1
|
Almeida DADF, Brant CF, da Costa Siqueira L, Soares LFF, Oliveira JA, de Oliveira DSB, Pigossi SC, Soares CJ. Genetic polymorphisms on temporomandibular disorders: Network meta-analysis. Arch Oral Biol 2025; 174:106235. [PMID: 40107000 DOI: 10.1016/j.archoralbio.2025.106235] [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: 08/03/2024] [Revised: 02/13/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVE The aim of this systematic review and network meta-analysis (NMA) is to compare and rank the effects of different genetic polymorphisms on the susceptibility of temporomandibular disorders (TMDs) occurrence. DESIGN The central question formulated was: "Are genetic polymorphisms involved in the etiology of TMDs?" Following PROSPERO registration (CRD42024507886), electronic searches were conducted in five databases for publications up to November 2024. RESULTS Sixty-three studies were included in the systematic review and 7 composed the NMA. The qualitative analysis summarized the association between 120 genes (and 206 polymorphisms) and TMDs. Thirty-two polymorphisms (in 24 genes) were linked to overall TMDs, while 22 polymorphisms (in 22 genes) with degenerative bone changes in the temporomandibular joint (TMJ). Additionally, 17 polymorphisms were identified in cases of painful chronic TMD, while 12 polymorphisms in intra-articular disorders. These polymorphisms were in genes related to neurotransmission (COMT, ADRB2, DRD2, ANKK1, SLC6A4 and HTR2A), inflammatory mediators (TNFα, IL10 and MMP1), sex hormones (ESR1and ESRRB), oxidative stress (GSTM1) and bone metabolism (VDR). A protective effect for myalgia occurrence with the COMT_rs165774 polymorphism compared to the wild-type genotype was found in the pairwise meta-analysis (AG genotype: OR: 0.33; 95 %CI: 0.14, 0.76; p < 0.01 and GG genotype: OR: 0.32; 95 %CI: 0.14, 0.74; p < 0.01) and this polymorphism showed the highest probability of being associated with the myalgia (97 %) and arthralgia (93 %) conditions. CONCLUSIONS Genetic polymorphisms in genes related to neurotransmission, inflammatory response, and sex hormones seem to be risk factors related to the TMDs pathogenesis.
Collapse
Affiliation(s)
- Daniel Augusto de Faria Almeida
- School of Dentistry, Alfenas Federal University (Unifal-MG), Alfenas, Minas Gerais, Brazil; Department of Operative Dentistry and Dental Materials, School of Dentistry, Federal University of Uberlândia - UFU, Uberlândia, Minas Gerais, Brazil.
| | - Camila Freire Brant
- School of Dentistry, Alfenas Federal University (Unifal-MG), Alfenas, Minas Gerais, Brazil; Department of Periodontology and Implantodontology, School of Dentistry, Federal University of Uberlândia - UFU, Uberlândia, Minas Gerais, Brazil
| | | | - Lélio Fernando Ferreira Soares
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State of University (FOAR/UNESP), São Paulo, Brazil
| | - Jovânia Alves Oliveira
- Department of Periodontology and Implantodontology, School of Dentistry, Federal University of Uberlândia - UFU, Uberlândia, Minas Gerais, Brazil
| | | | - Suzane Cristina Pigossi
- Department of Periodontology and Implantodontology, School of Dentistry, Federal University of Uberlândia - UFU, Uberlândia, Minas Gerais, Brazil
| | - Carlos José Soares
- Department of Operative Dentistry and Dental Materials, School of Dentistry, Federal University of Uberlândia - UFU, Uberlândia, Minas Gerais, Brazil
| |
Collapse
|
2
|
Study protocol for a randomised controlled trial of diacerein versus placebo to treat knee osteoarthritis with effusion-synovitis (DICKENS). Trials 2022; 23:768. [PMID: 36089595 PMCID: PMC9464426 DOI: 10.1186/s13063-022-06715-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/04/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
There is an unmet need for treatments for knee osteoarthritis (OA). Effusion-synovitis is a common inflammatory phenotype of knee OA and predicts knee pain and structural degradation. Anti-inflammatory therapies, such as diacerein, may be effective for this phenotype. While diacerein is recommended for alleviating pain in OA patients, evidence for its effectiveness is inconsistent, possibly because studies have not targeted patients with an inflammatory phenotype. Therefore, we will conduct a multi-centre, randomised, placebo-controlled double-blind trial to determine the effect of diacerein on changes in knee pain and effusion-synovitis over 24 weeks in patients with knee OA and magnetic resonance imaging (MRI)-defined effusion-synovitis.
Methods
We will recruit 260 patients with clinical knee OA, significant knee pain, and MRI-detected effusion-synovitis in Hobart, Melbourne, Adelaide, and Perth, Australia. They will be randomly allocated to receive either diacerein (50mg twice daily) or identical placebo for 24 weeks. MRI of the study knee will be performed at screening and after 24 weeks of intervention. The primary outcome is improvement in knee pain at 24 weeks as assessed by a 100-mm visual analogue scale (VAS). Secondary outcomes include improvement in volumetric (ml) and semi-quantitative (Whole-Organ Magnetic Resonance Imaging Score, 0–3) measurements of effusion-synovitis using MRI over 24 weeks, and improvement in knee pain (VAS) at 4, 8, 12, 16, and 20 weeks. Intention-to-treat analyses of primary and secondary outcomes will be performed as the primary analyses. Per protocol analyses will be performed as the secondary analyses.
Discussion
This study will provide high-quality evidence to determine whether diacerein improves pain, changes disease trajectory, and slows disease progression in OA patients with effusion-synovitis. If diacerein proves effective, this has the potential to significantly benefit the substantial proportion (up to 60%) of knee OA patients with an inflammatory phenotype.
Trial registration
Australian and New Zealand Clinical Trial Registry ACTRN12618001656224. Registered on 08 October 2018.
Collapse
|
3
|
Liu S, Deng Z, Chen K, Jian S, Zhou F, Yang Y, Fu Z, Xie H, Xiong J, Zhu W. Cartilage tissue engineering: From proinflammatory and anti‑inflammatory cytokines to osteoarthritis treatments (Review). Mol Med Rep 2022; 25:99. [PMID: 35088882 PMCID: PMC8809050 DOI: 10.3892/mmr.2022.12615] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/07/2021] [Indexed: 12/17/2022] Open
Abstract
Osteoarthritis (OA), one of the most common joint diseases, is characterized by fibrosis, rhagadia, ulcers and attrition of articular cartilage due to a number of factors. The etiology of OA remains unclear, but its occurrence has been associated with age, obesity, inflammation, trauma and genetic factors. Inflammatory cytokines are crucial for the occurrence and progression of OA. The intra-articular proinflammatory and anti-inflammatory cytokines jointly maintain a dynamic balance, in accordance with the physiological metabolism of articular cartilage. However, dynamic imbalance between proinflammatory and anti-inflammatory cytokines can cause abnormal metabolism in knee articular cartilage, which leads to deformation, loss and abnormal regeneration, and ultimately destroys the normal structure of the knee joint. The ability of articular cartilage to self-repair once damaged is limited, due to its inability to obtain nutrients from blood vessels, nerves and lymphatic vessels, as well as limitations in the extracellular matrix. There are several disadvantages inherent to conventional repair methods, while cartilage tissue engineering (CTE), which combines proinflammatory and anti-inflammatory cytokines, offers a new therapeutic approach for OA. The aim of the present review was to examine the proinflammatory factors implicated in OA, including IL-1β, TNF-α, IL-6, IL-15, IL-17 and IL-18, as well as the key anti-inflammatory factors reducing OA-related articular damage, including IL-4, insulin-like growth factor and TGF-β. The predominance of proinflammatory over anti-inflammatory cytokine effects ultimately leads to the development of OA. CTE, which employs mesenchymal stem cells and scaffolding technology, may prevent OA by maintaining the homeostasis of pro- and anti-inflammatory factors.
Collapse
Affiliation(s)
- Shuyu Liu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Kang Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Shengsheng Jian
- Department of Orthopedics, Luo Hu Hospital, Shenzhen, Guangdong 518001, P.R. China
| | - Feifei Zhou
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Yuan Yang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Zicai Fu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Huanyu Xie
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Jianyi Xiong
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| | - Weimin Zhu
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong 518035, P.R. China
| |
Collapse
|
4
|
Kim M, Choe YH, Lee SI. Lessons From the Success and Failure of Targeted Drugs for Rheumatoid Arthritis: Perspectives for Effective Basic and Translational Research. Immune Netw 2022; 22:e8. [PMID: 35291656 PMCID: PMC8901706 DOI: 10.4110/in.2022.22.e8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 12/01/2022] Open
Affiliation(s)
- Mingyo Kim
- Division of Rheumatology, Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju 52727, Korea
| | - Yong-ho Choe
- Division of Rheumatology, Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju 52727, Korea
| | - Sang-il Lee
- Division of Rheumatology, Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju 52727, Korea
| |
Collapse
|
5
|
Larsson PF, Karlsson R, Sarwar M, Miftakhova R, Wang T, Syed Khaja AS, Semenas J, Chen S, Hedblom A, Ali A, Ekström‐Holka K, Simoulis A, Kumar A, Wingren AG, Robinson B, Nyunt Wai S, Mongan NP, Heery DM, Öhlund D, Grundström T, Ødum N, Persson JL. FcγRIIIa receptor interacts with androgen receptor and PIP5K1α to promote growth and metastasis of prostate cancer. Mol Oncol 2021; 16:2496-2517. [PMID: 34932854 PMCID: PMC9251882 DOI: 10.1002/1878-0261.13166] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/23/2021] [Accepted: 12/20/2021] [Indexed: 11/08/2022] Open
Abstract
Low‐affinity immunoglobulin gamma Fc region receptor III‐A (FcγRIIIa) is a cell surface protein that belongs to a family of Fc receptors that facilitate the protective function of the immune system against pathogens. However, the role of FcγRIIIa in prostate cancer (PCa) progression remained unknown. In this study, we found that FcγRIIIa expression was present in PCa cells and its level was significantly higher in metastatic lesions than in primary tumors from the PCa cohort (P = 0.006). PCa patients with an elevated level of FcγRIIIa expression had poorer biochemical recurrence (BCR)‐free survival compared with those with lower FcγRIIIa expression, suggesting that FcγRIIIa is of clinical importance in PCa. We demonstrated that overexpression of FcγRIIIa increased the proliferative ability of PCa cell line C4‐2 cells, which was accompanied by the upregulation of androgen receptor (AR) and phosphatidylinositol‐4‐phosphate 5‐kinase alpha (PIP5Kα), which are the key players in controlling PCa progression. Conversely, targeted inhibition of FcγRIIIa via siRNA‐mediated knockdown or using its inhibitory antibody suppressed growth of xenograft PC‐3 and PC‐3M prostate tumors and reduced distant metastasis in xenograft mouse models. We further showed that elevated expression of AR enhanced FcγRIIIa expression, whereas inhibition of AR activity using enzalutamide led to a significant downregulation of FcγRIIIa protein expression. Similarly, inhibition of PIP5K1α decreased FcγRIIIa expression in PCa cells. FcγRIIIa physically interacted with PIP5K1α and AR via formation of protein–protein complexes, suggesting that FcγRIIIa is functionally associated with AR and PIP5K1α in PCa cells. Our study identified FcγRIIIa as an important factor in promoting PCa growth and invasion. Further, the elevated activation of FcγRIII and AR and PIP5K1α pathways may cooperatively promote PCa growth and invasion. Thus, FcγRIIIa may serve as a potential new target for improved treatment of metastatic and castration‐resistant PCa.
