1
|
Zhang J, Wise SG, Zuo S, Bao S, Zhang X. The distinct roles of IL-37 and IL-38 in non-small cell lung carcinoma and their clinical implications. Front Immunol 2025; 16:1564357. [PMID: 40191189 PMCID: PMC11968353 DOI: 10.3389/fimmu.2025.1564357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Lung cancer, a significant global health challenge, is primarily classified into non-small cell lung cancer (NSCLC) and small cell lung cancer. Despite advancements in targeted therapies and immunotherapies, NSCLC outcomes remain poor, with low five-year survival rates. Given the lung's constant exposure to the environment and the presence of mucosal-associated lymphoid tissues, immunity plays a crucial role in NSCLC development. Immune checkpoint inhibitors (ICIs) targeting PD-1/PD-L1 have shown promise. However, adverse immune events limit their efficacy. This review highlights the contrasting roles of IL-37 and IL-38 in NSCLC pathogenesis. IL-37, an anti-inflammatory cytokine, suppresses tumour growth. It achieves this by modulating macrophage polarization and dendritic cell maturation. Correlations between intra-tumoral IL-37 expression and improved survival suggest a protective role in NSCLC. This may be mediated through VEGF inhibition and immune regulation. Conversely, IL-38, while anti-inflammatory in certain contexts, exhibits a pro-tumorigenic role in NSCLC. IL-38 enhances tumour progression by increasing pro-inflammatory cytokine secretion and facilitating immune evasion, potentially through NF-κB signalling. Notably, IL-38 negatively regulates IL-37, further promoting tumorigenesis. Emerging data suggest that IL-37 has therapeutic potential in inhibiting NSCLC metastasis and supporting immune modulation. In contrast, IL-38 presents a potential target for mitigating pro-inflammatory microenvironment effects. The distinct roles of these cytokines emphasize the complex immune dynamics in NSCLC. Further exploration of their molecular mechanisms and therapeutic implications is warranted. Targeting IL-37 and IL-38 may offer novel strategies for enhancing NSCLC treatment outcomes.
Collapse
Affiliation(s)
- Jiwei Zhang
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Steven G. Wise
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NS, Australia
| | - Shunqing Zuo
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Shisan Bao
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Xufeng Zhang
- Department of Thoracic Surgery, Songjiang Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
2
|
Wu Q, Zeng Y, Geng K, Guo M, Teng FY, Yan PJ, Lei Y, Long Y, Jiang ZZ, Law BYK, Xu Y. The role of IL-1 family cytokines in diabetic cardiomyopathy. Metabolism 2025; 163:156083. [PMID: 39603339 DOI: 10.1016/j.metabol.2024.156083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Diabetic cardiomyopathy (DCM) is the primary cause of heart failure in patients with diabetes and is characterised by contractile dysfunction and left ventricular hypertrophy. The complex pathological and physiological mechanisms underlying DCM have contributed to a limited number of available treatment options. A substantial body of evidence has established that DCM is a low-grade inflammatory cardiovascular disorder, with the interleukin-1 (IL-1) family of cytokines playing crucial roles in initiating inflammatory responses and shaping innate and adaptive immunity. In this review, we aim to provide an overview of the underlying mechanisms of the IL-1 family and their relevance in DCM of various aetiologies. Furthermore, we highlighted potential therapeutic targets within the IL-1 family for the management of DCM.
Collapse
Affiliation(s)
- Qi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Pathology, and Luzhou Key Laboratory of Precision Pathology Diagnosis for Serious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Kang Geng
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Plastic and burns surgery, National Key Clinical Construction Specialty, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Man Guo
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Fang-Yuan Teng
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Pi-Jun Yan
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yi Lei
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yang Long
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zong-Zhe Jiang
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China.
| | - Yong Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
3
|
He Y, Shen X, Zhai K, Nian S. Advances in understanding the role of interleukins in pulmonary fibrosis (Review). Exp Ther Med 2025; 29:25. [PMID: 39650776 PMCID: PMC11619568 DOI: 10.3892/etm.2024.12775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/03/2024] [Indexed: 12/11/2024] Open
Abstract
Pulmonary fibrosis (PF) is a progressive, irreversible disease characterized by heterogeneous interstitial lung tissue damage. It originates from persistent or repeated lung epithelial injury and leads to the activation and differentiation of fibroblasts into myofibroblasts. Interleukins (ILs) are a group of lymphokines crucial for immunomodulation that are implicated in the pathogenesis of PF. However, different types of ILs exert disparate effects on PF. In the present review, based on the effect on PF, ILs are classified into three categories: i) Promotors of PF; ii) inhibitors of PF; and iii) those that exert dual effects on PF. Several types of ILs can promote PF by provoking inflammation, initiating proliferation and transdifferentiation of epithelial cells, exacerbating lung injury, while other ILs can inhibit PF through suppressing expression of inflammatory factors, modulating the Th1/Th2 balance and autophagy. The present review summarizes the association of ILs and PF, focusing on the roles and mechanisms of ILs underlying PF.
Collapse
Affiliation(s)
- Yuqing He
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Xuebin Shen
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| | - Kefeng Zhai
- School of Biological and Food Engineering, Engineering Research Center for Development and High Value Utilization of Genuine Medicinal Materials in North Anhui Province, Suzhou University, Suzhou, Anhui 234000, P.R. China
| | - Sihui Nian
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine, Institute of Health and Medicine, Wannan Medical College, Wuhu, Anhui 241002, P.R. China
| |
Collapse
|
4
|
Liu H, Zhang M, Gong W, Tian F, Zhang L, Zhao J, Xiang B, Hui J. Interleukin-38 as a potential diagnostic biomarker for heart failure. Biomark Med 2025; 19:105-111. [PMID: 39949181 PMCID: PMC11834501 DOI: 10.1080/17520363.2025.2459595] [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: 09/24/2024] [Accepted: 01/24/2025] [Indexed: 02/18/2025] Open
Abstract
OBJECTIVE Interleukin-38 (IL-38) attenuates inflammation, myocardial injury, and ventricular remodeling after myocardial infarction, which potentially reduces the incidence of heart failure (HF). This study aimed to evaluate the diagnostic value of IL-38 for HF. METHODS IL-38 was detected via enzyme-linked immunosorbent assay in the serum samples of 238 hF patients and 30 healthy controls when enrollment. RESULTS IL-38 was elevated in HF patients compared with controls (p = 0.002). Receiver operating characteristic (ROC) analysis showed that the area under the curve (AUC) of IL-38 for predicting HF risk was 0.676 (95% confidence interval = 0.594-0.758). The Youden index of IL-38 for diagnosing HF peaked at 0.329 when the cutoff value of IL-38 was set as 6 ng/mL, with sensitivity and specificity of 0.833 and 0.496, respectively. Before (odds ratio = 1.493, p = 0.002) and after (odds ratio = 1.500, p = 0.007) adjustment of age and gender by multivariable regression analysis, IL-38 was associated with a higher HF risk. Moreover, the combination of IL-38 with age and gender possessed a good value for predicting HF risk (AUC = 0.796, 95% confidence interval = 0.706-0.885). CONCLUSION IL-38 is increased in HF patients compared with controls, and its combination with age and gender shows a good value for the diagnosis of HF.
