity. Since an imbalance in expression and circulating levels of IL-18 is associated with disease, structural insights into how IL-18BP outcompetes binding of IL-18 by its cognate cell-surface receptors are highly desirable; however, the structure of human IL-18BP in complex with IL-18 has been elusive. Here, we elucidate the sequestration mechanism of human IL-18 mediated by IL-18BP based on the crystal structure of the IL-18:IL-18BP complex. These detailed structural snapshots reveal the interaction landscape leading to the ultra-high affinity of IL-18BP toward IL-18 and identify substantial differences with respect to previously characterized complexes of IL-18 with IL-18BP of viral origin. Furthermore, our structure captured a fortuitous higher-order assembly between IL-18 and IL-18BP coordinated by a disulfide-bond distal to the binding surface connecting IL-18 and IL-18BP molecules from different complexes, resulting in a novel tetramer with 2:2 stoichiometry. This tetrapartite assembly was found to restrain IL-18 activity more effectively than the canonical 1:1 complex. Collectively, our findings provide a framework for innovative, structure-driven therapeutic strategies and further functional interrogation of IL-18 in physiology and disease.
Collapse
|
61
|
Ohya T, Nishimura K, Murase A, Hattori S, Ohara A, Nozawa T, Hara R, Ito S. Impaired Interleukin-18 Signaling in Natural Killer Cells From Patients With Systemic Juvenile Idiopathic Arthritis. ACR Open Rheumatol 2022; 4:503-510. [PMID: 35275436 PMCID: PMC9190221 DOI: 10.1002/acr2.11426] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 01/07/2022] [Accepted: 02/01/2022] [Indexed: 12/03/2022] Open
Abstract
Objective Systemic juvenile idiopathic arthritis (sJIA) is characterized by fever, arthritis, rash, hepatosplenomegaly, and macrophage activation syndrome; however, its pathogenesis is still unclear. Elevated serum interleukin (IL)‐18 concentrations and decreased natural killer (NK) cell activity are characteristic of active disease; thus, we examined IL‐18 signaling in NK cells from sJIA. Methods We analyzed mitogen‐activated protein kinase (MAPK) p38 and nuclear factor κ light chain enhancer of activated B cells (NFκB) p65 phosphorylation in NK cells after in vitro recombinant IL‐18 (rIL‐18) stimulation in 31 patients with sJIA. Associations between clinical features, serum IL‐18, and phosphorylation intensity were analyzed. Furthermore, we investigated the effects of high IL‐18 concentrations on phosphorylation in NK cells. Results Patients were divided according to their disease activity: systemic features (n = 8), chronic arthritis (n = 7), remission on medication (n = 10), and remission off medication (n = 6). MAPK p38 and NFκB p65 phosphorylation intensity were the highest in healthy controls, followed by remission off medication, remission on medication (vs. control; MAPK p38, P < 0.01; NFκB p65, P < 0.05), chronic arthritis (P < 0.001, P < 0.001), and systemic features (P < 0.001, P < 0.001). The systemic features group showed a complete defect in phosphorylation. Serum IL‐18 was the highest in the systemic features group followed by chronic arthritis, remission on medication (P < 0.01), remission off medication (P < 0.01), and healthy controls (P < 0.01). Phosphorylation intensity was negatively correlated with serum IL‐18 (MAPK p38, r2 = 0.42; NFκB p65, r2 = 0.54). Furthermore, healthy control NK cells were cultured with rIL‐18; impaired phosphorylation was reproduced in vitro. Conclusion Impaired IL‐18 signaling in NK cells correlated with disease activity in sJIA. High serum IL‐18 exposure induces impaired MAPK and NFκB phosphorylation in NK cells.
Collapse
Affiliation(s)
- Takashi Ohya
- Yokohama City University, Yokohama, Kanagawa, Japan
| | | | - Ayako Murase
- Yokohama City University, Yokohama, Kanagawa, Japan
| | | | - Asami Ohara
- Yokohama City University, Yokohama, Kanagawa and Aichi Children's Health and Medical Center, Obu, Aichi, Japan
| | - Tomo Nozawa
- Yokohama City University, Yokohama, Kanagawa, Japan
| | - Ryoki Hara
- Yokohama City University and National Hospital Organization Yokohama Medical Center, Yokohama, Kanagawa, Japan
| | - Shuichi Ito
- Yokohama City University, Yokohama, Kanagawa, Japan
| |
Collapse
|
62
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
63
|
Prognostic tools and candidate drugs based on plasma proteomics of patients with severe COVID-19 complications. Nat Commun 2022; 13:946. [PMID: 35177642 PMCID: PMC8854716 DOI: 10.1038/s41467-022-28639-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
COVID-19 complications still present a huge burden on healthcare systems and warrant predictive risk models to triage patients and inform early intervention. Here, we profile 893 plasma proteins from 50 severe and 50 mild-moderate COVID-19 patients, and 50 healthy controls, and show that 375 proteins are differentially expressed in the plasma of severe COVID-19 patients. These differentially expressed plasma proteins are implicated in the pathogenesis of COVID-19 and present targets for candidate drugs to prevent or treat severe complications. Based on the plasma proteomics and clinical lab tests, we also report a 12-plasma protein signature and a model of seven routine clinical tests that validate in an independent cohort as early risk predictors of COVID-19 severity and patient survival. The risk predictors and candidate drugs described in our study can be used and developed for personalized management of SARS-CoV-2 infected patients. Prognostic markers for patients with COVID-19 are of critical importance in determining the course of SARS-CoV-2 infection and patient handling. Here the authors determine and apply a prognostic proteomic panel for risk and drug prediction in the management of SARS-CoV-2 infected patients.
Collapse
|
64
|
Supino D, Minute L, Mariancini A, Riva F, Magrini E, Garlanda C. Negative Regulation of the IL-1 System by IL-1R2 and IL-1R8: Relevance in Pathophysiology and Disease. Front Immunol 2022; 13:804641. [PMID: 35211118 PMCID: PMC8861086 DOI: 10.3389/fimmu.2022.804641] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022] Open
Abstract
Interleukin-1 (IL-1) is a primary cytokine of innate immunity and inflammation. IL-1 belongs to a complex family including ligands with agonist activity, receptor antagonists, and an anti-inflammatory cytokine. The receptors for these ligands, the IL-1 Receptor (IL-1R) family, include signaling receptor complexes, decoy receptors, and negative regulators. Agonists and regulatory molecules co-evolved, suggesting the evolutionary relevance of a tight control of inflammatory responses, which ensures a balance between amplification of innate immunity and uncontrolled inflammation. IL-1 family members interact with innate immunity cells promoting innate immunity, as well as with innate and adaptive lymphoid cells, contributing to their differentiation and functional polarization and plasticity. Here we will review the properties of two key regulatory receptors of the IL-1 system, IL-1R2, the first decoy receptor identified, and IL-1R8, a pleiotropic regulator of different IL-1 family members and co-receptor for IL-37, the anti-inflammatory member of the IL-1 family. Their complex impact in pathology, ranging from infections and inflammatory responses, to cancer and neurologic disorders, as well as clinical implications and potential therapeutic exploitation will be presented.
Collapse
Affiliation(s)
- Domenico Supino
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Luna Minute
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Italy
| | - Andrea Mariancini
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Italy
| | - Federica Riva
- Department of Veterinary Medicine, University of Milan, Milan, Italy
| | - Elena Magrini
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Cecilia Garlanda
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
65
|
Harel M, Fauteux-Daniel S, Girard-Guyonvarc'h C, Gabay C. Balance between Interleukin-18 and Interleukin-18 binding protein in auto-inflammatory diseases. Cytokine 2022; 150:155781. [DOI: 10.1016/j.cyto.2021.155781] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/03/2021] [Indexed: 02/07/2023]
|
66
|
Nimma S, Gu W, Manik MK, Ve T, Nanson JD, Kobe B. Crystal structure of the Toll/interleukin-1 receptor (TIR) domain of IL-1R10 provides structural insights into TIR domain signaling. FEBS Lett 2022; 596:886-897. [PMID: 35038778 DOI: 10.1002/1873-3468.14288] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/24/2021] [Accepted: 01/03/2022] [Indexed: 11/09/2022]
Abstract
The Toll/interleukin-1 receptor (TIR) domains are key innate immune signaling modules. Here, we present the crystal structure of the TIR domain of human Interleukin-1 receptor 10 (IL-1R10), also called IL-1RAPL2. It is similar to that of IL-1R9 (IL-1RAPL1) but shows significant structural differences to those from Toll-like receptors (TLRs) and the adaptor proteins MAL and MyD88. Interactions of TIR domains in their respective crystals and the higher-order assemblies (MAL and MyD88) reveal the presence of a common 'BCD surface', suggesting its functional significance. We also show that the TIR domains of IL-1R10 and IL-1R9 lack NADase activity, consistent with their structures. Our study provides a foundation for unraveling the functions of IL-1R9 and IL-1R10.
Collapse
Affiliation(s)
- Surekha Nimma
- The University of Queensland, School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre and Institute of Molecular Bioscience, Brisbane, Queensland, 4072, Australia
| | - Weixi Gu
- The University of Queensland, School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre and Institute of Molecular Bioscience, Brisbane, Queensland, 4072, Australia
| | - Mohammad K Manik
- The University of Queensland, School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre and Institute of Molecular Bioscience, Brisbane, Queensland, 4072, Australia
| | - Thomas Ve
- Griffith University, Institute for Glycomics, Southport, Queensland, 4222, Australia
| | - Jeffrey D Nanson
- The University of Queensland, School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre and Institute of Molecular Bioscience, Brisbane, Queensland, 4072, Australia
| | - Bostjan Kobe
- The University of Queensland, School of Chemistry and Molecular Biosciences, Australian Infectious Diseases Research Centre and Institute of Molecular Bioscience, Brisbane, Queensland, 4072, Australia
| |
Collapse
|
67
|
Girard-Guyonvarc’h C, Harel M, Gabay C. The Role of Interleukin 18/Interleukin 18-Binding Protein in Adult-Onset Still's Disease and Systemic Juvenile Idiopathic Arthritis. J Clin Med 2022; 11:jcm11020430. [PMID: 35054124 PMCID: PMC8781628 DOI: 10.3390/jcm11020430] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/02/2022] [Accepted: 01/10/2022] [Indexed: 02/06/2023] Open
Abstract
Interleukin 18 (IL-18) is a pro-inflammatory cytokine of the IL-1 family, whose activity is tightly controlled at the level of production, as well as signalization. Notably, it is buffered by its natural inhibitor, IL-18 binding protein (IL-18BP), which is massively present in circulation in normal and in most pathological conditions, thus preventing harmful pro-inflammatory systemic effects of IL-18. IL-18 has long been considered to be involved in the pathophysiology of various inflammatory diseases. However, a first clinical trial using recombinant IL-18BP for the treatment of rheumatoid arthritis and psoriasis gave disappointing results. Direct measurements of unbound, bioactive, free form of circulating IL-18 demonstrated that IL-18 was more specifically involved in adult-onset Still’s disease (AOSD) and systemic juvenile idiopathic arthritis (sJIA) but also in their most severe complication, macrophage activation syndrome (MAS). More importantly, administration of recombinant IL-18BP to patients with AOSD, and sJIA with MAS, showed promising results. This review summarizes available data regarding IL-18 and IL-18BP in AOSD and sJIA in mouse models and humans and shows the importance of IL-18/IL-18BP imbalance in these conditions, leading to the conclusion that IL-18, particularly free IL-18, may be a useful biomarker in these diseases and an interesting therapeutic target.
