1
|
Pamies A, Vallvé JC, Paredes S. New Cardiovascular Risk Biomarkers in Rheumatoid Arthritis: Implications and Clinical Utility-A Narrative Review. Biomedicines 2025; 13:870. [PMID: 40299461 PMCID: PMC12025197 DOI: 10.3390/biomedicines13040870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that not only causes joint inflammation but also significantly increases the risk of cardiovascular disease (CVD), leading to a higher morbidity and mortality. RA patients face an accelerated progression of atherosclerosis, attributed to both traditional cardiovascular risk factors and systemic inflammation. This review focuses on emerging biomarkers for cardiovascular risk assessment in RA, aiming to enhance early detection and treatment strategies. Specifically, we examine the roles of interleukin-32 (IL-32), Dickkopf-1 (DKK-1), galectin-3 (Gal-3), catestatin (CST), and fetuin-A (Fet-A) as potential markers for CVD in this patient population. IL-32, a proinflammatory cytokine, is elevated in RA patients and plays a significant role in inflammation and endothelial dysfunction, both of which contribute to atherosclerosis. DKK-1, a Wnt signaling pathway inhibitor, has been associated with both synovial inflammation and the development of atherosclerotic plaques. Elevated DKK-1 levels have been linked to an increased CV mortality and could serve as a marker for CVD progression in RA. Gal-3 is involved in immune modulation and fibrosis, with elevated levels in RA patients correlating with disease activity and cardiovascular outcomes. Catestatin, a peptide derived from chromogranin A, has protective anti-inflammatory and antioxidative properties, though its role in RA-related CVD remains under investigation. Finally, Fet-A, a glycoprotein involved in vascular calcification, shows potential as a biomarker for CV events in RA, though data on its role remain conflicting. These biomarkers provide deeper insights into the pathophysiology of RA and its cardiovascular comorbidities. Although some biomarkers show promise in improving CV risk stratification, further large-scale studies are required to validate their clinical utility. Currently, these biomarkers are in the research phase and are not yet implemented in standard care. Identifying and incorporating these biomarkers into routine clinical practice could lead to the better management of cardiovascular risk in RA patients, thus improving outcomes in this high-risk population. This review highlights the importance of continued research to establish reliable biomarkers that can aid in both diagnosis and the development of targeted therapies for cardiovascular complications in RA.
Collapse
Affiliation(s)
- Anna Pamies
- Secció de Reumatologia, Hospital de Tortosa Verge de la Cinta, 43500 Tortosa, Catalonia, Spain;
| | - Joan-Carles Vallvé
- Unitat de Recerca en Lípids i Arterioesclerosi, Universitat Rovira i Virgili, 43204 Reus, Catalonia, Spain;
- Institut Investigació Sanitaria Pere Virgili, 43204 Reus, Catalonia, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Silvia Paredes
- Unitat de Recerca en Lípids i Arterioesclerosi, Universitat Rovira i Virgili, 43204 Reus, Catalonia, Spain;
- Institut Investigació Sanitaria Pere Virgili, 43204 Reus, Catalonia, Spain
- Secció de Reumatologia, Hospital Universitari Sant Joan de Reus, 43204 Reus, Catalonia, Spain
| |
Collapse
|
2
|
Neto M, Mendes B, Albuquerque F, da Silva JAP. Novel biomarkers in RA: Implication for diagnosis, prognosis, and personalised treatment. Best Pract Res Clin Rheumatol 2025; 39:102021. [PMID: 39550250 DOI: 10.1016/j.berh.2024.102021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/18/2024]
Abstract
Over the past decades our understanding of rheumatoid arthritis (RA) pathogenesis has improved remarkably and major breakthroughs in the treatment of RA were made with the advent of numerous targeted therapies and new treatment strategies. Despite these advances, several unmet needs remain, namely in achieving earlier and more accurate diagnosis, monitoring disease activity, predicting disease prognosis and optimizing treatment. To address these gaps, recent research has focused on identifying biomarkers that may enhance diagnostic precision, predict disease prognosis, and optimize treatment strategies. In this narrative review we will describe recent developments in RA biomarkers with demonstrated or promising clinical relevance.
Collapse
Affiliation(s)
- Marcelo Neto
- Rheumatology Department, Unidade Local de Saúde de Coimbra, Portugal.
| | - Beatriz Mendes
- Rheumatology Department, Unidade Local de Saúde de Coimbra, Portugal.
| | | | - José António P da Silva
- Rheumatology Department, Unidade Local de Saúde de Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal.
| |
Collapse
|
3
|
Riitano G, Spinelli F, Manganelli V, Caissutti D, Capozzi A, Garufi C, Garofalo T, Misasi R, Sorice M, Conti F, Longo A, Alessandri C. Wnt signaling as a translational target in rheumatoid and psoriatic arthritis. J Transl Med 2025; 23:158. [PMID: 39905450 PMCID: PMC11796213 DOI: 10.1186/s12967-025-06174-2] [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: 11/12/2024] [Accepted: 01/25/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) and Psoriatic arthritis (PsA) are chronic inflammatory diseases mainly affecting joints. RA primarily targets the synovial joints and is characterized by cartilage and bone erosion, whereas PsA is associated with skin and nail psoriasis and is characterized by erosive bone damage with an exuberant bone formation and soft tissue involvement. Recent evidence described the involvement of the Wnt pathway in the pathogenesis of these diseases. Thus, we aimed to analyze some components of Wnt signaling, i.e. DKK1, Wnt 5a and β-catenin, and their association with disease activity indices, investigating possible differences between the two diseases. METHODS Sera from 18 RA patients naïve for biological therapy, 18 PsA patients and 20 matched healthy donors (HD) were tested for DKK1 by ELISA, Wnt 5a and β-catenin by Immunoblotting. Values were correlated with CTX-1, detected by ELISA, and with disease activity indices: Disease Activity Score on 28 joints (DAS28-CRP) for RA and the Disease Activity in Psoriatic Arthritis (DAPSA) score for PsA. RESULTS This study highlights significant increase in DKK1, Wnt 5a, and β-catenin levels in RA and PsA patients compared to HD, with distinct patterns of correlation with disease activity indices. Indeed, in RA patients, DKK1 levels positively correlated with DAS28-CRP score, whereas in PsA patients, DKK1 levels negatively correlated with DAPSA score. Our findings showed a strong correlation between DKK1 and CTX-1 levels in RA patients, supporting the relationship between DKK1 levels and the presence of joint erosions. Furthermore, a significant positive correlation was found between β-catenin and IL-6 levels in RA, indicating that β-catenin may be involved in the inflammatory cascade. CONCLUSION This study compares the involvement of Wnt signaling in RA and PsA, suggesting that Wnt signaling may represent a possible mechanism of disease activity. In particular, it indicates that DKK1 levels are correlated with CTX-1, a marker of bone resorption, and with disease activity in RA patients. These findings underscore the importance of these biomarkers in the potential monitoring of patients, offering insights into disease mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Gloria Riitano
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, Rome, 00161, Italy
| | - Francesca Spinelli
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Valeria Manganelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, Rome, 00161, Italy
| | - Daniela Caissutti
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, Rome, 00161, Italy
| | - Antonella Capozzi
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, Rome, 00161, Italy
| | - Cristina Garufi
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Tina Garofalo
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, Rome, 00161, Italy
| | - Roberta Misasi
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, Rome, 00161, Italy.
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, Rome, 00161, Italy
| | - Fabrizio Conti
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Agostina Longo
- Department of Experimental Medicine, "Sapienza" University of Rome, Viale Regina Elena 324, Rome, 00161, Italy
| | - Cristiano Alessandri
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
4
|
Iwamoto N, Sato S, Furukawa K, Michitsuji T, Shiraishi K, Watanabe K, Chiba K, Osaki M, Kawakami A. Association of denosumab with serum cytokines, chemokines, and bone-related factors in patients with rheumatoid arthritis: A post hoc analysis of a multicentre, open-label, randomised, parallel-group study. Mod Rheumatol 2024; 34:936-946. [PMID: 38226481 DOI: 10.1093/mr/roae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/12/2023] [Accepted: 12/26/2023] [Indexed: 01/17/2024]
Abstract
OBJECTIVES To clarify changes in serum cytokines, chemokines, and bone-related factors during denosumab treatment in rheumatoid arthritis (RA) patients. METHODS This was a post hoc analysis of a multicentre, open-label, randomised, parallel-group study. Patients were randomly assigned to continue treatment with conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs) plus receive treatment with denosumab (csDMARDs plus denosumab group) or to continue treatment with csDMARD therapy alone for 12 months. Serum biomarker levels were measured at baseline and at 6 and 12 months. RESULTS Baseline and 6-month data from the csDMARDs plus denosumab (n = 22) and csDMARD therapy alone (n = 22) groups were analysed. Statistically significant changes from baseline were seen: Dickkopf-related protein 1 decreased at 6 and 12 months (both groups); osteopontin decreased at 6 months in the csDMARDs plus denosumab group; osteopontin and soluble CD40 ligand increased at 6 and 12 months in the csDMARD therapy alone group; osteocalcin decreased at 6 and 12 months, epidermal growth factor decreased at 12 months, and macrophage-derived chemokine decreased at 6 months in the csDMARDs plus denosumab group; and interferon gamma-induced protein-10 increased at 12 months in the csDMARD therapy alone group. CONCLUSIONS Denosumab may inhibit bone destruction by suppressing bone-related factors/chemokines.
