1
|
Baryła M, Skrzycki M, Danielewicz R, Kosieradzki M, Struga M. Protein biomarkers in assessing kidney quality before transplantation‑current status and future perspectives (Review). Int J Mol Med 2024; 54:107. [PMID: 39370783 PMCID: PMC11448562 DOI: 10.3892/ijmm.2024.5431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/31/2024] [Indexed: 10/08/2024] Open
Abstract
To meet the demand for kidney transplants (KTx), organs are frequently retrieved not only from standard criteria donors (SCD; a donor who is aged <50 years and suffered brain death from any number of causes, such as traumatic injuries or a stroke) but also from expanded criteria donors (any donor aged >60 years or donors aged >50 years with two of the following: A history of high blood pressure, a creatinine serum level ≥1.5 mg/dl or death resulting from a stroke). This comes at the cost of a higher risk of primary non‑function (the permanent hyperkalemia, hyperuremia and fluid overload that result in the need for continuous dialysis after KTx), delayed graft function (the need for dialysis session at least once during the first week after KTx), earlier graft loss and urinary complications (vesico‑ureteral reflux, obstruction of the vesico‑ureteral anastomosis, urine leakage). At present, there are no commercially available diagnostic tools for assessing kidney quality prior to KTx. Currently available predictive models based on clinical data, such as the Kidney Donor Profile Index, are insufficient. One promising option is the application of perfusion solutions for protein biomarkers of kidney quality and predictors of short‑ and long‑term outcomes. However, to date, protein markers that can be detected with ELISA, western blotting and cytotoxic assays have not been identified to be a beneficial predictors of kidney quality. These include lactate dehydrogenases, glutathione S‑transferases, fatty acid binding proteins, extracellular histones, IL‑18, neutrophil gelatinase‑associated lipocalin, MMPs and kidney injury molecule‑1. However, novel methods, including liquid chromatography‑mass spectrometry (LC‑MS) and microarrays, allow the analysis of all renal proteins suspended/dissolved in the acellular preservation solution used for kidney storage before KTx (including hypothermic machine perfusion as one of kidney storage methods) e.g. Belzer University of Wisconsin. Recent proteomic studies utilizing LC‑MS have identified complement pathway elements (C3, C1QB, C4BPA, C1S, C1R and C1RL), desmoplakin, blood coagulation pathway elements and immunoglobulin heavy variable 2‑26 to be novel predictors of kidney quality before transplantation. This was because they were found to correlate with estimated glomerular filtration rate at 3 and 12 months after kidney transplantation. However, further proteomic studies focusing on distinct markers obtained from hypothermic and normothermic machine perfusion are needed to confirm their predictive value and to improve kidney storage methods. Therefore, the present literature review from PubMed, Scopus, Embase and Web of Science was performed with the aims of summarizing the current knowledge on the most frequently studied single protein biomarkers. In addition, novel analytical methods and insights into organ injury during preservation were documented, where future directions in assessing organ quality before kidney transplantation were also discussed.
Collapse
Affiliation(s)
- Maksymilian Baryła
- Chair and Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of General and Transplant Surgery, Infant Jesus Hospital, Medical University of Warsaw, 02-006 Warsaw, Poland
| | - Michał Skrzycki
- Chair and Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Roman Danielewicz
- Department of General and Transplant Surgery, Infant Jesus Hospital, Medical University of Warsaw, 02-006 Warsaw, Poland
| | - Maciej Kosieradzki
- Department of General and Transplant Surgery, Infant Jesus Hospital, Medical University of Warsaw, 02-006 Warsaw, Poland
| | - Marta Struga
- Department of General and Transplant Surgery, Infant Jesus Hospital, Medical University of Warsaw, 02-006 Warsaw, Poland
| |
Collapse
|
2
|
Yu C, Xu H, Jiang S, Sun L. IL-18 signaling is regulated by caspase 6/8 and IL-18BP in turbot (Scophthalmus maximus). Int J Biol Macromol 2024; 278:135015. [PMID: 39181350 DOI: 10.1016/j.ijbiomac.2024.135015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Interleukin (IL)-18 is synthesized as a precursor that requires intracellular processing to become functionally active. In human, IL-18 is processed by caspase 1 (CASP1). In teleost, the maturation and signal transduction mechanisms of IL-18 are unknown. We identified two IL-18 variants, IL-18a and IL-18b, in turbot. IL-18a, but not IL-18b, was processed by CASP6/8 cleavage. Mature IL-18a bound specifically to IL-18 receptor (IL-18R) α-expressing cells and induced IL-18Rα-IL-18Rβ association. Bacterial infection promoted IL-18a maturation in a manner that required CASP6 activation and correlated with gasdermin E activation. The mature IL-18a induced proinflammatory cytokine expression and enhanced bacterial clearance. IL-18a-mediated immune response was suppressed by IL-18 binding protein (IL-18BP), which functioned as a decoy receptor for IL-18a. IL-18BP also functioned as a pathogen pattern recognition receptor and directly inhibited pathogen infection. Our findings revealed unique mechanism of IL-18 maturation and conserved mechanism of IL-18 signaling and regulation in turbot, and provided new insights into the regulation and function of IL-18 related immune signaling.
Collapse
Affiliation(s)
- Chao Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; School of Foundational Education, University of Health and Rehabilitation Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China; College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hang Xu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China; College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shuai Jiang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China; College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing, China.
| | - Li Sun
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China; College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
3
|
Taheri M, Tehrani HA, Daliri F, Alibolandi M, Soleimani M, Shoari A, Arefian E, Ramezani M. Bioengineering strategies to enhance the interleukin-18 bioactivity in the modern toolbox of cancer immunotherapy. Cytokine Growth Factor Rev 2024; 75:65-80. [PMID: 37813764 DOI: 10.1016/j.cytogfr.2023.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/11/2023]
Abstract
Cytokines are the first modern immunotherapeutic agents used for activation immunotherapy. Interleukin-18 (IL-18) has emerged as a potent anticancer immunostimulatory cytokine over the past three decades. IL-18, structurally is a stable protein with very low toxicity at biological doses. IL-18 promotes the process of antigen presentation and also enhances innate and acquired immune responses. It can induce the production of proinflammatory cytokines and increase tumor infiltration of effector immune cells to revert the immunosuppressive milieu of tumors. Furthermore, IL-18 can reduce tumorigenesis, suppress tumor angiogenesis, and induce tumor cell apoptosis. These characteristics present IL-18 as a promising option for cancer immunotherapy. Although several preclinical studies have reported the immunotherapeutic potential of IL-18, clinical trials using it as a monotherapy agent have reported disappointing results. These results may be due to some biological characteristics of IL-18. Several bioengineering approaches have been successfully used to correct its defects as a bioadjuvant. Currently, the challenge with this anticancer immunotherapeutic agent is mainly how to use its capabilities in a rational combinatorial therapy for clinical applications. The present study discussed the strengths and weaknesses of IL-18 as an immunotherapeutic agent, followed by comprehensive review of various promising bioengineering approaches that have been used to overcome its disadvantages. Finally, this study highlights the promising application of IL-18 in modern combinatorial therapies, such as chemotherapy, immune checkpoint blockade therapy, cell-based immunotherapy and cancer vaccines to guide future studies, circumventing the barriers to administration of IL-18 for clinical applications, and bring it to fruition as a potent immunotherapy agent in cancer treatment.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | | | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoud Soleimani
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Iran
| | - Alireza Shoari
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran; Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Landy E, Carol H, Ring A, Canna S. Biological and clinical roles of IL-18 in inflammatory diseases. Nat Rev Rheumatol 2024; 20:33-47. [PMID: 38081945 DOI: 10.1038/s41584-023-01053-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 12/23/2023]
Abstract
Several new discoveries have revived interest in the pathogenic potential and possible clinical roles of IL-18. IL-18 is an IL-1 family cytokine with potent ability to induce IFNγ production. However, basic investigations and now clinical observations suggest a more complex picture. Unique aspects of IL-18 biology at the levels of transcription, activation, secretion, neutralization, receptor distribution and signalling help to explain its pleiotropic roles in mucosal and systemic inflammation. Blood biomarker studies reveal a cytokine for which profound elevation, associated with detectable 'free IL-18', defines a group of autoinflammatory diseases in which IL-18 dysregulation can be a primary driving feature, the so-called 'IL-18opathies'. This impressive specificity might accelerate diagnoses and identify patients amenable to therapeutic IL-18 blockade. Pathogenically, human and animal studies identify a preferential activation of CD8+ T cells over other IL-18-responsive lymphocytes. IL-18 agonist treatments that leverage the site of production or subversion of endogenous IL-18 inhibition show promise in augmenting immune responses to cancer. Thus, the unique aspects of IL-18 biology are finally beginning to have clinical impact in precision diagnostics, disease monitoring and targeted treatment of inflammatory and malignant diseases.
Collapse
Affiliation(s)
- Emily Landy
- Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hallie Carol
- Division of Rheumatology and Immune Dysregulation Program, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aaron Ring
- Translational Science and Therapeutics, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Scott Canna
- Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Division of Rheumatology and Immune Dysregulation Program, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Zhang Y, Yang Y, Li H, Feng Q, Ge W, Xu X. Investigating the Potential Mechanisms and Therapeutic Targets of Inflammatory Cytokines in Post-stroke Depression. Mol Neurobiol 2024; 61:132-147. [PMID: 37592185 DOI: 10.1007/s12035-023-03563-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023]
Abstract
Post-stroke depression (PSD) affects approximately one-third of stroke survivors, severely impacting general recovery and quality of life. Despite extensive studies, the exact mechanisms underlying PSD remain elusive. However, emerging evidence implicates proinflammatory cytokines, including interleukin-1β, interleukin-6, tumor necrosis factor-alpha, and interleukin-18, play critical roles in PSD development. These cytokines contribute to PSD through various mechanisms, including hypothalamic-pituitary-adrenal (HPA) axis dysfunction, neurotransmitter alterations, neurotrophic factor changes, gut microbiota imbalances, and genetic predispositions. This review is aimed at exploring the role of cytokines in stroke and PSD while identifying their potential as specific therapeutic targets for managing PSD. A more profound understanding of the mechanisms regulating inflammatory cytokine expression and anti-inflammatory cytokines like interleukin-10 in PSD may facilitate the development of innovative interventions to improve outcomes for stroke survivors experiencing depression.
Collapse
Affiliation(s)
- Yutong Zhang
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Yuehua Yang
- Department of Neurology, Suzhou Yongding Hospital, Suzhou, 215028, China
| | - Hao Li
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Qian Feng
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Wei Ge
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221600, China.
| | - Xingshun Xu
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
6
|
Yamanishi K, Hata M, Gamachi N, Watanabe Y, Yamanishi C, Okamura H, Matsunaga H. Molecular Mechanisms of IL18 in Disease. Int J Mol Sci 2023; 24:17170. [PMID: 38139000 PMCID: PMC10743479 DOI: 10.3390/ijms242417170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
Interleukin 18 (IL18) was originally identified as an inflammation-induced cytokine that is secreted by immune cells. An increasing number of studies have focused on its non-immunological functions, with demonstrated functions for IL18 in energy homeostasis and neural stability. IL18 is reportedly required for lipid metabolism in the liver and brown adipose tissue. Furthermore, IL18 (Il18) deficiency in mice leads to mitochondrial dysfunction in hippocampal cells, resulting in depressive-like symptoms and cognitive impairment. Microarray analyses of Il18-/- mice have revealed a set of genes with differential expression in liver, brown adipose tissue, and brain; however, the impact of IL18 deficiency in these tissues remains uncertain. In this review article, we discuss these genes, with a focus on their relationships with the phenotypic disease traits of Il18-/- mice.
