1
|
Ontiveros-Padilla L, Bachelder EM, Ainslie KM. Microparticle and nanoparticle-based influenza vaccines. J Control Release 2024; 376:880-898. [PMID: 39427775 DOI: 10.1016/j.jconrel.2024.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
Influenza infections are a health public problem worldwide every year with the potential to become the next pandemic. Vaccination is the most effective strategy to prevent future influenza outbreaks, however, influenza vaccines need to be reformulated each year to provide protection due to viral antigenic drift and shift. As more efficient influenza vaccines are needed, it is relevant to recapitulate strategies to improve the immunogenicity and broad reactivity of the current vaccines. Here, we review the current approved vaccines in the U.S. market and the platform used for their production. We discuss the different approaches to develop a broadly reactive vaccine as well as reviewing the adjuvant systems that are under study for being potentially included in future influenza vaccine formulations. The main components of the immune system involved in achieving a protective immune response are reviewed and how they participate in the trafficking of particles systemically and in the mucosa. Finally, we describe and classify, according to their physicochemical properties, some of the potential micro and nano-particulate platforms that can be used as delivery systems for parenteral and mucosal vaccinations.
Collapse
Affiliation(s)
- Luis Ontiveros-Padilla
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA; Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, USA; Department of Microbiology and Immunology, School of Medicine, UNC, Chapel Hill, NC, USA.
| |
Collapse
|
2
|
Rodriguez-Navarro C, Elert K, Ibañez-Velasco A, Monasterio-Guillot L, Andres M, Sivera F, Pascual E, Ruiz-Agudo E. Unraveling the pathological biomineralization of monosodium urate crystals in gout patients. Commun Biol 2024; 7:828. [PMID: 38972919 PMCID: PMC11228021 DOI: 10.1038/s42003-024-06534-6] [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: 03/14/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
Crystallization of monosodium urate monohydrate (MSU) leads to painful gouty arthritis. Despite extensive research it is still unknown how this pathological biomineralization occurs, which hampers its prevention. Here we show how inflammatory MSU crystals form after a non-inflammatory amorphous precursor (AMSU) that nucleates heterogeneously on collagen fibrils from damaged articular cartilage of gout patients. This non-classical crystallization route imprints a nanogranular structure to biogenic acicular MSU crystals, which have smaller unit cell volume, lower microstrain, and higher crystallinity than synthetic MSU. These distinctive biosignatures are consistent with the template-promoted crystallization of biotic MSU crystals after AMSU at low supersaturation, and their slow growth over long periods of time (possibly years) in hyperuricemic gout patients. Our results help to better understand gout pathophysiology, underline the role of cartilage damage in promoting MSU crystallization, and suggest that there is a time-window to treat potential gouty patients before a critical amount of MSU has slowly formed as to trigger a gout flare.
Collapse
Grants
- PID2021.125305NB.I00 Ministry of Economy and Competitiveness | Agencia Estatal de Investigación (Spanish Agencia Estatal de Investigación)
- B-RNM-574-UGR20 Consejería de Economía, Innovación, Ciencia y Empleo, Junta de Andalucía (Ministry of Economy, Innovation, Science and Employment, Government of Andalucia)
- RNM-179 Consejería de Economía, Innovación, Ciencia y Empleo, Junta de Andalucía (Ministry of Economy, Innovation, Science and Employment, Government of Andalucia)
- UCE-PP2016-05 Universidad de Granada (University of Granada)
Collapse
Affiliation(s)
- Carlos Rodriguez-Navarro
- Department of Mineralogy and Petrology, University of Granada, Fuentenueva s/n, 18002, Granada, Spain.
| | - Kerstin Elert
- Department of Mineralogy and Petrology, University of Granada, Fuentenueva s/n, 18002, Granada, Spain
- Escuela de Estudios Arabes, Consejo Superior de Investigaciones Científicas (EEA-CSIC), C. Chapiz 22, 18010, Granada, Spain
| | - Aurelia Ibañez-Velasco
- Department of Mineralogy and Petrology, University of Granada, Fuentenueva s/n, 18002, Granada, Spain
| | - Luis Monasterio-Guillot
- Department of Mineralogy and Petrology, University of Granada, Fuentenueva s/n, 18002, Granada, Spain
| | - Mariano Andres
- Department of Clinical Medicine, Miguel Hernandez University, CN 332 s/n, 03550, Alicante, Spain
- Department of Rheumatology, Dr. Balmis General University Hospital, Alicante Institute for Health and Biomedical Research, Av. Pintor Baeza s/n, 03010, Alicante, Spain
| | - Francisca Sivera
- Department of Clinical Medicine, Miguel Hernandez University, CN 332 s/n, 03550, Alicante, Spain
- Department of Rheumatology, Elda General University Hospital, Carretera Elda-Sax s/n, 03600, Elda, Spain
| | - Eliseo Pascual
- Department of Clinical Medicine, Miguel Hernandez University, CN 332 s/n, 03550, Alicante, Spain
- Department of Rheumatology, Dr. Balmis General University Hospital, Alicante Institute for Health and Biomedical Research, Av. Pintor Baeza s/n, 03010, Alicante, Spain
| | - Encarnación Ruiz-Agudo
- Department of Mineralogy and Petrology, University of Granada, Fuentenueva s/n, 18002, Granada, Spain
| |
Collapse
|
3
|
Mara AB, Rawat K, King WT, Jakubzick CV. Natural antibodies drive type 2 immunity in response to damage-associated molecular patterns. JCI Insight 2024; 9:e177230. [PMID: 38470489 PMCID: PMC11141869 DOI: 10.1172/jci.insight.177230] [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: 11/01/2023] [Accepted: 03/07/2024] [Indexed: 03/13/2024] Open
Abstract
Allergic airway disease (AAD) is an example of type 2 inflammation that leads to chronic airway eosinophilia controlled by CD4 Th2 cells. Inflammation is reinforced by mast cells and basophils armed with allergen-specific IgE made by allergen-specific B2 B cells of the adaptive immune system. Little is known about how AAD is affected by innate B1 cells, which produce natural antibodies (NAbs) that facilitate apoptotic cell clearance and detect damage- and pathogen-associated molecular patterns (DAMPS and PAMPS). We used transgenic mice lacking either B cells or NAbs in distinct mouse models of AAD that require either DAMPS or PAMPS as the initial trigger for type 2 immunity. In a DAMP-induced allergic model, driven by alum and uric acid, mouse strains lacking B cells (CD19DTA), NAbs (IgHEL MD4), or all secreted antibodies (sIgm-/-Aid-/-) displayed a significant reduction in both eosinophilia and Th2 priming compared with WT or Aid-/- mice lacking only germinal center-dependent high-affinity class-switched antibodies. Replenishing B cell-deficient mice with either unimmunized B1 B cells or NAbs during sensitization restored eosinophilia, suggesting that NAbs are required for licensing antigen-presenting cells to prime type 2 immunity. Conversely, PAMP-dependent type 2 priming to house dust mite or Aspergillus was not dependent on NAbs. This study reveals an underappreciated role of B1 B cell-generated NAbs in selectively driving DAMP-induced type 2 immunity.
Collapse
|
4
|
Heyndrickx I, Deswarte K, Verstraete K, Verschueren KHG, Smole U, Aegerter H, Dansercoer A, Hammad H, Savvides SN, Lambrecht BN. Ym1 protein crystals promote type 2 immunity. eLife 2024; 12:RP90676. [PMID: 38194250 PMCID: PMC10945506 DOI: 10.7554/elife.90676] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024] Open
Abstract
Spontaneous protein crystallization is a rare event, yet protein crystals are frequently found in eosinophil-rich inflammation. In humans, Charcot-Leyden crystals (CLCs) are made from galectin-10 (Gal10) protein, an abundant protein in eosinophils. Although mice do not encode Gal10 in their genome, they do form pseudo-CLCs, made from the chitinase-like proteins Ym1 and/or Ym2, encoded by Chil3 and Chil4 and made by myeloid and epithelial cells respectively. Here, we investigated the biological effects of pseudo-CLCs since their function is currently unknown. We produced recombinant Ym1 crystals which were shown to have identical crystal packing and structure by X-ray crystallography as in vivo native crystals derived from murine lung. When administered to the airways of mice, crystalline but not soluble Ym1 stimulated innate and adaptive immunity and acted as a type 2 immune adjuvant for eosinophilic inflammation via triggering of dendritic cells (DCs). Murine Ym1 protein crystals found at sites of eosinophilic inflammation reinforce type 2 immunity and could serve as a surrogate model for studying the biology of human CLCs.