Collapse
Affiliation(s)
- Hongman Liu
- Department of Cardiology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Mingliang Zhang
- Department of Cardiology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Wen Gong
- Department of Cardiology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Faqiang Tian
- Department of Cardiology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Lei Zhang
- Department of Cardiology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Jin Zhao
- Department of Cardiology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Bing Xiang
- Department of Cardiology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Jie Hui
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
5
|
Diaz-Barreiro A, Talabot-Ayer D, Huard A, Cereghetti G, Tonacini J, Maillasson M, Francés-Monerris A, Mortier E, Palmer G. Full-length and N-terminally truncated recombinant interleukin-38 variants are similarly inefficient in antagonizing interleukin-36 and interleukin-1 receptors. Cell Commun Signal 2025; 23:34. [PMID: 39833821 PMCID: PMC11744908 DOI: 10.1186/s12964-025-02035-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Interleukin (IL)-38 is an IL-1 family cytokine that was proposed to exert anti-inflammatory effects. However, its mechanisms of action are not well understood and the identity of the IL-38 receptor(s) remains debated. Proposed candidates include the IL-1 receptor (IL-1R1), the IL-36 receptor (IL-36R) and the orphan receptor IL-1RAPL1. Yet, in literature, IL-38 is often presented as an IL-36R antagonist. METHODS The N-terminus of the IL-38 protein produced in a human keratinocyte cell line and of endogenous epidermal IL-38 isolated from healthy human skin was characterized by mass spectrometry. The effects of various recombinant forms of IL-38 on IL-36R- and IL-1R1-mediated responses were assessed in IL-36R HEK Blue reporter cells and in a normal human keratinocyte cell line. IL-8 and IL-6 production was quantified by ELISA. Binding of recombinant IL-38 proteins to the IL-36R was assessed by surface plasmon resonance. RESULTS Analysis of its native N-terminus revealed that the IL-38 protein produced by human keratinocytes starts at cysteine 2. In cell-based assays, neither full-length amino acid 2-152 IL-38 nor two N-terminally truncated forms of the protein showed efficient antagonist activity on IL-36R- and IL-1R1-mediated responses. The recombinant IL-38 proteins bound to the IL-36R with only moderate affinity, which may provide a mechanistic explanation for inefficient IL-36R antagonism. CONCLUSIONS Our results argue against meaningful inhibitory effects of any of the recombinant IL-38 variants tested on IL-36R or IL-1R1-mediated responses. The mechanisms underlying reported anti-inflammatory effects of IL-38 are thus still unclear, but seem unlikely to be mediated by classical IL-36R or IL-1R1 antagonism.
Collapse
Affiliation(s)
- Alejandro Diaz-Barreiro
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1 rue Michel-Servet, Geneva, CH-1211, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Dominique Talabot-Ayer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1 rue Michel-Servet, Geneva, CH-1211, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Arnaud Huard
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1 rue Michel-Servet, Geneva, CH-1211, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Gea Cereghetti
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Jenna Tonacini
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1 rue Michel-Servet, Geneva, CH-1211, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Mike Maillasson
- Nantes Université, CNRS, Inserm, CRCI 2 NA, Nantes, France
- Nantes Université, Centre Hospitalo-Universitaire (CHU) Nantes, Inserm, CNRS, SFR Bonamy, UMS BioCore, Imp@ct Platform, Inserm, Nantes, France
| | | | - Erwan Mortier
- Nantes Université, CNRS, Inserm, CRCI 2 NA, Nantes, France
- Nantes Université, Centre Hospitalo-Universitaire (CHU) Nantes, Inserm, CNRS, SFR Bonamy, UMS BioCore, Imp@ct Platform, Inserm, Nantes, France
| | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1 rue Michel-Servet, Geneva, CH-1211, Switzerland.
- Geneva Centre for Inflammation Research, Geneva, Switzerland.
| |
Collapse
|
6
|
Chuang HC, Chang JH, Fan YY, Hsieh CL, Lee YL. Interleukin-38-overexpressing adenovirus infection in dendritic cell-based treatment enhances immunotherapy for allergic asthma via inducing Foxp3 + regulatory T cells. Biomed Pharmacother 2024; 181:117738. [PMID: 39667222 DOI: 10.1016/j.biopha.2024.117738] [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: 08/31/2024] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024] Open
Abstract
Allergic asthma is a chronic disease tied to unusual immune reactions involving type 2 T helper (Th2) cells specific to allergens. Dendritic cells (DCs) play a crucial role in guiding T-cell responses. Regulatory T (Treg) cells have the ability to suppress effector T-cell responses, and interleukin (IL)-38 is involved in Treg cell differentiation. In this study, we explored impacts of IL-38 on the activation and function of DCs, and we then developed an IL-38-overexpressing adenovirus (Ad-IL38) to evaluate its effectiveness in treating allergic asthma in mice through the adaptive transfer of Ad-IL38-infected DCs (IL38-DCs). Treating lipopolysaccharide (LPS)-activated bone marrow-derived DCs with recombinant IL-38 reduced cluster of differentiation 80 (CD80), CD86, and major histocompatibility complex (MHC) II expressions and decreased IL-1β, IL-6, and tumor necrosis factor (TNF)-α while increasing IL-10 secretion. The simultaneous culture of these semi-mature DCs with allogeneic CD4+ T cells facilitated the production of Forkhead box protein P3-positive (Foxp3+) Treg cells. A transcriptomic analysis revealed downregulation of the Chil3, Inhba, and Ctgf genes that are crucial for regulating inflammatory responses and cytokine-mediated signaling pathways in IL-38-treated DCs. In an animal model of asthma, IL38-DC treatment effectively decreased levels of an ovalbumin (OVA)-specific immunoglobulin E (IgE) antibody in serum, attenuated the severity of airway hyperresponsiveness, reduced the production of Th2-type cytokines (IL-4, IL-5, and IL-13) and proinflammatory cytokines (IL-6 and TNF-α) in bronchoalveolar lavage fluid, lowered expressions of the Th2-related cytokines IL-25 and thymic stromal-derived lymphopoietin (TSLP) by lung epithelial cells, and mitigated airway inflammation. Notably, enhanced expression of Foxp3+ Treg cells was linked to increased mRNA levels of transforming growth factor (TGF)-β production in vivo. In conclusion, we comprehensively clarified the immunomodulatory effects of IL-38 on DCs and provide a new treatment with IL-38 genetically modified DCs for alleviating Th2-mediated allergic diseases.