Collapse
Affiliation(s)
| | - Richard Karlsson
- Department of Molecular Biology Umeå University Umeå Sweden
- Division of Experimental Cancer Research Department of Translational Medicine Lund University Clinical Research Centre Malmö Sweden
| | - Martuza Sarwar
- Department of Molecular Biology Umeå University Umeå Sweden
| | | | - Tianyan Wang
- Department of Molecular Biology Umeå University Umeå Sweden
| | | | - Julius Semenas
- Department of Molecular Biology Umeå University Umeå Sweden
| | - Sa Chen
- Department of Molecular Biology Umeå University Umeå Sweden
| | - Andreas Hedblom
- Department of Molecular Biology Umeå University Umeå Sweden
- Division of Experimental Cancer Research Department of Translational Medicine Lund University Clinical Research Centre Malmö Sweden
| | - Amjad Ali
- Department of Molecular Biology Umeå University Umeå Sweden
| | | | - Athanasios Simoulis
- Department of Clinical Pathology and Cytology Skåne University Hospital Malmö Sweden
| | - Anjani Kumar
- Department of Molecular Biology Umeå University Umeå Sweden
| | | | - Brian Robinson
- Department of Pathology Weill Cornell Medical College New York NY USA
| | - Sun Nyunt Wai
- Department of Molecular Biology Umeå University Umeå Sweden
- Umeå Centre for Microbial Research (UCMR) Umeå University Umeå Sweden
| | - Nigel P Mongan
- Faculty of Medicine and Health Sciences School of Veterinary Medicine and Sciences
| | - David M Heery
- School of Pharmacy University of Nottingham Nottingham United Kingdom
| | - Daniel Öhlund
- Wallenberg Centre for Molecular Medicine, and Department of Radiation Sciences Umeå University Umeå Sweden
| | | | - Niels Ødum
- Department of Immunology and Microbiology University of Copenhagen Copenhagen Denmark
| | - Jenny L Persson
- Department of Molecular Biology Umeå University Umeå Sweden
- Division of Experimental Cancer Research Department of Translational Medicine Lund University Clinical Research Centre Malmö Sweden
- Faculty of Biomedicine Malmö University Malmö Sweden
| |
Collapse
|
6
|
Zhu G, Zhang T, Chen M, Yao K, Huang X, Zhang B, Li Y, Liu J, Wang Y, Zhao Z. Bone physiological microenvironment and healing mechanism: Basis for future bone-tissue engineering scaffolds. Bioact Mater 2021; 6:4110-4140. [PMID: 33997497 PMCID: PMC8091181 DOI: 10.1016/j.bioactmat.2021.03.043] [Citation(s) in RCA: 208] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/19/2021] [Accepted: 03/28/2021] [Indexed: 02/06/2023] Open
Abstract
Bone-tissue defects affect millions of people worldwide. Despite being common treatment approaches, autologous and allogeneic bone grafting have not achieved the ideal therapeutic effect. This has prompted researchers to explore novel bone-regeneration methods. In recent decades, the development of bone tissue engineering (BTE) scaffolds has been leading the forefront of this field. As researchers have provided deep insights into bone physiology and the bone-healing mechanism, various biomimicking and bioinspired BTE scaffolds have been reported. Now it is necessary to review the progress of natural bone physiology and bone healing mechanism, which will provide more valuable enlightenments for researchers in this field. This work details the physiological microenvironment of the natural bone tissue, bone-healing process, and various biomolecules involved therein. Next, according to the bone physiological microenvironment and the delivery of bioactive factors based on the bone-healing mechanism, it elaborates the biomimetic design of a scaffold, highlighting the designing of BTE scaffolds according to bone biology and providing the rationale for designing next-generation BTE scaffolds that conform to natural bone healing and regeneration.
Collapse
Affiliation(s)
- Guanyin Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Tianxu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Miao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Ke Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Xinqi Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Bo Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Yazhen Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Jun Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610041, PR China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| |
Collapse
|
7
|
Gupta R, Kapoor A, Khatri S, Sandal D, Masih GD. There is an Association of Synovial Interleukin-6 Levels With Chondral Damage in Anterior Cruciate Ligament-Deficient Knees. HSS J 2021; 17:145-149. [PMID: 34421423 PMCID: PMC8361593 DOI: 10.1177/1556331621992006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 11/15/2022]
Abstract
Background: Osteoarthritis (OA) in the anterior cruciate ligament (ACL)-deficient knee is seen in approximately 50% of affected patients. Possible causes include biochemical or biomechanical changes. Purpose: We sought to study the correlation between inflammatory cytokines and chondral damage in ACL-deficient knees. Methods: Seventy-six male patients who underwent ACL reconstruction were enrolled in a cross-sectional study. Synovial fluid was aspirated before surgery and analyzed for levels of the inflammatory cytokines tumor necrosis factor-α, interleukin-1 (IL-1), and interleukin-6 (IL-6). At the time of ACL reconstruction, the severity of chondral damage was documented as described by the Outerbridge classification. Results: Patients with grade 2 or higher chondral damage were observed to have elevated IL-6 levels when compared to patients who had no chondral damage. Interleukin-6 levels had no correlation with the duration of injury. Conclusion: Elevated levels of IL-6 in synovial fluid were associated with chondral damage in ACL-deficient knees. Further study is warranted to determine whether inflammatory cytokines contribute to the development of OA of the knee after ACL injury.
Collapse
Affiliation(s)
- Ravi Gupta
- Department of Orthopaedics, Government Medical College & Hospital, Chandigarh, India
| | - Anil Kapoor
- Department of Orthopaedics, Government Medical College & Hospital, Chandigarh, India,Anil Kapoor, MBBS, MS, MRCS, Department of Orthopaedics, Government Medical College & Hospital, Chandigarh 160030, India.
| | - Sourabh Khatri
- All India Institute of Medical Sciences, Rishikesh, India
| | - Dinesh Sandal
- Department of Orthopaedics, Government Medical College & Hospital, Chandigarh, India
| | - Gladson David Masih
- Department of Orthopaedics, Government Medical College & Hospital, Chandigarh, India
| |
Collapse
|
8
|
Cheon SY, Lee JE. Extracellular Vesicles and Immune System in Ageing and Immune Diseases. Exp Neurobiol 2021; 30:32-47. [PMID: 33632983 PMCID: PMC7926047 DOI: 10.5607/en20059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/06/2021] [Accepted: 01/17/2021] [Indexed: 02/06/2023] Open
Abstract
Immune system is essential for host homeostasis. Immune cells communicate with each other by binding to receptors or by releasing vesicles including chemokines and cytokines. Under healthy circumstances, immune cell-derived factors are critical for cellular growth, division and function, whereas under conditions such as ageing and inflammatory states, they can aggravate pathologies and cause disease. Cell-derived membranous extracellular vesicles mediate cell-to-cell communication and are implicated in various physiological and pathological processes involving ageing and age-related diseases. Extracellular vesicles are responsible for spreading detrimental factors to the surroundings and the propagation phase of inflammatory diseases. The regulation of extracellular vesicles is a putative target for treatment of inflammatory diseases. Moreover, their features are ideal for developing biomarkers and drug delivery systems modulated by bioengineering in inflammatory diseases. The present review summarizes the current understanding of extracellular vesicles in ageing and inflammatory diseases.
Collapse
Affiliation(s)
- So Yeong Cheon
- Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju 27478, Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Korea
- BK21 PLUS Project for Medical Science, and Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
9
|
Association of Estrogen Receptor 1 and Tumor Necrosis Factor α Polymorphisms with Temporomandibular Joint Anterior Disc Displacement without Reduction. DISEASE MARKERS 2020; 2020:6351817. [PMID: 33101543 PMCID: PMC7576356 DOI: 10.1155/2020/6351817] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 09/02/2020] [Accepted: 09/22/2020] [Indexed: 12/20/2022]
Abstract
Objectives The aim of this study was to investigate the role of ESR1 rs1643821 and TNF-α rs1800629 as potential genetic factors regulating anterior disc displacement without reduction-mediated inflammatory pathway. Background The temporomandibular joint is a complex synovial joint that allows mandibular movement in three directions. Although temporomandibular disorders are widespread, limited data is available on the biochemical characteristics of the displaced disc and quality of the surrounding soft tissue. Changes in degenerative tissue provoke disc displacement which involves secretion of inflammatory markers and sequential conversion of fibroblast-like cells into chondrocyte-like cells. Due to the high occurrence in female adolescents, the potential role of sex hormones in temporomandibular joint disorders has been speculated. Furthermore, anterior disc displacement without reduction severely affects the quality of life. Methods 124 Caucasian patients with a history of at least one anterior disc displacement without reduction within 3 months were enrolled. Anterior disc displacement without reduction was diagnosed based on clinical examination, diagnostic criteria (DC)/TMD, and cone-beam computed tomography/magnetic resonance imaging (CBCT/MRI). The control group consisted of 126 patients with no temporomandibular joint disorders. Genotyping of two single nucleotide polymorphisms, estrogen receptor 1 (ESR1) rs1643821, and tumor necrosis factor α (TNF-α) rs1800629 was performed. Results ESR1 rs1643821 showed significant P values (using chi-square analysis) revealing the difference in anterior disc displacement without reduction frequencies while TNF-α rs1800629 polymorphism was found to be statistically insignificant when compared to the control group. Furthermore, patients with a genotype of ESR1 rs1643821 showed a decreased probability (OR = 0.412) against anterior disc displacement without reduction when compared to the GG genotype (OR = 1). Conclusion ESR1 rs1643821 with A allele frequency was lower in patients with anterior disc displacement without reduction compared to the control group. Thus, the rs1643821 variant is significantly associated with susceptibility to the anterior disc displacement without a reduction in European Caucasians. Conversely, TNF-α rs1800629 was a statistically insignificant factor against anterior disc displacement without reduction when compared to the control group.
Collapse
|
10
|
Neferine Inhibits Expression of Inflammatory Mediators and Matrix Degrading Enzymes in IL-1β-Treated Rat Chondrocytes via Suppressing MAPK and NF-κB Signaling Pathways. Inflammation 2020; 43:1209-1221. [DOI: 10.1007/s10753-019-01143-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
11
|
Topping LM, Thomas BL, Rhys HI, Tremoleda JL, Foster M, Seed M, Voisin MB, Vinci C, Law HL, Perretti M, Norling LV, Azevedo HS, Nissim A. Targeting Extracellular Vesicles to the Arthritic Joint Using a Damaged Cartilage-Specific Antibody. Front Immunol 2020; 11:10. [PMID: 32117219 PMCID: PMC7033748 DOI: 10.3389/fimmu.2020.00010] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/06/2020] [Indexed: 01/18/2023] Open
Abstract
The targeted delivery of therapies to diseased tissues offers a safe opportunity to achieve optimal efficacy while limiting systemic exposure. These considerations apply to many disease indications but are especially relevant for rheumatoid arthritis (RA), as RA is a systemic autoimmune disease which affects multiple joints. We have identified an antibody that is specific to damaged arthritic cartilage (anti-ROS-CII) that can be used to deliver treatments specifically to arthritic joints, yielding augmented efficacy in experimental arthritis. In the current study, we demonstrate that scaffolds enriched with bioactive payloads can be delivered precisely to an inflamed joint and achieve superior efficacy outcomes consistent with the pharmacological properties of these payloads. As a scaffold, we have used extracellular vesicles (EVs) prepared from human neutrophils (PMNs), which possess intrinsic anti-inflammatory properties and the ability to penetrate inflamed arthritic cartilage. EV fortified with anti-ROS-CII (EV/anti-ROS-CII) retained anti-ROS-CII specificity and bound exclusively to the damaged cartilage. Following systemic administration, EV/anti-ROS-CII (a) exhibited the ability to localize specifically in the arthritic joint in vivo and (b) was able to specifically target single (viral IL-10 or anti-TNF) or combined (viral IL-10 and anti-TNF) anti-inflammatory treatments to the arthritic joint, which accelerated attenuation of clinical and synovial inflammation. Overall, this study demonstrates the attainability of targeting a pro-resolving biological scaffold to the arthritic joint. The potential of targeting scaffolds such as EV, nanoparticles, or a combination thereof alongside combined therapeutics is paramount for designing systemically administered broad-spectrum of anti-inflammatory treatments.
