1
|
Wang T, Shuai P, Wang Q, Guo C, Huang S, Li Y, Wu W, Yi L. α‑1 Antitrypsin is a potential target of inflammation and immunomodulation (Review). Mol Med Rep 2025; 31:107. [PMID: 40017119 PMCID: PMC11881679 DOI: 10.3892/mmr.2025.13472] [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] [Accepted: 12/12/2024] [Indexed: 03/01/2025] Open
Abstract
α‑1 Antitrypsin (AAT) is an acute phase protein encoded by the serine protease inhibitor family A member 1 gene. This multifunctional protein serves several roles, including anti‑inflammatory, antibacterial, antiapoptotic and immune regulatory functions. The primary role of AAT is to protect tissues and organs from protease‑induced damage due to its function as a serine protease inhibitor. AAT is associated with the development of lung inflammation, liver inflammation and immune‑mediated inflammatory diseases, which are influenced by environmental and genetic factors. For instance, AAT acts as an anti‑inflammatory protein to prevent and reverse type I diabetes. The present study briefly reviewed the molecular properties and mechanisms of AAT, as well as advances in the study of lung, liver and inflammatory diseases associated with AAT. The potential of AAT as a diagnostic and therapeutic target for inflammatory and immune‑mediated inflammatory diseases was reviewed. In addition, the damaging and protective effects of AAT, and its effects on organ function were discussed.
Collapse
Affiliation(s)
- Tiantian Wang
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Peimeng Shuai
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Qingyu Wang
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Caimao Guo
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Shuqi Huang
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yuanyuan Li
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wenyu Wu
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lan Yi
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
- Hengyang Key Laboratory of Cellular Stress Biology, Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
2
|
Yuan JSJ, Shashidhara A, Sutaria A, Tahir SH, Tahir H. An update on the pharmacotherapy of gout. Expert Opin Pharmacother 2025; 26:101-109. [PMID: 39665289 DOI: 10.1080/14656566.2024.2442028] [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: 10/23/2024] [Revised: 11/30/2024] [Accepted: 12/10/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION Gout is a common form of acute inflammatory arthritis caused by the deposition of monosodium urate crystals within synovium of joints. This leads to severe pain, reducing quality of life for patients with this condition. AREAS COVERED This review summarizes the treatment of both acute flares of gout and urate-lowering therapy based on guidance from various major international societies. We have also covered new emerging therapies that have not yet reached clinical practice. EXPERT OPINION Standard pharmacotherapies for gout flares include the options of colchicine, NSAIDs and oral or intramuscular corticosteroids, with IL-1 inhibitors newly established as an option for flare refractory to standard therapies. Urate-lowering therapies aim to prevent gout flares, with an emphasis on treat-to-target strategy; the escalation of therapies until the target serum uric acid is reached. Initial treatments include allopurinol and febuxostat, with uricosuric agents, probenecid, sulfinpyrazone and benzbromarone, as adjuncts. There are also emerging therapies in development. However, there is increasing personalization of treatment, adjusting pharmacotherapy depending on comorbidities, such as chronic kidney disease and ischemic heart disease, as well as patient preference.
Collapse
Affiliation(s)
- Jack Shi Jie Yuan
- Department of Rheumatology, Royal Free London NHS Foundation Trust, London, UK
- Department of Medicine, Barnet Hospital, London, UK
| | | | - Aman Sutaria
- UCL Medical School, University College London, London, UK
| | | | - Hasan Tahir
- Department of Rheumatology, Royal Free London NHS Foundation Trust, London, UK
- Division of Medicine, University College London, London, UK
| |
Collapse
|
3
|
Liu C, Yan Z, Zhang X, Xia T, Ashaolu JO, Olatunji OJ, Ashaolu TJ. Food-derived bioactive peptides potentiating therapeutic intervention in rheumatoid arthritis. Heliyon 2024; 10:e31104. [PMID: 38778960 PMCID: PMC11109807 DOI: 10.1016/j.heliyon.2024.e31104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that affects the joints of the human body and is projected to have a prevalence age-standardized rate of 1.5 million new cases worldwide by 2030. Several conventional and non-conventional preventive and therapeutic interventions have been suggested but they have their side effects including nausea, abdominal pain, liver damage, ulcers, heightened blood pressure, coagulation, and bleeding. Interestingly, several food-derived peptides (FDPs) from both plant and animal sources are increasingly gaining a reputation for their potential in the management or therapy of RA with little or no side effects. In this review, the concept of inflammation, its major types (acute and chronic), and RA identified as a chronic type were discussed based on its pathogenesis and pathophysiology. The conventional treatment options for RA were briefly outlined as the backdrop of introducing the FDPs that potentiate therapeutic effects in the management of RA.
Collapse
Affiliation(s)
- Chunhong Liu
- Second People's Hospital of Wuhu City, 241001, Anhui, China
| | - Zheng Yan
- Second People's Hospital of Wuhu City, 241001, Anhui, China
| | - Xiaohai Zhang
- Second People's Hospital of Wuhu City, 241001, Anhui, China
| | - Taibao Xia
- Second People's Hospital of Wuhu City, 241001, Anhui, China
| | - Joseph Opeoluwa Ashaolu
- Department of Public Health, Faculty of Basic Medical Sciences, Redeemers University, PMB 230, Ede, Osun State, Nigeria
| | | | - Tolulope Joshua Ashaolu
- Institute for Global Health Innovations, Duy Tan University, Da Nang, 550000, Viet Nam
- Faculty of Medicine, Duy Tan University, Da Nang, 550000, Viet Nam
| |
Collapse
|
4
|
Zhang WZ. Uric acid en route to gout. Adv Clin Chem 2023; 116:209-275. [PMID: 37852720 DOI: 10.1016/bs.acc.2023.05.003] [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] [Indexed: 10/20/2023]
Abstract
Gout and hyperuricemia (HU) have generated immense attention due to increased prevalence. Gout is a multifactorial metabolic and inflammatory disease that occurs when increased uric acid (UA) induce HU resulting in monosodium urate (MSU) crystal deposition in joints. However, gout pathogenesis does not always involve these events and HU does not always cause a gout flare. Treatment with UA-lowering therapeutics may not prevent or reduce the incidence of gout flare or gout-associated comorbidities. UA exhibits both pro- and anti-inflammation functions in gout pathogenesis. HU and gout share mechanistic and metabolic connections at a systematic level, as shown by studies on associated comorbidities. Recent studies on the interplay between UA, HU, MSU and gout as well as the development of HU and gout in association with metabolic syndromes, non-alcoholic fatty liver disease (NAFLD), and cardiovascular, renal and cerebrovascular diseases are discussed. This review examines current and potential therapeutic regimens and illuminates the journey from disrupted UA to gout.
Collapse
Affiliation(s)
- Wei-Zheng Zhang
- VIDRL, The Peter Doherty Institute, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Janciauskiene S, Tumpara S, Schebb NH, Buettner FFR, Mainka M, Sivaraman K, Immenschuh S, Grau V, Welte T, Olejnicka B. Indirect effect of alpha-1-antitrypsin on endotoxin-induced IL-1β secretion from human PBMCs. Front Pharmacol 2022; 13:995869. [PMID: 36249781 PMCID: PMC9564231 DOI: 10.3389/fphar.2022.995869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Human alpha-1-antitrypsin (AAT) encoded by the SERPINA1 gene, is an acute phase glycoprotein that regulates inflammatory responses via both protease inhibitory and non-inhibitory activities. We previously reported that AAT controls ATP-induced IL-1β release from human mononuclear cells by stimulating the release of small bioactive molecules. In the current study, we aimed to elucidate the identity of these putative effectors released from human PBMCs in response to AAT, which may inhibit the LPS-induced release of IL-1β. We pre-incubated human PBMCs alone or with different preparations of AAT (4 mg/ml) for 30 min at 37°C, 5% CO2, and collected cell supernatants filtered through centrifugal filters (cutoff 3 kDa) to eliminate AAT and other high molecular weight substances. Supernatants passed through the filters were used to culture PBMCs isolated from the autologous or a heterologous donors with or without adding LPS (1 μg/ml) for 6 h. Unexpectedly, supernatants from PBMCs pre-incubated with AAT (Zemaira®), but not with other AAT preparations tested or with oxidized AAT (Zemaira®), lowered the LPS-induced release of IL-1β by about 25%–60% without affecting IL1B mRNA. The reversed-phase liquid chromatography coupled with mass spectrometry did not confirm the hypothesis that small pro-resolving lipid mediators released from PBMCs after exposure to AAT (Zemaira®) are responsible for lowering the LPS-induced IL-1β release. Distinctively from other AAT preparations, AAT (Zemaira®) and supernatants from PBMCs pre-treated with this protein contained high levels of total thiols. In line, mass spectrometry analysis revealed that AAT (Zemaira®) protein contains freer Cys232 than AAT (Prolastin®). Our data show that a free Cys232 in AAT is required for controlling LPS-induced IL-1β release from human PBMCs. Further studies characterizing AAT preparations used to treat patients with inherited AAT deficiency remains of clinical importance.
Collapse
Affiliation(s)
- Sabina Janciauskiene
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Department of Experimental Medicine, Lund University, Lund, Sweden
- *Correspondence: Sabina Janciauskiene,
| | - Srinu Tumpara
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Falk F. R. Buettner
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Malwina Mainka
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Kokilavani Sivaraman
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Stephan Immenschuh
- Institute for Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Veronika Grau
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Center for Lung Research, Giessen, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Beata Olejnicka
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Department of Experimental Medicine, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Sun R, Xu Z, Zhu C, Chen T, Muñoz LE, Dai L, Zhao Y. Alpha-1 antitrypsin in autoimmune diseases: Roles and therapeutic prospects. Int Immunopharmacol 2022; 110:109001. [PMID: 35803133 DOI: 10.1016/j.intimp.2022.109001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 02/05/2023]
Abstract
Alpha-1 antitrypsin (A1AT) is a protease inhibitor in the serum. Its primary function is to inhibit the activity of a series of proteases, including proteinase 3, neutrophil elastase, metalloproteases, and cysteine-aspartate proteases. In addition, A1AT also has anti-inflammatory, anti-apoptotic, anti-oxidative stress, anti-viral, and anti-bacterial activities and plays essential roles in the regulation of tissue repair and lymphocyte differentiation and activation. The overactivation of the immune system characterizes the pathogenesis of autoimmune diseases. A1AT treatment shows beneficial effects on patients and animal models with autoimmune diseases such as rheumatoid arthritis and systemic lupus erythematosus. This review summarizes the functions and therapeutic prospects of A1AT in autoimmune diseases.
Collapse
Affiliation(s)
- Rui Sun
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiqiang Xu
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics and Department of General Practice, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chenxi Zhu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tao Chen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Luis E Muñoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lunzhi Dai
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics and Department of General Practice, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China; Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
7
|
Kim SK. The Mechanism of the NLRP3 Inflammasome Activation and Pathogenic Implication in the Pathogenesis of Gout. JOURNAL OF RHEUMATIC DISEASES 2022; 29:140-153. [PMID: 37475970 PMCID: PMC10324924 DOI: 10.4078/jrd.2022.29.3.140] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 07/22/2023]
Abstract
The NACHT, LRR, and PYD-domains-containing protein 3 (NLRP3) inflammasome is an intracellular multi-protein signaling platform that is activated by cytosolic pattern-recognition receptors such as NLRs against endogenous and exogenous pathogens. Once it is activated by a variety of danger signals, recruitment and assembly of NLRP3, ASC, and pro-caspase-1 trigger the processing and release of pro-inflammatory cytokines including interleukin-1β (IL-1β) and IL-18. Multiple intracellular and extracellular structures and molecular mechanisms are involved in NLRP3 inflammasome activation. Gout is an autoinflammatory disease induced by inflammatory response through production of NLRP3 inflammasome-mediated proinflammatory cytokines such as IL-1β by deposition of monosodium urate (MSU) crystals in the articular joints and periarticular structures. NLRP3 inflammasome is considered a main therapeutic target in MSU crystal-induced inflammation in gout. Novel therapeutic strategies have been proposed to control acute flares of gouty arthritis and prophylaxis for gout flares through modulation of the NLRP3/IL-1 axis pathway. This review discusses the basic mechanism of NLRP3 inflammasome activation and the IL-1-induced inflammatory cascade and explains the NLRP3 inflammasome-induced pathogenic role in the pathogenesis of gout.
