1
|
Baek HS, Hong VS, Kang H, Lee SJ, Lee JY, Kang H, Jeong S, Jung H, Park JW, Kwon TK, Son CN, Kim SH, Lee J, Kim KS, Kim S. Anti-rheumatic property and physiological safety of KMU-11342 in in vitro and in vivo models. Inflamm Res 2024; 73:1371-1391. [PMID: 38879731 PMCID: PMC11281989 DOI: 10.1007/s00011-024-01904-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 07/28/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic inflammatory disorder characterized by joint destruction due to synovial hypertrophy and the infiltration of inflammatory cells. Despite substantial progress in RA treatment, challenges persist, including suboptimal treatment responses and adverse effects associated with current therapies. This study investigates the anti-rheumatic capabilities of the newly identified multi-protein kinase inhibitor, KMU-11342, aiming to develop innovative agents targeting RA. In this study, we synthesized the novel multi-protein kinase inhibitor KMU-11342, based on indolin-2-one. We assessed its cardiac electrophysiological safety using the Langendorff system in rat hearts and evaluated its toxicity in zebrafish in vivo. Additionally, we examined the anti-rheumatic effects of KMU-11342 on human rheumatoid arthritis fibroblast-like synoviocytes (RA-FLS), THP-1 cells, and osteoclastogenesis in RAW264.7 cells. KMU-11342 demonstrated the ability to inhibit LPS-induced chemokine inhibition and the upregulation of pro-inflammatory cytokines, cyclooxygenase-2, inducible nitric oxide synthase, p-IKKα/β, p-NF-κB p65, and the nuclear translocation of NF-κB p65 in RA-FLS. It effectively suppressed the upregulation of NLR family pyrin domain containing 3 (NLRP3) and caspase-1 cleavage. Furthermore, KMU-11342 hindered the activation of osteoclast differentiation factors such as RANKL-induced TRAP, cathepsin K, NFATc-1, and c-Fos in RAW264.7 cells. KMU-11342 mitigates LPS-mediated inflammatory responses in THP-1 cells by inhibiting the activation of NLRP3 inflammasome. Notably, KMU-11342 exhibited minimal cytotoxicity in vivo and electrophysiological cardiotoxicity ex vivo. Consequently, KMU-11342 holds promise for development as a therapeutic agent in RA treatment.
Collapse
Affiliation(s)
- Hye Suk Baek
- Department of Immunology, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea
| | - Victor Sukbong Hong
- Department of Chemistry, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea
| | - Hyunsu Kang
- R&D Center for Advanced Pharmaceuticals & Evaluation, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, Republic of Korea
| | - Sang-Jin Lee
- Department of Biological Sciences, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jin-Young Lee
- Department of Biological Sciences, Keimyung University, Daegu, 42601, Republic of Korea
| | - Hyunju Kang
- Department of Food and Nutrition, Keimyung University, Daegu, 42601, Republic of Korea
| | - Seungik Jeong
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, Republic of Korea
| | - Hyunho Jung
- Department of Chemistry, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea
| | - Jong Wook Park
- Department of Immunology, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea
- Institute of Medical Science, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea
- Institute of Medical Science, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea
- Institute for Cancer Research, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea
| | - Chang-Nam Son
- Department of Rheumatology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, 712, Dongil-ro, Uijeongbu-si, 11759, Gyeonggi-do, Republic of Korea
| | - Sang Hyon Kim
- Division of Rheumatology, Department of Internal Medicine, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
| | - Jinho Lee
- Department of Chemistry, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea.
| | - Ki-Suk Kim
- R&D Center for Advanced Pharmaceuticals & Evaluation, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
| | - Shin Kim
- Department of Immunology, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea.
- Institute of Medical Science, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea.
- Institute for Cancer Research, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu, 42601, Republic of Korea.
| |
Collapse
|
2
|
Li ZC, Li M, Xiao LX, Zheng X, Li R, Dong SJ, Wang Y, Wen HY, Ruan KL, Cheng KG, Chen M, Tan YH. 6-O-angeloylplenolin inhibits osteoclastogenesis in vitro via suppressing c-Src/NF-κB/NFATc1 pathways and ameliorates bone resorption in collagen-induced arthritis mouse model. Biochem Pharmacol 2024; 224:116230. [PMID: 38643905 DOI: 10.1016/j.bcp.2024.116230] [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: 11/15/2023] [Revised: 04/02/2024] [Accepted: 04/19/2024] [Indexed: 04/23/2024]
Abstract
One of the effective therapeutic strategies to treat rheumatoid arthritis (RA)-related bone resorption is to target excessive activation of osteoclasts. We discovered that 6-O-angeloylplenolin (6-OAP), a pseudoguaianolide from Euphorbia thymifolia Linn widely used for the treatment of RA in traditional Chinese medicine, could inhibit RANKL-induced osteoclastogenesis and bone resorption in both RAW264.7 cells and BMMs from 1 μM and protect a collagen-induced arthritis (CIA) mouse model from bone destruction in vivo. The severity of arthritis and bone erosion observed in paw joints and the femurs of the CIA model were attenuated by 6-OAP administered at both dosages (1 or 5 mg/kg, i.g.). BMD, Tb.N and BV/TV were also improved by 6-OAP treatment. Histological analysis and TRAP staining of femurs further confirmed the protective effects of 6-OAP on bone erosion, which is mainly due to reduced osteoclasts. Molecular docking indicated that c-Src might be a target of 6-OAP and phosphorylation of c-Src was suppressed by 6-OAP treatment. CETSA and SPR assay further confirmed the potential interaction between 6-OAP and c-Src. Three signaling molecules downstream of c-Src that are vital to the differentiation and function of osteoclasts, NF-κB, c-Fos and NFATc1, were also suppressed by 6-OAP in vitro. In summary, the results demonstrated that the function of c-Src was disrupted by 6-OAP, which led to the suppression of downstream signaling vital to osteoclast differentiation and function. In conclusion, 6-OAP has the potential to be further developed for the treatment of RA-related bone erosion.
Collapse
Affiliation(s)
- Zhi-Chao Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China
| | - Min Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China
| | - Ling-Xiang Xiao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China
| | - Xi Zheng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China
| | - Rong Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China
| | - Shi-Jia Dong
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China
| | - Yue Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China
| | - Hong-Yu Wen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China
| | - Kun-Lin Ruan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China
| | - Ke-Guang Cheng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China.
| | - Ming Chen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China.
| | - Yan-Hui Tan
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, Guangxi, PR China.
| |
Collapse
|
3
|
Li JM, Yao YD, Luo JF, Liu JX, Lu LL, Liu ZQ, Dong Y, Xie Y, Zhou H. Pharmacological mechanisms of sinomenine in anti-inflammatory immunity and osteoprotection in rheumatoid arthritis: A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155114. [PMID: 37816287 DOI: 10.1016/j.phymed.2023.155114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/05/2023] [Accepted: 09/20/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND Sinomenine (SIN) is the main pharmacologically active component of Sinomenii Caulis and protects against rheumatoid arthritis (RA). In recent years, many studies have been conducted to elucidate the pharmacological mechanisms of SIN in the treatment of RA. However, the molecular mechanism of SIN in RA has not been fully elucidated. PURPOSE To summarize the pharmacological effects and molecular mechanisms of SIN in RA and clarify the most valuable regulatory mechanisms of SIN to provide clues and a basis for basic research and clinical applications. METHODS We systematically searched SciFinder, Web of Science, PubMed, China National Knowledge Internet (CNKI), the Wanfang Databases, and the Chinese Scientific Journal Database (VIP). We organized our work based on the PRISMA statement and selected studies for review based on predefined selection criteria. OUTCOME After screening, we identified 201 relevant studies, including 88 clinical trials and 113 in vivo and in vitro studies on molecular mechanisms. Among these studies, we selected key results for reporting and analysis. CONCLUSIONS We found that most of the known pharmacological mechanisms of SIN are indirect effects on certain signaling pathways or proteins. SIN was manifested to reduce the release of inflammatory cytokines such as Tumor necrosis factor-α (TNF-α), Interleukin-6 (IL-6), and IL-1β, thereby reducing the inflammatory response, and apparently blocking the destruction of bone and cartilage. The regulatory effects on inflammation and bone destruction make SIN a promising drug to treat RA. More notably, we believe that the modulation of α7nAChR and the regulation of methylation levels at specific GCG sites in the mPGES-1 promoter by SIN, and its mechanism of directly targeting GBP5, certainly enriches the possibilities and the underlying rationale for SIN in the treatment of inflammatory immune-related diseases.
Collapse
Affiliation(s)
- Juan-Min Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yun-Da Yao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Jin-Fang Luo
- Basic Medical College, Guizhou University of Traditional Chinese Medicine, Guian District, Guiyang, Guizhou, China
| | - Jian-Xin Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua, Hunan, China
| | - Lin-Lin Lu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhong-Qiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yan Dong
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510405, China.
| | - Ying Xie
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Hua Zhou
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, China; International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
4
|
Stefania S, Rotondo C, Mele A, Trotta A, Cantatore FP, Corrado A. Role of denosumab in bone erosions in rheumatoid arthritis. Postgrad Med J 2023; 99:976-984. [PMID: 36841226 DOI: 10.1093/postmj/qgad013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/05/2022] [Accepted: 01/17/2023] [Indexed: 02/27/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by systemic inflammation and synovitis which evolve into joint destruction and deformity. Bone abnormalities are represented by marginal bone erosions and iuxta-articular and generalized osteoporosis. Overactivation of osteoclasts along with dysregulation of osteoblasts are the key events. Bone resorption is mediated by the receptor activator of nuclear factor (NF)-κB (RANK) ligand (RANK-L), responsible for the differentiation, proliferation, and activation of osteoclasts. RANK-L binds its receptor RANK, localized on the surface of preosteoclasts and mature osteoclasts promoting osteoclastogenesis. High levels of RANK-L were demonstrated in active RA patients. Denosumab, a fully human monoclonal antibody, binds RANK-L and suppresses the RANK-RANK-L signaling pathway leading to the inhibition of osteoclastogenesis. A retrospective analysis of published studies such as clinical trials evidenced the efficacy of denosumab in preventing bone erosion progression in RA patients. Key messages Key questions to answer in future include the following: Could denosumab be associated with other biologic therapies in RA patients? Could denosumab block the progression of bone damage in RA? Could denosumab be used for the prevention of bone erosion in RA?
Collapse
Affiliation(s)
- Silvia Stefania
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| | - Cinzia Rotondo
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| | - Angiola Mele
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| | - Antonello Trotta
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| | - Francesco Paolo Cantatore
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| | - Addolorata Corrado
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, 71122, Italy
| |
Collapse
|
5
|
Manoharan AL, Jagadeesan G, Nataraj G, Muniyandi K, Guruswami G, Arunachalam K, Thangaraj P. Efficacy of Trevesia palmata (Roxb. ex Lindl.) Vis. Extract on MG 63 cell lines and arthritis-induced animal models. JOURNAL OF ETHNOPHARMACOLOGY 2023; 300:115742. [PMID: 36152784 DOI: 10.1016/j.jep.2022.115742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Despite widespread use of herbal remedies for treating arthritis and osteosarcoma, many plants are still not pharmacologically evaluated for their efficacy. Contrary to many non-steroidal, immunosuppressants, antibiotics, and antineoplastic drugs that have adverse effects, phytotherapeutic compounds have promising benefits with fewer complications. In this study the unexplored Northeastern India indigenous plant Trevesia palmata (Roxb. ex Lindl.) Vis. used in traditional medicine to cure bone fractures is chosen for studying anti-proliferative and anti-rheumatic properties. AIM OF THE STUDY This study designed to explore the polyphenolic composition, antioxidant, anti-inflammatory and anti-arthritic potential of T. palmata leaf extracts. Further, the cellular activity was studied using MG 63 osteoblast cell lines and pharmacologically evaluated using Complete Freund's Adjuvant (CFA) induced arthritic rat model. MATERIALS AND METHODS In vitro free radical scavenging activity, anti-inflammatory and anti-arthritic activities of extracts were analyzed using standardized methods. The polyphenolic profiling and apoptosis inducing ability of T. palmata ethyl acetate (TPEA) extract on MG 63 osteoblast cell lines were analyzed. The in vivo pharmacological studies were carried out with low dose 250 mg/kg and high dose of 500 mg/kg of T. palmata. The biochemical and haematological parameters and in vivo antioxidant activity were evaluated for the control and treated groups. Radiological and histological study were done to understand the impact and penetration of inflammatory arthritis from tissues to joint bones. RESULTS TPEA showed highest free radical scavenging activity (DPPH - 4.72 IC50, ABTS - 242.33 ± 6.81 mM TE/g extract), anti-inflammatory (40.04% inhibition of RBC lysis) and anti-arthritic activity (32.4% inhibition of protein denaturation) with the presence of gallic acid, catechin, caffeic acid, rutin, quercetin and naringenin. The TPEA extract inhibited cell proliferation of MG 63 osteoblast cells and induced apoptosis by arresting cell cycle at different phases. After acute toxicity studies the doses 250 mg/kg and 500 mg/kg were fixed and showed better results in CFA-induced arthritic animals. Thus, the extract phytoconstituents may have immense potential against chronic inflammation, joint ailments, bone cancer and arthritis which serves as a phytomedicine contrary to synthetic medications. CONCLUSIONS The potential treatment of polyphenolic compounds in the T. palmata extract on osteosarcoma and arthritis was demonstrated from this study. Thus, cellular inflammatory infiltrates are significantly reduced in bone and joint tissues as well.
Collapse
Affiliation(s)
- Ashwini Lydia Manoharan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| | - Gayathri Jagadeesan
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| | - Gayathri Nataraj
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| | - Kasipandi Muniyandi
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| | | | - Karuppusamy Arunachalam
- Centro de Estudos em Células Tronco, Terapia Celular e Genética Toxicológica (CeTroGen), Faculdade de Medicina (FAMED), Universidade Federal de Mato Grosso do Sul (UFMS), Campo Grande, Mato Grosso do Sul, Brazil; Programa de Pós-graduação em Saúde e Desenvolvimento na Região Centro-Oeste, Faculdade de Medicina Dr. Hélio Mandetta (FAMED), Universidade Federal de Mato Grosso do Sul (UFMS), Campo Grande, Mato Grosso do Sul, Brazil.
| | - Parimelazhagan Thangaraj
- Bioprospecting Laboratory, Department of Botany, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| |
Collapse
|
6
|
Harpagoside attenuates local bone Erosion and systemic osteoporosis in collagen-induced arthritis in mice. BMC Complement Med Ther 2022; 22:214. [PMID: 35948905 PMCID: PMC9364518 DOI: 10.1186/s12906-022-03694-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 08/01/2022] [Indexed: 11/10/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that causes local bone erosion and systemic osteoporosis. Harpagoside (HAR), an iridoid glycoside, has various pharmacological effects on pain, arthritis, and inflammation. Our previous study suggests that HAR is more deeply involved in the mechanism of bone loss caused by inflammatory stimuli than hormonal changes. Here, we identified the local and systemic bone loss inhibitory effects of HAR on RA and its intracellular mechanisms using a type 2 collagen-induced arthritis (CIA) mouse model. Methods The anti-osteoporosis and anti-arthritic effects of HAR were evaluated on bone marrow macrophage in vitro and CIA in mice in vivo by obtaining clinical scores, measuring hind paw thickness and inflammatory cytokine levels, micro-CT and histopathological assessments, and cell-based assay. Results HAR markedly reduced the clinical score and incidence rate of CIA in both the prevention and therapy groups. Histological analysis demonstrated that HAR locally ameliorated the destruction of bone and cartilage and the formation of pannus. In this process, HAR decreased the expression of inflammatory cytokines, such as tumor necrosis factor-α, interleukin (IL)-6, and IL-1β in the serum of CIA mice. Additionally, HAR downregulated the expression of receptor activator of nuclear factor-κB ligand and upregulated that of osteoprotegerin. HAR suppressed systemic bone loss by inhibiting osteoclast differentiation and osteoclast marker gene expression in a CIA mouse model. Conclusions Taken together, these findings show the beneficial effect of HAR on local symptoms and systemic bone erosion triggered by inflammatory arthritis. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-022-03694-y.
