1
|
Yu S, Qi R, Xiao L, Huang Y, Yu L. Single-cell transcriptome reveals potential mechanisms for gout in children. Front Immunol 2025; 16:1577109. [PMID: 40370447 PMCID: PMC12076732 DOI: 10.3389/fimmu.2025.1577109] [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: 02/15/2025] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Objective Pediatric gout is a condition that differs from traditional adult gout and has attracted significant attention. This study aims to explore the molecular mechanisms underlying pediatric gout. Methods We analyzed peripheral blood samples from pediatric gout patients and healthy controls using single-cell RNA sequencing (scRNA-seq). Statistical tests were employed to analyze the data and identify significant differences between the groups. Results Our findings revealed that CD14+ monocytes and DN T cells play crucial roles in pediatric gout. CD14+ monocytes are essential for recognizing and phagocytosing monosodium urate (MSU) crystals, triggering inflammation. DN T cells may be involved in the adaptive immune response within gouty joints. These cells not only contribute to the inflammatory response but also interact with other immune cells, amplifying the inflammatory cascade. Comparative analysis with adult gout studies highlighted both similarities and differences in cellular and molecular mechanisms between children and adults. The CD14+ monocytes may be interact with other immune cells through the TNF-α/NF-κB signaling pathway. Targeting this pathway may offer therapeutic potential for managing pediatric gout. Conclusion The results provide a foundation for new diagnostic markers and therapeutic targets for pediatric gout. They also pave the way for future research and the development of targeted therapies that can effectively manage and potentially prevent the debilitating effects of gout in children. Understanding the unique molecular mechanisms in pediatric gout could influence treatment strategies and improve patient outcomes.
Collapse
Affiliation(s)
- Shengyou Yu
- Department of Pediatrics, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - Ren Qi
- Department of Rheumatology and Immunology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Liang Xiao
- Department of Pediatrics, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, China
| | - YiHui Huang
- Department of Pediatrics, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Li Yu
- Department of Pediatrics, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Badii M, Nica V, Straton AR, Kischkel B, Gaal O, Cabău G, Klück V, Hotea I, Novakovic B, Pamfil C, Rednic S, Netea MG, Popp RA, Joosten LAB, Crișan TO. Downregulation of type I interferon signalling pathway by urate in primary human PBMCs. Immunology 2025; 174:100-112. [PMID: 39354748 PMCID: PMC11652411 DOI: 10.1111/imm.13858] [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: 12/05/2023] [Accepted: 08/23/2024] [Indexed: 10/03/2024] Open
Abstract
Type I interferons (IFN1s) mediate innate responses to microbial stimuli and regulate interleukin (IL)-1 and IL-1 receptor antagonist (Ra) production in human cells. This study explores interferon-stimulated gene (ISG) alterations in the transcriptome of patients with gout and stimulated human primary cells in vitro in relation to serum urate concentrations. Peripheral blood mononuclear cells (PBMCs) and monocytes of patients with gout were primed in vitro with soluble urate, followed by lipopolysaccharide (LPS) stimulation. Separately, PBMCs were stimulated with various toll-like receptor (TLR) ligands. RNA sequencing and IL-1Ra cytokine measurement were performed. STAT1 phosphorylation was assessed in urate-treated monocytes. Cytokine responses to IFN-β were evaluated in PBMCs cultured with or without urate and restimulated with LPS and monosodium urate (MSU) crystals. Transcriptomics revealed suppressed IFN-related signalling pathways in urate-exposed PBMCs or monocytes which was supported by diminishment of phosphorylated STAT1. The stimulation of PBMCs with IFN-β did not modify the urate-induced inflammation. Interestingly, in vivo, serum urate concentrations were inversely correlated to in vitro ISG expression upon stimulations with TLR ligands. These findings support a deficient IFN1 signalling in the presence of elevated serum urate concentrations, which could translate to increased susceptibility to infections.
Collapse
Affiliation(s)
- Medeea Badii
- Department of Medical GeneticsIuliu Hațieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Internal Medicine and Research Institute for Medical InnovationRadboud University Medical CentreNijmegenThe Netherlands
| | - Valentin Nica
- Department of Medical GeneticsIuliu Hațieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Ancuța R. Straton
- Department of Medical GeneticsIuliu Hațieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Brenda Kischkel
- Department of Internal Medicine and Research Institute for Medical InnovationRadboud University Medical CentreNijmegenThe Netherlands
| | - Orsolya Gaal
- Department of Medical GeneticsIuliu Hațieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Internal Medicine and Research Institute for Medical InnovationRadboud University Medical CentreNijmegenThe Netherlands
| | - Georgiana Cabău
- Department of Medical GeneticsIuliu Hațieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Viola Klück
- Department of Internal Medicine and Research Institute for Medical InnovationRadboud University Medical CentreNijmegenThe Netherlands
| | - Ioana Hotea
- Department of Medical GeneticsIuliu Hațieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Boris Novakovic
- Murdoch Children's Research Institute and Department of PaediatricsUniversity of Melbourne, Royal Children's HospitalParkvilleVictoriaAustralia
| | - Cristina Pamfil
- Department of RheumatologyIuliu Hațieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Simona Rednic
- Department of RheumatologyIuliu Hațieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Mihai G. Netea
- Department of Internal Medicine and Research Institute for Medical InnovationRadboud University Medical CentreNijmegenThe Netherlands
| | - Radu A. Popp
- Department of Medical GeneticsIuliu Hațieganu University of Medicine and PharmacyCluj‐NapocaRomania
| | - Leo A. B. Joosten
- Department of Medical GeneticsIuliu Hațieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Internal Medicine and Research Institute for Medical InnovationRadboud University Medical CentreNijmegenThe Netherlands
| | - Tania O. Crișan
- Department of Medical GeneticsIuliu Hațieganu University of Medicine and PharmacyCluj‐NapocaRomania
- Department of Internal Medicine and Research Institute for Medical InnovationRadboud University Medical CentreNijmegenThe Netherlands
| |
Collapse
|
3
|
Gaal OI, Leask M, Nica V, Cabău G, Badii M, Hotea I, de Graaf DM, Zhang Z, Li Y, Pamfil C, Rednic S, Merriman TR, Crișan TO, Joosten LAB. Gout-associated SNP at the IL1RN-IL1F10 region is associated with altered cytokine production in PBMCs of patients with gout and controls. Arthritis Res Ther 2024; 26:205. [PMID: 39568029 PMCID: PMC11577629 DOI: 10.1186/s13075-024-03436-0] [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: 08/26/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024] Open
Abstract
OBJECTIVES Gout is caused by the response of the innate immune system to monosodium urate (MSU) crystals. A recent gout GWAS identified a signal of genetic association at a locus encompassing IL1RN-IL1F10. Colocalisation analysis using Genotype Tissue Expression Database (GTEx) eQTL data showed that the signal overlaps with genetic control of IL1RN/IL1F10 gene expression. We assess the functional implications of IL1RN rs9973741, the lead gout-associated variant. METHODS We conducted functional validation of IL1RN rs9973741 in patients with gout and controls. The transcription level of IL1RN/IL1F10 was investigated in unstimulated or MSU-crystal co-stimulated PBMCs. Ex vivo functional assays were performed in PBMCs stimulated with C16 + MSU crystals or LPS for 24 h. Cytokine levels were assessed by ELISA. RESULTS In unstimulated PBMCs, no association of IL1RN rs9973741 (gout risk allele G) to IL1RN expression was observed while IL-1F10 was hindered by low expression at both transcriptional and protein levels. However, G allele carriers showed lower IL1RN expression in PBMCs stimulated with C16/MSU crystal and lower concentrations of circulating IL-1Ra in both controls and gout patients. PBMCs depicted less spontaneous IL-1Ra release in GG homozygous controls and lower IL-1Ra production in response to C16 + MSU crystal costimulation in patients with gout. The G allele was associated with elevated IL-1β cytokine production in response to C16 + MSU crystal stimulation in controls. CONCLUSIONS The gout risk allele G associates with lower circulating IL-1Ra, lower IL-1Ra production in PBMC assays and elevated IL-1β production in PBMCs challenged with C16 + MSU crystals but not in unchallenged cells. Our data indicate that the genetic signal that associates with gout at IL1RN-IL1F10 region functions to alter expression of IL-1Ra when stimulated by MSU crystals.