Collapse
Affiliation(s)
- Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Jer-Hwa Chang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yen-Yi Fan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ling Hsieh
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yueh-Lun Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
7
|
Diaz-Barreiro A, Cereghetti G, Ortega Sánchez FG, Tonacini J, Talabot-Ayer D, Kieffer-Jaquinod S, Kissling VM, Huard A, Swale C, Knowles TPJ, Couté Y, Peter M, Francés-Monerris A, Palmer G. Oxidation-sensitive cysteines drive IL-38 amyloid formation. Cell Rep 2024; 43:114940. [PMID: 39488827 DOI: 10.1016/j.celrep.2024.114940] [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: 08/19/2023] [Revised: 07/16/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024] Open
Abstract
Interleukin (IL)-1 family cytokines are essential for host defense at epithelial barriers. The IL-1 family member IL-33 was recently linked to stress granules (SGs). Formation of SGs and other biomolecular condensates is promoted by proteins containing low-complexity regions (LCRs). Computational analysis predicts LCRs in six of the 11 IL-1 family members. Among these, IL-38 contains a long LCR including two amyloid cores. IL-38 localizes to intracellular granules in keratinocytes under oxidative stress (OS) and forms OS-induced amyloid aggregates in cells and in vitro. Interestingly, soluble and aggregated IL-38 are released from keratinocytes in an exosome-enriched extracellular vesicle fraction. Disulfide-bond mapping, in silico modeling, and mutational analysis suggest that oxidation-sensitive cysteines act as redox switches to alter IL-38 conformation and promote its aggregation. Finally, the presence of IL-38 granules in human epidermis facing environmental OS suggests that oxidation-induced amyloidogenesis, as an intrinsic property of IL-38, supports barrier function.
Collapse
Affiliation(s)
- Alejandro Diaz-Barreiro
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland.
| | - Gea Cereghetti
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Francisco Gabriel Ortega Sánchez
- IBS Granada, Institute of Biomedical Research, Granada, Spain; Pulmonology Unit, Hospital Universitario Virgen de las Nieves, Granada, Spain; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - Jenna Tonacini
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Dominique Talabot-Ayer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| | | | - Vera Maria Kissling
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Empa (Swiss Federal Laboratories for Materials Science and Technology), St. Gallen, Switzerland
| | - Arnaud Huard
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Christopher Swale
- Institute for Advanced Biosciences (IAB), Team Host-Pathogen Interactions and Immunity to Infection, University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Grenoble, France
| | | | - Yohann Couté
- Université Grenoble Alpes, CEA, INSERM, UA13 BGE, CNRS, CEA, FR2048 Grenoble, France
| | - Matthias Peter
- Institute of Biochemistry, Department of Biology, ETH Zurich, Zurich, Switzerland
| | | | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| |
Collapse
|
8
|
da Silva P, Mora J, You X, Wiechmann S, Putyrski M, Garcia-Pardo J, Kannt A, Ernst A, Bruene B, Weigert A. Neutralizing IL-38 activates γδ T cell-dependent antitumor immunity and sensitizes for chemotherapy. J Immunother Cancer 2024; 12:e008641. [PMID: 39209451 PMCID: PMC11367332 DOI: 10.1136/jitc-2023-008641] [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] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The interleukin (IL)-1-family receptor antagonist IL-38 has emerged as a negative regulator of auto-inflammation. Given the intricate interplay between antitumor immunity and auto-inflammation, we hypothesized that blocking IL-38 may enhance tumor immune control. METHODS Our hypothesis was tested in the transgenic polyoma virus middle T oncoprotein mammary carcinoma model that is suitable for identifying strong immunomodulators. To investigate the effect of acute IL-38 blockade, we used a neutralizing antibody, alone or in combination with chemotherapy. Immune cell composition and location in tumors were determined by flow cytometry and immunohistochemistry, respectively. The role of γδ T cells was studied using an antibody blocking γδ T-cell receptor signaling. Whole transcriptome RNA sequencing and RNA expression analysis were employed to determine mechanisms downstream of IL-38 neutralization. Additionally, in vitro assays with γδ T cells, CD8+ T cells and cDC1, followed by in vivo CD8+ T cell depletion, were performed to study the underlying mechanistic pathways. RESULTS Both, genetic ablation of IL-38 and neutralization with the antibody, reduced tumorigenesis, and IL-38 blockade improved chemotherapy efficacy. This was accompanied by an augmented lymphocyte infiltrate dominated by γδ T cells and CD8+ T cells, and signaling through the γδ-T-cell receptor was required for CD8+ T cell infiltration. Rather than directly interacting with CD8+ T cells, γδ T cells recruited conventional dendritic cells (cDC1) into tumors via the chemokine Xcl1. cDC1 in turn activated CD8+ T cells via the Notch pathway. Moreover, IL-38 negatively correlated with cDC1, XCL1-producing γδ T cells, T-cell infiltrates and survival in patients with mammary carcinoma. CONCLUSIONS These data suggest that interfering with IL-38 improves antitumor immunity even in immunologically cold tumors.