Collapse
Affiliation(s)
- Charlotte Girard-Guyonvarc’h
- Division of Rheumatology, Department of Medicine, University Hospital of Geneva, 1206 Geneva, Switzerland; (M.H.); (C.G.)
- Correspondence:
| | - Mathilde Harel
- Division of Rheumatology, Department of Medicine, University Hospital of Geneva, 1206 Geneva, Switzerland; (M.H.); (C.G.)
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Cem Gabay
- Division of Rheumatology, Department of Medicine, University Hospital of Geneva, 1206 Geneva, Switzerland; (M.H.); (C.G.)
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1206 Geneva, Switzerland
| |
Collapse
|
68
|
Calabrese L, Fiocco Z, Satoh TK, Peris K, French LE. Therapeutic potential of targeting IL-1 family cytokines in chronic inflammatory skin diseases. Br J Dermatol 2022; 186:925-941. [PMID: 34990008 DOI: 10.1111/bjd.20975] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/15/2021] [Accepted: 12/30/2021] [Indexed: 11/27/2022]
Abstract
The interleukin-1 (IL-1) family of cytokines is a central regulator of a myriad of immunological responses. It comprises several cytokines, including those belonging to the IL-1, IL-36 and IL-18 subfamilies, as well as IL-33. The IL-1 family primarily plays a role in orchestrating innate immune responses but also in adaptive immunity. Increased interest in the IL-1 family occurred following the discovery that dysregulation of IL-1 signalling underlies the pathogenesis of several monogenic auto-inflammatory diseases, characterized by sterile inflammation involving the skin and other organs. This also provided increased understanding of the role of innate immunity and the IL-1 family in polygenic auto-inflammatory skin conditions, such as neutrophilic dermatoses, as well as in some of the most common chronic inflammatory skin diseases, such as psoriasis or hidradenitis suppurativa. Several therapeutic agents have been developed to inhibit the IL-1 family members and their signalling pathways. These have shown therapeutic efficacy in several chronic inflammatory skin disorders. The aim of this review is to thoroughly describe the consequences of pathological dysregulation of IL-1, IL-33, IL-36, IL-18 pathways in dermatological conditions and to provide a forward-looking update on therapeutic strategies targeting signalling by IL-1 family cytokines.
Collapse
Affiliation(s)
- Laura Calabrese
- Institute of Dermatology, Catholic University of the Sacred Heart, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Dermatology and Allergy, University Hospital, LMU, Munich, Germany
| | - Zeno Fiocco
- Department of Dermatology and Allergy, University Hospital, LMU, Munich, Germany
| | - Takashi K Satoh
- Department of Dermatology and Allergy, University Hospital, LMU, Munich, Germany
| | - Ketty Peris
- Institute of Dermatology, Catholic University of the Sacred Heart, Rome, Italy.,Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Lars E French
- Department of Dermatology and Allergy, University Hospital, LMU, Munich, Germany.,Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami, Miller School of Medicine, Miami, Florida
| |
Collapse
|
69
|
Time-series transcriptomic analysis of bronchoalveolar lavage cells from virulent and low virulent PRRSV-1-infected piglets. J Virol 2021; 96:e0114021. [PMID: 34851149 PMCID: PMC8826917 DOI: 10.1128/jvi.01140-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has evolved to escape the immune surveillance for a survival advantage leading to a strong modulation of host’s immune responses and favoring secondary bacterial infections. However, limited data are available on how the immunological and transcriptional responses elicited by virulent and low-virulent PRRSV-1 strains are comparable and how they are conserved during the infection. To explore the kinetic transcriptional signature associated with the modulation of host immune response at lung level, a time-series transcriptomic analysis was performed in bronchoalveolar lavage cells upon experimental in vivo infection with two PRRSV-1 strains of different virulence, virulent subtype 3 Lena strain or the low-virulent subtype 1 3249 strain. The time-series analysis revealed overlapping patterns of dysregulated genes enriched in T-cell signaling pathways among both virulent and low-virulent strains, highlighting an upregulation of co-stimulatory and co-inhibitory immune checkpoints that were disclosed as Hub genes. On the other hand, virulent Lena infection induced an early and more marked “negative regulation of immune system process” with an overexpression of co-inhibitory receptors genes related to T-cell and NK cell functions, in association with more severe lung lesion, lung viral load, and BAL cell kinetics. These results underline a complex network of molecular mechanisms governing PRRSV-1 immunopathogenesis at lung level, revealing a pivotal role of co-inhibitory and co-stimulatory immune checkpoints in the pulmonary disease, which may have an impact on T-cell activation and related pathways. These immune checkpoints, together with the regulation of cytokine-signaling pathways, modulated in a virulence-dependent fashion, orchestrate an interplay among pro- and anti-inflammatory responses. IMPORTANCE Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the major threats to swine health and global production, causing substantial economic losses. We explore the mechanisms involved in the modulation of host immune response at lung level performing a time-series transcriptomic analysis upon experimental infection with two PRRSV-1 strains of different virulence. A complex network of molecular mechanisms was revealed to control the immunopathogenesis of PRRSV-1 infection, highlighting an interplay among pro- and anti-inflammatory responses as a potential mechanism to restrict inflammation-induced lung injury. Moreover, a pivotal role of co-inhibitory and co-stimulatory immune checkpoints was evidenced, which may lead to progressive dysfunction of T cells, impairing viral clearance and leading to persistent infection, favoring as well secondary bacterial infections or viral rebound. However, further studies should be conducted to evaluate the functional role of immune checkpoints in advanced stages of PRRSV infection and explore a possible T-cell exhaustion state.
Collapse
|
70
|
Rood JE, Behrens EM. Inherited Autoinflammatory Syndromes. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2021; 17:227-249. [PMID: 34699263 DOI: 10.1146/annurev-pathmechdis-030121-041528] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Autoinflammation describes a collection of diverse diseases caused by indiscriminate activation of the immune system in an antigen-independent manner. The rapid advancement of genetic diagnostics has allowed for the identification of a wide array of monogenic causes of autoinflammation. While the clinical picture of these syndromes is diverse, it is possible to thematically group many of these diseases under broad categories that provide insight into the mechanisms of disease and therapeutic possibilities. This review covers archetypical examples of inherited autoinflammatory diseases in five major categories: inflammasomopathy, interferonopathy, unfolded protein/cellular stress response, relopathy, and uncategorized. This framework can suggest where future work is needed to identify other genetic causes of autoinflammation, what types of diagnostics need to be developed to care for this patient population, and which options might be considered for novel therapeutic targeting. Expected final online publication date for the Annual Review of Pathology: Mechanisms of Disease, Volume 17 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Julia E Rood
- Division of Rheumatology, Children's Hospital of Philadelphia, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Edward M Behrens
- Division of Rheumatology, Children's Hospital of Philadelphia, Perelman School of Medicine at The University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
71
|
Min HK, Kim S, Lee JY, Kim KW, Lee SH, Kim HR. IL-18 binding protein suppresses IL-17-induced osteoclastogenesis and rectifies type 17 helper T cell / regulatory T cell imbalance in rheumatoid arthritis. J Transl Med 2021; 19:392. [PMID: 34530864 PMCID: PMC8444577 DOI: 10.1186/s12967-021-03071-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/07/2021] [Indexed: 11/25/2022] Open
Abstract
Background Patients with rheumatoid arthritis (RA) have increased levels of interleukin-18 (IL-18) and decreased levels of IL-18 binding protein (IL-18BP) in the serum and synovial fluid (SF) compared to those in patients with osteoarthritis (OA) or in healthy controls. In this study, we evaluated the effects of IL-18BP on osteoclastogenesis and T cell differentiation in RA in vitro. Methods Serum and SF of patients with RA and OA were collected to compare IL-18 and IL-18BP levels by the enzyme-linked immunosorbent assay. Peripheral blood mononuclear cells (PBMCs) and SF mononuclear cells (SFMCs) of RA patients were cultured under type 17 helper T cell (Th17) polarisation conditions with or without IL-18BP. In addition, PBMCs were cultured in the presence of receptor activator of nuclear factor kappa-Β ligand (RANKL) or IL-17A with or without IL-18BP, and tartrate-resistant acid phosphatase (TRAP) staining and real-time quantitative polymerase chain reaction for expression levels of osteoclast-related genes were performed. Results IL-18 levels were higher in the serum and SF of patients with RA, whereas IL-18BP was lower in the SF of patients with RA than in the control group. Treatment of patients’ PBMCs with IL-18BP decreased the differentiation of CD4+ IL-17A+ and CD4+ RANKL+ T cells, whereas the differentiation of CD4+CD25highFOXP3+ T cell population increased in a dose-dependent manner. These changes in CD4+ T cell differentiation were also observed in the SFMCs of patients with RA. The levels IL-17A and soluble RANKL in the culture medium were significantly decreased by IL-18BP. IL-18BP administration decreased TRAP+ cell counts in a dose-dependent manner on the background of stimulation with RANKL-and IL-17A. In addition, expression levels of TRAP, NFATC1, CTSK, and TNFRSF11A (RANK) genes were lower in the IL-18BP treated cells. Conclusion We showed that IL-18BP can rectify the Th17/Treg imbalance and decrease IL-17-induced osteoclastogenesis in PBMCs from patients with RA. Therefore, IL-18BP may have therapeutic potential for RA treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03071-2.