Collapse
Affiliation(s)
- Louise M Topping
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Centre for Bioengineering, Life Sciences, Queen Mary University of London, London, United Kingdom
| | - Bethan L Thomas
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Hefin I Rhys
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Jordi L Tremoleda
- Barts and the London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Martyn Foster
- Experimental Pathology Consultancy, London, United Kingdom
| | - Michael Seed
- School of Health Sport and Bioscience, University of East London, London, United Kingdom
| | - Mathieu-Benoit Voisin
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Chiara Vinci
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Hannah L Law
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Mauro Perretti
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Centre for Bioengineering, Life Sciences, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Lucy V Norling
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| | - Helena S Azevedo
- Centre for Bioengineering, Life Sciences, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom.,School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary University of London, London, United Kingdom
| | - Ahuva Nissim
- Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.,Centre for Bioengineering, Life Sciences, Queen Mary University of London, London, United Kingdom.,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
12
|
Vitamin D as a Principal Factor in Mediating Rheumatoid Arthritis-Derived Immune Response. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3494937. [PMID: 31205940 PMCID: PMC6530219 DOI: 10.1155/2019/3494937] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic multifactorial autoimmune disorder. The interactions between diverse environmental and genetic factors lead to the onset of this complex autoimmune disorder. Serum levels of vitamin D (VD) are involved in the regulation of various immune responses. Vitamin D is a key signaling molecule in the human body that maintains calcium as well as phosphate homeostasis. It also regulates the functions of the immune system and, thus, can play a substantial role in the etiology of various autoimmune disorders, including RA. Low serum VD levels have been found to be associated with a higher risk of RA, although this finding has not been replicated consistently. The molecular mechanisms by which VD influences autoimmunity need to be further explored to understand how variation in plasma VD levels could affect the pathogenesis of RA. This mini-review focuses on the influence of VD and its serum levels on RA susceptibility, RA-associated complexities, treatment, and transcriptome products of key proinflammatory cytokines, along with other cytokines that are key regulators of inflammation in rheumatoid joints.
Collapse
|
13
|
Stefani RM, Halder SS, Estell EG, Lee AJ, Silverstein AM, Sobczak E, Chahine NO, Ateshian GA, Shah RP, Hung CT. A Functional Tissue-Engineered Synovium Model to Study Osteoarthritis Progression and Treatment. Tissue Eng Part A 2019; 25:538-553. [PMID: 30203722 PMCID: PMC6482911 DOI: 10.1089/ten.tea.2018.0142] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 08/31/2018] [Indexed: 01/15/2023] Open
Abstract
IMPACT STATEMENT The synovium envelops the diarthrodial joint and plays a key regulatory role in defining the composition of the synovial fluid through filtration and biosynthesis of critical boundary lubricants. Synovium changes often precede cartilage damage in osteoarthritis. We describe a novel in vitro tissue engineered model, validated against native synovium explants, to investigate the structure-function of synovium through quantitative solute transport measures. Synovium was evaluated in the presence of a proinflammatory cytokine, interleukin-1, or the clinically relevant corticosteroid, dexamethasone. We anticipate that a better understanding of synovium transport would support efforts to develop more effective strategies aimed at restoring joint health.
Collapse
Affiliation(s)
- Robert M. Stefani
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Saiti S. Halder
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Eben G. Estell
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Andy J. Lee
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Amy M. Silverstein
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Evie Sobczak
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Nadeen O. Chahine
- Department of Biomedical Engineering, Columbia University, New York, New York
- Department of Orthopedic Surgery, Columbia University, New York, New York
| | - Gerard A. Ateshian
- Department of Biomedical Engineering, Columbia University, New York, New York
- Department of Mechanical Engineering, Columbia University, New York, New York
| | - Roshan P. Shah
- Department of Orthopedic Surgery, Columbia University, New York, New York
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, New York
| |
Collapse
|
14
|
Jones IA, Togashi R, Wilson ML, Heckmann N, Vangsness CT. Intra-articular treatment options for knee osteoarthritis. Nat Rev Rheumatol 2019; 15:77-90. [PMID: 30498258 PMCID: PMC6390843 DOI: 10.1038/s41584-018-0123-4] [Citation(s) in RCA: 335] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Intra-articular drug delivery has a number of advantages over systemic administration; however, for the past 20 years, intra-articular treatment options for the management of knee osteoarthritis (OA) have been limited to analgesics, glucocorticoids, hyaluronic acid (HA) and a small number of unproven alternative therapies. Although HA and glucocorticoids can provide clinically meaningful benefits to an appreciable number of patients, emerging evidence indicates that the apparent effectiveness of these treatments is largely a result of other factors, including the placebo effect. Biologic drugs that target inflammatory processes are used to manage rheumatoid arthritis, but have not translated well into use in OA. A lack of high-level evidence and methodological limitations hinder our understanding of so-called 'stem' cell therapies and, although the off-label administration of intra-articular cell therapies (such as platelet-rich plasma and bone marrow aspirate concentrate) is common, high-quality clinical data are needed before these treatments can be recommended. A number of promising intra-articular treatments are currently in clinical development in the United States, including small-molecule and biologic therapies, devices and gene therapies. Although the prospect of new, non-surgical treatments for OA is exciting, the benefits of new treatments must be carefully weighed against their costs and potential risks.
Collapse
Affiliation(s)
- Ian A Jones
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Ryan Togashi
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Melissa L Wilson
- Department of Preventive Medicine, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Nathanael Heckmann
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - C Thomas Vangsness
- Department of Orthopaedic Surgery, Keck School of Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Abstract
The increase in global lifespan has in turn increased the prevalence of osteoarthritis which is now the most common type of arthritis. Cartilage tissue located on articular joints erodes during osteoarthritis which causes pain and may lead to a crippling loss of function in patients. The pathophysiology of osteoarthritis has been understudied and currently no disease modifying treatments exist. The only current end-point treatment remains joint replacement surgery. The primary risk factor for osteoarthritis is age. Clinical and basic research is now focused on understanding the ageing process of cartilage and its role in osteoarthritis. This chapter will outline the physiology of cartilage tissue, the clinical presentation and treatment options for the disease and the cellular ageing processes which are involved in the pathophysiology of the disease.
Collapse
|
16
|
Collins KH, Herzog W, MacDonald GZ, Reimer RA, Rios JL, Smith IC, Zernicke RF, Hart DA. Obesity, Metabolic Syndrome, and Musculoskeletal Disease: Common Inflammatory Pathways Suggest a Central Role for Loss of Muscle Integrity. Front Physiol 2018; 9:112. [PMID: 29527173 PMCID: PMC5829464 DOI: 10.3389/fphys.2018.00112] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/05/2018] [Indexed: 01/14/2023] Open
Abstract
Inflammation can arise in response to a variety of stimuli, including infectious agents, tissue injury, autoimmune diseases, and obesity. Some of these responses are acute and resolve, while others become chronic and exert a sustained impact on the host, systemically, or locally. Obesity is now recognized as a chronic low-grade, systemic inflammatory state that predisposes to other chronic conditions including metabolic syndrome (MetS). Although obesity has received considerable attention regarding its pathophysiological link to chronic cardiovascular conditions and type 2 diabetes, the musculoskeletal (MSK) complications (i.e., muscle, bone, tendon, and joints) that result from obesity-associated metabolic disturbances are less frequently interrogated. As musculoskeletal diseases can lead to the worsening of MetS, this underscores the imminent need to understand the cause and effect relations between the two, and the convergence between inflammatory pathways that contribute to MSK damage. Muscle mass is a key predictor of longevity in older adults, and obesity-induced sarcopenia is a significant risk factor for adverse health outcomes. Muscle is highly plastic, undergoes regular remodeling, and is responsible for the majority of total body glucose utilization, which when impaired leads to insulin resistance. Furthermore, impaired muscle integrity, defined as persistent muscle loss, intramuscular lipid accumulation, or connective tissue deposition, is a hallmark of metabolic dysfunction. In fact, many common inflammatory pathways have been implicated in the pathogenesis of the interrelated tissues of the musculoskeletal system (e.g., tendinopathy, osteoporosis, and osteoarthritis). Despite these similarities, these diseases are rarely evaluated in a comprehensive manner. The aim of this review is to summarize the common pathways that lead to musculoskeletal damage and disease that result from and contribute to MetS. We propose the overarching hypothesis that there is a central role for muscle damage with chronic exposure to an obesity-inducing diet. The inflammatory consequence of diet and muscle dysregulation can result in dysregulated tissue repair and an imbalance toward negative adaptation, resulting in regulatory failure and other musculoskeletal tissue damage. The commonalities support the conclusion that musculoskeletal pathology with MetS should be evaluated in a comprehensive and integrated manner to understand risk for other MSK-related conditions. Implications for conservative management strategies to regulate MetS are discussed, as are future research opportunities.