Collapse
Grants
- P_37_762, MySMIS 103587 Competitiveness Operational Programme grant of the Romanian Ministry of European Funds
- P_37_762, MySMIS 103587 Competitiveness Operational Programme grant of the Romanian Ministry of European Funds
- P_37_762, MySMIS 103587 Competitiveness Operational Programme grant of the Romanian Ministry of European Funds
- P_37_762, MySMIS 103587 Competitiveness Operational Programme grant of the Romanian Ministry of European Funds
- P_37_762, MySMIS 103587 Competitiveness Operational Programme grant of the Romanian Ministry of European Funds
- P_37_762, MySMIS 103587 Competitiveness Operational Programme grant of the Romanian Ministry of European Funds
- P_37_762, MySMIS 103587 Competitiveness Operational Programme grant of the Romanian Ministry of European Funds
- P_37_762, MySMIS 103587 Competitiveness Operational Programme grant of the Romanian Ministry of European Funds
- P_37_762, MySMIS 103587 Competitiveness Operational Programme grant of the Romanian Ministry of European Funds
- PNRR-III-C9-2022-I8, CF 85 / 15.11.2022 Romania's National Recovery and Resilience Plan grant of the Romanian Ministry of Investments and European Projects
- PNRR-III-C9-2022-I8, CF 85 / 15.11.2022 Romania's National Recovery and Resilience Plan grant of the Romanian Ministry of Investments and European Projects
- PNRR-III-C9-2022-I8, CF 85 / 15.11.2022 Romania's National Recovery and Resilience Plan grant of the Romanian Ministry of Investments and European Projects
- PNRR-III-C9-2022-I8, CF 85 / 15.11.2022 Romania's National Recovery and Resilience Plan grant of the Romanian Ministry of Investments and European Projects
- PNRR-III-C9-2022-I8, CF 85 / 15.11.2022 Romania's National Recovery and Resilience Plan grant of the Romanian Ministry of Investments and European Projects
- PNRR-III-C9-2022-I8, CF 85 / 15.11.2022 Romania's National Recovery and Resilience Plan grant of the Romanian Ministry of Investments and European Projects
- PNRR-III-C9-2022-I8, CF 85 / 15.11.2022 Romania's National Recovery and Resilience Plan grant of the Romanian Ministry of Investments and European Projects
- PNRR-III-C9-2022-I8, CF 85 / 15.11.2022 Romania's National Recovery and Resilience Plan grant of the Romanian Ministry of Investments and European Projects
- 2462/22 University of Medicine and Pharmacy "Iuliu Hațieganu", Cluj-Napoca, Romania
- University of Medicine and Pharmacy „Iuliu Hațieganu”, Cluj-Napoca, Romania
Collapse
Affiliation(s)
- Orsolya I Gaal
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur nr.6, Cluj, Napoca, 400349, Romania
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Megan Leask
- Department of Physiology, University of Otago, Dunedin, New Zealand
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Valentin Nica
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur nr.6, Cluj, Napoca, 400349, Romania
| | - Georgiana Cabău
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur nr.6, Cluj, Napoca, 400349, Romania
| | - Medeea Badii
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur nr.6, Cluj, Napoca, 400349, Romania
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ioana Hotea
- Department of Rheumatology, University of Medicine and Pharmay, Cluj-Napoca, Romania
| | - Dennis M de Graaf
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Zhenhua Zhang
- Centre for Individualised Infection Medicine (CiiM) & TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI), Hannover Medical School (MHH), Hannover, Germany
| | - Yang Li
- Centre for Individualised Infection Medicine (CiiM) & TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI), Hannover Medical School (MHH), Hannover, Germany
| | - Cristina Pamfil
- Department of Rheumatology, University of Medicine and Pharmay, Cluj-Napoca, Romania
| | - Simona Rednic
- Department of Rheumatology, University of Medicine and Pharmay, Cluj-Napoca, Romania
| | - Tony R Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Tania O Crișan
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur nr.6, Cluj, Napoca, 400349, Romania.
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Leo A B Joosten
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur nr.6, Cluj, Napoca, 400349, Romania
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
4
|
Major TJ, Takei R, Matsuo H, Leask MP, Sumpter NA, Topless RK, Shirai Y, Wang W, Cadzow MJ, Phipps-Green AJ, Li Z, Ji A, Merriman ME, Morice E, Kelley EE, Wei WH, McCormick SPA, Bixley MJ, Reynolds RJ, Saag KG, Fadason T, Golovina E, O'Sullivan JM, Stamp LK, Dalbeth N, Abhishek A, Doherty M, Roddy E, Jacobsson LTH, Kapetanovic MC, Melander O, Andrés M, Pérez-Ruiz F, Torres RJ, Radstake T, Jansen TL, Janssen M, Joosten LAB, Liu R, Gaal OI, Crişan TO, Rednic S, Kurreeman F, Huizinga TWJ, Toes R, Lioté F, Richette P, Bardin T, Ea HK, Pascart T, McCarthy GM, Helbert L, Stibůrková B, Tausche AK, Uhlig T, Vitart V, Boutin TS, Hayward C, Riches PL, Ralston SH, Campbell A, MacDonald TM, Nakayama A, Takada T, Nakatochi M, Shimizu S, Kawamura Y, Toyoda Y, Nakaoka H, Yamamoto K, Matsuo K, Shinomiya N, Ichida K, Lee C, Bradbury LA, Brown MA, Robinson PC, Buchanan RRC, Hill CL, Lester S, Smith MD, Rischmueller M, Choi HK, Stahl EA, Miner JN, Solomon DH, Cui J, Giacomini KM, Brackman DJ, Jorgenson EM, Liu H, Susztak K, Shringarpure S, So A, Okada Y, Li C, Shi Y, Merriman TR. A genome-wide association analysis reveals new pathogenic pathways in gout. Nat Genet 2024; 56:2392-2406. [PMID: 39406924 DOI: 10.1038/s41588-024-01921-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 08/21/2024] [Indexed: 10/18/2024]
Abstract
Gout is a chronic disease that is caused by an innate immune response to deposited monosodium urate crystals in the setting of hyperuricemia. Here, we provide insights into the molecular mechanism of the poorly understood inflammatory component of gout from a genome-wide association study (GWAS) of 2.6 million people, including 120,295 people with prevalent gout. We detected 377 loci and 410 genetically independent signals (149 previously unreported loci in urate and gout). An additional 65 loci with signals in urate (from a GWAS of 630,117 individuals) but not gout were identified. A prioritization scheme identified candidate genes in the inflammatory process of gout, including genes involved in epigenetic remodeling, cell osmolarity and regulation of NOD-like receptor protein 3 (NLRP3) inflammasome activity. Mendelian randomization analysis provided evidence for a causal role of clonal hematopoiesis of indeterminate potential in gout. Our study identifies candidate genes and molecular processes in the inflammatory pathogenesis of gout suitable for follow-up studies.