Collapse
Affiliation(s)
- Ines Heyndrickx
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation ResearchGhentBelgium
- Department of Internal Medicine and Pediatrics, Ghent UniversityGhentBelgium
| | - Kim Deswarte
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation ResearchGhentBelgium
- Department of Internal Medicine and Pediatrics, Ghent UniversityGhentBelgium
| | - Kenneth Verstraete
- Unit for Structural Biology, VIB-UGent Center for Inflammation ResearchGhentBelgium
- Department of Biochemistry and Microbiology, Ghent UniversityGhentBelgium
| | - Koen HG Verschueren
- Unit for Structural Biology, VIB-UGent Center for Inflammation ResearchGhentBelgium
- Department of Biochemistry and Microbiology, Ghent UniversityGhentBelgium
| | - Ursula Smole
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation ResearchGhentBelgium
- Department of Internal Medicine and Pediatrics, Ghent UniversityGhentBelgium
| | - Helena Aegerter
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation ResearchGhentBelgium
- Department of Internal Medicine and Pediatrics, Ghent UniversityGhentBelgium
| | - Ann Dansercoer
- Unit for Structural Biology, VIB-UGent Center for Inflammation ResearchGhentBelgium
- Department of Biochemistry and Microbiology, Ghent UniversityGhentBelgium
| | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation ResearchGhentBelgium
- Department of Internal Medicine and Pediatrics, Ghent UniversityGhentBelgium
| | - Savvas N Savvides
- Unit for Structural Biology, VIB-UGent Center for Inflammation ResearchGhentBelgium
- Department of Biochemistry and Microbiology, Ghent UniversityGhentBelgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation ResearchGhentBelgium
- Department of Internal Medicine and Pediatrics, Ghent UniversityGhentBelgium
- Department of Pulmonary Medicine, Erasmus University Medical Center RotterdamRotterdamNetherlands
| |
Collapse
|
5
|
Hao SH, Ye LY, Yang C. The landscape of pathophysiology guided therapeutic strategies for gout treatment. Expert Opin Pharmacother 2023; 24:1993-2003. [PMID: 38037803 DOI: 10.1080/14656566.2023.2291073] [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: 10/12/2023] [Accepted: 11/30/2023] [Indexed: 12/02/2023]
Abstract
INTRODUCTION Gout is a common autoinflammatory disease caused by hyperuricemia with acute and/or chronic inflammation as well as tissue damage. Currently, urate-lowering therapy (ULT) and anti-inflammatory therapy are used as first-line strategies for gout treatment. However, traditional drugs for gout treatment exhibit some unexpected side effects and are not suitable for certain patients due to their comorbidity with other chronic disease. AREAS COVERED In this review, we described the pathophysiology of hyperuricemia and monosodium urate (MSU) crystal induced inflammatory response during gout development in depth and comprehensively summarized the advances in the investigation of promising ULT drugs as well as anti-inflammatory drugs that might be safer and more effective for gout treatment. EXPERT OPINION New drugs that are developed based on these molecular mechanisms exhibited great efficacy on reduction of disease burden both in vitro and in vivo, implying their potential for clinical application. Moreover, hyperthermia also showed regulation effect on MSU crystals formation and the signaling pathways involved in inflammation.
Collapse
Affiliation(s)
- Sai Heng Hao
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lin Yan Ye
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chang Yang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Ma DW, Ha J, Yoon KS, Kang I, Choi TG, Kim SS. Innate Immune System in the Pathogenesis of Non-Alcoholic Fatty Liver Disease. Nutrients 2023; 15:2068. [PMID: 37432213 DOI: 10.3390/nu15092068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 07/12/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent condition characterized by lipid accumulation in hepatocytes with low alcohol consumption. The development of sterile inflammation, which occurs in response to a range of cellular stressors or injuries, has been identified as a major contributor to the pathogenesis of NAFLD. Recent studies of the pathogenesis of NAFLD reported the newly developed roles of damage-associated molecular patterns (DAMPs). These molecules activate pattern recognition receptors (PRRs), which are placed in the infiltrated neutrophils, dendritic cells, monocytes, or Kupffer cells. DAMPs cause the activation of PRRs, which triggers a number of immunological responses, including the generation of cytokines that promote inflammation and the localization of immune cells to the site of the damage. This review provides a comprehensive overview of the impact of DAMPs and PRRs on the development of NAFLD.
Collapse
Affiliation(s)
- Dae Won Ma
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung Sik Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
7
|
Tao H, Mo Y, Liu W, Wang H. A review on gout: Looking back and looking ahead. Int Immunopharmacol 2023; 117:109977. [PMID: 37012869 DOI: 10.1016/j.intimp.2023.109977] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/11/2023] [Accepted: 02/28/2023] [Indexed: 03/17/2023]
Abstract
Gout is a metabolic disease caused by the deposition of monosodium urate (MSU) crystals inside joints, which leads to inflammation and tissue damage. Increased concentration of serum urate is an essential step in the development of gout. Serum urate is regulated by urate transporters in the kidney and intestine, especially GLUT9 (SLC2A9), URAT1 (SLC22A12) and ABCG. Activation of NLRP3 inflammasome bodies and subsequent release of IL-1β by monosodium urate crystals induce the crescendo of acute gouty arthritis, while neutrophil extracellular traps (NETs) are considered to drive the self-resolving of gout within a few days. If untreated, acute gout may eventually develop into chronic tophaceous gout characterized by tophi, chronic gouty synovitis, and structural joint damage, leading the crushing burden of treatment. Although the research on the pathological mechanism of gout has been gradually deepened in recent years, many clinical manifestations of gout are still unable to be fully elucidated. Here, we reviewed the molecular pathological mechanism behind various clinical manifestations of gout, with a view to making contributions to further understanding and treatment.
Collapse
|
8
|
Golovanova OA. Characteristics of the Nucleation Process in a Model Solution of Synovial Fluid in the Presence of Organic Impurities. CRYSTALLOGR REP+ 2022. [DOI: 10.1134/s1063774522070197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
9
|
Natural antibodies and CRP drive anaphylatoxin production by urate crystals. Sci Rep 2022; 12:4483. [PMID: 35296708 PMCID: PMC8924570 DOI: 10.1038/s41598-022-08311-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/28/2022] [Indexed: 01/02/2023] Open
Abstract
In gout, crystallization of uric acid in the form of monosodium urate (MSU) leads to a painful inflammatory response. MSU crystals induce inflammation by activating the complement system and various immune cell types, and by inducing necrotic cell death. We previously found that the soluble pattern recognition molecule C-reactive protein (CRP) recognizes MSU crystals, while enhancing complement activation. In the absence of CRP, MSU crystals still induced complement activation, suggesting additional CRP-independent mechanisms of complement activation. In the present study, we searched for additional MSU crystal-binding complement activators. We found that all healthy individuals, even unborn children, have MSU crystal-specific immunoglobulin M (IgM) in their blood. This indicates that innate IgM, also known as natural IgM, recognizes these crystals. In serum lacking IgM and CRP, MSU crystals showed negligible complement activation as assessed by the production of the anaphylatoxins C4a, C3a, and C5a (listed in order of production via the classical complement pathway). We show that IgM and CRP both activate the classical complement pathway on MSU crystals. CRP was more efficient at fixating active C1 on the crystals and inducing release of the most inflammatory anaphylatoxin C5a, indicating non-redundant functions of CRP. Notably, while CRP recognizes MSU crystals but not the related calcium pyrophosphate dihydrate (CPPD) crystals, natural IgM bound to both, suggesting common and distinct mechanisms of recognition of individual crystal types by complement activators.