Collapse
Affiliation(s)
- Kelsey H. Collins
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Walter Herzog
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Graham Z. MacDonald
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Raylene A. Reimer
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Jaqueline L. Rios
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- CAPES Foundation, Brasilia, Brazil
| | - Ian C. Smith
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Ronald F. Zernicke
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Departments of Orthopaedic Surgery and Biomedical Engineering, School of Kinesiology, University of Michigan, Ann Arbor, MI, United States
- Department of Surgery, Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - David A. Hart
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
- Department of Family Practice, The Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC, Canada
- Alberta Health Services Bone and Joint Health Strategic Clinical Network, Calgary, AB, Canada
| |
Collapse
|
17
|
Peck Y, Leom LT, Low PFP, Wang DA. Establishment of an in vitro three-dimensional model for cartilage damage in rheumatoid arthritis. J Tissue Eng Regen Med 2017; 12:e237-e249. [PMID: 28079986 DOI: 10.1002/term.2399] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/15/2016] [Accepted: 01/09/2017] [Indexed: 11/09/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease that leads to progressive joint destruction. To further understand the process of rheumatoid cartilage damage, an in vitro model consisting of an interactive tri-culture of synovial fibroblasts (SFs), LPS-stimulated macrophages and a primary chondrocyte-based tissue-engineered construct was established. The tissue-engineered construct has a composition similar to that of human cartilage, which is rich in collagen type II and proteoglycans. Data generated from this model revealed that healthy chondrocytes were activated in the presence of SFs and macrophages. The activated chondrocytes subsequently displayed aberrant behaviours as seen in a disease state such as increased apoptosis, decreased gene expression for matrix components such as type II collagen and aggrecan, increased gene expression for tissue-degrading enzymes (MMP-1, -3, -13 and ADAMTS-4, -5), and upregulation of inflammatory mediator gene expression (TNF-α, IL-1β, IL-6 and IKBKB). Additionally, the inclusion of SFs and macrophages in the model enabled both cell types to more closely replicate an in vivo role in mediating cartilage destruction. This is evidenced by extensive matrix loss, detected in the model through immunostaining and biochemical analysis. Subsequent drug treatment with celecoxib has shown that the model was able to respond to the therapeutic effects of this drug by reversing cartilage damage. This study showed that the model was able to recapitulate certain pathological features of an RA cartilage. If properly validated, this model potentially can be used for screening new therapeutic drugs and strategies, thereby contributing to the improvement of anti-rheumatic treatment. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Yvonne Peck
- Division of BioEngineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Li Ting Leom
- Division of BioEngineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Pei Fen Patricia Low
- Division of BioEngineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Dong-An Wang
- Division of BioEngineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| |
Collapse
|
18
|
Martin P, Palmer G, Rodriguez E, Seemayer CA, Palomo J, Talabot-Ayer D, Gabay C. Deficiency in IL-1 Receptor Type 2 Aggravates K/BxN Serum Transfer-Induced Arthritis in Mice but Has No Impact on Systemic Inflammatory Responses. THE JOURNAL OF IMMUNOLOGY 2017; 198:2916-2926. [PMID: 28235865 DOI: 10.4049/jimmunol.1600855] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 01/30/2017] [Indexed: 01/27/2023]
Abstract
The biological activity of IL-1 is tightly regulated by the specific receptor antagonist (IL-1Ra) and the decoy receptor IL-1 receptor type 2 (IL-1R2). The role of IL-1Ra has been well demonstrated in IL-1Ra-deficient mice. In contrast, the role of endogenous IL-1R2 remains widely unknown. To define the functional role of endogenous IL-1R2 in the K/BxN serum transfer arthritis model and in IL-1β- or LPS-induced systemic inflammation in vivo, IL-1R2-/- mice were created and compared with wild type mice. IL-1R2-/- mice bred habitually and exhibited a normal phenotype. IL-1R2 deficiency aggravated arthritis severity and increased mRNA levels for key cytokines and chemokines such as IL-6, IL-1β, Cxcl-1, and Cxcl-2 significantly in ankles. There was no effect of IL-1R2 deficiency on the cell-autonomous cytokine response to IL-1β in the tested cell types, i.e., neutrophils, macrophages, and fibroblasts, but IL-1R2 deficiency on neutrophils increased the IL-1-induced response of fibroblasts in trans. Furthermore, IL-1β induced shedding of IL-1R2 in vivo. Inflammatory responses to IL-1β and LPS-induced mortality were not different in IL-1R2-/- compared with wild type mice. Our data demonstrate that the decoy receptor IL-1R2 plays an important inhibitory role in local IL-1- and neutrophil-dependent tissue inflammation as shown in the K/BxN serum transfer arthritis model. In contrast to IL-1Ra, IL-1R2 appears to be less crucial for systemic responses to acute administration of IL-1 or LPS.
Collapse
Affiliation(s)
- Praxedis Martin
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1211 Geneva 4, Switzerland.,Division of Rheumatology, University Hospital of Geneva, 1211 Geneva 4, Switzerland; and
| | - Gaby Palmer
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1211 Geneva 4, Switzerland.,Division of Rheumatology, University Hospital of Geneva, 1211 Geneva 4, Switzerland; and
| | - Emiliana Rodriguez
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1211 Geneva 4, Switzerland.,Division of Rheumatology, University Hospital of Geneva, 1211 Geneva 4, Switzerland; and
| | | | - Jennifer Palomo
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1211 Geneva 4, Switzerland.,Division of Rheumatology, University Hospital of Geneva, 1211 Geneva 4, Switzerland; and
| | - Dominique Talabot-Ayer
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1211 Geneva 4, Switzerland.,Division of Rheumatology, University Hospital of Geneva, 1211 Geneva 4, Switzerland; and
| | - Cem Gabay
- Department of Pathology and Immunology, School of Medicine, University of Geneva, 1211 Geneva 4, Switzerland; .,Division of Rheumatology, University Hospital of Geneva, 1211 Geneva 4, Switzerland; and
| |
Collapse
|
19
|
Noack M, Miossec P. Selected cytokine pathways in rheumatoid arthritis. Semin Immunopathol 2017; 39:365-383. [DOI: 10.1007/s00281-017-0619-z] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/31/2017] [Indexed: 12/13/2022]
|
20
|
Nicot R, Vieira AR, Raoul G, Delmotte C, Duhamel A, Ferri J, Sciote JJ. ENPP1 and ESR1 genotypes influence temporomandibular disorders development and surgical treatment response in dentofacial deformities. J Craniomaxillofac Surg 2016; 44:1226-37. [PMID: 27519661 DOI: 10.1016/j.jcms.2016.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 06/04/2016] [Accepted: 07/13/2016] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Dentofacial deformities are dys-morpho-functional disorders involving the temporomandibular joints (TMJ). Many authors have reported a TMJ improvement in dysfunctional subjects with malocclusion after orthodontic or combined orthodontic and surgical treatment particularly for the relief of pain. In particular, few studies have highlighted the demographic and clinical predictors of response to surgical treatment. To date, no genetic factor has yet been identified as a predictor of response to surgical treatment. The aim of this cohort study is therefore to identify single-nucleotide polymorphisms associated with postoperative temporomandibular disorders (TMD) or with TMJ symptoms after orthognathic surgery. Here, we found the AA genotype of SNP rs1643821 (ESR1 gene) as a risk factor for dysfunctional worsening after orthognathic surgery. In addition, we have identified TT genotype of SNP rs858339 (ENPP1 gene) as a protective factor against TMD in a population of patients with dentofacial deformities. Conversely, the heterozygous genotype AT was identified as a risk factor of TMD with respect to the rest of our population. All these elements are particularly important to bring new screening strategies and tailor future treatment. PERSPECTIVE This study allows us to identify sub-populations at high risk of developing postoperative temporomandibular disorders after orthognathic surgery procedures. Many other genes of interest could be potential factors influencing the dysfunctional response to orthognathic surgery, particularly genes of the Opera cohort.
Collapse
Affiliation(s)
- Romain Nicot
- Univ. Lille, Oral and Maxillofacial Department, Roger Salengro Hospital, CHU Lille, F-59000, Lille, France.
| | - Alexandre R Vieira
- Department of Oral Biology, University of Pittsburgh School of Dental Medicine, 3501 Terrace St, Pittsburgh, PA 15261, USA.
| | - Gwénaël Raoul
- Univ. Lille, Oral and Maxillofacial Department, Roger Salengro Hospital, CHU Lille, INSERM U 1008, Controlled Drug Delivery Systems and Biomaterials, F-59000, Lille, France.
| | - Constance Delmotte
- Univ. Lille, Oral and Maxillofacial Department, Roger Salengro Hospital, CHU Lille, F-59000, Lille, France.
| | - Alain Duhamel
- Univ. Lille, CHU Lille, EA 2694 - Santé publique: épidémiologie et qualité des soins, F-59000, Lille, France.
| | - Joël Ferri
- Univ. Lille, Oral and Maxillofacial Department, Roger Salengro Hospital, CHU Lille, INSERM U 1008, Controlled Drug Delivery Systems and Biomaterials, F-59000, Lille, France.
| | - James J Sciote
- Department of Orthodontics, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
21
|
Is Synovial Macrophage Activation the Inflammatory Link Between Obesity and Osteoarthritis? Curr Rheumatol Rep 2016; 18:57. [DOI: 10.1007/s11926-016-0605-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
22
|
Kour S, Garimella MG, Shiroor DA, Mhaske ST, Joshi SR, Singh K, Pal S, Mittal M, Krishnan HB, Chattopadhyay N, Ulemale AH, Wani MR. IL-3 Decreases Cartilage Degeneration by Downregulating Matrix Metalloproteinases and Reduces Joint Destruction in Osteoarthritic Mice. THE JOURNAL OF IMMUNOLOGY 2016; 196:5024-35. [PMID: 27183574 DOI: 10.4049/jimmunol.1500907] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 04/13/2016] [Indexed: 01/02/2023]
Abstract
Osteoarthritis (OA) is a chronic disease of articular joints that leads to degeneration of both cartilage and subchondral bone. These degenerative changes are further aggravated by proinflammatory cytokines including IL-1β and TNF-α. Previously, we have reported that IL-3, a cytokine secreted by activated T cells, protects cartilage and bone damage in murine models of inflammatory and rheumatoid arthritis. However, how IL-3 protects cartilage degeneration is not yet known. In this study, we investigated the role of IL-3 on cartilage degeneration under both in vitro and in vivo conditions. We found that both mouse and human chondrocytes show strong expression of IL-3R at gene and protein levels. IL-3 increases the expression of mouse chondrocyte-specific genes, Sox9 and collagen type IIa, which were downregulated by IL-1β. Moreover, IL-3 downregulated IL-1β- and TNF-α-induced expression of matrix metalloproteinases in both mouse and human chondrocytes. Interestingly, IL-3 reduces the degeneration of articular cartilage and subchondral bone microarchitecture in a mouse model of human OA. Moreover, IL-3 showed the preventive and therapeutic effects on cartilage degeneration induced by IL-1β in micromass pellet cultures of human mesenchymal stem cells. Thus, to our knowledge, we provide the first evidence that IL-3 has therapeutic potential in amelioration of degeneration of articular cartilage and subchondral bone microarchitecture associated with OA.
Collapse
Affiliation(s)
- Supinder Kour
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Manasa G Garimella
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Divya A Shiroor
- Department of Veterinary Surgery, Krantisinh Nana Patil College of Veterinary Science, Shirwal 412801, Satara, India
| | - Suhas T Mhaske
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Snehal R Joshi
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Kanupriya Singh
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411007, India
| | - Subhashis Pal
- Division of Endocrinology, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow 226031, India; and
| | - Monika Mittal
- Division of Endocrinology, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow 226031, India; and
| | - Hari B Krishnan
- Department of Orthopaedic Surgery, Armed Forces Medical College, Pune 411040, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow 226031, India; and
| | - Anil H Ulemale
- Department of Veterinary Surgery, Krantisinh Nana Patil College of Veterinary Science, Shirwal 412801, Satara, India
| | - Mohan R Wani
- National Centre for Cell Science, Savitribai Phule Pune University Campus, Ganeshkhind, Pune 411007, India;
| |
Collapse
|
23
|
Rainsford KD, Parke AL, Clifford-Rashotte M, Kean WF. Therapy and pharmacological properties of hydroxychloroquine and chloroquine in treatment of systemic lupus erythematosus, rheumatoid arthritis and related diseases. Inflammopharmacology 2015; 23:231-69. [PMID: 26246395 DOI: 10.1007/s10787-015-0239-y] [Citation(s) in RCA: 371] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 06/23/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVES This review examines the pharmacokinetics, modes of action and therapeutic properties of the anti-malarial drugs, hydroxychloroquine (HCQ) and chloroquine (CQ), in the treatment of systemic lupus erythematosus (SLE), rheumatoid arthritis (RA) and related conditions, as well as osteoarthritis (OA). KEY FINDINGS Both HCQ and CQ have historically been employed successfully for the treatment of SLE and RA for over 70 years. HCQ has been used extensively for SLE where it has a good reputation for controlling the dermatological complications in SLE. It has also been reported to effectively control the symptoms of Sjøgren's syndrome, as well as preventing thrombosis in phospholipid antibody (aPL) syndrome. In RA and SLE, HCQ is preferred because of the lower incidence of gastrointestinal adverse reactions compared with CQ and it might have a lower risk of ocular adverse reactions. There is increasing evidence that HCQ may reduce atherosclerosis and risks of cardiovascular disease in rheumatic patients. Both HCQ and CQ have been shown to improve glycaemia and reduce the risks of type II diabetes mellitus. Although both HCQ and CQ are effective in low-moderate RA, HCQ is now preferred as part of combination therapy for more severe disease. The advantages of combination therapy are that the doses of the individual drugs may be lowered so reducing adverse reactions. Both HCQ and CQ are diastereoisomers, have basic properties and are given as the sulphate and phosphate salts. While being relatively well absorbed orally and with good bioavailability, they have long and variable plasma terminal elimination half-lives (approximately 40-60 days). This reflects their high volume of distribution, V D (HCQ 44,000L; CQ 65,000L) which extends into aqueous compartments, long mean residence time (HCQ 1300 h; CQ 900 h) and with about half the drugs (metabolites) undergoing renal clearance. The strong binding to melanin reflects the ocular injury and dermatological properties of these drugs. The consensus is that the occurrence of ocular adverse reactions can be minimised by close attention to the dose (which should be set on a body weight basis) with regular (e.g. quarterly) retinal examination. Although HCQ and CQ can pass through the placenta, the use of these drugs during pregnancy does not appear to risk harm to the baby and might be beneficial to the mother with SLE and her child by controlling the SLE disease activity, which is known to be an important factor affecting pregnancy outcome. The modes of action of HCQ and CQ in these arthritides represent somewhat of an enigma. Undoubtedly, these drugs have multiple actions related, in part, their ability to accumulate in lysosomes and autophagosomes of phagocytic cells as well as affecting MHC Class II expression and antigen presentation; actions of the production of pro-inflammatory cytokines [e.g. interleukin-1 (IL-1) tumour necrosis factor-α (TNFα)]; control of toll-like receptor-9 activation; and leucocyte generation of reactive oxygen species (ROS); i.e. antioxidant activity. The actions of these drugs on T and B cells are less clear but may depend on these leucocyte-mediated actions. Anti-malarials also protect against cytokine-mediated cartilage resorption. This and other actions may underlie the potential benefits in treating OA. The exact relationships of these various actions, mostly determined in vitro, have not been specifically defined in vivo or ex vivo in relation to clinical efficacy. OUTCOMES HCQ and CQ have a good reputation for being effective and relatively safe treatments in SLE, mild-moderate RA and Sjøgren's syndrome. There is need for (a) more information on their mode of action in relation to the control of these diseases, (b) scope for developing formulations that have improved pharmacokinetic and therapeutic properties and safety, and (c) further exploring their use in drug combinations not only with other disease modifying agents but also with biologics.