Collapse
Affiliation(s)
- Tanya J Major
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Riku Takei
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
- Department of Biomedical Information Management, National Defense Medical College Research Institute, National Defense Medical College, Saitama, Japan
| | - Megan P Leask
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicholas A Sumpter
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ruth K Topless
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Yuya Shirai
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Wei Wang
- Genomics R&D, 23andMe, Inc, Sunnyvale, CA, USA
| | - Murray J Cadzow
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | | | - Zhiqiang Li
- The Biomedical Sciences Institute and The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, Shandong, China
| | - Aichang Ji
- Shandong Provincial Key Laboratory of Metabolic Diseases, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- The Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong, China
| | - Marilyn E Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Emily Morice
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric E Kelley
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Wen-Hua Wei
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand
| | | | - Matthew J Bixley
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Richard J Reynolds
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth G Saag
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tayaza Fadason
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Evgenia Golovina
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Justin M O'Sullivan
- Liggins Institute, University of Auckland, Auckland, New Zealand
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Singapore
- Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Lisa K Stamp
- Department of Medicine, University of Otago, Christchurch, Christchurch, New Zealand
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Abhishek Abhishek
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Michael Doherty
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Edward Roddy
- School of Medicine, Keele University, Keele, Staffordshire, United Kingdom
- Haywood Academic Rheumatology Centre, Midlands Partnership University NHS Foundation Trust, Stoke-on-Trent, UK
| | - Lennart T H Jacobsson
- Department of Rheumatology and Inflammation Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Meliha C Kapetanovic
- Department of Clinical Sciences Lund, Section of Rheumatology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Mariano Andrés
- Rheumatology Department, Dr Balmis General University Hospital-ISABIAL, Alicante, Spain
- Department of Clinical Medicine, Miguel Hernandez University, Alicante, Spain
| | - Fernando Pérez-Ruiz
- Osakidetza, OSI-EE-Cruces, BIOBizkaia Health Research Institute and Medicine Department of Medicine and Nursery School, University of the Basque Country, Biskay, Spain
| | - Rosa J Torres
- Department of Biochemistry, Hospital La Paz Institute for Health Research (IdiPaz), Madrid, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Timothy Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center, Utrecht, The Netherlands
| | - Timothy L Jansen
- Department of Rheumatology, VieCuri Medical Centre, Venlo, The Netherlands
| | - Matthijs Janssen
- Department of Rheumatology, VieCuri Medical Centre, Venlo, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Institute of Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ruiqi Liu
- Department of Internal Medicine and Radboud Institute of Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Orsolya I Gaal
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Tania O Crişan
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Simona Rednic
- Department of Rheumatology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Cluj, Romania
| | - Fina Kurreeman
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom W J Huizinga
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - René Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frédéric Lioté
- Rheumatology Department, Feel'Gout, GH Paris Saint Joseph, Paris, France
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Pascal Richette
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Thomas Bardin
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Hang Korng Ea
- Rheumatology Department, INSERM U1132, BIOSCAR, University Paris Cité, Lariboisière Hospital, Paris, France
| | - Tristan Pascart
- Department of Rheumatology, Hopital Saint-Philibert, Lille Catholic University, Lille, France
| | - Geraldine M McCarthy
- Department of Rheumatology, Mater Misericordiae University Hospital and School of Medicine, University College, Dublin, Ireland
| | - Laura Helbert
- Department of Rheumatology, Mater Misericordiae University Hospital and School of Medicine, University College, Dublin, Ireland
| | - Blanka Stibůrková
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
- Institute of Rheumatology, Prague, Czech Republic
| | - Anne-K Tausche
- Department of Rheumatology, University Clinic 'Carl Gustav Carus' at the Technical University, Dresden, Germany
| | - Till Uhlig
- Center for Treatment of Rheumatic and Musculoskeletal Diseases, Diakonhjemmet Hospital, Oslo, Norway
| | - Véronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Thibaud S Boutin
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Philip L Riches
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Stuart H Ralston
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Thomas M MacDonald
- MEMO Research, Division of Molecular and Clinical Medicine, University of Dundee Medical School, Ninewells Hospital, Dundee, United Kingdom
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Yusuke Kawamura
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
- Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Yu Toyoda
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Hirofumi Nakaoka
- Department of Cancer Genome Research, Sasaki Institute, Sasaki Foundation, Tokyo, Japan
| | - Ken Yamamoto
- Department of Medical Biochemistry, Kurume University School of Medicine, Fukuoka, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology & Prevention, Aichi Cancer Center, Aichi, Japan
- Division of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Aichi, Japan
- The Japan Multi-Institutional Collaborative Cohort (J-MICC) Study, Tokyo, Japan
| | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama, Japan
| | - Kimiyoshi Ichida
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Chaeyoung Lee
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, South Korea
| | - Linda A Bradbury
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Matthew A Brown
- Institute of Health and Biomedical Innovation, Translational Research Institute, Queensland University of Technology, Brisbane, Australia
| | - Philip C Robinson
- School of Clinical Medicine, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | | | - Catherine L Hill
- Rheumatology Department, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Susan Lester
- Rheumatology Department, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | | | - Maureen Rischmueller
- Rheumatology Department, The Queen Elizabeth Hospital, Woodville South, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Hyon K Choi
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eli A Stahl
- Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeff N Miner
- Viscient Biosciences, 5752 Oberlin Dr., Suite 111, San Diego, CA, 92121, USA
| | - Daniel H Solomon
- Division of Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jing Cui
- Division of Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences and Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Deanna J Brackman
- Department of Bioengineering and Therapeutic Sciences and Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Eric M Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Hongbo Liu
- Penn / The Children's Hospital of Pennsylvania Kidney Innovation Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
| | - Katalin Susztak
- Penn / The Children's Hospital of Pennsylvania Kidney Innovation Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
- Renal Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19101, USA
| | | | - Alexander So
- Service of Rheumatology, Center Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- University of Lausanne, Lausanne, Switzerland
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Changgui Li
- Shandong Provincial Key Laboratory of Metabolic Diseases, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- The Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong, China
| | - Yongyong Shi
- Affiliated Hospital of Qingdao University and Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai, China
| | - Tony R Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA.
- The Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong, China.
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
5
|
Merlo Pich LM, Ziogas A, Netea MG. Genetic and epigenetic dysregulation of innate immune mechanisms in autoinflammatory diseases. FEBS J 2024; 291:4414-4432. [PMID: 38468589 DOI: 10.1111/febs.17116] [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: 08/10/2023] [Revised: 01/17/2024] [Accepted: 03/01/2024] [Indexed: 03/13/2024]
Abstract
Dysregulation and hyperactivation of innate immune responses can lead to the onset of systemic autoinflammatory diseases. Monogenic autoinflammatory diseases are caused by inborn genetic errors and based on molecular mechanisms at play, can be divided into inflammasomopathies, interferonopathies, relopathies, protein misfolding, and endogenous antagonist deficiencies. On the other hand, more common autoinflammatory diseases are multifactorial, with both genetic and non-genetic factors playing an important role. During the last decade, long-term memory characteristics of innate immune responses have been described (also called trained immunity) that in physiological conditions provide enhanced host protection from pathogenic re-infection. However, if dysregulated, induction of trained immunity can become maladaptive, perpetuating chronic inflammatory activation. Here, we describe the mechanisms of genetic and epigenetic dysregulation of the innate immune system and maladaptive trained immunity that leads to the onset and perpetuation of the most common and recently described systemic autoinflammatory diseases.
Collapse
Affiliation(s)
- Laura M Merlo Pich
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Athanasios Ziogas
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Germany
| |
Collapse
|
6
|
Liu Z, Crișan TO, Qi C, Gupta MK, Liu X, Moorlag SJ, Koeken VA, de Bree LCJ, Mourits VP, Gao X, Baccarelli A, Schwartz J, Pessler F, Guzmán CA, Li Y, Netea MG, Joosten LA, Xu CJ. Sex-specific epigenetic signatures of circulating urate and its increase after BCG vaccination. RESEARCH SQUARE 2024:rs.3.rs-4498597. [PMID: 39108482 PMCID: PMC11302698 DOI: 10.21203/rs.3.rs-4498597/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Background Urate concentration and the physiological regulation of urate homeostasis exhibit clear sex differences. DNA methylation has been shown to explain a substantial proportion of serum urate variance, mediate the genetic effect on urate concentration, and co-regulate with cardiometabolic traits. However, whether urate concentration is associated with DNA methylation in a sex-dependent manner is unknown. Additionally, it is worth investigating if urate changes after perturbations, such as vaccination, are associated with DNA methylation in a sex-specific manner. Methods We investigated the association between DNA methylation and serum urate concentrations in a Dutch cohort of 325 healthy individuals. Urate concentration and DNA methylation were measured before and after Bacillus Calmette-Guérin (BCG) vaccination, used as a perturbation associated with increased gout flares. The association analysis included united, interaction, and sex-stratified analysis. Validation of the identified CpG sites was conducted using three independent cohorts. Results 215 CpG sites were associated with serum urate in males, while 5 CpG sites were associated with serum urate in females, indicating sex-specific associations. Circulating urate concentrations significantly increased after BCG vaccination, and baseline DNA methylation was associated with differences in urate concentration before and after vaccination in a sex-specific manner. The CpG sites associated with urate concentration in males were enriched in neuro-protection pathways, whereas in females, the urate change-associated CpG sites were related to lipid and glucose metabolism. Conclusion Our study enhances the understanding of how epigenetic factors contribute to regulating serum urate levels in a sex-specific manner. These insights have significant implications for the diagnosis, prevention, and treatment of various urate-related diseases and highlight the importance of personalized and sex-specific approaches in medicine.