Collapse
Affiliation(s)
- Naoki Iwamoto
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shuntaro Sato
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Kaori Furukawa
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Toru Michitsuji
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazuteru Shiraishi
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kounosuke Watanabe
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Ko Chiba
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Makoto Osaki
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
5
|
Garnero P, Gineyts E, Rousseau JC, Richette P, Sellam J, Chapurlat R. A new serum biochemical marker of synovium turnover predicts radiographic progression in patients with early arthritis. Rheumatology (Oxford) 2024; 63:874-881. [PMID: 37471609 DOI: 10.1093/rheumatology/kead375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/28/2023] [Accepted: 07/04/2023] [Indexed: 07/22/2023] Open
Abstract
OBJECTIVE To investigate whether serum Col 3-4, a new biochemical marker of synovial tissue turnover, was associated with progression of joint damage in patients with early arthritis. METHODS A total of 788 early arthritis patients (<6 months of symptoms, 82% diagnosis of RA, 18% undifferentiated arthritis) from the prospective ESPOIR study were investigated. Progression was defined as an increase of 1 or 5 unit(s) in radiographic van der Heijde modified Sharp score between baseline and 1 or 5 years, respectively. Associations between baseline Col 3-4 and progression were assessed by logistic regression. RESULTS Each standard deviation increase of baseline Col 3-4 levels was associated with an increased 5-yr total damage progression with an odds ratio (OR, 95% CI) of 1.51 (1.21, 1.88), which remained significant when DAS28, C-reactive protein and anti-citrullinated protein antibodies positivity were included in the model [OR (95% CI): 1.34 (1.01, 1.76)]. Further adjustment for bone erosion did not modify the association. Patients with both Col 3-4 in the highest quintile and bone erosion had a >2-fold higher risk of progression [OR (95% CI): 7.16 (2.31, 22)] than patients with either high Col 3-4 [2.91 (1.79, 4.73)] or bone erosion [2.36 (2.38, 3.70)] alone. Similar associations were observed for prediction of 12 months progression. CONCLUSIONS Increased serum Col 3-4 is associated with a higher risk of structural progression, independently of major risk factors. Col 3-4 may be useful in association with bone erosion to identify patients with early arthritis at higher risk.
Collapse
Affiliation(s)
- Patrick Garnero
- INSERM, UMR1033, Hôpital Edouard Herriot, Pavillon F, Lyon, France
| | - Evelyne Gineyts
- INSERM, UMR1033, Hôpital Edouard Herriot, Pavillon F, Lyon, France
| | | | - Pascal Richette
- Université Paris 7, UFR Médicale, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Lariboisière, Fédération de Rhumatologie, Paris, France
| | - Jérémie Sellam
- INSERM, UMRS 938, service de rhumatologie, Sorbonne Université, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Roland Chapurlat
- INSERM, UMR1033, Hôpital Edouard Herriot, Pavillon F, Lyon, France
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| |
Collapse
|
6
|
Zhao D, Wu L, Hong M, Zheng S, Wu X, Ye H, Chen F, Zhang D, Liu X, Meng X, Chen X, Chen S, Zhu J, Li J. DKK-1 and Its Influences on Bone Destruction: A Comparative Study in Collagen-Induced Arthritis Mice and Rheumatoid Arthritis Patients. Inflammation 2024; 47:129-144. [PMID: 37688661 DOI: 10.1007/s10753-023-01898-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/07/2023] [Accepted: 08/26/2023] [Indexed: 09/11/2023]
Abstract
Dickkopf-1 (DKK-1) has been considered a master regulator of bone remodeling. As precursors of osteoclasts (OCs), myeloid-derived suppressor cells (MDSCs) were previously shown to participate in the process of bone destruction in rheumatoid arthritis (RA). However, the role of DKK-1 and MDSCs in RA is not yet fully understood. We investigated the relevance between the level of DKK-1 and the expression of MDSCs in different tissues and joint destruction in RA patients and collagen-induced arthritis (CIA) mouse models. Furthermore, the CIA mice were administered recombinant DKK-1 protein. The arthritis scores, bone destruction, and the percentage of MDSCs in the peripheral blood and spleen were monitored. In vitro, the differentiation of MDSCs into OCs was intervened with recombinant protein and inhibitor of DKK-1. The number of OCs differentiated and the protein expression of the Wnt/β-catenin signaling pathway were explored. The level of DKK-1 positively correlates with the frequency of MDSCs and bone erosion in RA patients and CIA mice. Strikingly, recombinant DKK-1 intervention significantly exacerbated arthritis scores and bone destruction, increasing the percentage of MDSCs in the peripheral blood and spleen in CIA mice. In vitro experiments showed that recombinant DKK-1 promoted the differentiation of MDSCs into OCs, reducing the expression of β-catenin and TCF4 and increasing the expression of CyclinD1. In contrast, the DKK-1 inhibitor had the opposite effect. Our findings highlight that DKK-1 promoted MDSCs expansion in RA and enhanced the differentiation of MDSCs into OCs via targeting the Wnt/β-catenin pathway, aggravating the bone destruction in RA.
Collapse
Affiliation(s)
- Di Zhao
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lisheng Wu
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Mukeng Hong
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Songyuan Zheng
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xianghui Wu
- Laboratory Animal Research Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haixin Ye
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Feilong Chen
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Dingding Zhang
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xinhang Liu
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiangyun Meng
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoyun Chen
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Shixian Chen
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Junqing Zhu
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Juan Li
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China.
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Wang Q, Liu Y, Wu J, Chen S, Hu T, Liu Y, Li X, Li X, Wu Y, Yu J, Zeng T, Luo Y, Hu X, Tan LM. Potential significance of changes in serum levels of IL-17, TNF-α and DKK-1 in the progression of the rheumatoid arthritis. Autoimmunity 2023; 56:2276068. [PMID: 37909152 DOI: 10.1080/08916934.2023.2276068] [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: 03/26/2023] [Accepted: 10/21/2023] [Indexed: 11/02/2023]
Abstract
To detect the value of serum interleukin-17 (IL-17), tumour necrosis factor-α (TNF-α), and Dickkopf-1 (DKK-1) in rheumatoid arthritis (RA) at different disease stages. 141 RA patients were randomly obtained and diagnosed in a large tertiary first-class hospital in Jiangxi Province from November 2021 to January 2022. RA was divided into 38 low activity and remission phase (low remission patients), 72 moderate activity patients, 41 high activity patients, according to the disease activity score 28 (DAS28) of RA and 70 healthy controls. IL-17 and TNF-α in serum detected by flow cytometry; DKK-1by ELISA; rheumatoid factor (RF) and C-reactive protein (CRP) by rate scattering turbidimetry; erythrocyte sedimentation rate (ESR) by Widmanstat method; anti-cyclic citrullinated polypeptide antibody (Anti-CCP) by chemiluminescence. The changes among the groups were statistically analysed and evaluated their diagnostic value. ①Anti-CCP, CRP, and ESR levels in the moderate-to-high activity group were higher than controls, while IL-17, TNF-α, and DKK-1levels higher than low remission group, moderate activity group and controls (p < 0.05). ②IL-17, TNF-α and DKK-1 were positively correlated with RA disease activity, with the correlations of IL-17, TNF-α and DKK-1 all over 0.5 (p < 0.05). ③The ROC curve showed that among all indices the AUC of DKK-1 was the largest, 0. 922, and has the highest sensitivity and negative predictive value for RA, 0.965 and 0.953, respectively. The specificity and positive predictive value of TNF-α is highest, 0.918 and 0.921, respectively, combined them had the highest predictive value in moderate-to-high activity RA, with AUC of 0.968, and had the highest sensitivity of 0.965. The IL-17, TNF-α and DKK-1 levels were elevated in RA and positively correlated with disease activity, involved in the Wnt signalling pathway of inflammatory and joint destructive effects, combining them to monitor the RA disease process and biologically treat the cytokines in the pathogenesis of RA were valuable.
Collapse
Affiliation(s)
- Qunxia Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Yanzhao Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Jiazhen Wu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Simei Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Tingting Hu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Yuhan Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Xu Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Xiaohang Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Yang Wu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Jianlin Yu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Tingting Zeng
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Yi Luo
- The Second Affiliated Hospital of Jiangxi, University of Chinese Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Xiaoyan Hu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| | - Li-Ming Tan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Jiangxi Province's Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, People's Republic of China
| |
Collapse
|
8
|
Biedroń G, Czepiel M, Siedlar M, Korkosz M. Serum concentration of dickkopf-related protein 1 (DKK1) in psoriatic arthritis in the context of bone remodelling. Rheumatol Int 2023; 43:2175-2183. [PMID: 37750896 PMCID: PMC10587027 DOI: 10.1007/s00296-023-05452-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 08/30/2023] [Indexed: 09/27/2023]
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease, characterised by the pathological occurrence of two opposite phenomena-osteoresorption and osteogenesis. Dickkopf-related protein 1 (DKK1) which inhibits the Wingless protein (Wnt) signalling pathway has been shown to be a master regulator of bone remodeling in inflammatory rheumatic diseases. However, the exact relationship between DKK1 serum level and bone remodelling is not clear. The goal of this study is to review state-of-the-art knowledge on the association of serum DKK1 with a bone remodelling in PsA. The MEDLINE-PubMed, EMBASE, Scopus, Web of Science and DOAJ databases were searched for appropriate papers. The English terms: 'DKK1', 'Dickkopf-1' 'Dickkopf related protein 1', 'psoriatic arthritis' and 'PsA' were used for search purposes. Eight original articles and two reviews were identified up to August 2023. In four out of 8 discussed studies DKK1 serum level was higher in PsA patients than in healthy controls [Dalbeth, p < 0.01; Diani, p < 0.001; Chung, p < 0.01; Abd el Hamid, p < 0.001)], it was comparable in another (Daousiss, p = 0.430) and was lower in two (Fassio2017, p < 0.05; Fassio2019, p < 0.05). In one study, the comparative groups included patients with axial spondyloarthritis, where DKK1 serum levels were lower in PsA groups [Jadon, peripheral PsA, p = 0.01]. The true relative serum concentration of DKK1 in PsA, as well as its influence on osteogenesis and osteoresorption, is still equivocal. Further studies on this matter with consistent and stringent methodology are warranted.