Collapse
Affiliation(s)
- K D Rainsford
- Biomedical Research Centre, Sheffield Hallam University, Howard Street, Sheffield, S1 1WB, UK
| | - Ann L Parke
- Department of Rheumatology, St Francis Hospital and Medical Center, Hartford, CT, 06105, USA
| | | | - W F Kean
- Department of Medicine (Rheumatology), McMaster University Faculty of Health Sciences, Hamilton, ON, L8S 4K9, Canada.
- Department of Medicine (Rheumatology), McMaster University Faculty of Health Sciences, Suite #708, 1 Young Street, Hamilton, ON, L8N 1T8, Canada.
| |
Collapse
|
24
|
Zhu X, Song Y, Huo R, Zhang J, Sun S, He Y, Gao H, Zhang M, Sun X, Zhai T, Li H, Sun Y, Zhou Z, Shen B, Xiao L, Li N. Cyr61 participates in the pathogenesis of rheumatoid arthritis by promoting proIL-1β production by fibroblast-like synoviocytes through an AKT-dependent NF-κB signaling pathway. Clin Immunol 2015; 157:187-97. [PMID: 25728492 DOI: 10.1016/j.clim.2015.02.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/24/2014] [Accepted: 02/17/2015] [Indexed: 12/11/2022]
Abstract
IL-1β plays a major role in the development of rheumatoid arthritis (RA). We previously showed that Cyr61 participates in RA pathogenesis as a proinflammatory factor. Here, we found that the levels of IL-1β and Cyr61 were higher in RA SF than in osteoarthritis (OA) SF. IL-1β mRNA and proIL-1β protein levels were remarkably increased in Cyr61-stimulated FLS; however, IL-1β was hardly detectable in the supernatant. We also found that the level of adenosine triphosphate (ATP) in SF and ST was significantly increased in RA patients and that the level of IL-1β in supernatants from Cyr61-activated FLS increased significantly when we added exogenous ATP to the culture. Mechanistically, Cyr61 induced proIL-1β production in FLS via the AKT-dependent NF-κB signaling pathway, and ATP caused Cyr61-induced proIL-1β to generate IL-1β in a caspase-1-dependent manner. Our results reveal a novel role of Cyr61 in RA that involves the promotion of proIL-1β production in FLS.
Collapse
Affiliation(s)
- Xianjin Zhu
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Affiliated Union Hospital of Fujian Medical University, Fuzhou, PR China
| | - Yanfang Song
- Affiliated Renmin Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, PR China
| | - Rongfen Huo
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jie Zhang
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Songtao Sun
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, PR China
| | - Yong He
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, PR China
| | - Huali Gao
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, PR China
| | - Miaojia Zhang
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Xiaoxuan Sun
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China
| | - Tianhang Zhai
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Huidan Li
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yue Sun
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhou Zhou
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Baihua Shen
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Lianbo Xiao
- Institute of Arthritis Research, Shanghai Academy of Chinese Medical Sciences, Shanghai, PR China.
| | - Ningli Li
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
25
|
Role of genetic alterations in the NLRP3 and CARD8 genes in health and disease. Mediators Inflamm 2015; 2015:846782. [PMID: 25788762 PMCID: PMC4348606 DOI: 10.1155/2015/846782] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/05/2015] [Indexed: 12/30/2022] Open
Abstract
The complexity of a common inflammatory disease is influenced by multiple genetic and environmental factors contributing to the susceptibility of disease. Studies have reported that these exogenous and endogenous components may perturb the balance of innate immune response by activating the NLRP3 inflammasome. The multimeric NLRP3 complex results in the caspase-1 activation and the release of potent inflammatory cytokines, like IL-1β. Several studies have been performed on the association of the genetic alterations in genes encoding NLRP3 and CARD8 with the complex diseases with inflammatory background, like inflammatory bowel disease, cardiovascular diseases, rheumatoid arthritis, and type 1 diabetes. The aim of the present review is therefore to summarize the literature regarding genetic alterations in these genes and their association with health and disease.
Collapse
|
26
|
Elmallah RK, Cherian JJ, Jauregui JJ, Pierce TP, Beaver WB, Mont MA. Genetically modified chondrocytes expressing TGF-β1: a revolutionary treatment for articular cartilage damage? Expert Opin Biol Ther 2015; 15:455-64. [PMID: 25645308 DOI: 10.1517/14712598.2015.1009886] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Currently, joint arthroplasty remains the only definitive management of osteoarthritis, while other treatment modalities only provide temporary and symptomatic relief. The use of genetically engineered chondrocytes is currently undergoing clinical trials. Specifically, it has been designed to induce cartilage growth and differentiation in patients with degenerative arthritis, with the aim to play a curative role in the disease process. AREAS COVERED This treatment involves the incorporation of TGF-β1, which has been determined to play an influential role in chondrogenesis and extracellular matrix synthesis. Using genetic manipulation and viral transduction, TGF-β1 is incorporated into human chondrocytes and administered in a minimally invasive fashion directly to the affected joint. Following a database literature search, we evaluated the current evidence on this product and its outcomes. Furthermore, we also briefly reviewed other treatments developed for chondrogenesis and cartilage regeneration for comparison. EXPERT OPINION This treatment method has sustained positive effects on functional outcomes and cartilage growth in initial trials. It allows administration in a minimally invasive manner that does not require extended recovery time. Although several treatment modalities are currently under investigation and appear promising, we hope that these effects can be sustained in further studies. Ultimately, we anticipate that the results may be reproducible in many clinical settings and allow us to effectively treat cartilage damage in patients with degenerative arthritis.
Collapse
Affiliation(s)
- Randa K Elmallah
- Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Center for Joint Preservation and Replacement , 2401 West Belvedere Avenue, Baltimore, MD 21215 , USA +1 410 601 8500 ; +1 410 601 8501 ; ;
| | | | | | | | | | | |
Collapse
|
27
|
Zhang X, Sun J, Xin W, Li Y, Ni L, Ma X, Zhang D, Zhang D, Zhang T, Du G. Anti-inflammation effect of methyl salicylate 2-O-β-D-lactoside on adjuvant induced-arthritis rats and lipopolysaccharide (LPS)-treated murine macrophages RAW264.7 cells. Int Immunopharmacol 2015; 25:88-95. [PMID: 25637446 DOI: 10.1016/j.intimp.2015.01.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 01/23/2015] [Accepted: 01/23/2015] [Indexed: 01/12/2023]
Abstract
Methyl salicylate 2-O-β-D-lactoside (MSL) is a derivative of natural salicylate isolated from Gaultheria yunnanensis (Franch.) Rehder, which is widely used for treating rheumatoid arthritis (RA), swelling and pain. The aim of the present study was to investigate the effect of MSL on the progression of adjuvant-induced arthritis (AIA) in rat in vivo and explore the anti-inflammatory effects and mechanism of MSL in lipopolysaccharide (LPS)-treated murine macrophages RAW264.7 cells in vitro. Our results showed that MSL significantly inhibited the arthritis progression in AIA rats, decreasing the right hind paw swelling and ankle diameter, attenuating histopathological changes and suppressing the plasma levels of TNF-α and IL-1β in AIA rats. Besides, MSL had potent anti-inflammatory effects on the LPS-activated RAW264.7. MSL dose-dependently inhibited the activity of COX-1, and COX-2. Moreover, MSL prominently inhibited LPS-induced activation of MAPK in RAW264.7 cells by blocking phosphorylation of p38 and ERK. Our study suggests that MSL may be effective in the treatment of inflammatory diseases by inhibiting the pro-inflammatory cytokine production and regulating the MAPK signal pathway.
Collapse
Affiliation(s)
- Xue Zhang
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China.
| | - Jialin Sun
- Pharmacy Department of the Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Wenyu Xin
- Binzhou Medical University, Yantai 264003, China.
| | - Yongjie Li
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China.
| | - Lin Ni
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Xiaowei Ma
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Dan Zhang
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Dongming Zhang
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Tiantai Zhang
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China.
| | - Guanhua Du
- Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100050, China.
| |
Collapse
|
28
|
Lacy SE, Wu C, Ambrosi DJ, Hsieh CM, Bose S, Miller R, Conlon DM, Tarcsa E, Chari R, Ghayur T, Kamath RV. Generation and characterization of ABT-981, a dual variable domain immunoglobulin (DVD-Ig(TM)) molecule that specifically and potently neutralizes both IL-1α and IL-1β. MAbs 2015; 7:605-19. [PMID: 25764208 PMCID: PMC4622731 DOI: 10.1080/19420862.2015.1026501] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/24/2015] [Accepted: 03/01/2015] [Indexed: 12/15/2022] Open
Abstract
Interleukin-1 (IL-1) cytokines such as IL-1α, IL-1β, and IL-1Ra contribute to immune regulation and inflammatory processes by exerting a wide range of cellular responses, including expression of cytokines and chemokines, matrix metalloproteinases, and nitric oxide synthetase. IL-1α and IL-1β bind to IL-1R1 complexed to the IL-1 receptor accessory protein and induce similar physiological effects. Preclinical and clinical studies provide significant evidence for the role of IL-1 in the pathogenesis of osteoarthritis (OA), including cartilage degradation, bone sclerosis, and synovial proliferation. Here, we describe the generation and characterization of ABT-981, a dual variable domain immunoglobulin (DVD-Ig) of the IgG1/k subtype that specifically and potently neutralizes IL-1α and IL-1β. In ABT-981, the IL-1β variable domain resides in the outer domain of the DVD-Ig, whereas the IL-1α variable domain is located in the inner position. ABT-981 specifically binds to IL-1α and IL-1β, and is physically capable of binding 2 human IL-1α and 2 human IL-1β molecules simultaneously. Single-dose intravenous and subcutaneous pharmacokinetics studies indicate that ABT-981 has a half-life of 8.0 to 10.4 d in cynomolgus monkey and 10.0 to 20.3 d in rodents. ABT-981 exhibits suitable drug-like-properties including affinity, potency, specificity, half-life, and stability for evaluation in human clinical trials. ABT-981 offers an exciting new approach for the treatment of OA, potentially addressing both disease modification and symptom relief as a disease-modifying OA drug.