Collapse
Affiliation(s)
- Priscila da Silva
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Javier Mora
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
- Faculty of Microbiology, University of Costa Rica, San José, Costa Rica
- Centro de Investigación en Cirugía y Cancer (CICICA), University of Costa Rica, 2060 San José, Costa Rica
- Centro de Investigación en Enfermedades Tropicales (CIET), University of Costa Rica, San José, Costa Rica
| | - Xin You
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Svenja Wiechmann
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
| | - Mateusz Putyrski
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
| | - Javier Garcia-Pardo
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Institut de Biotecnologia i de Biomedicina (IBB) and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Aimo Kannt
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Faculty of Medicine, Institute of Clinical Pharmacology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Andreas Ernst
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Faculty of Medicine, Institute of Biochemistry II, Goethe-University Frankfurt, Frankfurt, Germany
| | - Bernhard Bruene
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
- Partner Site Frankfurt, German Cancer Consortium (DKTK), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany
| | - Andreas Weigert
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
- Partner Site Frankfurt, German Cancer Consortium (DKTK), Heidelberg, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany
| |
Collapse
|
9
|
Huard A, Rodriguez E, Talabot-Ayer D, Weigert A, Palmer G. Interleukin-38 overexpression in keratinocytes limits desquamation but does not affect the global severity of imiquimod-induced skin inflammation in mice. Front Immunol 2024; 15:1387921. [PMID: 39119344 PMCID: PMC11306934 DOI: 10.3389/fimmu.2024.1387921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 06/25/2024] [Indexed: 08/10/2024] Open
Abstract
Psoriasis is a common chronic inflammatory skin disease that significantly impacts the patients' quality of life. Recent studies highlighted the function of the interleukin (IL)-1 family member IL-38 in skin homeostasis and suggested an anti-inflammatory role for this cytokine in psoriasis. In this study, we generated mice specifically overexpressing the IL-38 protein in epidermal keratinocytes. We confirmed IL-38 overexpression in the skin by Western blotting. We further detected the protein by ELISA in the plasma, as well as in conditioned media of skin explants isolated from IL-38 overexpressing mice, indicating that IL-38 produced in the epidermis is released from keratinocytes and can be found in the circulation. Unexpectedly, epidermal IL-38 overexpression did not impact the global severity of imiquimod (IMQ)-induced skin inflammation, Similarly, keratinocyte activation and differentiation in IMQ-treated skin were not affected by increased IL-38 expression and there was no global effect on local or systemic inflammatory responses. Nevertheless, we observed a selective inhibition of CXCL1 and IL-6 production in response to IMQ in IL-38 overexpressing skin, as well as reduced Ly6g mRNA levels, suggesting decreased neutrophil infiltration. Epidermal IL-38 overexpression also selectively affected the desquamation process during IMQ-induced psoriasis, as illustrated by reduced plaque formation. Taken together, our results validate the generation of a new mouse line allowing for tissue-specific IL-38 overexpression. Interestingly, epidermal IL-38 overexpression selectively affected specific disease-associated readouts during IMQ-induced psoriasis, suggesting a more complex role of IL-38 in the inflamed skin than previously recognized. In particular, our data highlight a potential involvement of IL-38 in the regulation of skin desquamation.
Collapse
Affiliation(s)
- Arnaud Huard
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Emiliana Rodriguez
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Dominique Talabot-Ayer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Geneva Centre for Inflammation Research, Geneva, Switzerland
| |
Collapse
|
10
|
Wu H, Yang J, Yuan L, Tan Z, Zhang X, Hambly BD, Bao S, Tao K. IL-38 promotes the development of prostate cancer. Front Immunol 2024; 15:1384416. [PMID: 38779687 PMCID: PMC11109393 DOI: 10.3389/fimmu.2024.1384416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Introduction Prostate Cancer (PCa) remains a significant concern in male cancer-related mortality. Tumour development is intricately regulated by the complex interactions between tumour cells and their microenvironment, making it essential to determine which is/are key factor(s) that influence the progression of PCa within the tumour microenvironment. Materials and methods The current study utilised histopathology and immunohistochemistry to determine the expression of IL-38 in PCa and analysed the correlation between the expression level of IL-38 within PCa and clinical pathological characteristics. Results There was a significant increase in IL-38 expression in PCa tissues compared to adjacent non-PCa tissues (P < 0.0001). In addition, IL-38 expression was significantly higher in tumour cells with a high proliferation index compared to those with a low value-added index. ROC curve analysis demonstrated that IL-38 has high specificity and sensitivity for the diagnosis of PCa (AUC=0.76). Moreover, we Probed the cellular source of IL-38 in prostate cancer tissue by immunofluorescence double staining. Additionally, within PCa, the expression of IL-38 was inversely correlated with the expression levels of CD8 and PD-1. Survival analysis revealed a significantly lower overall survival rate for PCa patients with high IL-38 expression (P=0.0069), and when IL-38 was co-expressed with CD8, the survival rate of the IL-38high/CD8low group was decreased significantly. Multivariate analysis indicated that the expression level of IL-38 and TNM staging were independent predictors of survival in PCa patients. Conclusion These findings suggest that IL-38 plays a crucial role in the development of PCa, and the exploration of the correlation between IL-38 and various immune factors in the tumour microenvironment further reveals its mechanism of action, making it a potential target for immunotherapy in PCa.
Collapse
Affiliation(s)
- Huiyan Wu
- Department of Pathology, Tongji Hospital, Tongji University, Shanghai, China
- Department of Pathology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jing Yang
- Department of Pathology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Liuhong Yuan
- Department of Pathology, Tongji Hospital, Tongji University, Shanghai, China
| | - Zhenyu Tan
- Department of Pathology, Tongji Hospital, Tongji University, Shanghai, China
| | - Xiuqin Zhang
- Department of Pathology, Tongji Hospital, Tongji University, Shanghai, China
| | - Brett D. Hambly
- Department of Pathology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shisan Bao
- Department of Pathology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kun Tao
- Department of Pathology, Tongji Hospital, Tongji University, Shanghai, China
- Department of Pathology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Yuan L, Tan Z, Huang J, Chen F, Hambly BD, Bao S, Tao K. Exploring the clinical significance of IL-38 correlation with PD-1, CTLA-4, and FOXP3 in colorectal cancer draining lymph nodes. Front Immunol 2024; 15:1384548. [PMID: 38533512 PMCID: PMC10963446 DOI: 10.3389/fimmu.2024.1384548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
Introduction Colorectal cancer (CRC) presents a substantial challenge characterized by unacceptably high mortality and morbidity, primarily attributed to delayed diagnosis and reliance on palliative care. The immune response of the host plays a pivotal role in carcinogenesis, with IL-38 emerging as a potential protective factor in CRC. However, the precise involvement of IL-38 among various leucocytes, its interactions with PD-1/PD-L1, and its impact on metastasis require further elucidation. Results Our investigation revealed a significant correlation between IL-38 expression and metastasis, particularly concerning survival and interactions among diverse leucocytes within draining lymph nodes. In the mesentery lymph nodes, we observed an inverse correlation between IL-38 expression and stages of lymph node invasions (TNM), invasion depth, distance, and differentiation. This aligns with an overall survival advantage associated with higher IL-38 expression in CRC patients' nodes compared to lower levels, as well as elevated IL-38 expression on CD4+ or CD8+ cells. Notably, a distinct subset of patients characterized by IL-38high/PD-1low expression exhibited superior survival outcomes compared to other combinations. Discussion Our findings demonstrate that IL-38 expression in colorectal regional nodes from CRC patients is inversely correlated with PD-1/PD-L1 but positively correlated with infiltrating CD4+ or CD8+ lymphocytes. The combined assessment of IL-38 and PD-1 expression in colorectal regional nodes emerges as a promising biomarker for predicting the prognosis of CRC.