Collapse
Affiliation(s)
- Grzegorz Biedroń
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, Jakubowskiego 2, Krakow, Poland
| | - Marcin Czepiel
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Mariusz Korkosz
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, Jakubowskiego 2, Krakow, Poland
| |
Collapse
|
9
|
Ye L, Jin Y, Huang H, Ding F, Xu X, Liu C, Bao S, Wu J, Jin Y. The plasma levels of Dickkopf-1 elevated in patients with Juvenile Idiopathic Arthritis. Pediatr Res 2023; 94:1349-1354. [PMID: 37179434 DOI: 10.1038/s41390-023-02637-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/24/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND To explore the role of two major inhibitors of Wnt signal pathway, Dickkopf-1(DKK-1) and Sclerostin (SOST), in the pathogenesis of juvenile idiopathic arthritis (JIA). METHODS 88 patients with JIA, which including 49 patients with enthesitis-related arthritis (ERA), 21 oligoarthritis (oJIA) and 18 polyarthritis (pJIA), and 36 age-and sex-matched children as healthy controls (HC) were enrolled in this study. The plasma levels of DKK-1 and SOST, measured using commercially available ELISA kits, were analyzed the correlation between the levels of DKK-1/SOST and JIA, and were analyzed in 14 patients with JIA during before and after treatment. RESULTS Plasma levels of DKK-1 were significantly higher in the patients with JIA than that in HC, the elevation of DKK-1 level was positively correlated with HLA-B27 positive JIA. DKK-1 levels dropped significantly in patient with JIA after treatment (P < 0.05). There was no significant change in SOST levels among different subtypes of JIA, patients with JIA during before and after treatment, and HC. CONCLUSION It was suggested that the DKK-1 may have a certain correlation with the pathogenesis of JIA, and DKK-1 levels are more closely related to the HLA-B27 positive-ERA. IMPACT The abnormally elevated levels of Dickkopf-1 (DKK-1) may be involved in the pathogenesis of juvenile idiopathic arthritis (JIA). DKK-1 levels were more closely related to the HLA-B27 positive-enthesitis-related arthritis (ERA). DKK-1 is an inhibitor of Wnt signaling pathway that promotes osteoblastic new bone formation; it is very rare for pediatric patients with HLA-B27 positive-ERA to manifest typical spondylitis, while sacroiliac arthritis is relatively common, which may be related to the high levels of DKK-1, which is consistent with the early stage of ankylosing spondylitis (AS).
Collapse
Affiliation(s)
- Lili Ye
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Yingying Jin
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Hua Huang
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Fei Ding
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Xuemei Xu
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Chenxi Liu
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Shengfang Bao
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Jing Wu
- Institute of Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| | - Yanliang Jin
- Department of Rheumatology & Immunology, Shanghai Children's Medical Center, Shanghai Jiaotong University, School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Jaśkiewicz Ł, Chmielewski G, Kuna J, Stompór T, Krajewska-Włodarczyk M. The Role of Sclerostin in Rheumatic Diseases: A Review. J Clin Med 2023; 12:6248. [PMID: 37834893 PMCID: PMC10573925 DOI: 10.3390/jcm12196248] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Systemic connective tissue disorders constitute a heterogenous group of autoimmune diseases with the potential to affect a range of organs. Rheumatoid arthritis (RA) is a chronic, progressive, autoimmune inflammatory disease affecting the joints. Systemic lupus erythematosus (SLE) may manifest with multiple system involvement as a result of inflammatory response to autoantibodies. Spondyloarthropathies (SpAs) such as ankylosing spondylitis (AS) or psoriatic arthritis (PsA) are diseases characterised by the inflammation of spinal joints, paraspinal tissues, peripheral joints and enthesitis as well as inflammatory changes in many other systems and organs. Physiologically, sclerostin helps to maintain balance in bone tissue metabolism through the Wnt/β-catenin pathway, which represents a major intracellular signalling pathway. This review article aims to present the current knowledge on the role of sclerostin in the Wnt/β-catenin pathway and its correlation with clinical data from RA, SLE, AS and PsA patients.
Collapse
Affiliation(s)
- Łukasz Jaśkiewicz
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Grzegorz Chmielewski
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-900 Olsztyn, Poland
| | - Jakub Kuna
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-900 Olsztyn, Poland
| | - Tomasz Stompór
- Department of Nephrology, Hypertension and Internal Medicine, University of Warmia and Mazury in Olsztyn, 10-516 Olsztyn, Poland
| | - Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, 10-900 Olsztyn, Poland
| |
Collapse
|
11
|
Prajapati P, Doshi G. An Update on the Emerging Role of Wnt/β-catenin, SYK, PI3K/AKT, and GM-CSF Signaling Pathways in Rheumatoid Arthritis. Curr Drug Targets 2023; 24:1298-1316. [PMID: 38083893 DOI: 10.2174/0113894501276093231206064243] [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: 08/07/2023] [Revised: 10/14/2023] [Accepted: 11/14/2023] [Indexed: 01/06/2024]
Abstract
Rheumatoid arthritis is an untreatable autoimmune disorder. The disease is accompanied by joint impairment and anomalies, which negatively affect the patient's quality of life and contribute to a decline in manpower. To diagnose and treat rheumatoid arthritis, it is crucial to understand the abnormal signaling pathways that contribute to the disease. This understanding will help develop new rheumatoid arthritis-related intervention targets. Over the last few decades, researchers have given more attention to rheumatoid arthritis. The current review seeks to provide a detailed summary of rheumatoid arthritis, highlighting the basic description of the disease, past occurrences, the study of epidemiology, risk elements, and the process of disease progression, as well as the key scientific development of the disease condition and multiple signaling pathways and enumerating the most current advancements in discovering new rheumatoid arthritis signaling pathways and rheumatoid arthritis inhibitors. This review emphasizes the anti-rheumatoid effects of these inhibitors [for the Wnt/β-catenin, Phosphoinositide 3-Kinases (PI3K/AKT), Spleen Tyrosine Kinase (SYK), and Granulocyte-Macrophage Colony-Stimulating Factor (GM-CSF) signaling pathways], illustrating their mechanism of action through a literature search, current therapies, and novel drugs under pre-clinical and clinical trials.
Collapse
Affiliation(s)
- Pradyuman Prajapati
- SVKM's Dr Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, India
| | - Gaurav Doshi
- SVKM's Dr Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, India
| |
Collapse
|
12
|
Mao YM, Liao T, Ye QL, Wu GC, Zhang Q, Tao SS, Zhao CN, Wu Q, Dan YL, Pan HF, Ye DQ. Increased circulating sclerostin levels in rheumatoid arthritis patients: an updated meta-analysis. Z Rheumatol 2023; 82:51-58. [PMID: 34545431 DOI: 10.1007/s00393-021-01091-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2021] [Indexed: 01/13/2023]
Abstract
BACKGROUND Sclerostin, a regulator of bone metabolism and vascular calcification involved in regulating the Wnt/β-catenin signaling pathway, has been shown to be involved in the pathogenesis of rheumatoid arthritis (RA). However, current results regarding the circulating sclerostin level of RA patients are debatable. This study aimed to evaluate the circulating level of sclerostin in RA patients and briefly summarize its role. METHOD PubMed, EMBASE, and the Cochrane Library databases were systematically searched till May 27, 2021, for eligible articles. Useful data from all qualified papers were systematically extracted and analyzed using Stata 12.0 software (Stata Corp LP, College Station, TX, USA). RESULTS Overall, 13 qualifying studies including 1030 cases and 561 normal controls were analyzed in this updated meta-analysis. Forest plot of this meta-analysis showed that RA patients had higher circulating sclerostin levels (P < 0.001, standardized mean difference [SMD] = 0.916, 95% CI: 0.235-1.597) compared to normal controls. Subgroup analyses implied that age, region, and assay method were associated with sclerostin level in RA patients. CONCLUSION RA patients have higher circulating sclerostin levels, and these was influenced by age, region, and assay method.
Collapse
Affiliation(s)
- Yan-Mei Mao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, 230032, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Tao Liao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, 230032, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Qian-Ling Ye
- Department of Hematology, The Second Hospital, Anhui Medical University, Hefei, Anhui, China
| | - Guo-Cui Wu
- School of Nursing, Anhui Medical University, 15 Feicui Road, Hefei, Anhui, China
| | - Qin Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, 230032, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Sha-Sha Tao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, 230032, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Chan-Na Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, 230032, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Qian Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, 230032, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Yi-Lin Dan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, 230032, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, 230032, Hefei, Anhui, China. .,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China.