Collapse
Affiliation(s)
- Hong Ki Min
- Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, Seoul, 05030, Republic of Korea
| | - Sehee Kim
- Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, Seoul, 05030, Republic of Korea
| | - Ji-Yeon Lee
- The Rheumatism Research Center, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, 05030, Republic of Korea
| | | | - Sang-Heon Lee
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, 05030, Republic of Korea
| | - Hae-Rim Kim
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, 05030, Republic of Korea. .,Department of Rheumatology, Konkuk University Medical Center, 120-1 Neungdong-ro (Hwayang-dong), Gwangjin-gu, Seoul, 143-729, Republic of Korea.
| |
Collapse
|
72
|
Yu X, Newland SA, Zhao TX, Lu Y, Sage AS, Sun Y, Sriranjan RS, Ma MKL, Lam BYH, Nus M, Harrison JE, Bond SJ, Cheng X, Silvestre JS, Rudd JHF, Cheriyan J, Mallat Z. Innate Lymphoid Cells Promote Recovery of Ventricular Function After Myocardial Infarction. J Am Coll Cardiol 2021; 78:1127-1142. [PMID: 34503682 PMCID: PMC8434674 DOI: 10.1016/j.jacc.2021.07.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/06/2021] [Accepted: 07/13/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Innate lymphoid cells type 2 (ILC2s) play critical homeostatic functions in peripheral tissues. ILC2s reside in perivascular niches and limit atherosclerosis development. OBJECTIVES ILC2s also reside in the pericardium but their role in postischemic injury is unknown. METHODS We examined the role of ILC2 in a mouse model of myocardial infarction (MI), and compared mice with or without genetic deletion of ILC2. We determined infarct size using histology and heart function using echocardiography. We assessed cardiac ILC2 using flow cytometry and RNA sequencing. Based on these data, we devised a therapeutic strategy to activate ILC2 in mice with acute MI, using exogenous interleukin (IL)-2. We also assessed the ability of low-dose IL-2 to activate ILC2 in a double-blind randomized clinical trial of patients with acute coronary syndromes (ACS). RESULTS We found that ILC2 levels were increased in pericardial adipose tissue after experimental MI, and genetic ablation of ILC2 impeded the recovery of heart function. RNA sequencing revealed distinct transcript signatures in ILC2, and pointed to IL-2 axis as a major upstream regulator. Treatment of T-cell-deficient mice with IL-2 (to activate ILC2) significantly improved the recovery of heart function post-MI. Administration of low-dose IL-2 to patients with ACS led to activation of circulating ILC2, with significant increase in circulating IL-5, a prototypic ILC2-derived cytokine. CONCLUSIONS ILC2s promote cardiac healing and improve the recovery of heart function after MI in mice. Activation of ILC2 using low-dose IL-2 could be a novel therapeutic strategy to promote a reparative response after MI.
Collapse
Affiliation(s)
- Xian Yu
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom; Department of Cardiology, Union Hospital, Tongji, Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Stephen A Newland
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tian X Zhao
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Yuning Lu
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Andrew S Sage
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Yanyi Sun
- Université de Paris, PARCC, INSERM, F-75015 Paris, France
| | - Rouchelle S Sriranjan
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Marcella K L Ma
- The Wellcome Trust-MRC Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
| | - Brian Y H Lam
- The Wellcome Trust-MRC Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
| | - Meritxell Nus
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - James E Harrison
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Simon J Bond
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji, Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - James H F Rudd
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Joseph Cheriyan
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, United Kingdom; Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Ziad Mallat
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom; Université de Paris, PARCC, INSERM, F-75015 Paris, France.
| |
Collapse
|
73
|
Barnabei L, Laplantine E, Mbongo W, Rieux-Laucat F, Weil R. NF-κB: At the Borders of Autoimmunity and Inflammation. Front Immunol 2021; 12:716469. [PMID: 34434197 PMCID: PMC8381650 DOI: 10.3389/fimmu.2021.716469] [Citation(s) in RCA: 316] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/22/2021] [Indexed: 12/18/2022] Open
Abstract
The transcription factor NF-κB regulates multiple aspects of innate and adaptive immune functions and serves as a pivotal mediator of inflammatory response. In the first part of this review, we discuss the NF-κB inducers, signaling pathways, and regulators involved in immune homeostasis as well as detail the importance of post-translational regulation by ubiquitination in NF-κB function. We also indicate the stages of central and peripheral tolerance where NF-κB plays a fundamental role. With respect to central tolerance, we detail how NF-κB regulates medullary thymic epithelial cell (mTEC) development, homeostasis, and function. Moreover, we elaborate on its role in the migration of double-positive (DP) thymocytes from the thymic cortex to the medulla. With respect to peripheral tolerance, we outline how NF-κB contributes to the inactivation and destruction of autoreactive T and B lymphocytes as well as the differentiation of CD4+-T cell subsets that are implicated in immune tolerance. In the latter half of the review, we describe the contribution of NF-κB to the pathogenesis of autoimmunity and autoinflammation. The recent discovery of mutations involving components of the pathway has both deepened our understanding of autoimmune disease and informed new therapeutic approaches to treat these illnesses.
Collapse
Affiliation(s)
- Laura Barnabei
- INSERM UMR 1163, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute Paris Descartes Sorbonne Paris Cité University, Paris, France
| | - Emmanuel Laplantine
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| | - William Mbongo
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| | - Frédéric Rieux-Laucat
- INSERM UMR 1163, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute Paris Descartes Sorbonne Paris Cité University, Paris, France
| | - Robert Weil
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| |
Collapse
|
74
|
Kaur D, Chachi L, Gomez E, Sylvius N, Brightling CE. Interleukin-18, IL-18 binding protein and IL-18 receptor expression in asthma: a hypothesis showing IL-18 promotes epithelial cell differentiation. Clin Transl Immunology 2021; 10:e1301. [PMID: 34194747 PMCID: PMC8234286 DOI: 10.1002/cti2.1301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 01/15/2023] Open
Abstract
Objective In asthma, genome‐wide association studies have shown that interleukin‐18 (IL‐18) receptor 1 gene (IL‐18R1) and sputum IL‐18 are increased during exacerbations. However, the role of the IL‐18 axis in bronchial epithelial function is unclear. To investigate IL‐18, IL‐18 binding protein (BP) and IL‐18R expression in bronchial biopsies and sputum samples from patients with asthma, and to determine its functional role using in vitro bronchial epithelial cells. Methods The expression of IL‐18, IL‐18BP and IL‐18Rα was examined in subjects with asthma and healthy controls in bronchial biopsies by immunohistochemistry and IL‐18 and IL‐18BP release in sputum. In epithelial cells, the mRNA and protein expression of IL‐18, IL‐18BP, IL‐18Rα and IL‐18Rβ was assessed by qPCR, flow cytometry, Western blotting and immunofluorescence respectively. IL‐18 function in epithelial cells was examined by intracellular calcium, wound repair, synthetic activation and epithelial differentiation changes. Results In biopsies from subjects with asthma, the IL‐18 expression was not different in the lamina propria compared with controls but was decreased in the epithelium. In contrast, the IL‐18BP was decreased in the lamina propria in asthma and was absent in the bronchial epithelium. IL‐18 was released in sputum with IL‐18BP elevated in patients with asthma. The IL‐18Rα expression was not different between health and disease. In vitro, IL‐18‐stimulated bronchial epithelial cells increased intracellular calcium, wound repair, metabolic activity, morphological changes and epithelial cellular differentiation. Conclusion In asthma, the dynamic interaction between IL‐18, its cognate receptor and natural inhibitor is complex, with differences between airway compartments. Upregulation of IL‐18 can promote epithelial activation and cellular differentiation.
Collapse
Affiliation(s)
- Davinder Kaur
- Department of Respiratory Sciences Institute for Lung Health NIHR Biomedical Research Centre University of Leicester Leicester LE1 7RH UK
| | - Latifa Chachi
- Department of Respiratory Sciences Institute for Lung Health NIHR Biomedical Research Centre University of Leicester Leicester LE1 7RH UK
| | - Edith Gomez
- Department of Respiratory Sciences Institute for Lung Health NIHR Biomedical Research Centre University of Leicester Leicester LE1 7RH UK
| | - Nicolas Sylvius
- Genomic Core Facility Department of Genetics University of Leicester Adrian Building, University Road, G23 Leicester LE1 7RH UK
| | - Christopher E Brightling
- Department of Respiratory Sciences Institute for Lung Health NIHR Biomedical Research Centre University of Leicester Leicester LE1 7RH UK
| |
Collapse
|
75
|
Scimone C, Donato L, Alibrandi S, Vadalà M, Giglia G, Sidoti A, D'Angelo R. N-retinylidene-N-retinylethanolamine adduct induces expression of chronic inflammation cytokines in retinal pigment epithelium cells. Exp Eye Res 2021; 209:108641. [PMID: 34058230 DOI: 10.1016/j.exer.2021.108641] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/06/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022]
Abstract
Blindness due to photoreceptor degeneration is observed in both genetic and acquired eye disorders. Long blue light exposure can contribute to increase levels of oxidative compounds within the retinal pigment epithelium (RPE), enhancing risk of retinal damage. In retina, reactive oxygen species contribute to the activation of inflammatory cascade. If chronic, this inflammatory response can result in photoreceptor death. Therefore, we investigated the effects of the endogenous adduct N-retinylidene-N-retinylethanolamine (A2E) on RPE cells, in order to identify the most dysregulated cytokines and their related inflammatory pathways. RPE cells were exposed to A2E and blue light for 3h and 6h. By transcriptome analysis, we identified differentially expressed genes in A2E-treated cells, when compared to untreated ones. Expression values were quantified by the Limma R package. Enrichment analysis was performed according to the "Reactome" and the Gene Ontology databases. Expression of pro-inflammatory cytokines increased after 3h of A2E treatment and pathways related to IL-6 and IL-1 signaling resulted enriched. Also the up-regulation of genes having a protective role against inflammation was observed. Moreover, our results show that ferroptosis could contribute to RPE degeneration induced by A2E and blue light. Dysregulated genes related to retinal degeneration triggered by oxidative damage and inflammatory response activation identified in this study can be considered as potential biomarkers for targeted therapies.