Collapse
Affiliation(s)
- Liuhong Yuan
- Department of Pathology, Tongji Hospital, Tongji University, Shanghai, China
| | - Zhenyu Tan
- Department of Pathology, Tongji Hospital, Tongji University, Shanghai, China
| | - Junjie Huang
- Department of Pathology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Feier Chen
- Department of Pathology, Tongji Hospital, Tongji University, Shanghai, China
| | - Brett D. Hambly
- Department of Pathology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shisan Bao
- Department of Pathology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kun Tao
- Department of Pathology, Tongji Hospital, Tongji University, Shanghai, China
| |
Collapse
|
12
|
Gao Y, Cai L, Wu Y, Jiang M, Zhang Y, Ren W, Song Y, Li L, Lei Z, Wu Y, Zhu L, Li J, Li D, Li G, Luo C, Tao L. Emerging functions and therapeutic targets of IL-38 in central nervous system diseases. CNS Neurosci Ther 2024; 30:e14550. [PMID: 38334236 PMCID: PMC10853902 DOI: 10.1111/cns.14550] [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: 05/08/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 02/10/2024] Open
Abstract
Interleukin (IL)-38 is a newly discovered cytokine of the IL-1 family, which binds various receptors (i.e., IL-36R, IL-1 receptor accessory protein-like 1, and IL-1R1) in the central nervous system (CNS). The hallmark physiological function of IL-38 is competitive binding to IL-36R, as does the IL-36R antagonist. Emerging research has shown that IL-38 is abnormally expressed in the serum and brain tissue of patients with ischemic stroke (IS) and autism spectrum disorder (ASD), suggesting that IL-38 may play an important role in neurological diseases. Important advances include that IL-38 alleviates neuromyelitis optica disorder (NMOD) by inhibiting Th17 expression, improves IS by protecting against atherosclerosis via regulating immune cells and inflammation, and reduces IL-1β and CXCL8 release through inhibiting human microglial activity post-ASD. In contrast, IL-38 mRNA is markedly increased and is mainly expressed in phagocytes in spinal cord injury (SCI). IL-38 ablation attenuated SCI by reducing immune cell infiltration. However, the effect and underlying mechanism of IL-38 in CNS diseases remain inadequately characterized. In this review, we summarize the biological characteristics, pathophysiological role, and potential mechanisms of IL-38 in CNS diseases (e.g., NMOD, Alzheimer's disease, ASD, IS, TBI, and SCI), aiming to explore the therapeutic potential of IL-38 in the prevention and treatment of CNS diseases.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Forensic Medicine, School of Basic Medicine and Biological SciencesSoochow UniversitySuzhouChina
- Department of NeurosurgeryPennsylvania State University College of MedicineState CollegePennsylvaniaUSA
- Department of Forensic ScienceWenzhou Medical UniversityWenzhouZhejiangChina
| | - Luwei Cai
- Department of Forensic Medicine, School of Basic Medicine and Biological SciencesSoochow UniversitySuzhouChina
| | - Yulu Wu
- Department of Forensic Medicine, School of Basic Medicine and Biological SciencesSoochow UniversitySuzhouChina
| | - Min Jiang
- Department of Forensic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yidan Zhang
- Department of Forensic Medicine, School of Basic Medicine and Biological SciencesSoochow UniversitySuzhouChina
| | - Wenjing Ren
- Department of Forensic Medicine, School of Basic Medicine and Biological SciencesSoochow UniversitySuzhouChina
| | - Yirui Song
- Department of Forensic Medicine, School of Basic Medicine and Biological SciencesSoochow UniversitySuzhouChina
| | - Lili Li
- Department of Child and Adolescent HealthcareChildren's Hospital of Soochow UniversitySuzhouChina
| | - Ziguang Lei
- Department of Forensic ScienceWenzhou Medical UniversityWenzhouZhejiangChina
| | - Youzhuang Wu
- Department of Forensic ScienceWenzhou Medical UniversityWenzhouZhejiangChina
| | - Luwen Zhu
- Department of Forensic ScienceWenzhou Medical UniversityWenzhouZhejiangChina
| | - Jing Li
- Department of Forensic Medicine, School of Basic Medicine and Biological SciencesSoochow UniversitySuzhouChina
| | - Dongya Li
- Department of OrthopedicsThe Affiliated Hospital of Xuzhou Medical UniversityXuzhouChina
| | - Guohong Li
- Department of NeurosurgeryPennsylvania State University College of MedicineState CollegePennsylvaniaUSA
| | - Chengliang Luo
- Department of Forensic Medicine, School of Basic Medicine and Biological SciencesSoochow UniversitySuzhouChina
| | - Luyang Tao
- Department of Forensic Medicine, School of Basic Medicine and Biological SciencesSoochow UniversitySuzhouChina
| |
Collapse
|
13
|
Papareddy P, Selle M, Partouche N, Legros V, Rieu B, Olinder J, Ryden C, Bartakova E, Holub M, Jung K, Pottecher J, Herwald H. Identifying biomarkers deciphering sepsis from trauma-induced sterile inflammation and trauma-induced sepsis. Front Immunol 2024; 14:1310271. [PMID: 38283341 PMCID: PMC10820703 DOI: 10.3389/fimmu.2023.1310271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/22/2023] [Indexed: 01/30/2024] Open
Abstract
Objective The purpose of this study was to identify a panel of biomarkers for distinguishing early stage sepsis patients from non-infected trauma patients. Background Accurate differentiation between trauma-induced sterile inflammation and real infective sepsis poses a complex life-threatening medical challenge because of their common symptoms albeit diverging clinical implications, namely different therapies. The timely and accurate identification of sepsis in trauma patients is therefore vital to ensure prompt and tailored medical interventions (provision of adequate antimicrobial agents and if possible eradication of infective foci) that can ultimately lead to improved therapeutic management and patient outcome. The adequate withholding of antimicrobials in trauma patients without sepsis is also important in aspects of both patient and environmental perspective. Methods In this proof-of-concept study, we employed advanced technologies, including Matrix-Assisted Laser Desorption/Ionization (MALDI) and multiplex antibody arrays (MAA) to identify a panel of biomarkers distinguishing actual sepsis from trauma-induced sterile inflammation. Results By comparing patient groups (controls, infected and non-infected trauma and septic shock patients under mechanical ventilation) at different time points, we uncovered distinct protein patterns associated with early trauma-induced sterile inflammation on the one hand and sepsis on the other hand. SYT13 and IL1F10 emerged as potential early sepsis biomarkers, while reduced levels of A2M were indicative of both trauma-induced inflammation and sepsis conditions. Additionally, higher levels of TREM1 were associated at a later stage in trauma patients. Furthermore, enrichment analyses revealed differences in the inflammatory response between trauma-induced inflammation and sepsis, with proteins related to complement and coagulation cascades being elevated whereas proteins relevant to focal adhesion were diminished in sepsis. Conclusions Our findings, therefore, suggest that a combination of biomarkers is needed for the development of novel diagnostic approaches deciphering trauma-induced sterile inflammation from actual infective sepsis.