Collapse
Affiliation(s)
- Zhaoli Liu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
| | - Tania O. Crișan
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
- Department of Medical Genetics, „Iuliu Hațieganu” University of Medicine and Pharmacy. Cluj-Napoca, Romania
| | - Cancan Qi
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
| | - Manoj Kumar Gupta
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
| | - Xuan Liu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
| | - Simone J.C.F.M. Moorlag
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
| | - Valerie A.C.M. Koeken
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
- Research Centre Innovations in Care, Rotterdam University of Applied Sciences, Rotterdam, the Netherlands
| | - L. Charlotte J. de Bree
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
| | - Vera P. Mourits
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
| | - Xu Gao
- Department of Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Andrea Baccarelli
- Department of Environmental Health, Mailman School of Public Health, Columbia University, New York, NY, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Joel Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Frank Pessler
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- Research Group Biomarkers for Infectious Diseases, TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
| | - Carlos A. Guzmán
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- Department Vaccinology and Applied Microbiology, Helmholtz-Centre for Infection Research (HZI), Braunschweig, Germany
| | - Yang Li
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
- Cluster of Excellence RESIST (EXC 2155), Hanover Medical School, Hannover, Germany
- Lower Saxony center for artificial intelligence and causal methods in medicine (CAIMed). Hannover, Germany
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES). University of Bonn. Bonn, Germany
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center. Nijmegen, the Netherlands
- Department of Medical Genetics, „Iuliu Hațieganu” University of Medicine and Pharmacy. Cluj-Napoca, Romania
| | - Cheng-Jian Xu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
- TWINCORE, a joint venture between the Helmholtz-Centre for Infection Research (HZI) and Hannover Medical School (MHH). Hannover, Germany
| |
Collapse
|
7
|
Xu W, Yuan Y, Shu Z, Guo T, Liu B, Xiao J, Li L, Yin Y, Zhang X. Streptococcus pneumoniae endopeptidase O induces trained immunity and confers protection against various pathogenic infections. Clin Immunol 2024; 263:110226. [PMID: 38663493 DOI: 10.1016/j.clim.2024.110226] [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: 01/06/2024] [Revised: 03/30/2024] [Accepted: 04/16/2024] [Indexed: 05/12/2024]
Abstract
Antibiotic resistance and the surge of infectious diseases during the pandemic present significant threats to human health. Trained immunity emerges as a promising and innovative approach to address these infections. Synthetic or natural fungal, parasitic and viral components have been reported to induce trained immunity. However, it is not clear whether bacterial virulence proteins can induce protective trained immunity. Our research demonstrates Streptococcus pneumoniae virulence protein PepO, is a highly potent trained immunity inducer for combating broad-spectrum infection. Our findings showcase that rPepO training confers robust protection to mice against various pathogenic infections by enhancing macrophage functionality. rPepO effectively re-programs macrophages, re-configures their epigenetic modifications and bolsters their immunological responses, which is independent of T or B lymphocytes. In vivo and in vitro experiments confirm that trained macrophage-secreted complement C3 activates peritoneal B lymphocyte and enhances its bactericidal capacity. In addition, we provide the first evidence that granulocyte colony-stimulating factor (G-CSF) derived from trained macrophages plays a pivotal role in shaping central-trained immunity. In summation, our research demonstrates the capability of rPepO to induce both peripheral and central trained immunity in mice, underscoring its potential application in broad-spectrum anti-infection therapy. Our research provides a new molecule and some new target options for infectious disease prevention.
Collapse
Affiliation(s)
- Wenlong Xu
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Yuan Yuan
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Zhaoche Shu
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Ting Guo
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Bichen Liu
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Jiangming Xiao
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Lian Li
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Yibin Yin
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing 400016, China
| | - Xuemei Zhang
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
8
|
Badii M, Klück V, Gaal O, Cabău G, Hotea I, Nica V, Mirea AM, Bojan A, Zdrenghea M, Novakovic B, Merriman TR, Liu Z, Li Y, Xu CJ, Pamfil C, Rednic S, Popp RA, Crişan TO, Joosten LAB. Regulation of SOCS3-STAT3 in urate-induced cytokine production in human myeloid cells. Joint Bone Spine 2024; 91:105698. [PMID: 38309518 DOI: 10.1016/j.jbspin.2024.105698] [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: 09/28/2023] [Revised: 01/10/2024] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
OBJECTIVE Hyperuricaemia is necessary for gout. High urate concentrations have been linked to inflammation in mononuclear cells. Here, we explore the role of the suppressor of cytokine signaling 3 (SOCS3) in urate-induced inflammation. METHODS Peripheral blood mononuclear cells (PBMCs) from gout patients, hyperuricemic and normouricemic individuals were cultured for 24h with varying concentrations of soluble urate, followed by 24h restimulation with lipopolysaccharides (LPS)±monosodium urate (MSU) crystals. Transcriptomic profiling was performed using RNA-Sequencing. DNA methylation was assessed using Illumina Infinium® MethylationEPIC BeadChip system (EPIC array). Phosphorylation of signal transducer and activator of transcription 3 (STAT3) was determined by flow cytometry. Cytokine responses were also assessed in PBMCs from patients with JAK2 V617F tyrosine kinase mutation. RESULTS PBMCs pre-treated with urate produced more interleukin-1beta (IL-1β) and interleukin-6 (IL-6) and less interleukin-1 receptor anatagonist (IL-1Ra) after LPS simulation. In vitro, urate treatment enhanced SOCS3 expression in control monocytes but no DNA methylation changes were observed at the SOCS3 gene. A dose-dependent reduction in phosphorylated STAT3 concomitant with a decrease in IL-1Ra was observed with increasing concentrations of urate. PBMCs with constitutively activated STAT3 (JAK2 V617F mutation) could not be primed by urate. CONCLUSION In vitro, urate exposure increased SOCS3 expression, while urate priming, and subsequent stimulation resulted in decreased STAT3 phosphorylation and IL-1Ra production. There was no evidence that DNA methylation constitutes a regulatory mechanism of SOCS3. Elevated SOCS3 and reduced pSTAT3 could play a role in urate-induced hyperinflammation since urate priming had no effect in PBMCs from patients with constitutively activated STAT3.