Collapse
Affiliation(s)
- Seong-Kyu Kim
- Division of Rheumatology, Department of Internal Medicine, Daegu Catholic University School of Medicine, Daegu, Korea
| |
Collapse
|
8
|
Mellati A, Lo Faro L, Dumbill R, Meertens P, Rozenberg K, Shaheed S, Snashall C, McGivern H, Ploeg R, Hunter J. Kidney Normothermic Machine Perfusion Can Be Used as a Preservation Technique and a Model of Reperfusion to Deliver Novel Therapies and Assess Inflammation and Immune Activation. Front Immunol 2022; 13:850271. [PMID: 35720316 PMCID: PMC9198253 DOI: 10.3389/fimmu.2022.850271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
Ischaemia-reperfusion injury (IRI) is an inevitable process in transplantation and results in inflammation and immune system activation. Alpha-1 antitrypsin (AAT) has anti-inflammatory properties. Normothermic machine perfusion (NMP) can be used to deliver therapies and may help in assessing the effects of IRI and immunity. This study investigated the effects of AAT on IRI and inflammation in pig kidneys when administered during preservation, followed by normothermic reperfusion (NR) with autologous whole blood, as a surrogate for transplant. Two different models were used to deliver AAT or placebo to paired slaughterhouse pig kidneys: Model 1: 7-h static cold storage (SCS) + 3-h NR (n = 5 pairs), where either AAT (10 mg/ml) or placebo was delivered in the flush following retrieval; Model 2: 4-h SCS + 3-h NMP + 3-h NR (n = 5 pairs), where either AAT or placebo was delivered during NMP. Injury markers and cytokines levels were analysed in the perfusate, and heat shock protein 70 KDa (HSP-70) was analysed in biopsies. AAT delivered to kidneys showed no adverse effects on perfusion parameters. HSP-70 fold changes were significantly lower in the AAT group during NMP (P < 0.01, paired t-test) but not during NR. Interleukin-1 receptor antagonist (IL-1ra) fold changes were significantly higher in the AAT group during NR model 1 (p < 0.05, two-way ANOVA). In contrast to the AAT group, significant upregulation of interleukin-1 beta (IL-1β) and interleukin-6 (IL-6) between t = 90 min and t = 180 min and interleukin-8 (IL-8) between baseline and t = 90 min was observed in the control group in NR model 2 (p < 0.05, Tukey's multiple comparison test). However, overall inflammatory cytokines and injury markers showed similar levels between groups. Delivery of AAT to pig kidneys was safe without any detrimental effects. NMP and NR provided excellent methods for comparison of inflammation and immune activation in the delivery of a novel therapy.
Collapse
Affiliation(s)
- Azita Mellati
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Letizia Lo Faro
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Richard Dumbill
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Pommelien Meertens
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Leiden University Medical Centre, Leiden University, Leiden, Netherlands
| | - Kaithlyn Rozenberg
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Sadr Shaheed
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Corinna Snashall
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Hannah McGivern
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Rutger Ploeg
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Leiden University Medical Centre, Leiden University, Leiden, Netherlands
- Oxford University Hospital National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - James Hunter
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- University Hospitals of Coventry and Warwickshire National Health Service (NHS) Trust, Coventry, United Kingdom
| |
Collapse
|
9
|
Ouyang X, Li NZ, Guo MX, Zhang MM, Cheng J, Yi LT, Zhu JX. Active Flavonoids From Lagotis brachystachya Attenuate Monosodium Urate-Induced Gouty Arthritis via Inhibiting TLR4/MyD88/NF-κB Pathway and NLRP3 Expression. Front Pharmacol 2021; 12:760331. [PMID: 34803702 PMCID: PMC8602055 DOI: 10.3389/fphar.2021.760331] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/11/2021] [Indexed: 01/05/2023] Open
Abstract
Lagotis brachystachya Maxim is a characteristic herb commonly used in Tibetan medicine. Tibetan medicine records it as an important medicine for the clinical treatment of "Yellow Water Disease," the symptoms of which are similar to that of arthritis. Our previous study showed that the flavonoid fraction extracted from L. brachystachya could attenuate hyperuricemia. However, the effects of the active flavonoids on gouty arthritis remain elusive, and the underlying mechanism is not understood. In the present study, the effects of the active flavonoids were evaluated in rats or Raw264.7 cells with gouty arthritis induced by monosodium urate (MSU) crystal, followed by the detection of TLR4, MyD88, pNF-κB, and NLR family pyrin domain-containing 3 (NLRP3) expression. The swelling of the ankle joint induced by MSU crystal began to be relieved 6 h post the administration with the active flavonoids. In addition, the active flavonoids not only alleviated MSU crystal-induced inflammation in synovial tissues by histopathological examination but also reduced tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β) levels in the joint tissue fluid of MSU crystal-induced rats. Furthermore, Western blot analysis indicated that the active flavonoids reduced the production of these cytokines by inhibiting the TLR4/MyD88/NF-κB pathway and decreasing NLRP3 expression in synovial tissues of rats. More importantly, the inhibition of TLR4/MyD88/NF-κB pathway and NLRP3 expression was also confirmed in MSU-induced Raw264.7 cells. In conclusion, these results indicated that the active flavonoids from L. brachystachya could effectively attenuate gouty arthritis induced by MSU crystal through the TLR4/MyD88/NF-κB pathway and NLRP3 expression in vivo and in vitro, suggesting several potential candidates for the treatment of gouty arthritis.
Collapse
Affiliation(s)
- Xiang Ouyang
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Na-Zhi Li
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Min-Xia Guo
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Man-Man Zhang
- Department of Chemical and Pharmaceutical Engineering, Huaqiao University, Xiamen, China
| | - Jie Cheng
- Department of Chemical and Pharmaceutical Engineering, Huaqiao University, Xiamen, China
| | - Li-Tao Yi
- Department of Chemical and Pharmaceutical Engineering, Huaqiao University, Xiamen, China
| | - Ji-Xiao Zhu
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
10
|
Georgel PT, Georgel P. Where Epigenetics Meets Food Intake: Their Interaction in the Development/Severity of Gout and Therapeutic Perspectives. Front Immunol 2021; 12:752359. [PMID: 34603340 PMCID: PMC8484966 DOI: 10.3389/fimmu.2021.752359] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/31/2021] [Indexed: 01/02/2023] Open
Abstract
Gout is the most frequent form of inflammatory arthritis in the world. Its prevalence is particularly elevated in specific geographical areas such as in the Oceania/Pacific region and is rising in the US, Europe, and Asia. Gout is a severe and painful disease, in which co-morbidities are responsible for a significant reduction in life expectancy. However, gout patients remain ostracized because the disease is still considered "self-inflicted", as a result of unhealthy lifestyle and excessive food and alcohol intake. While the etiology of gout flares is clearly associated with the presence of monosodium urate (MSU) crystal deposits, several major questions remain unanswered, such as the relationships between diet, hyperuricemia and gout flares or the mechanisms by which urate induces inflammation. Recent advances have identified gene variants associated with gout incidence. Nevertheless, genetic origins of gout combined to diet-related possible uric acid overproduction account for the symptoms in only a minor portion of patients. Hence, additional factors must be at play. Here, we review the impact of epigenetic mechanisms in which nutrients (such as ω-3 polyunsaturated fatty acids) and/or dietary-derived metabolites (like urate) trigger anti/pro-inflammatory responses that may participate in gout pathogenesis and severity. We propose that simple dietary regimens may be beneficial to complement therapeutic management or contribute to the prevention of flares in gout patients.
Collapse
Affiliation(s)
- Philippe T Georgel
- Department of Biological Sciences, Cell Differentiation and Development Center, Joan C. Edwards School of Medicine, Byrd Biotechnology Science Center, Marshall University, Huntington, WV, United States
| | - Philippe Georgel
- Laboratoire d'ImmunoRhumatologie Moléculaire, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,Unité de Recherche et d'Expertise Immunity and Inflammation, Institut Pasteur in New Caledonia, Pasteur Network, Nouméa, New Caledonia
| |
Collapse
|
11
|
Kaneva MK. Neutrophil elastase and its inhibitors-overlooked players in osteoarthritis. FEBS J 2021; 289:113-116. [PMID: 34580987 DOI: 10.1111/febs.16194] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 12/13/2022]
Abstract
Cartilage homeostasis is maintained by a delicate balance between anabolism and catabolism. In osteoarthritis, pathological biomechanics or injury triggers cartilage breakdown, nonresolving synovial inflammation, and bone changes, causing reduced joint mobility and incapacitating pain. Undoubtedly, the most important cartilage degrading collagenase during osteoarthritis, matrix metalloproteinase (MMP)-13, is activated by an unlikely player: neutrophil elastase. Although primarily associated with inflammatory arthritis, neutrophil elastase is present in the osteoarthritic joint, and through activating MMP-13, spurs a cascade of events leading not just to the aberrant destruction of the cartilage itself, but to the proteolysis of its own inhibitor, alpha-1-antitrypsin, as described in the new study by Wilkinson et al. Endowed with potent chondrogenic and cartilage-protective properties, the loss of alpha-1-antitrypsin from cartilage will have major consequences for osteoarthritis progression, and strategies to prevent its loss, or replace it, might provide an innovative treatment opportunity that should not be ignored. Comment on: https://doi.org/10.1111/febs.16127.
Collapse
Affiliation(s)
- Magdalena K Kaneva
- The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, UK.,Centre for Inflammation and Therapeutic Innovation (CiTI), Queen Mary University of London, UK
| |
Collapse
|
12
|
Kaneva MK, Muley MM, Krustev E, Reid AR, Souza PR, Dell'Accio F, McDougall JJ, Perretti M. Alpha-1-antitrypsin reduces inflammation and exerts chondroprotection in arthritis. FASEB J 2021; 35:e21472. [PMID: 33788977 DOI: 10.1096/fj.202001801r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022]
Abstract
While new treatments have been developed to control joint disease in rheumatoid arthritis, they are partially effective and do not promote structural repair of cartilage. Following an initial identification of α-1-Antitrypsin (AAT) during the resolution phase of acute inflammation, we report here the properties of this protein in the context of cartilage protection, joint inflammation, and associated pain behavior. Intra-articular and systemic administration of AAT reversed joint inflammation, nociception, and cartilage degradation in the KBxN serum and neutrophil elastase models of arthritis. Ex vivo analyses of arthritic joints revealed that AAT promoted transcription of col2a1, acan, and sox9 and downregulated mmp13 and adamts5 gene expression. In vitro studies using human chondrocytes revealed that SERPINA1 transfection and rAAT protein promoted chondrogenic differentiation through activation of PKA-dependent CREB signaling and inhibition of Wnt/β-catenin pathways. Thus, AAT is endowed with anti-inflammatory, analgesic, and chondroprotective properties that are partially inter-related. We propose that AAT could be developed for new therapeutic strategies to reduce arthritic pain and repair damaged cartilage.