Collapse
|
7
|
Nooreen R, Nene S, Jain H, Prasannanjaneyulu V, Chitlangya P, Otavi S, Khatri DK, Raghuvanshi RS, Singh SB, Srivastava S. Polymer nanotherapeutics: A versatile platform for effective rheumatoid arthritis therapy. J Control Release 2022; 348:397-419. [PMID: 35660632 DOI: 10.1016/j.jconrel.2022.05.054] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 11/27/2022]
Abstract
Rheumatoid arthritis is an aggressive and severely debilitating disorder that is characterized by joint pain and cartilage damage. It restricts mobility in patients, leaving them unable to carry out simple tasks. RA presents itself with severe lasting pain, swelling and stiffness in the joints and may cause permanent disability in patients. Treatment regimens currently employed for rheumatoid arthritis revolve around keeping clinical symptoms like joint pain, inflammation, swelling and stiffness at bay. The current therapeutic interventions in rheumatoid arthritis involve the use of non-steroidal anti-inflammatory drugs, glucocorticoids, disease-modifying anti-rheumatic drugs and newer biological drugs that are engineered for inhibiting the expression of pro-inflammatory mediators. These conventional drugs are plagued with severe adverse effects because of their higher systemic distribution, lack of specificity and higher doses. Oral, intra-articular, and intravenous routes are routinely used for drug delivery which is associated with decreased patient compliance, high cost, poor bioavailability and rapid systemic clearance. All these drawbacks have enticed researchers to create novel strategies for drug delivery, the main approach being nanocarrier-based systems. In this article, we aim to consolidate the remarkable contributions of polymeric carrier systems including microneedle technology and smart trigger-responsive polymeric carriers in the management of rheumatoid arthritis along with its detailed pathophysiology. This review also briefly describes the safety and regulatory aspects of polymer therapeutics.
Collapse
Affiliation(s)
- Rimsha Nooreen
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Shweta Nene
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Harsha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Velpula Prasannanjaneyulu
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Palak Chitlangya
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Shivam Otavi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Dharmendra Kumar Khatri
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Rajeev Singh Raghuvanshi
- Indian Pharmacopoeia Commission, Ministry of Health & Family Welfare, Government of India, India
| | - Shashi Bala Singh
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana 500037, India.
| |
Collapse
|
8
|
Wang X, Sun T, Mao X. mascRNA promotes macrophage apoptosis, inhibits osteoclast differentiation and attenuates disease progression in a murine model of arthritis. Biochem Biophys Res Commun 2022; 611:151-157. [PMID: 35489201 DOI: 10.1016/j.bbrc.2022.04.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/12/2022] [Accepted: 04/19/2022] [Indexed: 11/17/2022]
Abstract
Macrophages play a crucial role in the pathogenesis of rheumatoid arthritis (RA) and have been considered as a therapeutic target of this disease. Here we show that mascRNA, a tRNA-like cytoplasmic small noncoding RNA, promoted RIPK1-dependent apoptosis (RDA) in RAW267.4 macrophages in response to the TAK1 inhibitor 5Z-7-oxozeaenol (5Z-7) alone as well as in combination with TNF. Moreover, mascRNA suppressed RANKL-induced expression of osteoclast marker genes and attenuated RANKL signaling. Using a murine model of collagen-induced arthritis (CIA), we demonstrated that mascRNA, administered either alone or in combination with 5Z-7, alleviated joint inflammation in CIA mice. Thus, mascRNA might be a promising agent for the treatment of RA.
Collapse
Affiliation(s)
- Xuxu Wang
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Tao Sun
- School of Life Science and Technology, Key Laboratory of Ministry of Education for Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu, China
| | - Xiaohua Mao
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, Jiangsu, China; School of Life Science and Technology, Key Laboratory of Ministry of Education for Developmental Genes and Human Disease, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
9
|
Ebeling PR, Nguyen HH, Aleksova J, Vincent AJ, Wong P, Milat F. Secondary Osteoporosis. Endocr Rev 2022; 43:240-313. [PMID: 34476488 DOI: 10.1210/endrev/bnab028] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 02/07/2023]
Abstract
Osteoporosis is a global public health problem, with fractures contributing to significant morbidity and mortality. Although postmenopausal osteoporosis is most common, up to 30% of postmenopausal women, > 50% of premenopausal women, and between 50% and 80% of men have secondary osteoporosis. Exclusion of secondary causes is important, as treatment of such patients often commences by treating the underlying condition. These are varied but often neglected, ranging from endocrine to chronic inflammatory and genetic conditions. General screening is recommended for all patients with osteoporosis, with advanced investigations reserved for premenopausal women and men aged < 50 years, for older patients in whom classical risk factors for osteoporosis are absent, and for all patients with the lowest bone mass (Z-score ≤ -2). The response of secondary osteoporosis to conventional anti-osteoporosis therapy may be inadequate if the underlying condition is unrecognized and untreated. Bone densitometry, using dual-energy x-ray absorptiometry, may underestimate fracture risk in some chronic diseases, including glucocorticoid-induced osteoporosis, type 2 diabetes, and obesity, and may overestimate fracture risk in others (eg, Turner syndrome). FRAX and trabecular bone score may provide additional information regarding fracture risk in secondary osteoporosis, but their use is limited to adults aged ≥ 40 years and ≥ 50 years, respectively. In addition, FRAX requires adjustment in some chronic conditions, such as glucocorticoid use, type 2 diabetes, and HIV. In most conditions, evidence for antiresorptive or anabolic therapy is limited to increases in bone mass. Current osteoporosis management guidelines also neglect secondary osteoporosis and these existing evidence gaps are discussed.
Collapse
Affiliation(s)
- Peter R Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia
| | - Hanh H Nguyen
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Department of Endocrinology and Diabetes, Western Health, Victoria 3011, Australia
| | - Jasna Aleksova
- Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Amanda J Vincent
- Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Monash Centre for Health Research and Implementation, School of Public Health and Preventative Medicine, Monash University, Clayton, Victoria 3168, Australia
| | - Phillip Wong
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Frances Milat
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Endocrinology, Monash Health, Clayton, Victoria 3168, Australia.,Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| |
Collapse
|
10
|
Chan YH, Ngai MC, Chen Y, Wu MZ, Yu YJ, Zhen Z, Lai K, Chung HY, Lau CS, Tse HF, Yiu KH. Osteogenic Circulating Endothelial Progenitor Cells are Associated with Vascular Aging of the Large Arteries in Rheumatoid Arthritis. Clin Interv Aging 2022; 17:287-294. [PMID: 35321150 PMCID: PMC8937309 DOI: 10.2147/cia.s337118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 01/18/2022] [Indexed: 11/25/2022] Open
Abstract
Background and Aim Rheumatoid arthritis is associated with both abnormal bone metabolism and accelerated vascular aging but a mechanistic link was lacking. This study aims to investigate the role of osteocalcin (OCN)-expressing circulating endothelial progenitor cells (EPCs) in vascular aging, as determined by arterial calcifications in rheumatoid arthritis. Methods We performed flow cytometry studies in 145 consecutive patients with rheumatoid arthritis to determine osteogenic circulating levels of OCN-positive (OCN+) CD34+KDR+ and OCN+CD34+ versus conventional early EPC CD34+CD133+KDR+. Total calcium load of the thoracic aorta (ascending plus descending) and the carotid arteries were assessed by non-contrast computed tomography (CT) and contrast CT angiography. Results Osteogenic EPCs OCN+CD34+KDR+ (P = 0.002) and OCN+CD34+ (P = 0.001), together with clinical parameters of age, history of hypertension, systolic blood pressure, serum levels of triglycerides, HbA1c and creatinine, use of leflunomide and brachial-ankle pulse-wave velocity (all P < 0.05), were associated with the clustered presence of aortic and carotid calcification. Multivariable analyses revealed that circulating OCN+CD34+KDR+ (B = 14.4 [95% CI 4.0 to 24.8], P = 0.007) and OCN+CD34+ (B = 9.6 [95% CI 4.9 to 14.3], P < 0.001) remained independently associated with increased aortic calcium load. OCN+CD34+ EPC (B = 0.8 [95% CI 0.1 to 1.5], P = 0.023), but not OCN+CD34+KDR+ EPC (B = 1.2 [95% CI −0.2 to 2.6], P = 0.09), was further independently associated with carotid calcium load. In comparison, conventional early EPC CD34+CD133+KDR+ had no significant association with aortic or carotid calcium load (P = 0.46 and 0.88, respectively). Conclusion Circulating level of osteogenic EPC is associated with increased vascular aging in terms of calcification of the large arteries in patients with rheumatoid arthritis. The findings may suggest a role of the bone-vascular axis underlying vascular aging in rheumatic diseases. Further research is needed to characterize the mechanistic links and basis of these observations.
Collapse
Affiliation(s)
- Yap-Hang Chan
- Cardiology Division, Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People’s Republic of China
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, Special Administrative Regions, People’s Republic of China
| | - Michael Cheong Ngai
- Division of Hematology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, Special Administrative Regions, People’s Republic of China
| | - Yan Chen
- Cardiology Division, Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People’s Republic of China
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, Special Administrative Regions, People’s Republic of China
| | - Mei-Zhen Wu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, Special Administrative Regions, People’s Republic of China
| | - Yu-Juan Yu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, Special Administrative Regions, People’s Republic of China
| | - Zhe Zhen
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, Special Administrative Regions, People’s Republic of China
| | - Kevin Lai
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, Special Administrative Regions, People’s Republic of China
| | - Ho-Yin Chung
- Division of Rheumatology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, Special Administrative Regions, People’s Republic of China
| | - Chak-Sing Lau
- Division of Rheumatology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, Special Administrative Regions, People’s Republic of China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People’s Republic of China
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, Special Administrative Regions, People’s Republic of China
| | - Kai-Hang Yiu
- Cardiology Division, Department of Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People’s Republic of China
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong, Special Administrative Regions, People’s Republic of China
- Correspondence: Kai-Hang Yiu, Cardiology Division, Department of Medicine, The University of Hong Kong-Shenzhen Hospital, 1 Haiyuan 1st Road, Futian District, Shenzhen, 518053, Guangdong, People’s Republic of China, Tel +852 22553633, Fax +852 28186304, Email
| |
Collapse
|
11
|
Jiang H, Xu F, Zeng L, Li C, Chen Y, Wang L, Li Z, Liu R. Saponins from Nigella glandulifera seeds attenuate collagen-induced rheumatoid arthritis in rats via the OPG/RANKL/NF-κB and Ang/Tie-2 pathways. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114714. [PMID: 34624452 DOI: 10.1016/j.jep.2021.114714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/25/2021] [Accepted: 10/04/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nigella glandulifera Freyn et Sint. (N. glandulifera) seeds are widely used in traditional Uyghur medicine for a variety of immuno-inflammatory diseases. The total saponins from N. glandulifera seeds (TSNGS) have been shown to have analgesic, antioxidant, and anti-inflammatory effects that can alleviate joint pain and swelling. AIM OF THE STUDY Rheumatoid arthritis (RA) is a chronic and progressive, debilitating autoimmune disease for which current treatments are not sufficiently effective and result in unsatisfactory side effects. This study aimed to mechanistically investigate the therapeutic effects of TSNGS on RA. MATERIALS AND METHODS Qualitative analysis of TSNGS was performed using ultra-high-performance liquid chromatography-Q-Orbitrap-high-resolution mass spectrometry. Rats with collagen-induced arthritis (CIA), IL-1β-induced HFLS-RAs, and VEGF-induced HUVECs were analyzed to determine the efficacy and mechanism of TSNGS on RA. RESULTS Twenty-one compounds were identified in TSNGS. TSNGS (10, 50, or 250 mg/kg) reduced the severity of arthritis, indicated by a lower arthritis score, reduced paw swelling, and body weight in rats with CIA. TSNGS ameliorated histopathological changes involving inflammatory infiltration, bone degeneration, and angiogenesis in knee and ankle joints. TSNGS improved the immuno-inflammatory response by restoring the levels of the cytokines IFN-γ, TNF-α, IL-1β, IL-6, IL-17A, IL-4, and IL-10, and increasing the number of CD4+CD25+ Tregs in the peripheral circulation and Foxp3 levels in knee joints in rats with CIA. Furthermore, TSNGS increased the OPG/RANKL ratio and downregulated p-p65 in serum and joint synovia. Inhibition of angiogenesis by TSNGS was associated with recovery of the angiogenesis-related Ang/Tie-2 signaling pathway. CONCLUSIONS It was established that TSNGS provides a therapeutic effect on RA by alleviating synovitis, bone degeneration, and angiogenesis via the OPG/RANKL/NF-κB and Ang/Tie-2 pathways and may be used for the treatment of RA.
Collapse
Affiliation(s)
- Hailun Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Fang Xu
- Key Laboratory of Uighur Medicine of Xinjiang Uygur Autonomous Region, Xinjiang Institute of Materia Medica, Urumqi, 830004, PR China
| | - Li Zeng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Chenyang Li
- Key Laboratory of Uighur Medicine of Xinjiang Uygur Autonomous Region, Xinjiang Institute of Materia Medica, Urumqi, 830004, PR China
| | - Yan Chen
- Key Laboratory of Uighur Medicine of Xinjiang Uygur Autonomous Region, Xinjiang Institute of Materia Medica, Urumqi, 830004, PR China
| | - Linlin Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China
| | - Zhuorong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Rui Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| |
Collapse
|
12
|
Perez MO, Figueiredo CP, Sales LP, Medeiros-Ribeiro AC, Takayama L, Domiciano DS, Bonfiglioli K, Caparbo VF, Pereira RM. Bone erosions and osteophytes in premenopausal women with long-standing rheumatoid arthritis: association with systemic bone involvement by HR-Pqct. Arthritis Rheumatol 2021; 74:407-417. [PMID: 34463054 DOI: 10.1002/art.41961] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 07/13/2021] [Accepted: 08/26/2021] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To evaluate premenopausal women with long-standing RA and to explore the relationship between parameters of systemic and localized bone involvement. METHODS Eighty consecutive RA premenopausal women were evaluated. Volumetric bone mineral density (vBMD), microarchitecture and finite element analysis were performed using high-resolution peripheral quantitative computed tomography (HR-pQCT) at the distal radius (DR) and tibia (DT) and compared with parameters from 160 female healthy controls matched by age and body mass index. Localized bone involvement was also analyzed using HR-pQCT in the metacarpophalangeal and proximal interphalangeal joints to identify erosions and osteophytes. RESULTS The mean age of RA patients was 39.4±6.7years and disease duration 9.8±5.3years. RA patients had impaired trabecular, cortical and bone strength parameters, at DR and DT, compared with healthy controls (p<0.05). Bone erosions and osteophytes were found in 75% and 41.3% of patients, respectively. Comparing patients with and without erosions, at DR and DT, a lower cortical vBMD (DR: 980±72 versus 1021±47mgHA/cm3 , p=0.03; DT:979±47 versus 1003±34 mgHA/cm3 , p=0.04) and higher cortical porosity (DR:2.8±2.5 versus 1.8±1.6%, p=0.04; and DT:3.7±1.6 versus 2.7±1.6%, p=0.01) were observed in patients with erosions. At the DR, osteophyte volume was positively correlated with trabecular vBMD (0.392, p=0.02), trabecular number (0.381, p=0.03) and stiffness (0.411, p=0.02), and negatively with trabecular separation (-0.364, p=0.04). CONCLUSION Our findings showed that premenopausal women with long-standing RA had systemic bone fragility at peripheral sites. Moreover, erosions were associated mainly with cortical bone fragility at the DR and DT, and osteophytes correlated with repair of trabecular bone at the radius.