Collapse
Affiliation(s)
- Susan E Lacy
- AbbVie Bioresearch Center; Global Biologics; Worcester, MA USA
| | - Chengbin Wu
- Shanghai CP Guojian Pharmaceutical Co., Ltd.; Shanghai, China
| | | | | | - Sahana Bose
- AbbVie Bioresearch Center; Global Biologics; Worcester, MA USA
| | - Renee Miller
- AbbVie Bioresearch Center; Global Biologics; Worcester, MA USA
| | - Donna M Conlon
- AbbVie Bioresearch Center; Immunology Pharmacology; Worcester, MA USA
| | - Edit Tarcsa
- AbbVie Bioresearch Center; DMPK-BA; Worcester, MA USA
| | - Ravi Chari
- AbbVie Bioresearch Center; Drug Product Development; Worcester, MA USA
| | - Tariq Ghayur
- AbbVie Bioresearch Center; Global Biologics; Worcester, MA USA
| | - Rajesh V Kamath
- AbbVie Bioresearch Center; Foundational Immunology; Worcester, MA USA
| |
Collapse
|
29
|
Abstract
Gout is a common crystal-induced arthritis, in which monosodium urate (MSU) crystals precipitate within joints and soft tissues and elicit an inflammatory response. The causes of elevated serum urate and the inflammatory pathways activated by MSU crystals have been well studied, but less is known about the processes leading to crystal formation and growth. Uric acid, the final product of purine metabolism, is a weak acid that circulates as the deprotonated urate anion under physiologic conditions, and combines with sodium ions to form MSU. MSU crystals are known to have a triclinic structure, in which stacked sheets of purine rings form the needle-shaped crystals that are observed microscopically. Exposed, charged crystal surfaces are thought to allow for interaction with phospholipid membranes and serum factors, playing a role in the crystal-mediated inflammatory response. While hyperuricemia is a clear risk factor for gout, local factors have been hypothesized to play a role in crystal formation, such as temperature, pH, mechanical stress, cartilage components, and other synovial and serum factors. Interestingly, several studies suggest that MSU crystals may drive the generation of crystal-specific antibodies that facilitate future MSU crystallization. Here, we review MSU crystal biology, including a discussion of crystal structure, effector function, and factors thought to play a role in crystal formation. We also briefly compare MSU biology to that of uric acid stones causing nephrolithasis, and consider the potential treatment implications of MSU crystal biology.
Collapse
Affiliation(s)
- Miguel A Martillo
- Divisions of Rheumatology, Department of Medicine, NYU School of Medicine, New York, USA
| | | | | |
Collapse
|
30
|
Hacibeyoğlu G, Saritaş TB, Saritaş ZK, Korkmaz M, Sevimli A, Mehmetoğlu İ, Otelcioğlu Ş. The determination of histopathological and biochemical effects of the rabbit knee joint injected dexketoprofen trometamol. Fundam Clin Pharmacol 2014; 29:79-85. [PMID: 24673725 DOI: 10.1111/fcp.12074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 03/03/2014] [Accepted: 03/14/2014] [Indexed: 11/28/2022]
Abstract
This study was conducted to investigate possible histopathological effects and biochemical reflections of intra-articular dexketoprofen trometamol. A total of 24 New Zealand rabbits were included in the study. Blood sampling was carried out from all animals on the first day, then they were randomly allocated either to the control group (Group C, n = 9) or the dexketoprofen trometamol group (Group D, n = 15). Group C underwent each two intra-articular injections of saline, 0.25 mL into right and 0.50 mL into left knee. Group D was injected 0.25 mL (6.25 mg) dexketoprofen trometamol into the right knee and 0.50 mL (12.5 mg) into the left. The groups were divided randomly into three. Tissue and blood samples were collected from Groups C1 and D1 on the first day, C2 and D2 on the second day and C3 and D3 on the 10th day of the study. Interleukin-1 (IL-1β), interleukin-6 (IL-6), tumour necrosis factor-alpha (TNF-α) and C-reactive protein (CRP) levels were studied. The histopathological examination of C and D groups did not present any deterioration. IL-6 basal levels were significantly higher in Group D2 compared with C2 and C3 compared with D3. Basal TNF-α levels were higher compared with day 1 in Group C1, and IL-6 and CRP levels were higher in Group D3. Also, none of the increases in these values are supported by histopathological evaluation results. Consequently, we suppose that dexketoprofen trometamol does not cause histopathological deterioration in articular cartilage of rabbits, and the increases in biochemical parameters exclusively are not clinically significant.
Collapse
Affiliation(s)
- Gülçin Hacibeyoğlu
- Anesthesiology and Reanimation Department, Meram Medical School, Necmettin Erbakan University, Konya, Turkey
| | | | | | | | | | | | | |
Collapse
|
31
|
Parekh R, Lorenzo MK, Shin SY, Pozzi A, Clark AL. Integrin α1β1 differentially regulates cytokine-mediated responses in chondrocytes. Osteoarthritis Cartilage 2014; 22:499-508. [PMID: 24418674 PMCID: PMC4028170 DOI: 10.1016/j.joca.2013.12.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 12/07/2013] [Accepted: 12/25/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To elucidate the role of integrin α1β1 in chondrocyte responses to inflammatory interleukin-1α (IL-1) and anabolic transforming growth factor-β1 (TGF-β1) in the knee. METHODS Intracellular calcium transient responses to IL-1 and TGF-β1 were measured in wild type and integrin α1-null chondrocytes using real time ex vivo confocal microscopy, and immunohistochemistry was performed to analyze TGF-β1-mediated activation of Smad2/3 in tibial and femoral chondrocytes. RESULTS Loss of integrin α1β1 reduces intracellular calcium transient response to IL-1, while it enhances chondrocyte responses to TGF-β1 as measured by intracellular calcium transients and activation of downstream Smad2/3. CONCLUSIONS Integrin α1β1 plays a vital role in mediating chondrocyte responses to two contrasting factors that are critical players in the onset and progression of osteoarthritis - inflammatory IL-1 and anabolic TGF-β. Further investigation into the molecular mechanisms by which integrin α1β1 mediates these responses will be an important next step in understanding the influence of increased expression of integrin α1β1 during the early stages of osteoarthritis on disease progression.
Collapse
Affiliation(s)
- R Parekh
- Faculty of Science, The University of Calgary, Calgary, AB, Canada
| | - MK Lorenzo
- Faculty of Science, The University of Calgary, Calgary, AB, Canada
| | - SY Shin
- Faculty of Kinesiology, The University of Calgary, Calgary, AB, Canada
| | - A Pozzi
- Department of Medicine, Vanderbilt University, Nashville, TN, USA,Department of Medicine, Veterans Affairs Hospital, Nashville, TN, USA
| | - AL Clark
- Faculty of Kinesiology, The University of Calgary, Calgary, AB, Canada,Department of Surgery, Faculty of Medicine, The University of Calgary, Calgary, AB, Canada
| |
Collapse
|
32
|
[New therapies for rheumatoid arthritis]. Med Clin (Barc) 2014; 143:461-6. [PMID: 24461738 DOI: 10.1016/j.medcli.2013.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 11/07/2013] [Accepted: 11/14/2013] [Indexed: 12/20/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic inflammatory disease characterized by inflammation of the synovial membrane and progressive destruction of the articular cartilage and bone. Advances in the knowledge of disease pathogenesis allowed the identification of novel therapeutic targets such as tumor necrosis factor (TNF), interleukin (IL)-1, IL-6 or the system JAK/STAT phosphorylation. At present there are 5 TNF antagonists approved for RA. Tocilizumab blocks the pathway of IL-6 and is the only biological with proven efficacy in monotherapy. Rituximab modulates B cell response in RA. Abatacept provided new data on T cell involvement in the pathogenesis of RA. Tofacitinib is the first kinase inhibitor approved for this disease. Biologic drugs have proven efficacy, almost always in combination with methotrexate, and even halt radiographic progression. Monitoring infection is the main precaution in handling these patients.
Collapse
|
33
|
Chamberlain CS, Leiferman EM, Frisch KE, Brickson SL, Murphy WL, Baer GS, Vanderby R. Interleukin expression after injury and the effects of interleukin-1 receptor antagonist. PLoS One 2013; 8:e71631. [PMID: 23936523 PMCID: PMC3731283 DOI: 10.1371/journal.pone.0071631] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 07/01/2013] [Indexed: 01/31/2023] Open
Abstract
Ligament healing follows a series of complex coordinated events involving various cell types, cytokines, as well as other factors, producing a mechanically inferior tissue more scar-like than native tissue. Macrophages provide an ongoing source of cytokines to modulate inflammatory cell adhesion and migration as well as fibroblast proliferation. Studying interleukins inherent to ligament healing during peak macrophage activation and angiogenesis may elucidate inflammatory mediators involved in subsequent scar formation. Herein, we used a rat healing model assayed after surgical transection of their medial collateral ligaments (MCLs). On days 3 and 7 post-injury, ligaments were collected and used for microarray analysis. Of the 12 significantly modified interleukins, components of the interleukin-1 family were significantly up-regulated. We therefore examined the influence of interleukin-1 receptor antagonist (IL-1Ra) on MCL healing. Transected rat MCLs received PBS or IL-1Ra at the time of surgery. Inhibition of IL-1 activation decreased pro-inflammatory cytokines (IL-1α, IL-1β, IL-12, IL-2, and IFN-γ), myofibroblasts, and proliferating cells, as well as increased anti-inflammatory cytokines (IL-10), endothelial cells/blood vessel lumen, M2 macrophages, and granulation tissue size without compromising the mechanical properties. These results support the concept that IL-1Ra modulates MCL-localized granulation tissue components and cytokine production to create a transient environment that is less inflammatory. Overall, IL-1Ra may have therapeutic potential early in the healing cascade by stimulating the M2 macrophages and altering the granulation tissue components. However, the single dose of IL-1Ra used in this study was insufficient to maintain the more regenerative early response. Due to the transient influence on most of the healing components tested, IL-1Ra may have greater therapeutic potential with sustained delivery.