Collapse
Affiliation(s)
- Concetta Scimone
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125, Messina, Italy; Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, I.E.ME.S.T., Via Michele Miraglia, 90139, Palermo, Italy
| | - Luigi Donato
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125, Messina, Italy; Department of Biomolecular Strategies, Genetics and Avant-Garde Therapies, I.E.ME.S.T., Via Michele Miraglia, 90139, Palermo, Italy.
| | - Simona Alibrandi
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125, Messina, Italy; Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d'Alcontres 31, 98166, Messina, Italy
| | - Maria Vadalà
- Department of Experimental Biomedicine and Clinical Neuroscience, Ophthalmology Section, University of Palermo, 90127, Palermo, Italy
| | - Giuseppe Giglia
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), Section of Human Physiology, University of Palermo, 90134, Palermo, Italy
| | - Antonina Sidoti
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125, Messina, Italy
| | - Rosalia D'Angelo
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria 1, 98125, Messina, Italy
| |
Collapse
|
76
|
Malhotra H, Garg V, Singh G. Biomarker Approach Towards Rheumatoid Arthritis Treatment. Curr Rheumatol Rev 2021; 17:162-175. [PMID: 33327920 DOI: 10.2174/1573397116666201216164013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/02/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023]
Abstract
Rheumatoid arthritis is an auto-immune disorder, recognized by cartilage as well as bone destruction, which causes irreversible joint deformities, which further results in functional limitations in the patient. Genes like HLA-DRB1 and PTPN22 are likely implicated in the genetic predisposition of rheumatoid arthritis pathology. The first and foremost clinical manifestation in a person with rheumatoid arthritis is joint destruction followed by cartilage and bone destruction caused by cell-cell interactions. The cell-cell interactions are thought to be initialized through the contact of antigen-presenting cells (APC) with CD4+ cells, leading to the progression of the disease. APC includes a complex of class ІІ major histocompatibility complex molecules along with peptide antigens and binds to the receptors present on the surface of T-cells. Further, the activation of macrophages is followed by the release of various pro-inflammatory cytokines such as IL-1 and TNF-α, which lead to the secretion of enzymes that degrade proteoglycan and collagen, which in turn, increase tissue degradation. Biomarkers like IL-6, IL-12, IL-8 and IL-18, 14-3-3η, RANKL, IFN-γ, IFN-β and TGF-β have been designated as key biomarkers in disease development and progression. The study of these biomarkers is very important as they act as a molecular indicator of pathological processes that aggravate the disease.
Collapse
Affiliation(s)
- Hitesh Malhotra
- Chandigarh College of Pharmacy Landran, Mohali, Punjab, India
| | - Vandana Garg
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
77
|
A novel anti-human IL-1R7 antibody reduces IL-18-mediated inflammatory signaling. J Biol Chem 2021; 296:100630. [PMID: 33823154 PMCID: PMC8018910 DOI: 10.1016/j.jbc.2021.100630] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 12/17/2022] Open
Abstract
Unchecked inflammation can result in severe diseases with high mortality, such as macrophage activation syndrome (MAS). MAS and associated cytokine storms have been observed in COVID-19 patients exhibiting systemic hyperinflammation. Interleukin-18 (IL-18), a proinflammatory cytokine belonging to the IL-1 family, is elevated in both MAS and COVID-19 patients, and its level is known to correlate with the severity of COVID-19 symptoms. IL-18 binds its specific receptor IL-1 receptor 5 (IL-1R5, also known as IL-18 receptor alpha chain), leading to the recruitment of the coreceptor, IL-1 receptor 7 (IL-1R7, also known as IL-18 receptor beta chain). This heterotrimeric complex then initiates downstream signaling, resulting in systemic and local inflammation. Here, we developed a novel humanized monoclonal anti-IL-1R7 antibody to specifically block the activity of IL-18 and its inflammatory signaling. We characterized the function of this antibody in human cell lines, in freshly obtained peripheral blood mononuclear cells (PBMCs) and in human whole blood cultures. We found that the anti-IL-1R7 antibody significantly suppressed IL-18-mediated NFκB activation, reduced IL-18-stimulated IFNγ and IL-6 production in human cell lines, and reduced IL-18-induced IFNγ, IL-6, and TNFα production in PBMCs. Moreover, the anti-IL-1R7 antibody significantly inhibited LPS- and Candida albicans–induced IFNγ production in PBMCs, as well as LPS-induced IFNγ production in whole blood cultures. Our data suggest that blocking IL-1R7 could represent a potential therapeutic strategy to specifically modulate IL-18 signaling and may warrant further investigation into its clinical potential for treating IL-18-mediated diseases, including MAS and COVID-19.
Collapse
|
78
|
Martin P, Goldstein JD, Mermoud L, Diaz-Barreiro A, Palmer G. IL-1 Family Antagonists in Mouse and Human Skin Inflammation. Front Immunol 2021; 12:652846. [PMID: 33796114 PMCID: PMC8009184 DOI: 10.3389/fimmu.2021.652846] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 12/23/2022] Open
Abstract
Interleukin (IL)-1 family cytokines initiate inflammatory responses, and shape innate and adaptive immunity. They play important roles in host defense, but excessive immune activation can also lead to the development of chronic inflammatory diseases. Dysregulated IL-1 family signaling is observed in a variety of skin disorders. In particular, IL-1 family cytokines have been linked to the pathogenesis of psoriasis and atopic dermatitis. The biological activity of pro-inflammatory IL-1 family agonists is controlled by the natural receptor antagonists IL-1Ra and IL-36Ra, as well as by the regulatory cytokines IL-37 and IL-38. These four anti-inflammatory IL-1 family members are constitutively and highly expressed at steady state in the epidermis, where keratinocytes are a major producing cell type. In this review, we provide an overview of the current knowledge concerning their regulatory roles in skin biology and inflammation and their therapeutic potential in human inflammatory skin diseases. We further highlight some common misunderstandings and less well-known observations, which persist in the field despite recent extensive interest for these cytokines.
Collapse
Affiliation(s)
- Praxedis Martin
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Jérémie D. Goldstein
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Loïc Mermoud
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alejandro Diaz-Barreiro
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
79
|
Ortutay Z, Grönholm A, Laitinen M, Keresztes-Andrei M, Hermelo I, Pesu M. Identification of Novel Genetic Regulatory Region for Proprotein Convertase FURIN and Interferon Gamma in T Cells. Front Immunol 2021; 12:630389. [PMID: 33679774 PMCID: PMC7930619 DOI: 10.3389/fimmu.2021.630389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/22/2021] [Indexed: 12/30/2022] Open
Abstract
The proprotein convertase enzyme FURIN promotes the proteolytic maturation of pro-proteins and thereby it serves as an important factor for maintaining cellular homeostasis. In T cells, FURIN is critical for maintaining the T regulatory cell dependent peripheral immune tolerance and intact T helper cell polarization. The enzymatic activity of FURIN is directly associated with its expression levels, but genetic determinants for cell-type specific Furin gene regulation have remained elusive. By exploring the histone acetyltransferase p300 binding patterns in T helper cells, a putative regulatory region at ca. 20kB upstream of Furin gene was identified. When this region was deleted with CRISPR/Cas9 the production of Furin mRNA was significantly reduced in activated mouse T cells. Genome-wide RNA profiling by sequencing revealed that the novel Furin regulator region also impacted the expression of several genes that have previously been associated with the Th1 type hall mark cytokine IFNγ regulation or function. Finally, Furin genetic regulatory region was found to specifically promote the secretion of IFNγ by activated T cells. In sum, our data unravels the presence of Furin expression regulatory region in T cells that has characteristics of a super-enhancer for Th1 cell fate.
Collapse
Affiliation(s)
- Zsuzsanna Ortutay
- Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Anna Grönholm
- Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Melina Laitinen
- Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Melinda Keresztes-Andrei
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Ismail Hermelo
- Computational Biology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marko Pesu
- Immunoregulation, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Fimlab Laboratories, Tampere, Finland
| |
Collapse
|
80
|
Van Den Eeckhout B, Tavernier J, Gerlo S. Interleukin-1 as Innate Mediator of T Cell Immunity. Front Immunol 2021; 11:621931. [PMID: 33584721 PMCID: PMC7873566 DOI: 10.3389/fimmu.2020.621931] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/08/2020] [Indexed: 12/19/2022] Open
Abstract
The three-signal paradigm tries to capture how the innate immune system instructs adaptive immune responses in three well-defined actions: (1) presentation of antigenic peptides in the context of MHC molecules, which allows for a specific T cell response; (2) T cell co-stimulation, which breaks T cell tolerance; and (3) secretion of polarizing cytokines in the priming environment, thereby specializing T cell immunity. The three-signal model provides an empirical framework for innate instruction of adaptive immunity, but mainly discusses STAT-dependent cytokines in T cell activation and differentiation, while the multi-faceted roles of type I IFNs and IL-1 cytokine superfamily members are often neglected. IL-1α and IL-1β are pro-inflammatory cytokines, produced following damage to the host (release of DAMPs) or upon innate recognition of PAMPs. IL-1 activity on both DCs and T cells can further shape the adaptive immune response with variable outcomes. IL-1 signaling in DCs promotes their ability to induce T cell activation, but also direct action of IL-1 on both CD4+ and CD8+ T cells, either alone or in synergy with prototypical polarizing cytokines, influences T cell differentiation under different conditions. The activities of IL-1 form a direct bridge between innate and adaptive immunity and could therefore be clinically translatable in the context of prophylactic and therapeutic strategies to empower the formation of T cell immunity. Understanding the modalities of IL-1 activity during T cell activation thus could hold major implications for rational development of the next generation of vaccine adjuvants.
Collapse
Affiliation(s)
- Bram Van Den Eeckhout
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jan Tavernier
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Orionis Biosciences BV, Ghent, Belgium
| | - Sarah Gerlo
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
81
|
Giudice V, Cardamone C, Triggiani M, Selleri C. Bone Marrow Failure Syndromes, Overlapping Diseases with a Common Cytokine Signature. Int J Mol Sci 2021; 22:ijms22020705. [PMID: 33445786 PMCID: PMC7828244 DOI: 10.3390/ijms22020705] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/06/2021] [Accepted: 01/09/2021] [Indexed: 12/19/2022] Open
Abstract
Bone marrow failure (BMF) syndromes are a heterogenous group of non-malignant hematologic diseases characterized by single- or multi-lineage cytopenia(s) with either inherited or acquired pathogenesis. Aberrant T or B cells or innate immune responses are variously involved in the pathophysiology of BMF, and hematological improvement after standard immunosuppressive or anti-complement therapies is the main indirect evidence of the central role of the immune system in BMF development. As part of this immune derangement, pro-inflammatory cytokines play an important role in shaping the immune responses and in sustaining inflammation during marrow failure. In this review, we summarize current knowledge of cytokine signatures in BMF syndromes.
Collapse
Affiliation(s)
- Valentina Giudice
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (V.G.); (C.C.); (C.S.)
- Clinical Pharmacology, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
| | - Chiara Cardamone
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (V.G.); (C.C.); (C.S.)
- Internal Medicine and Clinical Immunology, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
| | - Massimo Triggiani
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (V.G.); (C.C.); (C.S.)