Collapse
Affiliation(s)
- Praveen Papareddy
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Michael Selle
- Genomics and Bioinformatics of Infectious Diseases, Institute for Animal Genomics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Nicolas Partouche
- Hôpitaux Universitaires de Strasbourg, Service d’Anesthésie-Réanimation & Médecine Péri-opératoire - Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Vincent Legros
- Département d’Anesthésie-Réanimation et Médecine Peri-Operatoire, Centre Hospitalier et Universitaire (CHU) de Reims, Université de Reims Champagne-Ardenne, Reims, France
| | - Benjamin Rieu
- Réanimation Médico-Chirurgicale, Trauma Center, Pôle Médecine Péri-Opératoire, Centre Hospitalier et Universitaire (CHU) de Clermont-Ferrand, Clermont Ferrand, France
| | - Jon Olinder
- Division of Infection Medicine, Helsingborg Hospital and Department of Clinical Sciences Helsingborg, Lund University, Helsingborg, Sweden
| | - Cecilia Ryden
- Division of Infection Medicine, Helsingborg Hospital and Department of Clinical Sciences Helsingborg, Lund University, Helsingborg, Sweden
| | - Eva Bartakova
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| | - Klaus Jung
- Genomics and Bioinformatics of Infectious Diseases, Institute for Animal Genomics, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Julien Pottecher
- Hôpitaux Universitaires de Strasbourg, Service d’Anesthésie-Réanimation & Médecine Péri-opératoire - Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Heiko Herwald
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
14
|
Kiełbowski K, Herian M, Bakinowska E, Banach B, Sroczyński T, Pawlik A. The Role of Genetics and Epigenetic Regulation in the Pathogenesis of Osteoarthritis. Int J Mol Sci 2023; 24:11655. [PMID: 37511413 PMCID: PMC10381003 DOI: 10.3390/ijms241411655] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Osteoarthritis (OA) is progressive disease characterised by cartilage degradation, subchondral bone remodelling and inflammation of the synovium. The disease is associated with obesity, mechanical load and age. However, multiple pro-inflammatory immune mediators regulate the expression of metalloproteinases, which take part in cartilage degradation. Furthermore, genetic factors also contribute to OA susceptibility. Recent studies have highlighted that epigenetic mechanisms may regulate the expression of OA-associated genes. This review aims to present the mechanisms of OA pathogenesis and summarise current evidence regarding the role of genetics and epigenetics in this process.
Collapse
Affiliation(s)
| | | | | | | | | | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (M.H.); (E.B.); (B.B.); (T.S.)
| |
Collapse
|
15
|
Huard A, Wilmes C, Kiprina A, Netzer C, Palmer G, Brüne B, Weigert A. Cell Intrinsic IL-38 Affects B Cell Differentiation and Antibody Production. Int J Mol Sci 2023; 24:ijms24065676. [PMID: 36982750 PMCID: PMC10053218 DOI: 10.3390/ijms24065676] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/18/2023] Open
Abstract
IL-38 is an IL-1 family receptor antagonist with an emerging role in chronic inflammatory diseases. IL-38 expression has been mainly observed not only in epithelia, but also in cells of the immune system, including macrophages and B cells. Given the association of both IL-38 and B cells with chronic inflammation, we explored if IL-38 affects B cell biology. IL-38-deficient mice showed higher amounts of plasma cells (PC) in lymphoid organs but, conversely, lower levels of plasmatic antibody titers. Exploring underlying mechanisms in human B cells revealed that exogenously added IL-38 did not significantly affect early B cell activation or differentiation into plasma cells, even though IL-38 suppressed upregulation of CD38. Instead, IL-38 mRNA expression was transiently upregulated during the differentiation of human B cells to plasma cells in vitro, and knocking down IL-38 during early B cell differentiation increased plasma cell generation, while reducing antibody production, thus reproducing the murine phenotype. Although this endogenous role of IL-38 in B cell differentiation and antibody production did not align with an immunosuppressive function, autoantibody production induced in mice by repeated IL-18 injections was enhanced in an IL-38-deficient background. Taken together, our data suggest that cell-intrinsic IL-38 promotes antibody production at baseline but suppresses the production of autoantibodies in an inflammatory context, which may partially explain its protective role during chronic inflammation.
Collapse
Affiliation(s)
- Arnaud Huard
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Christian Wilmes
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Anastasiia Kiprina
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Christoph Netzer
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Cardio-Pulmonary Institute (CPI), 60590 Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Cardio-Pulmonary Institute (CPI), 60590 Frankfurt, Germany
- Correspondence: ; Tel.: +49-69-6301-4593; Fax: +49-69-6301-420
| |
Collapse
|
16
|
Baker KJ, Buskiewicz E, Finucane M, Chelliah A, Burke L, Houston A, Brint E. IL-36 expression is increased in NSCLC with IL-36 stimulation of lung cancer cells promoting a pro-tumorigenic phenotype. Cytokine 2023; 165:156170. [PMID: 36931148 DOI: 10.1016/j.cyto.2023.156170] [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: 11/08/2022] [Revised: 02/28/2023] [Accepted: 02/28/2023] [Indexed: 03/17/2023]
Abstract
The IL-36 cytokines are a recently described subset of the IL-1 family of cytokines, and have been shown to play a role in the pathogenesis of respiratory diseases such as asthma and COPD. Given the common aetiological links between COPD and lung cancer development, as well as the involvement of other IL-1 family members in lung tumorigenesis, the aim of this work was to investigate the role of IL-36 cytokines in the pathogenesis of lung cancer. In this study we demonstrate that expression of IL-36 cytokines and receptor mRNA and protein are significantly increased in lung cancer tissue compared to adjacent non-tumour tissue. In vitro assays showed that stimulation of two lung cancer cell lines, SKMES-1 human squamous cell and LLC murine lung cancer, with IL-36R agonists resulted in increased cellular migration and proliferation. All IL-36 cytokines induced the expression of pro-inflammatory chemokines in both lung cancer cell lines with synergistic effects identified upon co-stimulation of cells with IL-17, IL-22 and TNFα. Furthermore, we report that IL-36 cytokines induce protein expression of the immune checkpoint inhibitor protein PD-L1 on lung cancer cells. Taken together, this data indicates that targeting IL-36R signalling may be a useful targeted therapy for lung cancer patients with IL-36R+ cancer cells.