Collapse
|
10
|
Feldman D. The Effect of Size of Materials Formed or Implanted In Vivo on the Macrophage Response and the Resultant Influence on Clinical Outcome. MATERIALS (BASEL, SWITZERLAND) 2021; 14:4572. [PMID: 34443095 PMCID: PMC8402017 DOI: 10.3390/ma14164572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/23/2021] [Accepted: 08/02/2021] [Indexed: 11/24/2022]
Abstract
Both the chemistry and size of a material formed in vivo, or an implanted biomaterial, can alter the in vivo host response. Within the size range covered within this review, over 1 μm, chemistry is only important if the solid material is unstable and leeching small molecules. The macrophage activity and the resultant inflammatory response, however, are related to the size of the solid material. The premise of this review is that differences in size of the solid material, in different cases, can be the reason why there is some individual-to-individual variation in response. Specifically, the inflammatory response is enhanced when the size is between 1-50 μm. This will be looked at for three configurations: spherical particulate (silicone oil or gel from breast implants), elongated particulate (monosodium urate [MSU] crystals in gout or in kidney stones), and fibers (e.g., polyester used in fabric implants). These specific examples were selected because many still believe that the clinical outcome for each is controlled by the surface chemistry, when in fact it is the size. In each case, specific studies will be highlighted to either show a mechanism for creating different sizes and therefore a differential biological response (first three) or how changing the size and shape (diameter and spacing of fibers, in this example) can affect the response and can help explain the different responses to fabric implants found in vivo within the 1-50 μm size range. It was found that polyester fibers under 70 μm had a significant increase in macrophage response. Further, it was found that compounds found in synovial fluid could limit MSU crystal size. In addition, it was shown that plasma with low triglyceride levels emulsifies silicone oils to a greater extent than plasma with higher triglyceride levels. Therefore, in three cases it appears that differences in the inflammatory response between individuals and between different implants could be explained just by the size of the material formed or implanted.
Collapse
Affiliation(s)
- Dale Feldman
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
11
|
Goldenberg M, Wang H, Walker T, Kaffenberger BH. Clinical and immunologic differences in cellulitis vs. pseudocellulitis. Expert Rev Clin Immunol 2021; 17:1003-1013. [PMID: 34263717 DOI: 10.1080/1744666x.2021.1953982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: The immunologic mechanisms between cellulitis and pseudocellulitis differ greatly, even though their clinical presentations may overlap.Areas covered: This article discusses cellulitis and common entities within the pseudocellulitis spectrum including acute lymphedema, superficial venous thrombosis, allergic contact dermatitis, lipodermatosclerosis, stasis dermatitis, erythema nodosum, cutaneous gout, and bursitis. The literature search was conducted from PubMed search engine between March and May 2021.Expert commentary: While immunologic differences in cellulitis and the various entities of pseudocellulitis are clear, there is a practice gap in applying these differences to the clinic and hospital setting. Further, existing studies are weakened by the lack of a gold-standard diagnosis in this disease category. Additional work is necessary in developing a gold-standard for the diagnosis and secondly, to project these immunologic differences as biomarkers to differentiate sterile inflammation from a potential life threatening bacterial or fungal infection.
Collapse
Affiliation(s)
- Michael Goldenberg
- Division of Dermatology, Ohio State University College of Medicine, the Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Henry Wang
- Department of Emergency Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Trent Walker
- Division of Dermatology, Ohio State University College of Medicine, the Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Benjamin H Kaffenberger
- Division of Dermatology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
12
|
Bousoik E, Qadri M, Elsaid KA. CD44 Receptor Mediates Urate Crystal Phagocytosis by Macrophages and Regulates Inflammation in A Murine Peritoneal Model of Acute Gout. Sci Rep 2020; 10:5748. [PMID: 32238827 PMCID: PMC7113258 DOI: 10.1038/s41598-020-62727-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 03/16/2020] [Indexed: 02/04/2023] Open
Abstract
Gout is a chronic arthritis caused by the deposition of poorly soluble monosodium urate monohydrate (MSU) crystals in peripheral joints. Resident macrophages initiate inflammation in response to MSU mediated by NF-κB nuclear translocation and NLRP3 inflammasome activation. We investigated the role of CD44, a transmembrane receptor, in mediating MSU phagocytosis by macrophages. We used an antibody that sheds the extracellular domain (ECD) of CD44 to study the role of the receptor and its associated protein phosphatase 2A (PP2A) in macrophage activation. We also studied the significance of CD44 in mediating MSU inflammation in-vivo. Cd44−/− BMDMs showed reduced MSU phagocytosis, LDH release, IL-1β expression and production compared to Cd44+/+ BMDMs. Elevated CD44 staining was detected intracellularly and CD44 colocalized with α-tubulin as a result of MSU exposure and ECD-shedding reduced MSU phagocytosis in murine and human macrophages. Anti-CD44 antibody treatment reduced NF-κB p65 subunit nuclear levels, IL-1β expression, pro-IL-1β and IL-8 production in MSU stimulated THP-1 macrophages (p < 0.01). The effect of the antibody was mediated by an enhancement in PP2A activity. CD44 ECD-shedding reduced the conversion of procaspase-1 to active caspase-1, caspase-1 activity and resultant generation of mature IL-1β in macrophages. Neutrophil and monocyte influx and upregulated production of IL-1β was evident in wildtype mice. MSU failed to trigger neutrophil and monocyte recruitment in Cd44−/− mice and lower IL-1β levels were detected in peritoneal lavages from Cd44−/− mice (p < 0.01). Anti-CD44 antibody treatment reduced neutrophil and monocyte recruitment and resulted in reduced lavage IL-1β levels in the same model. CD44 plays a biologically significant role in mediating phagocytosis of MSU and downstream inflammation and is a novel target in gout treatment.
Collapse
Affiliation(s)
- Emira Bousoik
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Chapman University, Irvine, CA, USA.,School of Pharmacy, Omar-Al-Mukhtar University, Derna, Libya
| | - Marwa Qadri
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Chapman University, Irvine, CA, USA.,Department of Pharmacology, College of Pharmacy, Jazan University, Jazan, 82826, Saudi Arabia
| | - Khaled A Elsaid
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Chapman University, Irvine, CA, USA.
| |
Collapse
|
13
|
Abstract
Extensive studies have suggested a central role of B cells in the autoimmune pathogenesis, as loss of B cell tolerance results in increased serum levels of autoantibodies, enhanced effector T cell response and tissue damages. Here, we provide an overview of dysregulated B cell responses in the development of autoimmunity. In addition to their presence in the target organs, autoreactive B cells can promote the formation of ectopic lymphoid structures and differentiate into plasma cells that produce large amounts of autoantibodies and cytokines. In animal models that recapitulate the key features of human autoimmune disease, mechanistic studies have indicated two categories of autoantibodies: (1) serological markers for disease diagnosis and prognosis; (2) effector molecules that induce organ hypofunction or damage directly in an epitope-specific manner, or indirectly by activating other immune cell subsets. Moreover, B cell-derived cytokines usually promote the autoreactive T cell response during autoimmune development, but there is compelling evidence that a subpopulation of B cells negatively regulates immune responses, also known as regulatory B cells via secreting anti-inflammatory cytokines (IL-10, IL-35, etc.) or a contact-dependent fashion. Although B cell depletion could eliminate most circulating B cells in the periphery, the clinical outcomes of B cell depletion therapy for autoimmune diseases vary among individuals due to differential activation or survival signals for B cells provided by tissue microenvironment. Thus, therapeutic combinations that target immune checkpoints and B cell activation may represent a promising strategy for the effective treatment of human autoimmune diseases.
Collapse
|
14
|
Brovold H, Lund T, Svistounov D, Solbu MD, Jenssen TG, Ytrehus K, Zykova SN. Crystallized but not soluble uric acid elicits pro-inflammatory response in short-term whole blood cultures from healthy men. Sci Rep 2019; 9:10513. [PMID: 31324844 PMCID: PMC6642259 DOI: 10.1038/s41598-019-46935-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/03/2019] [Indexed: 12/27/2022] Open
Abstract
Several epidemiological studies have pointed at serum uric acid (SUA) as an independent risk factor for mortality, diabetes, hypertension, cardiovascular and kidney disease; however, no clear pathogenic pathway is established. Uric acid (UA) crystals show pro-inflammatory properties and can thus create or contribute to the state of chronic low-grade inflammation, a widely accepted pathogenic mechanism in several of the above-mentioned pathologies. On the other hand, soluble uric acid possesses antioxidant properties that might attenuate inflammatory responses. We aimed to explore the net effects of experimentally rising SUA in human whole blood cultures on several mediators of inflammation. Production of TNF-α, IL-1ß, IL-1RA, MCP-1 and IL-8 was assessed upon addition of 200 µM UA, 500 µM UA or monosodium urate (MSU) crystals in the presence or absence of 5 ng/ml lipopolysaccharide (LPS). RT-qPCR and multiplex bead based immunoassay were used to measure mRNA expression and cytokine release at 2 and 4 h of culture, respectively. 14C labeled UA was used to assess intracellular uptake of UA. We show that crystallized, but not soluble, UA induces production of pro-inflammatory mediators in human whole blood. Soluble UA is internalized in blood cells but does not potentiate or reduce LPS-induced release of cytokines.