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, 230032, Hefei, Anhui, China. .,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, 81 Meishan Road, Hefei, Anhui, China.
| |
Collapse
|
13
|
Tao SS, Cao F, Sam NB, Li HM, Feng YT, Ni J, Wang P, Li XM, Pan HF. Dickkopf-1 as a promising therapeutic target for autoimmune diseases. Clin Immunol 2022; 245:109156. [DOI: 10.1016/j.clim.2022.109156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/24/2022] [Accepted: 10/06/2022] [Indexed: 11/03/2022]
|
14
|
Maeda K, Yoshida K, Nishizawa T, Otani K, Yamashita Y, Okabe H, Hadano Y, Kayama T, Kurosaka D, Saito M. Inflammation and Bone Metabolism in Rheumatoid Arthritis: Molecular Mechanisms of Joint Destruction and Pharmacological Treatments. Int J Mol Sci 2022; 23:2871. [PMID: 35270012 PMCID: PMC8911191 DOI: 10.3390/ijms23052871] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/31/2022] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease characterized by a variety of symptoms and pathologies often presenting with polyarthritis. The primary symptom in the initial stage is joint swelling due to synovitis. With disease progression, cartilage and bone are affected to cause joint deformities. Advanced osteoarticular destruction and deformation can cause irreversible physical disabilities. Physical disabilities not only deteriorate patients' quality of life but also have substantial medical economic effects on society. Therefore, prevention of the progression of osteoarticular destruction and deformation is an important task. Recent studies have progressively improved our understanding of the molecular mechanism by which synovitis caused by immune disorders results in activation of osteoclasts; activated osteoclasts in turn cause bone destruction and para-articular osteoporosis. In this paper, we review the mechanisms of bone metabolism under physiological and RA conditions, and we describe the effects of therapeutic intervention against RA on bone.
Collapse
Affiliation(s)
- Kazuhiro Maeda
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Ken Yoshida
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Tetsuro Nishizawa
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Kazuhiro Otani
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Yu Yamashita
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Hinako Okabe
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Yuka Hadano
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Tomohiro Kayama
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| | - Daitaro Kurosaka
- Division of Rheumatology, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.Y.); (K.O.); (D.K.)
| | - Mitsuru Saito
- Department of Orthopaedic Surgery, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (T.N.); (Y.Y.); (H.O.); (Y.H.); (T.K.); (M.S.)
| |
Collapse
|
15
|
Romero-Sánchez C, Giraldo S, Heredia-P AM, De Avila J, Chila-Moreno L, Londoño J, Valle-Oñate R, Bello-Gualtero JM, Bautista-Molano W. Association of Serum and Crevicular Fluid Dickkopf-1 Levels with Disease Activity and Periodontitis in Patients with Early Rheumatoid Arthritis. Curr Rheumatol Rev 2021; 18:124-135. [PMID: 34784873 DOI: 10.2174/1573397117666211116105118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/30/2021] [Accepted: 08/10/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The aim of this study was to assess DKK-1 levels, in Gingival Crevicular Fluid (GCF) and serum, as a biomarker for bone loss and disease activity in periodontitis and early RA (eRA). METHODS In this cross-sectional study, we obtained serum and GCF from 10 interproximal sites (Distal Buccal I/S, Mesio Buccal I/S, Distal Palatal/Lingual, Mesio Palatal/Lingual) according to the highest degree of inflammation by a patient for 240 sites from eRA patients. Patients received a periodontal assessment, a radiographic evaluation, tomography of interproximal sites, and DKK1 levels were determined by ELISA. Comparisons were performed by the Mann-Whitney U test and analysis by Chi2 test, and a logistic regression model was applied. RESULTS The mean age was 46.33 ± 12.0 years, the Disease Activity Score (DAS-28-ESR) was 4.08 ± 1.4. Periodontitis was present in 65.2% of the patients, and 59.6% of these patients had bone loss in interproximal sites. Higher GCF-DKK1 levels were associated with serum-DKK1 (OR:2.41 IC95% 1.14-5.09, p=0.021) and were related with DAS28-ESR (p=0.001), Routine Assessment of Patient Index Data 3 (RAPID 3) (p=0.001), and tender joints (p=0.040). Foot bone erosion and juxta-articular osteopenia were associated with high levels of serum-DKK1 (p=0.009 and 0.001, respectively). Serum-DKK1 were associated with SDAI (OR: 2.38 IC95% 1.03-5.52, p=0.043), RAPID 3 (p=0.001), and rheumatoid factor (p=0.018). The GCF-DKK1 levels were associated with periodontal bone loss (p=0.011), periodontitis (p=0.070) and its severity (OR: 2.58 IC95% 2.28-7.28, p=0.001). Bone loss was more frequent in buccal sites (73.5%) and was associated with increased levels of DKK1 (p=0.033). CONCLUSION In the early stages of the eRA disease, serum and GCF-DKK1 could be a biomarker for clinical disease activity and periodontal and articular bone erosion.
Collapse
Affiliation(s)
- Consuelo Romero-Sánchez
- Cellular and Molecular Immunology Group/ INMUBO, School of Dentistry, Universidad El Bosque, Bogotá. Colombia
| | - Sebastián Giraldo
- Clinical Immunology Group, Rheumatology and Immunology Department Hospital Militar Central/School of Medicine, Universidad Militar Nueva Granada/, Bogotá. Colombia
| | - Ana María Heredia-P
- Cellular and Molecular Immunology Group/ INMUBO, School of Dentistry, Universidad El Bosque, Bogotá. Colombia
| | - Juliette De Avila
- Cellular and Molecular Immunology Group/ INMUBO, School of Dentistry, Universidad El Bosque, Bogotá. Colombia
| | - Lorena Chila-Moreno
- Cellular and Molecular Immunology Group/ INMUBO, School of Dentistry, Universidad El Bosque, Bogotá. Colombia
| | - John Londoño
- Spondyloarthropathy Group, Rheumatology Department, Hospital Militar Central/Universidad de La Sabana, Bogotá. Colombia
| | - Rafael Valle-Oñate
- Clinical Immunology Group, Rheumatology and Immunology Department Hospital Militar Central/School of Medicine, Universidad Militar Nueva Granada/, Bogotá. Colombia
| | - Juan Manuel Bello-Gualtero
- Clinical Immunology Group, Rheumatology and Immunology Department Hospital Militar Central/School of Medicine, Universidad Militar Nueva Granada/, Bogotá. Colombia
| | - Wilson Bautista-Molano
- Cellular and Molecular Immunology Group/ INMUBO, School of Dentistry, Universidad El Bosque, Bogotá. Colombia
| |
Collapse
|
16
|
Radial BMD and serum CTX-I can predict the progression of carotid plaque in rheumatoid arthritis: a 3-year prospective cohort study. Arthritis Res Ther 2021; 23:258. [PMID: 34641970 PMCID: PMC8513174 DOI: 10.1186/s13075-021-02642-4] [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: 01/22/2021] [Accepted: 10/04/2021] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Patients with rheumatoid arthritis (RA) are almost twice as likely to develop cardiovascular disease (CVD) as those without. However, traditional CVD risks have been shown to underperform in RA patients; thus, we aimed to identify new surrogate risk factors to better reflect their atherosclerotic burden. METHODS A total of 380 RA patients with carotid atherosclerosis data were analyzed in this prospective cohort study. The primary outcome was carotid plaque progression over the 3-year follow-up period. Risk parameters assessed for the progression of carotid plaque were categorized as demographics, traditional CVD risks, RA-related risks, and bone parameters. RESULTS The progression of carotid plaque was associated with the level of rheumatoid factor (p = 0.025), serum C-terminal telopeptide of type-I collagen (CTX-I) (p = 0.014), and femur and distal radius bone mass density (BMD) (p = 0.007 and 0.004, respectively), as well as traditional CVD risk factors. In multivariable analyses, the bone parameters of serum CTX-I and distal radius BMD proved to be independent predictors of the progression of carotid plaque along with hyperlipidemia, smoking, and baseline carotid plaque (all, p < 0.05). Adding both serum CTX-I and distal radius BMD increased the carotid plaque progression prediction model's percentage of explained variance from 24 to 30%. CONCLUSION High serum CTX-I and lower radius BMD, reflecting high bone turnover, were independent risk factors for the progression of carotid plaque in RA patients, implicating the direct or indirect role of bone metabolism on the atherosclerotic burden.
Collapse
|
17
|
Elevated serum Dickkopf-1 is a biomarker for bone erosion in patients with psoriatic arthritis. Chin Med J (Engl) 2021; 134:2583-2588. [PMID: 34267065 PMCID: PMC8577657 DOI: 10.1097/cm9.0000000000001612] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background: Psoriatic arthritis (PsA) is an inflammatory arthropathy characterized by psoriasis and bone erosion on radiology. Dickkopf-1 (Dkk-1) is considered to be the main inhibitor of the Wnt signaling pathway and results in reduced osteoblast proliferation. The aim of this study was to investigate the serum level of Dkk-1 and its association with bone erosion in PsA patients. Methods: Serum Dkk-1 levels were measured by enzyme-linked immunosorbent assay (ELISA) in 69 patients with PsA and 60 controls, including 39 rheumatoid arthritis (RA) patients, and 21 healthy controls (HCs). Rheumatoid factor and anti-cyclic citrullinated peptide levels were also determined by ELISA. The association of Dkk-1 level with clinical and laboratory features of PsA was analyzed. Logistic regression analysis was used to analyze the risk factors for bone erosion in PsA. Results: Dkk-1 was elevated in 68.1% (47/69) of the patients with PsA, 46.2% (18/39) of RA patients, and 9.5% (2/21) of HCs. Serum Dkk-1 concentration was significantly higher in PsA patients compared with that in HCs. The level of serum Dkk-1 was correlated with a swollen joint count, and levels of complement components 3 and 4. Elevated Dkk-1 level (odds ratio = 4.440, 95% confidence interval: 1.246–15.817, P = 0.021) was identified as the risk factor for bone erosion in PsA. Conclusions: The serum level of Dkk-1 is abnormally elevated in PsA patients. The elevation of Dkk-1 might be involved in the mechanism of bone erosion in patients with PsA.