Collapse
Affiliation(s)
- Kevin James Baker
- Department of Pathology, UCC, Cork, Ireland; Department of Medicine, UCC, Cork, Ireland
| | | | - Méabh Finucane
- Department of Pathology, UCC, Cork, Ireland; Department of Medicine, UCC, Cork, Ireland
| | | | - Louise Burke
- Department of Pathology, UCC, Cork, Ireland; Dept. Of Pathology CUH, Ireland
| | - Aileen Houston
- Department of Medicine, UCC, Cork, Ireland; APC Microbiome Ireland, UCC, Cork, Ireland
| | - Elizabeth Brint
- Department of Pathology, UCC, Cork, Ireland; APC Microbiome Ireland, UCC, Cork, Ireland.
| |
Collapse
|
17
|
Dowling JP, Nikitin PA, Shen F, Shukla H, Finn JP, Patel N, Swider C, Bingaman-Steele JL, Nicolescu C, Sikorski EL, Greenawalt EJ, Morin MJ, Robinson MK, Lundgren K, Harman BC. IL-38 blockade induces anti-tumor immunity by abrogating tumor-mediated suppression of early immune activation. MAbs 2023; 15:2212673. [PMID: 37216961 DOI: 10.1080/19420862.2023.2212673] [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: 08/02/2022] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/24/2023] Open
Abstract
Immune checkpoint inhibitors that overcome T cell suppressive mechanisms in tumors have revolutionized the treatment of cancer but are only efficacious in a small subset of patients. Targeting suppressive mechanisms acting on innate immune cells could significantly improve the incidence of clinical response by facilitating a multi-lineage response against the tumor involving both adaptive and innate immune systems. Here, we show that intra-tumoral interleukin (IL)-38 expression is a feature of a large frequency of head and neck, lung and cervical squamous cancers and correlates with reduced immune cell numbers. We generated IMM20324, an antibody that binds human and mouse IL-38 proteins and inhibits the binding of IL-38 to its putative receptors, interleukin 1 receptor accessory protein-like 1 (IL1RAPL) and IL-36R. In vivo, IMM20324 demonstrated a good safety profile, delayed tumor growth in a subset of mice in an EMT6 syngeneic model of breast cancer, and significantly inhibited tumor expansion in a B16.F10 melanoma model. Notably, IMM20324 treatment resulted in the prevention of tumor growth following re-implantation of tumor cells, indicating the induction of immunological memory. Furthermore, exposure of IMM20324 correlated with decreased tumor volume and increased levels of intra-tumoral chemokines. Together, our data suggest that IL-38 is expressed in a high frequency of cancer patients and allows tumor cells to suppress anti-tumor immunity. Blockade of IL-38 activity using IMM20324 can re-activate immunostimulatory mechanisms in the tumor microenvironment leading to immune infiltration, the generation of tumor-specific memory and abrogation of tumor growth.
Collapse
Affiliation(s)
| | | | - Fang Shen
- Research & Development, Immunome Inc, Exton, PA, USA
| | - Halley Shukla
- Research & Development, Immunome Inc, Exton, PA, USA
| | - James P Finn
- Research & Development, Immunome Inc, Exton, PA, USA
| | - Nirja Patel
- Research & Development, Immunome Inc, Exton, PA, USA
| | - Cezary Swider
- Research & Development, Immunome Inc, Exton, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Frenay J, Bellaye PS, Oudot A, Helbling A, Petitot C, Ferrand C, Collin B, Dias AMM. IL-1RAP, a Key Therapeutic Target in Cancer. Int J Mol Sci 2022; 23:ijms232314918. [PMID: 36499246 PMCID: PMC9735758 DOI: 10.3390/ijms232314918] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer is a major cause of death worldwide and especially in high- and upper-middle-income countries. Despite recent progress in cancer therapies, such as chimeric antigen receptor T (CAR-T) cells or antibody-drug conjugate (ADC), new targets expressed by the tumor cells need to be identified in order to selectively drive these innovative therapies to tumors. In this context, IL-1RAP recently showed great potential to become one of these new targets for cancer therapy. IL-1RAP is highly involved in the inflammation process through the interleukins 1, 33, and 36 (IL-1, IL-33, IL-36) signaling pathways. Inflammation is now recognized as a hallmark of carcinogenesis, suggesting that IL-1RAP could play a role in cancer development and progression. Furthermore, IL-1RAP was found overexpressed on tumor cells from several hematological and solid cancers, thus confirming its potential involvement in carcinogenesis. This review will first describe the structure and genetics of IL-1RAP as well as its role in tumor development. Finally, a focus will be made on the therapies based on IL-1RAP targeting, which are now under preclinical or clinical development.