Collapse
Affiliation(s)
- Henrik Brovold
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Trine Lund
- Cardiovascular Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Dmitri Svistounov
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway.,Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
| | - Marit D Solbu
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway.,Section of Nephrology, University Hospital of North Norway, Tromsø, Norway
| | - Trond G Jenssen
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Transplantation Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Kirsti Ytrehus
- Cardiovascular Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Svetlana N Zykova
- Metabolic and Renal Research Group, UiT The Arctic University of Norway, Tromsø, Norway. .,Center for Quality Assurance and Development, University Hospital of North Norway, Tromsø, Norway. .,Department of Blood Bank and Medical Biochemistry, Innlandet Hospital Trust, Lillehammer, Norway.
| |
Collapse
|
15
|
Integrative Genome-Wide Association Studies of eQTL and GWAS Data for Gout Disease Susceptibility. Sci Rep 2019; 9:4981. [PMID: 30899057 PMCID: PMC6428872 DOI: 10.1038/s41598-019-41434-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/26/2019] [Indexed: 01/08/2023] Open
Abstract
There is a paucity of genome-wide association study on Han Chinese gout patients. We performed a genome-wide association meta-analysis on two Taiwanese cohorts consisting of 758 gout cases and 14166 controls of Han Chinese ancestry. All the participants were recruited from the Taiwan Biobank. For pathway analysis, we applied ICSNPathway (Identify candidate Causal SNPs and Pathways) analysis, and to investigate whether expression-associated genetic variants contribute to gout susceptibility, we systematically integrated lymphoblastoid expression quantitative trait loci (eQTL) and genome-wide association data of gout using Sherlock, a Bayesian statistical frame-work. In the meta-analysis, we found 4 SNPs that reached genome-wide statistical significance (P < 5.0 × 10−8). These SNPs are in or close to ABCG2, PKD2 and NUDT9 gene on chromosome 4. ICSNPathway analysis identified rs2231142 as the candidate causal SNP, and ABCG2 as the candidate gene. Sherlcok analysis identified three genes, which were significantly associated with the risk of gout (PKD2, NUTD9, and NAP1L5). To conclude, we reported novel susceptible loci for gout that has not been previously addressed in the literature.
Collapse
|
16
|
Abstract
In contrast to adaptive antibodies, natural antibodies are present in a non-immunised organism from birth, and they do not include anti-Gal antibodies and/or anti-Gal natural antibodies, which are developed as a result of the effect of the α-Gal epitope and physiological flora. Natural antibodies are the first line of the organism’s defence before the formation of the immunity created via the stimulation of elements that determine specific and non-specific immunity. This is especially important in the case of infants. Despite the fact that natural antibodies differ in their function from adaptive antibodies, they are polyreactive and they detect autoantigens and new antigenic determinants. Natural antibodies are formed from the subpopulation of B lymphocytes, mainly B1 lymphocytes and B lymphocytes of the marginal zone. This phenomenon is supported by the fact that when the quantity of these cells in the organism decreases, which happens with age, the level of natural antibodies also decreases and the risk of illnesses of old age becomes higher. During ontogenesis, these antibodies participate in many physiological processes, including the “support” of the immune system and homeostasis, the prevention of inflammation, infections and other pathological states, such as autoimmune and cardiovascular diseases, or the process of carcinogenesis. The best known natural antibody is IgM, but the role of IgGs and IgAs is also considered important. Nowadays, many researchers also mention intravenous immunoglobulins, which are used in the treatment of numerous illnesses, and there are discussions on the possibility of increasing their potential if they were based on natural antibodies.
Collapse
|
17
|
Robinson PC. Gout - An update of aetiology, genetics, co-morbidities and management. Maturitas 2018; 118:67-73. [PMID: 30415758 DOI: 10.1016/j.maturitas.2018.10.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/22/2018] [Accepted: 10/25/2018] [Indexed: 02/07/2023]
Abstract
Gout is an increasingly common chronic disorder of urate crystal deposition that manifests as flares of acute inflammatory arthritis. Hyperuricaemia is a prerequisite and a fifth of both men and woman are hyperuricaemic. The prevalence of gout is much lower than the prevalence of hyperuricaemia for reasons that are not currently clear. Gout is more common in men than women prior to menopause due to the uricosuric effects of oestrogen, but after menopause the incidence of gout rises substantially in women. Co-morbidities are an important issue in gout, with cardiovascular disease, diabetes mellitus, obesity and chronic kidney disease all common in patients with gout. Environmental factors like diet affect the incidence of gout but there is little evidence to support an emphasis on diet in treating established gout. The diagnosis of gout is often made without the use of joint aspiration and validated diagnostic rules are available for both primary and secondary care as well as classification criteria for research use. The overarching principle of the management of gout with pharmacotherapy is the need to reduce serum urate levels to below a target of 0.30 mmol/L or 0.36 mmol/L depending on whether it is tophaceous or non-tophaceous respectively. The use of allopurinol has been researched extensively and newer strategies for safer effective dosing are now recommended. Newer agents have been introduced for the treatment of gout, including febuxostat and lesinurad. A number of important questions in the field are under current investigation.
Collapse
Affiliation(s)
- Philip C Robinson
- University of Queensland School of Clinical Medicine, Royal Brisbane and Women's Hospital, Butterfield Street, Herston, Brisbane, Queensland, 4006, Australia.
| |
Collapse
|
18
|
Shu F, Shi Y. Systematic Overview of Solid Particles and Their Host Responses. Front Immunol 2018; 9:1157. [PMID: 29892295 PMCID: PMC5985299 DOI: 10.3389/fimmu.2018.01157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/08/2018] [Indexed: 12/17/2022] Open
Abstract
Crystalline/particulate substances trigger a plethora of signaling events in host cells. The most prominent consequence is the inflammatory reactions that underlie crystal arthropathies, such as gout and pseudogout. However, their impact on our health was underestimated. Recent work on the role of cholesterol crystal in the development of atherosclerosis and the harm of environmental particulates has set up new frontiers in our defense against their detrimental effects. On the other hand, in the last 100 years, crystalline/particulate substances have been used with increasing frequencies in our daily lives as a part of new industrial manufacturing and engineering. Importantly, they have become a tool in modern medicine, used as vaccine adjuvants and drug delivery vehicles. Their biological effects are also being dissected in great detail, particularly with regard to their inflammatory signaling pathways. Solid structure interaction with host cells is far from being uniform, with outcomes dependent on cell types and chemical/physical properties of the particles involved. In this review, we offer a systematic and broad outlook of this landscape and a sage analysis of the complex nature of this topic.