Collapse
Affiliation(s)
- Magdalena K Kaneva
- The William Harvey Research Institute, The London School of Medicine, Queen Mary University of London, London, UK
| | - Milind M Muley
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Eugene Krustev
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Allison R Reid
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Patricia R Souza
- The William Harvey Research Institute, The London School of Medicine, Queen Mary University of London, London, UK
| | - Francesco Dell'Accio
- The William Harvey Research Institute, The London School of Medicine, Queen Mary University of London, London, UK.,Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| | - Jason J McDougall
- Departments of Pharmacology and Anaesthesia, Pain Management & Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| | - Mauro Perretti
- The William Harvey Research Institute, The London School of Medicine, Queen Mary University of London, London, UK.,Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| |
Collapse
|
13
|
Galozzi P, Bindoli S, Luisetto R, Sfriso P, Ramonda R, Scanu A, Oliviero F. Regulation of crystal induced inflammation: current understandings and clinical implications. Expert Rev Clin Immunol 2021; 17:773-787. [PMID: 34053376 DOI: 10.1080/1744666x.2021.1937129] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Accumulation of abnormal crystals in the body, derived from endogenous or exogenous materials can drive a wide spectrum of inflammatory disease states. It is well established that intra-articular deposition of monosodium urate (MSU) and calcium pyrophoshate (CPP) crystals contributes to joint destruction through pro-inflammatory processes.Areas covered: This review will focus on current understanding and recent novelty about the mechanisms and the clinical implications of the inflammation induced by MSU and CPP crystals.Expert opinion: Advances in molecular biology reveal that at the base of the inflammatory cascade, stimulated by MSU or CPP crystals, there are many complex cellular mechanisms mainly involving the NLRP3 inflammasome, the hallmark of autoinflammatory syndromes. The extensive studies carried out through in vitro and in vivo models along with a better clinical definition of the disease has led to an optimized use of existing drugs and the introduction of novel therapeutic strategies. In particular, the identification of IL-1 as the most important target in gout and pseudogout has made it possible to expand the pharmacological indications of anti-IL-1 biological drugs, opening new therapeutic perspectives for patients.
Collapse
Affiliation(s)
- Paola Galozzi
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Sara Bindoli
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Roberto Luisetto
- Department of Surgery, Oncology and Gastroenterology-DISCOG, University of Padova, Padova, Italy
| | - Paolo Sfriso
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Roberta Ramonda
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Anna Scanu
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| | - Francesca Oliviero
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova, Padova, Italy
| |
Collapse
|
14
|
McElvaney OJ, Curley GF, Rose-John S, McElvaney NG. Interleukin-6: obstacles to targeting a complex cytokine in critical illness. THE LANCET. RESPIRATORY MEDICINE 2021; 9:643-654. [PMID: 33872590 PMCID: PMC8051931 DOI: 10.1016/s2213-2600(21)00103-x] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 02/07/2023]
Abstract
Circulating concentrations of the pleiotropic cytokine interleukin-6 (IL-6) are known to be increased in pro-inflammatory critical care syndromes, such as sepsis and acute respiratory distress syndrome. Elevations in serum IL-6 concentrations in patients with severe COVID-19 have led to renewed interest in the cytokine as a therapeutic target. However, although the pro-inflammatory properties of IL-6 are widely known, the cytokine also has a series of important physiological and anti-inflammatory functions. An adequate understanding of the complex processes by which IL-6 signalling occurs is crucial for the correct interpretation of IL-6 concentrations in the blood or lung, the use of IL-6 as a critical care biomarker, or the design of effective anti-IL-6 strategies. Here, we outline the role of IL-6 in health and disease, explain the different types of IL-6 signalling and their contribution to the net biological effect of the cytokine, describe the approaches to IL-6 inhibition that are currently available, and discuss implications for the future use of treatments such as tocilizumab in the critical care setting.
Collapse
Affiliation(s)
- Oliver J McElvaney
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Beaumont Hospital, Dublin, Ireland
| | - Gerard F Curley
- Beaumont Hospital, Dublin, Ireland; Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Stefan Rose-John
- Biochemical Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Noel G McElvaney
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Beaumont Hospital, Dublin, Ireland.
| |
Collapse
|
15
|
Bianchera A, Alomari E, Bruno S. Augmentation therapy with alpha 1-antitrypsin: present and future of production, formulation, and delivery. Curr Med Chem 2021; 29:385-410. [PMID: 34036902 DOI: 10.2174/0929867328666210525161942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/24/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
Alpha 1-antitrypsin is one of the first protein therapeutics introduced on the market - more than 30 years ago - and, to date, it is indicated only for the treatment of the severe forms of a genetic condition known as alpha-1 antitrypsin deficiency. The only approved preparations are derived from plasma, posing potential problems associated with its limited supply and high processing costs. Moreover, augmentation therapy with alpha 1-antitrypsin is still limited to intravenous infusions, a cumbersome regimen for patients. Here, we review the recent literature on its possible future developments, focusing on i) the recombinant alternatives to the plasma-derived protein, ii) novel formulations, and iii) novel administration routes. Regulatory issues and the still unclear noncanonical functions of alpha 1-antitrypsin - possibly associated with the glycosylation pattern found only in the plasma-derived protein - have hindered the introduction of new products. However, potentially new therapeutic indications other than the treatment of alpha-1 antitrypsin deficiency might open the way to new sources and new formulations.
Collapse
Affiliation(s)
- Annalisa Bianchera
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| | - Esraa Alomari
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| | - Stefano Bruno
- Dipartimento di Scienze degli Alimenti e del Farmaco, University of Parma, Parma, Italy
| |
Collapse
|
16
|
de Albuquerque Wanderley Sales V, Timóteo TRR, da Silva NM, de Melo CG, Ferreira AS, de Oliveira MVG, de Oliveira Silva E, Dos Santos Mendes LM, Rolim LA, Neto PJR. A Systematic Review of the Anti-inflammatory Effects of Gallium Compounds. Curr Med Chem 2021; 28:2062-2076. [PMID: 32484099 DOI: 10.2174/0929867327666200525160556] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/16/2020] [Accepted: 04/27/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Inflammation is an essential response provided by the immune system, ensuring the survival during microbial infection, tissue injury and other noxious conditions. However, prolonged inflammatory processes are often associated with severe side effects on health. OBJECTIVE This systematic review aimed to provide the evidence in the literature of the preclinical and human anti-inflammatory activity of gallium compounds from 2000 to 2019 focused on elucidating the mechanisms involved in the inflammatory process. METHODS Seven bibliographical databases were consulted (PubMed, Medline, ScienceDirect, Scopus, Springer, Web of Science, and EBSCOhost). The selection of appropriate publications and writing of this systematic review were based on the guidelines mentioned in the PRISMA statement. Moreover, the assessment of the methodological quality of the selected studies was also performed. RESULTS From a total of 3018 studies, 16 studies were included in this paper based on our eligibility criteria, which showed promising and consistent results. CONCLUSION Further research concerning specific inflammatory conditions is required.
Collapse
Affiliation(s)
| | | | - Natália Millena da Silva
- Laboratorio de Tecnologia dos Medicamentos, Federal University of Pernambuco (UFPE), Recife-PE, Brazil
| | - Camila Gomes de Melo
- Laboratorio de Tecnologia dos Medicamentos, Federal University of Pernambuco (UFPE), Recife-PE, Brazil
| | - Aline Silva Ferreira
- Laboratorio de Tecnologia dos Medicamentos, Federal University of Pernambuco (UFPE), Recife-PE, Brazil
| | | | - Emerson de Oliveira Silva
- Laboratorio de Tecnologia dos Medicamentos, Federal University of Pernambuco (UFPE), Recife-PE, Brazil
| | | | - Larissa Araújo Rolim
- Central de Analise de Farmacos, Medicamentos e Alimentos, Federal University of Vale do Sao Francisco (UNIVASF), Petrolina-PE, Brazil
| | - Pedro José Rolim Neto
- Laboratorio de Tecnologia dos Medicamentos, Federal University of Pernambuco (UFPE), Recife-PE, Brazil
| |
Collapse
|
17
|
Wang L, Balmat TJ, Antonia AL, Constantine FJ, Henao R, Burke TW, Ingham A, McClain MT, Tsalik EL, Ko ER, Ginsburg GS, DeLong MR, Shen X, Woods CW, Hauser ER, Ko DC. An atlas connecting shared genetic architecture of human diseases and molecular phenotypes provides insight into COVID-19 susceptibility. Genome Med 2021; 13:83. [PMID: 34001247 PMCID: PMC8127495 DOI: 10.1186/s13073-021-00904-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/05/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND While genome-wide associations studies (GWAS) have successfully elucidated the genetic architecture of complex human traits and diseases, understanding mechanisms that lead from genetic variation to pathophysiology remains an important challenge. Methods are needed to systematically bridge this crucial gap to facilitate experimental testing of hypotheses and translation to clinical utility. RESULTS Here, we leveraged cross-phenotype associations to identify traits with shared genetic architecture, using linkage disequilibrium (LD) information to accurately capture shared SNPs by proxy, and calculate significance of enrichment. This shared genetic architecture was examined across differing biological scales through incorporating data from catalogs of clinical, cellular, and molecular GWAS. We have created an interactive web database (interactive Cross-Phenotype Analysis of GWAS database (iCPAGdb)) to facilitate exploration and allow rapid analysis of user-uploaded GWAS summary statistics. This database revealed well-known relationships among phenotypes, as well as the generation of novel hypotheses to explain the pathophysiology of common diseases. Application of iCPAGdb to a recent GWAS of severe COVID-19 demonstrated unexpected overlap of GWAS signals between COVID-19 and human diseases, including with idiopathic pulmonary fibrosis driven by the DPP9 locus. Transcriptomics from peripheral blood of COVID-19 patients demonstrated that DPP9 was induced in SARS-CoV-2 compared to healthy controls or those with bacterial infection. Further investigation of cross-phenotype SNPs associated with both severe COVID-19 and other human traits demonstrated colocalization of the GWAS signal at the ABO locus with plasma protein levels of a reported receptor of SARS-CoV-2, CD209 (DC-SIGN). This finding points to a possible mechanism whereby glycosylation of CD209 by ABO may regulate COVID-19 disease severity. CONCLUSIONS Thus, connecting genetically related traits across phenotypic scales links human diseases to molecular and cellular measurements that can reveal mechanisms and lead to novel biomarkers and therapeutic approaches. The iCPAGdb web portal is accessible at http://cpag.oit.duke.edu and the software code at https://github.com/tbalmat/iCPAGdb .
Collapse
Affiliation(s)
- Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
| | - Thomas J Balmat
- Duke Research Computing, Duke University, Durham, NC, 27710, USA
| | - Alejandro L Antonia
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
| | - Florica J Constantine
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Ricardo Henao
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Thomas W Burke
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Andy Ingham
- Duke Research Computing, Duke University, Durham, NC, 27710, USA
| | - Micah T McClain
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC, 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Ephraim L Tsalik
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC, 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Emily R Ko
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
- Department of Hospital Medicine, Duke Regional Hospital, Durham, NC, 27705, USA
| | - Geoffrey S Ginsburg
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Mark R DeLong
- Duke Research Computing, Duke University, Durham, NC, 27710, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Woo Center for Big Data and Precision Health, Duke University, Durham, NC, 27710, USA
| | - Christopher W Woods
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC, 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC, 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Elizabeth R Hauser
- Duke Molecular Physiology Institute and Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, 27710, USA
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, NC, 27705, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA.
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA.
| |
Collapse
|
18
|
Abstract
Multiple interacting checkpoints are involved in the pathophysiology of gout. Hyperuricemia is the key risk factor for gout and is considered a prerequisite for monosodium urate (MSU) crystal formation. Urate underexcretion through renal and gut mechanisms is the major mechanism for hyperuricemia in most people. Multiple genetic, environmental, and metabolic factors are associated with serum urate and alter urate transport or synthesis. Urate supersaturation is the most important factor for MSU crystal formation, and other factors such as temperature, pH, and connective tissue components also play a role. The nucleotide-binding oligomerization domain leucine-rich repeats and pyrin domain-containing protein 3 inflammasome plays a pivotal role in the inflammatory response to MSU crystals, and interleukin 1β is the key cytokine mediating the inflammatory cascade. Variations in the regulatory mechanisms of this inflammatory response may affect an individual's susceptibility to developing gout. Tophus formation is the cardinal feature of advanced gout, and both MSU crystals and the inflammatory tissue component of the tophus contribute to the development of structural joint damage owing to gout. In this article, we review the pathophysiologic mechanisms of hyperuricemia, MSU crystal formation and the associated inflammatory response, tophus formation, and structural joint damage in gout.