Collapse
Affiliation(s)
- Kyosuke Yamanishi
- Department of Neuropsychiatry, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Masaki Hata
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Naomi Gamachi
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Yuko Watanabe
- Hirakata General Hospital for Developmental Disorders, Hirakata 573-0122, Osaka, Japan; (Y.W.); (C.Y.)
| | - Chiaki Yamanishi
- Hirakata General Hospital for Developmental Disorders, Hirakata 573-0122, Osaka, Japan; (Y.W.); (C.Y.)
| | - Haruki Okamura
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| | - Hisato Matsunaga
- Department of Neuropsychiatry, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
- Department of Psychoimmunology, Hyogo Medical University, 1-1 Mukogawa, Nishinomiya 663-8501, Hyogo, Japan
| |
Collapse
|
7
|
Baggio C, Bindoli S, Guidea I, Doria A, Oliviero F, Sfriso P. IL-18 in Autoinflammatory Diseases: Focus on Adult Onset Still Disease and Macrophages Activation Syndrome. Int J Mol Sci 2023; 24:11125. [PMID: 37446301 DOI: 10.3390/ijms241311125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Interleukin-18 (IL-18) is a potent pro-inflammatory cytokine that is involved in various innate and adaptive immune processes related to infection, inflammation, and autoimmunity. Therefore, it is described as a key mediator of autoinflammatory diseases associated with the development of macrophage activation syndrome (MAS), including systemic juvenile idiopathic arthritis and adult-onset Still's disease. This review focuses on the role of IL-18 in inflammatory responses, placing emphasis on autoinflammatory diseases associated with chronic excess of serum IL-18, which correlate with clinical and biological signs of the disease. Therefore, it is useful for the diagnosis and monitoring of disease activity. Researchers are currently investigating IL-18's role as a therapeutic target for the treatment of inflammatory diseases. The inhibition of IL-18 signaling through recombinant human IL-18BP (IL-18 binding protein) seems to be an effective therapeutic strategy, though further studies are necessary to clarify its importance as a therapeutic target.
Collapse
Affiliation(s)
- Chiara Baggio
- Rheumatology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Sara Bindoli
- Rheumatology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Irina Guidea
- Rheumatology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Francesca Oliviero
- Rheumatology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy
| | - Paolo Sfriso
- Rheumatology Unit, Department of Medicine, University of Padova, 35128 Padova, Italy
| |
Collapse
|
8
|
Cirella A, Olivera I, Luri-Rey C, Bolaños E, Berraondo P, Melero I. Interleukin-18 in cancer immunology and immunotherapy. Expert Opin Ther Targets 2023; 27:1035-1042. [PMID: 37993172 DOI: 10.1080/14728222.2023.2287574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
INTRODUCTION Interleukin-18 (IL-18) is a myeloid leukocyte inflammatory mediator whose main known function is to elicit IFNγ secretion from T and NK cells. AREAS COVERED This function offers potential in cancer immunotherapy but as a single treatment, preclinical and clinical antitumor activities are modest. IL-18 bioactivity is chiefly downregulated by a decoy soluble receptor named IL18-binding protein (IL-18BP) that is induced by IFNγ as a negative feedback mechanism. Recent advances indicate promising efficacy of IL-18 at armoring CAR-T cells for the treatment of hematological malignancies. Preclinical research has also yielded IL-18 constructs that do not bind IL-18BP but have preserved activity on the receptor and exert markedly increased antitumor effects. Indeed, agents of this kind are undergoing clinical trials. The synergistic effects of IL-18 and IL-12 in combination to induce IFNγ are extremely potent but are toxic if systemically delivered. In mouse models, IL-12 and decoy-resistant variants of IL-18 can be efficaciously used as local treatments for tumors by exploiting mRNA intratumoral co-delivery. Moreover, antitumor T cells can be transiently engineered with mRNAs encoding this combination of cytokines to attain efficacious synergistic effects also upon intratumoral delivery. EXPERT OPINION IL-18 certainly holds promise for immunotherapy in combination with other agents and for local approaches.
Collapse
Affiliation(s)
- Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Irene Olivera
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Elixabet Bolaños
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
- Department of Immunology and immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
- Department of oncology, Clínica Universidad de Navarra, Madrid, Spain
- Centro del Cancer de la Universidad de Navarra (CCUN), Pamplona, Spain
- Nuffield Department of Medicine (NDM), University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Johnson AF, Sands JS, Trivedi KM, Russell R, LaRock DL, LaRock CN. Constitutive secretion of pro-IL-18 allows keratinocytes to initiate inflammation during bacterial infection. PLoS Pathog 2023; 19:e1011321. [PMID: 37068092 PMCID: PMC10138833 DOI: 10.1371/journal.ppat.1011321] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/27/2023] [Accepted: 03/27/2023] [Indexed: 04/18/2023] Open
Abstract
Group A Streptococcus (GAS, Streptococcus pyogenes) is a professional human pathogen that commonly infects the skin. Keratinocytes are one of the first cells to contact GAS, and by inducing inflammation, they can initiate the earliest immune responses to pathogen invasion. Here, we characterized the proinflammatory cytokine repertoire produced by primary human keratinocytes and surrogate cell lines commonly used in vitro. Infection induces several cytokines and chemokines, but keratinocytes constitutively secrete IL-18 in a form that is inert (pro-IL-18) and lacks proinflammatory activity. Canonically, IL-18 activation and secretion are coupled through a single proteolytic event that is regulated intracellularly by the inflammasome protease caspase-1 in myeloid cells. The pool of extracellular pro-IL-18 generated by keratinocytes is poised to sense extracellular proteases. It is directly processed into a mature active form by SpeB, a secreted GAS protease that is a critical virulent factor during skin infection. This mechanism contributes to the proinflammatory response against GAS, resulting in T cell activation and the secretion of IFN-γ. Under these conditions, isolates of several other major bacterial pathogens and microbiota of the skin were found to not have significant IL-18-maturing ability. These results suggest keratinocyte-secreted IL-18 is a sentinel that sounds an early alarm that is highly sensitive to GAS, yet tolerant to non-invasive members of the microbiota.
Collapse
Affiliation(s)
- Anders F Johnson
- Department of Microbiology and Immunology and Department of Medicine, Emory School of Medicine, Atlanta, Georgia, United States of America
| | - Jenna S Sands
- Department of Microbiology and Immunology and Department of Medicine, Emory School of Medicine, Atlanta, Georgia, United States of America
| | - Keya M Trivedi
- Department of Microbiology and Immunology and Department of Medicine, Emory School of Medicine, Atlanta, Georgia, United States of America
| | - Raedeen Russell
- Department of Microbiology and Immunology and Department of Medicine, Emory School of Medicine, Atlanta, Georgia, United States of America
| | - Doris L LaRock
- Department of Microbiology and Immunology and Department of Medicine, Emory School of Medicine, Atlanta, Georgia, United States of America
| | - Christopher N LaRock
- Department of Microbiology and Immunology and Department of Medicine, Emory School of Medicine, Atlanta, Georgia, United States of America
- Department of Medicine, Division of Infectious Diseases, Emory School of Medicine, Atlanta, Georgia, United States of America
- Emory Antibiotic Resistance Center, Atlanta, Georgia, United States of America
| |
Collapse
|
10
|
Yang YC, Chen SN, Gan Z, Huang L, Li N, Wang KL, Nie P. Functional characterization of IL-18 receptor subunits, IL-18Rα and IL-18Rβ, and its natural inhibitor, IL-18 binding protein (IL-18BP) in rainbow trout Oncorhynchus mykiss. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 140:104610. [PMID: 36496012 DOI: 10.1016/j.dci.2022.104610] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 06/17/2023]
Abstract
As an important proinflammation and immunomodulatory cytokine, IL-18 has been reported in several species of fish, but its receptor subunits, IL-18Rα and IL-18Rβ, and its decoy receptor, IL-18BP, have not been functionally characterized in fish. In the present study, IL-18Rα, IL-18Rβ and IL-18BP were cloned from rainbow trout Oncorhynchus mykiss, and they possess common conserved domains with their mammalian orthologues. In tested organs/tissues, IL-18Rα and IL-18Rβ exhibit basal expression levels, and IL-18BP has a pattern of constitutive expression. When transfected with different combinations of chimeric receptors in HEK293T cells, recombinant IL-18 (rIL-18) can induce the activation of NF-κB only when pcDNA3.1-IL-18Rα/IL-1R1 and pcDNA3.1-IL-18Rβ/IL-1RAP were both expressed. On the other hand, recombinant receptors, including rIL-18BP, rIL-18Rα-ECD-Fc and rIL-18Rβ-ECD-Fc can down-regulate significantly the activity of NF-κB, suggesting the participation of IL-18Rα, IL-18Rβ and IL-18BP in rainbow trout IL-18 signal transduction. Co-IP assays indicated that IL-18Rβ may form a complex with MyD88, IRAK4, IRAK1, TRAF6 and TAB2 in HEK293T cells, indicating that IL-18Rβ, in IL-18 signalling pathway, is associated with these signalling molecules. In conclusion, IL-18Rα, IL-18Rβ and IL-18BP in rainbow trout are conserved in function and signalling pathway with their mammalian orthologues.
Collapse
Affiliation(s)
- Yue Chong Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Shan Nan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Zhen Gan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Lin Huang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Nan Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - Kai Lun Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China
| | - P Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, Shandong Province, 266237, China; School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China.
| |
Collapse
|
11
|
Deraison C, Bonnart C, Langella P, Roget K, Vergnolle N. Elafin and its precursor trappin-2: What is their therapeutic potential for intestinal diseases? Br J Pharmacol 2023; 180:144-160. [PMID: 36355635 PMCID: PMC10098471 DOI: 10.1111/bph.15985] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/22/2022] [Accepted: 05/07/2022] [Indexed: 11/12/2022] Open
Abstract
Elafin and its precursor trappin-2 are known for their contribution to the physiological mucosal shield against luminal microbes. Such a contribution seems to be particularly relevant in the gut, where the exposure of host tissues to heavy loads of microbes is constant and contributes to mucosa-associated pathologies. The expression of trappin-2/elafin has been shown to be differentially regulated in diseases associated with gut inflammation. Accumulating evidence has demonstrated the protective effects of trappin-2/elafin in gut intestinal disorders associated with acute or chronic inflammation, or with gluten sensitization disorders. The protective effects of trappin-2/elafin in the gut are discussed in terms of their pleiotropic modes of action: acting as protease inhibitors, transglutaminase substrates, antimicrobial peptides or as a regulator of pro-inflammatory transcription factors. Further, the question of the therapeutic potential of trappin-2/elafin delivery at the intestinal mucosa surface is raised. Whether trappin-2/elafin mucosal delivery should be considered to ensure intestinal tissue repair is also discussed.
Collapse
Affiliation(s)
- Céline Deraison
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Chrystelle Bonnart
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Philippe Langella
- Université Paris-Saclay, AgroParisTech, Micalis Institute, INRAE, Jouy-en-Josas, France
| | | | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France.,Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
12
|
Harju N. Regulation of oxidative stress and inflammatory responses in human retinal pigment epithelial cells. Acta Ophthalmol 2022; 100 Suppl 273:3-59. [DOI: 10.1111/aos.15275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Niina Harju
- School of Pharmacy University of Eastern Finland Kuopio Finland
| |
Collapse
|
13
|
Acute stress induces severe neural inflammation and overactivation of glucocorticoid signaling in interleukin-18-deficient mice. Transl Psychiatry 2022; 12:404. [PMID: 36151082 PMCID: PMC9508168 DOI: 10.1038/s41398-022-02175-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 12/01/2022] Open
Abstract
Interleukin-18 (IL18) is an inflammatory cytokine that is related to psychiatric disorders such as depression and cognitive impairment. We previously found that IL18 deficiency may cause hippocampal impairment, resulting in depression-like behavioral changes. However, the potential role of IL18 in stressful conditions remains uncertain. In the present study, we examined the effect of IL18 on neural inflammation and stress tolerance during acute stress. Littermate Il18+/+ and Il18-/- mice were exposed to a single restraint stress for 6 h, and all assessments were performed 18 h after the mice were released from the restraint. In Il18-/- mice exposed to acute stress, the immobility times in both the forced swim test and tail suspension test were decreased, although no difference was observed in Il18+/+ mice. Il1β, Il6, and Tnfα expression levels in the hippocampus of stressed Il18-/- mice were significantly higher than those in the other groups. Moreover, the numbers of astrocytes and microglia, including those in the active form, were also increased compared with those in other groups. Regarding the molecular mechanism, the HSF5 and TTR genes were specifically expressed in stressed Il18-/- mice. As a potential treatment, intracerebral administration of IL18 to Il18-/- mice resulted in partial recovery of changes in behavioral assessments. Our results revealed that IL18-deficient mice were more sensitive and had a longer response to acute stress than that in normal mice. In addition, neural inflammation and augmentation of glucocorticoid signals caused by stress were more intense and remained longer in Il18-/- mice, resulting in behavioral changes. In conclusion, IL18 might be an indispensable factor that modulates the stress response and maintains balance between neural inflammation and glucocorticoid signaling.