- Internal Medicine and Clinical Immunology, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
- Correspondence: ; Tel.: +39-089-672810
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (V.G.); (C.C.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84131 Salerno, Italy
| |
Collapse
|
82
|
Quader M, Mezzaroma E, Kenning K, Toldo S. Modulation of Interleukin-1 and -18 Mediated Injury in Donation after Circulatory Death Mouse Hearts. J Surg Res 2021; 257:468-476. [PMID: 32896815 PMCID: PMC8687874 DOI: 10.1016/j.jss.2020.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/14/2020] [Accepted: 08/02/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND Donation after circulatory death donors (DCD) can expand the donor pool for heart transplantation, which primarily depends on brain death donors. Ischemia and reperfusion injury are inherent to the DCD process. We hypothesize that pharmacologic inhibition of interleukin-1 (IL-1) and/or IL-18 is protective to DCD hearts. MATERIALS AND METHODS Following clinical protocol, in-situ ischemia time in control beating-heart donor (CBD) and DCD groups was less than 5 and 40 min, respectively. Wild type (WT) C57Bl6/j, IL-1 receptor type I knockout (IL-1RI-KO), and IL-18 KO mice were used. Hearts were reanimated for 90 min on a Langendorff system with Krebs-Henseleit buffer at 37°C, to assess physiologic parameters. Recombinant IL-1 receptor antagonist (IL-1Ra) and/or IL-18 binding protein (IL-18BP) were added to the Krebs-Henseleit buffer to inhibit IL-1 and/or the IL-18 signaling, respectively. RESULTS Developed pressure and ± dP/dt were significantly impaired in the DCD-WT group compared to CBD-WT (P ≤ 0.05). Troponin release was higher in DCD-WT groups. Functional parameters were preserved, and troponin release was significantly less in the DCD knockout groups. Heart function was improved in DCD groups treated with IL-1Ra or IL-18BP compared to the DCD-WT group. CONCLUSIONS Heart function was significantly impaired in the DCD-WT group compared to CBD-WT. Genetic deletion or pharmacologic blockade of IL-1 or IL-18 was protective to DCD hearts.
Collapse
Affiliation(s)
- Mohammed Quader
- Cardiovascular Surgery, Virginia Commonwealth University, Richmond, Virginia; Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia.
| | - Eleonora Mezzaroma
- Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Kristine Kenning
- Cardiovascular Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Stefano Toldo
- Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia; Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
83
|
Gibson MS, Steyn A, Kealy D, Kaspers B, Fife MS. Molecular cloning and characterisation of chicken IL-18 binding protein. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 114:103850. [PMID: 32918930 PMCID: PMC7661785 DOI: 10.1016/j.dci.2020.103850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 06/11/2023]
Abstract
The human IL-1 receptor family is comprised of 11 membrane bound or soluble receptors and the IL-18 binding protein (IL-18BP). These receptors are dispersed across seven genomic loci, with the majority at a single locus. Direct orthologues were identified in the chicken at conserved genomic loci; however, the IL-18BP remained absent from the first four builds of the chicken genome sequence. Subsequent assemblies identified the gene at a locus syntenic with mammals; however, these predicted sequences differed between genome builds and contained multiple errors. A partial IL-18BP-like sequence in the NCBI EST database was used to clone the full-length cDNA. A splice variant, which lacks the exon that encodes part of the signal peptide, was also cloned. Human IL-18BP is differentially spliced to produce a number of variants, which are all secreted. By contrast, the spliced chicken isoform was predicted to be intracellular, and we identified similar variants with the same exon missing in a limited number of divergent vertebrate species. Mammalian and viral IL-18BPs inhibit IL-18 activity by directly binding to this cytokine. Full-length and intracellular chicken IL-18BPs were equally effective at inhibiting IL-18-mediated IFN-γ release from an avian B-cell line. Analysis of the predicted chIL-18BP protein sequence revealed two crucial residues, which account for 50% of the binding affinity between human IL-18 and IL-18BP, are conserved in the chicken and a fowlpox-encoded homologue, fpv214. This suggests specific fowlpox viruses used in humans as a vaccine vector have the potential to dampen anti-viral host immune responses.
Collapse
Affiliation(s)
- Mark S Gibson
- BioISI - Biosystems and Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Lisboa, Portugal
| | | | - David Kealy
- York Biomedical Research Institute and Department of Biology, University of York, Heslington, York, UK
| | - Bernd Kaspers
- Department of Veterinary Science, Ludwig-Maximilians-Universität, Munich, Germany
| | - Mark S Fife
- The Pirbright Institute, Pirbright, Woking, UK; Aviagen Ltd, Newbridge, UK.
| |
Collapse
|
84
|
Machahua C, Buendia-Roldan I, Ocaña-Guzman R, Molina-Molina M, Pardo A, Chavez-Galan L, Selman M. CD4+T cells in ageing-associated interstitial lung abnormalities show evidence of pro-inflammatory phenotypic and functional profile. Thorax 2020; 76:152-160. [PMID: 33298584 PMCID: PMC7815886 DOI: 10.1136/thoraxjnl-2020-215520] [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: 06/11/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022]
Abstract
Background Interstitial lung abnormalities (ILA) occur in around 10% of subjects over 60 years, and are associated with a higher rate of all-cause mortality. The pathogenic mechanisms are unclear, and the putative contribution of alterations in the immune response has not been explored. Normal ageing is associated with immune deficiencies, including Naïve T-cell decrease and greater expression of the proliferative-limiting, co-inhibitory receptor killer-cell lectin-like receptor G1 (KLRG1). Objective To evaluate the frequency and activation state of different T-cell subpopulations in ILA subjects. Methods Peripheral blood mononuclear cells were obtained from 15 individuals with ILA, 21 age-matched controls and 28 healthy young subjects. T-cells phenotype was characterised by flow cytometry, and proliferation and activation by stimulation with anti-CD3/anti-CD28 or phorbol myristate acetate/ionomycin; KLRG1 isoforms were evaluated by western blot and cytokines were quantified by ELISA and Multiplex. Results A significant increase of Naïve CD4+T cells together with a decrease of central and effector memory CD4+T cells was observed in ILA compared with age-matched controls. CD4+T cells from ILA subjects exhibited greater basal proliferation, which raised after anti-CD3/anti-CD28 stimulation. Additionally, a significant increase in the levels of interleukin-6 and interferon gamma was observed in isolated CD4+T cells and plasma of ILA subjects. They also displayed fewer KLRG1+/CD4+T cells with an increase of circulating E-cadherin, the ligand of KLRG1+. No changes were observed with CD8+T cell subsets. Conclusion CD4+T cells from ILA subjects are highly proliferative and show an excessive functional activity, likely related to the loss of KLRG1 expression, which may contribute to an inflammatory state and the development of ILA.
Collapse
Affiliation(s)
- Carlos Machahua
- Servei de Pneumologia, Grup de Recerca Pneumològic, Institut d'Investigacions Biomèdiques de Bellvitge (IDIBELL), Hospital Universitari de Bellvitge, Hospital de Llobregat, Barcelona, Spain
| | - Ivette Buendia-Roldan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Ranferi Ocaña-Guzman
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - María Molina-Molina
- Servei de Pneumologia, Grup de Recerca Pneumològic, Institut d'Investigacions Biomèdiques de Bellvitge (IDIBELL), Hospital Universitari de Bellvitge, Hospital de Llobregat, Barcelona, Spain
| | - Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Leslie Chavez-Galan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Moises Selman
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| |
Collapse
|
85
|
IL-18 binding protein (IL-18BP) as a novel radiation countermeasure after radiation exposure in mice. Sci Rep 2020; 10:18674. [PMID: 33122671 PMCID: PMC7596073 DOI: 10.1038/s41598-020-75675-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/07/2020] [Indexed: 12/22/2022] Open
Abstract
Recent studies suggested that radiation exposure causes local and systemic inflammatory responses and induces cell and tissue damage. We have reported that IL-18 plays an important role in radiation-induced injury. Here, we demonstrate that IL-18 binding protein (IL-18BP), a natural antagonist of IL-18, was significantly increased (1.7-63 fold) in mouse serum on day 1 after 0.5-10 Gy TBI. However, this high level of IL-18BP was not sufficient to neutralize the active IL-18 in irradiated mice, resulting in a radiation dose-dependent free IL-18 increase in these mice's serum which led to pathological alterations to the irradiated cells and tissues and finally caused animal death. Administration of recombinant human (rh) IL-18BP (1.5 mg/kg) with single (24, 48 or 72 h post-TBI) or double doses (48 h and 5 days post-TBI) subcutaneous (SC) injection increased 30-day survival of CD2F1 mice after 9 Gy TBI 12.5-25% compared with the vehicle control treated group, respectively. Furthermore, the mitigative effects of rhIL-18BP included balancing the ratio of IL-18/IL-18BP and decreasing the free IL-18 levels in irradiated mouse serum and significantly increasing blood cell counts, BM hematopoietic cellularity and stem and progenitor cell clonogenicity in mouse BM. Furthermore, IL-18BP treatment inhibited the IL-18 downstream target interferon (IFN)-γ expression in mouse BM, decreased reactive oxygen species (ROS) level in the irradiated mouse heart tissues, attenuated the stress responsive factor GDF-15 (growth differentiation factor-15) and increased the intestine protector citrulline level in total body irradiated mouse serum, implicating that IL-18BP may protect multiple organs from radiation-induced inflammation and oxidative stress. Our data suggest that IL-18 plays a key role in radiation-induced cell and tissue damage and dysfunction; and for the first time demonstrated that IL-18BP counters IL-18 activation and therefore may mitigate/treat radiation-induced multiple organ injuries and increase animal survival with a wider therapeutic window from 24 h and beyond after lethal doses of radiation exposure.