Collapse
|
18
|
Dickkopf-1 perpetuated synovial fibroblast activation and synovial angiogenesis in rheumatoid arthritis. Clin Rheumatol 2021; 40:4279-4288. [PMID: 34013491 DOI: 10.1007/s10067-021-05766-9] [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: 02/21/2021] [Revised: 04/26/2021] [Accepted: 05/05/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Dickkopf-1 (Dkk-1), a regulatory molecule of the Wnt pathway, is elevated and leads to bone resorption in patients with RA. This study is aimed to investigate the contribution of Dkk-1 to synovial inflammation and synovial fibroblast-mediated angiogenesis in RA. METHODS The expression of Dkk-1 in RA synovial fibroblasts (RASF) and osteoarthritis synovial fibroblasts (OASF) was detected by real-time PCR and ELISA, respectively. RASF were stimulated with different pro-inflammatory factors. The expression of angiogenic factors, pro-inflammatory cytokines, and MMPs in RASF was analyzed by real-time PCR when Dkk-1 was inhibited or overexpressed. Meanwhile, the concentrations of MCP-1, IL-6, IL-8, and MMP-3 in the cell culture supernatant were assessed by ELISA. The effects of Dkk-1 on the MAPK signaling pathway were evaluated by western blot. Matrigel tube formation assay was employed to reveal the direct and indirect effects of Dkk-1 on synovial angiogenesis. RESULTS Dkk-1 expression was elevated in synovial fluids and synovial fibroblasts of RA patients. Treatment with various pro-inflammatory cytokines significantly promoted DKK-1 expression in RASF. The production of potent angiogenic factors, pro-inflammatory cytokines, and MMPs in RASF was elevated, whereas the reverse results were found in the inhibitor groups. Silenced Dkk-1expression in RASF dampened capillary tube organization in both direct and indirect manners, resulting in restrained ERK, JNK, and p38 signaling pathway activation. CONCLUSION We concluded that Dkk-1 exacerbated the inflammation, cartilage erosion, and angiogenesis mediated by synovial fibroblasts in RA. Modulation of DKK-1 expression may facilitate development of novel strategies to control RA. Key points • Dkk-1 expression was elevated in synovial fluids and synovial fibroblasts of RA patients. • Treatment with various pro-inflammatory cytokines significantly promoted DKK-1 expression. • Silenced Dkk-1expression in RASF dampened capillary tube organization.
Collapse
|
19
|
Surmiak M, Kosałka‐Węgiel J, Polański S, Sanak M. Endothelial cells response to neutrophil-derived extracellular vesicles miRNAs in anti-PR3 positive vasculitis. Clin Exp Immunol 2021; 204:267-282. [PMID: 33527387 PMCID: PMC8062988 DOI: 10.1111/cei.13581] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
In vasculitis disorders, inflammation affects blood vessels. Granulomatosis with polyangiitis (GPA) is a chronic systemic vasculitis distinguished by the presence of anti-proteinase-3 autoantibodies (anti-PR3). In this study we analyzed the molecular signature of human umbilical endothelial cells (HUVECs) in response to neutrophil-derived extracellular vesicles (EVs). EVs were obtained from anti-PR3-activated neutrophils, purified and characterized by flow cytometry, nanoparticle tracking and miRNA screening. HUVECs were stimulated with EVs and miRNA/mRNA expression was measured. Cell culture media proteins were identified by antibody microarrays and selected cytokines were measured. Comparison of differentially expressed miRNAs/mRNAs between non-stimulated and EV-stimulated HUVECs revealed two regulatory patterns. Significant up-regulation of 14 mRNA transcripts (including CXCL8, DKK1, IL1RL1, ANGPT-2, THBS1 and VCAM-1) was accompanied by 11 miRNAs silencing (including miR-661, miR-664a-3p, miR-377-3p, miR-30d-5p). Significant down-regulation was observed for nine mRNA transcripts (including FASLG, CASP8, STAT3, GATA3, IRAK1 and IL6) and accompanied by up-regulation of 10 miRNAs (including miR-223-3p, miR-142-3p, miR-211-5p). Stimulated HUVECs released IL-8, Dickkopf-related protein 1 (DKK-1), soluble interleukin (IL)-1 like receptor-1 (ST2), growth differentiation factor 15 (GDF-15), angiopoietin-2, endoglin, thrombospondin-1 and vascular adhesion molecule-1 (VCAM-1). Moreover, transfection of HUVECs with mimics of highly expressed in EVs miR-223-3p or miR-142-3p, stimulated production of IL-8, ST2 and endoglin. Cytokines released by HUVECs were also elevated in blood of patients with GPA. The most increased were IL-8, DKK-1, ST2, angiopoietin-2 and IL-33. In-vitro stimulation of HUVECs by neutrophil-derived EVs recapitulates contribution of endothelium in autoimmune vasculitis. Proinflammatory phenotype of released cytokines corresponds with the regulatory network of miRNAs/mRNAs comprising both EVs miRNA and endothelial cell transcripts.
Collapse
Affiliation(s)
- M. Surmiak
- Department of Internal MedicineJagiellonian University Medical CollegeKrakówPoland
| | - J. Kosałka‐Węgiel
- Department of Internal MedicineJagiellonian University Medical CollegeKrakówPoland
| | - S. Polański
- Department of Biochemical and Molecular DiagnosticsUniversity HospitalKrakówPoland
| | - M. Sanak
- Department of Internal MedicineJagiellonian University Medical CollegeKrakówPoland
| |
Collapse
|
20
|
Bruno D, Fedele AL, Tolusso B, Barini A, Petricca L, Di Mario C, Barini A, Mirone L, Ferraccioli G, Alivernini S, Gremese E. Systemic Bone Density at Disease Onset Is Associated With Joint Erosion Progression in Early Naive to Treatment Rheumatoid Arthritis: A Prospective 12-Month Follow-Up Open-Label Study. Front Med (Lausanne) 2021; 8:613889. [PMID: 33732715 PMCID: PMC7959810 DOI: 10.3389/fmed.2021.613889] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 01/18/2021] [Indexed: 12/14/2022] Open
Abstract
Objectives: Osteoporosis and bone erosions are hallmarks of rheumatoid arthritis (RA) since disease onset is underpinned by the inflammatory burden. In this observational study, we aimed to dissect the putative RA-related parameters and bone-derived biomarkers associated with systemic and focal bone loss at disease onset and with their progression. Methods: One-hundred twenty-eight patients with early rheumatoid arthritis (ERA) were recruited at disease onset. At study entry, demographic, clinical, and immunological parameters were recorded. Each ERA patient underwent plain X-rays of the hands and feet at study entry and after 12 months to assess the presence of erosions. After enrollment, each patient was treated according to the recommendations for RA management and followed up based on a treat-to-target (T2T) strategy. At baseline, blood samples for soluble biomarkers were collected from each patient, and plasma levels of osteoprotegerin (OPG), receptor activator of nuclear factor κB ligand (RANKL), Dickkopf-1 (DKK1), and interleukin 6 (IL-6) were assessed by enzyme-linked immunosorbent assay (ELISA). Seventy-one ERA patients underwent bone mineral density (BMD) measurement at the left femoral neck and second to fourth lumbar spine vertebrae (L2–L4) by dual-energy X-ray absorptiometry (DXA). Results: Among the whole cohort, 34 (26.6%) ERA patients with bone erosions at study entry had a higher disease activity (p = 0.02) and IL-6 plasma levels (p = 0.03) than non-erosive ones. Moreover, at DXA, 33 (46.5%) ERA patients had osteopenia, and 16 (22.5%) had osteoporosis; patients with baseline bone erosions were more likely osteopenic/osteoporotic than non-erosive ones (p = 0.03), regardless of OPG, RANKL, and DKK1 plasma levels. Obese ERA patients were less likely osteopenic/osteoporotic than normal weight ones (p = 0.002), whereas anti-citrullinated protein antibodies (ACPA) positive ERA patients were more likely osteopenic/osteoporotic than ACPA negative ones (p = 0.034). At logistic regression analysis, baseline Disease Activity Score measured on 44 joints (DAS44) [OR: 2.46 (1.11–5.44)] and osteopenic/osteoporosis status [OR: 7.13 (1.27–39.94)] arose as independent factors of erosiveness. Baseline osteopenic/osteoporotic status and ACPA positivity were associated with bone damage progression during the follow-up. Conclusions: Bone erosions presence is associated with systemic bone loss since the earliest phases of RA, suggesting that the inflammatory burden and autoimmune biology, underpinning RA, represent crucial enhancers of bone remodeling either locally as at systemic level.
Collapse
Affiliation(s)
- Dario Bruno
- Division of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Laura Fedele
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Barbara Tolusso
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Angelina Barini
- Istituto di Biochimica e Biochimica Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luca Petricca
- Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Clara Di Mario
- Division of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonella Barini
- Istituto di Biochimica e Biochimica Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Luisa Mirone
- Division of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy.,Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Stefano Alivernini
- Division of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy.,Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elisa Gremese
- Division of Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy.,Division of Rheumatology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
21
|
Ibrahim NH, Abdel-Monem SM, Elbarashy AWSE, Elhussieny HAW, Elsayed RAH. Study of serum and synovial fluid Dickkopf-1 levels in patients with primary osteoarthritis of the knee joint in correlation with disease activity and severity. EGYPTIAN RHEUMATOLOGY AND REHABILITATION 2020. [DOI: 10.1186/s43166-020-00019-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Osteoarthritis (OA) is a typical complex degenerative articular ailment that shows focal cartilage loss, new bone formation with involvement of entire joint tissues. Dkk-1 assumes a job in controlling the pattern of bone repair and regeneration in both OA and RA. This study aimed to determine serum and synovial fluid levels of Dickkopf-1 (Dkk-1) in patients with primary OA of the knee joint and study their correlations with disease activity and severity. This study included 45 patients, 30 of them were diagnosed as primary knee OA. Fifteen rheumatoid arthritis patients as well as 15 healthy subjects were enrolled in the study as control groups, serum and synovial levels of Dkk-1 were estimated utilizing the ELISA technique.