Collapse
Affiliation(s)
- Medeea Badii
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, 6525GA Nijmegen, The Netherlands
| | - Viola Klück
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, 6525GA Nijmegen, The Netherlands
| | - Orsolya Gaal
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, 6525GA Nijmegen, The Netherlands
| | - Georgiana Cabău
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ioana Hotea
- Department of Rheumatology, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Valentin Nica
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Andreea M Mirea
- Department of Genetics, Clinical Emergency Hospital for Children, 400535 Cluj-Napoca, Romania
| | - Anca Bojan
- Department of Haematology, The Oncology Institute, "Prof. Dr. Ion Chiricuță", 400015 Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department of Haematology, The Oncology Institute, "Prof. Dr. Ion Chiricuță", 400015 Cluj-Napoca, Romania
| | - Boris Novakovic
- Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Tony R Merriman
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, 35294, United States; Department of Biochemistry, University of Otago, 9016 Dunedin, New Zealand
| | - Zhaoli Liu
- Centre for Individualized Infection Medicine (CiiM), a joint venture between Hannover Medical School and Helmholtz Centre for Infection Research, 30625 Hannover, Germany
| | - Yang Li
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, 6525GA Nijmegen, The Netherlands; Centre for Individualized Infection Medicine (CiiM), a joint venture between Hannover Medical School and Helmholtz Centre for Infection Research, 30625 Hannover, Germany
| | - Cheng-Jian Xu
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, 6525GA Nijmegen, The Netherlands; Centre for Individualized Infection Medicine (CiiM), a joint venture between Hannover Medical School and Helmholtz Centre for Infection Research, 30625 Hannover, Germany
| | - Cristina Pamfil
- Department of Rheumatology, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Simona Rednic
- Department of Rheumatology, Iuliu Hațieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
| | - Radu A Popp
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Tania O Crişan
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, 6525GA Nijmegen, The Netherlands.
| | - Leo A B Joosten
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; Department of Internal Medicine and Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, 6525GA Nijmegen, The Netherlands
| |
Collapse
|
9
|
Gaal OI, Liu R, Marginean D, Badii M, Cabău G, Hotea I, Nica V, Colcear D, Pamfil C, Merriman TR, Rednic S, Popp RA, Crișan TO, Joosten LAB. GWAS-identified hyperuricemia-associated IGF1R variant rs6598541 has a limited role in urate mediated inflammation in human mononuclear cells. Sci Rep 2024; 14:3565. [PMID: 38347000 PMCID: PMC10861580 DOI: 10.1038/s41598-024-53209-7] [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: 06/29/2023] [Accepted: 01/29/2024] [Indexed: 02/15/2024] Open
Abstract
Gout is a common autoinflammatory joint diseases characterized by deposition of monosodium urate (MSU) crystals which trigger an innate immune response mediated by inflammatory cytokines. IGF1R is one of the loci associated with both urate levels and gout susceptibility in GWAS to date, and IGF-1-IGF-1R signaling is implicated in urate control. We investigate the role of IGF-1/IGF1R signaling in the context of gouty inflammation. Also, we test the gout and urate-associated IGF1R rs6598541 polymorphism for association with the inflammatory capacity of mononuclear cells. For this, freshly isolated human peripheral blood mononuclear cells (PBMCs) were exposed to recombinant IGF-1 or anti-IGF1R neutralizing antibody in the presence or absence of solubilized urate, stimulated with LPS/MSU crystals. Also, the association of rs6598541 with IGF1R and protein expression and with ex vivo cytokine production levels after stimulation with gout specific stimuli was tested. Urate exposure was not associated with IGF1R expression in vitro or in vivo. Modulation of IGF1R did not alter urate-induced inflammation. Developing urate-induced trained immunity in vitro was not influenced in cells challenged with IGF-1 recombinant protein. Moreover, the IGF1R rs6598541 SNP was not associated with cytokine production. Our results indicate that urate-induced inflammatory priming is not regulated by IGF-1/IGF1R signaling in vitro. IGF1R rs6598541 status was not asociated with IGF1R expression or cytokine production in primary human PBMCs. This study suggests that the role of IGF1R in gout is tissue-specific and may be more relevant in the control of urate levels rather than in inflammatory signaling in gout.
Collapse
Affiliation(s)
- Orsolya I Gaal
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur Nr.6, 400349, Cluj-Napoca, Romania
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ruiqi Liu
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dragoș Marginean
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur Nr.6, 400349, Cluj-Napoca, Romania
| | - Medeea Badii
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur Nr.6, 400349, Cluj-Napoca, Romania
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Georgiana Cabău
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur Nr.6, 400349, Cluj-Napoca, Romania
| | - Ioana Hotea
- Department of Rheumatology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Valentin Nica
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur Nr.6, 400349, Cluj-Napoca, Romania
| | - Doina Colcear
- Clinical Infectious Disease Hospital, Cluj-Napoca, Romania
| | - Cristina Pamfil
- Department of Rheumatology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Tony R Merriman
- Department of Microbiology, University of Otago, Dunedin, New Zealand
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Simona Rednic
- Department of Rheumatology, Iuliu Hațieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Radu A Popp
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur Nr.6, 400349, Cluj-Napoca, Romania
| | - Tania O Crișan
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur Nr.6, 400349, Cluj-Napoca, Romania.
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Leo A B Joosten
- Department of Medical Genetics, Iuliu Hațieganu University of Medicine and Pharmacy, Str. Pasteur Nr.6, 400349, Cluj-Napoca, Romania
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
10
|
Riksen NP, Bekkering S, Mulder WJM, Netea MG. Trained immunity in atherosclerotic cardiovascular disease. Nat Rev Cardiol 2023; 20:799-811. [PMID: 37322182 DOI: 10.1038/s41569-023-00894-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2023] [Indexed: 06/17/2023]
Abstract
Trained immunity, also known as innate immune memory, is a persistent hyper-responsive functional state of innate immune cells. Accumulating evidence implicates trained immunity as an underlying mechanism of chronic inflammation in atherosclerotic cardiovascular disease. In this context, trained immunity is induced by endogenous atherosclerosis-promoting factors, such as modified lipoproteins or hyperglycaemia, causing broad metabolic and epigenetic reprogramming of the myeloid cell compartment. In addition to traditional cardiovascular risk factors, lifestyle factors, including unhealthy diets, sedentary lifestyle, sleep deprivation and psychosocial stress, as well as inflammatory comorbidities, have been shown to activate trained immunity-like mechanisms in bone marrow haematopoietic stem cells. In this Review, we discuss the molecular and cellular mechanisms of trained immunity, its systemic regulation through haematopoietic progenitor cells in the bone marrow, and the activation of these mechanisms by cardiovascular disease risk factors. We also highlight other trained immunity features that are relevant for atherosclerotic cardiovascular disease, including the diverse cell types that show memory characteristics and transgenerational inheritance of trained immunity traits. Finally, we propose potential strategies for the therapeutic modulation of trained immunity to manage atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Niels P Riksen
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Siroon Bekkering
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Willem J M Mulder
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department for Genomics and Immunoregulation, University of Bonn, Bonn, Germany
| |
Collapse
|
11
|
Li D, Yuan S, Deng Y, Wang X, Wu S, Chen X, Li Y, Ouyang J, Lin D, Quan H, Fu X, Li C, Mao W. The dysregulation of immune cells induced by uric acid: mechanisms of inflammation associated with hyperuricemia and its complications. Front Immunol 2023; 14:1282890. [PMID: 38053999 PMCID: PMC10694226 DOI: 10.3389/fimmu.2023.1282890] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/26/2023] [Indexed: 12/07/2023] Open
Abstract
Changes in lifestyle induce an increase in patients with hyperuricemia (HUA), leading to gout, gouty arthritis, renal damage, and cardiovascular injury. There is a strong inflammatory response in the process of HUA, while dysregulation of immune cells, including monocytes, macrophages, and T cells, plays a crucial role in the inflammatory response. Recent studies have indicated that urate has a direct impact on immune cell populations, changes in cytokine expression, modifications in chemotaxis and differentiation, and the provocation of immune cells by intrinsic cells to cause the aforementioned conditions. Here we conducted a detailed review of the relationship among uric acid, immune response, and inflammatory status in hyperuricemia and its complications, providing new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Delun Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Siyu Yuan
- Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiyao Deng
- Department of Nephrology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Xiaowan Wang
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Shouhai Wu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Xuesheng Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Yimeng Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Jianting Ouyang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Danyao Lin
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Haohao Quan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Xinwen Fu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Chuang Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| | - Wei Mao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Nephrology Institute of Guangdong Provincial Academy of Chinese Medical Sciences (NIGH-CM), Guangzhou, China
| |
Collapse
|
12
|
Cabău G, Gaal O, Badii M, Nica V, Mirea AM, Hotea I, HINT-consortium, Pamfil C, Popp RA, Netea MG, Rednic S, Crișan TO, Joosten LA. Hyperuricemia remodels the serum proteome toward a higher inflammatory state. iScience 2023; 26:107909. [PMID: 37810213 PMCID: PMC10550725 DOI: 10.1016/j.isci.2023.107909] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/01/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023] Open
Abstract
Gout is an autoinflammatory disease triggered by a complex innate immune response to MSU crystals and inflammatory triggers. While hyperuricemia is an obligatory risk factor for the development of gout, the majority of individuals with hyperuricemia never develop gout but have an increased risk of developing cardiometabolic disorders. Current management of gout aims at MSU crystal dissolution by lowering serum urate. We apply a targeted proteomic analysis, using Olink inflammation panel, to a large group of individuals with gout, asymptomatic hyperuricemia, and normouricemic controls, and we show a urate-driven inflammatory signature. We add in vivo evidence of persistent immune activation linked to urate exposure and describe immune pathways involved in the pathogenesis of gout. Our results support a pro-inflammatory effect of asymptomatic hyperuricemia and pave the way for new research into targetable mechanisms in gout and cardiometabolic complications of asymptomatic hyperuricemia.