Collapse
Affiliation(s)
- Mariana O Perez
- Division of Rheumatology, Hospital das Clinicas HCFMUSP Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Camille P Figueiredo
- Division of Rheumatology, Hospital das Clinicas HCFMUSP Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Lucas P Sales
- Division of Rheumatology, Hospital das Clinicas HCFMUSP Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Ana Cristina Medeiros-Ribeiro
- Division of Rheumatology, Hospital das Clinicas HCFMUSP Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Liliam Takayama
- Division of Rheumatology, Hospital das Clinicas HCFMUSP Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Diogo S Domiciano
- Division of Rheumatology, Hospital das Clinicas HCFMUSP Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Karina Bonfiglioli
- Division of Rheumatology, Hospital das Clinicas HCFMUSP Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Valeria F Caparbo
- Division of Rheumatology, Hospital das Clinicas HCFMUSP Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Rosa Mr Pereira
- Division of Rheumatology, Hospital das Clinicas HCFMUSP Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
13
|
Murata K, Uozumi R, Hashimoto M, Ebina K, Akashi K, Onishi A, Nagai K, Yoshikawa A, Katayama M, Son Y, Amuro H, Hara R, Yamamoto W, Watanabe R, Murakami K, Tanaka M, Ito H, Morinobu A, Matsuda S. The real-world effectiveness of anti-RANKL antibody denosumab on the clinical fracture prevention in patients with rheumatoid arthritis: The ANSWER cohort study. Mod Rheumatol 2021; 32:834-838. [PMID: 34910199 DOI: 10.1093/mr/roab043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 11/14/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by localized and generalized bone loss. The risk of fractures is doubled in patients with RA. Denosumab, an anti-RANKL monoclonal antibody, is used for those with osteoporosis at high risk fracture and it has inhibitory effect of progressive bone erosion in patients with RA. While the increase in bone mineral density by denosumab has been reported in patients with RA, preventive effect of fracture by denosumab remains unknown. This study aimed to evaluate the efficacy of denosumab in treating clinical fracture risk in patients with RA. METHODS Patients with RA who received denosumab treatment between 2013 and 2019 were retrospectively evaluated using the ANSWER (Kansai Consortium for the Well-Being of Rheumatic Disease Patients) cohort data. Fracture rates were evaluated between 0 and 6 months (reference period) versus > 6 months (post-reference period) of denosumab use. RESULTS A total of 873 patients with RA received denosumab, and their characteristics were as follows: 88% females, mean age 68 years, and average disease duration 14.5 years. The hazard rates of all clinical fractures were 0.69 (per 100 person-years) in the reference period and 0.35 in the post-reference period, indicating a 49.2% decrease (p = 0.03). CONCLUSIONS Denosumab suppresses the risk of clinical fractures in patients with RA.
Collapse
Affiliation(s)
- Koichi Murata
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryuji Uozumi
- Department of Biomedical and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Motomu Hashimoto
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kosuke Ebina
- Department of Musculoskeletal Regenerative Medicine, Osaka University, Graduate School of Medicine, Osaka, Japan
| | - Kengo Akashi
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Akira Onishi
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Koji Nagai
- Department of Internal Medicine (IV), Osaka Medical College, Osaka, Japan
| | - Ayaka Yoshikawa
- Department of Internal Medicine (IV), Osaka Medical College, Osaka, Japan
| | - Masaki Katayama
- Department of Rheumatology, Osaka Red Cross Hospital, Osaka, Japan
| | - Yonsu Son
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Hideki Amuro
- First Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Ryota Hara
- The Center for Rheumatic Diseases, Department of Orthopaedic Surgery, Nara Medical University, Nara, Japan
| | - Wataru Yamamoto
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Health Information Management, Kurashiki Sweet Hospital, Okayama, Japan
| | - Ryu Watanabe
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kosaku Murakami
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masao Tanaka
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiromu Ito
- Department of Advanced Medicine for Rheumatic Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
14
|
Guo J, Zhai W, Wei L, Zhang J, Jin L, Yan H, Huang Z, Jia Y. Radiological and clinical outcomes of balloon kyphoplasty for osteoporotic vertebral compression fracture in patients with rheumatoid arthritis. J Orthop Surg Res 2021; 16:435. [PMID: 34229715 PMCID: PMC8259446 DOI: 10.1186/s13018-021-02573-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/20/2021] [Indexed: 11/25/2022] Open
Abstract
Background This study was conducted to investigate the outcomes and complications of balloon kyphoplasty (KP) for the treatment of osteoporotic vertebral compression fracture (OVCF) in patients with rheumatoid arthritis (RA) and compare its radiological and clinical effects with OVCF patients without RA. Methods Ninety-eight patients in the RA group with 158 fractured vertebrae and 114 patients in the control group with 150 vertebrae were involved in this study. Changes in compression rate, local kyphotic angle, visual analog scale (VAS) and Oswestry disability index (ODI) scores, conditions of bone cement leakage, refracture of the operated vertebrae, and new adjacent vertebral fractures were examined after KP. In addition, patients in the RA group were divided into different groups according to the value of erythrocyte sedimentation rate (ESR), c-reactive protein (CRP), and whether they were glucocorticoid users or not to evaluate their influence on the outcomes of KP. Results KP procedure significantly improved the compression rate, local kyphotic angle, and VAS and ODI scores in both RA and control groups (p<0.05). Changes in compression rate and local kyphotic angle in the RA group were significantly larger than that in the control group (p<0.05), and patients with RA suffered more new adjacent vertebral fractures after KP. The outcomes and complications of KP from different ESR or CRP groups did not show significant differences. The incidence of cement leakage in RA patients with glucocorticoid use was significantly higher than those who did not take glucocorticoids. In addition, RA patients with glucocorticoid use suffered more intradiscal leakage and new adjacent vertebral fractures. Conclusions OVCF patients with RA obtained more improvement in compression rate and local kyphotic angle after KP when compared to those without RA, but they suffered more new adjacent vertebral fractures. Intradiscal leakage and new adjacent vertebral fractures occurred more in RA patients with glucocorticoid use. Trial registration Retrospectively registered.
Collapse
Affiliation(s)
- Ji Guo
- Department of Spine Surgery, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.540 Xinhua Road, Shanghai, 200052, China.,Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, 200052, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Weifeng Zhai
- Department of Spine Surgery, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.540 Xinhua Road, Shanghai, 200052, China.,Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, 200052, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Licheng Wei
- Department of Spine Surgery, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.540 Xinhua Road, Shanghai, 200052, China.,Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, 200052, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Jianpo Zhang
- Department of Spine Surgery, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.540 Xinhua Road, Shanghai, 200052, China.,Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, 200052, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Lang Jin
- Department of Spine Surgery, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.540 Xinhua Road, Shanghai, 200052, China.,Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, 200052, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Hao Yan
- Department of Spine Surgery, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.540 Xinhua Road, Shanghai, 200052, China.,Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, 200052, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Zheng Huang
- Department of Spine Surgery, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.540 Xinhua Road, Shanghai, 200052, China.,Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, 200052, China.,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Yongwei Jia
- Department of Spine Surgery, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.540 Xinhua Road, Shanghai, 200052, China. .,Shanghai Guanghua Hospital of Integrative Medicine, Shanghai, 200052, China. .,Institute of Arthritis Research in Integrative Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200052, China.
| |
Collapse
|
15
|
Tanaka Y, Soen S, Ishiguro N, Yamanaka H, Yoneda T, Tanaka S, Ohira T, Nitta T, Okubo N, Genant H, van der Heijde D, Takeuchi T. Identifying the preferable rheumatoid arthritis subgroups for intervention with the anti-RANKL antibody denosumab to reduce progression of joint destruction. RMD Open 2021; 6:rmdopen-2020-001249. [PMID: 32732353 PMCID: PMC7722273 DOI: 10.1136/rmdopen-2020-001249] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/06/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES To clarify which rheumatoid arthritis (RA) patients benefit most from the anti-receptor activator of nuclear factor-κB ligand antibody denosumab to reduce the progression of joint destruction. METHODS We pooled patient data from the 12-month, double-blind, placebo-controlled DRIVE (phase II) and DESIRABLE (phase III) studies. In DRIVE, concomitant treatment was limited to methotrexate, salazosulfapyridine and bucillamine. In DESIRABLE, patients could receive any disease-modifying antirheumatic drug. RA patients were randomised to denosumab 60 mg every 6 months (Q6M), every 3 months (Q3M) or placebo. Efficacy was assessed by van der Heijde-modified total Sharp score (mTSS), bone erosion score (ES) and joint space narrowing score (JSNS). Change in mTSS was assessed in subgroups stratified by risk factors for radiographic damage if the interaction factor was significant. RESULTS The pooled analysis included 909 patients. Denosumab reduced worsening of mTSS (mean (SD)) at 12 months in the Q6M (0.88 (3.30), p=0.0024) and Q3M (0.66 (2.16), p=0.0002) groups versus placebo (1.50 (3.73)). This reduction in mTSS progression was due to the change in ES (Q6M, 0.44 (1.89), p=0.0006; Q3M, 0.20 (0.86), p<0.0001) versus placebo (0.98 (2.54)); no effect was observed on JSNS. Anti-cyclic citrullinated peptide (CCP) antibodies, glucocorticoid use and baseline ES showed a significant interaction. Denosumab was particularly effective in patients who were anti-CCP antibody positive (p<0.05). Changes in mTSS versus placebo were observed in all denosumab dose groups, regardless of glucocorticoid use and baseline ES. CONCLUSIONS Denosumab broadly reduced the progression of joint destruction in RA patients with risk factors for radiographic damage such as especially anti-CCP antibody positivity.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Satoshi Soen
- Department of Orthopaedic Surgery and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan
| | - Naoki Ishiguro
- Department of Orthopaedics, Nagoya University Graduate School of Medicine, Aichi, Japan
| | | | - Toshiyuki Yoneda
- Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, The University of Tokyo, Tokyo, Japan
| | - Takeshi Ohira
- Clinical Development Department, Daiichi Sankyo Co Ltd, Tokyo, Japan
| | - Takaya Nitta
- Clinical Development Department, Daiichi Sankyo Co Ltd, Tokyo, Japan
| | - Naoki Okubo
- Data Intelligence Department, Daiichi Sankyo Co Ltd, Tokyo, Japan
| | - Harry Genant
- Departments of Radiology, University of California San Francisco, California, USA
| | | | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
Tanaka Y, Takeuchi T, Soen S, Yamanaka H, Yoneda T, Tanaka S, Nitta T, Okubo N, Genant HK, van der Heijde D. Effects of Denosumab in Japanese Patients With Rheumatoid Arthritis Treated With Conventional Antirheumatic Drugs: 36-month Extension of a Phase III Study. J Rheumatol 2021; 48:1663-1671. [PMID: 33858976 DOI: 10.3899/jrheum.201376] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To evaluate the safety and efficacy of long-term denosumab 60 mg every 6 months (Q6M) or every 3 months (Q3M) in patients with rheumatoid arthritis (RA). METHODS This 12-month, randomized, double-blind, placebo-controlled, multicenter, phase III trial with an open-label extension period from 12 to 36 months (DESIRABLE) enrolled Japanese patients with RA treated with placebo (P) for 12 months followed by either denosumab Q6M (P/Q6M) or denosumab Q3M (P/Q3M) for 24 months; denosumab Q6M for 36 months (Q6M/Q6M); or denosumab Q3M for 36 months (Q3M/Q3M). Efficacy was assessed by van der Heijde modified total Sharp score (mTSS), bone erosion score (BES), and joint space narrowing ( JSN) score. RESULTS Long-term treatment better maintained mTSS and BES suppression in the P/Q3M and Q3M/ Q3M vs P/Q6M and Q6M/Q6M groups; changes from baseline in total mTSS (standard error) at 36 months were 2.8 (0.4) and 1.7 (0.3) vs 3.0 (0.4) and 2.4 (0.3), respectively, and corresponding changes in BES were 1.3 (0.2) and 0.4 (0.2) vs 1.4 (0.2) and 1.1 (0.2), respectively. No JSN effect was observed. Bone mineral density consistently increased in all groups after denosumab initiation, regardless of concomitant glucocorticoid administration. Serum C-terminal telopeptide of type I collagen decreased rapidly at 1 month postdenosumab administration (in both the initial 12-month [Q3M and Q6M groups] and long-term treatment [P/Q3M and P/Q6M groups] phases). Adverse event incidence leading to study drug discontinuation was similar across treatment groups. CONCLUSION Denosumab treatment maintained inhibition of progression of joint destruction up to 36 months. Based on effects on BES progression, higher dosing frequency at an earlier treatment stage may be needed to optimize treatment. Denosumab was generally well tolerated. (ClinicalTrials.gov: NCT01973569).
Collapse
Affiliation(s)
- Yoshiya Tanaka
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Tsutomu Takeuchi
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Satoshi Soen
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Hisashi Yamanaka
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Toshiyuki Yoneda
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Sakae Tanaka
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Takaya Nitta
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Naoki Okubo
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Harry K Genant
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| | - Désirée van der Heijde
- This study was funded by Daiichi Sankyo Co., Ltd. Y. Tanaka, MD, PhD, First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan; T. Takeuchi, MD, PhD, Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan; S. Soen, MD, PhD, Department of Orthopedics and Rheumatology, Kindai University Nara Hospital, Ikoma, Japan; H. Yamanaka, MD, PhD, Director of Rheumatology, Sanno Medical Center, Tokyo, Japan; T. Yoneda, DDS, PhD, Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan; S. Tanaka, MD, PhD, Orthopedic Surgery and Spinal Surgery, The University of Tokyo, Tokyo, Japan; T. Nitta, MS, N. Okubo, PhD, Daiichi Sankyo Co., Ltd, Japan; H.K. Genant, MD, Departments of Radiology, Medicine, and Orthopedic Surgery, University of California, San Francisco, California, USA; D. van der Heijde, MD, PhD, Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands. Y. Tanaka and T. Takeuchi contributed equally to this work. H.K. Genant passed away on January 14, 2021. YT has received speakers bureau fees from Daiichi Sankyo, Eli Lilly, Novartis, YL Biologics, BMS, Eisai, Chugai, AbbVie GK, Astellas, Pfizer, Sanofi, GSK, Mitsubishi Tanabe, Gilead, and Janssen. TT has received grant/research support from Daiichi Sankyo, Takeda, Nippon Kayaku, JCR Pharma, Astellas, Chugai, AbbVie GK, Asahi Kasei Pharma, Mitsubishi Tanabe, UCB Japan, and Eisai, and consulting fees from Astellas, AbbVie GK, Gilead, Daiichi Sankyo, Taisho Pharma, Nippon Kayaku, BMS, GSK, Eli Lilly, Novartis, Mitsubishi Tanabe, and Chugai, and speakers bureau fees from Mitsubishi Tanabe, Pfizer, Astellas, Daiichi Sankyo, Eisai, Sanofi, Novartis, Eli Lilly, Gilead, AbbVie GK, BMS, and Chugai. SS has received grant/ research support from Chugai and Daiichi Sankyo and has served on speakers bureaus for Asahi Kasei Pharma, Astellas, Daiichi Sankyo, Takeda, Chugai, Eisai, Pfizer, Eli Lilly, and Ono. HY has received speakers bureau fees from Pfizer, YL Biologics, Teijin, Astellas, Tanabe Mitsubishi, and BMS. TY has received grants-in-aid for scientific research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan (MEXT #17H04377) and has acted as a consultant for Daiichi Sankyo. ST has received grant/research support from Kyocera Corp and Asahi Kasei Corp and consulting fees from Amgen Astellas BioPharma KK, Kyocera Corp, Daiichi Sankyo, and Pfizer, and speakers bureau fees from Asahi Kasei Corp, Astellas, Ayumi, Eisai, Ono, Daiichi Sankyo, Taisho Pharma, Mitsubishi Tanabe, Chugai, Teijin, Eli Lilly, Hisamitsu, Pfizer, and BMS. TN and NO report fulltime employment with Daiichi Sankyo. HG has received consulting fees or other remuneration from Daiichi Sankyo, Amgen, Eli Lilly, Biomarin, Clementia, Agnovos, Regeneron, Medtronic, QED, and Bioclinica. DvdH has acted as a consultant for AbbVie, Amgen, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Cyxone, Daiichi Sankyo, Eisai, Eli Lilly, Galapagos, Gilead, GSK, Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi, Takeda, and UCB, and is a director at Imaging Rheumatology BV. Address correspondence to Dr. Y. Tanaka, First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan. . Accepted for publication April 8, 2021
| |
Collapse
|
17
|
Choi Y, Lee EG, Jeong JH, Yoo WH. 4-Phenylbutyric acid, a potent endoplasmic reticulum stress inhibitor, attenuates the severity of collagen-induced arthritis in mice via inhibition of proliferation and inflammatory responses of synovial fibroblasts. Kaohsiung J Med Sci 2021; 37:604-615. [PMID: 33759334 DOI: 10.1002/kjm2.12376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 02/02/2021] [Accepted: 02/07/2021] [Indexed: 11/11/2022] Open
Abstract
4-Phenylbutyric acid (4-PBA) exerts potent pharmacological effects, including anti-inflammatory properties, via inhibition of endoplasmic reticulum (ER) stress. However, it is not known whether 4-PBA attenuates the severity of rheumatoid arthritis. The present study aimed to determine whether the inhibition of ER stress by 4-PBA ameliorated experimentally induced arthritis. The proliferation of synovial fibroblasts (SFs) and expression of matrix metalloproteinases (MMPs) were evaluated in the presence of interleukin (IL)-1β with or without 4-PBA. The effect of 4-PBA on the phosphorylation of Mitogen-activated protein kinase (MAPK) and the activation of Nuclear factor-κB (NF-κB) in IL-1β-stimulated SFs was assessed. In an in vivo study, the effects of 4-PBA were investigated using DBA/1 mice with collagen-induced arthritis (CIA). Clinical, histological, and serological assessments of CIA treated with 4-PBA were performed to determine the therapeutic effect of 4-PBA. In vitro, 4-PBA inhibited the proliferation and expression of IL-1β-stimulated SFs and MMP-1 and MMP-3 through the suppression of both the phosphorylation of MAPKs and NF-κB in IL-1β-stimulated SFs. The 4-PBA treatment markedly attenuated the severity of arthritis in CIA mice. The 4-PBA treatment ameliorated joint swelling and the degree of bone erosion and destruction and decreased the level of inflammatory cytokines and MMP-3 and Cox-2. Furthermore, remarkable improvements in histopathological findings occurred in 4-PBA-treated mice. These findings suggested that 4-PBA could attenuate the severity of arthritis in CIA mice by partially blocking the phosphorylation of MAPKs and the activation of NF-κB in SFs. Thus, through the inhibition of ER stress, 4-PBA may be a potent agent for the treatment of RA.