Collapse
|
14
|
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: 100] [Impact Index Per Article: 50.0] [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
|
15
|
Pan H, Huan C, Zhang W, Hou Y, Zhou Z, Yao J, Gao S. PDZK1 upregulates nitric oxide production through the PI3K/ERK2 pathway to inhibit porcine circovirus type 2 replication. Vet Microbiol 2022; 272:109514. [PMID: 35917623 DOI: 10.1016/j.vetmic.2022.109514] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 07/01/2022] [Accepted: 07/14/2022] [Indexed: 10/17/2022]
Abstract
Porcine circovirus type 2 (PCV2) is the causative agent of porcine circovirus-associated disease. Changes in host cell gene expression are induced by PCV2 infection. Here, we showed that porcine PDZ Domain-Containing 1 (PDZK1) expression was enhanced during PCV2 infection and that overexpression of PDZK1 inhibited the expression of PCV2 Cap protein. PCV2 genomic DNA copy number and viral titers were decreased in PDZK1-overexpressing PK-15B6 cells. PDZK1 knockdown enhanced the replication of PCV2. Overexpression of PDZK1 activated the phosphoinositide 3-kinase (PI3K)/ERK2 signaling pathway to enhance nitric oxide (NO) levels, while PDZK1 knockdown had the opposite effects. A PI3K inhibitor (LY294002) and a NO synthase inhibitor (L-NAME hydrochloride) decreased the activity of PDZK1 in restricting PCV2 replication. ERK2 knockdown enhanced the proliferation of PCV2 by decreasing levels of NO. Levels of interleukin (IL)- 4 mRNA were reduced in PDZK1 knockdown and ERK2 knockdown PK-15B6 cells. Increased IL-4 mRNA levels were unable to decrease NO production in PDZK1-overexpressing cells. Thus, we conclude that PDZK1 affected PCV2 replication by regulating NO production via PI3K/ERK2 signaling. PDZK1 affected IL-4 expression through the PI3K/ERK2 pathway, but PDZK1 modulation of PCV2 replication occurred independently of IL-4. Our results contribute to understanding the biological functions of PDZK1 and provide a theoretical basis for the pathogenic mechanisms of PCV2.
Collapse
Affiliation(s)
- Haochun Pan
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Changchao Huan
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Wei Zhang
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Yutong Hou
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Ziyan Zhou
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Jingting Yao
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China
| | - Song Gao
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, Jiangsu, China; Key Laboratory of Avian Bioproduct Development, Ministry of Agriculture and Rural Affairs, Yangzhou 225009, Jiangsu, China; College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, China.
| |
Collapse
|
16
|
Martynova E, Rizvanov A, Urbanowicz RA, Khaiboullina S. Inflammasome Contribution to the Activation of Th1, Th2, and Th17 Immune Responses. Front Microbiol 2022; 13:851835. [PMID: 35369454 PMCID: PMC8969514 DOI: 10.3389/fmicb.2022.851835] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/22/2022] [Indexed: 12/24/2022] Open
Abstract
Inflammasomes are cytosolic polyprotein complexes formed in response to various external and internal stimuli, including viral and bacterial antigens. The main product of the inflammasome is active caspase 1 which proteolytically cleaves, releasing functional interleukin-1 beta (IL-1β) and interleukin-18 (IL-18). These cytokines play a central role in shaping immune response to pathogens. In this review, we will focus on the mechanisms of inflammasome activation, as well as their role in development of Th1, Th2, and Th17 lymphocytes. The contribution of cytokines IL-1β, IL-18, and IL-33, products of activated inflammasomes, are summarized. Additionally, the role of cytokines released from tissue cells in promoting differentiation of lymphocyte populations is discussed.
Collapse
Affiliation(s)
| | | | - Richard A. Urbanowicz
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | | |
Collapse
|
17
|
Rastogi S, Briken V. Interaction of Mycobacteria With Host Cell Inflammasomes. Front Immunol 2022; 13:791136. [PMID: 35237260 PMCID: PMC8882646 DOI: 10.3389/fimmu.2022.791136] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/13/2022] [Indexed: 12/17/2022] Open
Abstract
The inflammasome complex is important for host defense against intracellular bacterial infections. Mycobacterium tuberculosis (Mtb) is a facultative intracellular bacterium which is able to survive in infected macrophages. Here we discuss how the host cell inflammasomes sense Mtb and other related mycobacterial species. Furthermore, we describe the molecular mechanisms of NLRP3 inflammasome sensing of Mtb which involve the type VII secretion system ESX-1, cell surface lipids (TDM/TDB), secreted effector proteins (LpqH, PPE13, EST12, EsxA) and double-stranded RNA acting on the priming and/or activation steps of inflammasome activation. In contrast, Mtb also mediates inhibition of the NLRP3 inflammasome by limiting exposure of cell surface ligands via its hydrolase, Hip1, by inhibiting the host cell cathepsin G protease via the secreted Mtb effector Rv3364c and finally, by limiting intracellular triggers (K+ and Cl- efflux and cytosolic reactive oxygen species production) via its serine/threonine kinase PknF. In addition, Mtb inhibits the AIM2 inflammasome activation via an unknown mechanism. Overall, there is good evidence for a tug-of-war between Mtb trying to limit inflammasome activation and the host cell trying to sense Mtb and activate the inflammasome. The detailed molecular mechanisms and the importance of inflammasome activation for virulence of Mtb or host susceptibility have not been fully investigated.
Collapse
Affiliation(s)
| | - Volker Briken
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States
| |
Collapse
|
18
|
Bonaventura A, Vecchié A, Dagna L, Tangianu F, Abbate A, Dentali F. Colchicine for COVID-19: targeting NLRP3 inflammasome to blunt hyperinflammation. Inflamm Res 2022; 71:293-307. [PMID: 35113170 PMCID: PMC8811745 DOI: 10.1007/s00011-022-01540-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/14/2021] [Accepted: 01/13/2022] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is capable of inducing the activation of NACHT, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome, a macromolecular structure sensing the danger and amplifying the inflammatory response. The main product processed by NLRP3 inflammasome is interleukin (IL)-1β, responsible for the downstream production of IL-6, which has been recognized as an important mediator in coronavirus disease 2019 (COVID-19). Since colchicine is an anti-inflammatory drug with the ability to block NLRP3 inflammasome oligomerization, this may prevent the release of active IL-1β and block the detrimental effects of downstream cytokines, i.e. IL-6. To date, few randomized clinical trials and many observational studies with colchicine have been conducted, showing interesting signals. As colchicine is a nonspecific inhibitor of the NLRP3 inflammasome, compounds specifically blocking this molecule might provide increased advantages in reducing the inflammatory burden and its related clinical manifestations. This may occur through a selective blockade of different steps preceding NLRP3 inflammasome oligomerization as well as through a reduced release of the main cytokines (IL-1β and IL-18). Since most evidence is based on observational studies, definitive conclusion cannot be drawn and additional studies are needed to confirm preliminary results and further dissect how colchicine and other NLRP3 inhibitors reduce the inflammatory burden and evaluate the timing and duration of treatment.
Collapse
Affiliation(s)
- Aldo Bonaventura
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy.
| | - Alessandra Vecchié
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Lorenzo Dagna
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS Ospedale San Raffaele, Milan, Italy
| | - Flavio Tangianu
- Medicina Generale 1, Medical Center, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | - Antonio Abbate
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Francesco Dentali
- Department of Medicine and Surgery, Insubria University, Varese, Italy
| |
Collapse
|
19
|
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: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/03/2021] [Indexed: 02/07/2023]
|
20
|
Azargoon A, Kharazmkia A, Kordalivand N, Birjandi M, Mir S. Evaluation of exposure to secondhand smoke and serum level of interleukin 18 in non-smokers. Ann Med Surg (Lond) 2022; 73:103238. [PMID: 35079372 PMCID: PMC8767295 DOI: 10.1016/j.amsu.2021.103238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 11/24/2022] Open
Abstract
Objective Smoking is one significant global health care problems, that not only affects the users but also endangers the health of people inhaling the smoke (passive smoking/secondhand smoke). The serum level of IL-18, an important regulator of inherent and acquired immune response, is affected by cigarette smoking. The aim of this study was to evaluate the effect of secondhand smoke (SHS) exposure on IL-18 serum level in non-smoker adults. Methods In a case-control study, using easy sampling method, 76 non-smokers who were exposed to cigarette smoke for at least 1 h daily during the past year were considered as exposure group, while 76 of their companions without exposure to cigarette smoke (after matching age) were considered as non-exposure group. Serum IL-18 levels were measured for all participants and finally compared between the two groups using Chi-square test. P value < 0.05 was considered to be statistically significant. Results The exposure and non-exposure groups included 58 (76.3%) and 25 (32.9%) males, respectively (P < 0.001). The mean ± SD of age for the exposure and non-exposure groups was 35.42 ± 10.37 and 38.47 ± 12.49 years, respectively (P = 0.102). There was no significant difference between the mean serum levels of IL-18 in the exposure (54.81 ± 57.03 ng/ml) and non-exposure (41.49 ± 42.14 ng/ml) groups (P = 0.104). Conclusion The exposure to secondhand smoke has no significant effect on serum level of IL-18 in exposed adult individuals. However, more studies with larger sample sizes on different populations are required to confirm these results. Smoking is one significant global health care problems. That not only affects the users but also endangers the health of people inhaling the smoke. The serum level of IL-18, an important regulator of inherent and acquired immune response. The exposure to secondhand smoke has no significant effect on serum level of IL-18.
Collapse
Affiliation(s)
- Alireza Azargoon
- Department of Internal Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Kharazmkia
- Department of Clinical Pharmacy, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Nazanin Kordalivand
- Department of Internal Medicine, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mehdi Birjandi
- Department of Biostatistics and Epidemiology, School of Health and Nutrition, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Samareh Mir
- Nutritional Health Research Center, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
21
|
Thomas JM, Ling YH, Huuskes B, Jelinic M, Sharma P, Saini N, Ferens DM, Diep H, Krishnan SM, Kemp-Harper BK, O'Connor PM, Latz E, Arumugam TV, Guzik TJ, Hickey MJ, Mansell A, Sobey CG, Vinh A, Drummond GR. IL-18 (Interleukin-18) Produced by Renal Tubular Epithelial Cells Promotes Renal Inflammation and Injury During Deoxycorticosterone/Salt-Induced Hypertension in Mice. Hypertension 2021; 78:1296-1309. [PMID: 34488433 DOI: 10.1161/hypertensionaha.120.16437] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
IL-18 (interleukin-18) is elevated in hypertensive patients, but its contribution to high blood pressure and end-organ damage is unknown. We examined the role of IL-18 in the development of renal inflammation and injury in a mouse model of low-renin hypertension. Hypertension was induced in male C57BL6/J (WT) and IL-18−/− mice by uninephrectomy, deoxycorticosterone acetate (2.4 mg/d, s.c.) and 0.9% drinking saline (1K/DOCA/salt). Normotensive controls received uninephrectomy and placebo (1K/placebo). Blood pressure was measured via tail cuff or radiotelemetry. After 21 days, kidneys were harvested for (immuno)histochemical, quantitative-PCR and flow cytometric analyses of fibrosis, inflammation, and immune cell infiltration. 1K/DOCA/salt-treated WT mice developed hypertension, renal fibrosis, upregulation of proinflammatory genes, and accumulation of CD3+ T cells in the kidneys. They also displayed increased expression of IL-18 on tubular epithelial cells. IL-18−/− mice were profoundly protected from hypertension, renal fibrosis, and inflammation. Bone marrow transplantation between WT and IL-18−/− mice revealed that IL-18-deficiency in non-bone marrow-derived cells alone afforded equivalent protection against hypertension and renal injury as global IL-18 deficiency. IL-18 receptor subunits—interleukin-18 receptor 1 and IL-18R accessory protein—were upregulated in kidneys of 1K/DOCA/salt-treated WT mice and localized to T cells and tubular epithelial cells. T cells from kidneys of 1K/DOCA/salt-treated mice produced interferon-γ upon ex vivo stimulation with IL-18, whereas those from 1K/placebo mice did not. In conclusion, IL-18 production by tubular epithelial cells contributes to elevated blood pressure, renal inflammation, and fibrosis in 1K/DOCA/salt-treated mice, highlighting it as a promising therapeutic target for hypertension and kidney disease.