Collapse
Affiliation(s)
- Georgiana Cabău
- Department of Medical Genetics, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Orsolya Gaal
- Department of Medical Genetics, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Internal Medicine, Radboudumc, Nijmegen, the Netherlands
| | - Medeea Badii
- Department of Medical Genetics, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Internal Medicine, Radboudumc, Nijmegen, the Netherlands
| | - Valentin Nica
- Department of Medical Genetics, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Ioana Hotea
- Department of Rheumatology, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - HINT-consortium
- Department of Medical Genetics, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Internal Medicine, Radboudumc, Nijmegen, the Netherlands
- Department of Rheumatology, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Cristina Pamfil
- Department of Rheumatology, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Radu A. Popp
- Department of Medical Genetics, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G. Netea
- Department of Internal Medicine, Radboudumc, Nijmegen, the Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Simona Rednic
- Department of Rheumatology, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Tania O. Crișan
- Department of Medical Genetics, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Internal Medicine, Radboudumc, Nijmegen, the Netherlands
| | - Leo A.B. Joosten
- Department of Medical Genetics, “Iuliu Haţieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Internal Medicine, Radboudumc, Nijmegen, the Netherlands
| |
Collapse
|
13
|
de Lima JD, de Paula AGP, Yuasa BS, de Souza Smanioto CC, da Cruz Silva MC, Dos Santos PI, Prado KB, Winter Boldt AB, Braga TT. Genetic and Epigenetic Regulation of the Innate Immune Response to Gout. Immunol Invest 2023; 52:364-397. [PMID: 36745138 DOI: 10.1080/08820139.2023.2168554] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gout is a disease caused by uric acid (UA) accumulation in the joints, causing inflammation. Two UA forms - monosodium urate (MSU) and soluble uric acid (sUA) have been shown to interact physically with inflammasomes, especially with the nod-like receptor (NLR) family pyrin domain containing 3 (NLRP3), albeit the role of the immune response to UA is poorly understood, given that asymptomatic hyperuricemia does also exist. Macrophage phagocytosis of UA activate NLRP3, lead to cytokines release, and ultimately, lead to chemoattract neutrophils and lymphocytes to the gout flare joint spot. Genetic variants of inflammasome genes and of genes encoding their molecular partners may influence hyperuricemia and gout susceptibility, while also influencing other comorbidities such as metabolic syndrome and cardiovascular diseases. In this review, we summarize the inflammatory responses in acute and chronic gout, specifically focusing on innate immune cell mechanisms and genetic and epigenetic characteristics of participating molecules. Unprecedently, a novel UA binding protein - the neuronal apoptosis inhibitor protein (NAIP) - is suggested as responsible for the asymptomatic hyperuricemia paradox.Abbreviation: β2-integrins: leukocyte-specific adhesion molecules; ABCG2: ATP-binding cassete family/breast cancer-resistant protein; ACR: American college of rheumatology; AIM2: absent in melanoma 2, type of pattern recognition receptor; ALPK1: alpha-protein kinase 1; ANGPTL2: angiopoietin-like protein 2; ASC: apoptosis-associated speck-like protein; BIR: baculovirus inhibitor of apoptosis protein repeat; BIRC1: baculovirus IAP repeat-containing protein 1; BIRC2: baculoviral IAP repeat-containing protein 2; C5a: complement anaphylatoxin; cAMP: cyclic adenosine monophosphate; CARD: caspase activation and recruitment domains; CARD8: caspase recruitment domain-containing protein 8; CASP1: caspase 1; CCL3: chemokine (C-C motif) ligand 3; CD14: cluster of differentiation 14; CD44: cluster of differentiation 44; Cg05102552: DNA-methylation site, usually cytosine followed by guanine nucleotides; contains arbitrary identification code; CIDEC: cell death-inducing DNA fragmentation factor-like effector family; CKD: chronic kidney disease; CNV: copy number variation; CPT1A: carnitine palmitoyl transferase - type 1a; CXCL1: chemokine (CXC motif) ligand 1; DAMPs: damage associated molecular patterns; DC: dendritic cells; DNMT(1): maintenance DNA methyltransferase; eQTL: expression quantitative trait loci; ERK1: extracellular signal-regulated kinase 1; ERK2: extracellular signal-regulated kinase 2; EULAR: European league against rheumatism; GMCSF: granulocyte-macrophage colony-stimulating factor; GWAS: global wide association studies; H3K27me3: tri-methylation at the 27th lysine residue of the histone h3 protein; H3K4me1: mono-methylation at the 4th lysine residue of the histone h3 protein; H3K4me3: tri-methylation at the 4th lysine residue of the histone h3 protein; HOTAIR: human gene located between hoxc11 and hoxc12 on chromosome 12; IκBα: cytoplasmatic protein/Nf-κb transcription inhibitor; IAP: inhibitory apoptosis protein; IFNγ: interferon gamma; IL-1β: interleukin 1 beta; IL-12: interleukin 12; IL-17: interleukin 17; IL18: interleukin 18; IL1R1: interleukin-1 receptor; IL-1Ra: interleukin-1 receptor antagonist; IL-22: interleukin 22; IL-23: interleukin 23; IL23R: interleukin 23 receptor; IL-33: interleukin 33; IL-6: interleukin 6; IMP: inosine monophosphate; INSIG1: insulin-induced gene 1; JNK1: c-jun n-terminal kinase 1; lncRNA: long non-coding ribonucleic acid; LRR: leucine-rich repeats; miR: mature non-coding microRNAs measuring from 20 to 24 nucleotides, animal origin; miR-1: miR followed by arbitrary identification code; miR-145: miR followed by arbitrary identification code; miR-146a: miR followed by arbitrary identification code, "a" stands for mir family; "a" family presents similar mir sequence to "b" family, but different precursors; miR-20b: miR followed by arbitrary identification code; "b" stands for mir family; "b" family presents similar mir sequence to "a" family, but different precursors; miR-221: miR - followed by arbitrary identification code; miR-221-5p: miR followed by arbitrary identification code; "5p" indicates different mature miRNAs generated from the 5' arm of the pre-miRNA hairpin; miR-223: miR followed by arbitrary identification code; miR-223-3p: mir followed by arbitrary identification code; "3p" indicates different mature miRNAs generated from the 3' arm of the pre-miRNA hairpin; miR-22-3p: miR followed by arbitrary identification code, "3p" indicates different mature miRNAs generated from the 3' arm of the pre-miRNA hairpin; MLKL: mixed lineage kinase domain-like pseudo kinase; MM2P: inductor of m2-macrophage polarization; MSU: monosodium urate; mTOR: mammalian target of rapamycin; MyD88: myeloid differentiation primary response 88; n-3-PUFAs: n-3-polyunsaturated fatty-acids; NACHT: acronym for NAIP (neuronal apoptosis inhibitor protein), C2TA (MHC class 2 transcription activator), HET-E (incompatibility locus protein from podospora anserina) and TP1 (telomerase-associated protein); NAIP: neuronal apoptosis inhibitory protein (human); Naip1: neuronal apoptosis inhibitory protein type 1 (murine); Naip5: neuronal apoptosis inhibitory protein type 5 (murine); Naip6: neuronal apoptosis inhibitory protein type 6 (murine); NBD: nucleotide-binding domain; Nek7: smallest NIMA-related kinase; NET: neutrophil extracellular traps; Nf-κB: nuclear factor kappa-light-chain-enhancer of activated b cells; NFIL3: nuclear-factor, interleukin 3 regulated protein; NIIMA: network of immunity in infection, malignancy, and autoimmunity; NLR: nod-like receptor; NLRA: nod-like receptor NLRA containing acidic domain; NLRB: nod-like receptor NLRA containing BIR domain; NLRC: nod-like receptor NLRA containing CARD