Collapse
Affiliation(s)
- Yunjung Choi
- Division of Rheumatology, Department of Internal Medicine, Jeonbuk National University Hospital, Jeonju, Republic of Korea.,Research Institute of Clinical Medicine, Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea.,Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Eun-Gyeong Lee
- Research Institute of Clinical Medicine, Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea.,Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Ji-Hyeon Jeong
- Research Institute of Clinical Medicine, Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea.,Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| | - Wan-Hee Yoo
- Division of Rheumatology, Department of Internal Medicine, Jeonbuk National University Hospital, Jeonju, Republic of Korea.,Research Institute of Clinical Medicine, Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Republic of Korea.,Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, Republic of Korea
| |
Collapse
|
18
|
Managing Osteoporosis and Joint Damage in Patients with Rheumatoid Arthritis: An Overview. J Clin Med 2021; 10:jcm10061241. [PMID: 33802804 PMCID: PMC8002542 DOI: 10.3390/jcm10061241] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/06/2021] [Accepted: 03/12/2021] [Indexed: 01/04/2023] Open
Abstract
In rheumatoid arthritis, a representative systemic autoimmune disease, immune abnormality and accompanying persistent synovitis cause bone and cartilage destruction and systemic osteoporosis. Biologics targeting tumor necrosis factor, which plays a central role in the inflammatory process, and Janus kinase inhibitors have been introduced in the treatment of rheumatoid arthritis, making clinical remission a realistic treatment goal. These drugs can prevent structural damage to bone and cartilage. In addition, osteoporosis, caused by factors such as menopause, aging, immobility, and glucocorticoid use, can be treated with bisphosphonates and the anti-receptor activator of the nuclear factor-κB ligand antibody. An imbalance in the immune system in rheumatoid arthritis induces an imbalance in bone metabolism. However, osteoporosis and bone and cartilage destruction occur through totally different mechanisms. Understanding the mechanisms underlying osteoporosis and joint destruction in rheumatoid arthritis leads to improved care and the development of new treatments.
Collapse
|
19
|
The Role of Collagen Triple Helix Repeat-Containing 1 Protein (CTHRC1) in Rheumatoid Arthritis. Int J Mol Sci 2021; 22:ijms22052426. [PMID: 33670905 PMCID: PMC7957534 DOI: 10.3390/ijms22052426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 01/15/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease causing inflammation of joints, cartilage destruction and bone erosion. Biomarkers and new drug targets are actively sought and progressed to improve available options for patient treatment. The Collagen Triple Helix Repeat Containing 1 protein (CTHRC1) may have an important role as a biomarker for rheumatoid arthritis, as CTHRC1 protein concentration is significantly elevated in the peripheral blood of rheumatoid arthritis patients compared to osteoarthritis (OA) patients and healthy individuals. CTHRC1 is a secreted glycoprotein that promotes cell migration and has been implicated in arterial tissue-repair processes. Furthermore, high CTHRC1 expression is observed in many types of cancer and is associated with cancer metastasis to the bone and poor patient prognosis. However, the function of CTHRC1 in RA is still largely undefined. The aim of this review is to summarize recent findings on the role of CTHRC1 as a potential biomarker and pathogenic driver of RA progression. We will discuss emerging evidence linking CTHRC1 to the pathogenic behavior of fibroblast-like synoviocytes and to cartilage and bone erosion through modulation of the balance between bone resorption and repair.
Collapse
|
20
|
Yang B, Luo L, Chen L, Niu Q, Zhang J, Xu H, Wu Y, Huang Z. ETS1 polymorphism rs73013527 in relation to serum RANKL levels among patients with RA. Medicine (Baltimore) 2021; 100:e24562. [PMID: 33592912 PMCID: PMC7870260 DOI: 10.1097/md.0000000000024562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 01/11/2021] [Indexed: 02/05/2023] Open
Abstract
We previously identified E26 transformation specific sequence 1 (ETS1) rs73013527 single nucleotide polymorphism associated with RA susceptibility and disease activity. In the present study, we aims to further investigate the association between ETS1 rs73013527 and receptor activator of nuclear factor kappa B ligand (RANKL), an index related to bone destruction and was reported to elevate in RA.We determined genotypes of ETS1 rs73013527, serum RANKL concentration, clinical characteristics (disease duration, disease activity score for 28 painful/swollen joints), and laboratory markers (rheumatoid factor, anti-citrullinated protein antibody, anti-keratin antibody, c-reactive protein, erythrocyte sedimentation rate) of 254 RA cases. Univariate and multivariate analysis were employed to explore the association between ETS1 rs73013527 and serum RANKL levels in RA patients.Univariate and multivariate analysis indicated no association of serum RANKL levels with patient age, gender, clinical characteristics, and laboratory markers. Univariate analysis, not multivariate analysis indicated genotype CT/TT of ETS1 rs73013527 was significantly associated with elevated RANKL levels in RA patients.ETS1 rs73013527 is in relation to serum RANKL levels among patients with RA. ETS1 probably might be an indirect factors involved in RANKL regulation in RA.
Collapse
Affiliation(s)
- Bin Yang
- Department of Laboratory Medcine, West China Hospital, Sichuan University
| | - Limei Luo
- Department of Laboratory Medcine, West China Hospital, Sichuan University
| | - Lin Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qian Niu
- Department of Laboratory Medcine, West China Hospital, Sichuan University
| | - Junlong Zhang
- Department of Laboratory Medcine, West China Hospital, Sichuan University
| | - Huan Xu
- Department of Laboratory Medcine, West China Hospital, Sichuan University
| | - Yifeng Wu
- Department of Laboratory Medcine, West China Hospital, Sichuan University
| | - Zhuochun Huang
- Department of Laboratory Medcine, West China Hospital, Sichuan University
| |
Collapse
|
21
|
Damerau A, Gaber T, Ohrndorf S, Hoff P. JAK/STAT Activation: A General Mechanism for Bone Development, Homeostasis, and Regeneration. Int J Mol Sci 2020; 21:E9004. [PMID: 33256266 PMCID: PMC7729940 DOI: 10.3390/ijms21239004] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
The Janus kinase (JAK) signal transducer and activator of transcription (STAT) signaling pathway serves as an important downstream mediator for a variety of cytokines, hormones, and growth factors. Emerging evidence suggests JAK/STAT signaling pathway plays an important role in bone development, metabolism, and healing. In this light, pro-inflammatory cytokines are now clearly implicated in these processes as they can perturb normal bone remodeling through their action on osteoclasts and osteoblasts at both intra- and extra-articular skeletal sites. Here, we summarize the role of JAK/STAT pathway on development, homeostasis, and regeneration based on skeletal phenotype of individual JAK and STAT gene knockout models and selective inhibition of components of the JAK/STAT signaling including influences of JAK inhibition in osteoclasts, osteoblasts, and osteocytes.
Collapse
Affiliation(s)
- Alexandra Damerau
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany; (A.D.); (S.O.); (P.H.)
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Timo Gaber
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany; (A.D.); (S.O.); (P.H.)
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Sarah Ohrndorf
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany; (A.D.); (S.O.); (P.H.)
| | - Paula Hoff
- Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany; (A.D.); (S.O.); (P.H.)
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
- Endokrinologikum Berlin am Gendarmenmarkt, 10117 Berlin, Germany
| |
Collapse
|
22
|
Shatunova EA, Korolev MA, Omelchenko VO, Kurochkina YD, Davydova AS, Venyaminova AG, Vorobyeva MA. Aptamers for Proteins Associated with Rheumatic Diseases: Progress, Challenges, and Prospects of Diagnostic and Therapeutic Applications. Biomedicines 2020; 8:biomedicines8110527. [PMID: 33266394 PMCID: PMC7700471 DOI: 10.3390/biomedicines8110527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023] Open
Abstract
Nucleic acid aptamers capable of affine and specific binding to their molecular targets have now established themselves as a very promising alternative to monoclonal antibodies for diagnostic and therapeutic applications. Although the main focus in aptamers’ research and development for biomedicine is made on cardiovascular, infectious, and malignant diseases, the use of aptamers as therapeutic or diagnostic tools in the context of rheumatic diseases is no less important. In this review, we consider the main features of aptamers that make them valuable molecular tools for rheumatologists, and summarize the studies on the selection and application of aptamers for protein biomarkers associated with rheumatic diseases. We discuss the progress in the development of aptamer-based diagnostic assays and targeted therapeutics for rheumatic disorders, future prospects in the field, and issues that have yet to be addressed.
Collapse
Affiliation(s)
- Elizaveta A. Shatunova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.S.); (A.S.D.); (A.G.V.)
| | - Maksim A. Korolev
- Research Institute of Clinical and Experimental Lymphology, Affiliated Branch of Federal Research Center of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, 630060 Novosibirsk, Russia; (M.A.K.); (V.O.O.); (Y.D.K.)
| | - Vitaly O. Omelchenko
- Research Institute of Clinical and Experimental Lymphology, Affiliated Branch of Federal Research Center of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, 630060 Novosibirsk, Russia; (M.A.K.); (V.O.O.); (Y.D.K.)
| | - Yuliya D. Kurochkina
- Research Institute of Clinical and Experimental Lymphology, Affiliated Branch of Federal Research Center of Cytology and Genetics, Siberian Division of the Russian Academy of Sciences, 630060 Novosibirsk, Russia; (M.A.K.); (V.O.O.); (Y.D.K.)
| | - Anna S. Davydova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.S.); (A.S.D.); (A.G.V.)
| | - Alya G. Venyaminova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.S.); (A.S.D.); (A.G.V.)
| | - Mariya A. Vorobyeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Division of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.A.S.); (A.S.D.); (A.G.V.)
- Correspondence:
| |
Collapse
|
23
|
Hu H, Yang W, Zeng Q, Chen W, Zhu Y, Liu W, Wang S, Wang B, Shao Z, Zhang Y. Promising application of Pulsed Electromagnetic Fields (PEMFs) in musculoskeletal disorders. Biomed Pharmacother 2020; 131:110767. [PMID: 33152929 DOI: 10.1016/j.biopha.2020.110767] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/06/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence suggests that an exogenous electromagnetic field might be involved in many biologic processes which are of great importance for therapeutic interventions. Pulsed electromagnetic fields (PEMFs) are known to be a noninvasive, safe and effective therapy agent without apparent side effects. Numerous studies have shown that PEMFs possess the potential to become a stand-alone or adjunctive treatment modality for treating musculoskeletal disorders. However, several issues remain unresolved. Prior to their widely clinical application, further researches from well-designed, high-quality studies are still required to standardize the treatment parameters and derive the optimal protocol for health-care decision making. In this review, we aim to provide current evidence on the mechanism of action, clinical applications, and controversies of PEMFs in musculoskeletal disorders.
Collapse
Affiliation(s)
- Hongzhi Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China.
| | - Wenbo Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Qianwen Zeng
- Department of Pediatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Wei Chen
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - YanBin Zhu
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Weijian Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shangyu Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Baichuan Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yingze Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China.
| |
Collapse
|
24
|
Azouz AA, Saleh E, Abo-Saif AA. Aliskiren, tadalafil, and cinnamaldehyde alleviate joint destruction biomarkers; MMP-3 and RANKL; in complete Freund's adjuvant arthritis model: Downregulation of IL-6/JAK2/STAT3 signaling pathway. Saudi Pharm J 2020; 28:1101-1111. [PMID: 32922141 PMCID: PMC7474170 DOI: 10.1016/j.jsps.2020.07.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/28/2020] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease, which is accompanied by progressive joint damage and disability. The intolerability of conventional antirheumatic drugs by some patients necessitates the search for effective antirheumatic agents having better tolerability. In the current work, we aimed to investigate the efficacy of cinnamaldehyde, tadalafil, and aliskiren as potential antirheumatic candidates and to explore their modulatory effects on joint destruction, inflammatory response, and intracellular signaling. Arthritis was induced in female Wistar rats by complete Freund's adjuvant (CFA) 0.4 ml s.c. on days 1, 4, and 7. Treated groups received their respective drugs, starting from day 13, daily for 3 weeks. Methotrexate and prednisolone were the standard antirheumatic drugs, while cinnamaldehyde, tadalafil, and aliskiren were the test agents. Treatment with cinnamaldehyde, tadalafil, or aliskiren reduced serum levels of rheumatoid factor, and pro-inflammatory cytokines; tumor necrosis factor-alpha and interleukin-6 (IL-6), along with elevated level of IL-10 which is an anti-inflammatory cytokine. Besides, cartilage and bone destruction biomarkers; matrix metalloproteinase-3 (MMP-3) and receptor activator of nuclear factor-kappa B ligand (RANKL); were significantly reduced after treatment with the test agents, which was further confirmed by histopathological investigation. The elevated protein expressions of phosphorylated-Janus kinase 2 (p-JAK2), phosphorylated-signal transducer and activator of transcription 3 (p-STAT3), and inducible nitric oxide synthase (iNOS) in articular tissue were markedly attenuated after treatment with cinnamaldehyde, tadalafil, or aliskiren, while that of endothelial nitric oxide synthase (eNOS) was greatly enhanced. In addition, oxidative stress and inflammatory markers such as malondialdehyde, nitric oxide, and myeloperoxidase were reduced in joint tissue after treatment with the test agents, while glutathione content was elevated. Furthermore, the renin inhibitor aliskiren produced effects close to those of the normal and methotrexate, the gold standard antirheumatic drug, in most of the measured parameters. Collectively, these findings led to the assumption that the downregulation of IL-6/JAK2/STAT3 signaling by cinnamaldehyde, tadalafil, and aliskiren could alleviate joint destruction by MMP-3 and RANKL, reduce iNOS, and enhance eNOS expressions. Moreover, aliskiren could be a promising therapeutic agent for RA, because of its ability to normalize most of the measured parameters after CFA-induced arthritis.
Collapse
Key Words
- Aliskiren
- CFA, complete Freund's adjuvant
- CFA-induced arthritis
- DMARD, disease-modifying antirheumatic drug
- GSH, reduced glutathione
- H&E, hematoxylin and eosin
- IL-10, interleukin-10
- IL-6, interleukin-6
- IL-6/JAK2/STAT3 signaling
- JAK2, Janus kinase 2
- MDA, malondialdehyde
- MMP-3
- MMP-3, matrix metalloproteinase-3
- MPO, myeloperoxidase
- NO, nitric oxide
- PDE, phosphodiesterase
- RA, rheumatoid arthritis
- RANKL
- RANKL, receptor activator of nuclear factor-kappa B ligand
- RAS, renin angiotensin system
- STAT3, signal transducer and activator of transcription 3
- TNF-α, tumor necrosis factor-alpha
- eNOS, endothelial nitric oxide synthase
- iNOS, inducible nitric oxide synthase
Collapse
Affiliation(s)
- Amany A Azouz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Esraa Saleh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.,Operations Pharmacy, General Fayoum Hospital, Fayoum, Egypt
| | - Ali A Abo-Saif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| |
Collapse
|
25
|
Kuang Z, Bai J, Ni L, Hang K, Xu J, Ying L, Xue D, Pan Z. Withanolide B promotes osteogenic differentiation of human bone marrow mesenchymal stem cells via ERK1/2 and Wnt/β-catenin signaling pathways. Int Immunopharmacol 2020; 88:106960. [PMID: 32919219 DOI: 10.1016/j.intimp.2020.106960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND The treatment of bone defects has always been a problem for clinicians. In recent years, research on human bone mesenchymal stem cells (hBMSCs) has found that promoting their osteogenic differentiation could be a useful therapeutic strategy for bone healing. Previous studies have been reported that Withania somnifera Dunal inhibits osteoclastogenesis by inhibiting the NF-κB signaling pathway. Withanolide B is an active component of W. somnifera Dunal, but its role in osteogenic differentiation of hBMSCs remains unknown. Here, we performed a preliminary study on the role of Withanolide B in promoting osteogenic differentiation and its possible mechanism. METHODS We investigated the effect of Withanolide B on osteogenic differentiation of hBMSCs in vitro and in vivo. The effect of Withanolide B on the activity of hBMSCs was verified by CCK-8 assay and quantitative Real-time polymerase chain reaction (qPCR) and Western blotting analysis were used to verify the effect of Withanolide B on osteogenic differentiation-specific genes and proteins. The effect of Withanolide B on ALP activity and mineral deposition was verified by ALP and ARS staining. We then used a rat tibial osteotomy model to observe the effect of Withanolide B on bone healing. RESULTS Withanolide B is noncytotoxic to hBMSCs and can effectively promote their osteogenic differentiation. Moreover, we found that Withanolide B can regulate the osteogenic differentiation of hBMSCs through the ERK1/2 and Wnt/β-catenin signaling pathways. When inhibitors of the ERK1/2 and Wnt/β-catenin signaling pathways were used, the enhancement of osteogenic differentiation induced by Withanolide B was attenuated. Withanolide B also effectively promoted bone healing in the rat tibial osteotomy model. CONCLUSIONS Our results suggest that Withanolide B can promote the osteogenic differentiation of hBMSCs through the ERK1/2 and Wnt/β-catenin signaling pathways and can effectively promote bone defect healing.