Collapse
Affiliation(s)
- Jordyn M Thomas
- Centre for Cardiovascular Biology and Disease Research and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia (J.M.T., B.M.H., M.J., P.S., N.S., H.D., T.V.A., C.G.S., A.V., G.R.D.)
| | - Yeong H Ling
- Department of Pharmacology, Biomedicine Discovery Institute, Cardiovascular Disease Program, Monash University, Clayton, Australia (Y.H.L., D.M.F., S.M.K., B.K.K.-H.)
| | - Brooke Huuskes
- Centre for Cardiovascular Biology and Disease Research and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia (J.M.T., B.M.H., M.J., P.S., N.S., H.D., T.V.A., C.G.S., A.V., G.R.D.)
| | - Maria Jelinic
- Centre for Cardiovascular Biology and Disease Research and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia (J.M.T., B.M.H., M.J., P.S., N.S., H.D., T.V.A., C.G.S., A.V., G.R.D.)
| | - Prerna Sharma
- Centre for Cardiovascular Biology and Disease Research and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia (J.M.T., B.M.H., M.J., P.S., N.S., H.D., T.V.A., C.G.S., A.V., G.R.D.)
| | - Narbada Saini
- Centre for Cardiovascular Biology and Disease Research and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia (J.M.T., B.M.H., M.J., P.S., N.S., H.D., T.V.A., C.G.S., A.V., G.R.D.)
| | - Dorota M Ferens
- Department of Pharmacology, Biomedicine Discovery Institute, Cardiovascular Disease Program, Monash University, Clayton, Australia (Y.H.L., D.M.F., S.M.K., B.K.K.-H.)
| | - Henry Diep
- Centre for Cardiovascular Biology and Disease Research and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia (J.M.T., B.M.H., M.J., P.S., N.S., H.D., T.V.A., C.G.S., A.V., G.R.D.)
| | - Shalini M Krishnan
- Department of Pharmacology, Biomedicine Discovery Institute, Cardiovascular Disease Program, Monash University, Clayton, Australia (Y.H.L., D.M.F., S.M.K., B.K.K.-H.)
| | - Barbara K Kemp-Harper
- Department of Pharmacology, Biomedicine Discovery Institute, Cardiovascular Disease Program, Monash University, Clayton, Australia (Y.H.L., D.M.F., S.M.K., B.K.K.-H.)
| | - Paul M O'Connor
- Department of Physiology, Medical College of Georgia, Augusta University (P.M.O.)
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital, University of Bonn, Germany (E.L.)
- German Center for Neurodegenerative Diseases, Bonn, Germany (E.L.)
| | - Thiruma V Arumugam
- Centre for Cardiovascular Biology and Disease Research and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia (J.M.T., B.M.H., M.J., P.S., N.S., H.D., T.V.A., C.G.S., A.V., G.R.D.)
| | - Tomasz J Guzik
- Department of Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland (T.J.G.)
- BHF Centre of Research Excellence, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (T.J.G.)
| | - Michael J Hickey
- Department of Medicine, Centre for Inflammatory Diseases, Monash University, Clayton, Australia (M.J.H.)
| | - Ashley Mansell
- Hudson Institute of Medical Research, Clayton, Australia (A.M.)
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia (J.M.T., B.M.H., M.J., P.S., N.S., H.D., T.V.A., C.G.S., A.V., G.R.D.)
- Baker Heart and Diabetes Institute, Prahran, Australia (C.G.S., G.R.D.)
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia (J.M.T., B.M.H., M.J., P.S., N.S., H.D., T.V.A., C.G.S., A.V., G.R.D.)
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research and Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Bundoora, Australia (J.M.T., B.M.H., M.J., P.S., N.S., H.D., T.V.A., C.G.S., A.V., G.R.D.)
- Baker Heart and Diabetes Institute, Prahran, Australia (C.G.S., G.R.D.)
| |
Collapse
|
22
|
Exploring Molecular Mechanisms Involved in the Development of the Depression-Like Phenotype in Interleukin-18-Deficient Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9975865. [PMID: 34708129 PMCID: PMC8545524 DOI: 10.1155/2021/9975865] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 09/18/2021] [Indexed: 11/18/2022]
Abstract
Interleukin-18 (IL-18) is an inflammatory cytokine that has been linked to energy homeostasis and psychiatric symptoms such as depression and cognitive impairment. We previously revealed that deficiency in IL-18 led to hippocampal abnormalities and resulted in depression-like symptoms. However, the impact of IL-18 deficiency on other brain regions remains to be clarified. In this study, we first sought to confirm that IL-18 expression in neural cells can be found in human brain tissue. Subsequently, we examined the expression of genes in the prefrontal cortex of Il18−/− mice and compared it with gene expression in mice subjected to a chronic mild stress model of depression. Extracted genes were further analyzed using Ingenuity® Pathway Analysis, in which 18 genes common to both the chronic mild stressed model and Il18−/− mice were identified. Of those, 16 were significantly differentially expressed between Il18+/+ and Il18−/− mice. We additionally measured protein expression of α-2-HS-glycoprotein (AHSG) and transthyretin (TTR) in serum and the brain. In the prefrontal cortex of Il18−/− mice, TTR but not AHSG was significantly decreased. Conversely, in the serum of Il18−/− mice, AHSG was significantly increased but not TTR. Therefore, our results suggest that in IL-18-deficit conditions, TTR in the brain is one of the mediators causally related to depression, and AHSG in peripheral organs is one of the regulators inducing energy imbalance. Moreover, this study suggests a possible “signpost” to clarify the molecular mechanisms commonly underlying the immune system, energy metabolism, neural function, and depressive disorders.
Collapse
|
23
|
Wittmann N, Behrendt AK, Mishra N, Bossaller L, Meyer-Bahlburg A. Instructions for Flow Cytometric Detection of ASC Specks as a Readout of Inflammasome Activation in Human Blood. Cells 2021; 10:2880. [PMID: 34831104 PMCID: PMC8616555 DOI: 10.3390/cells10112880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/14/2021] [Accepted: 10/21/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammasome activation is linked to the aggregation of the adaptor protein ASC into a multiprotein complex, known as the ASC speck. Redistribution of cytosolic ASC to this complex has been widely used as a readout for inflammasome activation and precedes the downstream proteolytic release of the proinflammatory cytokines, IL-1β and IL-18. Although inflammasomes are important for many diseases such as periodic fever syndromes, COVID-19, gout, sepsis, atherosclerosis and Alzheimer's disease, only a little knowledge exists on the precise and cell type specific occurrence of inflammasome activation in patient samples ex vivo. In this report, we provide detailed information about the optimal conditions to reliably identify inflammasome activated monocytes by ASC speck formation using a modified flow cytometric method introduced by Sester et al. in 2015. Since no protocol for optimal sample processing exists, we tested human blood samples for various conditions including anticoagulant, time and temperature, the effect of one freeze-thaw cycle for PBMC storage, and the fast generation of a positive control. We believe that this flow cytometric protocol will help researchers to perform high quality translational research in multicenter studies, and therefore provide a basis for investigating the role of the inflammasome in the pathogenesis of various diseases.
Collapse
Affiliation(s)
- Nico Wittmann
- Pediatric Rheumatology, Department Pediatric and Adolescent Medicine, University Medicine, University of Greifswald, 17489 Greifswald, Germany; (N.W.); (A.-K.B.)
| | - Ann-Kathrin Behrendt
- Pediatric Rheumatology, Department Pediatric and Adolescent Medicine, University Medicine, University of Greifswald, 17489 Greifswald, Germany; (N.W.); (A.-K.B.)
| | - Neha Mishra
- Section of Rheumatology, Department of Medicine A, University Medicine, University of Greifswald, 17489 Greifswald, Germany;
| | - Lukas Bossaller
- Section of Rheumatology, Department of Medicine A, University Medicine, University of Greifswald, 17489 Greifswald, Germany;
| | - Almut Meyer-Bahlburg
- Pediatric Rheumatology, Department Pediatric and Adolescent Medicine, University Medicine, University of Greifswald, 17489 Greifswald, Germany; (N.W.); (A.-K.B.)
| |
Collapse
|
24
|
Cui ZW, Li DQ, Zhao F, Tan AP, Deng YT, Lai YT, Huang ZB, Jiang L. Molecular characterization and functional analysis of IL-18 in snakehead (Channa argus) during Aeromonas schubertii and Nocardia seriolae infections. Mol Immunol 2021; 137:212-220. [PMID: 34280771 DOI: 10.1016/j.molimm.2021.07.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 11/16/2022]
Abstract
As a proinflammatory cytokine of the interleukin-1 (IL-1) family, IL-18 plays important roles in host protection against bacterial, viral, and fungal infection. We cloned the open reading frame of snakehead (Channa argus) IL-18 (shIL-18) and found that it contained 609 base pairs and encoded 202 amino acid residues. The shIL-18 included a conserved IL-1-like family signature and two potential IL-1β-converting enzyme cutting sites; one was conserved in all analyzed IL-18s, but the other was unique to shIL-18. Unlike other IL-18s, shIL-18 also contained a predicted signal peptide. In this study, shIL-18 was constitutively expressed in all tested tissues, and its expression was induced by Aeromonas schubertii and Nocardia seriolae in the head kidney and spleen in vivo and by lipoteichoic acid, lipopolysaccharides, and polyinosinic-polycytidylic acid in head kidney leukocytes in vitro. Moreover, recombinant shIL-18 upregulated the expression of interferon-γ, IL-1β, and tumor necrosis factor-α1 and -α2 and promoted the proliferation of leukocytes. Taken together, these results showed that IL-18 played crucial roles in host defense against bacterial infection in fish, as it does in mammals.
Collapse
Affiliation(s)
- Zheng-Wei Cui
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Dong-Qi Li
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Fei Zhao
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China.
| | - Ai-Ping Tan
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Yu-Ting Deng
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Ying-Tiao Lai
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Zhi-Bin Huang
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| | - Lan Jiang
- Key Laboratory of Fishery Drug Development of Ministry of Agriculture and Rural Affairs, Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, 510380, China
| |
Collapse
|
25
|
Zhang WJ, Chen SJ, Zhou SC, Wu SZ, Wang H. Inflammasomes and Fibrosis. Front Immunol 2021; 12:643149. [PMID: 34177893 PMCID: PMC8226128 DOI: 10.3389/fimmu.2021.643149] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
Fibrosis is the final common pathway of inflammatory diseases in various organs. The inflammasomes play an important role in the progression of fibrosis as innate immune receptors. There are four main members of the inflammasomes, such as NOD-like receptor protein 1 (NLRP1), NOD-like receptor protein 3 (NLRP3), NOD-like receptor C4 (NLRC4), and absent in melanoma 2 (AIM2), among which NLRP3 inflammasome is the most studied. NLRP3 inflammasome is typically composed of NLRP3, ASC and pro-caspase-1. The activation of inflammasome involves both "classical" and "non-classical" pathways and the former pathway is better understood. The "classical" activation pathway of inflammasome is that the backbone protein is activated by endogenous/exogenous stimulation, leading to inflammasome assembly. After the formation of "classic" inflammasome, pro-caspase-1 could self-activate. Caspase-1 cleaves cytokine precursors into mature cytokines, which are secreted extracellularly. At present, the "non-classical" activation pathway of inflammasome has not formed a unified model for activation process. This article reviews the role of NLRP1, NLRP3, NLRC4, AIM2 inflammasome, Caspase-1, IL-1β, IL-18 and IL-33 in the fibrogenesis.