Collapse
Affiliation(s)
- Connie S. Chamberlain
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Ellen M. Leiferman
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Kayt E. Frisch
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Stacey L. Brickson
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, United States of America
| | - William L. Murphy
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Geoffrey S. Baer
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Ray Vanderby
- Department of Orthopedics and Rehabilitation, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
34
|
Mechanisms of adhesion and subsequent actions of a haematopoietic stem cell line, HPC-7, in the injured murine intestinal microcirculation in vivo. PLoS One 2013; 8:e59150. [PMID: 23554986 PMCID: PMC3595270 DOI: 10.1371/journal.pone.0059150] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 02/11/2013] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Although haematopoietic stem cells (HSCs) migrate to injured gut, therapeutic success clinically remains poor. This has been partially attributed to limited local HSC recruitment following systemic injection. Identifying site specific adhesive mechanisms underpinning HSC-endothelial interactions may provide important information on how to enhance their recruitment and thus potentially improve therapeutic efficacy. This study determined (i) the integrins and inflammatory cyto/chemokines governing HSC adhesion to injured gut and muscle (ii) whether pre-treating HSCs with these cyto/chemokines enhanced their adhesion and (iii) whether the degree of HSC adhesion influenced their ability to modulate leukocyte recruitment. METHODS Adhesion of HPC-7, a murine HSC line, to ischaemia-reperfused (IR) injured mouse gut or cremaster muscle was monitored intravitally. Critical adhesion molecules were identified by pre-treating HPC-7 with blocking antibodies to CD18 and CD49d. To identify cyto/chemokines capable of recruiting HPC-7, adhesion was monitored following tissue exposure to TNF-α, IL-1β or CXCL12. The effects of pre-treating HPC-7 with these cyto/chemokines on surface integrin expression/clustering, adhesion to ICAM-1/VCAM-1 and recruitment in vivo was also investigated. Endogenous leukocyte adhesion following HPC-7 injection was again determined intravitally. RESULTS IR injury increased HPC-7 adhesion in vivo, with intestinal adhesion dependent upon CD18 and muscle adhesion predominantly relying on CD49d. Only CXCL12 pre-treatment enhanced HPC-7 adhesion within injured gut, likely by increasing CD18 binding to ICAM-1 and/or CD18 surface clustering on HPC-7. Leukocyte adhesion was reduced at 4 hours post-reperfusion, but only when local HPC-7 adhesion was enhanced using CXCL12. CONCLUSION This data provides evidence that site-specific molecular mechanisms govern HPC-7 adhesion to injured tissue. Importantly, we show that HPC-7 adhesion is a modulatable event in IR injury and further demonstrate that adhesion instigated by injury alone is not sufficient for mediating anti-inflammatory effects. Enhancing local HSC presence may therefore be essential to realising their clinical potential.
Collapse
|
35
|
Edwards CK, Green JS, Volk HD, Schiff M, Kotzin BL, Mitsuya H, Kawaguchi T, Sakata KM, Cheronis J, Trollinger D, Bankaitis-Davis D, Dinarello CA, Norris DA, Bevilacqua MP, Fujita M, Burmester GR. Combined anti-tumor necrosis factor-α therapy and DMARD therapy in rheumatoid arthritis patients reduces inflammatory gene expression in whole blood compared to DMARD therapy alone. Front Immunol 2012; 3:366. [PMID: 23264777 PMCID: PMC3525111 DOI: 10.3389/fimmu.2012.00366] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 11/17/2012] [Indexed: 11/13/2022] Open
Abstract
Periodic assessment of gene expression for diagnosis and monitoring in rheumatoid arthritis (RA) may provide a readily available and useful method to detect subclinical disease progression and follow responses to therapy with disease modifying anti-rheumatic agents (DMARDs) or anti-TNF-α therapy. We used quantitative real-time PCR to compare peripheral blood gene expression profiles in active (“unstable”) RA patients on DMARDs, stable RA patients on DMARDs, and stable RA patients treated with a combination of a disease-modifying anti-rheumatoid drug (DMARD) and an anti-TNF-α agent (infliximab or etanercept) to healthy human controls. The expression of 48 inflammatory genes were compared between healthy controls (N = 122), unstable DMARD patients (N = 18), stable DMARD patients (N = 26), and stable patients on combination therapy (N = 20). Expression of 13 genes was very low or undetectable in all study groups. Compared to healthy controls, patients with unstable RA on DMARDs exhibited increased expression of 25 genes, stable DMARD patients exhibited increased expression of 14 genes and decreased expression of five genes, and combined therapy patients exhibited increased expression of six genes and decreased expression of 10 genes. These findings demonstrate that active RA is associated with increased expression of circulating inflammatory markers whereas increases in inflammatory gene expression are diminished in patients with stable disease on either DMARD or anti-TNF-α therapy. Furthermore, combination DMARD and anti-TNF-α therapy is associated with greater reductions in circulating inflammatory gene expression compared to DMARD therapy alone. These results suggest that assessment of peripheral blood gene expression may prove useful to monitor disease progression and response to therapy.
Collapse
Affiliation(s)
- Carl K Edwards
- Department of Dermatology, University of Colorado Denver, Anschutz Medical Campus Aurora, CO, USA ; Department of Rheumatology, University of Colorado Denver, Anschutz Medical Campus Aurora, CO, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Longo UG, Loppini M, Fumo C, Rizzello G, Khan WS, Maffulli N, Denaro V. Osteoarthritis: new insights in animal models. Open Orthop J 2012; 6:558-63. [PMID: 23248728 PMCID: PMC3522504 DOI: 10.2174/1874325001206010558] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2012] [Revised: 09/14/2012] [Accepted: 09/23/2012] [Indexed: 11/22/2022] Open
Abstract
Osteoarthritis (OA) is the most frequent and symptomatic health problem in the middle-aged and elderly population, with over one-half of all people over the age of 65 showing radiographic changes in painful knees. The aim of the present study was to perform an overview on the available animal models used in the research field on the OA. Discrepancies between the animal models and the human disease are present. As regards human 'idiopathic' OA, with late onset and slow progression, it is perhaps wise not to be overly enthusiastic about animal models that show severe chondrodysplasia and very early OA. Advantage by using genetically engineered mouse models, in comparison with other surgically induced models, is that molecular etiology is known. Find potential molecular markers for the onset of the disease and pay attention to the role of gender and environmental factors should be very helpful in the study of mice that acquire premature OA. Surgically induced destabilization of joint is the most widely used induction method. These models allow the temporal control of disease induction and follow predictable progression of the disease. In animals, ACL transection and meniscectomy show a speed of onset and severity of disease higher than in humans after same injury.
Collapse
Affiliation(s)
- Umile Giuseppe Longo
- Department of Orthopaedic and Trauma Surgery. Campus Bio-Medico University, Via Alvaro del Portillo, 200, 00128 Trigoria, Rome, Italy ; Centro Integrato di Ricerca (CIR) Campus Bio-Medico University, Via Alvaro del Portillo, 21, 00128, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
37
|
Cytokine and Proteinase Inhibitors in the Modulation of Connective Tissue Destruction in Rheumatoid Arthritis. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/bf03259265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
38
|
Alfieri A, Watson JJ, Kammerer RA, Tasab M, Progias P, Reeves K, Brown NJ, Brookes ZL. Angiopoietin-1 variant reduces LPS-induced microvascular dysfunction in a murine model of sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2012; 16:R182. [PMID: 23036162 PMCID: PMC3682284 DOI: 10.1186/cc11666] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 10/04/2012] [Indexed: 11/10/2022]
Abstract
Introduction Severe sepsis is characterised by intravascular or extravascular infection with microbial agents, systemic inflammation and microcirculatory dysfunction, leading to tissue damage, organ failure and death. The growth factor angiopoietin (Ang-1) has therapeutic potential but recombinant Ang-1 tends to aggregate and has a short half-life in vivo. This study aimed to investigate the acute effects of the more stable Ang-1 variant matrilin-1-angiopoietin-1 (MAT.Ang-1) on the function of the microcirculation in an experimental model of sepsis, and whether any protection by MAT-Ang-1 was associated with modulation of inflammatory cytokines, angiogenic factors or the endothelial nitric oxide synthase (eNOS)-Akt and vascular endothelial (VE)-cadherin pathways. Methods Aluminium window chambers were implanted into the dorsal skinfold of male C3H/HeN mice (7 to 10 weeks old) to expose the striated muscle microcirculation. Endotoxemia was induced by intraperitoneal injection of lipopolysaccharide (LPS, 1 mg/kg at 0 and 19 hours). MAT.Ang-1 was administered intravenously 20 hours after the onset of sepsis. Microcirculatory function was evaluated by intravital microscopy and Doppler fluximetry. Results Endotoxemia resulted in macromolecular leak, which was ameliorated by MAT.Ang-1 post-treatment. LPS induced a dramatic reduction in tissue perfusion, which was improved by MAT.Ang-1. Proteome profiler array analysis of skeletal muscle also demonstrated increased inflammatory and reduced angiogenic factors during endotoxemia. MAT.Ang-1 post-treatment reduced the level of IL-1β but did not significantly induce the expression of angiogenic factors. MAT.Ang-1 alone did not induce leak or increase angiogenic factors but did reduce vascular endothelial growth factor expression in controls. Conclusion Administration of MAT.Ang-1 after the onset of sepsis protects the microcirculation from endotoxemia-induced vascular dysfunction through reducing inflammation but without pro-angiogenic actions, thus representing a novel, potential pharmacotherapeutic agent for the treatment of sepsis.
Collapse
|
39
|
Abstract
Many studies in animals have examined biochemical, immune and histological changes during arthritis; however, the study of the effects of arthritis on mobility has been largely neglected. Interleukin-1, administered by the intraarticular route into hamster knee joints, resulted in inhibition of spontaneous wheel running activity; however, the effect was transient, lasting only through the evening following IL-1 administration. A further injection of IL-1 2 days later showed still greater inhibition of running. The effect again did not extend beyond the first evening after injection. IL-1alpha and IL-1beta showed equivalent effects on mobility, and no evidence was seen for cooperative interaction between them. A 50% inhibition of running occurred at a dose of approximately 10 ng/knee of IL-1alpha. The effect appeared not to be systemic since intraperitoneal injection required microgram amounts of IL-1 for an equivalent inhibition. At the time mobility had been restored to normal, histological examination showed the continued presence of inflammatory cells, soft tissue swelling and cartilage proteoglycan loss. These results suggest a lack of correlation between inhibition of mobility and histopathological changes in cartilage and soft tissue.
Collapse
|
40
|
COMMUNICATION. Br J Pharmacol 2012. [DOI: 10.1111/j.1476-5381.1987.tb16603.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
41
|
POSTER COMMUNICATIONS. Br J Pharmacol 2012. [DOI: 10.1111/j.1476-5381.1991.tb14726.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
42
|
Effects of (-)-epigallocatechin gallate on HMGB1 and interleukin-1β-mediated barrier disruption in human endothelial cells. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/s13765-012-1041-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Lamacchia C, Rodriguez E, Palmer G, Vigne S, Martin P, Talabot-Ayer D, Seemayer CA, Gabay C. Articular inflammation is controlled by myeloid cell-derived interleukin 1 receptor antagonist during the acute phase of arthritis in mice. Ann Rheum Dis 2012; 71:281-7. [PMID: 22072017 DOI: 10.1136/annrheumdis-2011-200429] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVES To define the cell type (myeloid vs other cells) specific effect of interleukin 1 (IL-1) receptor antagonist (IL-1Ra) deficiency on the acute inflammatory phase of arthritis. METHODS Arthritis was induced by K/BxN serum transfer in wild-type (WT), IL-1Ra-deficient (IL-1Ra(-/-)) and conditional knockout mice. In the latter, IL-1Ra production was specifically targeted in myeloid cells (IL-1Ra(ΔM)) or in both hepatocytes and myeloid cells (IL-1Ra(ΔH+M)). Arthritis severity was clinically evaluated and ankle sections were scored for synovial inflammation and cartilage erosion. Quantitative RT-PCR, western blot and immunohistochemical analyses measured expression, localisation and cellular sources of the different IL-1Ra isoforms in arthritic joints. RESULTS Total and myeloid cell-specific IL-1Ra deficiency was associated with increased arthritis severity, although disease incidence was similar to that of WT mice. Increased clinical scores were associated with exacerbated synovial inflammation. All IL-1Ra isoforms, except for intracellular (ic)IL-1Ra2, were expressed in arthritic joints of WT mice. In contrast, production of secreted (s)IL-1Ra and icIL-1Ra3 isoforms was markedly decreased in arthritic joints of both IL-1Ra(ΔM) and IL-1Ra(ΔH+M) mice. Immunohistochemical and western blot analyses suggested that the icIL-1Ra1 isoform is produced primarily by synovial fibroblasts. CONCLUSION Myeloid cell-derived IL-1Ra, including both sIL-1Ra and icIL-1Ra3 isoforms, controls articular inflammation during the acute phase of K/BxN serum transfer-induced arthritis.