Collapse
|
19
|
McNulty MJ, Silberstein DZ, Kuhn BT, Padgett HS, Nandi S, McDonald KA, Cross CE. Alpha-1 antitrypsin deficiency and recombinant protein sources with focus on plant sources: Updates, challenges and perspectives. Free Radic Biol Med 2021; 163:10-30. [PMID: 33279618 DOI: 10.1016/j.freeradbiomed.2020.11.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022]
Abstract
Alpha-1 antitrypsin deficiency (A1ATD) is an autosomal recessive disease characterized by low plasma levels of A1AT, a serine protease inhibitor representing the most abundant circulating antiprotease normally present at plasma levels of 1-2 g/L. The dominant clinical manifestations include predispositions to early onset emphysema due to protease/antiprotease imbalance in distal lung parenchyma and liver disease largely due to unsecreted polymerized accumulations of misfolded mutant A1AT within the endoplasmic reticulum of hepatocytes. Since 1987, the only FDA licensed specific therapy for the emphysema component has been infusions of A1AT purified from pooled human plasma at the 2020 cost of up to US $200,000/year with the risk of intermittent shortages. In the past three decades various, potentially less expensive, recombinant forms of human A1AT have reached early stages of development, one of which is just reaching the stage of human clinical trials. The focus of this review is to update strategies for the treatment of the pulmonary component of A1ATD with some focus on perspectives for therapeutic production and regulatory approval of a recombinant product from plants. We review other competitive technologies for treating the lung disease manifestations of A1ATD, highlight strategies for the generation of data potentially helpful for securing FDA Investigational New Drug (IND) approval and present challenges in the selection of clinical trial strategies required for FDA licensing of a New Drug Approval (NDA) for this disease.
Collapse
Affiliation(s)
- Matthew J McNulty
- Department of Chemical Engineering, University of California, Davis, CA, USA
| | - David Z Silberstein
- Department of Chemical Engineering, University of California, Davis, CA, USA
| | - Brooks T Kuhn
- Department of Internal Medicine, University of California, Davis, CA, USA; University of California, Davis, Alpha-1 Deficiency Clinic, Sacramento, CA, USA
| | | | - Somen Nandi
- Department of Chemical Engineering, University of California, Davis, CA, USA; Global HealthShare Initiative®, University of California, Davis, CA, USA
| | - Karen A McDonald
- Department of Chemical Engineering, University of California, Davis, CA, USA; Global HealthShare Initiative®, University of California, Davis, CA, USA
| | - Carroll E Cross
- Department of Internal Medicine, University of California, Davis, CA, USA; University of California, Davis, Alpha-1 Deficiency Clinic, Sacramento, CA, USA; Department of Physiology and Membrane Biology, University of California, Davis, CA, USA.
| |
Collapse
|
20
|
de Graaf DM, Maas RJA, Smeekens SP, Eisenmesser E, Redzic JS, Helsen MM, Powers NE, Li S, Kalabokis V, Gresnigt MS, Joosten LAB, Dinarello CA, van de Veerdonk FL. Human recombinant interleukin-38 suppresses inflammation in mouse models of local and systemic disease. Cytokine 2021; 137:155334. [PMID: 33128926 PMCID: PMC7725974 DOI: 10.1016/j.cyto.2020.155334] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 09/18/2020] [Accepted: 10/01/2020] [Indexed: 12/16/2022]
Abstract
Interleukin (IL)-38 belongs to the IL-1 family and is part of the IL-36 subfamily due to its binding to the IL-36 Receptor (IL-1R6). In the current study, we assessed the anti-inflammatory properties of IL-38 in murine models of arthritis and systemic inflammation. First, the anti-inflammatory properties of mouse and human IL-38 precursors were compared to forms with a truncated N-terminus. In mouse bone marrow derived dendritic cells (BMDC), human and mouse IL-38 precursors with a truncation of the two N-terminal amino acids (3-152) suppressed LPS-induced IL-6. Recombinant human IL-38 (3-152) was further investigated for its immunomodulatory potential using four murine models of inflammatory disease: streptococcal cell wall (SCW)-induced arthritis, monosodium urate (MSU) crystal-induced arthritis, MSU crystal-induced peritonitis, and systemic endotoxemia. In each of these models IL-38 significantly reduced inflammation. In SCW and MSU crystal-induced arthritis, joint swelling, inflammatory cell influx, and synovial levels of IL-1β, IL-6, and KC were reduced by 50% or greater. These suppressive properties of IL-38 in SCW-induced arthritis were independent of the anti-inflammatory co-receptor IL-1R8, as IL-38 reduced arthritis equally in IL-1R8 deficient and WT mice. In MSU crystal-induced peritonitis, IL-38 reduced hypothermia, while plasma IL-6 and KC and peritoneal KC levels were reduced by 65-70%. In the LPS endotoxemia model, IL-38 pretreatment reduced systemic IL-6, TNFα and KC. Furthermore, in ex vivo cultured bone marrow, LPS-induced IL-6, TNFα and KC were reduced by 75-90%. Overall, IL-38 exhibits broad anti-inflammatory properties in models of systemic and local inflammation and therefore may be an effective cytokine therapy.
Collapse
Affiliation(s)
- Dennis M de Graaf
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Ralph J A Maas
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Sanne P Smeekens
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Elan Eisenmesser
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, USA.
| | - Jasmina S Redzic
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO, USA.
| | - Monique M Helsen
- Department of Rheumatology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands.
| | - Nicholas E Powers
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA.
| | - Suzhao Li
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA.
| | | | - Mark S Gresnigt
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA; Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Jena, Germany.
| | - Leo A B Joosten
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Frank L van de Veerdonk
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
21
|
Wang L, Balmat TJ, Antonia AL, Constantine FJ, Henao R, Burke TW, Ingham A, McClain MT, Tsalik EL, Ko ER, Ginsburg GS, DeLong MR, Shen X, Woods CW, Hauser ER, Ko DC. An atlas connecting shared genetic architecture of human diseases and molecular phenotypes provides insight into COVID-19 susceptibility. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.12.20.20248572. [PMID: 33398303 PMCID: PMC7781346 DOI: 10.1101/2020.12.20.20248572] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
While genome-wide associations studies (GWAS) have successfully elucidated the genetic architecture of complex human traits and diseases, understanding mechanisms that lead from genetic variation to pathophysiology remains an important challenge. Methods are needed to systematically bridge this crucial gap to facilitate experimental testing of hypotheses and translation to clinical utility. Here, we leveraged cross-phenotype associations to identify traits with shared genetic architecture, using linkage disequilibrium (LD) information to accurately capture shared SNPs by proxy, and calculate significance of enrichment. This shared genetic architecture was examined across differing biological scales through incorporating data from catalogs of clinical, cellular, and molecular GWAS. We have created an interactive web database (interactive Cross-Phenotype Analysis of GWAS database (iCPAGdb); http://cpag.oit.duke.edu) to facilitate exploration and allow rapid analysis of user-uploaded GWAS summary statistics. This database revealed well-known relationships among phenotypes, as well as the generation of novel hypotheses to explain the pathophysiology of common diseases. Application of iCPAGdb to a recent GWAS of severe COVID-19 demonstrated unexpected overlap of GWAS signals between COVID-19 and human diseases, including with idiopathic pulmonary fibrosis driven by the DPP9 locus. Transcriptomics from peripheral blood of COVID-19 patients demonstrated that DPP9 was induced in SARS-CoV-2 compared to healthy controls or those with bacterial infection. Further investigation of cross-phenotype SNPs with severe COVID-19 demonstrated colocalization of the GWAS signal of the ABO locus with plasma protein levels of a reported receptor of SARS-CoV-2, CD209 (DC-SIGN), pointing to a possible mechanism whereby glycosylation of CD209 by ABO may regulate COVID-19 disease severity. Thus, connecting genetically related traits across phenotypic scales links human diseases to molecular and cellular measurements that can reveal mechanisms and lead to novel biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
- Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | | | - Alejandro L. Antonia
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Florica J. Constantine
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Ricardo Henao
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Thomas W. Burke
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Andy Ingham
- Duke Research Computing, Duke University, Durham, NC 27710, USA
| | - Micah T. McClain
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Ephraim L. Tsalik
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Emily R. Ko
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
- Department of Hospital Medicine, Duke Regional Hospital, Durham, NC, 27705, USA
| | - Geoffrey S. Ginsburg
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Mark R. DeLong
- Duke Research Computing, Duke University, Durham, NC 27710, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Woo Center for Big Data and Precision Health, Duke University, Durham, NC 27710, USA
| | - Christopher W. Woods
- Center for Applied Genomics and Precision Medicine, Department of Medicine, Duke University, Durham, NC 27710, USA
- Durham Veterans Affairs Health Care System, Durham, NC 27705, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Elizabeth R. Hauser
- Duke Molecular Physiology Institute and Department of Biostatistics and Bioinformatics, Duke University Medical Center Durham, NC 27710, USA
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, NC 27705, USA
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Lead contact
| |
Collapse
|
22
|
Klück V, Liu R, Joosten LAB. The role of interleukin-1 family members in hyperuricemia and gout. Joint Bone Spine 2020; 88:105092. [PMID: 33129923 DOI: 10.1016/j.jbspin.2020.105092] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Interleukin (IL)-1 family cytokines and their receptors have important roles in innate and partly in adaptive immunity. The family consists of 11 members of which IL-1α, IL-1β, IL-18, IL-33, IL-36α, IL-36β and IL-36γ are considered pro-inflammatory and IL-1Ra, IL-36Ra, IL-37 and IL-38 anti-inflammatory. Whereas IL-1β has a known pivotal role in gout, increasing evidence suggests other IL-1 family members are also involved in the pathogenesis of hyperuricemia and gout flares. FINDINGS Studies indicate IL-1α, like IL-1β, plays an essential role in the pathogenesis of gout flares. IL-18, although elevated in patients with gout, does not contribute to MSU crystal-induced inflammation, but may be involved in the subsequent development of cardiovascular disease in individuals with gout. The role of the pro-inflammatory cytokine IL-36 in gout remains elusive. In contrast, IL-1Ra, IL-33, IL-37 and IL-38 inhibit MSU crystal-induced inflammation and therefore have therapeutic potential for treatment of gout flares. In addition to existing IL-1β blockers, several new therapeutics to treat gout are being developed either inhibiting the transcription or maturation of IL-1β. CONCLUSION In this review, IL-1 family cytokines are discussed in the context of hyperuricemia and gout. Finally, current and novel therapeutic options for targeting IL-1 are reviewed.
Collapse
Affiliation(s)
- Viola Klück
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands; Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands.
| | - Ruiqi Liu
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands; Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands.