Collapse
Affiliation(s)
- Wen-Juan Zhang
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Shu-Juan Chen
- Department of Immunology, School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Shun-Chang Zhou
- Department of Experimental Animals, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Su-Zhen Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
26
|
Lebratti T, Lim YS, Cofie A, Andhey P, Jiang X, Scott J, Fabbrizi MR, Ozantürk AN, Pham C, Clemens R, Artyomov M, Dinauer M, Shin H. A sustained type I IFN-neutrophil-IL-18 axis drives pathology during mucosal viral infection. eLife 2021; 10:e65762. [PMID: 34047696 PMCID: PMC8163503 DOI: 10.7554/elife.65762] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/21/2021] [Indexed: 12/14/2022] Open
Abstract
Neutrophil responses against pathogens must be balanced between protection and immunopathology. Factors that determine these outcomes are not well-understood. In a mouse model of genital herpes simplex virus-2 (HSV-2) infection, which results in severe genital inflammation, antibody-mediated neutrophil depletion reduced disease. Comparative single-cell RNA-sequencing analysis of vaginal cells against a model of genital HSV-1 infection, which results in mild inflammation, demonstrated sustained expression of interferon-stimulated genes (ISGs) only after HSV-2 infection primarily within the neutrophil population. Both therapeutic blockade of IFNα/β receptor 1 (IFNAR1) and genetic deletion of IFNAR1 in neutrophils concomitantly decreased HSV-2 genital disease severity and vaginal IL-18 levels. Therapeutic neutralization of IL-18 also diminished genital inflammation, indicating an important role for this cytokine in promoting neutrophil-dependent immunopathology. Our study reveals that sustained type I interferon (IFN) signaling is a driver of pathogenic neutrophil responses and identifies IL-18 as a novel component of disease during genital HSV-2 infection.
Collapse
MESH Headings
- Animals
- Antibodies/pharmacology
- Chlorocebus aethiops
- Disease Models, Animal
- Female
- Herpes Genitalis/immunology
- Herpes Genitalis/metabolism
- Herpes Genitalis/prevention & control
- Herpes Genitalis/virology
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/pathogenicity
- Herpesvirus 2, Human/immunology
- Herpesvirus 2, Human/pathogenicity
- Host-Pathogen Interactions
- Immunity, Mucosal/drug effects
- Interferon Type I/metabolism
- Interleukin-18/metabolism
- Mice, Inbred C57BL
- Mice, Transgenic
- Mucous Membrane/drug effects
- Mucous Membrane/innervation
- Mucous Membrane/metabolism
- Mucous Membrane/virology
- Neutrophil Activation/drug effects
- Neutrophils/drug effects
- Neutrophils/immunology
- Neutrophils/metabolism
- Neutrophils/virology
- Receptor, Interferon alpha-beta/antagonists & inhibitors
- Receptor, Interferon alpha-beta/metabolism
- Signal Transduction
- Vagina/drug effects
- Vagina/immunology
- Vagina/metabolism
- Vagina/virology
- Vero Cells
- Mice
Collapse
Affiliation(s)
- Tania Lebratti
- Department of Medicine/Division of Infectious Diseases, Washington University School of MedicineSt LouisUnited States
| | - Ying Shiang Lim
- Department of Medicine/Division of Infectious Diseases, Washington University School of MedicineSt LouisUnited States
| | - Adjoa Cofie
- Department of Medicine/Division of Infectious Diseases, Washington University School of MedicineSt LouisUnited States
| | - Prabhakar Andhey
- Department of Pathology and Immunology, Washington University School of MedicineSt LouisUnited States
| | - Xiaoping Jiang
- Department of Medicine/Division of Infectious Diseases, Washington University School of MedicineSt LouisUnited States
| | - Jason Scott
- Department of Medicine/Division of Infectious Diseases, Washington University School of MedicineSt LouisUnited States
| | - Maria Rita Fabbrizi
- Department of Medicine/Division of Infectious Diseases, Washington University School of MedicineSt LouisUnited States
| | - Ayşe Naz Ozantürk
- Department of Medicine/Division of Infectious Diseases, Washington University School of MedicineSt LouisUnited States
| | - Christine Pham
- Department of Medicine/Division of Rheumatology, Washington University School of MedicineSt LouisUnited States
| | - Regina Clemens
- Department of Pediatrics/Division of Critical Care Medicine, Washington University School of MedicineSt LouisUnited States
| | - Maxim Artyomov
- Department of Pathology and Immunology, Washington University School of MedicineSt LouisUnited States
| | - Mary Dinauer
- Department of Pediatrics/Hematology and Oncology, Washington University School of MedicineSt LouisUnited States
| | - Haina Shin
- Department of Medicine/Division of Infectious Diseases, Washington University School of MedicineSt LouisUnited States
| |
Collapse
|
27
|
Ren H, Yang H, Yang X, Zhang G, Rong X, Huang J, Zhang L, Fu Y, Allain JP, Li C, Wang W. Brucella Outer Membrane Lipoproteins 19 and 16 Differentially Induce IL-18 Response or Pyroptosis in Human Monocytic Cells. J Infect Dis 2021; 224:2148-2159. [PMID: 34013337 DOI: 10.1093/infdis/jiab272] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/14/2021] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Brucella species (B. spp.) are Gram-negative intracellular bacteria, causing severe inflammatory diseases in animals and humans. Two major lipoproteins (L19) and (L16) of Brucella outer membrane proteins (OMPs) were extensively explored in associating with inflammatory response of human monocytes (THP-1). METHODS Activated THP-1 cells induced with recombinant L19 and L16 were analyzed in comparison with unlipidated forms (U19 and U16) and lipopolysaccharide (LPS) of B. melitensis, respectively. RESULTS Secretion of inflammatory factors TNF-α, IL-6 and IL-1β was significantly increased from L19, L16 or both stimulated THP-1 cells. High secretion of IL-18 was detected only from L19-induced cells. Signaling of those cytokine responses was identified mainly through P38-MAPK pathway, and signaling of L19-induced IL-1β response was partly occurred via NF-κB. Exploration for different forms of IL-18 found that L19-induced production of active IL-18 (18 kD) was through up-regulating NLRP3 and activating caspase-1, while L16-induced production of inactive IL-18 fragments (15 kD and 16 kD) occurred through activating caspase-8/3. Additionally, L19 up-regulated phosphorylation of XIAP for inhibiting caspase-3 activity to cleave IL-18, while L16 activated caspase-3 for producing GSDME-N and leading to pyroptosis of THP-1 cells. CONCLUSION Brucella L19 and L16 differentially induce IL-18 response or pyroptosis in THP-1 cells, respectively.
Collapse
Affiliation(s)
- Hui Ren
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Heng Yang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China.,Department of blood Transfusion, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xin Yang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Guoxia Zhang
- Department of Infectious Disease, Hei Longjiang General Hospital of Agriculture reclamation Bureau, Harbin 150088, China
| | - Xia Rong
- Guangzhou Blood Center, Guangzhou 510095, China
| | - Jiaheng Huang
- Department of Surgery, First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ling Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Yongshui Fu
- Guangzhou Blood Center, Guangzhou 510095, China
| | - Jean-Pierre Allain
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China.,Emeritus professor of Transfusion Medicine, University of Cambridge, Cambridge CB2 2PT, Cambridge, UK
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Wenjing Wang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
28
|
Kurihara C, Lecuona E, Wu Q, Yang W, Núñez-Santana FL, Akbarpour M, Liu X, Ren Z, Li W, Querrey M, Ravi S, Anderson ML, Cerier E, Sun H, Kelly ME, Abdala-Valencia H, Shilatifard A, Mohanakumar T, Budinger GRS, Kreisel D, Bharat A. Crosstalk between nonclassical monocytes and alveolar macrophages mediates transplant ischemia-reperfusion injury through classical monocyte recruitment. JCI Insight 2021; 6:147282. [PMID: 33621212 PMCID: PMC8026186 DOI: 10.1172/jci.insight.147282] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/17/2021] [Indexed: 12/24/2022] Open
Abstract
Primary graft dysfunction (PGD) is the predominant cause of early graft loss following lung transplantation. We recently demonstrated that donor pulmonary intravascular nonclassical monocytes (NCM) initiate neutrophil recruitment. Simultaneously, host-origin classical monocytes (CM) permeabilize the vascular endothelium to allow neutrophil extravasation necessary for PGD. Here, we show that a CCL2-CCR2 axis is necessary for CM recruitment. Surprisingly, although intravital imaging and multichannel flow cytometry revealed that depletion of donor NCM abrogated CM recruitment, single cell RNA sequencing identified donor alveolar macrophages (AM) as predominant CCL2 secretors. Unbiased transcriptomic analysis of murine tissues combined with murine KOs and chimeras indicated that IL-1β production by donor NCM was responsible for the early activation of AM and CCL2 release. IL-1β production by NCM was NLRP3 inflammasome dependent and inhibited by treatment with a clinically approved sulphonylurea. Production of CCL2 in the donor AM occurred through IL-1R-dependent activation of the PKC and NF-κB pathway. Accordingly, we show that IL-1β-dependent paracrine interaction between donor NCM and AM leads to recruitment of recipient CM necessary for PGD. Since depletion of donor NCM, IL-1β, or IL-1R antagonism and inflammasome inhibition abrogated recruitment of CM and PGD and are feasible using FDA-approved compounds, our findings may have potential for clinical translation.
Collapse
Affiliation(s)
| | | | - Qiang Wu
- Division of Thoracic Surgery and
| | | | | | | | | | - Ziyou Ren
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Wenjun Li
- Department of Surgery, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | | | | | | | | | | | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ali Shilatifard
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - G R Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Daniel Kreisel
- Department of Surgery, Washington University in St. Louis, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ankit Bharat
- Division of Thoracic Surgery and.,Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
29
|
Betancur D, Muñoz Grez C, Oñate A. Comparative Analysis of Cytokine Expression in Oral Keratinocytes and THP-1 Macrophages in Response to the Most Prevalent Serotypes of Aggregatibacter actinomycetemcomitans. Microorganisms 2021; 9:622. [PMID: 33802988 PMCID: PMC8002688 DOI: 10.3390/microorganisms9030622] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Periodontitis is a chronic inflammatory disease associated with a dysbiotic biofilm. Many pathogens have been related with its progression and severity, one of which is Aggregatibacter actinomycetemcomitans, a Gram-negative bacteria with seven serotypes (a-g) according with the structure of its LPS, with serotype b defined as the most virulent compared with the other serotypes. The aim of this study was to evaluate the response of oral keratinocytes and macrophages to A. actinomycetemcomitans. METHODS Oral keratinocytes (OKF6/TERT2) and macrophages (THP-1) were infected with A. actinomycetemcomitans serotypes a, b and c. The expression of IL-1β, IL-6, IL-8, IL-18, TNF-α, MMP-9, RANKL, TLR-2, TLR-4, TLR-6, thymic stromal lymphopoietin (TSLP), and ICAM-1 was evaluated by qPCR at 2 and 24 h after infection. RESULTS An increase in the expression of these molecules was induced by all serotypes at both times of infection, with macrophages showing higher levels of expression at 24 h compared to epithelial cells in which the highest levels were observed in the first hours after infection. CONCLUSIONS Keratinocytes and macrophages contribute to the inflammation in periodontitis from the early stages of infection, producing the first waves of cytokines, acting as the first signal for professional immune cell recruitment and modulation of more specific immune responses.
Collapse
Affiliation(s)
| | | | - Angel Oñate
- Laboratory of Molecular Immunology, Department of Microbiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción 4030000, Chile; (D.B.); (C.M.G.)
| |
Collapse
|
30
|
Griffiths JS, Camilli G, Kotowicz NK, Ho J, Richardson JP, Naglik JR. Role for IL-1 Family Cytokines in Fungal Infections. Front Microbiol 2021; 12:633047. [PMID: 33643264 PMCID: PMC7902786 DOI: 10.3389/fmicb.2021.633047] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Fungal pathogens kill approximately 1.5 million individuals per year and represent a severe disease burden worldwide. It is estimated over 150 million people have serious fungal disease such as recurrent mucosal infections or life-threatening systemic infections. Disease can ensue from commensal fungi or new infection and involves different fungal morphologies and the expression of virulence factors. Therefore, anti-fungal immunity is complex and requires coordination between multiple facets of the immune system. IL-1 family cytokines are associated with acute and chronic inflammation and are essential for the innate response to infection. Recent research indicates IL-1 cytokines play a key role mediating immunity against different fungal infections. During mucosal disease, IL-1R and IL-36R are required for neutrophil recruitment and protective Th17 responses, but function through different mechanisms. During systemic disease, IL-18 drives protective Th1 responses, while IL-33 promotes Th2 and suppresses Th1 immunity. The IL-1 family represents an attractive anti-fungal immunotherapy target. There is a need for novel anti-fungal therapeutics, as current therapies are ineffective, toxic and encounter resistance, and no anti-fungal vaccine exists. Furthering our understanding of the IL-1 family cytokines and their complex role during fungal infection may aid the development of novel therapies. As such, this review will discuss the role for IL-1 family cytokines in fungal infections.