Collapse
Affiliation(s)
- Zhihui Kuang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Jinwu Bai
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Licheng Ni
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Kai Hang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Jianxiang Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Li Ying
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China
| | - Deting Xue
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China.
| | - Zhijun Pan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China; Orthopedics Research Institute of Zhejiang University, No. 88, Jiefang Road, Hangzhou 310009, China.
| |
Collapse
|
26
|
Abstract
Rheumatoid arthritis is an autoimmune inflammatory disease primarily characterized by synovitis which is accompanied by extra-articular organ involvement, such as interstitial pneumonia, in addition to clinical symptoms including pain, swelling, stiffness of multiple joints, fever, and malaise. Joint destruction progresses soon after the onset, and once the affected joints are deformed, the development of irreversible physical dysfunction is noted. Thus, proper diagnosis and treatment are required from the early stages of the disease. Although palliative therapy with glucocorticoids and anti-inflammatory drugs had been used, disease-modifying antirheumatic drugs (DMARDs) are currently used to suppress immune abnormalities and to control disease activity. DMARDs are classified into different groups, such as conventional synthetic DMARD, targeted synthetic DMARD, and biologic DMARD. The appropriate use of these drugs has allowed remission to be the therapeutic goal in all patients. By maintaining remission, these drugs have also been shown to prevent the progression of joint destruction and physical dysfunction over a long period. The advent of molecular-targeted therapies has allowed for the use of treatments based on pathological mechanisms, and such therapeutic strategies have also been applied to the treatment of various autoimmune inflammatory diseases. In the future, safer and more effective treatments, therapeutic strategies aimed at drug holidays or cure, and the introduction of precision medicine are expected.
Collapse
|
27
|
Salvio G, Gianfelice C, Firmani F, Lunetti S, Balercia G, Giacchetti G. Bone Metabolism in SARS-CoV-2 Disease: Possible Osteoimmunology and Gender Implications. Clin Rev Bone Miner Metab 2020; 18:51-57. [PMID: 32904892 PMCID: PMC7459260 DOI: 10.1007/s12018-020-09274-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/18/2020] [Indexed: 01/08/2023]
Abstract
Even though inflammatory conditions are known to exert adverse effects on bone metabolism, there are no published data regarding SARS-CoV-2 infection and subsequent fracture risk. We present a brief review of the molecular mechanisms linking inflammatory diseases to increased fracture risk/osteoporosis and of the therapeutic strategies that can prevent bone resorption in patients with inflammatory disease, focusing on the RANK-RANKL system. We also make some considerations on gender differences in infection response and on their implications for survival and for the consequences of COVID-19. Several inflammatory cytokines, especially IL-1, IL-6, and TNF-α, stimulate osteoclast activity, favoring bone resorption through the RANK-RANKL system. Data from the previous SARS-CoV outbreak suggest that the present disease also has the potential to act directly on bone resorption units, although confirmation is clearly needed. Even though the available data are limited, the RANK-RANKL system may provide the best therapeutic target to prevent bone resorption after COVID-19 disease. Vitamin D supplementation in case of deficiency could definitely be beneficial for bone metabolism, as well as for the immune system. Supplementation of vitamin D in case of deficiency could be further advantageous. In COVID-19 patients, it would be useful to measure the bone metabolism markers and vitamin D. Targeting the RANK-RANKL system should be a priority, and denosumab could represent a safe and effective choice. In the near future, every effort should be made to investigate the fracture risk after SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Gianmaria Salvio
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Via Conca 71, Umberto I Hospital, 60126 Ancona, Italy
| | - Claudio Gianfelice
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Via Conca 71, Umberto I Hospital, 60126 Ancona, Italy
| | - Francesca Firmani
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Via Conca 71, Umberto I Hospital, 60126 Ancona, Italy
| | - Stefano Lunetti
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Via Conca 71, Umberto I Hospital, 60126 Ancona, Italy
| | - Giancarlo Balercia
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Via Conca 71, Umberto I Hospital, 60126 Ancona, Italy
| | - Gilberta Giacchetti
- Division of Endocrinology, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Via Conca 71, Umberto I Hospital, 60126 Ancona, Italy
| |
Collapse
|
28
|
Chan YH, Ngai MC, Chen Y, Wu MZ, Yu YJ, Zhen Z, Lai K, Cheung T, Ho LM, Chung HY, Lau CS, Tse HF, Yiu KH. Cumulative Rheumatic Inflammation Modulates the Bone-Vascular Axis and Risk of Coronary Calcification. J Am Heart Assoc 2020; 8:e011540. [PMID: 31130038 PMCID: PMC6585350 DOI: 10.1161/jaha.118.011540] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Background Rheumatic diseases are related to both abnormal bone turnover and atherogenesis, but a mechanistic link was missing. Methods and Results We investigated the effect of cumulative rheumatic inflammation (CRI) on risk of coronary calcification in a retrospective cohort of 145 rheumatoid arthritis patients. A time‐adjusted aggregate CRI score was derived by conglomerating all quarterly biomarker encounters of serum C‐reactive protein over 60 months immediately preceding computed tomography coronary angiography. Flow cytometry was performed to measure the osteocalcin‐positive (OCN+) CD34+KDR+ and OCN+CD34+ circulating endothelial progenitor cells (EPCs). Conventional early circulating EPCs CD34+CD133+KDR+ was determined. Coronary calcification was defined as any Agatston score >0. 50% of patients (n=72/145) had coronary calcification. CRI score was associated with presence of coronary calcification (P=0.004) (multivariable‐adjusted: highest versus lowest quartile: odds ratio=5.6 [95% CI 1.1–28.9], P=0.041). Receiver operating characteristics curve revealed divergent behavior of OCN‐expressing circulating EPCs (OCN+CD34+EPCs: area under the curve=0.60, P=0.034; OCN+CD34+KDR+EPCs: area under the curve=0.59, P=0.053, positive predictors) versus conventional early EPCs (CD34+CD133+KDR+: area under the curve=0.60, P=0.034, negative predictor) for coronary calcification, which persisted after multivariable adjustments (OCN+CD34+KDR+ [>75th percentile]: odds ratio=7.2 [95% CI 1.8–27.9], P=0.005; OCN+CD34+EPCs [>75th percentile]: odds ratio=6.0 [95% CI 1.5–23.3], P=0.010; CD34+CD133+KDR+ [>75th percentile: odds ratio=0.3 [95% CI 0.1–1.0], P=0.053). Intriguingly, the CRI score was associated with increased OCN+CD34+EPCs (highest versus lowest quartile: B=+25.6 [95% CI 0.8–50.5] [×103/mL peripheral blood], P=0.043), but reduced CD34+CD133+KDR+EPCs (highest versus lowest quartile: B=−16.2 [95% CI −31.5 to −0.9], P=0.038). Conclusions Preceding 60 months of CRI is associated with increased risk of coronary calcification and altered OCN expression in circulating EPCs.
Collapse
Affiliation(s)
- Yap-Hang Chan
- 1 Cardiology Division Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China
| | - Michael Cheong Ngai
- 1 Cardiology Division Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China
| | - Yan Chen
- 1 Cardiology Division Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China.,2 Cardiology Division Department of Medicine University of Hong Kong Shenzhen Hospital Shenzhen China
| | - Mei-Zhen Wu
- 1 Cardiology Division Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China
| | - Yu-Juan Yu
- 1 Cardiology Division Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China
| | - Zhe Zhen
- 1 Cardiology Division Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China
| | - Kevin Lai
- 1 Cardiology Division Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China
| | - Tommy Cheung
- 3 Division of Rheumatology Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China
| | - Lai-Ming Ho
- 4 School of Public Health The University of Hong Kong Hong Kong
| | - Ho-Yin Chung
- 3 Division of Rheumatology Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China
| | - Chak-Sing Lau
- 3 Division of Rheumatology Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China
| | - Hung-Fat Tse
- 1 Cardiology Division Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China.,2 Cardiology Division Department of Medicine University of Hong Kong Shenzhen Hospital Shenzhen China
| | - Kai-Hang Yiu
- 1 Cardiology Division Department of Medicine Queen Mary Hospital University of Hong Kong Hong Kong SAR China.,2 Cardiology Division Department of Medicine University of Hong Kong Shenzhen Hospital Shenzhen China
| |
Collapse
|
29
|
Qian H, Lei T, Ye Z, Hu Y, Lei P. From the Performance to the Essence: The Biological Mechanisms of How Tantalum Contributes to Osteogenesis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5162524. [PMID: 32802853 PMCID: PMC7403943 DOI: 10.1155/2020/5162524] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022]
Abstract
Despite the brilliant bioactive performance of tantalum as an orthopedic biomaterial verified through laboratory researches and clinical practice in the past decades, scarce evidences about the essential mechanisms of how tantalum contributes to osteogenesis were systematically discussed. Up to now, a few studies have uncovered preliminarily the biological mechanism of tantalum in osteogenic differentiation and osteogenesis; it is of great necessity to map out the panorama through which tantalum contributes to new bone formation. This minireview summarized current advances to demonstrate the probable signaling pathways and underlying molecular cascades through which tantalum orchestrates osteogenesis, which mainly contain Wnt/β-catenin signaling pathway, BMP signaling pathway, TGF-β signaling pathway, and integrin signaling pathway. Limits of subsistent studies and further work are also discussed, providing a novel vision for the study and application of tantalum.
Collapse
Affiliation(s)
- Hu Qian
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
- Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Changsha, 410008 Hunan, China
| | - Ting Lei
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Zhimin Ye
- Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Changsha, 410008 Hunan, China
| | - Yihe Hu
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Pengfei Lei
- Department of Orthopedics, Xiangya Hospital of Central South University, 87 Xiangya Road, Changsha, Hunan 410008, China
| |
Collapse
|
30
|
Osteoclasts and their circulating precursors in rheumatoid arthritis: Relationships with disease activity and bone erosions. Bone Rep 2020; 12:100282. [PMID: 32478145 PMCID: PMC7251539 DOI: 10.1016/j.bonr.2020.100282] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/01/2020] [Accepted: 05/11/2020] [Indexed: 12/29/2022] Open
Abstract
Patients with rheumatoid arthritis (RA) have very different outcomes, particularly with regard to bone erosions. Since osteoclasts are responsible for bone destruction adjacent to rheumatoid synovium, profiling osteoclasts from circulating precursors in RA could help identify patients at risk for bone destruction. In this study, we sought to determine whether the functional characteristics of osteoclasts generated from their blood precursors were modified by RA activity or were intrinsic to osteoclasts and associated with the RA phenotype (erosive or not). Osteoclasts were generated in vitro from peripheral blood mononuclear cells (PBMCs) of subjects with RA (n = 140), as well as sex- and age-matched healthy controls (n = 101). Osteoclastic parameters were analyzed at baseline and during the follow-up for up to 4 years, with regular assessment of RA activity, bone erosions, and bone mineral density (BMD). As a validation cohort, we examined RA patients from the Early Undifferentiated PolyArthritis (EUPA) study (n = 163). The proportion of CD14+ PBMC was higher in RA than in control subjects, but inversely correlated with the 28-joint disease activity score (DAS28). Also surprisingly, in osteoclast cultures from PBMCs, active RA was associated with lower osteoclastogenic capacity, while in vitro bone resorption per osteoclast and resistance to apoptosis were similar in both active and quiescent RA. In a small subgroup analysis, osteoclasts from subjects with recent RA that had progressed at four years to an erosive RA exhibited at baseline greater resistance to apoptosis than those from patients remaining non-erosive. Our findings establish that when RA is active, circulating monocytes have a reduced potential to generate osteoclasts from PBMCs in vitro. In addition, osteoclasts associated with erosive disease had resistance to apoptosis from the start of RA. Osteoclasts are derived in vitro from circulating monocytes in rheumatoid arthritis. Blood CD14+ monocytes (%) are higher but inversely correlated with disease activity. Active rheumatoid arthritis is associated with reduced osteoclast formation in vitro. Inflammation alters the ability to generate osteoclasts from circulating monocytes. Osteoclast resistance to apoptosis is stable and associated with long-term erosions.
Collapse
|
31
|
Gao J, Zhang G, Xu K, Ma D, Ren L, Fan J, Hou J, Han J, Zhang L. Bone marrow mesenchymal stem cells improve bone erosion in collagen-induced arthritis by inhibiting osteoclasia-related factors and differentiating into chondrocytes. Stem Cell Res Ther 2020; 11:171. [PMID: 32381074 PMCID: PMC7203805 DOI: 10.1186/s13287-020-01684-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is characterized by joint inflammation and damage to the cartilage and bone in collagen-induced arthritis (CIA). Mesenchymal stem cells (MSCs) can improve articular symptoms and reduce bone erosion in CIA rats; however, the underlying mechanism remains unknown. This study aimed to investigate the mechanism underlying MSC-induced improvement of bone destruction in CIA. Methods Wistar rats were divided into a normal group, CIA control group, MTX intervention group, and BMSC intervention group, each comprising 8 rats. Serum RANKL, OPG, and CXCL10 levels of all groups were determined via flow cytometry after 42 days of interventions. RANKL, OPG, TRAF6, CXCL10, and CXCR3 were detected on the synovial membrane via immunohistochemistry, and their relative mRNA levels were determined via RT-PCR analysis. BMSCs were labeled with GFP and administered to CIA rats via the tail vein. At different time points, the distribution of implanted GFP-MSCs in synovial tissues was observed using a fluorescence microscope, and the potential of GFP-MSCs to differentiate into chondrocytes was assessed via immunofluorescence analysis. Results BMSC transplantation improved joint inflammation and inhibited bone destruction in CIA rats. BMSCs inhibited the expression of serum CXCL10 and CXCL10 and CXCR3 expression at the synovial membrane. Moreover, protein and mRNA expression analyses revealed that BMSCs potentially regulated RANKL/OPG expression levels in the serum and synovial tissue. Upon implantation into CIA rats, GFP-MSCs were traced in the joints. GFP-positive cells were observed in the cartilage tissue from day 11 and until 42 days after transplantation. Anti-type II collagen/GFP double-positive cells were observed in the articular cartilage (especially damaged cartilage) upon immunofluorescence staining of anti-type II collagen. Conclusions BMSCs improve bone destruction in CIA by inhibiting the CXCL10/CXCR3 chemotactic axis, regulating the RANKL/OPG ratio, and directly differentiating into chondrocytes.