domain; NLRC4: nod-like receptor family CARD domain containing 4; NLRP: nod-like receptor NLRA containing PYD domain; NLRP1: nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 1; NLRP12: nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 12; NLRP3: nod-like receptor family pyrin domain containing 3; NOD2: nucleotide-binding oligomerization domain; NRBP1: nuclear receptor-binding protein; Nrf2: nuclear factor erythroid 2-related factor 2; OR: odds ratio; P2X: group of membrane ion channels activated by the binding of extracellular; P2X7: p2x purinoceptor 7 gene; p38: member of the mitogen-activated protein kinase family; PAMPs: pathogen associated molecular patters; PBMC: peripheral blood mononuclear cells; PGGT1B: geranylgeranyl transferase type-1 subunit beta; PHGDH: phosphoglycerate dehydrogenase; PI3-K: phospho-inositol; PPARγ: peroxisome proliferator-activated receptor gamma; PPARGC1B: peroxisome proliferative activated receptor, gamma, coactivator 1 beta; PR3: proteinase 3 antigen; Pro-CASP1: inactive precursor of caspase 1; Pro-IL1β: inactive precursor of interleukin 1 beta; PRR: pattern recognition receptors; PYD: pyrin domain; RAPTOR: regulatory associated protein of mTOR complex 1; RAS: renin-angiotensin system; REDD1: regulated in DNA damage and development 1; ROS: reactive oxygen species; rs000*G: single nuclear polymorphism, "*G" is related to snp where replaced nucleotide is guanine, usually preceded by an id number; SLC2A9: solute carrier family 2, member 9; SLC7A11: solute carrier family 7, member 11; SMA: smooth muscular atrophy; Smac: second mitochondrial-derived activator of caspases; SNP: single nuclear polymorphism; Sp3: specificity protein 3; ST2: serum stimulation-2; STK11: serine/threonine kinase 11; sUA: soluble uric acid; Syk: spleen tyrosine kinase; TAK1: transforming growth factor beta activated kinase; Th1: type 1 helper T cells; Th17: type 17 helper T cells; Th2: type 2 helper T cells; Th22: type 22 helper T cells; TLR: tool-like receptor; TLR2: toll-like receptor 2; TLR4: toll-like receptor 4; TNFα: tumor necrosis factor alpha; TNFR1: tumor necrosis factor receptor 1; TNFR2: tumor necrosis factor receptor 2; UA: uric acid; UBAP1: ubiquitin associated protein; ULT: urate-lowering therapy; URAT1: urate transporter 1; VDAC1: voltage-dependent anion-selective channel 1.
Collapse
Affiliation(s)
- Jordana Dinorá de Lima
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | | | - Bruna Sadae Yuasa
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | | | - Maria Clara da Cruz Silva
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | | | - Karin Braun Prado
- Genetics Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | - Angelica Beate Winter Boldt
- Program of Internal Medicine, Universidade Federal do Parana (UFPR), Curitiba, Brazil
- Genetics Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
| | - Tárcio Teodoro Braga
- Microbiology, Parasitology and Pathology Program, Universidade Federal do Parana (UFPR), Curitiba, Brazil
- Biosciences and Biotechnology Program, Instituto Carlos Chagas (ICC), Fiocruz-Parana, Brazil
| |
Collapse
|
14
|
Devi S, Indramohan M, Jäger E, Carriere J, Chu LH, de Almeida L, Greaves DR, Stehlik C, Dorfleutner A. CARD-only proteins regulate in vivo inflammasome responses and ameliorate gout. Cell Rep 2023; 42:112265. [PMID: 36930645 PMCID: PMC10151391 DOI: 10.1016/j.celrep.2023.112265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 01/10/2023] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
Inflammatory responses are crucial for controlling infections and initiating tissue repair. However, excessive and uncontrolled inflammation causes inflammatory disease. Processing and release of the pro-inflammatory cytokines interleukin-1β (IL-1β) and IL-18 depend on caspase-1 activation within inflammasomes. Assembly of inflammasomes is initiated upon activation of cytosolic pattern recognition receptors (PRRs), followed by sequential polymerization of pyrin domain (PYD)-containing and caspase recruitment domain (CARD)-containing proteins mediated by homotypic PYD and CARD interactions. Small PYD- or CARD-only proteins (POPs and COPs, respectively) evolved in higher primates to target these crucial interactions to limit inflammation. Here, we show the ability of COPs to regulate inflammasome activation by modulating homotypic CARD-CARD interactions in vitro and in vivo. CARD16, CARD17, and CARD18 displace crucial CARD interactions between caspase-1 proteins through competitive binding and ameliorate uric acid crystal-mediated NLRP3 inflammasome activation and inflammatory disease. COPs therefore represent an important family of inflammasome regulators and ameliorate inflammatory disease.
Collapse
Affiliation(s)
- Savita Devi
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mohanalaxmi Indramohan
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Elisabeth Jäger
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jessica Carriere
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Lan H Chu
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Driskill Graduate Program in Life Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lucia de Almeida
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - David R Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Christian Stehlik
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; The Kao Autoimmunity Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - Andrea Dorfleutner
- Department of Academic Pathology, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA; The Kao Autoimmunity Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
15
|
Klück V, Cabău G, Mies L, Bukkems F, van Emst L, Bakker R, van Caam A, Crişan TO, Joosten LAB. TGF-β is elevated in hyperuricemic individuals and mediates urate-induced hyperinflammatory phenotype in human mononuclear cells. Arthritis Res Ther 2023; 25:30. [PMID: 36850003 PMCID: PMC9969669 DOI: 10.1186/s13075-023-03001-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 01/29/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND Soluble urate leads to a pro-inflammatory phenotype in human monocytes characterized by increased production of IL-1β and downregulation of IL-1 receptor antagonist, the mechanism of which remains to be fully elucidated. Previous transcriptomic data identified differential expression of genes in the transforming growth factor (TGF)-β pathway in monocytes exposed to urate in vitro. In this study, we explore the role of TGF-β in urate-induced hyperinflammation in peripheral blood mononuclear cells (PBMCs). METHODS TGF-β mRNA in unstimulated PBMCs and protein levels in plasma were measured in individuals with normouricemia, hyperuricemia and gout. For in vitro validation, PBMCs of healthy volunteers were isolated and treated with a dose ranging concentration of urate for assessment of mRNA and pSMAD2. Urate and TGF-β priming experiments were performed with three inhibitors of TGF-β signalling: SB-505124, 5Z-7-oxozeaenol and a blocking antibody against TGF-β receptor II. RESULTS TGF-β mRNA levels were elevated in gout patients compared to healthy controls. TGF-β-LAP levels in serum were significantly higher in individuals with hyperuricemia compared to controls. In both cases, TGF-β correlated positively to serum urate levels. In vitro, urate exposure of PBMCs did not directly induce TGF-β but did enhance SMAD2 phosphorylation. The urate-induced pro-inflammatory phenotype of monocytes was partly reversed by blocking TGF-β. CONCLUSIONS TGF-β is elevated in individuals with hyperuricemia and correlated to serum urate concentrations. In addition, the urate-induced pro-inflammatory phenotype in human monocytes is mediated by TGF-β signalling. Future studies are warranted to explore the intracellular pathways involved and to assess the clinical significance of urate-TGF-β relation.