Collapse
Affiliation(s)
- James S Griffiths
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Giorgio Camilli
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Natalia K Kotowicz
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jemima Ho
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Jonathan P Richardson
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, United Kingdom
| |
Collapse
|
31
|
Leon G, Hussey S, Walsh PT. The Diverse Roles of the IL-36 Family in Gastrointestinal Inflammation and Resolution. Inflamm Bowel Dis 2021; 27:440-450. [PMID: 32860042 DOI: 10.1093/ibd/izaa232] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Indexed: 12/16/2022]
Abstract
The interleukin (IL)-36 family is a member of the IL-1 superfamily of cytokines and, in common with other IL-1 family members, has been shown to exhibit pleiotropic effects in homeostasis and inflammation. Although the important role these cytokines play in the skin has been widely reported, recent evidence suggests that IL-36 family members are expressed and can also exert significant influence at the intestinal mucosa. In this review, we summarize current knowledge surrounding the role of the IL-36 in the intestines. In particular, we examine its likely dichotomous role as a mediator of both inflammation and resolution, highlighting its overlapping roles in innate and adaptive inflammation at the mucosa and its contribution to pathophysiology of inflammatory bowel disease. We also summarize the complexities of targeting this cytokine family in a clinical setting.
Collapse
Affiliation(s)
- Gemma Leon
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - Seamus Hussey
- National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - Patrick T Walsh
- Department of Clinical Medicine, School of Medicine, Trinity College Dublin, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| |
Collapse
|
32
|
Tyrkalska SD, Candel S, Mulero V. The neutrophil inflammasome. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 115:103874. [PMID: 32987011 DOI: 10.1016/j.dci.2020.103874] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 06/11/2023]
Abstract
Since inflammasomes were discovered in the early 21st century, knowledge about their biology has multiplied exponentially. These cytosolic multiprotein complexes alert the immune system about the presence of infection or tissue damage, and regulate the subsequent inflammatory responses. As inflammasome dysregulation is increasingly associated with numerous autoinflammatory disorders, there is an urgent need for further research into the inflammasome's involvement in the pathogenesis of such diseases in order to identify novel therapeutic targets and treatments. The zebrafish has become a widely used animal model to study human diseases in recent years, and has already provided relevant findings in the field of inflammasome biology including the identification of new components and pathways. We provide a detailed analysis of current knowledge on neutrophil inflammasome biology and compare its features with those of the better known macrophage inflammasome, focusing on its contribution to innate immunity and its relevance for human health. Importantly, a large body of evidence points to a link between neutrophil inflammasome dysfunction and many neutrophil-mediated human diseases, but the real contribution of the neutrophil inflammasome to the pathogenesis of these disorders is largely unknown. Although neutrophils have remained in the shadow of macrophages and monocytes in the field of inflammasome research since the discovery of these multiprotein platforms, recent studies strongly suggest that the importance of the neutrophil inflammasome has been underestimated.
Collapse
Affiliation(s)
- Sylwia D Tyrkalska
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain.
| | - Sergio Candel
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain.
| | - Victoriano Mulero
- Departamento de Biología Celular e Histología, Facultad de Biología, Universidad de Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Spain.
| |
Collapse
|
33
|
Gigon L, Yousefi S, Karaulov A, Simon HU. Mechanisms of toxicity mediated by neutrophil and eosinophil granule proteins. Allergol Int 2021; 70:30-38. [PMID: 33277190 DOI: 10.1016/j.alit.2020.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Neutrophils and eosinophils are granulocytes which are characterized by the presence of granules in the cytoplasm. Granules provide a safe storage site for granule proteins that play important roles in the immune function of granulocytes. Upon granulocytes activation, diverse proteins are released from the granules into the extracellular space and contribute to the fight against infections. In this article, we describe granule proteins of both neutrophils and eosinophils able to kill pathogens and review their anticipated mechanism of antimicrobial toxicity. It should be noted that an excess of granules protein release can lead to tissue damage of the host resulting in chronic inflammation and organ dysfunction.
Collapse
|
34
|
Rodrigues TS, de Sá KSG, Ishimoto AY, Becerra A, Oliveira S, Almeida L, Gonçalves AV, Perucello DB, Andrade WA, Castro R, Veras FP, Toller-Kawahisa JE, Nascimento DC, de Lima MHF, Silva CMS, Caetite DB, Martins RB, Castro IA, Pontelli MC, de Barros FC, do Amaral NB, Giannini MC, Bonjorno LP, Lopes MIF, Santana RC, Vilar FC, Auxiliadora-Martins M, Luppino-Assad R, de Almeida SCL, de Oliveira FR, Batah SS, Siyuan L, Benatti MN, Cunha TM, Alves-Filho JC, Cunha FQ, Cunha LD, Frantz FG, Kohlsdorf T, Fabro AT, Arruda E, de Oliveira RDR, Louzada-Junior P, Zamboni DS. Inflammasomes are activated in response to SARS-CoV-2 infection and are associated with COVID-19 severity in patients. J Exp Med 2020; 218:211560. [PMID: 33231615 PMCID: PMC7684031 DOI: 10.1084/jem.20201707] [Citation(s) in RCA: 536] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/19/2020] [Accepted: 11/10/2020] [Indexed: 12/26/2022] Open
Abstract
Severe cases of COVID-19 are characterized by a strong inflammatory process that may ultimately lead to organ failure and patient death. The NLRP3 inflammasome is a molecular platform that promotes inflammation via cleavage and activation of key inflammatory molecules including active caspase-1 (Casp1p20), IL-1β, and IL-18. Although participation of the inflammasome in COVID-19 has been highly speculated, the inflammasome activation and participation in the outcome of the disease are unknown. Here we demonstrate that the NLRP3 inflammasome is activated in response to SARS-CoV-2 infection and is active in COVID-19 patients. Studying moderate and severe COVID-19 patients, we found active NLRP3 inflammasome in PBMCs and tissues of postmortem patients upon autopsy. Inflammasome-derived products such as Casp1p20 and IL-18 in the sera correlated with the markers of COVID-19 severity, including IL-6 and LDH. Moreover, higher levels of IL-18 and Casp1p20 are associated with disease severity and poor clinical outcome. Our results suggest that inflammasomes participate in the pathophysiology of the disease, indicating that these platforms might be a marker of disease severity and a potential therapeutic target for COVID-19.
Collapse
Affiliation(s)
- Tamara S Rodrigues
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Keyla S G de Sá
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Adriene Y Ishimoto
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Amanda Becerra
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Samuel Oliveira
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Leticia Almeida
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.,Center of Research in Inflammatory Diseases, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Augusto V Gonçalves
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Debora B Perucello
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Warrison A Andrade
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Ricardo Castro
- Departamento de Análises Clínicas, Toxicológicas e Bromatologia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Flavio P Veras
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Juliana E Toller-Kawahisa
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Daniele C Nascimento
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Mikhael H F de Lima
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Camila M S Silva
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Diego B Caetite
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Ronaldo B Martins
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Italo A Castro
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Marjorie C Pontelli
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Fabio C de Barros
- Departamento de Biologia, Faculdade de Filosofia Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.,Departamento de Ecologia e Biologia Evolutiva, Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Universidade Federal de São Paulo, Diadema, Brazil
| | - Natália B do Amaral
- Divisão de Imunologia Clinica, Emergência, Doenças Infecciosas e Unidade de Terapia Intensiva, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Marcela C Giannini
- Divisão de Imunologia Clinica, Emergência, Doenças Infecciosas e Unidade de Terapia Intensiva, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Letícia P Bonjorno
- Divisão de Imunologia Clinica, Emergência, Doenças Infecciosas e Unidade de Terapia Intensiva, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Maria Isabel F Lopes
- Divisão de Imunologia Clinica, Emergência, Doenças Infecciosas e Unidade de Terapia Intensiva, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Rodrigo C Santana
- Divisão de Imunologia Clinica, Emergência, Doenças Infecciosas e Unidade de Terapia Intensiva, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Fernando C Vilar
- Divisão de Imunologia Clinica, Emergência, Doenças Infecciosas e Unidade de Terapia Intensiva, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Maria Auxiliadora-Martins
- Divisão de Medicina Intensiva, Departamento de Cirurgia e Anatomia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Rodrigo Luppino-Assad
- Divisão de Imunologia Clinica, Emergência, Doenças Infecciosas e Unidade de Terapia Intensiva, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Sergio C L de Almeida
- Divisão de Imunologia Clinica, Emergência, Doenças Infecciosas e Unidade de Terapia Intensiva, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Fabiola R de Oliveira
- Divisão de Imunologia Clinica, Emergência, Doenças Infecciosas e Unidade de Terapia Intensiva, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Sabrina S Batah
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Li Siyuan
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Maira N Benatti
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Thiago M Cunha
- Center of Research in Inflammatory Diseases, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.,Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - José C Alves-Filho
- Center of Research in Inflammatory Diseases, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.,Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Fernando Q Cunha
- Center of Research in Inflammatory Diseases, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.,Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Larissa D Cunha
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Fabiani G Frantz
- Departamento de Análises Clínicas, Toxicológicas e Bromatologia, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Tiana Kohlsdorf
- Departamento de Biologia, Faculdade de Filosofia Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Alexandre T Fabro
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Eurico Arruda
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Renê D R de Oliveira
- Divisão de Imunologia Clinica, Emergência, Doenças Infecciosas e Unidade de Terapia Intensiva, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Paulo Louzada-Junior
- Divisão de Imunologia Clinica, Emergência, Doenças Infecciosas e Unidade de Terapia Intensiva, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Dario S Zamboni
- Departamento de Biologia Celular e Molecular e Bioagentes Patogênicos, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil.,Center of Research in Inflammatory Diseases, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
35
|
IL-18 and related function proteins associated with tuberculosis severity and screening for active TB among patients with non-mycobacterial community-acquired pneumonia (CAP). Saudi J Biol Sci 2020; 27:3035-3045. [PMID: 33100863 PMCID: PMC7569146 DOI: 10.1016/j.sjbs.2020.09.003] [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: 03/23/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 01/04/2023] Open
Abstract
Background Differentiation of active pulmonary tuberculosis (TB) from non-mycobacterial community-acquired pneumonia (CAP) still remains a diagnostic challenge. Objective The study aimed to quantify the IL-18, IFN-γ, IL-18BP, IL-37, and IP-10 levels in serum and Mycobacterium tuberculosis (M.tb) antigens-stimulated blood cultures from TB or CAP patients and explore if the proteins can be a useful basis for discriminating these diseases. Methods In total, 124 Polish adults, including mild/moderate (M/MTB) or advanced (ATB) TB patients, and CAP patients, were enrolled in the study. The concentrations of IL-18, IL-18BP, IFN-γ, IL-37, and IP-10 in sera and M.tb-stimulated cultures were measured by ELISA. Results The most specific and sensitive serum proteins discriminating TB from CAP were IP-10 and IL-18BP; however, IP-10 had the highest AUC in the ROC curve for the diagnosis. Serum IP-10 and IL-18BP levels increased significantly in M/MTB or ATB groups. The IL-18BP elevation in ATB group was accompanied by an increase in IL-18. No single protein measured in M.tb-stimulated cultures differed TB from CAP patients. Conclusions The combined analysis of serum IL-18BP and IP-10 might be considered as an auxiliary tool in the differentiation of TB from CAP.