Collapse
Affiliation(s)
- Fei Shu
- Department of Basic Medical Sciences, Institute for Immunology, Center for Life Sciences, Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Peking University, Beijing, China
| | - Yan Shi
- Department of Basic Medical Sciences, Institute for Immunology, Center for Life Sciences, Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
19
|
Chen CJ, Tseng CC, Yen JH, Chang JG, Chou WC, Chu HW, Chang SJ, Liao WT. ABCG2 contributes to the development of gout and hyperuricemia in a genome-wide association study. Sci Rep 2018; 8:3137. [PMID: 29453348 PMCID: PMC5816657 DOI: 10.1038/s41598-018-21425-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 01/25/2018] [Indexed: 01/31/2023] Open
Abstract
Although many genome-wide association studies (GWASs) of hyperuricemia or gout have been reported, the related genetic factors and the mechanisms from hyperuricemia to gouty attack remain unclear. This study aimed to identify genetic factors and pathogenesis of gout from hyperuricemia by genome-wide association study (GWAS). 747 gout patients, 747 hyperuricemia and 2071 age-matched controls were recruited and analyzed with Affymetrix 650 K chip to find the related genetic variants. The functions of the related genes were investigated in an endothelial cell (EC) with urate crystal stimulation. The GWAS results showed 36 SNPs to be strongly associated with gout compared to controls (all p-values < 10−7). Whereas the rs2231142 in ABCG2 gene had significant associations between gout and controls, between gout and hyperuricemia, and between hyperuricemia and controls (all p-values < 10−7), and the ORs were 4.34, 3.37 and 2.15 (all p-values < 0.001) after adjustment of potential confounders, respectively. The cell model showed significantly higher IL-8 release from EC combined with ABCG2 knockdown. We concluded that ABCG2 gene contributed to hyperuricemia but also gout, and that it was involved in the inflammation dysregulation via augmented IL-8 release in EC.
Collapse
Affiliation(s)
- Chung-Jen Chen
- Division of General Internal Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Internal Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Chun Tseng
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jeng-Hsien Yen
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jan-Gowth Chang
- Department of Laboratory Medicine and Epigenome Research Center, China Medical University Hospital, China Medical University, Kaohsiung, Taichung, Taiwan
| | - Wen-Cheng Chou
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hou-Wei Chu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shun-Jen Chang
- Department of Kinesiology, Health and Leisure Studies, National University of Kaohsiung, Kaohsiung, Taiwan.
| | - Wei-Ting Liao
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan. .,Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
20
|
El Ridi R, Tallima H. Physiological functions and pathogenic potential of uric acid: A review. J Adv Res 2017; 8:487-493. [PMID: 28748115 PMCID: PMC5512149 DOI: 10.1016/j.jare.2017.03.003] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 03/11/2017] [Accepted: 03/11/2017] [Indexed: 12/13/2022] Open
Abstract
Uric acid is synthesized mainly in the liver, intestines and the vascular endothelium as the end product of an exogenous pool of purines, and endogenously from damaged, dying and dead cells, whereby nucleic acids, adenine and guanine, are degraded into uric acid. Mentioning uric acid generates dread because it is the established etiological agent of the severe, acute and chronic inflammatory arthritis, gout and is implicated in the initiation and progress of the metabolic syndrome. Yet, uric acid is the predominant anti-oxidant molecule in plasma and is necessary and sufficient for induction of type 2 immune responses. These properties may explain its protective potential in neurological and infectious diseases, mainly schistosomiasis. The pivotal protective potential of uric acid against blood-borne pathogens and neurological and autoimmune diseases is yet to be established.
Collapse
Affiliation(s)
- Rashika El Ridi
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Hatem Tallima
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt.,Department of Chemistry, School of Science and Engineering, American University in Cairo, New Cairo 11835, Cairo, Egypt
| |
Collapse
|
21
|
P2X7 receptor-mediated TG2 externalization: a link to inflammatory arthritis? Amino Acids 2016; 49:453-460. [PMID: 27562793 PMCID: PMC5332493 DOI: 10.1007/s00726-016-2319-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 08/18/2016] [Indexed: 12/15/2022]
Abstract
Transglutaminases have important roles in stabilizing extracellular protein assemblies in tissue repair processes but some reaction products can stimulate immune activation, leading to chronic inflammatory conditions or autoimmunity. Exacerbated disease in models of inflammatory arthritis has been ascribed to sustained extracellular enzyme activity alongside formation of select protein modifications. Here, we review the evidence, with a focus on the link between P2X7R signaling and TG2 export, a pathway that we have recently discovered which ties extracellular protein modifications into the danger signal-mediated innate immune response. These recent insights offer new opportunities for therapeutic intervention.
Collapse
|
22
|
Synovial Fluid Findings and Demographic Analysis of Patients With Coexistent Intra-articular Monosodium Urate and Calcium Pyrophosphate Crystals. J Clin Rheumatol 2016; 22:68-70. [DOI: 10.1097/rhu.0000000000000321] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
23
|
Panda S, Ding JL. Natural antibodies bridge innate and adaptive immunity. THE JOURNAL OF IMMUNOLOGY 2016; 194:13-20. [PMID: 25527792 DOI: 10.4049/jimmunol.1400844] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Natural Abs, belonging to isotypes IgM, IgG3, and IgA, were discovered nearly half a century ago. Despite knowledge about the role of the polyreactive natural IgM in pathogen elimination, B cell survival and homeostasis, inflammatory diseases, and autoimmunity, there is a lack of clarity about the physiological role of natural IgG and natural IgA because they appear incapable of recognizing Ags on their own and are perceived as nonreactive. However, recent research revealed exciting functions of natural IgG in innate immunity. Natural IgG:lectin collaboration swiftly and effectively kills invading pathogens. These advances prompt further examination of natural Abs in immune defense and homeostasis, with the potential for developing novel therapeutics. This review provides new insights into the interaction between natural Abs and lectins, with implications on how interactions between molecules of the innate and adaptive immune systems bridge these two arms of immunity.
Collapse
Affiliation(s)
- Saswati Panda
- Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Jeak L Ding
- Department of Biological Sciences, National University of Singapore, Singapore 117543
| |
Collapse
|
24
|
|
25
|
Chhana A, Lee G, Dalbeth N. Factors influencing the crystallization of monosodium urate: a systematic literature review. BMC Musculoskelet Disord 2015; 16:296. [PMID: 26467213 PMCID: PMC4606994 DOI: 10.1186/s12891-015-0762-4] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 10/08/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Gout is a chronic disease of monosodium urate (MSU) crystal deposition. Although hyperuricaemia is the central risk factor for development of gout, not all people with hyperuricaemia have subclinical MSU crystal deposition or indeed, symptomatic disease. The aim of this systematic literature review was to identify factors that contribute to MSU crystallization. METHODS A search was conducted of the electronic databases PubMed, Science Direct and Scopus. Articles were included if they contained original data related to MSU crystallization. The methods and results were summarized and categorized into articles describing at least one of the three key steps in MSU crystallization (reduced urate solubility, nucleation and growth). RESULTS A total of 2175 articles were initially identified in our systematic search with 35 of these articles included in the final analysis. Elevated urate concentration was identified as a central factor driving all three stages of MSU crystallization. Factors that were found to consistently reduce urate solubility were reduced temperatures, pH 7-9 and various ions including sodium ions. Connective tissue factors including bovine cartilage homogenates and healthy human synovial fluid and serum all enhanced urate solubility. MSU nucleation was found to be increased by a number of factors, including sodium ions, uric acid binding antibodies, and synovial fluid or serum from patients with gout. Other than elevated urate concentrations, no other specific factors were identified as promoters of MSU crystal growth. CONCLUSIONS Increased urate concentration is the key factor required at each stage of MSU crystallization. Different proteins and factors within connective tissues may promote MSU crystallization and may be important for determining the sites at which MSU crystallization occurs in the presence of elevated urate concentrations.
Collapse
Affiliation(s)
- Ashika Chhana
- Bone & Joint Research Group, Department of Medicine, University of Auckland, Auckland, New Zealand.
| | - Gerald Lee
- Bone & Joint Research Group, Department of Medicine, University of Auckland, Auckland, New Zealand.
| | - Nicola Dalbeth
- Bone & Joint Research Group, Department of Medicine, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
26
|
Etiology and pathogenesis of gout. Rheumatology (Oxford) 2015. [DOI: 10.1016/b978-0-323-09138-1.00187-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
27
|
Chiang SL, Ou TT, Wu YJ, Tu HP, Lu CY, Huang CM, Kuo TM, Wang TN, Chou CH, Ko YC. Increased level of MSU crystal-bound protein apolipoprotein A-I in acute gouty arthritis. Scand J Rheumatol 2014; 43:498-502. [PMID: 25178483 DOI: 10.3109/03009742.2014.903994] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Gout is a common form of inflammatory arthritis that is triggered by the crystallization of monosodium urate (MSU). We investigated the potential proteins that relate to the pathogenesis or the spontaneous resolution of acute gouty arthritis. METHOD We screened for differentially expressed proteins in the plasma of patients with acute gouty arthritis using two-dimensional gel electrophoresis (2-DE) and mass spectrometry (MS) identification. We confirmed these findings in a population study of 209 subjects, and further determined the protein profile of the synovial fluid (SF) from 24 gouty patients during acute attack by liquid chromatography coupled with tandem MS (LC/MS/MS). RESULTS The highly expressed apolipoprotein A-I (apoA-I) was identified in the plasma of acute gouty patients compared with healthy controls. Moreover, we detected high levels of SF apoA-I in 83.3% of acute gouty patients during attack. From the population study, apoA-I was increasingly associated with normouricaemia, hyperuricaemia, and acute gouty arthritis (ptrend < 0.001), and plasma uric acid (UA) and apoA-I were positively correlated (p = 0.0156). We used a human liver cell model and found that UA enhanced the hepatic apoA-I mRNA expression level (ptrend < 0.01) and apoA-I secretion level (ptrend = 0.002) in a dose-dependent manner. An elevated MSU concentration caused the endogenous apoA-I to deplete gradually. CONCLUSIONS Based on the role of apoA-I in anti-inflammation, our observational data in acute gout support the hypothesis that apoA-I expression can be induced under the condition of a high concentration of UA and its elevated level may be implicated in the spontaneous resolution of acute gouty arthritis.