Collapse
Affiliation(s)
- Céline Lamacchia
- Division of Rheumatology, University Hospitals of Geneva, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Mueller MB, Tuan RS. Anabolic/Catabolic balance in pathogenesis of osteoarthritis: identifying molecular targets. PM R 2011; 3:S3-11. [PMID: 21703577 DOI: 10.1016/j.pmrj.2011.05.009] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 05/10/2011] [Indexed: 10/18/2022]
Abstract
Osteoarthritis is the most common degenerative musculoskeletal disease. In healthy cartilage, a low turnover of extracellular matrix molecules occurs. Proper balance of anabolic and catabolic activities is thus crucial for the maintenance of cartilage tissue integrity and for the repair of molecular damages sustained during daily usage. In persons with degenerative diseases such as osteoarthritis, this balance of anabolic and catabolic activities is compromised, and the extent of tissue degradation predominates over the capacity of tissue repair. This mismatch eventually results in cartilage loss in persons with osteoarthritis. Tissue homeostasis is controlled by coordinated actions and crosstalk among a number of proanabolic and antianabolic and procatabolic and anticatabolic factors. In osteoarthritis, an elevation of antianabolic and catabolic factors occurs. Interestingly, anabolic activity is also increased, but this response fails to repair the tissue because of both quantitative and qualitative insufficiency. This review presents an overview of the anabolic and catabolic activities involved in cartilage degeneration and the interplay among different signaling and metabolic factors. Understanding the basic molecular mechanisms responsible for tissue degeneration is critical to identifying and developing means to efficiently block or reverse the pathobiological symptoms of osteoarthritis.
Collapse
Affiliation(s)
- Michael B Mueller
- Department of Trauma Surgery, University of Regensburg Medical Center, Regensburg, Germany
| | | |
Collapse
|
45
|
Lawrence JTR, Birmingham J, Toth AP. Emerging ideas: prevention of posttraumatic arthritis through interleukin-1 and tumor necrosis factor-alpha inhibition. Clin Orthop Relat Res 2011; 469:3522-6. [PMID: 21161742 PMCID: PMC3210259 DOI: 10.1007/s11999-010-1699-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 11/12/2010] [Indexed: 01/31/2023]
Abstract
BACKGROUND Despite surgical and mechanical stabilization of an acutely injured joint through ligament reconstruction, meniscus repair, or labral repair, the risk of posttraumatic arthritis remains high. Joint injury triggers three phases of pathogenic events: the early (acute) phase involves joint swelling, hemarthrosis, expression of inflammatory cytokines (especially interleukin-1 [IL-1] and tumor necrosis factor-α [TNF-α]), and biomarkers of cartilage catabolism; an intermediate phase is characterized by reduction of joint inflammation, ongoing joint catabolism, but no evidence yet for typical features of radiographic osteoarthritis (OA); and a late phase characterized by radiographic OA. HYPOTHESES We hypothesize that the early phase of acute knee injury represents a window of opportunity for providing biologic treatment to promote healing and to slow or prevent a subsequent cascade of destructive joint processes leading to OA. PROPOSED PROGRAM We propose a phase II, randomized, placebo-controlled, double-blinded, clinical trial to treat acute knee injuries with intraarticular injection of an IL-1 inhibitor. Patient-centered outcomes will include pain reduction and improvement of knee function. MR imaging and measurement of biochemical markers will be monitored during the subsequent 2 years to determine if the structural response to injury can be reversed. SIGNIFICANCE If this model is validated, modulation of the molecular pathways responsible for articular cartilage breakdown will augment current reconstructive procedures in the treatment of acute joint injuries and prevent the development of injury-related arthritis.
Collapse
Affiliation(s)
- J. Todd R. Lawrence
- Division of Orthopaedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA USA ,Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA USA ,Division of Orthopaedic Surgery, Duke University, Durham, NC USA
| | - James Birmingham
- Pediatric and Adult Rheumatology, West Michigan Rheumatology, PLLC, Grand Rapids, MI USA ,Helen Devos Pediatric Rheumatology, Grand Rapids, MI USA
| | - Alison P. Toth
- Division of Orthopaedic Surgery, Duke University, DUMC 3970, Durham, NC 27710 USA
| |
Collapse
|
46
|
Ohtani T, Habu M, Khanal A, Yoshioka I, Matsukawa A, Tominaga K. Local effects of intra-articular injection of anti-rabbit tumor necrosis factor alpha monoclonal antibody in antigen-induced arthritis of the rabbit temporomandibular joint. J Oral Pathol Med 2011; 41:96-105. [DOI: 10.1111/j.1600-0714.2011.01056.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
47
|
Carroll GJ, Breidahl WH, Bulsara MK, Olynyk JK. Hereditary hemochromatosis is characterized by a clinically definable arthropathy that correlates with iron load. ACTA ACUST UNITED AC 2010; 63:286-94. [PMID: 20954257 DOI: 10.1002/art.30094] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- G J Carroll
- University of Notre Dame Australia, Fremantle Campus, and Fremantle Hospital, Fremantle, Western Australia, Australia.
| | | | | | | |
Collapse
|
48
|
Hahn YS, Kim JG. Pathogenesis and clinical manifestations of juvenile rheumatoid arthritis. KOREAN JOURNAL OF PEDIATRICS 2010; 53:921-30. [PMID: 21218013 PMCID: PMC3012271 DOI: 10.3345/kjp.2010.53.11.921] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 10/19/2010] [Indexed: 12/24/2022]
Abstract
Juvenile rheumatoid arthritis (JRA) is the most common rheumatic childhood disease; its onset is before 16 years of age and it persists for at least 6 weeks. JRA encompasses a heterogeneous group of diseases that is classified according to 3 major presentations: oligoarthritis, polyarthritis, and systemic onset diseases. These presentations may originate from the same or different causes that involve interaction with specific immunogenetic predispositions, and result in heterogeneous clinical manifestations. An arthritic joint exhibits cardinal signs of joint inflammation, such as swelling, pain, heat, and loss of function; any joint can be arthritic, but large joints are more frequently affected. Extra-articular manifestations include high fever, skin rash, serositis, and uveitis. The first 2 types of JRA are regarded as T helper 1 (Th1) cell-mediated inflammatory disorders, mainly based on the abundance of activated Th1 cells in the inflamed synovium and the pathogenetic role of proinflammatory cytokines that are mainly produced by Th1 cell-stimulated monocytes. In contrast, the pathogenesis of systemic onset disease differs from that of other types of JRA in several respects, including the lack of association with human leukocyte antigen type and the absence of autoantibodies or autoreactive T cells. Although the precise mechanism that leads to JRA remains unclear, proinflammatory cytokines are thought to be responsible for at least part of the clinical symptoms in all JRA types. The effectiveness of biologic therapy in blocking the action of these cytokines in JRA patients provides strong evidence that they play a fundamental role in JRA inflammation.
Collapse
Affiliation(s)
- Youn-Soo Hahn
- Department of Pediatrics, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Joong-Gon Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
49
|
Abstract
Arthropathy is a frequent and serious complication of repeated joint bleeding in patients with hemophilia, resulting in pain, deformity, and disability. Although the pathogenesis of hemophilic arthropathy has not been fully elucidated, it appears to have similarities with the degenerative joint damage that occurs in osteoarthritis and the inflammatory processes associated with rheumatoid arthritis. This article reviews the potential actions of various blood constituents on joint components that culminate in the development of hemophilic arthropathy.
Collapse
Affiliation(s)
- L A Valentino
- Rush Hemophilia and Thrombophilia Center, Department of Pediatrics, Rush Children's Hospital and Rush University Medical Center, Chicago, IL USA.
| |
Collapse
|
50
|
Jacobs JP, Ortiz-Lopez A, Campbell JJ, Gerard CJ, Mathis D, Benoist C. Deficiency of CXCR2, but not other chemokine receptors, attenuates autoantibody-mediated arthritis in a murine model. ARTHRITIS AND RHEUMATISM 2010; 62:1921-32. [PMID: 20506316 PMCID: PMC2994550 DOI: 10.1002/art.27470] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE Chemokines coordinate leukocyte trafficking in homeostasis and during immune responses. Prior studies of their role in arthritis have used animal models with both an initial adaptive immune response and an inflammatory effector phase. We undertook analysis of chemokines and their receptors in the effector phase of arthritis using the K/BxN mouse serum-transfer model. METHODS A time-course microarray analysis of serum-transferred arthritis was performed, examining ankle tissue, synovial fluid, and peripheral blood leukocytes. Up-regulation of chemokines was confirmed by quantitative reverse transcriptase-polymerase chain reaction. The functional relevance of chemokine induction was assessed by transferring serum into mice deficient in CCR1-7, CCR9, CXCR2, CXCR3, CXCR5, CX(3)CR1, CCL2, or CCL3. Further mechanistic analysis of CXCR2 involved treatment of arthritic mice with a CXCR2 antagonist, bone marrow (BM) cell transfers with CXCR2(+/-) and CXCR2(-/-) donors and recipients, flow cytometry of synovial cells, and competition experiments measuring enrichment of CXCR2-expressing neutrophils in arthritic joints of mice with mixed CXCR2(+/+) and CXCR2(-/-) BM cells. RESULTS Gene expression profiling revealed up-regulation of the CXCR2 ligands CXCL1, CXCL2, and CXCL5 in the joint in parallel with disease activity. CXCR2(-/-) mice had attenuated disease relative to CXCR2(+/-) littermates, as did mice receiving the CXCR2 inhibitor, while deficiency of other chemokine receptors did not affect arthritis severity. CXCR2 was required only on hematopoietic cells and was widely expressed on synovial neutrophils. CXCR2-expressing neutrophils were preferentially recruited to arthritic joints in the presence of CXCR2-deficient neutrophils. CONCLUSION CXCR2 (but not other chemokine receptors) is critical for the development of autoantibody-mediated arthritis, exhibiting a cell-autonomous role in neutrophil recruitment to inflamed joints.
Collapse
Affiliation(s)
- Jonathan P. Jacobs
- Section on Immunology and Immunogenetics, Joslin Diabetes Center; Department of Medicine, Brigham and Women’s Hospital; Department of Pathology; Harvard Medical School, Boston, MA 02115
| | - Adriana Ortiz-Lopez
- Section on Immunology and Immunogenetics, Joslin Diabetes Center; Department of Medicine, Brigham and Women’s Hospital; Department of Pathology; Harvard Medical School, Boston, MA 02115
| | - James J. Campbell
- Brigham and Women’s Hospital, Department of Dermatology, Boston, MA 02115
| | | | - Diane Mathis
- Section on Immunology and Immunogenetics, Joslin Diabetes Center; Department of Medicine, Brigham and Women’s Hospital; Department of Pathology; Harvard Medical School, Boston, MA 02115
| | - Christophe Benoist
- Section on Immunology and Immunogenetics, Joslin Diabetes Center; Department of Medicine, Brigham and Women’s Hospital; Department of Pathology; Harvard Medical School, Boston, MA 02115
| |
Collapse
|