Collapse
|
86
|
Harms RZ, Ostlund KR, Cabrera MS, Edwards E, Fisher M, Sarvetnick N. Confirmation and Identification of Biomarkers Implicating Environmental Triggers in the Pathogenesis of Type 1 Diabetes. Front Immunol 2020; 11:1922. [PMID: 33042112 PMCID: PMC7523316 DOI: 10.3389/fimmu.2020.01922] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/17/2020] [Indexed: 12/16/2022] Open
Abstract
Multiple environmental triggers have been proposed to explain the increased incidence of type 1 diabetes (T1D). These include viral infections, microbiome disturbances, metabolic disorders, and vitamin D deficiency. Here, we used ELISA to examine blood plasma from juvenile T1D subjects and age-matched controls for the abundance of several circulating factors relevant to these hypotheses. We screened plasma for sCD14, mannose binding lectin (MBL), lipopolysaccharide binding protein (LBP), c-reactive protein (CRP), fatty acid binding protein 2 (FABP2), human growth hormone, leptin, total adiponectin, high molecular weight (HMW) adiponectin, total IgG, total IgA, total IgM, endotoxin core antibodies (EndoCAbs), 25(OH) vitamin D, vitamin D binding protein, IL-7, IL-10, IFN-γ, TNF-α, IL-17A, IL-18, and IL-18BPa. Subjects also were tested for prevalence of antibodies targeting adenovirus, parainfluenza 1/2/3, Coxsackievirus, cytomegalovirus, Epstein-Barr virus viral capsid antigen (EBV VCA), herpes simplex virus 1, and Saccharomyces cerevisiae. Finally, all subjects were screened for presence and abundance of autoantibodies targeting islet cell cytoplasmic proteins (ICA), glutamate decarboxylase 2 (GAD65), zinc transporter 8 (ZNT8), insulinoma antigen 2 (IA-2), tissue transglutaminase, and thyroid peroxidase, while β cell function was gauged by measuring c-peptide levels. We observed few differences between control and T1D subjects. Of these, we found elevated sCD14, IL-18BPa, and FABP2, and reduced total IgM. Female T1D subjects were notably elevated in CRP levels compared to control, while males were similar. T1D subjects also had significantly lower prevalence of EBV VCA antibodies compared to control. Lastly, we observed that c-peptide levels were significantly correlated with leptin levels among controls, but this relationship was not significant among T1D subjects. Alternatively, adiponectin levels were significantly correlated with c-peptide levels among T1D subjects, while controls showed no relationship between these two factors. Among T1D subjects, the highest c-peptide levels were associated with the lowest adiponectin levels, an indication of insulin resistance. In total, from our examination we found limited data that strongly support any of the hypotheses investigated. Rather, we observed an indication of unexplained monocyte/macrophage activation in T1D subjects judging from elevated levels of sCD14 and IL-18BPa. These observations were partnered with unique associations between adipokines and c-peptide levels among T1D subjects.
Collapse
Affiliation(s)
- Robert Z Harms
- Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
| | - Katie R Ostlund
- Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
| | - Monina S Cabrera
- Endocrine Clinic, Children's Hospital and Medical Center, Omaha, NE, United States
| | - Earline Edwards
- Endocrine Clinic, Children's Hospital and Medical Center, Omaha, NE, United States
| | - Marisa Fisher
- Endocrine Clinic, Children's Hospital and Medical Center, Omaha, NE, United States
| | - Nora Sarvetnick
- Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States.,Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
87
|
Han Y, Huard A, Mora J, da Silva P, Brüne B, Weigert A. IL-36 family cytokines in protective versus destructive inflammation. Cell Signal 2020; 75:109773. [PMID: 32898612 DOI: 10.1016/j.cellsig.2020.109773] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/14/2022]
Abstract
The IL-1 family of cytokines and receptors are critical regulators of inflammation. Within the IL-1 family and in contrast to its IL-1 and IL-18 subfamilies, the IL-36 subfamily is still poorly characterized. Three pro-inflammatory agonists IL-36α, IL-36β, IL-36γ, one IL-36 receptor (IL-1R6) antagonist, IL-36RA, and one putative IL-1R6 antagonist, IL-38, have been grouped into the IL-36 cytokine subfamily. IL-36 agonists signal through a common receptor complex to serve as early triggers of inflammatory responses by activating and cross-regulating a number of inflammatory pathways including NF-κB, MAPK and IFN signaling. IL-36RA binds to IL-1R6 to limit inflammatory signaling, while IL-38 may be an antagonist of more than one IL-1 family receptor. Expression patterns of IL-36 family cytokines, being most prominently expressed in epithelial barrier tissues such as the skin and intestines as well as in immune cells, suggest a role in protecting these barriers from infection. Dysregulation of IL-36 family cytokine signaling at physiological barriers, most prominently the skin, induces autoimmune inflammation. However, transferring the potential of IL-36 to induce tissue damage to tumors might benefit cancer patients. Here we summarize signaling pathways regulated by IL-36 family cytokines, including IL-38, and the consequences for physiological protective and pathophysiological destructive inflammation. Moreover, we discuss the limits of current knowledge on IL-36 family function to open potential avenues for research in the future.
Collapse
Affiliation(s)
- Yingying Han
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt 60590, Germany; Special Key Laboratory of Oral Diseases Research, Higher Education Institutions of Guizhou Province, Zunyi Medical University, Zunyi 563006, Guizhou, China; School of Stomatology, Zunyi Medical University, Zunyi 563006, Guizhou, China
| | - Arnaud Huard
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt 60590, Germany
| | - Javier Mora
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt 60590, Germany; Faculty of Microbiology, University of Costa Rica, San José 2060, Costa Rica
| | - Priscila da Silva
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt 60590, Germany; Translational Medicine and Pharmacology (TMP), Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt 60590, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt 60590, Germany; Translational Medicine and Pharmacology (TMP), Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt 60590, Germany; Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt 60596, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt 60590, Germany; Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt 60596, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany.
| |
Collapse
|
88
|
Vecchié A, Bonaventura A, Toldo S, Dagna L, Dinarello CA, Abbate A. IL-18 and infections: Is there a role for targeted therapies? J Cell Physiol 2020; 236:1638-1657. [PMID: 32794180 DOI: 10.1002/jcp.30008] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/24/2020] [Accepted: 08/01/2020] [Indexed: 01/08/2023]
Abstract
Interleukin (IL)-18 is a pro-inflammatory cytokine belonging to the IL-1 family, first identified for its interferon-γ-inducing properties. IL-18 regulates both T helper (Th) 1 and Th2 responses. It acts synergistically with IL-12 in the Th1 paradigm, whereas with IL-2 and without IL-12 it can induce Th2 cytokine production from cluster of differentation (CD)4+ T cells, natural killer (NK cells, NKT cells, as well as from Th1 cells. IL-18 also plays a role in the hemophagocytic lymphohistiocytosis, a life-threatening condition characterized by a cytokine storm that can be secondary to infections. IL-18-mediated inflammation was largely studied in animal models of bacterial, viral, parasitic, and fungal infections. These studies highlight the contribution of either IL-18 overproduction by the host or overresponsiveness of the host to IL-18 causing an exaggerated inflammatory burden and leading to tissue injury. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the coronavirus disease 2019 (COVID-19). The damage in the later phase of the disease appears to be driven by a cytokine storm, including interleukin IL-1 family members and secondary cytokines like IL-6. IL-18 may participate in this hyperinflammation, as it was previously found to be able to cause injury in the lung tissue of infected animals. IL-18 blockade has become an appealing therapeutic target and has been tested in some IL-18-mediated rheumatic diseases and infantile-onset macrophage activation syndrome. Given its role in regulating the immune response to infections, IL-18 blockade might represent a therapeutic option for COVID-19, although further studies are warranted to investigate more in detail the exact role of IL-18 in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Alessandra Vecchié
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Aldo Bonaventura
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia.,Department of Internal Medicine, First Clinic of Internal Medicine, University of Genoa, Genoa, Italy
| | - Stefano Toldo
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Charles A Dinarello
- Department of Medicine and Immunology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Antonio Abbate
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
89
|
Interleukin-18 from neurons and microglia mediates depressive behaviors in mice with post-stroke depression. Brain Behav Immun 2020; 88:411-420. [PMID: 32272223 DOI: 10.1016/j.bbi.2020.04.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/23/2020] [Accepted: 04/04/2020] [Indexed: 02/07/2023] Open
Abstract
Post-stroke depression (PSD) is a common and serious complication that is affecting one thirds of stroke patients which leaves them with a poor quality of life, high mortality rate, high recurrent rate, and slow recovery. Recent studies showed that serum interleukin-18 (IL-18) level is a biomarker for patients with PSD. However, the role of IL-18 in the pathology of PSD is still unclear. In this study, we demonstrated that the IL-18 level in the ischemic brain significantly increased in mice with depression-like behaviors that were caused by the combined use of chronic spatial restraint stress and middle cerebral artery occlusion. Interestingly, IL-18 expression was mainly found in neurons at early phase and in microglia at a later phase. Injection of the exogenous IL-18 into the amygdala, but not the hippocampus or the striatum caused severe depression-like behaviors. On the contrary, the blockage of endogenous IL-18 by IL-18 binding protein, a specific antagonist of IL-18, repressed depressive phenotypes in SIR mice. IL-18 KO mice exhibited the resistance to spatial restraint stress and cerebral ischemia injury. Finally, we found that IL-18 mediated depressive behaviors by the interaction of IL-18 receptor and NKCC1, a sodium-potassium chloride co-transporter that is related to GABAergic inhibition. Administration of NKCC1 antagonist bumetanide exerted a therapeutic effect on the in IL-18-induced depressive mice. In conclusion, we demonstrated that increased IL-18 in the brain causes depression-like behaviors by promoting the IL-18 receptor/NKCC1 signaling pathway. Targeting IL-18 and its downstream pathway is a promising strategy for the prevention and treatment of PSD.
Collapse
|
90
|
Spalinger MR, Schwarzfischer M, Scharl M. The Role of Protein Tyrosine Phosphatases in Inflammasome Activation. Int J Mol Sci 2020; 21:E5481. [PMID: 32751912 PMCID: PMC7432435 DOI: 10.3390/ijms21155481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/13/2022] Open
Abstract
Inflammasomes are multi-protein complexes that mediate the activation and secretion of the inflammatory cytokines IL-1β and IL-18. More than half a decade ago, it has been shown that the inflammasome adaptor molecule, ASC requires tyrosine phosphorylation to allow effective inflammasome assembly and sustained IL-1β/IL-18 release. This finding provided evidence that the tyrosine phosphorylation status of inflammasome components affects inflammasome assembly and that inflammasomes are subjected to regulation via kinases and phosphatases. In the subsequent years, it was reported that activation of the inflammasome receptor molecule, NLRP3, is modulated via tyrosine phosphorylation as well, and that NLRP3 de-phosphorylation at specific tyrosine residues was required for inflammasome assembly and sustained IL-1β/IL-18 release. These findings demonstrated the importance of tyrosine phosphorylation as a key modulator of inflammasome activity. Following these initial reports, additional work elucidated that the activity of several inflammasome components is dictated via their phosphorylation status. Particularly, the action of specific tyrosine kinases and phosphatases are of critical importance for the regulation of inflammasome assembly and activity. By summarizing the currently available literature on the interaction of tyrosine phosphatases with inflammasome components we here provide an overview how tyrosine phosphatases affect the activation status of inflammasomes.
Collapse
Affiliation(s)
- Marianne R. Spalinger
- Department of Gastroenterology and Hepatology, University Hospital Zurich, 8091 Zurich, Switzerland; (M.S.); (M.S.)
| | - Marlene Schwarzfischer
- Department of Gastroenterology and Hepatology, University Hospital Zurich, 8091 Zurich, Switzerland; (M.S.); (M.S.)
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, 8091 Zurich, Switzerland; (M.S.); (M.S.)