Collapse
Affiliation(s)
- S-L Chiang
- Environment-Omics-Disease Research Centre, China Medical University Hospital , Taichung, Taiwan , ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Dalbeth N, Lauterio TJ, Wolfe HR. Mechanism of action of colchicine in the treatment of gout. Clin Ther 2014; 36:1465-79. [PMID: 25151572 DOI: 10.1016/j.clinthera.2014.07.017] [Citation(s) in RCA: 259] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/07/2014] [Accepted: 07/22/2014] [Indexed: 12/25/2022]
Abstract
PURPOSE The aims of this article were to systematically review the literature about the mechanism of action of colchicine in the multimodal pathology of acute inflammation associated with gout and to consider the clinical utility of colchicine in other chronic inflammatory diseases. METHODS The English-language literature on PubMed was searched for articles published between 1990 and October 2013, with a cross-reference to citations across all years. Relevant articles pertaining to the mechanism of action of colchicine and the clinical applications of colchicine in gout and other inflammatory conditions were identified and reviewed. FINDINGS The molecular pathology of acute inflammation associated with gouty arthritis involves several concurrent pathways triggered by a variety of interactions between monosodium urate crystals and the surface of cells. Colchicine modulates multiple pro- and antiinflammatory pathways associated with gouty arthritis. Colchicine prevents microtubule assembly and thereby disrupts inflammasome activation, microtubule-based inflammatory cell chemotaxis, generation of leukotrienes and cytokines, and phagocytosis. Many of these cellular processes can be found in other diseases involving chronic inflammation. The multimodal mechanism of action of colchicine suggests potential efficacy of colchicine in other comorbid conditions associated with gout, such as osteoarthritis and cardiovascular disease. IMPLICATIONS Colchicine has multiple mechanisms of action that affect inflammatory processes and result in its utility for treating and preventing acute gout flare. Other chronic inflammatory diseases that invoke these molecular pathways may represent new therapeutic applications for colchicine.
Collapse
Affiliation(s)
- Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand.
| | | | | |
Collapse
|
29
|
Kataoka H, Kono H, Patel Z, Rock KL. Evaluation of the contribution of multiple DAMPs and DAMP receptors in cell death-induced sterile inflammatory responses. PLoS One 2014; 9:e104741. [PMID: 25127469 PMCID: PMC4134227 DOI: 10.1371/journal.pone.0104741] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 07/16/2014] [Indexed: 02/01/2023] Open
Abstract
When cells die by necrosis in vivo they stimulate an inflammatory response. It is thought that this response is triggered when the injured cells expose proinflammatory molecules, collectively referred to as damage associated molecular patterns (DAMPs), which are recognized by cells or soluble molecules of the innate or adaptive immune system. Several putative DAMPs and/or their receptors have been identified, but whether and how much they participate in responses in vivo is incompletely understood, and they have not previously been compared side-by-side in the same models. This study focuses on evaluating the contribution of multiple mechanisms that have been proposed to or potentially could participate in cell death-induced inflammation: The third component of complement (C3), ATP (and its receptor P2X7), antibodies, the C-type lectin receptor Mincle (Clec4e), and protease-activated receptor 2 (PAR2). We investigate the role of these factors in cell death-induced inflammation to dead cells in the peritoneum and acetaminophen-induced liver damage. We find that mice deficient in antibody, C3 or PAR2 have impaired inflammatory responses to dying cells. In contrast there was no reduction in inflammation to cell death in the peritoneum or liver of mice that genetically lack Mincle, the P2X7 receptor or that were treated with apyrase to deplete ATP. These results indicate that antibody, complement and PAR2 contribute to cell death-induced inflammation but that Mincle and ATP- P2X7 receptor are not required for this response in at least 2 different in vivo models.
Collapse
Affiliation(s)
- Hiroshi Kataoka
- Department of Pathology, UMass Medical School, Worcester, Massachusetts, United States of America
| | - Hajime Kono
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Zubin Patel
- Department of Pathology, UMass Medical School, Worcester, Massachusetts, United States of America
| | - Kenneth L. Rock
- Department of Pathology, UMass Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
30
|
Abstract
Gout is a common crystal-induced arthritis, in which monosodium urate (MSU) crystals precipitate within joints and soft tissues and elicit an inflammatory response. The causes of elevated serum urate and the inflammatory pathways activated by MSU crystals have been well studied, but less is known about the processes leading to crystal formation and growth. Uric acid, the final product of purine metabolism, is a weak acid that circulates as the deprotonated urate anion under physiologic conditions, and combines with sodium ions to form MSU. MSU crystals are known to have a triclinic structure, in which stacked sheets of purine rings form the needle-shaped crystals that are observed microscopically. Exposed, charged crystal surfaces are thought to allow for interaction with phospholipid membranes and serum factors, playing a role in the crystal-mediated inflammatory response. While hyperuricemia is a clear risk factor for gout, local factors have been hypothesized to play a role in crystal formation, such as temperature, pH, mechanical stress, cartilage components, and other synovial and serum factors. Interestingly, several studies suggest that MSU crystals may drive the generation of crystal-specific antibodies that facilitate future MSU crystallization. Here, we review MSU crystal biology, including a discussion of crystal structure, effector function, and factors thought to play a role in crystal formation. We also briefly compare MSU biology to that of uric acid stones causing nephrolithasis, and consider the potential treatment implications of MSU crystal biology.
Collapse
Affiliation(s)
- Miguel A Martillo
- Divisions of Rheumatology, Department of Medicine, NYU School of Medicine, New York, USA
| | | | | |
Collapse
|
31
|
Kubes P, Mehal WZ. Sterile inflammation in the liver. Gastroenterology 2012; 143:1158-1172. [PMID: 22982943 DOI: 10.1053/j.gastro.2012.09.008] [Citation(s) in RCA: 513] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 08/10/2012] [Accepted: 09/13/2012] [Indexed: 12/13/2022]
Abstract
Inflammation In the absence of pathogens occurs in all tissues in response to a wide range of stimuli that cause tissue stress and injury. Such sterile inflammation (SI) is a key process in drug-induced liver injury, nonalcoholic steatohepatitis, and alcoholic steatohepatitis and is a major determinant of fibrosis and carcinogenesis. In SI, endogenous damage-associated molecular patterns (DAMPS), which are usually hidden from the extracellular environment, are released on tissue injury and activate receptors on immune cells. More than 20 such DAMPS have been identified and activate cellular pattern recognition receptors, which were originally identified as sensors of pathogen-associated molecular patterns. Activation of pattern recognition receptors by DAMPS results in a wide range of immune responses, including production of proinflammatory cytokines and localization of immune cells to the site of injury. DAMPS result in the assembly of a cytosolic protein complex termed the inflammasome, which activates the serine protease caspase-1, resulting in activation and secretion of interleukin-1β and other cytokines. SI-driven liver diseases are responsible for the majority of liver pathology in industrially developed countries and lack specific therapy. Identification of DAMPS, their receptors, signaling pathways, and cytokines now provides a wide range of therapeutic targets for which many antagonists are already available.