Collapse
|
36
|
Wang LZ, Xu YP, Zhou YL, Liu ZP, Li B, Gu WB, Zhao XF, Dong WR, Shu MA. The first evidence of four transcripts from two Interleukin 18 genes in animal and their involvement in immune responses in the largest amphibian Andrias davidianus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 106:103598. [PMID: 31881236 DOI: 10.1016/j.dci.2019.103598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 12/22/2019] [Accepted: 12/22/2019] [Indexed: 06/10/2023]
Abstract
Interleukin 18 (IL-18), a member of IL-1 cytokine superfamily, is an important proinflammatory cytokine with multiple functions in both innate immunity and acquired immunity. However, the characteristics and functional roles of IL-18 remain largely unknown in amphibians, which were classed as major group of vertebrates. In the present study, two IL-18 genes (AdIL-18A and AdIL-18B) and four transcripts (AdIL-18A1, AdIL-18A2, AdIL-18B1 and AdIL-18B2) were firstly identified and characterized from Chinese giant salamander (Andrias davidianus). To the best of our knowledge, this is the first report on the presence of more than one gene copy or two transcripts of IL-18 in one species. The complete open reading frames of AdIL-18A1, AdIL-18A2, AdIL-18B1 and AdIL-18B2 were 588 bp, 603 bp, 591 bp and 606 bp, respectively. The putative AdIL-18 proteins possessed the typical IL-1 domains and phylogenetic analysis indicated that AdIL-18s grouped together with other vertebrate IL-18 proteins. The expression profiles of AdIL-18s were investigated under the challenges of Aeromonas hydrophila, Staphylococcus ureae and Poly (I:C) respectively, and the results suggested that AdIL-18s were involved in the immune responses against both bacterial and viral infections. Moreover, the expression levels of two NF-κBs (P100 and P105) and four proinflammatory cytokines (IL-1β, IL-6, TNF-α and IFN-γ) were inhibited in AdIL-18A1/A2-silenced cells when treated with bacteria and viral RNA analog. Additionally, the transcription levels of these immune-related cytokine genes were markedly induced when the lymphocytes were treated with recombinant AdIL-18A1 or AdIL-18A2 proteins, implying the involvement of AdIL-18s in triggering NF-κB signaling and proinflammatory responses. These results might provide new insights into the origin or evolution of IL-18 in amphibians and even in vertebrates.
Collapse
Affiliation(s)
- Lan-Zhi Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ya-Ping Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yi-Lian Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ze-Peng Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Li
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wen-Bin Gu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiao-Feng Zhao
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Wei-Ren Dong
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Miao-An Shu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
37
|
|
38
|
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: 1.0] [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
|
39
|
Rex DAB, Agarwal N, Prasad TSK, Kandasamy RK, Subbannayya Y, Pinto SM. A comprehensive pathway map of IL-18-mediated signalling. J Cell Commun Signal 2019; 14:257-266. [PMID: 31863285 DOI: 10.1007/s12079-019-00544-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 12/06/2019] [Indexed: 02/07/2023] Open
Abstract
Interleukin-18 (IL-18) is a member of the IL-1 family of cytokines and was initially described as an IFN-γ-inducing factor derived from anti-CD3-stimulated T-helper (Th)1 cells. IL-18 plays a significant role in the activation of hematopoietic cell types mediating both Th1 and Th2 responses and is the primary inducer of interferon-γ in these cells. The biological activity of IL-18 is mediated through its binding to the IL-18 receptor complex and activation of nuclear factor-κB (NF-κB), culminating in the production and release of several cytokines, chemokines, and cellular adhesion molecules. In certain cell types, IL-18 also activates mitogen-activated protein kinases (MAPKs) and phosphoinositide 3-kinase/ AKT serine/threonine kinase (PI3K/AKT) signaling modules leading to the production and release of proinflammatory cytokines. IL-18-mediated signaling acts as one of the vital components of the immunomodulatory cytokine networks involved in host defense, inflammation, and tissue regeneration. Albeit its biomedical importance, a comprehensive resource of IL-18 mediated signaling pathway is currently lacking. In this study, we report on the development of an integrated pathway map of IL-18/IL-18R signaling. The pathway map was developed through literature mining from published literature based on manual curation guidelines adapted from NetPath and includes information on 16 protein-protein interaction events, 38 enzyme-catalysis events, 12 protein translocation events, 26 activations/inhibition events, transcriptional regulators, 230 gene regulation events and 84 induced protein expression events. The IL-18 signaling pathway can be freely accessed through the WikiPathways database (https://www.wikipathways.org/index.php/Pathway:WP4754).
Collapse
Affiliation(s)
- D A B Rex
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Nupur Agarwal
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Richard K Kandasamy
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, N-7491, Trondheim, Norway
| | - Yashwanth Subbannayya
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India. .,Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, N-7491, Trondheim, Norway.
| | - Sneha M Pinto
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India. .,Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, N-7491, Trondheim, Norway.
| |
Collapse
|
40
|
Li Z, Yu X, Werner J, Bazhin AV, D'Haese JG. The role of interleukin-18 in pancreatitis and pancreatic cancer. Cytokine Growth Factor Rev 2019; 50:1-12. [PMID: 31753718 DOI: 10.1016/j.cytogfr.2019.11.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023]
Abstract
Originally described as an interferon (IFN)-γ-inducing factor, interleukin (IL)-18 has been reported to be involved in Th1 and Th2 immune responses, as well as in activation of NK cells and macrophages. There is convincing evidence that IL-18 plays an important role in various pathologies (i.e. inflammatory diseases, cancer, chronic obstructive pulmonary disease, Crohn's disease and others). Recently, IL-18 has also been shown to execute specific effects in pancreatic diseases, including acute and chronic pancreatitis, as well as pancreatic cancer. The aim of this study was to give a profound review of recent data on the role of IL-18 and its potential as a therapeutic target in pancreatic diseases. The existing data on this topic are in part controversial and will be discussed in detail. Future studies should aim to confirm and clarify the role of IL-18 in pancreatic diseases and unravel their molecular mechanisms.
Collapse
Affiliation(s)
- Zhiqiang Li
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; Department of Hepatopancreatobiliary Surgery, The third Xiangya hospital, Central south university, Changsha 410013, Hunan, China
| | - Xiao Yu
- Department of Hepatopancreatobiliary Surgery, The third Xiangya hospital, Central south university, Changsha 410013, Hunan, China
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany
| | - Alexandr V Bazhin
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany; German Cancer Consortium (DKTK), Partner Site Munich, 81377 Munich, Germany.
| | - Jan G D'Haese
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| |
Collapse
|
41
|
Mühl H, Bachmann M. IL-18/IL-18BP and IL-22/IL-22BP: Two interrelated couples with therapeutic potential. Cell Signal 2019; 63:109388. [PMID: 31401146 DOI: 10.1016/j.cellsig.2019.109388] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023]
Abstract
Interleukin (IL)-18 and IL-22 are key components of cytokine networks that play a decisive role in (pathological) inflammation, host defense, and tissue regeneration. Tight regulation of cytokine-driven signaling, inflammation, and immunoactivation is supposed to enable nullification of a given deleterious trigger without mediating overwhelming collateral tissue damage or even activating a cancerous face of regeneration. In fact, feedback regulation by specific cytokine opponents is regarded as a major means by which the immune system is kept in balance. Herein, we shine a light on the interplay between IL-18 and IL-22 and their opponents IL-18 binding protein (IL-18BP) and IL-22BP in order to provide integrated information on their biology, pathophysiological significance, and prospect as targets and/or instruments of therapeutic intervention.
Collapse
Affiliation(s)
- Heiko Mühl
- pharmazentrum frankfurt/ZAFES, University Hospital Goethe University Frankfurt am Main, Theodor-Stern- Kai 7, 60590 Frankfurt am Main, Germany.
| | - Malte Bachmann
- pharmazentrum frankfurt/ZAFES, University Hospital Goethe University Frankfurt am Main, Theodor-Stern- Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
42
|
Abstract
Inflammation is an important driver of atherosclerosis, the underlying pathology of cardiovascular diseases. Therefore, therapeutic targeting of inflammatory pathways is suggested to improve cardiovascular outcomes in patients with cardiovascular diseases. This concept was recently proven by CANTOS (Canakinumab Anti-Inflammatory Thrombosis Outcomes Study), which demonstrated the therapeutic potential of the monoclonal IL (interleukin)-1β-neutralizing antibody canakinumab. IL-1β and other IL-1 family cytokines are important vascular and systemic inflammatory mediators, which contribute to atherogenesis. The NLRP3 (NOD [nucleotide oligomerization domain]-, LRR [leucine-rich repeat]-, and PYD [pyrin domain]-containing protein 3) inflammasome, an innate immune signaling complex, is the key mediator of IL-1 family cytokine production in atherosclerosis. NLRP3 is activated by various endogenous danger signals abundantly present in atherosclerotic lesions, such as oxidized low-density lipoprotein and cholesterol crystals. Consequently, NLRP3 inflammasome activation contributes to the vascular inflammatory response driving atherosclerosis development and progression. Here, we review the mechanisms of NLRP3 inflammasome activation and proinflammatory IL-1 family cytokine production in the context of atherosclerosis and discuss treatment possibilities in light of the positive outcomes of the CANTOS trial.
Collapse
Affiliation(s)
- Alena Grebe
- From the Institute of Innate Immunity, University Hospital Bonn, Germany (A.G., F.H., E.L.)
| | - Florian Hoss
- From the Institute of Innate Immunity, University Hospital Bonn, Germany (A.G., F.H., E.L.)
| | - Eicke Latz
- From the Institute of Innate Immunity, University Hospital Bonn, Germany (A.G., F.H., E.L.) .,Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester (E.L.).,German Center of Neurodegenerative Diseases (DZNE), Bonn, Germany (E.L.).,Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway (E.L.)
| |
Collapse
|
43
|
Clancy DM, Sullivan GP, Moran HBT, Henry CM, Reeves EP, McElvaney NG, Lavelle EC, Martin SJ. Extracellular Neutrophil Proteases Are Efficient Regulators of IL-1, IL-33, and IL-36 Cytokine Activity but Poor Effectors of Microbial Killing. Cell Rep 2019. [PMID: 29539422 DOI: 10.1016/j.celrep.2018.02.062] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neutrophil granule proteases are thought to function as anti-microbial effectors, cooperatively hydrolyzing microorganisms within phagosomes, or upon deployment into the extracellular space. However, evidence also suggests that neutrophil proteases play an important role in the coordination and escalation of inflammatory reactions, but how this is achieved has been obscure. IL-1 family cytokines are important initiators of inflammation and are typically released via necrosis but require proteolytic processing for activation. Here, we show that proteases liberated from activated neutrophils can positively or negatively regulate the activity of six IL-1 family cytokines (IL-1α, IL-1β, IL-33, IL-36α, IL-36β, and IL-36γ) with exquisite sensitivity. In contrast, extracellular neutrophil proteases displayed very poor bactericidal activity, exhibiting 100-fold greater potency toward cytokine processing than bacterial killing. Thus, in addition to their classical role as phagocytes, neutrophils play an important immunoregulatory role through deployment of their granule proteases into the extracellular space to process multiple IL-1 family cytokines.
Collapse
Affiliation(s)
- Danielle M Clancy
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland
| | - Graeme P Sullivan
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland
| | - Hannah B T Moran
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland
| | - Conor M Henry
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland
| | - Emer P Reeves
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland
| | - Seamus J Martin
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
44
|
Hernandez-Santana YE, Giannoudaki E, Leon G, Lucitt MB, Walsh PT. Current perspectives on the interleukin-1 family as targets for inflammatory disease. Eur J Immunol 2019; 49:1306-1320. [PMID: 31250428 DOI: 10.1002/eji.201848056] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/15/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022]
Abstract
Since the first description of interleukin-1 (IL-1) and the genesis of the field of cytokine biology, the understanding of how IL-1 and related cytokines play central orchestrating roles in the inflammatory response has been an area of intense investigation. As a consequence of these endeavours, specific strategies have been developed to target the function of the IL-1 family in human disease realizing significant impacts for patients. While the most significant advances to date have been associated with inhibition of the prototypical family members IL-1α/β, approaches to target more recently identified family members such as IL-18, IL-33 and the IL-36 subfamily are now beginning to come to fruition. This review summarizes current knowledge surrounding the roles of the IL-1 family in human disease and describes the rationale and strategies which have been developed to target these cytokines to inhibit the pathogenesis of a wide range of diseases in which inflammation plays a centrally important role.