Results
Serum levels of Dkk-1 were significantly higher in OA patients than healthy subjects (p < 0.001), although it was even significantly higher in RA patients than OA patients (p < 0.001). There was a highly significant decrease in the median synovial level of Dkk-1 in OA patients compared to the RA control group (p < 0.001). There was a highly statistically significant inverse correlation between circulating as well as synovial fluid Dkk-1 levels and radiological disease grading in knee OA (p < 0.001). There was a statistically significant decrease in serum levels of Dkk-1 in patients with severe OA (grade 3, 4) compared to those with mild OA (Grade 2) (p < 0.001).
Conclusion
Dkk-1 is an interesting marker that is related to articular disease .It could play an important role in decelerating the degenerative process of OA and can reflects radiographic severity of the disease as well.
Collapse
|
22
|
Association of Dickkopf-1 Polymorphisms With Radiological Damage and Periodontal Disease in Patients With Early Rheumatoid Arthritis. J Clin Rheumatol 2020; 26:S187-S194. [DOI: 10.1097/rhu.0000000000001391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
23
|
Subclinical inflammation in the preclinical phase of rheumatoid arthritis might contribute to articular joint damage. Hum Immunol 2020; 81:726-731. [PMID: 32690328 DOI: 10.1016/j.humimm.2020.07.003] [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: 01/03/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 01/26/2023]
Abstract
The first degree relatives of rheumatoid arthritis (RA) patients have a higher risk of developing RA, which is related to the expression of autoantibodies against citrullinated proteins (ACPA). Remarkably, prior to the onset of RA, cartilage damage is already initiated, whereas ACPA autoantibodies are already expressed. Here we show that both TNF-α and IL-6 are also increased prior to the onset of RA. Furthermore, when the levels of DKK1 and Sclerostin were evaluated in first degree relatives of RA patients, we found that the serum levels of TNF- α correlate with the expression levels of both DKK1 and Sclerostin. Interestingly, when the disease is already established, the correlation of TNF- α with DKK1 is lost in RA patients, whereas the correlation of Sclerostin with both TNF- α and IL-6 is further increased. Our data suggest a subclinical inflammation in patients at high risk of developing RA, which might lead to an increase in the levels of both DKK1 and Sclerostin, contributing to joint damage in the preclinical phase of the disease linked to the expression of ACPA autoantibodies.
Collapse
|
24
|
Möller B, Kollert F, Sculean A, Villiger PM. Infectious Triggers in Periodontitis and the Gut in Rheumatoid Arthritis (RA): A Complex Story About Association and Causality. Front Immunol 2020; 11:1108. [PMID: 32582191 PMCID: PMC7283532 DOI: 10.3389/fimmu.2020.01108] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic immune mediated inflammatory disease of unknown origin, which is predominantly affecting the joints. Antibodies against citrullinated peptides are a rather specific immunological hallmark of this heterogeneous entity. Furthermore, certain sequences of the third hypervariable region of human leukocyte antigen (HLA)-DR class II major histocompatibility (MHC) molecules, the so called "shared epitope" sequences, appear to promote autoantibody positive types of RA. However, MHC-II molecule and other genetic associations with RA could not be linked to immune responses against specific citrullinated peptides, nor do genetic factors fully explain the origin of RA. Consequently, non-genetic factors must play an important role in the complex interaction of endogenous and exogenous disease factors. Tobacco smoking was the first environmental factor that was associated with onset and severity of RA. Notably, smoking is also an established risk factor for oral diseases. Furthermore, smoking is associated with extra-articular RA manifestations such as interstitial lung disease in anatomical proximity to the airway mucosa, but also with subcutaneous rheumatoid nodules. In the mouth, Porphyromonas gingivalis is a periodontal pathogen with unique citrullinating capacity of foreign microbial antigens as well as candidate RA autoantigens. Although the original hypothesis that this single pathogen is causative for RA remained unproven, epidemiological as well as experimental evidence linking periodontitis (PD) with RA is rapidly accumulating. Other periopathogens such as Aggregatibacter actinomycetemcomitans and Prevotella intermedia were also proposed to play a specific immunodominant role in context of RA. However, demonstration of T cell reactivity against citrullinated, MHC-II presented autoantigens from RA synovium coinciding with immunity against Prevotella copri (Pc.), a gut microbe attracted attention to another mucosal site, the intestine. Pc. was accumulated in the feces of clinically healthy subjects with citrulline directed immune responses and was correlated with RA onset. In conclusion, we retrieved more than one line of evidence for mucosal sites and different microbial taxa to be potentially involved in the development of RA. This review gives an overview of infectious agents and mucosal pathologies, and discusses the current evidence for causality between different exogenous or mucosal factors and systemic inflammation in RA.
Collapse
Affiliation(s)
- Burkhard Möller
- Department for Rheumatology, Immunology and Allergology, Inselspital-University Hospital of Bern, Bern, Switzerland
| | - Florian Kollert
- Department for Rheumatology, Immunology and Allergology, Inselspital-University Hospital of Bern, Bern, Switzerland
| | - Anton Sculean
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Peter M Villiger
- Department for Rheumatology, Immunology and Allergology, Inselspital-University Hospital of Bern, Bern, Switzerland
| |
Collapse
|
25
|
Evaluating the role of serum sclerostin as an indicator of activity and damage in Egyptian patients with rheumatoid arthritis: university hospital experience. Clin Rheumatol 2019; 39:1121-1130. [DOI: 10.1007/s10067-019-04878-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 11/25/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022]
|
26
|
Heredia-P AM, Lafaurie GI, Bautista-Molano W, Trujillo TG, Chalem-Choueka P, Bello-Gualtero JM, Pacheco-Tena C, Chila-Moreno L, Romero-Sánchez C. Predictive factors related to the progression of periodontal disease in patients with early rheumatoid arthritis: a cohort study. BMC Oral Health 2019; 19:240. [PMID: 31703715 PMCID: PMC6842164 DOI: 10.1186/s12903-019-0939-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/24/2019] [Indexed: 02/07/2023] Open
Abstract
Background Rheumatoid arthritis (RA) and periodontal disease are inter-related conditions. However, factors predictive of periodontal disease progression in patients with early rheumatoid arthritis (eRA) are lacking. The aim of this study was to identify factors associated with the progression of clinical attachment loss (CAL) in interproximal dental sites of eRA patients. Methods Twenty-eight eRA patients were evaluated for the progression of CAL at 280 interproximal dental sites at 1 year of follow-up. Markers of RA activity (rheumatoid factor, erythrocyte sedimentation rate, and C-reactive protein), a marker of bone resorption (Dickkopf-related protein 1), Disease Activity Score 28 and Simple Disease Activity Index were included as potential systemic predictive factors. Plaque index, gingival index, pocket depth, clinical attachment level and Dickkopf-related protein 1 in crevicular fluid at baseline were included as potential local predictive factors. Data were analysed in a hierarchical structure using generalised linear mixed models for progression at each site (> 2 mm) during follow-up. Results C-reactive protein level was the most important predictive systemic factor for the progression of CAL. The mean CAL and a high degree of gingival inflammation in interproximal sites at baseline were important predictive local factors (p < 0.0001). Patients who received combined treatment with disease-modifying antirheumatic drugs and corticosteroids exhibited less CAL (p < 0.0001). The predictive value of the generalised linear mixed model for progression was 85%. Conclusions Systemic factors, including RA disease activity and baseline periodontal condition, were associated with periodontal progression. Pharmacological treatment may affect periodontal progression in patients with early RA.
Collapse
Affiliation(s)
- Ana María Heredia-P
- Unit of Basic Oral Investigation-UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | - Gloria Inés Lafaurie
- Unit of Basic Oral Investigation-UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | - Wilson Bautista-Molano
- Clinical Immunology Group, Rheumatology and Immunology Department Hospital Militar Central/School of Medicine, Universidad Militar Nueva Granada, Transversal 3ª #, 49-00, Bogotá, Colombia.,Cellular and Molecular Immunology Group/ INMUBO, School of Dentistry, Universidad El Bosque, Cra 9 No. 131 A-02, Bogotá, Colombia
| | - Tamy Goretty Trujillo
- Unit of Basic Oral Investigation-UIBO, School of Dentistry, Universidad El Bosque, Bogotá, Colombia
| | | | - Juan M Bello-Gualtero
- Clinical Immunology Group, Rheumatology and Immunology Department Hospital Militar Central/School of Medicine, Universidad Militar Nueva Granada, Transversal 3ª #, 49-00, Bogotá, Colombia
| | | | - Lorena Chila-Moreno
- Cellular and Molecular Immunology Group/ INMUBO, School of Dentistry, Universidad El Bosque, Cra 9 No. 131 A-02, Bogotá, Colombia
| | - Consuelo Romero-Sánchez
- Clinical Immunology Group, Rheumatology and Immunology Department Hospital Militar Central/School of Medicine, Universidad Militar Nueva Granada, Transversal 3ª #, 49-00, Bogotá, Colombia. .,Cellular and Molecular Immunology Group/ INMUBO, School of Dentistry, Universidad El Bosque, Cra 9 No. 131 A-02, Bogotá, Colombia.
| |
Collapse
|
27
|
Wnt Signaling and Biological Therapy in Rheumatoid Arthritis and Spondyloarthritis. Int J Mol Sci 2019; 20:ijms20225552. [PMID: 31703281 PMCID: PMC6888549 DOI: 10.3390/ijms20225552] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/02/2019] [Accepted: 11/06/2019] [Indexed: 12/17/2022] Open
Abstract
The Wnt signaling pathway plays a key role in several biological processes, such as cellular proliferation and tissue regeneration, and its dysregulation is involved in the pathogenesis of many autoimmune diseases. Several evidences support its role especially in bone complications of rheumatic diseases. In Rheumatoid Arthritis (RA), the Wnt signaling is implicated in systemic and localized bone loss, while available data of its role in Spondyloarthritis (SpA) are conflicting. In the last few decades, the quality of life of rheumatic patients has been dramatically improved by biological therapy, targeting cytokines involved in the pathogenesis of these diseases like tumor necrosis factor (TNF)α, interleukin (IL)-1, IL-6, IL-17. In this review, we reviewed the role of Wnt signaling in RA and SpA, focusing on the effect of biological therapy on this pathway and its possible clinical implications.