Collapse
Affiliation(s)
- Jame Frenay
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Pierre-Simon Bellaye
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Alexandra Oudot
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Alex Helbling
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Camille Petitot
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Christophe Ferrand
- INSERM UMR1098, EFS BFC, Université de Bourgogne Franche-Comté, 25000 Besançon, France
- CanCell Therapeutics, 25000 Besançon, France
| | - Bertrand Collin
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
- Institut de Chimie Moléculaire de l'Université de Bourgogne, UMR CNRS 6302, 21000 Dijon, France
| | - Alexandre M M Dias
- Plateforme d'Imagerie et Radiothérapie Précliniques, Médecine Nucléaire, Centre Georges-François Leclerc, 21000 Dijon, France
| |
Collapse
|
19
|
Talabot-Ayer D, Diaz-Barreiro A, Modarressi A, Palmer G. Epigenetic remodeling of downstream enhancer regions is linked to selective expression of the IL1F10 gene in differentiated human keratinocytes. Gene 2022; 842:146800. [PMID: 35961432 DOI: 10.1016/j.gene.2022.146800] [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: 03/23/2022] [Revised: 07/02/2022] [Accepted: 08/05/2022] [Indexed: 11/04/2022]
Abstract
Interleukin (IL)-38, encoded by the IL1F10 gene, is a member of the IL-1 family of cytokines. IL-38 is constitutively expressed in epithelia in healthy humans, and in particular in epidermal keratinocytes in the skin. IL-38 expression is closely correlated with keratinocyte differentiation. The aim of this study was to further characterize the regulation of IL1F10 expression and the mechanisms involved in its selective induction in differentiated human keratinocytes. We observed coordinated expression of two IL1F10 transcripts, transcribed from two different promoters, upon differentiation of primary human keratinocytes. Using ENCODE datasets and ChIP-qPCR on ex vivo isolated normal human epidermis, we identified regulatory regions located downstream of the IL1F10 gene, which displayed features of differentiated keratinocyte-specific enhancers. Expression of the IL1F10 gene was linked to changes in the epigenetic landscape at these downstream enhancer regions in human epidermis. Overexpression of the transcription factors KLF4 and TAp63β in an immortalized normal human keratinocyte (iNHK) cell line promoted the expression of mRNA encoding the differentiation markers keratin 10 and involucrin, and of IL1F10. ChIP-qPCR experiments indicated that KLF4 and TAp63β overexpression also modified the chromatin state of the proximal downstream enhancer region, suggesting a role for KLF4 and TAp63β in directly or indirectly regulating IL1F10 transcription. In conclusion, expression of the IL1F10 gene in differentiated keratinocytes in normal human epidermis involves coordinated transcription from two promoters and is linked to epigenetic remodeling of enhancer regions located downstream of the gene.
Collapse
Affiliation(s)
- Dominique Talabot-Ayer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Alejandro Diaz-Barreiro
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ali Modarressi
- Division of Plastic, Reconstructive & Aesthetic Surgery, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
20
|
Haghshenas MR, Zamir MR, Sadeghi M, Fattahi MJ, Mirshekari K, Ghaderi A. Clinical relevance and therapeutic potential of IL-38 in immune and non-immune-related disorders. Eur Cytokine Netw 2022; 33:54-69. [PMID: 37052152 PMCID: PMC10134710 DOI: 10.1684/ecn.2022.0480] [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] [Accepted: 07/30/2022] [Indexed: 04/14/2023]
Abstract
Interleukin-38 (IL-38) is the most recent member of the IL-1 family that acts as a natural inflammatory inhibitor by binding to cognate receptors, particularly the IL-36 receptor. In vitro, animal and human studies on autoimmune, metabolic, cardiovascular and allergic diseases, as well sepsis and respiratory viral infections, have shown that IL-38 exerts an anti-inflammatory activity by modulating the generation and function of inflammatory cytokines (e.g. IL-6, IL-8, IL-17 and IL-36) and regulating dendritic cells, M2 macrophages and regulatory T cells (Tregs). Accordingly, IL-38 may possess therapeutic potential for these types of diseases. IL-38 down-regulates CCR3+ eosinophil cells, CRTH2+ Th2 cells, Th17 cells, and innate lymphoid type 2 cells (ILC2), but up-regulates Tregs, and this has influenced the design of immunotherapeutic strategies based on regulatory cells/cytokines for allergic asthma in future studies. In auto-inflammatory diseases, IL-38 alleviates skin inflammation by regulating γδ T cells and limiting the production of IL-17. Due to its ability to suppress IL-1β, IL-6 and IL-36, this cytokine could reduce COVID-19 severity, and might be employed as a therapeutic tool. IL-38 may also influence host immunity and/or the components of the cancer microenvironment, and has been shown to improve the outcome of colorectal cancer, and may participate in tumour progression in lung cancer possibly by modulating CD8 tumour infiltrating T cells and PD-L1 expression. In this review, we first briefly present the biological and immunological functions of IL-38, and then discuss the important roles of IL-38 in various types of diseases, and finally highlight its use in therapeutic strategies.
Collapse
Affiliation(s)
- Mohammad Reza Haghshenas
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Roshan Zamir
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboubeh Sadeghi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Fattahi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kimia Mirshekari
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
21
|
Special Issue "IL-1 family cytokines in host defense, inflammation and cancer". Cytokine 2022; 158:156010. [PMID: 36029747 DOI: 10.1016/j.cyto.2022.156010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Teufel LU, de Graaf DM, Netea MG, Dinarello CA, Joosten LAB, Arts RJW. Circulating interleukin-38 concentrations in healthy adults. Front Immunol 2022; 13:964365. [PMID: 36016926 PMCID: PMC9396651 DOI: 10.3389/fimmu.2022.964365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Interleukin (IL)-38 is the latest discovered member of the interleukin-1 family, which has anti-inflammatory properties similar to IL-36Ra. Several studies compared circulating IL-38 concentrations in healthy and diseased populations to characterize its role in both auto-immune and inflammatory pathologies, with both higher and lower concentrations being associated with certain diseases. However, in order to use IL-38 as a biomarker, a reference range in healthy adults is needed. To establish a reference IL-38 circulating concentration, accessible data from 25 eligible studies with IL-38 concentrations in healthy adults was collected. To validate the values found in literature, we measured IL-38 concentrations by enzyme-linked immunosorbent assay (ELISA) in several cohorts from our own institute. Additionally, the effect of blood collection techniques, freeze thawing cycles, and hemolysis on IL-38 measurements was assessed. To evaluate the importance of the genetic background of individuals as confounding factor of IL-38 synthesis, we used publicly available eQTL databases with matched data on allele frequencies in individuals of different ethnicities. Mean IL-38 concentrations in the various studies were weighted by their corresponding sample size, resulting in a weighted mean, and weighted upper and lower limits were calculated by mean ± 2 SD. Differences of over 10.000-fold were found in the weighted means between studies, which could not be attributed to the blood collection method or assessment of IL-38 in plasma or serum. Although IL-38 concentrations were markedly higher in Chinese then in European population studies, we could not show an association with the genetic background. From our analysis, a reference range for circulating IL-38 in healthy adults could thus not yet be established.
Collapse
Affiliation(s)
- Lisa U. Teufel
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Dennis M. de Graaf
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
- Department of Medicine, University of Colorado, Aurora, CO, United States
- Institute of Innate Immunity, University Hospital, University of Bonn, Bonn, Germany
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Charles A. Dinarello
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Leo A. B. Joosten
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
- Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rob J. W. Arts
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
- *Correspondence: Rob J. W. Arts,
| |
Collapse
|