Collapse
Affiliation(s)
- Paul Kubes
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Wajahat Z Mehal
- Section of Digestive Diseases, Yale University, and West Haven Veterans Medical Center, New Haven, Connecticut.
| |
Collapse
|
32
|
Amaral FA, Costa VV, Tavares LD, Sachs D, Coelho FM, Fagundes CT, Soriani FM, Silveira TN, Cunha LD, Zamboni DS, Quesniaux V, Peres RS, Cunha TM, Cunha FQ, Ryffel B, Souza DG, Teixeira MM. NLRP3 inflammasome-mediated neutrophil recruitment and hypernociception depend on leukotriene B(4) in a murine model of gout. ACTA ACUST UNITED AC 2012; 64:474-84. [PMID: 21952942 DOI: 10.1002/art.33355] [Citation(s) in RCA: 201] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Deposition of monosodium urate monohydrate (MSU) crystals in the joints promotes an intense inflammatory response and joint dysfunction. This study evaluated the role of the NLRP3 inflammasome and 5-lipoxygenase (5-LOX)-derived leukotriene B(4) (LTB(4) ) in driving tissue inflammation and hypernociception in a murine model of gout. METHODS Gout was induced by injecting MSU crystals into the joints of mice. Wild-type mice and mice deficient in NLRP3, ASC, caspase 1, interleukin-1β (IL-1β), IL-1 receptor type I (IL-1RI), IL-18R, myeloid differentiation factor 88 (MyD88), or 5-LOX were used. Evaluations were performed to assess neutrophil influx, LTB(4) activity, cytokine (IL-1β, CXCL1) production (by enzyme-linked immunosorbent assay), synovial microvasculature cell adhesion (by intravital microscopy), and hypernociception. Cleaved caspase 1 and production of reactive oxygen species (ROS) were analyzed in macrophages by Western blotting and fluorometric assay, respectively. RESULTS Injection of MSU crystals into the knee joints of mice induced neutrophil influx and neutrophil-dependent hypernociception. MSU crystal-induced neutrophil influx was CXCR2-dependent and relied on the induction of CXCL1 in an NLRP3/ASC/caspase 1/IL-1β/MyD88-dependent manner. LTB(4) was produced rapidly after injection of MSU crystals, and this was necessary for caspase 1-dependent IL-1β production and consequent release of CXCR2-acting chemokines in vivo. In vitro, macrophages produced LTB(4) after MSU crystal injection, and LTB(4) was relevant in the MSU crystal-induced maturation of IL-1β. Mechanistically, LTB(4) drove MSU crystal-induced production of ROS and ROS-dependent activation of the NLRP3 inflammasome. CONCLUSION These results reveal the role of the NLRP3 inflammasome in mediating MSU crystal-induced inflammation and dysfunction of the joints, and highlight a previously unrecognized role of LTB(4) in driving NLRP3 inflammasome activation in response to MSU crystals, both in vitro and in vivo.
Collapse
|
33
|
Ea HK. [Mechanisms of gout inflammation]. Presse Med 2011; 40:836-43. [PMID: 21684103 DOI: 10.1016/j.lpm.2011.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2011] [Revised: 03/15/2011] [Accepted: 03/22/2011] [Indexed: 01/14/2023] Open
Abstract
Gout inflammation is an acute and self-resolving reaction. MSU crystals can stimulate cells through either crystal-cell membrane interaction or after their phagocytosis. The onset of gout inflammation relies on non-hematopoietic resident cells whereas the amplification of the reaction is driven by phagocytic cells of immune innate system. Interleukin-1β (IL-1β) and polynuclear neutrophils play central role in gout inflammation. In vitro, MSU crystal-induced IL-1β secretion is secondary mainly to NLRP3 inflammasome activation although numerous proteases are also involved. Mechanisms of NLRP3 inflammasome activation remain unclear involving mostly reactive oxygen species production. Gout resolution involves several mechanisms including monocyte differentiation into macrophage, clearance of apoptotic neutrophils by macrophages, production of Transforming Growth Factor (TGF-β) and modification of protein coating on MSU crystal surface.
Collapse
Affiliation(s)
- Hang-Korng Ea
- Assistance-publique-Hôpitaux de Paris, hôpital Lariboisière, fédération de rhumatologie, pôle locomoteur, centre Viggo-Petersen, Inserm U606 (IFR139), Paris, France; Université médecine Paris 7, Paris, France.
| |
Collapse
|
34
|
Abstract
Gout is an ancient disease that still plagues us. Its pathogenic culprit, uric acid crystal deposition in tissues, is a strong inflammatory stimulant. In recent years, the mechanisms through which uric acid crystals promote inflammation have been a subject of increasing interest among rheumatologists and immunologists. Uric acid has been identified as an endogenous adjuvant that drives immune responses in the absence of microbial stimulation. Because uric acid is a ubiquitous metabolite that is produced in high quantities upon cellular injury, the ramifications of its effects may be considerable in health and in disease. Uric acid crystals also have been shown to trigger interleukin-1β-mediated inflammation via activation of the NOD-like receptor protein (NLRP)3 inflammasome, a multimolecular complex whose activation appears to be central to many pathological inflammatory conditions. In this article, we review the possible mechanisms of uric acid-mediated inflammation and offer some historical perspectives on what has been learned about the complex effects of a relatively simple substance.
Collapse
Affiliation(s)
- Faranak Ghaemi-Oskouie
- Department of Microbiology and Infectious Disease and Immunology Research Group, University of Calgary, 4A18 HRIC, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada
| | - Yan Shi
- Department of Microbiology and Infectious Disease and Immunology Research Group, University of Calgary, 4A18 HRIC, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
35
|
Flach TL, Ng G, Hari A, Desrosiers MD, Zhang P, Ward SM, Seamone ME, Vilaysane A, Mucsi AD, Fong Y, Prenner E, Ling CC, Tschopp J, Muruve DA, Amrein MW, Shi Y. Alum interaction with dendritic cell membrane lipids is essential for its adjuvanticity. Nat Med 2011; 17:479-87. [PMID: 21399646 DOI: 10.1038/nm.2306] [Citation(s) in RCA: 299] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Accepted: 01/18/2011] [Indexed: 12/16/2022]
Abstract
As an approved vaccine adjuvant for use in humans, alum has vast health implications, but, as it is a crystal, questions remain regarding its mechanism. Furthermore, little is known about the target cells, receptors, and signaling pathways engaged by alum. Here we report that, independent of inflammasome and membrane proteins, alum binds dendritic cell (DC) plasma membrane lipids with substantial force. Subsequent lipid sorting activates an abortive phagocytic response that leads to antigen uptake. Such activated DCs, without further association with alum, show high affinity and stable binding with CD4(+) T cells via the adhesion molecules intercellular adhesion molecule-1 (ICAM-1) and lymphocyte function-associated antigen-1 (LFA-1). We propose that alum triggers DC responses by altering membrane lipid structures. This study therefore suggests an unexpected mechanism for how this crystalline structure interacts with the immune system and how the DC plasma membrane may behave as a general sensor for solid structures.
Collapse
Affiliation(s)
- Tracy L Flach
- Immunology Research Group, Department of Microbiology & Infectious Diseases, and Snyder Institute, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Uric acid is a danger signal of increasing risk for osteoarthritis through inflammasome activation. Proc Natl Acad Sci U S A 2011; 108:2088-93. [PMID: 21245324 DOI: 10.1073/pnas.1012743108] [Citation(s) in RCA: 193] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Uric acid (UA) is known to activate the NLRP3 (Nacht, leucine-rich repeat and pyrin domain containing protein 3) inflammasome. When activated, the NLRP3 (also known as NALP3) inflammasome leads to the production of IL-18 and IL-1β. In this cohort of subjects with knee osteoarthritis (OA), synovial fluid uric acid was strongly correlated with synovial fluid IL-18 and IL-1β. Synovial fluid uric acid and IL-18 were strongly and positively associated with OA severity as measured by both radiograph and bone scintigraphy, and synovial fluid IL-1β was associated with OA severity but only by radiograph. Furthermore, synovial fluid IL-18 was associated with a 3-y change in OA severity, on the basis of the radiograph. We conclude that synovial fluid uric acid is a marker of knee OA severity. The correlation of synovial fluid uric acid with the two cytokines (IL-18 and IL-1β) known to be produced by uric acid-activated inflammasomes and the association of synovial fluid IL-18 with OA progression, lend strong support to the potential involvement of the innate immune system in OA pathology and OA progression.