Collapse
|
28
|
Nácher-Juan J, Terencio MC, Alcaraz MJ, Ferrándiz ML. Osteostatin Inhibits Collagen-Induced Arthritis by Regulation of Immune Activation, Pro-Inflammatory Cytokines, and Osteoclastogenesis. Int J Mol Sci 2019; 20:E3845. [PMID: 31394717 PMCID: PMC6721041 DOI: 10.3390/ijms20163845] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/04/2019] [Accepted: 08/05/2019] [Indexed: 01/05/2023] Open
Abstract
In chronic inflammatory joint diseases, such as rheumatoid arthritis, there is an important bone loss. Parathyroid hormone-related protein (PTHrP) and related peptides have shown osteoinductive properties in bone regeneration models, but there are no data on inflammatory joint destruction. We have investigated whether the PTHrP (107-111) C-terminal peptide (osteostatin) could control the development of collagen-induced arthritis in mice. Administration of osteostatin (80 or 120 μg/kg s.c.) after the onset of disease decreased the severity of arthritis as well as cartilage and bone degradation. This peptide reduced serum IgG2a levels as well as T cell activation, with the downregulation of RORγt+CD4+ T cells and upregulation of FoxP3+CD8+ T cells in lymph nodes. The levels of key cytokines, such as interleukin(IL)-1β, IL-2, IL-6, IL-17, and tumor necrosis factor-α in mice paws were decreased by osteostatin treatment, whereas IL-10 was enhanced. Bone protection was related to reductions in receptor activator of nuclear factor-κB ligand, Dickkopf-related protein 1, and joint osteoclast area. Osteostatin improves arthritis and controls bone loss by inhibiting immune activation, pro-inflammatory cytokines, and osteoclastogenesis. Our results support the interest of osteostatin for the treatment of inflammatory joint conditions.
Collapse
Affiliation(s)
- Josep Nácher-Juan
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain
| | - María Carmen Terencio
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain
| | - María José Alcaraz
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain.
| | - María Luisa Ferrándiz
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Av. Vicent A. Estellés s/n, 46100 Burjasot, Valencia, Spain.
| |
Collapse
|
29
|
Singh A, Gupta MK, Mishra SP. Study of correlation of level of expression of Wnt signaling pathway inhibitors sclerostin and dickkopf-1 with disease activity and severity in rheumatoid arthritis patients. Drug Discov Ther 2019; 13:22-27. [PMID: 30880318 DOI: 10.5582/ddt.2019.01011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This study was done with aim to assess the serum sclerostin and dickkopf-1 (DKK-1) level in patients of rheumatoid arthritis (RA) and to correlate their level with disease activity and bone mineral density. Fifty patients of RA and equal age and sex matched healthy controls were included in the study. Patients were evaluated clinically and investigated with routine blood tests along with rheumatoid factor (RF), anti-citrullinated protein antibody (anti-CCP2), radiographs and bone mineral density (BMD). Serum sclerostin and DKK-1 levels of both cases and controls was assayed by using enzyme-linked immunosorbent assay (ELISA) assay [RayBio®, Georgia, USA with coefficient of variation percent (CV %), < 10%] and compared with disease activity and bone mineral density. Disease activity was measured by Disease Activity Score 28 (DAS28) along with Modified Health Assessment Questionnaire (MHAQ) score. Mean serum sclerostin and DKK-1 was significantly higher in study group as compared to control group. Serum sclerostin showed significant correlation with disease activity scores (DAS score and MHAQ score), erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) level. Serum sclerostin at level of 394 pg/mL was found to have diagnostic significance with sensitivity of 100% and specificity of 90%. DKK-1 level shows significantly positive correlation with larson score which denotes radiological progression (r value 0.468; p value 0.001). More studies with larger sample size of RA patients are needed for better determination of the role of sclerostin and DKK-1 in RA. Also, the correlation of these and other bone turn over markers will help decipher their role with disease progression in RA patients.
Collapse
Affiliation(s)
- Anup Singh
- Department of Geriatric Medicine, Institute of Medical Sciences, Banaras Hindu University
| | - Manish Kumar Gupta
- Department of General Medicine, Institute of Medical Sciences, Banaras Hindu University
| | | |
Collapse
|
30
|
Fassio A, Adami G, Gatti D, Orsolini G, Giollo A, Idolazzi L, Benini C, Vantaggiato E, Rossini M, Viapiana O. Inhibition of tumor necrosis factor-alpha (TNF-alpha) in patients with early rheumatoid arthritis results in acute changes of bone modulators. Int Immunopharmacol 2018; 67:487-489. [PMID: 30599401 DOI: 10.1016/j.intimp.2018.12.050] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/17/2018] [Accepted: 12/22/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Dicckopf-1 (Dkk-1) is a potent inhibitor of the Wnt canonical pathway. In rheumatoid arthritis (RA), Dkk-1 is upregulated by tumor necrosis factor-α (TNF). Certolizumab pegol (CMZ) is a biologic TNF-inhibitor (TNFi) effective in RA and slows radiographic progression. Data on the immediate effects (≤1-8 weeks) of TNFi on Wnt modulators are lacking. This study investigated the acute influence of TNFi treatment on Wnt modulators (Dkk-1 and sclerostin) and bone turnover markers (BTM), including intact N-terminal propeptide of collagen type I (PINP) and C-terminal telopeptide of type I collagen (CTX-I). METHODS This longitudinal, uncontrolled study involved female RA patients with inadequate response to conventional methotrexate who underwent treatment with CMZ. ESR, Dkk-1, sclerostin, BTM, parathyroid hormone (PTH), and 25OH-vitamin D levels were evaluated at baseline, week 1, week 4, and week 8. Radiographs of the hands and feet were obtained at baseline and the total and erosion scores were assessed using the Simple Erosion Narrowing Score method (SENS). RESULTS Seventeen patients were enrolled. Dkk-1 and CTX-I significantly decreased after one week of treatment with CMZ (-49.1 ± 17.1% and -25.0 ± 20.6%, respectively, p < 0.01), whereas PINP increased (+43.2 ± 31.5%, p < 0.01). These changes persisted at week 4 and 8. CONCLUSIONS Our study showed that TNF-alpha inhibition with CMZ promptly results in a rapid decline of serum Dkk-1 levels, alongside decreased bone resorption and increased bone formation.
Collapse
Affiliation(s)
- A Fassio
- Rheumatology Unit, University of Verona, Ospedale Civile Maggiore, Piazzale A. Scuro, 37134 Verona, Italy.
| | - G Adami
- Rheumatology Unit, University of Verona, Ospedale Civile Maggiore, Piazzale A. Scuro, 37134 Verona, Italy
| | - D Gatti
- Rheumatology Unit, University of Verona, Ospedale Civile Maggiore, Piazzale A. Scuro, 37134 Verona, Italy
| | - G Orsolini
- Rheumatology Unit, University of Verona, Ospedale Civile Maggiore, Piazzale A. Scuro, 37134 Verona, Italy
| | - A Giollo
- Rheumatology Unit, University of Verona, Ospedale Civile Maggiore, Piazzale A. Scuro, 37134 Verona, Italy
| | - L Idolazzi
- Rheumatology Unit, University of Verona, Ospedale Civile Maggiore, Piazzale A. Scuro, 37134 Verona, Italy
| | - C Benini
- Rheumatology Unit, University of Verona, Ospedale Civile Maggiore, Piazzale A. Scuro, 37134 Verona, Italy
| | - E Vantaggiato
- Rheumatology Unit, University of Verona, Ospedale Civile Maggiore, Piazzale A. Scuro, 37134 Verona, Italy
| | - M Rossini
- Rheumatology Unit, University of Verona, Ospedale Civile Maggiore, Piazzale A. Scuro, 37134 Verona, Italy
| | - O Viapiana
- Rheumatology Unit, University of Verona, Ospedale Civile Maggiore, Piazzale A. Scuro, 37134 Verona, Italy
| |
Collapse
|
31
|
Ha YJ, Han DW, Kim JH, Chung SW, Kang EH, Song YW, Lee YJ. Circulating Semaphorin 4D as a Marker for Predicting Radiographic Progression in Patients with Rheumatoid Arthritis. DISEASE MARKERS 2018; 2018:2318386. [PMID: 30538782 PMCID: PMC6261241 DOI: 10.1155/2018/2318386] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022]
Abstract
Semaphorin 3A (Sema3A) and semaphorin 4D (Sema4D) are molecules which regulate immune responses as well as bone remodeling process. The aim of this study was to evaluate the serum levels of Sema3A and Sema4D and to investigate their clinical significance in rheumatoid arthritis (RA). The serum levels of Sema3A and Sema4D were measured in 130 patients with RA and 65 sex- and age-matched healthy individuals. Circulating levels of biomarkers of RA-related inflammation and bone turnover such as tumor necrosis factor- (TNF-) α, interleukin- (IL-) 6, IL-22, IL-34, osteopontin, Dkk-1, and sclerostin were also measured. Disease activity was determined by the 28-joint disease activity score (DAS28), and radiographic joint damage was assessed by the modified Sharp van der Heijde score (SHS). The serum levels of Sema3A were significantly higher in patients with RA than those in healthy controls (p < 0.001), whereas serum4D levels did not differ between the two groups. The levels of Sema4D showed a positive correlation with C-reactive protein (p = 0.001) and IL-6 (p < 0.001) levels, whereas the levels of Sema3A showed a negative correlation with Dkk-1 (p = 0.007) and TNF-α (p = 0.001). Even though Sema3A and Sema4D levels were comparable between RA patients with DAS28> 3.2 and with DAS28 ≤ 3.2, RA patients with radiographic progression (ΔSHS change/year ≥ 1) had significantly higher baseline levels of Sema4D than those without progression (p = 0.029). Additionally, when RA patients were divided into 3 groups using tertiles of Sema4D levels, the percentage of progressors was significantly increased (p = 0.045). In multivariate logistic regression analysis, serum Sema4D levels were an independent risk factor for radiographic progression. Our results suggest that the baseline levels of Sema4D might be a useful marker to identify RA patients with subsequent radiographic progression and that Sema4D may be an active mediator involved in RA-induced joint damage.