Collapse
Affiliation(s)
- Jinfang Gao
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China.,Department of Rheumatology, Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Gailian Zhang
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Ke Xu
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Dan Ma
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China.,Department of Rheumatology, Bethune Hospital Affiliated to Shanxi Medical University, Taiyuan, 030032, Shanxi, China
| | - Limin Ren
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Jingjing Fan
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Jianwen Hou
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Jian Han
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China
| | - Liyun Zhang
- Department of Rheumatology, Shanxi Bethune Hospital, Taiyuan, 030032, Shanxi, China.
| |
Collapse
|
32
|
Zhang X, Zhang X, Wang X, Wang T, Bai B, Zhang N, Zhao Y, Yu Y, Wang B. Efficient Delivery of Triptolide Plus a miR-30-5p Inhibitor Through the Use of Near Infrared Laser Responsive or CADY Modified MSNs for Efficacy in Rheumatoid Arthritis Therapeutics. Front Bioeng Biotechnol 2020; 8:170. [PMID: 32258008 PMCID: PMC7092621 DOI: 10.3389/fbioe.2020.00170] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 02/19/2020] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disease for which treatment focuses on suppressing an overactive immune system and maintaining the physiological balance of synovial fibroblasts (SFs). We found that miR-30-5p was highly expressed in rheumatoid arthritis synovial fibroblasts (RASFs). Subsequently, we predicted that phosphatidylinositol 3-kinase regulatory subunit 2 (PIK3R2) might be a putative target of miR-30-5p. Recent studies have reported that PIK3R2 can maintain the physiological homeostasis of RASFs. Therefore, miR-30-5p inhibitor has the potential to be used in the treatment of RA, but low levels of miR-30-5p inhibitor internalization affect its application. Triptolide (TP) is an effective drug in the treatment of RA but induces severe toxicity and has a narrow therapeutic window. In this study, the cell internalization performance of miR-30-5p inhibitor was improved by loading it into cell membrane penetrating peptide (CADY)-modified mesoporous silica nanoparticles (MSNs), and the toxicity of TP was decreased by loading it into a controlled drug release system based on MSNs. The nanodrug carrier was constructed by filling a phase-change material (PCM) of 1-tetradecanol and drugs into MSNs that could be triggered by an NIR laser with thermo-chemo combination RA therapy. Our results show that the miR-30-5p inhibitor-loaded MSNs@CADY significantly inhibited RASF proliferation and increased apoptosis. In addition, MSNs@PCM@TP under 808 nm laser irradiation were effective in downregulating immune system activation in an RA rat model. Finally, the results of a pharmacodynamics study showed that the combination of MSNs@CADY@miR-30-5p inhibitor and MSNs@PCM@TP under 808 nm laser significantly increased the effectiveness of RA treatment. These findings provide a novel understanding of RA pathogenesis and a theoretical basis for RA treatment.
Collapse
Affiliation(s)
- Xiaonan Zhang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Xin Zhang
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xipeng Wang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Tao Wang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Bin Bai
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Na Zhang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yanjiao Zhao
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yang Yu
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Bing Wang
- Institute of Biochemistry and Molecular Biology, College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
33
|
Molecular and Cellular Pathways Contributing to Joint Damage in Rheumatoid Arthritis. Mediators Inflamm 2020; 2020:3830212. [PMID: 32256192 PMCID: PMC7103059 DOI: 10.1155/2020/3830212] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 02/24/2020] [Indexed: 01/19/2023] Open
Abstract
Rheumatoid arthritis is a chronic autoimmune syndrome associated with several genetic, epigenetic, and environmental factors affecting the articular joints contributing to cartilage and bone damage. Although etiology of this disease is not clear, several immune pathways, involving immune (T cells, B cells, dendritic cells, macrophages, and neutrophils) and nonimmune (fibroblasts and chondrocytes) cells, participate in the secretion of many proinflammatory cytokines, chemokines, proteases (MMPs, ADAMTS), and other matrix lysing enzymes that could disturb the immune balance leading to cartilage and bone damage. The presence of autoantibodies preceding the clinical onset of arthritis and the induction of bone erosion early in the disease course clearly suggest that initiation events damaging the cartilage and bone start very early during the autoimmune phase of the arthritis development. During this process, several signaling molecules (RANKL-RANK, NF-κB, MAPK, NFATc1, and Src kinase) are activated in the osteoclasts, cells responsible for bone resorption. Hence, comprehensive knowledge on pathogenesis is a prerequisite for prevention and development of targeted clinical treatment for RA patients that can restore the immune balance improving clinical therapy.
Collapse
|
34
|
Liu D, Fang Y, Rao Y, Tan W, Zhou W, Wu X, Zhang C, Zhang Y, Liu Y, Sunagawa M, Hisamitsu T, Li G. Synovial fibroblast-derived exosomal microRNA-106b suppresses chondrocyte proliferation and migration in rheumatoid arthritis via down-regulation of PDK4. J Mol Med (Berl) 2020; 98:409-423. [PMID: 32152704 DOI: 10.1007/s00109-020-01882-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 01/02/2020] [Accepted: 01/21/2020] [Indexed: 12/12/2022]
Abstract
Fibroblast-derived exosomes have been reported to transfer microRNAs to recipient cells, where they regulate target gene expression, which is of interest for understanding the basic biology of inflammation, tissue homeostasis, and development of therapeutic approaches. Initial microarray-based analysis carried out in this study identified the rheumatoid arthritis (RA)-related differentially expressed gene pyruvate dehydrogenase kinase 4 (PDK4). Subsequently, the upstream regulatory microRNA-106b (miR-106b) of PDK4 was predicted with bioinformatic analyses. A collagen-induced arthritis (CIA)-induced mouse model was established, and exosomes were isolated from synovial fibroblasts (SFs) and transferred into chondrocytes to identify the role of exosomes in rheumatoid arthritis (RA). We found that PDK4 was poorly expressed in RA cartilage tissues and chondrocytes, while miR-106b was highly expressed in RA SFs and SF-derived exosomes. Notably, PDK4 was confirmed as a target gene of miR-106b. Over-expression of PDK4 promoted the proliferation and migration abilities of chondrocytes and inhibited their apoptosis as well as affected the receptor activator of nuclear factor kappa B ligand (RANKL)/RANK/osteoprotegerin (OPG) system. Meanwhile, miR-106b was delivered from SFs to chondrocytes through exosomes, which suppressed chondrocyte proliferation and migration and accelerated apoptosis as well as affected the RANKL/RANK/OPG system via down-regulation of PDK4. Furthermore, in vivo results validated that miR-106b inhibition could relieve CIA-induced RA. Taken together, SF-derived exosomal miR-106b stimulates RA initiation by targeting PDK4, indicating a physiologically validated potential approach for the prevention and treatment of RA. KEY MESSAGES: PDK4 is decreased in chondrocytes of RA, while miR-106b is increased in SFBs. PDK4 promotes proliferation and migration of chondrocytes. miR-106b could target 3'UTR of PDK4 gene. SFB-exosomal miR-106b inhibits proliferation and migration of chondrocytes. Inhibition of miR-106b attenuates RA progression in a CIA mouse model.
Collapse
Affiliation(s)
- Dan Liu
- Department of Pathology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Yuxuan Fang
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368, Hanjiang Middle Road, Yangzhou, 225000, Jiangsu Province, People's Republic of China
| | - Yujun Rao
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368, Hanjiang Middle Road, Yangzhou, 225000, Jiangsu Province, People's Republic of China
| | - Wei Tan
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368, Hanjiang Middle Road, Yangzhou, 225000, Jiangsu Province, People's Republic of China
| | - Wei Zhou
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368, Hanjiang Middle Road, Yangzhou, 225000, Jiangsu Province, People's Republic of China
| | - Xia Wu
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368, Hanjiang Middle Road, Yangzhou, 225000, Jiangsu Province, People's Republic of China
- Clinical Medical College, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Chunwang Zhang
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368, Hanjiang Middle Road, Yangzhou, 225000, Jiangsu Province, People's Republic of China
- Clinical Medical College, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Yu Zhang
- Medical College of Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Yanqing Liu
- Medical College of Yangzhou University, Yangzhou, 225000, People's Republic of China
| | - Masataka Sunagawa
- Department of Physiology, School of Medicine, Showa University, Tokyo, 142-8555, Japan
| | - Tadashi Hisamitsu
- Department of Physiology, School of Medicine, Showa University, Tokyo, 142-8555, Japan
| | - Guoqing Li
- Department of Rheumatology and Immunology, Affiliated Hospital of Yangzhou University, Yangzhou University, No. 368, Hanjiang Middle Road, Yangzhou, 225000, Jiangsu Province, People's Republic of China.
| |
Collapse
|
35
|
Gaber T, Brinkman ACK, Pienczikowski J, Diesing K, Damerau A, Pfeiffenberger M, Lang A, Ohrndorf S, Burmester GR, Buttgereit F, Hoff P. Impact of Janus Kinase Inhibition with Tofacitinib on Fundamental Processes of Bone Healing. Int J Mol Sci 2020; 21:ijms21030865. [PMID: 32013232 PMCID: PMC7037633 DOI: 10.3390/ijms21030865] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/20/2020] [Accepted: 01/27/2020] [Indexed: 01/08/2023] Open
Abstract
Both inflammatory diseases like rheumatoid arthritis (RA) and anti-inflammatory treatment of RA with glucocorticoids (GCs) or non-steroidal anti-inflammatory drugs (NSAIDs) negatively influence bone metabolism and fracture healing. Janus kinase (JAK) inhibition with tofacitinib has been demonstrated to act as a potent anti-inflammatory therapeutic agent in the treatment of RA, but its impact on the fundamental processes of bone regeneration is currently controversially discussed and at least in part elusive. Therefore, in this study, we aimed to examine the effects of tofacitinib on processes of bone healing focusing on recruitment of human mesenchymal stromal cells (hMSCs) into the inflammatory microenvironment of the fracture gap, chondrogenesis, osteogenesis and osteoclastogenesis. We performed our analyses under conditions of reduced oxygen availability in order to mimic the in vivo situation of the fracture gap most optimal. We demonstrate that tofacitinib dose-dependently promotes the recruitment of hMSCs under hypoxia but inhibits recruitment of hMSCs under normoxia. With regard to the chondrogenic differentiation of hMSCs, we demonstrate that tofacitinib does not inhibit survival at therapeutically relevant doses of 10–100 nM. Moreover, tofacitinib dose-dependently enhances osteogenic differentiation of hMSCs and reduces osteoclast differentiation and activity. We conclude from our data that tofacitinib may influence bone healing by promotion of hMSC recruitment into the hypoxic microenvironment of the fracture gap but does not interfere with the cartilaginous phase of the soft callus phase of fracture healing process. We assume that tofacitinib may promote bone formation and reduce bone resorption, which could in part explain the positive impact of tofacitinib on bone erosions in RA. Thus, we hypothesize that it will be unnecessary to stop this medication in case of fracture and suggest that positive effects on osteoporosis are likely.
Collapse
Affiliation(s)
- Timo Gaber
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Antonia Clara Katharina Brinkman
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Justyna Pienczikowski
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Karoline Diesing
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Alexandra Damerau
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Moritz Pfeiffenberger
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Annemarie Lang
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Sarah Ohrndorf
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
| | - Gerd-Rüdiger Burmester
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Frank Buttgereit
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
| | - Paula Hoff
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, 10117 Berlin, Germany
- Endokrinologikum Berlin, 10117 Berlin, Germany
| |
Collapse
|
36
|
Tokunaga T, Mokuda S, Kohno H, Yukawa K, Kuranobu T, Oi K, Yoshida Y, Hirata S, Sugiyama E. TGFβ1 Regulates Human RANKL-Induced Osteoclastogenesis via Suppression of NFATc1 Expression. Int J Mol Sci 2020; 21:ijms21030800. [PMID: 31991837 PMCID: PMC7038124 DOI: 10.3390/ijms21030800] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/31/2022] Open
Abstract
Osteoclasts are multinucleated giant cells responsible for bone resorption. Various mediators involved in osteoclast differentiation have been investigated as possible therapeutic targets for osteoporosis and rheumatoid arthritis (RA). Although transforming growth factor beta1 (TGFβ1) has been described as one such multifunctional cytokine essential for bone remodeling, its effect on osteoclastogenesis remains controversial. Therefore, we sought to examine the effect of TGFβ1 on osteoclast generation induced by receptor activator of nuclear factor (NF)-κB ligand (RANKL) in humans. Peripheral blood monocytes, isolated using magnetic bead sorting, were cultured with macrophage-colony stimulating factor (M-CSF) or RANKL with or without TGFβ1. Tartrate-resistant acid phosphatase (TRAP) staining, as well as bone resorption assays, revealed that TGFβ1 suppressed RANKL-mediated human osteoclast development. Real-time reverse transcription PCR and Western blotting revealed that TGFβ1 reduced the gene and protein expression of nuclear factor of activated T cells, cytoplasmic 1 (NFATc1), the master regulator of osteoclast differentiation, respectively. Luciferase assays indicated that TGFβ1 inhibited the NF-κB p65-stimulated promoter activity of NFATc1. Immunofluorescence analysis demonstrated that TGFβ1 abrogated RANKL-induced nuclear translocation of p65. Thus, TGFβ1 regulates human RANKL-induced osteoclastogenesis via downregulation of NFATc1 by blocking nuclear translocation of NF-κB, suggesting that TGFβ1 may be a potential therapeutic target for RA.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Eiji Sugiyama
- Correspondence: ; Tel.: +81 82 257 5539; Fax: +81 82 257 1584
| |
Collapse
|
37
|
Takeuchi T, Soen S, Ishiguro N, Yamanaka H, Tanaka S, Kobayashi M, Okubo N, Nitta T, Tanaka Y. Predictors of new bone erosion in rheumatoid arthritis patients receiving conventional synthetic disease-modifying antirheumatic drugs: Analysis of data from the DRIVE and DESIRABLE studies. Mod Rheumatol 2020; 31:34-41. [PMID: 31826682 DOI: 10.1080/14397595.2019.1703484] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate new bone erosion and cartilage destruction predictors in rheumatoid arthritis (RA) patients treated with conventional synthetic disease-modifying antirheumatic drugs (csDMARDs). METHODS Placebo-treated patient data from two 12-month, randomized, double-blind, phase 2 (DRIVE) and 3 (DESIRABLE) trials that evaluated denosumab efficacy in csDMARD-treated RA patients were used. Change from baseline in erosion score (ES) of ≥1.0 at 12 months was considered new bone erosion; predictors were identified using a multivariate model. RESULTS Among 306 patients, mean ± standard deviation disease activity score 28-C-reactive protein (CRP) at baseline was 3.58 ± 1.03. New bone erosion was observed in 90 patients (29.4%). Univariate analysis identified female sex, anti-cyclic citrullinated peptide (CCP) antibody positivity, rheumatoid factor (RF) positivity, tender joint count ≥6, CRP ≥0.3 mg/dL, erythrocyte sedimentation rate (ESR) ≥28 mm/h, and baseline ES ≥3 as significant predictors for new bone erosion. In multivariate analysis, predictors were anti-CCP antibody positivity, CRP ≥0.3 mg/dL, and baseline ES ≥3; RF and ESR were excluded as they strongly correlated with anti-CCP antibody and CRP, respectively. CONCLUSION In RA patients treated with csDMARDs, new bone erosion predictors were seropositivity, elevated inflammatory markers, and baseline ES ≥3. TRIAL REGISTRATION NUMBER DRIVE, JapicCTI-101263; DESIRABLE, NCT01973569.