Collapse
Affiliation(s)
- Viola Klück
- Department of Internal Medicine, Radboud UMC, Nijmegen, The Netherlands.,Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Georgiana Cabău
- Department of Medical Genetics, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Linda Mies
- Department of Internal Medicine, Radboud UMC, Nijmegen, The Netherlands
| | - Femke Bukkems
- Departement of Rheumatology, Radboud UMC, Nijmegen, The Netherlands
| | - Liesbeth van Emst
- Department of Internal Medicine, Radboud UMC, Nijmegen, The Netherlands
| | - René Bakker
- Departement of Rheumatology, Radboud UMC, Nijmegen, The Netherlands
| | - Arjan van Caam
- Departement of Rheumatology, Radboud UMC, Nijmegen, The Netherlands
| | | | - Tania O Crişan
- Department of Medical Genetics, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud UMC, Nijmegen, The Netherlands. .,Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands. .,Department of Medical Genetics, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj Napoca, Romania.
| |
Collapse
|
16
|
Single-cell RNA sequencing of immune cells in patients with acute gout. Sci Rep 2022; 12:22130. [PMID: 36550178 PMCID: PMC9772586 DOI: 10.1038/s41598-022-25871-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Cell subpopulations in the blood and joint fluid of patients with gout are poorly understood. Single-cell RNA sequencing and bioinformatic tools were used to identify cell subsets and their gene signatures in blood and synovial fluid (SF) cells, determine their relationships, characterize the diversity, and evaluate interactions among specific cell types. We identified 34 subpopulations (5 types of B cells, 16 types of T and natural killer cells, 9 types of monocytes, and 4 other cell types) in the blood of five healthy subjects and seven patients with acute gouty, and the SF of three patients with acute gout. We found that naïve CD4 T cells and classical monocytes cell populations were enriched in patients with gout, whereas plasmacytoid dendritic cells and intermediate monocytes were more abundant in healthy subjects. SF was enriched in Th1/Th17 cells, effector memory CD8 T cells, mucosal-associated invariant T cells, and macrophages. Subclusters of these cell subpopulations showed different compositions between healthy subjects and those with acute gout, according to blood and SF samples. At the cellular level, the inflammation score of a subpopulation or subcluster was highest in SF, following by the blood of acute gout patients and healthy person, whereas energy score showed the opposite trend. We also detected specific cell-cell interactions for interleukin-1, tumor necrosis factor-α, and transforming growth factor-β1 expression in the cells of patients with acute gout. Our study reveals cellular and molecular insights on inflammatory responses to hyperuricemia or uric crystal and may provide therapeutic guidance to improve treatments for gout.
Collapse
|
17
|
|
18
|
Copur S, Demiray A, Kanbay M. Uric acid in metabolic syndrome: Does uric acid have a definitive role? Eur J Intern Med 2022; 103:4-12. [PMID: 35508444 DOI: 10.1016/j.ejim.2022.04.022] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/18/2022] [Accepted: 04/27/2022] [Indexed: 12/25/2022]
Abstract
Increased serum uric acid (SUA) levels are commonly seen in patients with metabolic syndrome and are widely accepted as risk factors for hypertension, gout, non-alcoholic fatty liver disease, chronic kidney disease (CKD), and cardiovascular diseases. Although some ambiguity for the exact role of uric acid (UA) in these diseases is still present, several pathophysiological mechanisms have been identified such as increased oxidative stress, inflammation, and apoptosis. Accumulating evidence in genomics enlightens genetic variabilities and some epigenetic changes that can contribute to hyperuricemia. Here we discuss the role of UA within metabolism and the consequences of asymptomatic hyperuricemia while providing newfound evidence for the associations between UA and gut microbiota and vitamin D. Increased SUA levels and beneficial effects of lowering SUA levels need to be elucidated more to understand its complicated function within different metabolic pathways and set optimal target levels for SUA for reducing risks for metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Atalay Demiray
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Mehmet Kanbay
- Division of Nephrology, Department of Medicine, Koc University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
19
|
Birefringent Crystals Deposition and Inflammasome Expression in Human Atheroma Plaques by Levels of Uricemia. Joint Bone Spine 2022; 89:105423. [PMID: 35714832 DOI: 10.1016/j.jbspin.2022.105423] [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: 03/07/2022] [Revised: 04/20/2022] [Accepted: 06/01/2022] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To verify the monosodium urate (MSU) crystal deposition in artery walls following a structure assessment and to assess NLRP3 inflammasome expression in human atheroma plaques by levels of uricemia. METHODS Patients with peripheral arterial disease who were candidates for amputation were recruited and classified as normouricemic or hyperuricemic. During surgery, an artery segment from the amputated limb was sampled, divided and fixed separately by cryo-embedding, 100% ethanol or Glyo-fixx. Samples were assessed by compensated polarized-light microscopy to identify MSU crystals on the artery walls. Afterwards, macrophages, neutrophils and NLRP3 inflammasome components at the plaque were categorized by immunostaining and compared between normouricemics and hyperuricemics. RESULTS Thirty artery samples from 27 patients were studied; 10 (37.0%) participants were hyperuricemic. Birefringent needle-shaped crystals were found in three samples (10.0%), all processed by frozen sectioning. Other methods showed no crystals. No accompanying inflammatory process was noted, and the presence of crystals was equally distributed across ranges of uricemia, making it unlikely they were MSU crystals. Regarding immunostaining, 28 artery samples were available for analysis, with similar infiltration of macrophages and neutrophils. NLRP3 and gasdermin-D expression were significantly greater in hyperuricemics compared to normouricemics (p=0.044 and p=0.017, respectively). ASC content was numerically larger in hyperuricemics as well, while caspase-1 and IL-1beta expression were similar between groups. CONCLUSIONS The presence of MSU crystals on artery walls was not confirmed. Hyperuricemia was associated with greater NLRP3 and gasdermin-D expression on human atheroma plaques in patients with peripheral artery disease.
Collapse
|
20
|
Badii M, Gaal O, Popp RA, Crisan TO, Joosten LAB. Trained immunity and inflammation in rheumatic diseases. Joint Bone Spine 2022; 89:105364. [DOI: 10.1016/j.jbspin.2022.105364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/01/2022] [Accepted: 02/09/2022] [Indexed: 11/27/2022]
|
21
|
Role of Damage-Associated Molecular Pattern/Cell Death Pathways in Vaccine-Induced Immunity. Viruses 2021; 13:v13122340. [PMID: 34960608 PMCID: PMC8708515 DOI: 10.3390/v13122340] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022] Open
Abstract
Immune responses induced by natural infection and vaccination are known to be initiated by the recognition of microbial patterns by cognate receptors, since microbes and most vaccine components contain pathogen-associated molecular patterns. Recent discoveries on the roles of damage-associated molecular patterns (DAMPs) and cell death in immunogenicity have improved our understanding of the mechanism underlying vaccine-induced immunity. DAMPs are usually immunologically inert, but can transform into alarming signals to activate the resting immune system in response to pathogenic infection, cellular stress and death, or tissue damage. The activation of DAMPs and cell death pathways can trigger local inflammation, occasionally mediating adaptive immunity, including antibody- and cell-mediated immune responses. Emerging evidence indicates that the components of vaccines and adjuvants induce immunogenicity via the stimulation of DAMP/cell death pathways. Furthermore, strategies for targeting this pathway to enhance immunogenicity are being investigated actively. In this review, we describe various DAMPs and focus on the roles of DAMP/cell death pathways in the context of vaccines for infectious diseases and cancer.
Collapse
|