- Zurich Center for Integrative Human Physiology, University of Zurich, 8006 Zurich, Switzerland
| |
Collapse
|
91
|
Harel M, Girard-Guyonvarc'h C, Rodriguez E, Palmer G, Gabay C. Production of IL-18 Binding Protein by Radiosensitive and Radioresistant Cells in CpG-Induced Macrophage Activation Syndrome. THE JOURNAL OF IMMUNOLOGY 2020; 205:1167-1175. [PMID: 32651219 DOI: 10.4049/jimmunol.2000168] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/15/2020] [Indexed: 12/20/2022]
Abstract
IL-18 binding protein (IL-18BP) acts as a naturally occurring IL-18 decoy receptor. If the balance between IL-18 and IL-18BP is dysregulated, abnormal levels of free bioactive IL-18 are detected, such as in the sera of Il-18bp knockout (KO) mice with CpG-induced macrophage activation syndrome. To determine the cellular sources of Il-18bp in vivo, we selectively depleted Il-18bp expression in either radiosensitive or radioresistant cells using bone marrow transfer between wild-type (WT) and Il-18bp KO mice. Following repeated CpG injections, Il-18bp KO (donor)→ Il-18bp KO (recipient) chimeric mice exhibited more severe disease, with an enhanced Ifn-γ signature and circulating free Il-18 levels, in comparison with WT→WT chimeras. Interestingly, the phenotype of KO→WT and WT→KO mice did not differ from that of WT→WT mice. Consistent with this finding, serum Il-18bp levels were similar in these three groups of mice. The contribution of radioresistant and radiosensitive cells to Il-18bp production varied markedly according to the organ examined, with a major contribution of radiosensitive cells in the spleen as opposed to a major contribution of radioresistant cells in the lung. Finally, Ifn-γ blockade abrogated the CpG-induced but not the constitutive Il-18bp production. Our results demonstrate that circulating Il-18bp is induced in response to Ifn-γ during CpG-induced macrophage activation syndrome and is present at high levels in the circulation to prevent the deleterious systemic effects of Il-18.
Collapse
Affiliation(s)
- Mathilde Harel
- Department of Pathology and Immunology, School of Medicine, University of Geneva, CH-1211 Geneva 4, Switzerland; and.,Division of Rheumatology, Department of Medicine, University Hospitals, CH-1211 Geneva 14, Switzerland
| | | | - Emiliana Rodriguez
- Department of Pathology and Immunology, School of Medicine, University of Geneva, CH-1211 Geneva 4, Switzerland; and.,Division of Rheumatology, Department of Medicine, University Hospitals, CH-1211 Geneva 14, Switzerland
| | - Gaby Palmer
- Department of Pathology and Immunology, School of Medicine, University of Geneva, CH-1211 Geneva 4, Switzerland; and.,Division of Rheumatology, Department of Medicine, University Hospitals, CH-1211 Geneva 14, Switzerland
| | - Cem Gabay
- Department of Pathology and Immunology, School of Medicine, University of Geneva, CH-1211 Geneva 4, Switzerland; and .,Division of Rheumatology, Department of Medicine, University Hospitals, CH-1211 Geneva 14, Switzerland
| |
Collapse
|
92
|
Italiani P, Mosca E, Della Camera G, Melillo D, Migliorini P, Milanesi L, Boraschi D. Profiling the Course of Resolving vs. Persistent Inflammation in Human Monocytes: The Role of IL-1 Family Molecules. Front Immunol 2020; 11:1426. [PMID: 32754155 PMCID: PMC7365847 DOI: 10.3389/fimmu.2020.01426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 06/03/2020] [Indexed: 12/18/2022] Open
Abstract
Monocytes and macrophages have a central role in all phases of an inflammatory reaction. To understanding the regulation of monocyte activation during a physiological or pathological inflammation, we propose two in vitro models that recapitulate the different phases of the reaction (recruitment, initiation, development, and resolution vs. persistence of inflammation), based on human primary blood monocytes exposed to sequential modifications of microenvironmental conditions. These models exclusively describe the functional development of blood-derived monocytes that first enter an inflammatory site. All reaction phases were profiled by RNA-Seq, and the two models were validated by studying the modulation of IL-1 family members. Genes were differentially modulated, and distinct clusters were identified during the various phases of inflammation. Pathway analysis revealed that both models were enriched in pathways involved in innate immune activation. We observe that monocytes acquire an M1-like profile during early inflammation, and switch to a deactivated M2-like profile during both the resolving and persistent phases. However, during persistent inflammation they partially maintain an M1 profile, although they lose the ability to produce inflammatory cytokines compared to M1 cells. The production of IL-1 family molecules by ELISA reflected the transcriptomic profiles in the distinct phases of the two inflammatory reactions. Based on the results, we hypothesize that persistence of inflammatory stimuli cannot maintain the M1 activated phenotype of incoming monocytes for long, suggesting that the persistent presence of M1 cells and effects in a chronically inflamed tissue is mainly due to activation of newly incoming cells. Moreover, being IL-1 family molecules mainly expressed and secreted by monocytes during the early stages of the inflammatory response (within 4-14 h), and the rate of their production decreasing during the late phase of both resolving and persistent inflammation, we suppose that IL-1 factors are key regulators of the acute defensive innate inflammatory reaction that precedes establishment of longer-term adaptive immunity, and are mainly related to the presence of recently recruited blood monocytes. The well-described role of IL-1 family cytokines and receptors in chronic inflammation is therefore most likely dependent on the continuous influx of blood monocytes into a chronically inflamed site.
Collapse
Affiliation(s)
- Paola Italiani
- Institute of Protein Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Ettore Mosca
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Giacomo Della Camera
- Institute of Protein Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Daniela Melillo
- Institute of Protein Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Paola Migliorini
- Clinical Immunology Unit, Department Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luciano Milanesi
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Diana Boraschi
- Institute of Protein Biochemistry and Cell Biology, National Research Council, Naples, Italy
| |
Collapse
|
93
|
Abstract
PURPOSE OF REVIEW IL-18 is a pleiotropic cytokine involved in the regulation of innate and adaptive immune responses. IL-18 pro-inflammatory activities are finely regulated in vivo by the inhibitory effects of the soluble IL-18-binding protein (IL-18BP). The elevation of circulating levels of IL-18 has been described in children with systemic juvenile idiopathic arthritis (sJIA). In the recent years, the role of IL-18 in the pathogenesis of secondary haemophagocytic lymphohistiocytosis (sHLH), also referred to as macrophage activation syndrome (MAS), in the context of autoinflammatory diseases, including sJIA, is emerging. RECENT FINDINGS A large number of studies in patients and animal models pointed to the imbalance in IL-18/IL-18BP levels, causing increased systemic levels of free bioactive IL-18, as a predisposing factor in the development of MAS. Although the exact mechanisms involved in the development of MAS are not clearly understood, increasing evidence demonstrate the role of IL-18 in upregulating the production of interferon (IFN)-γ. SUMMARY On the basis of the first emerging data on the possibility of blocking IL-18, we here discuss the scientific rationale for neutralizing the IL-18/IFNγ axis in the prevention and treatment of sHLH and MAS.
Collapse
|
94
|
Koga T, Sumiyoshi R, Furukawa K, Sato S, Migita K, Shimizu T, Umeda M, Endo Y, Fukui S, Kawashiri SY, Iwamoto N, Ichinose K, Tamai M, Nakamura H, Origuchi T, Nonaka F, Yachie A, Kondo H, Maeda T, Kawakami A. Interleukin-18 and fibroblast growth factor 2 in combination is a useful diagnostic biomarker to distinguish adult-onset Still's disease from sepsis. Arthritis Res Ther 2020; 22:108. [PMID: 32381117 PMCID: PMC7206754 DOI: 10.1186/s13075-020-02200-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/27/2020] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To identify potential biomarkers to distinguish adult-onset Still's disease (AOSD) from sepsis. METHOD We recruited 70 patients diagnosed with AOSD according to the Yamaguchi criteria, 22 patients with sepsis, and 118 age-matched controls. Serum levels of 40 cytokines were analyzed using multi-suspension cytokine array. We performed a cluster analysis of each cytokine in the AOSD and sepsis groups in order to identify specific molecular networks. Further, multivariate classification (random forest analysis) and logistic regression analysis were used to rank the cytokines by their importance and determine specific biomarkers for distinguishing AOSD from sepsis. RESULTS Seventeen of the 40 cytokines were found to be suitable for further analyses. The serum levels of eleven were significantly higher in patients with AOSD than healthy controls. Levels of serum fibroblast growth factor 2 (FGF-2), vascular endothelial growth factor (VEGF), granulocyte colony-stimulating factor (G-CSF), and interleukin (IL)-18 were significantly elevated in patients with AOSD compared with those with sepsis, and cytokine clustering patterns differed between these two groups. Multivariate classification followed by logistic regression analysis revealed that measurement of both FGF-2 and IL-18 could distinguish AOSD from sepsis with high accuracy (cutoff value for FGF-2 = 36 pg/mL; IL-18 = 543 pg/mL, sensitivity 100%, specificity 72.2%, accuracy 93.8%). CONCLUSION Determination of FGF-2 and IL-18 levels in combination may represent a biomarker for the differential diagnosis of AOSD from sepsis, based on the measurement of multiple cytokines.
Collapse
Affiliation(s)
- Tomohiro Koga
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan. .,Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan.
| | - Remi Sumiyoshi
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Kaori Furukawa
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shuntaro Sato
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Toshimasa Shimizu
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Masataka Umeda
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yushiro Endo
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shoichi Fukui
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shin-Ya Kawashiri
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naoki Iwamoto
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kunihiro Ichinose
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Mami Tamai
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hideki Nakamura
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomoki Origuchi
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Fumiaki Nonaka
- Department of Internal Medicine, Sasebo City General Hospital, Sasebo, Japan
| | - Akihiro Yachie
- Department of Pediatrics, School of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Hideaki Kondo
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takahiro Maeda
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Department of General Medicine, Nagasaki University Hospital, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
95
|
Abbate A, Toldo S, Marchetti C, Kron J, Van Tassell BW, Dinarello CA. Interleukin-1 and the Inflammasome as Therapeutic Targets in Cardiovascular Disease. Circ Res 2020; 126:1260-1280. [PMID: 32324502 DOI: 10.1161/circresaha.120.315937] [Citation(s) in RCA: 455] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intracellular sensing protein termed NLRP3 (for NACHT, LRR, and PYD domains-containing protein 3) forms a macromolecular structure called the NLRP3 inflammasome. The NLRP3 inflammasome plays a major role in inflammation, particularly in the production of IL (interleukin)-1β. IL-1β is the most studied of the IL-1 family of cytokines, including 11 members, among which are IL-1α and IL-18. Here, we summarize preclinical and clinical findings supporting the key pathogenetic role of the NLRP3 inflammasome and IL-1 cytokines in the formation, progression, and complications of atherosclerosis, in ischemic (acute myocardial infarction), and nonischemic injury to the myocardium (myocarditis) and the progression to heart failure. We also review the clinically available IL-1 inhibitors, although not currently approved for cardiovascular indications, and discuss other IL-1 inhibitors, not currently approved, as well as oral NLRP3 inflammasome inhibitors currently in clinical development. Canakinumab, IL-1β antibody, prevented the recurrence of ischemic events in patients with prior acute myocardial infarction in a large phase III clinical trial, including 10 061 patients world-wide. Phase II clinical trials show promising data with anakinra, recombinant IL-1 receptor antagonist, in patients with ST-segment-elevation acute myocardial infarction or heart failure with reduced ejection fraction. Anakinra also improved outcomes in patients with pericarditis, and it is now considered standard of care as second-line treatment for patients with recurrent/refractory pericarditis. Rilonacept, a soluble IL-1 receptor chimeric fusion protein neutralizing IL-1α and IL-1β, has also shown promising results in a phase II study in recurrent/refractory pericarditis. In conclusion, there is overwhelming evidence linking the NLRP3 inflammasome and the IL-1 cytokines with the pathogenesis of cardiovascular diseases. The future will likely include targeted inhibitors to block the IL-1 isoforms, and possibly oral NLRP3 inflammasome inhibitors, across a wide spectrum of cardiovascular diseases.