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands; Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, Geert Grooteplein Zuid, 8, 6525 GA, Nijmegen, The Netherlands; Department of Medical Genetics, Iuliu Hatieganu University of Medicine and Pharmacy, Street Pasteur nr. 6, 400349 Cluj-Napoca, Romania.
| |
Collapse
|
23
|
Distinct anti-inflammatory properties of alpha1-antitrypsin and corticosteroids reveal unique underlying mechanisms of action. Cell Immunol 2020; 356:104177. [DOI: 10.1016/j.cellimm.2020.104177] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/31/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
|
24
|
McElvaney OJ, McEvoy NL, McElvaney OF, Carroll TP, Murphy MP, Dunlea DM, Ní Choileáin O, Clarke J, O'Connor E, Hogan G, Ryan D, Sulaiman I, Gunaratnam C, Branagan P, O'Brien ME, Morgan RK, Costello RW, Hurley K, Walsh S, de Barra E, McNally C, McConkey S, Boland F, Galvin S, Kiernan F, O'Rourke J, Dwyer R, Power M, Geoghegan P, Larkin C, O'Leary RA, Freeman J, Gaffney A, Marsh B, Curley GF, McElvaney NG. Characterization of the Inflammatory Response to Severe COVID-19 Illness. Am J Respir Crit Care Med 2020; 202:812-821. [PMID: 32584597 PMCID: PMC7491404 DOI: 10.1164/rccm.202005-1583oc] [Citation(s) in RCA: 455] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/25/2020] [Indexed: 12/22/2022] Open
Abstract
Rationale: Coronavirus disease (COVID-19) is a global threat to health. Its inflammatory characteristics are incompletely understood.Objectives: To define the cytokine profile of COVID-19 and to identify evidence of immunometabolic alterations in those with severe illness.Methods: Levels of IL-1β, IL-6, IL-8, IL-10, and sTNFR1 (soluble tumor necrosis factor receptor 1) were assessed in plasma from healthy volunteers, hospitalized but stable patients with COVID-19 (COVIDstable patients), patients with COVID-19 requiring ICU admission (COVIDICU patients), and patients with severe community-acquired pneumonia requiring ICU support (CAPICU patients). Immunometabolic markers were measured in circulating neutrophils from patients with severe COVID-19. The acute phase response of AAT (alpha-1 antitrypsin) to COVID-19 was also evaluated.Measurements and Main Results: IL-1β, IL-6, IL-8, and sTNFR1 were all increased in patients with COVID-19. COVIDICU patients could be clearly differentiated from COVIDstable patients, and demonstrated higher levels of IL-1β, IL-6, and sTNFR1 but lower IL-10 than CAPICU patients. COVID-19 neutrophils displayed altered immunometabolism, with increased cytosolic PKM2 (pyruvate kinase M2), phosphorylated PKM2, HIF-1α (hypoxia-inducible factor-1α), and lactate. The production and sialylation of AAT increased in COVID-19, but this antiinflammatory response was overwhelmed in severe illness, with the IL-6:AAT ratio markedly higher in patients requiring ICU admission (P < 0.0001). In critically unwell patients with COVID-19, increases in IL-6:AAT predicted prolonged ICU stay and mortality, whereas improvement in IL-6:AAT was associated with clinical resolution (P < 0.0001).Conclusions: The COVID-19 cytokinemia is distinct from that of other types of pneumonia, leading to organ failure and ICU need. Neutrophils undergo immunometabolic reprogramming in severe COVID-19 illness. Cytokine ratios may predict outcomes in this population.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jennifer Clarke
- Department of Anaesthesia and Critical Care
- Beaumont Hospital, Dublin, Ireland; and
| | | | | | | | | | | | | | | | | | | | | | | | - Eoghan de Barra
- Department of International Health and Tropical Medicine, and
| | | | - Samuel McConkey
- Department of International Health and Tropical Medicine, and
| | - Fiona Boland
- Data Science Centre, Division of Biostatistics and Population Health Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | | | | | | | | | | | | | | | | | | | - Brian Marsh
- Department of Critical Care Medicine, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Gerard F Curley
- Department of Anaesthesia and Critical Care
- Beaumont Hospital, Dublin, Ireland; and
| | | |
Collapse
|
25
|
Mollaei M, Abbasi A, Hassan ZM, Pakravan N. The intrinsic and extrinsic elements regulating inflammation. Life Sci 2020; 260:118258. [PMID: 32818542 DOI: 10.1016/j.lfs.2020.118258] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/07/2020] [Accepted: 08/08/2020] [Indexed: 12/14/2022]
Abstract
Inflammation is a sophisticated biological tissue response to both extrinsic and intrinsic stimuli. Although the pathological aspects of inflammation are well appreciated, there are still rooms for understanding the physiological functions of the inflammation. Recent studies have focused on mechanisms, context and the role of physiological inflammation. Besides, there have been progress in the comprehension of commensal microbiota, immunometabolism, cancer and intracellular signaling events' roles that impact on the regulation of inflammation. Despite the fact that inflammatory responses are vital through tissue damage, understanding the mechanisms to turn off the finished or unnecessary inflammation is crucial for restoring homeostasis. Inflammation seems to be a smart process that acts like two edges of a sword, meaning that it has both protective and deleterious consequences. Knowing both edges and the regulation processes will help the future understanding and therapy for various diseases.
Collapse
Affiliation(s)
- M Mollaei
- Department of Immunology, School of Medicine, Tarbiat Modares University, Iran.
| | - A Abbasi
- Department of Immunology, School of Medicine, Tarbiat Modares University, Iran
| | - Z M Hassan
- Department of Immunology, School of Medicine, Tarbiat Modares University, Iran
| | - N Pakravan
- Department of Immunology, School of Medicine, Alborz University of Medical Science, Iran
| |
Collapse
|
26
|
Zhang X, Zou Y, Zheng J, Ji S, Wen X, Ye F, Liu J, Li X, Lei J, Qiu M. lncRNA‑MM2P downregulates the production of pro‑inflammatory cytokines in acute gouty arthritis. Mol Med Rep 2020; 22:2227-2234. [PMID: 32705194 PMCID: PMC7411394 DOI: 10.3892/mmr.2020.11314] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 04/17/2020] [Indexed: 12/18/2022] Open
Abstract
Acute gouty arthritis (AGA) is characterized by the accumulation of pro‑inflammatory cytokines, which are immunological responses to monosodium urate (MSU) crystals. It has been demonstrated that long non‑coding RNA (lncRNA)‑MM2P is a novel regulator of M2 polarization of macrophages. The aim of the present study was to investigate whether lncRNA‑MM2P regulates the MSU‑induced inflammatory process. In cell models of RAW 264.7 and THP‑1‑derived macrophages, decreased expression of lncRNA‑MM2P was observed in lipopolysaccharide‑ and MSU‑treated macrophages, which was accompanied with obvious inflammatory responses. Using small interfering RNA to knockdown lncRNA‑MM2P led to the upregulation of MSU‑mediated inflammatory responses, both in RAW 264.7 and THP‑1‑derived macrophages. In conclusion, lncRNA‑MM2P could be an important regulator of MSU‑induced inflammation, and therefore could be involved in the development of AGA.
Collapse
Affiliation(s)
- Xifeng Zhang
- Department of Rheumatism and Immunology of The First People's Hospital, Jiujiang, Jiangxi 332000, P.R. China
| | - Ying Zou
- Department of Rheumatology, Xuzhou Hospital of Traditional Chinese Medicine, Xuzhou, Jiangsu 221009, P.R. China
| | - Jiangxia Zheng
- Department of Rheumatism and Immunology of The First People's Hospital, Jiujiang, Jiangxi 332000, P.R. China
| | - Senguo Ji
- Department of Rheumatism and Immunology of The First People's Hospital, Jiujiang, Jiangxi 332000, P.R. China
| | - Xiuzhen Wen
- Department of Rheumatism and Immunology of The First People's Hospital, Jiujiang, Jiangxi 332000, P.R. China
| | - Feng Ye
- Department of Rheumatism and Immunology of The First People's Hospital, Jiujiang, Jiangxi 332000, P.R. China
| | - Ju Liu
- Department of Rheumatism and Immunology of The First People's Hospital, Jiujiang, Jiangxi 332000, P.R. China
| | - Xueyong Li
- Department of Rheumatism and Immunology of The First People's Hospital, Jiujiang, Jiangxi 332000, P.R. China
| | - Jin Lei
- Department of Rheumatism and Immunology of The First People's Hospital, Jiujiang, Jiangxi 332000, P.R. China
| | - Mingliang Qiu
- Department of Rheumatology, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
27
|
Ye SM, Zhou MZ, Jiang WJ, Liu CX, Zhou ZW, Sun MJ, Hu QH. Silencing of Gasdermin D by siRNA-Loaded PEI-Chol Lipopolymers Potently Relieves Acute Gouty Arthritis through Inhibiting Pyroptosis. Mol Pharm 2020; 18:667-678. [PMID: 32579365 DOI: 10.1021/acs.molpharmaceut.0c00229] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gasdermin D (GSDMD) plays a causal role in NOD-like receptor protein 3 (NLRP3) inflammasome-mediated pyroptosis eruption, which has been regarded as a potential therapeutic target for pyroptosis-related diseases including acute gouty arthritis. In the present study, the synthesized PEI-Chol (cholesterol grafted polyethylenimine) was assembled with GSDMD small interfering RNA (siRNA) to form PEI-Chol/siGSDMD polyplexes, which provided high transfection efficiency for siRNA-mediated GSDMD knockdown. Then we evaluated the effect of GSDMD siRNA-loaded PEI-Chol on inflammatory cascades in bone-marrow-derived macrophages (BMDMs) and acute gouty arthritis animal models under MSU exposure. When accompanied by pyroptosis blockade and decreased release of interleukin-1 beta (IL-1β), NLRP3 inflammasome activation was also suppressed by GSDMD knockdown in vivo and in vitro. Moreover, in MSU-induced acute gouty arthritis mice, blocking GSDMD with siRNA significantly improved ankle swelling and inflammatory infiltration observed in histopathological analysis. Furthermore, investigation using a mouse air pouch model verified the effect of siGSDMD-loaded PEI-Chol on pyroptosis of recruited macrophages and related signaling pathways in response to MSU. These novel findings exhibited that GSDMD knockdown relieved acute gouty arthritis through inhibiting pyroptosis, providing a possible therapeutic approach for MSU-induced acute gouty arthritis molecular therapy using PEI-Chol as a nucleic acid delivery carrier.
Collapse
Affiliation(s)
- Shu-Min Ye
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China.,School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PRChina
| | - Meng-Ze Zhou
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wen-Jiao Jiang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Chun-Xiao Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China.,School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PRChina
| | - Zhan-Wei Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PRChina
| | - Min-Jie Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PRChina
| | - Qing-Hua Hu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, PR China.,School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PRChina
| |
Collapse
|
28
|
Li H, Jiang W, Ye S, Zhou M, Liu C, Yang X, Hao K, Hu Q. P2Y 14 receptor has a critical role in acute gouty arthritis by regulating pyroptosis of macrophages. Cell Death Dis 2020; 11:394. [PMID: 32457291 PMCID: PMC7250907 DOI: 10.1038/s41419-020-2609-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/11/2020] [Accepted: 05/14/2020] [Indexed: 12/11/2022]
Abstract
Nod-like receptor protein 3 (NLRP3)-mediated pyroptosis has a causal role in the pathogenesis of gout. P2Y14 receptor (P2Y14R) distributed in immune cells including macrophages is a Gi-coupled receptor that inhibits the synthesis of cAMP, which has been regarded as a potential regulator of inflammatory response. Nevertheless, the role of P2Y14R in MSU-induced pyroptosis of macrophages involved in acute gouty arthritis is still unclear. In our present study, P2Y14R knockout (P2Y14R-KO) disrupted MSU-induced histopathologic changes in rat synoviums, accompanied with a significant inhibition of pyroptotic cell death characterized by Caspase-1/PI double-positive and blockade of NLRP3 inflammasome activation in synovial tissues, which was consistent with that observed in in vitro studies. Owing to the interaction of NLRP3 inflammasome and cAMP, we then investigated the effect of adenylate cyclase activator (Forskolin) on macrophage pyroptosis and gout flare caused by MSU stimulation. The reversal effect of Forskolin verified the negative regulatory role of cAMP in MSU-induced pyroptosis. More importantly, adenylate cyclase inhibitor (SQ22536) intervention led to a reversal of protection attributed to P2Y14R deficiency. Findings in air pouch animal models also verified aforementioned experimental results. Our study first identified the role of P2Y14R/cAMP/NLRP3 signaling pathway in acute gouty arthritis, which provides a novel insight into the pathological mechanisms of pyroptosis-related diseases.