Collapse
|
37
|
|
38
|
Abstract
The existence of IgM has been known for more than a century, but its importance in immunity and autoimmunity continues to emerge. Studies of mice deficient in secreted IgM have provided unexpected insights into its role in several diverse processes, from B cell survival to atherosclerosis, as well as in autoimmunity and protection against infection. Among the various distinct properties that underlie the functions of IgM, two stand out: its polyreactivity and its ability to facilitate the removal of apoptotic cells. In addition, new B cell-targeted therapies for the treatment of autoimmunity have been shown to cause a reduction in serum IgM, potentially disrupting the functions of this immunoregulatory molecule and increasing susceptibility to infection.
Collapse
|
39
|
Abstract
Gout is a common metabolic disorder characterized by elevated uric acid leading to the formation and accumulation of uric acid crystals in synovial fluids. An attack of gout is characterized by intense, self-limited bouts of acute arthritis with excruciating pain. The mechanisms regulating initiation and resolution of gouty inflammation are still unclear. A significant though incomplete body of information implicating the innate immune system as a central component of immune and inflammatory cell activation in gout has been accumulated over the past few years. In this review, advances in the understanding of the basic biology of crystal-mediated inflammation are summarized. The emerging role of the inflammasome and the cytokine interleukin-1 in the initiation of acute gout is highlighted. How these findings may open a door to a new approach for therapy with the development of interleukin-1 antagonists is discussed.
Collapse
|
40
|
Dalbeth N, Pool B, Gamble GD, Smith T, Callon KE, McQueen FM, Cornish J. Cellular characterization of the gouty tophus: a quantitative analysis. ACTA ACUST UNITED AC 2010; 62:1549-56. [PMID: 20131281 DOI: 10.1002/art.27356] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To characterize the cellular architecture of the tophus and to determine the presence of cytokines implicated in the initiation and resolution of gouty inflammation. METHODS Sixteen fixed, paraffin-embedded, uninfected tophus samples were surgically obtained from 12 patients with microscopically proven gout and were analyzed by quantitative immunohistochemistry. The number of cells present in the corona and fibrovascular zones of the tophus was analyzed by Genmod mixed models analysis. RESULTS Numerous CD68+ mononucleated and multinucleated cells were present within the corona zone. Mast cells were identified in all tophus samples and at similar densities throughout the corona and fibrovascular zones. In contrast, neutrophils were rarely observed. Plasma cells were present in very high numbers within the corona zone. The overall number of CD20+ B cells was much lower. However, in 6 of 12 patients (50%), at least 1 B cell aggregate was present in the fibrovascular zone. Large numbers of cells expressing interleukin-1beta (IL-1beta) were observed in the corona zone. Transforming growth factor beta1 (TGFbeta1)-expressing mononucleated cells were also identified. The number of CD68+ cells correlated with the number of cells expressing IL-1beta (r = 0.691, P = 0.009) and the number expressing TGFbeta1 (r = 0.518, P = 0.04). CONCLUSION The tophus represents a complex and organized chronic inflammatory tissue response to monosodium urate monohydrate crystals involving both innate and adaptive immune cells. The coexpression of IL-1beta and TGFbeta1 suggests that both proinflammatory and antiinflammatory factors present within the tophus contribute to a cycle of chronic inflammation, attempted resolution, and tissue remodeling.
Collapse
|
41
|
Ng G, Chau EMT, Shi Y. Recent Developments in Immune Activation by Uric Acid Crystals. Arch Immunol Ther Exp (Warsz) 2010; 58:273-7. [DOI: 10.1007/s00005-010-0082-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Accepted: 02/11/2010] [Indexed: 12/21/2022]
|
42
|
Abstract
Inflammation occurs in response to both pathogenic insult and tissue damage under sterile conditions, with the latter contributing to the pathogenesis of many diseases. Although several endogenous substances, including uric acid, have been suggested to alert the body to danger and to stimulate inflammation, little is known about their contribution to such responses in vivo. In this issue of the JCI, Kono et al. use newly generated mice with reduced levels of uric acid to investigate its role as an endogenous signal of tissue damage in inflammatory responses to hepatic injury. They find that uric acid is released from dying tissues and induces inflammation to cell death but is not involved in the response to microbial molecules or sterile irritant particles. I believe this to be the first report of an endogenous danger signal acting as a physiological regulator of inflammation.
Collapse
Affiliation(s)
- Yan Shi
- Immunology Research Group and Department of Microbiology & Infectious Diseases, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
43
|
Abstract
Uric acid crystals [monosodium urate (MSU)] have emerged as an important factor for both gouty arthritis and immune regulation. This simple crystalline structure appears to activate innate host defense mechanisms in multiple ways and triggers robust inflammation and immune activation. The recognition mechanisms of MSU following its phase change from soluble uric acid are diverse, involving both protein receptors and non-specific plasma membrane attachment. Upon contact with host cells, MSU induces a set of membrane events that trigger Syk and PI3K activation, phagocytosis, and cytokine production. Having entered the cell, MSU further triggers NALP3 inflammasome activation and induces the production of IL-1 beta, likely inducing a full spectrum of inflammation. This review describes the recognition mechanisms and activation pathways involved in MSU-mediated inflammation and adjuvanticity and hypothesizes that direct membrane binding by solid surfaces, such as MSU, may function as a generic mechanism in tissue responses to particulate and crystalline structures.
Collapse
Affiliation(s)
- Yan Shi
- Department of Microbiology and Infectious Diseases, and Immunology Research Group, Faculty of Medicine, University of Calgary, Calgary, AB, Canada.
| | | | | |
Collapse
|
44
|
Abstract
Gout is an arthritis characterized by elevated uric acid in the bloodstream. In this condition, crystals of uric acid are formed and accumulate in the synovial fluids. Crystal deposition leads to acute inflammation, which is associated with the spontaneous resolution of the disease. Recent studies have led to significant advances in the understanding of the basic biology of crystal-mediated inflammation. Uric acid has been identified as a danger signal that triggers a cytosolic sensor, the inflammasome. This signaling platform is required for the activation of interleukin-1, a cytokine that is critical to the initiation of acute inflammation in gout. Importantly, both molecular and pathological evidence support the notion that gout is a prototypical member of the growing family of autoinflammatory diseases. This review discusses the role of the inflammasome in gout and the emerging new therapeutic strategies aimed at controlling inflammation in crystal arthritis.
Collapse
Affiliation(s)
- Fabio Martinon
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Merriman TR, Dalbeth N. The genetic basis of hyperuricaemia and gout. Joint Bone Spine 2010; 78:35-40. [PMID: 20472486 DOI: 10.1016/j.jbspin.2010.02.027] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 02/10/2010] [Indexed: 12/14/2022]
Abstract
Gout results from elevated urate concentrations in the blood (hyperuricaemia). When super-saturation of urate is reached, monosodium urate crystals form within the joint. In some individuals, these crystals elicit a painful self-limiting inflammatory response that is characteristic of acute gouty arthritis. The most important cause of hyperuricaemia is reduced excretion of uric acid in the urine. Uric acid excretion is coordinated by a suite of urate transport molecules expressed in the renal collecting tubules, and is a key physiological checkpoint in gout. Other checkpoints in gout are hepatic production of urate, monosodium urate crystal formation, and initiation of the acute inflammatory response. Genome-wide association scans for genes regulating serum urate concentrations have identified two major regulators of hyperuricaemia- the renal urate transporters SLC2A9 and ABCG2. The risk variants at each gene approximately double the risk for gout in people of Caucasian ancestry, with SLC2A9 also resulting in higher risk for gout in people of Polynesian ancestry, a diverse population characterized by a high prevalence of gout. Ongoing genetic association studies are identifying and confirming other genes controlling serum urate concentrations; although genome-wide association studies in gout per se await recruitment of suitable case sample sets.
Collapse
Affiliation(s)
- Tony R Merriman
- Department of Biochemistry, University of Otago, PO Box 56, Dunedin, New Zealand.
| | | |
Collapse
|