Collapse
Affiliation(s)
- Yasmina E Hernandez-Santana
- Trinity Translational Medicine Institute, Department of Clinical Medicine, School of Medicine, Trinity College, Dublin.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin
| | - Eirini Giannoudaki
- Trinity Translational Medicine Institute, Department of Clinical Medicine, School of Medicine, Trinity College, Dublin.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin
| | - Gemma Leon
- Trinity Translational Medicine Institute, Department of Clinical Medicine, School of Medicine, Trinity College, Dublin.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin
| | - Margaret B Lucitt
- Department of Pharmacology and Therapeutics, School of Medicine, Trinity College, Dublin
| | - Patrick T Walsh
- Trinity Translational Medicine Institute, Department of Clinical Medicine, School of Medicine, Trinity College, Dublin.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin
| |
Collapse
|
45
|
McEntee CP, Finlay CM, Lavelle EC. Divergent Roles for the IL-1 Family in Gastrointestinal Homeostasis and Inflammation. Front Immunol 2019; 10:1266. [PMID: 31231388 PMCID: PMC6568214 DOI: 10.3389/fimmu.2019.01266] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/17/2019] [Indexed: 12/11/2022] Open
Abstract
Inflammatory disorders of the gastro-intestinal tract are a major cause of morbidity and significant burden from a health and economic perspective in industrialized countries. While the incidence of such conditions has a strong environmental component, in particular dietary composition, epidemiological studies have identified specific hereditary mutations which result in disequilibrium between pro- and anti-inflammatory factors. The IL-1 super-family of cytokines and receptors is highly pleiotropic and plays a fundamental role in the pathogenesis of several auto-inflammatory conditions including rheumatoid arthritis, multiple sclerosis and psoriasis. However, the role of this super-family in the etiology of inflammatory bowel diseases remains incompletely resolved despite extensive research. Herein, we highlight the currently accepted paradigms as they pertain to specific IL-1 family members and focus on some recently described non-classical roles for these pathways in the gastrointestinal tract. Finally, we address some of the shortcomings and sources of variance in the field which to date have yielded several conflicting results from similar studies and discuss the potential effect of these factors on data interpretation.
Collapse
Affiliation(s)
- Craig P McEntee
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Conor M Finlay
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Faculty of Biology, Medicine and Health, Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
46
|
Kaneko N, Kurata M, Yamamoto T, Morikawa S, Masumoto J. The role of interleukin-1 in general pathology. Inflamm Regen 2019; 39:12. [PMID: 31182982 PMCID: PMC6551897 DOI: 10.1186/s41232-019-0101-5] [Citation(s) in RCA: 328] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/07/2019] [Indexed: 12/19/2022] Open
Abstract
Interleukin-1, an inflammatory cytokine, is considered to have diverse physiological functions and pathological significances and play an important role in health and disease. In this decade, interleukin-1 family members have been expanding and evidence is accumulating that highlights the importance of interleukin-1 in linking innate immunity with a broad spectrum of diseases beyond inflammatory diseases. In this review, we look back on the definition of "inflammation" in traditional general pathology and discuss new insights into interleukin-1 in view of its history and the molecular bases of diseases, as well as current progress in therapeutics.
Collapse
Affiliation(s)
- Naoe Kaneko
- Department of Pathology, Ehime University Graduate School of Medicine and Proteo-Science Center, Shitsukawa 454, Toon, Ehime 791-0295 Japan
| | - Mie Kurata
- Department of Pathology, Ehime University Graduate School of Medicine and Proteo-Science Center, Shitsukawa 454, Toon, Ehime 791-0295 Japan
| | - Toshihiro Yamamoto
- Department of Pathology, Ehime University Graduate School of Medicine and Proteo-Science Center, Shitsukawa 454, Toon, Ehime 791-0295 Japan
| | - Shinnosuke Morikawa
- Department of Pathology, Ehime University Graduate School of Medicine and Proteo-Science Center, Shitsukawa 454, Toon, Ehime 791-0295 Japan
| | - Junya Masumoto
- Department of Pathology, Ehime University Graduate School of Medicine and Proteo-Science Center, Shitsukawa 454, Toon, Ehime 791-0295 Japan
| |
Collapse
|
47
|
Interleukin-18-deficient mice develop hippocampal abnormalities related to possible depressive-like behaviors. Neuroscience 2019; 408:147-160. [PMID: 30981863 DOI: 10.1016/j.neuroscience.2019.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 12/31/2022]
Abstract
Interleukin-18 (IL-18) is an inflammatory cytokine linked to major depressive disorder (MDD). MDD is closely related to metabolic disorders, such as diabetes mellitus (DM) and obesity. Moreover, DM is associated with cognitive impairment and promotes apoptosis of hippocampal cells by activating pro-apoptotic and inhibiting anti-apoptotic factors. IL-18-deficient (Il18-/-) mice are obese and have DM. Therefore, we hypothesized a close relationship between IL-18 and death of hippocampal cells, affecting neurogenesis related to behavioral changes such as MDD. Il18-/- male mice were generated on the C57Bl/6 background and Il18+/+ mice were used as controls. Behavioral, histopathological, and molecular responses, as well as responses to intracerebral recombinant IL-18 administration, were examined. Compared with Il18+/+ mice, Il18-/- mice had impaired learning and memory and exhibited lower motivation. In the Il18-/- mice, degenerated mitochondria were detected in synaptic terminals in the molecular layer, the polymorphic layer, and in mossy fibers in the dentate gyrus, suggesting mitochondrial abnormalities. Because of the degeneration of mitochondria in the dentate gyrus, in which pro-apoptotic molecules were upregulated and anti-apoptotic factors were decreased, apoptosis inducers were not cleaved, indicating inhibition of apoptosis. In addition, neurogenesis in the dentate gyrus and the maturity of neuronal cells were decreased in the Il18-/- mice, while intracerebral administration of recombinant IL-18 promoted significant recovery of neurogenesis. Our findings suggested that IL-18 was indispensable for mitochondrial homeostasis, sustaining clearance of degenerative neural cells, and supporting neurogenesis, normal neuronal maturation and hippocampal function.
Collapse
|
48
|
Celias DP, Corvo I, Silvane L, Tort JF, Chiapello LS, Fresno M, Arranz A, Motrán CC, Cervi L. Cathepsin L3 From Fasciola hepatica Induces NLRP3 Inflammasome Alternative Activation in Murine Dendritic Cells. Front Immunol 2019; 10:552. [PMID: 30967874 PMCID: PMC6438957 DOI: 10.3389/fimmu.2019.00552] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/01/2019] [Indexed: 12/19/2022] Open
Abstract
The production of IL-1-family cytokines such as IL-1β and IL-18 is finely regulated by inflammasome activation after the recognition of pathogens associated molecular pattern (PAMPs) and danger associated molecular patterns (DAMPs). However, little is known about the helminth-derived molecules capable of activating the inflammasome. In the case of the helminth trematode Fasciola hepatica, the secretion of different cathepsin L cysteine peptidases (FhCL) is crucial for the parasite survival. Among these enzymes, cathepsin L3 (FhCL3) is expressed mainly in the juvenile or invasive stage. The ability of FhCL3 to digest collagen has demonstrated to be critical for intestinal tissue invasion during juvenile larvae migration. However, there is no information about the interaction of FhCL3 with the immune system. It has been shown here that FhCL3 induces a non-canonical inflammasome activation in dendritic cells (DCs), leading to IL-1β and IL-18 production without a previous microbial priming. Interestingly, this activation was depending on the cysteine protease activity of FhCL3 and the NLRP3 receptor, but independent of caspase activation. We also show that FhCL3 is internalized by DCs, promoting pro-IL-1β cleavage to its mature and biologically active form IL-1β, which is released to the extracellular environment. The FhCL3-induced NLRP3 inflammasome activation conditions DCs to promote a singular adaptive immune response, characterized by increased production of IFN-γ and IL-13. These data reveal an unexpected ability of FhCL3, a helminth-derived molecule, to activate the NLRP3 inflammasome, which is independent of the classical mechanism involving caspase activation.
Collapse
Affiliation(s)
- Daiana Pamela Celias
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Ileana Corvo
- Laboratorio de Investigación y Desarrollo de Moléculas Bioactivas, CENUR Litoral Norte - Sede Paysandú, Universidad de la República, Paysandú, Uruguay
| | - Leonardo Silvane
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - José Francisco Tort
- Departmento de Genética, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Laura Silvina Chiapello
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa (CSIC), Madrid, Spain
| | - Alicia Arranz
- Centro de Biología Molecular Severo Ochoa (CSIC), Madrid, Spain
| | - Claudia Cristina Motrán
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| | - Laura Cervi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Córdoba, Argentina
| |
Collapse
|
49
|
Interleukin-18 in Health and Disease. Int J Mol Sci 2019; 20:ijms20030649. [PMID: 30717382 PMCID: PMC6387150 DOI: 10.3390/ijms20030649] [Citation(s) in RCA: 322] [Impact Index Per Article: 64.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-18 was originally discovered as a factor that enhanced IFN-γ production from anti-CD3-stimulated Th1 cells, especially in the presence of IL-12. Upon stimulation with Ag plus IL-12, naïve T cells develop into IL-18 receptor (IL-18R) expressing Th1 cells, which increase IFN-γ production in response to IL-18 stimulation. Therefore, IL-12 is a commitment factor that induces the development of Th1 cells. In contrast, IL-18 is a proinflammatory cytokine that facilitates type 1 responses. However, IL-18 without IL-12 but with IL-2, stimulates NK cells, CD4+ NKT cells, and established Th1 cells, to produce IL-3, IL-9, and IL-13. Furthermore, together with IL-3, IL-18 stimulates mast cells and basophils to produce IL-4, IL-13, and chemical mediators such as histamine. Therefore, IL-18 is a cytokine that stimulates various cell types and has pleiotropic functions. IL-18 is a member of the IL-1 family of cytokines. IL-18 demonstrates a unique function by binding to a specific receptor expressed on various types of cells. In this review article, we will focus on the unique features of IL-18 in health and disease in experimental animals and humans.
Collapse
|
50
|
Nariai Y, Kamino H, Obayashi E, Kato H, Sakashita G, Sugiura T, Migita K, Koga T, Kawakami A, Sakamoto K, Kadomatsu K, Nakakido M, Tsumoto K, Urano T. Generation and characterization of antagonistic anti-human interleukin (IL)-18 monoclonal antibodies with high affinity: Two types of monoclonal antibodies against full-length IL-18 and the neoepitope of inflammatory caspase-cleaved active IL-18. Arch Biochem Biophys 2019; 663:71-82. [PMID: 30615852 DOI: 10.1016/j.abb.2019.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 12/29/2018] [Accepted: 01/03/2019] [Indexed: 01/16/2023]
Abstract
Interleukin-18 (IL-18) is a pro-inflammatory cytokine that evokes both innate and acquired immune responses. IL-18 is initially synthesized as an inactive precursor and the cleavage for processing into a mature, active molecule is mediated by pro-inflammatory caspases following the activation of inflammasomes. Two types of monoclonal antibodies were raised: anti-IL-1863-68 antibodies which recognize full-length1-193 and cleaved IL-18; and anti-IL-18 neoepitope antibodies which specifically recognize the new N-terminal 37YFGKLESK44 of IL-18 cleaved by pro-inflammatory caspase-1/4. These mAbs were suitable for Western blotting, capillary Western immunoassay (WES), immunofluorescence, immunoprecipitation, and function-blocking assays. WES analysis of these mAbs allowed visualization of the IL-18 bands and provided a molecular weight corresponding to the pro-inflammatory caspase-1/4 cleaved, active form IL-1837-193, and not to the inactive precursor IL-18, in the serum of patients with adult-onset Still's disease (6/14, 42%) and hemophagocytic activation syndrome (2/6, 33%). These monoclonal antibodies will be very useful in IL-18 and inflammasome biology and for diagnostic and therapeutic strategies for inflammatory diseases.
Collapse
Affiliation(s)
- Yuko Nariai
- Department of Biochemistry, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Hiroki Kamino
- Department of Biochemistry, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Eiji Obayashi
- Department of Biochemistry, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Hiroaki Kato
- Department of Biochemistry, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Gyosuke Sakashita
- Department of Biochemistry, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Tomoko Sugiura
- Department of Biochemistry, Shimane University School of Medicine, Izumo, 693-8501, Japan
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, 960-1247, Japan
| | - Tomohiro Koga
- Department of Rheumatology, Unit of Advanced Preventive Medical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8501, Japan
| | - Atsushi Kawakami
- Department of Rheumatology, Unit of Advanced Preventive Medical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8501, Japan
| | - Kazuma Sakamoto
- Department of Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Makoto Nakakido
- Department of Bioengineering, School of Engineering, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Kouhei Tsumoto
- Department of Bioengineering, School of Engineering, The University of Tokyo, Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan; Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Takeshi Urano
- Department of Biochemistry, Shimane University School of Medicine, Izumo, 693-8501, Japan; mAbProtein Co. Ltd, Izumo, 693-8501, Japan.
| |
Collapse
|