Collapse
Affiliation(s)
- Hanwen Li
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China.,Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Wenjiao Jiang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Shumin Ye
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China.,Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Mengze Zhou
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Chunxiao Liu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China.,Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Xiping Yang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China.,Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Kun Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Qinghua Hu
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, PR China. .,Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China.
| |
Collapse
|
29
|
Klück V, Jansen TLTA, Janssen M, Comarniceanu A, Efdé M, Tengesdal IW, Schraa K, Cleophas MCP, Scribner CL, Skouras DB, Marchetti C, Dinarello CA, Joosten LAB. Dapansutrile, an oral selective NLRP3 inflammasome inhibitor, for treatment of gout flares: an open-label, dose-adaptive, proof-of-concept, phase 2a trial. THE LANCET. RHEUMATOLOGY 2020; 2:e270-e280. [PMID: 33005902 PMCID: PMC7523621 DOI: 10.1016/s2665-9913(20)30065-5] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
SUMMARY BACKGROUND Gout flares are driven by interleukin (IL)-1β. Dapansutrile inhibits the NLRP3 inflammasome and subsequent activation of IL-1β. In this study we aimed to investigate the safety and efficacy of orally administered dapansutrile in patients with a gout flare. METHODS In this open-label, proof-of-concept, phase 2a trial, adult patients (aged 18-80 years) with a monoarticular monosodium urate crystal-proven gout flare were enrolled at an outpatient clinic in the Netherlands and sequentially assigned using a dose-adaptive design to receive 100 mg/day, 300 mg/day, 1000 mg/day, or 2000 mg/day oral dapansutrile for 8 days. The coprimary outcomes were change in patient-reported target joint pain from baseline to day 3 and from baseline to day 7, assessed in the per-protocol population (all patients who received at least 80% of the study drug and had no major protocol deviations). Safety was assessed in the intention-to-treat population. This trial is registered with the EU Clinical Trials Register, EudraCT 2016-000943-14, and is completed. FINDINGS Between May 18, 2017, and Jan 21, 2019, 144 patients were assessed for eligibility, of whom 34 were enrolled and 29 were included in the per-protocol population (three patients were excluded due to receiving <80% of study drug and two had major protocol deviations): eight patients received 100 mg/day, seven received 300 mg/day, six received 1000 mg/day, and eight received 2000 mg/day. Between baseline and day 3, there was a mean reduction in patient-reported target joint pain of 52·4% (SD 32·94; p=0∙016) for the 100 mg/day group, 68·4% (34·29; p=0∙016) for the 300 mg/day group, 55·8% (44·90; p=0∙063) for the 1000 mg/day group, and 57·6% (38·72; p=0∙016) for the 2000 mg/day group. At day 7, there was a mean reduction of 82·1% (22·68; p=0∙031) for the 100 mg/day group, 84·2% (16·33; p=0∙016) for the 300 mg/day group, 68·9% (34·89; p=0∙031) for the 1000 mg/day group, and 83·9% (15·44; p=0∙008) for the 2000 mg/day group, compared to baseline. 25 (73·5%) of 34 patients reported a total of 45 treatment-emergent adverse events, most of which were metabolism and nutrition disorders (17 [37·8%]) and gastrointestinal disorders (ten [22·2%]). Two serious adverse events occurred during the study, admission to hospital because of worsening of gout flare at day 3, and admission to hospital because of coronary stenosis 18 days after the patient received their last dose; these were considered moderate in severity and unrelated to the study drug. INTERPRETATION Dapansutrile is a specific NLRP3 inflammasome inhibitor with a satisfactory safety profile and efficacy in the reduction of target joint pain in this study. Future studies are needed to confirm the clinical potential of dapansutrile.
Collapse
Affiliation(s)
| | | | - Matthijs Janssen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Antoaneta Comarniceanu
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Monique Efdé
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Isak W Tengesdal
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Kiki Schraa
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Maartje C P Cleophas
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Curtis L Scribner
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Damaris B Skouras
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Carlo Marchetti
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Charles A Dinarello
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands (V Klück MD, K Schraa BSc, M C P Cleophas PhD, Prof C A Dinarello MD, Prof L A B Joosten PhD); Department of Rheumatology, VieCuri Medical Center, Venlo, Netherlands (T L Th A Jansen PhD, M Janssen PhD, A Comarniceanu MD, M Efdé MD); Olatec Therapeutics, New York, NY, USA (C L Scribner MD, D B Skouras MBA); Department of Medicine, University of Colorado, Aurora, CO, USA (I W Tengesdal MSc, C Marchetti PhD, Prof C A Dinarello); and Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania (Prof L A B Joosten)
| |
Collapse
|
30
|
Oliviero F, Bindoli S, Scanu A, Feist E, Doria A, Galozzi P, Sfriso P. Autoinflammatory Mechanisms in Crystal-Induced Arthritis. Front Med (Lausanne) 2020; 7:166. [PMID: 32426360 PMCID: PMC7203538 DOI: 10.3389/fmed.2020.00166] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
Crystal-induced arthritides have been classified as “type-1 autoinflammatory diseases” for their main features which resemble those of the monogenic autoinflammatory syndromes. They are in fact characterized by spontaneous onset, recurrence of the episodes, self-limitation and resolution, inflammasome activation with huge production of IL-1β and a prevalent involvement of the innate immune system. The term “auto” refers also to the induction of IL-1β gene expression, processing and secretion by IL-1β itself. The concept of autoinflammation in crystal-induced arthritis has been finally reinforced by the efficacy of IL-1 blockade in treating acute and chronic state of this disease. The aim of this article is to review the autoinflammatory mechanisms in crystal-induced arthritis, considering both clinical and molecular aspects.
Collapse
Affiliation(s)
- Francesca Oliviero
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Sara Bindoli
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Anna Scanu
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Eugen Feist
- Department of Rheumatology, Cooperation Partner of the Otto-von-Guericke, University Magdeburg, Helios Clinic, Vogelsang-Gommern, Germany
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Paola Galozzi
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| | - Paolo Sfriso
- Rheumatology Unit, Department of Medicine-DIMED, University of Padova, Padova, Italy
| |
Collapse
|
31
|
Joosten LAB, Crişan TO, Bjornstad P, Johnson RJ. Asymptomatic hyperuricaemia: a silent activator of the innate immune system. Nat Rev Rheumatol 2020; 16:75-86. [PMID: 31822862 PMCID: PMC7075706 DOI: 10.1038/s41584-019-0334-3] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2019] [Indexed: 12/22/2022]
Abstract
Asymptomatic hyperuricaemia affects ~20% of the general population in the USA, with variable rates in other countries. Historically, asymptomatic hyperuricaemia was considered a benign laboratory finding with little clinical importance in the absence of gout or kidney stones. Yet, increasing evidence suggests that asymptomatic hyperuricaemia can predict the development of hypertension, obesity, diabetes mellitus and chronic kidney disease and might contribute to disease by stimulating inflammation. Although urate has been classically viewed as an antioxidant with beneficial effects, new data suggest that both crystalline and soluble urate activate various pro-inflammatory pathways. This Review summarizes what is known about the role of urate in the inflammatory response. Further research is needed to define the role of asymptomatic hyperuricaemia in these pro-inflammatory pathways.
Collapse
Affiliation(s)
- Leo A B Joosten
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Internal Medicine and Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Tania O Crişan
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Petter Bjornstad
- Department of Medicine of the University of Colorado School of Medicine of the University Hospital, Aurora, CO, USA
| | - Richard J Johnson
- Department of Medicine of the University of Colorado School of Medicine of the University Hospital, Aurora, CO, USA.
| |
Collapse
|
32
|
Mariotte A, De Cauwer A, Po C, Abou-Faycal C, Pichot A, Paul N, Aouadi I, Carapito R, Frisch B, Macquin C, Chatelus E, Sibilia J, Armspach JP, Bahram S, Georgel P. A mouse model of MSU-induced acute inflammation in vivo suggests imiquimod-dependent targeting of Il-1β as relevant therapy for gout patients. Am J Cancer Res 2020; 10:2158-2171. [PMID: 32104502 PMCID: PMC7019178 DOI: 10.7150/thno.40650] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 10/24/2019] [Indexed: 12/11/2022] Open
Abstract
Rationale: The role of Monosodium Urate (MSU) crystals in gout pathophysiology is well described, as is the major impact of IL-1β in the inflammatory reaction that constitutes the hallmark of the disease. However, despite the discovery of the NLRP3 inflammasome and its role as a Pattern Recognition Receptor linking the detection of a danger signal (MSU) to IL-1β secretion in vitro, the precise mechanisms leading to joint inflammation in gout patients are still poorly understood. Methods: Acute urate crystal inflammation was obtained by subcutaneous injections of MSU crystals in mice. Symptoms were followed by scoring, cytokine quantification by ELISA and western blot, gene expression by RT-qPCR and RNAseq; Magnetic Resonance Imaging was also used to assess inflammation. Results: We provide an extensive clinical, biological and molecular characterization of an acute uratic inflammation mouse model which accurately mimics human gout. We report the efficacy of topical imiquimod treatment and its impact on Interferon-dependent down modulation of Il-1β gene expression in this experimental model. Conclusion: Our work reveals several key features of MSU-dependent inflammation and identifies novel therapeutic opportunities for gout patients.
Collapse
|
33
|
Scott BM, Sheffield WP. Engineering the serpin α 1 -antitrypsin: A diversity of goals and techniques. Protein Sci 2019; 29:856-871. [PMID: 31774589 DOI: 10.1002/pro.3794] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/19/2019] [Accepted: 11/19/2019] [Indexed: 12/19/2022]
Abstract
α1 -Antitrypsin (α1 -AT) serves as an archetypal example for the serine proteinase inhibitor (serpin) protein family and has been used as a scaffold for protein engineering for >35 years. Techniques used to engineer α1 -AT include targeted mutagenesis, protein fusions, phage display, glycoengineering, and consensus protein design. The goals of engineering have also been diverse, ranging from understanding serpin structure-function relationships, to the design of more potent or more specific proteinase inhibitors with potential therapeutic relevance. Here we summarize the history of these protein engineering efforts, describing the techniques applied to engineer α1 -AT, specific mutants of interest, and providing an appended catalog of the >200 α1 -AT mutants published to date.
Collapse
Affiliation(s)
- Benjamin M Scott
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland.,Biosystems and Biomaterials Division, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - William P Sheffield
- Canadian Blood Services, Centre for Innovation, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
34
|
Pye A, Turner AM. Experimental and investigational drugs for the treatment of alpha-1 antitrypsin deficiency. Expert Opin Investig Drugs 2019; 28:891-902. [PMID: 31550938 DOI: 10.1080/13543784.2019.1672656] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Introduction: Alpha-1 antitrypsin deficiency (AATD) is most often associated with chronic lung disease, early onset emphysema, and liver disease. The standard of care in lung disease due to AATD is alpha-1 antitrypsin augmentation but there are several new and emerging treatment options under investigation for both lung and liver manifestations. Areas covered: We review therapeutic approaches to lung and liver disease in alpha-1 antitrypsin deficiency (AATD) and the agents in clinical development according to their mode of action. The focus is on products in clinical trials, but data from pre-clinical studies are described where relevant, particularly where progression to trials appears likely. Expert opinion: Clinical trials directed at lung and liver disease separately are now taking place. Multimodality treatment may be the future, but this could be limited by treatment costs. The next 5-10 years may reveal new guidance on when to use therapeutics for slowing disease progression with personalized treatment regimes coming to the forefront.