Collapse
Affiliation(s)
- Tsutomu Takeuchi
- Department of Internal Medicine, Division of Rheumatology, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Soen
- Department of Orthopaedic Surgery and Rheumatology, Kindai University Nara Hospital, Nara, Japan
| | - Naoki Ishiguro
- Department of Orthopaedics/Rheumatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | - Sakae Tanaka
- Orthopaedic Surgery and Spinal Surgery, The University of Tokyo Hospital, Tokyo, Japan
| | - Makiko Kobayashi
- Medical Science Department, Medical Affairs Division, Daiichi Sankyo Co. Ltd, Tokyo, Japan
| | - Naoki Okubo
- Biostatistics and Data Management Department, R&D Division, Daiichi Sankyo Co. Ltd, Tokyo, Japan
| | - Takaya Nitta
- Clinical Development Department, R&D Division, Daiichi Sankyo Co. Ltd, Tokyo, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, Graduate School of Medical Science, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
38
|
Xie J, Li S, Xiao L, Ouyang G, Zheng L, Gu Y, Gao C, Han X. Zoledronic acid ameliorates the effects of secondary osteoporosis in rheumatoid arthritis patients. J Orthop Surg Res 2019; 14:421. [PMID: 31823822 PMCID: PMC6902494 DOI: 10.1186/s13018-019-1492-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/27/2019] [Indexed: 01/16/2023] Open
Abstract
Background Secondary osteoporosis may occur in patients with rheumatoid arthritis (RA), causing irreversible joint damage and disability. Bisphosphonates, the recently developed bone resorption inhibitors, have demonstrated significant therapeutic effects on senile and postmenopausal osteoporosis. This study evaluated the efficacy and safety of zoledronic acid (ZOL), with or without methotrexate (MTX), for the prevention and treatment of bone destruction in RA patients. Methods We recruited 66 RA patients with symptoms of secondary osteoporosis. They were randomized into three treatment groups—combined treatment with MTX and ZOL, ZOL monotherapy, or MTX monotherapy—in two consecutive 6-month periods. The participants were followed for 12 months. At the end of each treatment period, improvement in disease activity, bone destruction, and fracture risk were evaluated. Results Combined treatment with ZOL and MTX had significantly better clinical efficacy compared with either ZOL or MTX monotherapy (P < 0.05). The combination significantly improved the lumbar spine and hip BMD and reduced FRAX scores, suggesting that ZOL combined with MTX reduces bone loss and risk of hip fracture in RA patients with secondary osteoporosis. Conclusion ZOL has a synergistic effect when combined with MTX, inhibiting RA disease activity, reducing fracture risk, and improving quality of life in RA patients with secondary osteoporosis. Trial registration Chinese Clinical Trial Registry, ChiCTR1800019290. Registered 3 November 2018–Retrospective registered, http://www.chictr.org.cn/showproj.aspx?proj = 31758
Collapse
Affiliation(s)
- Jun Xie
- Department of Orthopedics, The Tenth People's Hospital Affiliated to Nanjing Medical University, No. 301 Yanchangzhong Road, Shanghai, 200000, China.,Department of Joint Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 504 Xinhua Road, Shanghai, 200052, China
| | - Shaohua Li
- Department of Orthopedics, The Tenth People's Hospital Affiliated to Nanjing Medical University, No. 301 Yanchangzhong Road, Shanghai, 200000, China.
| | - Lianbo Xiao
- Department of Joint Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 504 Xinhua Road, Shanghai, 200052, China.
| | - Guilin Ouyang
- Department of Joint Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 504 Xinhua Road, Shanghai, 200052, China
| | - Lin Zheng
- Department of Joint Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 504 Xinhua Road, Shanghai, 200052, China
| | - Yubiao Gu
- Department of Joint Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 504 Xinhua Road, Shanghai, 200052, China
| | - Chengxin Gao
- Department of Joint Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 504 Xinhua Road, Shanghai, 200052, China
| | - Xiuwei Han
- Department of Joint Orthopedics, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No. 504 Xinhua Road, Shanghai, 200052, China
| |
Collapse
|
39
|
Li F, Li X, Liu G, Gao C, Li X. Bone Marrow Mesenchymal Stem Cells Decrease the Expression of RANKL in Collagen-Induced Arthritis Rats via Reducing the Levels of IL-22. J Immunol Res 2019; 2019:8459281. [PMID: 31828174 PMCID: PMC6885301 DOI: 10.1155/2019/8459281] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 09/05/2019] [Accepted: 09/19/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE To investigate the transplantation effect of bone marrow mesenchymal stem cells (MSCs) on the expression of interlukin-22 (IL-22) and RANKL in collagen-induced arthritis (CIA) rats. METHODS 32 CIA models were established. 16 CIA rats were transplanted with MSCs, and others were used as nontreatment CIA controls. The concentrations of IL-22 and RANKL in serum were detected by ELISA and those in synovial tissue of rats' joints by immunohistochemical staining. In addition, the expression of RANKL mRNA was measured by RT-PCR in the fibroblast-like synoviocytes (FLSs), cultured with IL-22 in vitro, which were delivered from the joints of CIA rats treated with or without MSCs. RESULTS The transplantation of MSCs into CIA rats relieved the destruction of joints, measured by AI score, X-ray, and histopathology. MSCs also reduced the expression of IL-22 and RANKL in serum by ELISA (P < 0.001) and similarly in FLSs by immunohistochemical staining. In vitro, IL-22 induced significantly the expression of RANKL mRNA in cultured FLSs in a dose-dependent manner, whereas this induction was significantly reduced in FLSs derived from CIA rats transplanted with MSCs (normal controls: F = 79.33, P < 0.001; CIA controls: F = 712.72, P < 0.001; and CIA-MSC rats: F = 139.04, P < 0.001). CONCLUSION Our results suggest that the transplantation of MSCs can reduce the expression of RANKL in vivo by downregulating the levels of IL-22, thereby ameliorating the degree of RA bone destruction. This study provides a theoretical basis for a potential therapy of RA with MSCs, and IL-22 and RANKL may become two new targets to treat RA.
Collapse
Affiliation(s)
- Fang Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Xin Li
- Endocrine Metabolism and Immune Center, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China
| | - Guiyan Liu
- Department of Nephrology, Changzhi People's Hospital, Changzhi 046000, China
| | - Chong Gao
- Department of Pathology, Joint Program in Transfusion Medicine, Brigham and Women's Hospital/Children's Hospital Boston, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
40
|
Tong JJ, Xu SQ, Zong HX, Pan MJ, Teng YZ, Xu JH. Prevalence and risk factors associated with vertebral osteoporotic fractures in patients with rheumatoid arthritis. Clin Rheumatol 2019; 39:357-364. [PMID: 31691041 DOI: 10.1007/s10067-019-04787-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/16/2019] [Accepted: 09/21/2019] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To explore the prevalence and risk factors of osteoporosis (OP) and vertebral osteoporotic fracture (VOPF) in patients with rheumatoid arthritis (RA). METHODS Anteroposterior and lateral X-ray examination of the vertebral column (T4-L4) was used for the semi-quantitative assessment of VOPF. Bone mineral density was measured by dual-energy X-ray absorptiometry. RESULTS Of 865 RA patients, the prevalence of OP and VOPF was 33.6% and 20.2%, respectively. Patients with OP or VOPF were older, and had longer term use and a larger daily amount and cumulative dose of glucocorticoids (GCs), longer disease duration, and higher Health Assessment Questionnaire (HAQ) scores and Sharp scores than patients without OP or VOPF (P < 0.05). OP was also correlated with higher disease activity. The patients treated with GCs had higher incidences of OP and VOPF than the patients without GCs (P < 0.05). The cutoff values in the area under curve (AUC) of the daily dose or treatment course of GCs-VOPF were 9 mg and 37.5 days. Older age, female sex, and a higher Sharp score were risk factors for OP in RA patients, while higher BMI was a protective factor. Older age and a high GC daily dose were risk factors for VOPF in RA patients. CONCLUSIONS RA patients have a high prevalence of OP and VOPF. Older age, female sex, lower BMI, and higher activity and severity of RA are closely related with OP. Older age and a higher GC daily dose are risk factors for VOPF in RA patients. Key Points • Older age, female sex, lower BMI, and a higher Sharp score were risk factors for OP in RA patients. • Older age and a high GC daily dose were risk factors for VOPF in RA patients. • OP and VOPF in RA patients were correlated with longer disease duration and higher severity of RA.
Collapse
Affiliation(s)
- Jing-Jing Tong
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Sheng-Qian Xu
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| | - He-Xiang Zong
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Mei-Juan Pan
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Yu-Zhu Teng
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Jian-Hua Xu
- Department of Rheumatology & Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| |
Collapse
|
41
|
Song G, Feng T, Zhao R, Lu Q, Diao Y, Guo Q, Wang Z, Zhang Y, Ge L, Pan J, Wang L, Han J. CD109 regulates the inflammatory response and is required for the pathogenesis of rheumatoid arthritis. Ann Rheum Dis 2019; 78:1632-1641. [PMID: 31455659 PMCID: PMC6900259 DOI: 10.1136/annrheumdis-2019-215473] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 12/20/2022]
Abstract
Objective The aim of this study was to investigate the role of CD109 in rheumatoid arthritis (RA) fibroblast-like synoviocytes (FLSs) and to evaluate its potential as a therapeutic target. Methods CD109 expression was examined in synovial tissues and FLSs from RA patients and collagen-induced arthritis (CIA) model mice. CD109-deficient mice were developed to evaluate the severity of CIA. Small interfering RNAs and a neutralising antibody against CD109 (anti-CD109) were designed for functional or treatment studies in RA FLSs and CIA. Results CD109 was found to be abundantly expressed in the synovial tissues from RA patients and CIA mice. CD109 expression in RA FLSs was upregulated by inflammatory stimuli, such as interleukin-1β and tumour necrosis factor-α. Silencing of CD109 or anti-CD109 treatment reduced proinflammatory factor production, cell migration, invasion, chemoattractive potential and osteoclast differentiation, thereby reducing the deleterious inflammatory response of RA FLSs in vitro. Mice lacking CD109 were protected against arthritis in the CIA model. Anti-CD109 treatment prevented the onset and ameliorated the severity of CIA lesions. Conclusion Our study uncovers an antiarthritic role for CD109 and suggests that CD109 inhibition might serve as a promising novel therapeutic strategy for RA.
Collapse
Affiliation(s)
- Guanhua Song
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Tingting Feng
- Department of Pathology, Shandong University Medical School, Jinan, China
| | - Ru Zhao
- Department of Pathology, Shandong University Medical School, Jinan, China
| | - Qiqi Lu
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, China
| | - Yutao Diao
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Qingwei Guo
- Department of Hematology, Qilu Children's Hospital of Shandong University, Jinan, China
| | - Zhaoxia Wang
- Institute of Basic Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuang Zhang
- School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, China
| | - Luna Ge
- Shandong Medicinal Biotechnology Centre, Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Lab for Biotechnology Drugs of Ministry of Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jihong Pan
- Shandong Medicinal Biotechnology Centre, Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Lab for Biotechnology Drugs of Ministry of Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Lin Wang
- Shandong Medicinal Biotechnology Centre, Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Lab for Biotechnology Drugs of Ministry of Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jinxiang Han
- Shandong Medicinal Biotechnology Centre, Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Key Lab for Biotechnology Drugs of Ministry of Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
42
|
Olímpio RMC, Moretto FCF, De Sibio MT, de Oliveira M, Mathias LS, Gonçalves BM, Deprá IC, Tilli HP, Rodrigues BM, Saraiva PP, Maria DA, Nogueira CR. The importance of estrogen for bone protection in experimental hyperthyroidism in human osteoblasts. Life Sci 2019; 231:116556. [PMID: 31194990 DOI: 10.1016/j.lfs.2019.116556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/31/2019] [Accepted: 06/09/2019] [Indexed: 11/16/2022]
Abstract
Triiodothyronine (T3) and estrogen (E2) play important roles in the bone remodeling process and signaling of receptor activator of the nuclear factor-kappa β (RANKL) and osteoprotegerin (OPG) expressed by osteoblasts. However, little is known of the molecular action of these hormones in conditions of hyperthyroidism and associated E2 in human cells. AIMS This study evaluated the effects of the physiological concentration of E2 (10 nM), alone or in association with physiological (1 nM) and supraphysiological (10 nM) concentrations of T3, on RANKL and OPG gene expression in human osteoblasts. MAIN METHODS Alkaline phosphatase and osteocalcin assays were performed to verify the presence of mature osteoblasts. After mimicking the experimental hyperthyroidism in osteoblasts untreated or treated with E2, RANKL and OPG gene expression was analyzed by real-time PCR and protein expression by western Blot and ELISA. Alizarin Red staining analyzed the amount of bone matrix after hormonal treatments. KEY FINDINGS E2 enhanced the gene expression of OPG when associated with 1 nM and 10 nM T3. E2 was able to restore the bone matrix after an initial decrease using 1 nM and 10 nM T3. The protective effect of E2 on the RANKL and OPG signaling pathway was demonstrated. E2 restored the bone matrix induced by experimental hyperthyroidism. SIGNIFICANCE The data highlight the importance of E2 to maintain OPG expression and osteoblast activity against possible loss of bone mass, especially in conditions where T3 is in excess.
Collapse
Affiliation(s)
- Regiane Marques Castro Olímpio
- Department of Internal Medicine, Botucatu Medical School, Univ Estadual Paulista - UNESP, Botucatu, SP 18618-970, Brazil.
| | | | - Maria Teresa De Sibio
- Department of Internal Medicine, Botucatu Medical School, Univ Estadual Paulista - UNESP, Botucatu, SP 18618-970, Brazil
| | - Miriane de Oliveira
- Department of Internal Medicine, Botucatu Medical School, Univ Estadual Paulista - UNESP, Botucatu, SP 18618-970, Brazil
| | - Lucas Solla Mathias
- Department of Internal Medicine, Botucatu Medical School, Univ Estadual Paulista - UNESP, Botucatu, SP 18618-970, Brazil
| | - Bianca Mariani Gonçalves
- Department of Internal Medicine, Botucatu Medical School, Univ Estadual Paulista - UNESP, Botucatu, SP 18618-970, Brazil
| | - Igor Carvalho Deprá
- Department of Internal Medicine, Botucatu Medical School, Univ Estadual Paulista - UNESP, Botucatu, SP 18618-970, Brazil
| | - Helena Paim Tilli
- Department of Internal Medicine, Botucatu Medical School, Univ Estadual Paulista - UNESP, Botucatu, SP 18618-970, Brazil
| | - Bruna Moretto Rodrigues
- Department of Internal Medicine, Botucatu Medical School, Univ Estadual Paulista - UNESP, Botucatu, SP 18618-970, Brazil
| | - Patrícia Pinto Saraiva
- Department of Internal Medicine, Botucatu Medical School, Univ Estadual Paulista - UNESP, Botucatu, SP 18618-970, Brazil
| | - Durvanei Augusto Maria
- Laboratory Biochemistry and Biophysics, Butantan Institute, 1500, Avenue Vital Brazil, São Paulo, Brazil
| | - Célia Regina Nogueira
- Department of Internal Medicine, Botucatu Medical School, Univ Estadual Paulista - UNESP, Botucatu, SP 18618-970, Brazil
| |
Collapse
|
43
|
Gossec L, Soubrier M, Foissac F, Molto A, Richette P, Beauvais C, Ruyssen-Witrand A, Perdriger A, Chary-Valckenaere I, Mouterde G, Dernis E, Euller-Ziegler L, Flipo RM, Gilson M, Guis S, Mariette X, Pouplin S, Marhadour T, Schaeverbeke T, Sordet C, Fayet F, Dougados M. Screening for and management of comorbidities after a nurse-led program: results of a 3-year longitudinal study in 769 established rheumatoid arthritis patients. RMD Open 2019; 5:e000914. [PMID: 31275607 PMCID: PMC6579559 DOI: 10.1136/rmdopen-2019-000914] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/03/2019] [Accepted: 04/23/2019] [Indexed: 12/20/2022] Open
Abstract
Background/purpose Cardiovascular (CV) risk, cancer, infections and osteoporosis should be screened for in rheumatoid arthritis (RA). The objective was to assess 3-year effects of a nurse visit for comorbidity counselling. Methods This was an open long-term (3 years) extension of the Comorbidities and Education in Rheumatoid Arthritis 6-month randomised controlled trial in which patients with definite, stable RA were visiting a nurse for comorbidity counselling. Comorbidity status was assessed and nurses provided advice on screening and management, at baseline and 3 years later. A score was developed to quantify comorbidity screening and management: 0–100, where lower scores indicate better screening and management. The score was compared between baseline and 3-year assessment using a Wilcoxon test for paired data. Results Of the 970 recruited patients, 776 (80%) were followed-up at 2–4 years and 769 (79%) had available data for comorbidities at both time points: mean (±SD) age 58 (±11) years and mean disease duration 14 (±10) years; 614 (80%) were women, the mean Disease Activity Score 28 was 3.0±1.3, and 538 (70%) were receiving a biologic. At baseline, the mean comorbidity screening score was 36.6 (±19.9) and it improved at 3 years to 24.3 (±17.8) (p<0.0001), thus with a relative improvement of 33% (improvement of 12 points). CV risk screening, vaccination status and bone densitometry performance improved the most. Conclusions Comorbidity screening was suboptimal but improved notably over 3 years, after a nurse-led programme aiming at checking systematically for comorbidity screening and giving patient advice. This long-term efficacy pleads in favour of nurse-led interventions to better address comorbidities in RA. Trial registration number NCT01315652
Collapse
Affiliation(s)
- Laure Gossec
- Sorbonne Universite, Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, Paris, France.,Rheumatology, Pitie Salpetriere Hospital, AP-HP, Paris, France
| | - Martin Soubrier
- Rheumatology, Gabriel Montpied University Hospital, Clermont-Ferrand, France
| | - Frantz Foissac
- Clinical Research Unit Necker-Cochin, Assistance Publique-Hopitaux de Paris, Paris, Île-de-France, France
| | - Anna Molto
- Rheumatology, Cochin Hospital, AP-HP, Paris, France.,INSERM (U1153), Clinical Epidemiology and Biostatistics, PRES Sorbonne Paris-Cité, Paris Descartes University, Paris, France
| | - Pascal Richette
- Rheumatology, Lariboisiere Hospital AP-HP, Paris, France.,Inserm UMR1132 Bioscar, Universite Paris Diderot, Paris, France
| | - Catherine Beauvais
- Rheumatology, Sorbonne Université and Hopital Saint-Antoine AP-HP, Paris, France
| | - Adeline Ruyssen-Witrand
- Rheumatology, UMR 1027 Inserm, Paul Sabatier University and Purpan Hospital, Toulouse, France
| | - Aleth Perdriger
- Rheumatology, South Hospital, Rennes 1 University, Rennes, France
| | | | - Gael Mouterde
- Immuno-rhumatologie, Lapeyronie Hospital and Montpellier University, Montpellier, France
| | | | | | - René-Marc Flipo
- Rheumatology, R Salengro Hospital, University of Lille, Lille, France
| | - Mélanie Gilson
- Rheumatology, GREPI-CNRS, Grenoble Hospital and Université Joseph Fourier, Echirolles, France
| | - Sandrine Guis
- Rheumatology, University Hospital, AP-HM, Marseille, France
| | - Xavier Mariette
- Rheumatology, HôpitauxUniversitaires Paris-Sud, Université Paris-Sud, INSERM UMR1184, Le Kremlin Bicetre, France
| | | | - Thierry Marhadour
- Rheumatology, Cavale Blanche Hospital and INSERM 1227, Université Bretagne Occidentale, Brest, France
| | - Thierry Schaeverbeke
- Rheumatology, Pellegrin Hospital, Bordeaux University, CNRS 5164, Bordeaux, France
| | - Christelle Sordet
- Rheumatology, Hautepierre Hospital, Fédération de médecine translationnelle, UMR INSERM 1109, Strasbourg, France
| | - Françoise Fayet
- Rheumatology, Gabriel Montpied University Hospital, Clermont-Ferrand, France
| | - Maxime Dougados
- Rheumatology, Cochin Hospital, AP-HP, Paris, France.,INSERM (U1153), Clinical Epidemiology and Biostatistics, PRES Sorbonne Paris-Cité, Paris Descartes University, Paris, France
| |
Collapse
|
44
|
Margheri F, Laurenzana A, Giani T, Maggi L, Cosmi L, Annunziato F, Cimaz R, Del Rosso M. The protease systems and their pathogenic role in juvenile idiopathic arthritis. Autoimmun Rev 2019; 18:761-766. [PMID: 31181328 DOI: 10.1016/j.autrev.2019.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 02/11/2019] [Indexed: 12/20/2022]
Abstract
Numerous proteases produced by synovial cells of arthritic joints, chondrocytes, macrophages and polymorphonuclear cells have been identified as responsible for the joint damage in rheumatoid arthritis. There are few scientific contributions aimed to identify similar mechanisms in the joints of patients with juvenile idiopathic arthritis. Recently, some mechanisms emerged, triggered by the TH17 and TH1/TH17 lymphocytes, which could shed new light on unexpected pathogenic pathways of joint damage in the JIA, mainly regarding the RANK-RANKL pathway. Other novelties are linked to the mechanisms of acidification of the synovial fluid, which create a microenvironment suitable for the extracellular activity of lysosomal enzymes. Some biological drugs currently used in the therapy of JIA can interfere with these mechanisms.