Collapse
Affiliation(s)
- Antonio Abbate
- From the VCU Pauley Heart Center, Virginia Commonwealth University, Richmond (A.A., S.T., J.K.)
| | - Stefano Toldo
- From the VCU Pauley Heart Center, Virginia Commonwealth University, Richmond (A.A., S.T., J.K.)
| | - Carlo Marchetti
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy, Richmond, VA (C.M., C.A.D.)
| | - Jordana Kron
- From the VCU Pauley Heart Center, Virginia Commonwealth University, Richmond (A.A., S.T., J.K.)
| | | | - Charles A Dinarello
- Department of Pharmacotherapy and Outcome Sciences, School of Pharmacy, Richmond, VA (C.M., C.A.D.)
| |
Collapse
|
96
|
Wedervang-Resell K, Friis S, Lonning V, Smelror RE, Johannessen C, Reponen EJ, Lyngstad SH, Lekva T, Aukrust P, Ueland T, Andreassen OA, Agartz I, Myhre AM. Increased interleukin 18 activity in adolescents with early-onset psychosis is associated with cortisol and depressive symptoms. Psychoneuroendocrinology 2020; 112:104513. [PMID: 31761332 DOI: 10.1016/j.psyneuen.2019.104513] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/14/2019] [Accepted: 11/12/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Evidence indicates that the pathophysiology of adult psychosis involves immune dysregulation, but its associations with stress are often not considered. The inflammatory cytokine interleukin (IL)-18, which is elevated in adult schizophrenia, is suggested to be sensitive to stress. We compared the associations of IL-18 with cortisol and clinical variables in adolescents with early-onset psychosis (EOP) aged 12-18 years and age-matched healthy controls (HC). METHOD We measured serum IL-18, IL-18 binding protein (IL-18BP), IL-18 receptor accessory protein (IL-18RAP), IL-18 receptor 1 (IL-18R1) and cortisol, and calculated the IL-18/IL-18BP ratio in patients (n = 31) and HC (n = 60). Psychotic symptoms were assessed using the Positive and Negative Syndrome Scale and depressive symptoms by the Mood and Feelings Questionnaire-Child version (MFQ-C). Bivariate correlation analysis was used to explore relationships between IL-18/IL-18BP ratio and cortisol, depression and other clinical characteristics. Hierarchical multiple linear regression analysis was used to assess their individual contributions to the variance of the IL-18/IL-18BP ratio. RESULTS Patients had significantly higher IL-18 levels and IL-18/IL-18BP ratios than HC, but similar IL-18BP, IL-18RAP and IL-18R1. Both cortisol (R2 change = 0.05) and the MFQ-C score (R2 change = 0.09) contributed significantly to the variance in IL-18/IL-18BP ratios after controlling for confounders. CONCLUSION We found increased IL-18 system activity in adolescents with EOP. Cortisol and depressive symptoms each contributed to the variance in the IL-18/IL-18BP ratio. Our findings support activation of inflammatory pathways in adolescent psychosis and suggest interactions between stress, inflammation and depressive symptoms in EOP.
Collapse
Affiliation(s)
- Kirsten Wedervang-Resell
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Division of Mental Health and Addiction, Department of Psychiatric Research and Development, Oslo University Hospital, Oslo, Norway.
| | - Svein Friis
- Division of Mental Health and Addiction, Department of Psychiatric Research and Development, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vera Lonning
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Runar E Smelror
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Cecilie Johannessen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Elina J Reponen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Siv H Lyngstad
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tove Lekva
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ingrid Agartz
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway; Centre for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anne M Myhre
- Division of Mental Health and Addiction, Department of Psychiatric Research and Development, Oslo University Hospital, Oslo, Norway; Child and Adolescent Psychiatry Unit, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
97
|
Liu S, Miersch S, Li P, Bai B, Liu C, Qin W, Su J, Huang H, Pan J, Sidhu SS, Wu D. A Synthetic Human Antibody Antagonizes IL-18Rβ Signaling Through an Allosteric Mechanism. J Mol Biol 2020; 432:1169-1182. [PMID: 31954129 DOI: 10.1016/j.jmb.2020.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 01/01/2023]
Abstract
The interleukin-18 subfamily belongs to the interleukin-1 family and plays an important role in modulating innate and adaptive immune responses. Dysregulation of IL-18 has been implicated in or correlated with numerous diseases, including inflammatory diseases, autoimmune disorders, and cancer. Thus, blockade of IL-18 signaling may offer therapeutic benefits in many pathological settings. Here, we report the development of synthetic human antibodies that target human IL-18Rβ and block IL-18-mediated IFN-γ secretion by inhibiting NF-κB and MAPK dependent pathways. The crystal structure of a potent antagonist antibody in complex with IL-18Rβ revealed inhibition through an unexpected allosteric mechanism. Our findings offer a novel means for therapeutic intervention in the IL-18 pathway and may provide a new strategy for targeting cytokine receptors.
Collapse
Affiliation(s)
- Shusu Liu
- Laboratory of Antibody Engineering, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Shane Miersch
- Banting and Best Department of Medical Research, Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Ping Li
- Laboratory of Antibody Engineering, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China; Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, China
| | - Bingxin Bai
- Laboratory of Antibody Engineering, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Chunchun Liu
- Laboratory of Antibody Engineering, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Wenming Qin
- National Facility for Protein Science (Shanghai), Shanghai Advanced Research Institute (Zhangjiang Lab), Chinese Academy of Sciences, Shanghai, China
| | - Jie Su
- Laboratory of Antibody Engineering, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Haiming Huang
- Banting and Best Department of Medical Research, Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada; Shanghai Asian United Antibody Medical Co., Shanghai, China
| | - James Pan
- Banting and Best Department of Medical Research, Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Sachdev S Sidhu
- Laboratory of Antibody Engineering, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China; Banting and Best Department of Medical Research, Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada.
| | - Donghui Wu
- Laboratory of Antibody Engineering, Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
98
|
Kiltz U, Kiefer D, Braun J, Schiffrin EJ, Girard-Guyonvarc'h C, Gabay C. Prolonged treatment with Tadekinig alfa in adult-onset Still's disease. Ann Rheum Dis 2020; 79:e10. [PMID: 30352887 DOI: 10.1136/annrheumdis-2018-214496] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 09/29/2018] [Indexed: 11/03/2022]
Affiliation(s)
- Uta Kiltz
- Rheumazentrum Ruhrgebiet, Herne, Germany
| | | | | | | | - Charlotte Girard-Guyonvarc'h
- Division of Rheumatology, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland
| | - Cem Gabay
- Division of Rheumatology, Department of Internal Medicine Specialties, University of Geneva, Geneva, Switzerland
| |
Collapse
|
99
|
Emerging Roles for Interleukin-18 in the Gastrointestinal Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1240:59-72. [PMID: 32060888 DOI: 10.1007/978-3-030-38315-2_5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Interleukin (IL)-18, a member of the IL-1 family of cytokines, has emerged as a key regulator of mucosal homeostasis within the gastrointestinal tract. Like other members of this family, IL-18 is secreted as an inactive protein and is processed into its active form by caspase-1, although other contributors to precursor processing are emerging.Numerous studies have evaluated the role of IL-18 within the gastrointestinal tract using genetic or complementary pharmacological tools and have revealed multiple roles in tumorigenesis. Most striking among these are the divergent roles for IL-18 in colon and gastric cancers. Here, we review our current understanding of IL-18 biology and how this applies to colorectal and gastric cancers.
Collapse
|
100
|
Pan Y, Wen X, Hao D, Wang Y, Wang L, He G, Jiang X. The role of IL-37 in skin and connective tissue diseases. Biomed Pharmacother 2019; 122:109705. [PMID: 31918276 DOI: 10.1016/j.biopha.2019.109705] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/12/2019] [Accepted: 11/24/2019] [Indexed: 02/05/2023] Open
Abstract
IL-37 was discovered as an anti-inflammatory and immunosuppressive cytokine of the IL-1 family. Significant advancements in the understanding of signaling pathways associated with IL-37 have been made in recent years. IL-37 binds to IL-18R and recruits IL-1R8 to form the IL-37/IL-1R8/IL-18Rα complex. Capase-1 plays a key role in the nuclear transduction of IL-37 signal, processing precursor IL-37 into the mature isoform, and interacting with Smad3. IL-37 exerts its role by activating anti-inflammation pathways including AMPK, PTEN, Mer, STAT3 and p62, and promoting tolerogenic dendritic cells and Tregs. In addition, IL-37 inhibits pro-inflammatory cytokines such as IL-1, IL-6, IL-8, IL-17, IL-23, TNF-α, and IFN-γ, and suppresses Fyn, MAPK, TAK1, NFκB, and mTOR signaling. The final effects of IL-37 depend on the interaction among IL-18R, IL-1R8, IL-37 and IL-18BP. Previous studies have deciphered the role of IL-37 in the development and pathogenesis of autoimmune diseases, chronic infections and cancer. In this review, we discuss the role of IL-37 in psoriasis, atopic dermatitis, Behcet's diseases, systemic lupus erythematosus, and other skin and connective tissue diseases.
Collapse
Affiliation(s)
- Yu Pan
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xiang Wen
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Dan Hao
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yujia Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lian Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Gu He
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, PR China.
| | - Xian Jiang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|