Collapse
Affiliation(s)
- Anita Pye
- Institute of Applied Health Research, University of Birmingham , Birmingham , UK
| | - Alice M Turner
- Institute of Applied Health Research, University of Birmingham , Birmingham , UK
| |
Collapse
|
35
|
The IL-1 family of cytokines and receptors in rheumatic diseases. Nat Rev Rheumatol 2019; 15:612-632. [DOI: 10.1038/s41584-019-0277-8] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
|
36
|
Szekanecz Z, Szamosi S, Kovács GE, Kocsis E, Benkő S. The NLRP3 inflammasome - interleukin 1 pathway as a therapeutic target in gout. Arch Biochem Biophys 2019; 670:82-93. [DOI: 10.1016/j.abb.2019.01.031] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/22/2019] [Accepted: 01/27/2019] [Indexed: 02/07/2023]
|
37
|
Nasonov EL. The role of interleukin 1 in the development of human diseases. RHEUMATOLOGY SCIENCE AND PRACTICE 2019. [DOI: 10.14412/1995-4484-2018-19-27] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Human immuno-inflammatory diseases (IID), depending on the predominant mechanisms of immune activation, are divided into two main categories: autoimmune and autoinflammatory. It is assumed that hyperproduction of "proinflammatory" and immunoregulatory cytokine-interleukin 1 (IL 1) largely determines the "intersection" between the mechanisms underlying autoimmunity and autoinflammation in many IID. This review discusses the role of IL1 in the pathogenesis of IID, primarily those associated with the activation of NLRP3-inflammasome, and therapeutic perspectives of IL1β inhibition with monoclonal antibodies to IL1β – canakinumab. The study of the IL1 role in the regulation of interactions between innate (TLR activation, inflammasome) and adaptive (Th1 – and Th17-types of immune response) immunity and the efficacy of IL1 inhibitors may be important in terms of decoding the pathogenetic mechanisms of IID and the development of new approaches to personalized therapy.
Collapse
Affiliation(s)
- E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology;
I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia
| |
Collapse
|
38
|
Abstract
A significant part of patients with gout has contraindications to taking nonsteroidal anti-inflammatory drugs, colchicine and glucocorticoids. Such therapy is often ineffective, particularly in patients with the severe tophaceous gout what hampers treatment of acute arthritis attack assuming the need for other methods of therapy. During the last years several medications have been introduced the mechanism of anti-inflammatory action of which is associated with inhibition of interleukin 1 (IL1) playing a key role in the development of acute gouty arthritis. To date, the most well-studied and the only registered drug for relief of acute arthritis attack is canakinumab, recommended for use in situations where other therapy options are unacceptable. Despite these limitations, the use of IL1 inhibitors, in particular canakinumab, seems promising due to the high efficiency of the drug, the ability to use it in patients with comorbid diseases, as well as a favorable effect on the risk of cardiovascular disease.
Collapse
Affiliation(s)
| | - E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology; I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia
| |
Collapse
|
39
|
Kaner Z, Engelman R, Schuster R, Rider P, Greenberg D, Av-Gay Y, Benhar M, Lewis EC. S-Nitrosylation of α1-Antitrypsin Triggers Macrophages Toward Inflammatory Phenotype and Enhances Intra-Cellular Bacteria Elimination. Front Immunol 2019; 10:590. [PMID: 31001247 PMCID: PMC6454134 DOI: 10.3389/fimmu.2019.00590] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/05/2019] [Indexed: 01/01/2023] Open
Abstract
Background: Human α1-antitrypsin (hAAT) is a circulating anti-inflammatory serine-protease inhibitor that rises during acute phase responses. in vivo, hAAT reduces bacterial load, without directly inhibiting bacterial growth. In conditions of excess nitric-oxide (NO), hAAT undergoes S-nitrosylation (S-NO-hAAT) and gains antibacterial capacity. The impact of S-NO-hAAT on immune cells has yet to be explored. Aim: Study the effects of S-NO-hAAT on immune cells during bacterial infection. Methods: Clinical-grade hAAT was S-nitrosylated and then compared to unmodified hAAT, functionally, and structurally. Intracellular bacterial clearance by THP-1 macrophages was assessed using live Salmonella typhi. Murine peritoneal macrophages were examined, and signaling pathways were evaluated. S-NO-hAAT was also investigated after blocking free mambranal cysteine residues on cells. Results: S-NO-hAAT (27.5 uM) enhances intracellular bacteria elimination by immunocytes (up to 1-log reduction). S-NO-hAAT causes resting macrophages to exhibit a pro-inflammatory and antibacterial phenotype, including release of inflammatory cytokines and induction of inducible nitric oxide synthase (iNOS) and TLR2. These pro-inflammatory effects are dependent upon cell surface thiols and activation of MAPK pathways. Conclusions: hAAT duality appears to be context-specific, involving S-nitrosylation in a nitric oxide rich environment. Our results suggest that S-nitrosylation facilitates the antibacterial activity of hAAT by promoting its ability to activate innate immune cells. This pro-inflammatory effect may involve transferring of nitric oxide from S-NO-hAAT to a free cysteine residue on cellular targets.
Collapse
Affiliation(s)
- Ziv Kaner
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Rotem Engelman
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ronen Schuster
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Peleg Rider
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - David Greenberg
- The Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer-Sheva, Israel
| | - Yossef Av-Gay
- Division of Infectious Diseases, Departments of Medicine and Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Moran Benhar
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Eli C Lewis
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
40
|
Mirea AM, Tack CJ, Chavakis T, Joosten LAB, Toonen EJM. IL-1 Family Cytokine Pathways Underlying NAFLD: Towards New Treatment Strategies. Trends Mol Med 2018; 24:458-471. [PMID: 29665983 PMCID: PMC5939989 DOI: 10.1016/j.molmed.2018.03.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/11/2018] [Accepted: 03/12/2018] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease worldwide. Pathways responsible for the activation of IL-1 family cytokines are key in the development of NAFLD but underlying mechanisms are not fully understood. Many studies have focused on the inflammasome-caspase-1 pathway and have shown that this pathway is an important inducer of inflammation in NAFLD. However, this pathway is not solely responsible for the activation of proinflammatory cytokines. Also, neutrophil serine proteases (NSPs) are capable of activating cytokines and recent studies reported that these proteases also contribute to NAFLD. These studies provided, for the first time, evidence that this inflammasome-independent pathway is involved in NAFLD. In our opinion, these new insights open up new approaches for therapeutic intervention.
Collapse
Affiliation(s)
- Andreea-Manuela Mirea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erik J M Toonen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; R&D Department, Hycult Biotech, Uden, The Netherlands.
| |
Collapse
|
41
|
Li MH, Xiao R, Li JB, Zhu Q. Regenerative approaches for cartilage repair in the treatment of osteoarthritis. Osteoarthritis Cartilage 2017; 25:1577-1587. [PMID: 28705606 DOI: 10.1016/j.joca.2017.07.004] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/09/2017] [Accepted: 07/01/2017] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) as a debilitating affliction of joints currently affects millions of people and remains an unsolved problem. The disease involves multiple cellular and molecular pathways that converge on the progressive destruction of cartilage. Activation of cartilage regenerative potential and specific targeting pathogenic mediators have been the major focus of research efforts aimed at slowing the progression of cartilage degeneration and preserve joint function. This review will summarize recent key discoveries toward better understanding of the complex mechanisms behind OA development and highlight the latest advances in basic and clinical research in the approach for cartilage regeneration. Prospectively, more potent therapeutic strategies against progressive cartilage deterioration may use a combination of cytotherapy, pharmacotherapy, and bioscaffoldings for improved chondrogenic differentiation and stem/progenitor cell homing as well as the concomitant reduced enzymatic matrix degradation and inflammation. Further, treatments need to be provided with increased preciseness of targeted therapy. One might expect that the regenerative therapies could potentially control or even possibly cure OA if performed at early stages of the disease.
Collapse
Affiliation(s)
- M H Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - R Xiao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - J B Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Q Zhu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
42
|
Abstract
The acute symptoms of gout are triggered by the inflammatory response to monosodium urate crystals, mediated principally by macrophages and neutrophils. Innate immune pathways are of key importance in the pathogenesis of gout, in particular the activation of the NLRP3 inflammasome, which leads to the release of IL-1β and other pro-inflammatory cytokines. The orchestration of this pro-inflammatory cascade involves multiple intracellular and extracellular receptors and enzymes interacting with environmental influences that modulate the inflammatory state. Furthermore, the resolution of inflammation in gout is becoming better understood. This Review highlights recent advances in our understanding of both positive and negative regulatory pathways, as well as the genetic and environmental factors that modulate the inflammatory response. Some of these pathways can be manipulated and present novel therapeutic opportunities for the treatment of acute gout attacks.
Collapse
Affiliation(s)
- Alexander K So
- Service of Rheumatology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Avenue Pierre Decker 4, 1011 Lausanne, Switzerland
| | - Fabio Martinon
- Department of Biochemistry, University of Lausanne, 155 Chemin des Boveresses, 1066 Epalinges, Switzerland
| |
Collapse
|
43
|
Abstract
Acute gout arthritis flares contribute dominantly to gout-specific impaired health-related quality of life, representing a progressively increasing public health problem. Flares can be complex and expensive to treat, partly due to the frequent comorbidities. Unmet needs in gout management are more pressing given the markedly increasing gout flare hospital admission rates. In addition, chronic gouty arthritis can cause joint damage and functional impairment. This review addresses new knowledge on the basis for the marked, inherent variability of responses to deposited urate crystals, including the unpredictable and self-limited aspects of many gout flares. Specific topics reviewed include how innate immunity and two-signal inflammasome activation intersect with diet, metabolism, nutritional biosensing, the microbiome, and the phagocyte cytoskeleton and cell fate. The paper discusses the roles of endogenous constitutive regulators of inflammation, including certain nutritional biosensors, and emerging genetic and epigenetic factors. Recent advances in the basis of variability in responses to urate crystals in gout provide information about inflammatory arthritis, and have identified potential new targets and strategies for anti-inflammatory prevention and treatment of gouty arthritis.
Collapse
Affiliation(s)
- Robert Terkeltaub
- VA San Diego Healthcare System, 111K, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA. .,Department of Medicine, University of California San Diego, San Diego, CA, USA.
| |
Collapse
|
44
|
Netea MG, Balkwill F, Chonchol M, Cominelli F, Donath MY, Giamarellos-Bourboulis EJ, Golenbock D, Gresnigt MS, Heneka MT, Hoffman HM, Hotchkiss R, Joosten LA, Kastner DL, Korte M, Latz E, Libby P, Mandrup-Poulsen T, Mantovani A, Mills KHG, Nowak KL, O’Neill LA, Pickkers P, van der Poll T, Ridker PM, Schalkwijk J, Schwartz DA, Siegmund B, Steer CJ, Tilg H, van der Meer JW, van de Veerdonk FL, Dinarello CA. A guiding map for inflammation. Nat Immunol 2017; 18:826-831. [PMID: 28722720 PMCID: PMC5939996 DOI: 10.1038/ni.3790] [Citation(s) in RCA: 524] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Biologists, physicians and immunologists have contributed to the understanding of the cellular participants and biological pathways involved in inflammation. Here, we provide a general guide to the cellular and humoral contributors to inflammation as well as to the pathways that characterize inflammation in specific organs and tissues.