Collapse
Affiliation(s)
- Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Anna Laurenzana
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Teresa Giani
- Department of Pediatrics, Rheumatology Unit, Anna Meyer Children's Hospital, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | | | - Rolando Cimaz
- Department of Clinical Sciences and Community Health, University of Milano, Italy.
| | - Mario Del Rosso
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| |
Collapse
|
45
|
Chan YH, Ngai MC, Chen Y, Wu MZ, Yu YJ, Zhen Z, Lai K, Cheung T, Ho LM, Chung HY, Lau CS, Lau CP, Tse HF, Yiu KH. Osteogenic circulating endothelial progenitor cells are linked to electrocardiographic conduction abnormalities in rheumatic patients. Ann Noninvasive Electrocardiol 2019; 24:e12651. [PMID: 31017736 DOI: 10.1111/anec.12651] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 03/02/2019] [Accepted: 03/06/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Osteogenic circulating endothelial progenitor cells (EPC) play a pathogenic role in cardiovascular system degeneration through promulgating vasculature calcification, but its role in conduction disorders as part of the cardiovascular degenerative continuum remained unknown. AIM To investigate the role of osteocalcin (OCN)-expressing circulating EPCs in cardiac conduction disorders in the unique clinical sample of rheumatoid arthritis (RA) susceptible to both abnormal bone metabolism and cardiac conduction disorders. METHODS We performed flow cytometry studies in 134 consecutive asymptomatic patients with rheumatoid arthritis to derive osteogenic circulating OCN-positive (OCN+) CD34+KDR+ vs. CD34+CD133+KDR+ conventional EPC. Study endpoint was the prespecified combined endpoint of electrocardiographic conduction abnormalities. RESULTS Total prevalence of cardiac conduction abnormality was 9% (n = 12). All patients except one had normal sinus rhythm. One patient had atrial fibrillation. No patient had advanced atrioventricular (AV) block. Prevalence of first-degree heart block (>200 ms), widened QRS duration (>120 ms) and right bundle branch block were 6.7%, 2.1%, and 2.2% respectively. Circulating osteogenic OCN+ CD34+ KDR+ EPCs were significantly higher among patients with cardiac conduction abnormalities (p = 0.039). Elevated OCN+ CD34+ KDR+ EPCs> 75th percentile was associated with higher prevalence of cardiac conduction abnormalities (58.3% vs. 20.02%, p = 0.003). Adjusted for potential confounders, elevated OCN+ CD34+ KDR+ EPCs> 75th percentile remained independently associated with increased risk of cardiac conduction abnormalities (OR = 4.4 [95%CI 1.2-16.4], p = 0.028). No significant relation was found between conventional EPCs CD34+CD133+KDR+ and conduction abnormalities (p = 0.36). CONCLUSIONS Elevated osteogenic OCN+ CD34+ KDR+ EPCs are independently associated with the presence of electrocardiographic conduction abnormalities in patients with rheumatoid arthritis, unveiling a potential novel pathophysiological mechanism.
Collapse
Affiliation(s)
- Yap-Hang Chan
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| | - Michael Cheong Ngai
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| | - Yan Chen
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China.,Cardiology Division, Department of Medicine, University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Mei-Zhen Wu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| | - Yu-Juan Yu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| | - Zhe Zhen
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| | - Kevin Lai
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| | - Tommy Cheung
- Division of Rheumatology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| | - Lai-Ming Ho
- School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Ho-Yin Chung
- Division of Rheumatology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| | - Chak-Sing Lau
- Division of Rheumatology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| | - Chu-Pak Lau
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China.,Cardiology Division, Department of Medicine, University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Kai-Hang Yiu
- Cardiology Division, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China.,Cardiology Division, Department of Medicine, University of Hong Kong Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
46
|
Sobacchi C, Menale C, Villa A. The RANKL-RANK Axis: A Bone to Thymus Round Trip. Front Immunol 2019; 10:629. [PMID: 30984193 PMCID: PMC6450200 DOI: 10.3389/fimmu.2019.00629] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/08/2019] [Indexed: 12/13/2022] Open
Abstract
The identification of Receptor activator of nuclear factor kappa B ligand (RANKL) and its cognate receptor Receptor activator of nuclear factor kappa B (RANK) during a search for novel tumor necrosis factor receptor (TNFR) superfamily members has dramatically changed the scenario of bone biology by providing the functional and biochemical proof that RANKL signaling via RANK is the master factor for osteoclastogenesis. In parallel, two independent studies reported the identification of mouse RANKL on activated T cells and of a ligand for osteoprotegerin on a murine bone marrow-derived stromal cell line. After these seminal findings, accumulating data indicated RANKL and RANK not only as essential players for the development and activation of osteoclasts, but also for the correct differentiation of medullary thymic epithelial cells (mTECs) that act as mediators of the central tolerance process by which self-reactive T cells are eliminated while regulatory T cells are generated. In light of the RANKL-RANK multi-task function, an antibody targeting this pathway, denosumab, is now commonly used in the therapy of bone loss diseases including chronic inflammatory bone disorders and osteolytic bone metastases; furthermore, preclinical data support the therapeutic application of denosumab in the framework of a broader spectrum of tumors. Here, we discuss advances in cellular and molecular mechanisms elicited by RANKL-RANK pathway in the bone and thymus, and the extent to which its inhibition or augmentation can be translated in the clinical arena.
Collapse
Affiliation(s)
- Cristina Sobacchi
- Milan Unit, Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy.,Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | - Ciro Menale
- Milan Unit, Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy.,Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | - Anna Villa
- Milan Unit, Institute for Genetic and Biomedical Research (CNR-IRGB), Milan, Italy.,San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
47
|
Jafarzadeh A, Nemati M, Khorramdelazad H, Hassan ZM. Immunomodulatory properties of cimetidine: Its therapeutic potentials for treatment of immune-related diseases. Int Immunopharmacol 2019; 70:156-166. [PMID: 30802678 DOI: 10.1016/j.intimp.2019.02.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/06/2019] [Accepted: 02/13/2019] [Indexed: 12/27/2022]
Abstract
Histamine exerts potent modulatory impacts on the cells of innate- [including neutrophils, monocytes, macrophages, dendritic cells (DCs), natural killer (NK) cells and NKT cells] and adaptive immunity (such as Th1-, Th2-, Th17-, regulatory T-, CD8+ cytotoxic T cells, and B cells) through binding to histamine receptor 2 (H2R). Cimetidine, as an H2R antagonist, reverses the histamine-mediated immunosuppression, as it has powerful stimulatory effects on the effector functions of neutrophils, monocytes, macrophages, DCs, NK cells, NKT cells, Th1-, Th2-, Th17-, and CD8+ cytotoxic T cells. However, cimetidine reduces the regulatory/suppressor T cell-mediated immunosuppression. Experimentally, cimetidine potentiate some immunologic activities in vitro and in vivo. The therapeutic potentials of cimetidine as an immunomodulatory agent were also investigated in a number of human diseases (such as cancers, viral warts, allergic disorders, burn, and bone resorption) and vaccination. This review aimed to provide a concise summary regarding the impacts of cimetidine on the immune system and highlight the cellular mechanisms of action and the immunomodulatory effects of this drug in various diseases to give novel insights regarding the therapeutic potentials of this drug for treatment of immune-related disorders. The review encourages more investigations to consider the immunomodulatory characteristic of cimetidine for managing of immune-related disorders.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Maryam Nemati
- Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossain Khorramdelazad
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | | |
Collapse
|
48
|
Wang T, Yang L, Jiang J, Liu Y, Fan Z, Zhong C, He C. Pulsed electromagnetic fields: promising treatment for osteoporosis. Osteoporos Int 2019; 30:267-276. [PMID: 30603841 DOI: 10.1007/s00198-018-04822-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 12/18/2018] [Indexed: 02/05/2023]
Abstract
Osteoporosis (OP) is considered to be a well-defined disease which results in high morbidity and mortality. In patients diagnosed with OP, low bone mass and fragile bone strength have been demonstrated to significantly increase risk of fragility fractures. To date, various anabolic and antiresorptive therapies have been applied to maintain healthy bone mass and strength. Pulsed electromagnetic fields (PEMFs) are employed to treat patients suffering from delayed fracture healing and nonunions. Although PEMFs stimulate osteoblastogenesis, suppress osteoclastogenesis, and influence the activity of bone marrow mesenchymal stem cells (BMSCs) and osteocytes, ultimately leading to retention of bone mass and strength. However, whether PEMFs could be taken into clinical use to treat OP is still unknown. Furthermore, the deeper signaling pathways underlying the way in which PEMFs influence OP remain unclear.
Collapse
Affiliation(s)
- T Wang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - L Yang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - J Jiang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Y Liu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Z Fan
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - C Zhong
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - C He
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu, 610041, Sichuan, People's Republic of China.
- Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
49
|
Ma Z, Yu R, Zhao J, Sun L, Jian L, Li C, Liu X. Constant hypoxia inhibits osteoclast differentiation and bone resorption by regulating phosphorylation of JNK and IκBα. Inflamm Res 2019; 68:157-166. [PMID: 30604211 DOI: 10.1007/s00011-018-1209-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/19/2018] [Accepted: 12/17/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Osteoclasts are responsible for the bone loss in rheumatoid arthritis (RA). Hypoxia has been suggested to play key roles in pathological bone loss. However, the current understanding of the effects of hypoxia on osteoclastogenesis is controversial. Effects of hypoxia on both the formation and function of osteoclasts requires examination. In the current study, we aimed to explore the effect of hypoxia on osteoclast differentiation and the underlying mechanisms. METHODS RAW264.7 cells and murine bone-marrow-derived monocytes were used to induce osteoclastogenesis in the presence of macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa B ligand (RANKL). Hypoxic conditions were maintained in a hypoxic chamber at 5% CO2 and 1% O2, balanced with N2. Osteoclasts were detected by tartrate-resistant acid phosphatase (TRAP) staining. A bone resorption assay was carried out in vitro using bone slices. RT-PCR was conducted to detect osteoclast markers and transcription factors. The phosphorylation of nuclear factor-κBα (IκBα), c-Jun N-terminal kinase (JNK), extracellular regulated protein kinase (ERK), and p38 was detected by western blotting. Mann-Whitney U test or Student's t test was used to compare differences between the two groups. RESULTS TRAP staining and the bone resorption assay revealed that hypoxia-restrained osteoclast differentiation and bone resorption. Expression of osteoclast markers including cathepsin K, RANK, and TRAP decreased during osteoclast differentiation under hypoxic conditions (all P < 0.05). Hypoxia at 1% O2 did not affect cell viability, whereas it dramatically abated RANKL-dependent phosphorylation of the JNK-mitogen-activated protein kinases (MAPK) and IκBα pathways. Moreover, the expression of nuclear factor of activated T-cell cytoplasmic 1 (NFATc1) was inhibited under hypoxic conditions (all P < 0.05). CONCLUSIONS These results suggest that constant hypoxia at 1% O2 significantly restrains osteoclast formation and resorbing function without affecting cell viability. Constant hypoxia might inhibit RANKL-induced osteoclastogenesis by regulating NFATc1 expression via interfering the phosphorylation of JNK and IκBα.
Collapse
Affiliation(s)
- Zhenzhen Ma
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China
| | - Ruohan Yu
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China
| | - Jinxia Zhao
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China
| | - Lin Sun
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China
| | - Leilei Jian
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China
| | - Changhong Li
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China.
| | - Xiangyuan Liu
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
50
|
Abstract
The immune system and bone metabolism influence each other. An imbalance in the immune system, resulting in inflammatory stimuli may induce an imbalance in bone turnover via induction of osteoclast differentiation and inhibition of osteoblast differentiation, leading to various pathological conditions including osteoporosis. T-cell subsets, helper T (Th)1 and Th17, which activate the immune system, induce osteoclasts, whereas regulatory T (Treg) cells, responsible for immunosuppression, inhibit osteoclastic differentiation. In addition, inflammatory cytokines, such as the tumor necrosis factor (TNF), also cause an imbalance in bone turnover, induction of osteoclasts and inhibition of osteoblasts. Treatments targeting the immune system may regulate abnormalities in bone metabolism, while also controlling immune abnormalities. In rheumatoid arthritis (RA), a representative autoimmune disease, immune abnormality and accompanying prolongation of synovial inflammation cause bone and cartilage destruction, periarticular osteoporosis, and systemic osteoporosis. Joint damage and osteoporosis in RA occur through totally different mechanisms. Stimulation by inflammatory cytokines induces the expression of the receptor activator for nuclear factor-κB ligand (RANKL) in T cells and synovial cells, thereby inducing bone destruction due to osteoblast-independent osteoclast maturation. However, biological products targeting TNF or interleukin-6 not only control disease activity, but also inhibit joint destruction. However, these biological products are not effective for osteoporosis. Conversely, anti-RANKL antibody inhibits osteoporosis and bone destruction, but exerts no influence on RA disease activity. Such differences in therapeutic efficacy may indicate the necessity for rethinking current theories on the mechanism of bone metabolism abnormality and joint destruction. Understanding the mechanisms underlying these pathologies via commonalities existing between the immune system and the metabolic system may lead to the development of new treatments.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Kitakyushu, 807-8555, Japan.
| |
Collapse
|