Collapse
Affiliation(s)
- Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania
| | - Frances Balkwill
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, UK
| | - Michel Chonchol
- Division of Renal Diseases and Hypertension, University of Colorado, Denver, USA
| | - Fabio Cominelli
- Digestive Health Research Institute, Case Western Reserve University, Cleveland, OH, USA
| | - Marc Y. Donath
- Clinic of Endocrinology, Diabetes and Metabolism University Hospital and University of Basel, Switzerland
| | | | - Douglas Golenbock
- Division of Infectious Diseases and Immunology, University of Massacchussetts Medical School, Worchester, USA
| | - Mark S. Gresnigt
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michael T. Heneka
- Department of Neurodegenerative Disease and Gerontopsychiatry/Neurology, University of Bonn, Bonn, Germany
| | - Hal M. Hoffman
- Division of Pediatric Allergy, Immunology, and Rheumatology, University of California at San Diego and Rady Children’s Hospital of San Diego, USA
| | - Richard Hotchkiss
- Department of Anesthesiology, Medicine, and Surgery, Washington University School of Medicine, St Louis, MO, USA
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca Romania
| | - Daniel L. Kastner
- Inflammatory Disease Section, Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Martin Korte
- TU Braunschweig, Zoological Institute, Braunschweig, Germany and HZI, AG NIND, Braunschweig, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn, Germany
- Department of Infectious Diseases & Immunology, UMass Medical School, Worcester, MA, USA
| | - Peter Libby
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Alberto Mantovani
- Humanitas University and Humanitas Clinica Research Center, Rozzano, Milano, Italy
| | - Kingston H. G. Mills
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Kristen L. Nowak
- Division of Renal Diseases and Hypertension, University of Colorado, Denver, USA
| | - Luke A. O’Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Paul M. Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Joost Schalkwijk
- Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - David A. Schwartz
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Denver, USA
| | - Britta Siegmund
- Department of Medicine (Gastroenterology, Infectious Diseases, Rheumatology), Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Clifford J. Steer
- Departments of Medicine and Genetics, Cell Biology and Development, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology & Endocrinology, Medical University Innsbruck, Austria
| | - Jos W.M. van der Meer
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frank L. van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Charles A. Dinarello
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
45
|
Dayer JM, Oliviero F, Punzi L. A Brief History of IL-1 and IL-1 Ra in Rheumatology. Front Pharmacol 2017; 8:293. [PMID: 28588495 PMCID: PMC5440542 DOI: 10.3389/fphar.2017.00293] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 05/08/2017] [Indexed: 11/13/2022] Open
Abstract
The history of what, in 1979, was called interleukin-1 (IL-1), orchestrator of leukocyte inter-communication, began many years before then, initially by the observation of fever induction via the endogenous pyrogen (EP) (1974) and then in rheumatology on the role in tissue destruction in rheumatoid diseases via the induction of collagenase and PGE2 in human synovial cells by a mononuclear cell factor (MCF) (1977). Since then, the family has exploded to presently 11 members as well as many membrane-bound and soluble receptor forms. The discovery of a natural Interleukin-1 receptor antagonist (IL-1Ra) in human biological fluids has highlighted the importance of IL-1 and IL-1Ra in human diseases. Evidence delineating its role in autoinflammatory syndromes and the elucidation of the macromolecular complex referred to as "inflammasome" have been instrumental to our understanding of the link with IL-1. At present, the IL-1blockade as therapeutic approach is crucial for many hereditary autoinflammatory diseases, as well as for adult-onset Still's disease, crystal-induced arthropathies, certain skin diseases including neutrophil-triggered skin diseases, Behçet's disease and deficiency of IL-1Ra and other rare fever syndromes. Its role is only marginally important in rheumatoid arthritis and is still under debate with regard to osteoarthritis, type 2 diabetes mellitus, cardiovascular diseases and cancer. This brief historical review focuses on some aspects of IL-1, mainly IL-1β and IL-Ra, in rheumatology. There are many excellent reviews focusing on the IL-1 family in general or with regard to specific diseases or biological discoveries.
Collapse
Affiliation(s)
| | | | - Leonardo Punzi
- Department of Medicine, University of PadovaPadova, Italy
| |
Collapse
|
46
|
Uric acid priming in human monocytes is driven by the AKT-PRAS40 autophagy pathway. Proc Natl Acad Sci U S A 2017; 114:5485-5490. [PMID: 28484006 DOI: 10.1073/pnas.1620910114] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Metabolic triggers are important inducers of the inflammatory processes in gout. Whereas the high serum urate levels observed in patients with gout predispose them to the formation of monosodium urate (MSU) crystals, soluble urate also primes for inflammatory signals in cells responding to gout-related stimuli, but also in other common metabolic diseases. In this study, we investigated the mechanisms through which uric acid selectively lowers human blood monocyte production of the natural inhibitor IL-1 receptor antagonist (IL-1Ra) and shifts production toward the highly inflammatory IL-1β. Monocytes from healthy volunteers were first primed with uric acid for 24 h and then subjected to stimulation with lipopolysaccharide (LPS) in the presence or absence of MSU. Transcriptomic analysis revealed broad inflammatory pathways associated with uric acid priming, with NF-κB and mammalian target of rapamycin (mTOR) signaling strongly increased. Functional validation did not identify NF-κB or AMP-activated protein kinase phosphorylation, but uric acid priming induced phosphorylation of AKT and proline-rich AKT substrate 40 kDa (PRAS 40), which in turn activated mTOR. Subsequently, Western blot for the autophagic structure LC3-I and LC3-II (microtubule-associated protein 1A/1B-light chain 3) fractions, as well as fluorescence microscopy of LC3-GFP-overexpressing HeLa cells, revealed lower autophagic activity in cells exposed to uric acid compared with control conditions. Interestingly, reactive oxygen species production was diminished by uric acid priming. Thus, the Akt-PRAS40 pathway is activated by uric acid, which inhibits autophagy and recapitulates the uric acid-induced proinflammatory cytokine phenotype.
Collapse
|
47
|
Ter Horst R, Jaeger M, Smeekens SP, Oosting M, Swertz MA, Li Y, Kumar V, Diavatopoulos DA, Jansen AFM, Lemmers H, Toenhake-Dijkstra H, van Herwaarden AE, Janssen M, van der Molen RG, Joosten I, Sweep FCGJ, Smit JW, Netea-Maier RT, Koenders MMJF, Xavier RJ, van der Meer JWM, Dinarello CA, Pavelka N, Wijmenga C, Notebaart RA, Joosten LAB, Netea MG. Host and Environmental Factors Influencing Individual Human Cytokine Responses. Cell 2017; 167:1111-1124.e13. [PMID: 27814508 DOI: 10.1016/j.cell.2016.10.018] [Citation(s) in RCA: 319] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/03/2016] [Accepted: 10/11/2016] [Indexed: 02/08/2023]
Abstract
Differences in susceptibility to immune-mediated diseases are determined by variability in immune responses. In three studies within the Human Functional Genomics Project, we assessed the effect of environmental and non-genetic host factors of the genetic make-up of the host and of the intestinal microbiome on the cytokine responses in humans. We analyzed the association of these factors with circulating mediators and with six cytokines after stimulation with 19 bacterial, fungal, viral, and non-microbial metabolic stimuli in 534 healthy subjects. In this first study, we show a strong impact of non-genetic host factors (e.g., age and gender) on cytokine production and circulating mediators. Additionally, annual seasonality is found to be an important environmental factor influencing cytokine production. Alpha-1-antitrypsin concentrations partially mediate the seasonality of cytokine responses, whereas the effect of vitamin D levels is limited. The complete dataset has been made publicly available as a comprehensive resource for future studies. PAPERCLIP.
Collapse
Affiliation(s)
- Rob Ter Horst
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Martin Jaeger
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Sanne P Smeekens
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Marije Oosting
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Morris A Swertz
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Groningen 9700RB, the Netherlands
| | - Yang Li
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Groningen 9700RB, the Netherlands
| | - Vinod Kumar
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Groningen 9700RB, the Netherlands
| | - Dimitri A Diavatopoulos
- Laboratory of Pediatric Infectious Diseases and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Anne F M Jansen
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Heidi Lemmers
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Helga Toenhake-Dijkstra
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Antonius E van Herwaarden
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Matthijs Janssen
- Department of Rheumatology, Rijnstate Hospital, Arnhem, Gelderland 6815AD, the Netherlands
| | - Renate G van der Molen
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Irma Joosten
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Fred C G J Sweep
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Johannes W Smit
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands; Division of Endocrinology, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Romana T Netea-Maier
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands; Division of Endocrinology, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Mieke M J F Koenders
- Elkerliek Hospital, Clinical Chemistry, Helmond, Noord-Brabant 5700AB, the Netherlands
| | - Ramnik J Xavier
- Broad Institute of Massachusetts Institute of Technology (MIT), Cambridge, MA 02142, USA; Harvard University, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02139, USA
| | - Jos W M van der Meer
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Charles A Dinarello
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands; Division of Medicine, University of Colorado Denver, Aurora, CO 80045, USA
| | - Norman Pavelka
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands; Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore 138648, Singapore
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Groningen 9700RB, the Netherlands; Centre for Immune Regulation and Department of Immunology, University of Oslo, Oslo University Hospital, Oslo, Oslo 0027, Norway
| | - Richard A Notebaart
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands.
| | - Mihai G Netea
- Department of Internal Medicine and Radboudumc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Gelderland 6500HB, the Netherlands.
| |
Collapse
|
48
|
Oliviero F, Scanu A. How Factors Involved in the Resolution of Crystal-Induced Inflammation Target IL-1β. Front Pharmacol 2017; 8:164. [PMID: 28400732 PMCID: PMC5368178 DOI: 10.3389/fphar.2017.00164] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/13/2017] [Indexed: 12/12/2022] Open
Abstract
One of the main clinical features characterizing crystal-induced inflammation is its spontaneous resolution. The aim of this review is to outline the various factors involved in the self-limiting course of crystal-induced inflammation focusing on their effect on IL-1β production. Endogenous molecules that are induced or locally recruited by the process itself, inhibitory proteins naturally present in the joint and exogenous dietary factors are discussed. Aside from the classical well-known molecules involved in the resolution of crystal-induced acute attack such as TGFβ, IL-10, IL-1Ra, and lipoproteins, particular attention is paid to recently uncovered mechanisms such as the aggregation of neutrophil extracellular traps, the release of ectosomes from neutrophil surface, and alpha-1-anti-trypsin-mediated IL-1 inhibition.
Collapse
Affiliation(s)
- Francesca Oliviero
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova Padova, Italy
| | - Anna Scanu
- Rheumatology Unit, Department of Medicine - DIMED, University of Padova Padova, Italy
| |
Collapse
|
49
|
|
50
|
Kaneva MK, Greco KV, Headland SE, Montero-Melendez T, Mori P, Greenslade K, Pitzalis C, Moore A, Perretti M. Identification of Novel Chondroprotective Mediators in Resolving Inflammatory Exudates. THE JOURNAL OF IMMUNOLOGY 2017; 198:2876-2885. [PMID: 28242648 DOI: 10.4049/jimmunol.1601111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/29/2017] [Indexed: 12/11/2022]
Abstract
We hypothesized that exudates collected at the beginning of the resolution phase of inflammation might be enriched for tissue protective molecules; thus an integrated cellular and molecular approach was applied to identify novel chondroprotective bioactions. Exudates were collected 6 h (inflammatory) and 24 h (resolving) following carrageenan-induced pleurisy in rats. The resolving exudate was subjected to gel filtration chromatography followed by proteomics, identifying 61 proteins. Fractions were added to C28/I2 chondrocytes, grown in micromasses, ions with or without IL-1β or osteoarthritic synovial fluids for 48 h. Three proteins were selected from the proteomic analysis, α1-antitrypsin (AAT), hemopexin (HX), and gelsolin (GSN), and tested against catabolic stimulation for their effects on glycosaminoglycan deposition as assessed by Alcian blue staining, and gene expression of key anabolic proteins by real-time PCR. In an in vivo model of inflammatory arthritis, cartilage integrity was determined histologically 48 h after intra-articular injection of AAT or GSN. The resolving exudate displayed protective activities on chondrocytes, using multiple readouts: these effects were retained in low m.w. fractions of the exudate (46.7% increase in glycosaminoglycan deposition; ∼20% upregulation of COL2A1 and aggrecan mRNA expression), which reversed the effect of IL-1β. Exogenous administration of HX, GSN, or AAT abrogated the effects of IL-1β and osteoarthritic synovial fluids on anabolic gene expression and increased glycosaminoglycan deposition. Intra-articular injection of AAT or GSN protected cartilage integrity in mice with inflammatory arthritis. In summary, the strategy for identification of novel chondroprotective activities in resolving exudates identified HX, GSN and AAT as potential leads for new drug discovery programs.
Collapse
Affiliation(s)
- Magdalena K Kaneva
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| | - Karin V Greco
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| | - Sarah E Headland
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| | - Trinidad Montero-Melendez
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| | | | | | - Costantino Pitzalis
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| | | | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom; and
| |
Collapse
|