Collapse
Affiliation(s)
- You-Jung Ha
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Dong Woo Han
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ji Hyoun Kim
- Division of Rheumatology, Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Sang Wan Chung
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Eun Ha Kang
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yeong Wook Song
- WCU Department of Molecular Medicine and Biopharmaceutical Sciences, Medical Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yun Jong Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Translational Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
32
|
Dickkopf-1 (Dkk-1) serum levels in systemic sclerosis and rheumatoid arthritis patients: correlation with the Trabecular Bone Score (TBS). Clin Rheumatol 2018; 37:3057-3062. [PMID: 30291470 DOI: 10.1007/s10067-018-4322-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/25/2018] [Accepted: 10/01/2018] [Indexed: 12/17/2022]
Abstract
The aim of this research was to determine any correlations between Dickkopf-1 serum levels (Dkk-1, a natural inhibitor of the Wnt signaling pathway promoting osteoclastogenesis) and the Trabecular Bone Score (TBS), in systemic sclerosis (SSc) and rheumatoid arthritis (RA) patients. It also aimed at determining any difference in Dkk-1 serum levels between SSc and RA patients and a control group (CNT) of healthy subjects. A prospective study was carried out in 60 SSc and 60 RA patients and 60 CNT. Dkk-1 serum levels were evaluated by the ELISA method (Quantikine Human Dkk-1 Immunoassay, R&D System, Minneapolis, USA). The severity of microvascular damage was evaluated by nailfold videocapillaroscopy (NVC patterns: "Early," "Active," "Late"), in the SSc patients. TBS analysis and bone mineral density (BMD, g/cm2) were measured in all patients/subjects. The SSc patients showed higher Dkk-1 serum levels than RA (p < 0.004) and CNT (p < 0.0001) patients. SSc patients, showing the "Late" NVC pattern had statistically higher Dkk-1 serum levels than patients with either the "Active" or "Early" (p < 0.004) patterns. Only in the "Late" NVC pattern group of SSc patients was there a significant negative correlation between Dkk-1 and TBS values (p < 0.0001). The increased Dkk-1 serum levels and decreased TBS values observed suggest a diffuse bone damage in SSc patients with advanced disease, as demonstrated by the concomitant presence of the "Late" NVC pattern. Moreover, the bone remodeling in SSc seems even higher than that in RA patients.
Collapse
|
33
|
Bone erosions in rheumatoid arthritis: recent developments in pathogenesis and therapeutic implications. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2018; 18:304-319. [PMID: 30179207 PMCID: PMC6146189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Bone erosions develop early in the course of rheumatoid arthritis (RA) and deteriorate progressively, causing joint damage and resulting in impaired functional capacity of patients. During the last years, considerable number of studies has increased our understanding of the pathogenetic mechanisms mediating the development of bone erosions in RA. Increased production of RANKL and other cytokines, dysregulation of innate immune mechanisms, autoantibodies specific to RA and alterations of microRNA expression stimulate differentiation and function of osteoclasts, which are responsible for the development of bone erosions. Besides, increased levels of cytokines, overproduction of antagonists of the canonical Wnt signaling pathway and deficient production of bone morphogenetic proteins result in impaired osteoblast differentiation and function, undermining the capacity of bone erosions to repair. Disease-modifying antirheumatic drugs, synthetic or biological, currently used in the treatment of RA, can halt the progression of bone erosions and may even lead to partial repair, although complete repair is unattainable. Targeting pathogenetic mechanisms participating in the erosive process may add to the therapeutic effect of DMARDs and help in the prevention or repair of bone erosions. However, more studies are still needed to confirm whether such therapeutic strategies are effective.
Collapse
|
34
|
Iwamoto N, Fukui S, Takatani A, Shimizu T, Umeda M, Nishino A, Igawa T, Koga T, Kawashiri SY, Ichinose K, Tmai M, Nakamura H, Origuchi T, Chiba K, Osaki M, Jüngel A, Gay S, Kawakami A. Osteogenic differentiation of fibroblast-like synovial cells in rheumatoid arthritis is induced by microRNA-218 through a ROBO/Slit pathway. Arthritis Res Ther 2018; 20:189. [PMID: 30157923 PMCID: PMC6116572 DOI: 10.1186/s13075-018-1703-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 08/16/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Fibroblast-like synovial cells (FLS) have multilineage differentiation potential including osteoblasts. We aimed to investigate the role of microRNAs during the osteogenic differentiation of rheumatoid arthritis (RA)-FLS. METHODS RA-FLS were differentiated in osteogenic medium for 21 days. Osteogenic differentiation was evaluated by alkaline phosphatase (ALP) staining and Alizarin Red staining. MicroRNA (miRNA) array analysis was performed to investigate the differentially expressed miRNAs during osteogenic differentiation. Expression of miR-218-5p (miR-218) during the osteogenic differentiation was determined by quantitative real-time PCR. Transfections with an miR-218 precursor and inhibitor were used to confirm the targets of miR-218 and to analyze the ability of miR-218 to induce osteogenic differentiation. Secreted Dickkopf-1 (DKK1) from FLS transfected with miR-218 precursor/inhibitor or roundabout 1 (ROBO1) knockdown FLS established using ROBO1-small interfering RNA (siRNA) were measured by ELISA. RESULTS The miRNA array revealed that 12 miRNAs were upregulated and 24 miRNAs were downregulated after osteogenic differentiation. We observed that the level of miR-218 rose in the early phase of osteogenic differentiation and then decreased. Pro-inflammatory cytokines modified the expression of miR-218. The induction of miR-218 in RA-FLS decreased ROBO1 expression, and promoted osteogenic differentiation. Both the overexpression of miR-218 and the knockdown of ROBO1 in RA-FLS decreased DKK1 secretion. CONCLUSION We identified miR-218 as a crucial inducer of the osteogenic differentiation of RA-FLS. MiR-218 modulates the osteogenic differentiation of RA-FLS through the ROBO1/DKK-1 axis. The induction of the osteogenic differentiation of proliferating RA-FLS through the provision of miR-218 into RA-FLS or by boosting the cellular reservoir of miR-218 might thus become a therapeutic strategy for RA.
Collapse
Affiliation(s)
- Naoki Iwamoto
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Shoichi Fukui
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Ayuko Takatani
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Toshimasa Shimizu
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Masataka Umeda
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
- Medical Education Development Center, Nagasaki University School Hospital, Nagasaki, Japan
| | - Ayako Nishino
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
- Center for Comprehensive Community Care Education, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Takashi Igawa
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Tomohiro Koga
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
- Center for Bioinformatics and Molecular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shin-ya Kawashiri
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
- Departments of Community Medicine, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kunihiro Ichinose
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Mami Tmai
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Hideki Nakamura
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| | - Tomoki Origuchi
- Department of Physical Therapy, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Ko Chiba
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Makoto Osaki
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Astrid Jüngel
- Center of Experimental Rheumatology, University Hospital Zurich and University of Zurich, Schlieren, Zurich, Switzerland
| | - Steffen Gay
- Center of Experimental Rheumatology, University Hospital Zurich and University of Zurich, Schlieren, Zurich, Switzerland
| | - Atsushi Kawakami
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501 Japan
| |
Collapse
|
35
|
Orsolini G, Adami G, Rossini M, Ghellere F, Caimmi C, Fassio A, Idolazzi L, Gatti D, Viapiana O. Parathyroid hormone is a determinant of serum Dickkopf-1 levels in ankylosing spondylitis. Clin Rheumatol 2018; 37:3093-3098. [PMID: 29995219 DOI: 10.1007/s10067-018-4205-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/12/2018] [Accepted: 07/09/2018] [Indexed: 12/15/2022]
Abstract
Available studies reported contradictory results about serum levels Dickkopf-1 (DKK1), an inhibitor of Wnt signaling in patients with ankylosing spondylitis (AS). In previous studies, we observed in other conditions that parathyroid hormone (PTH) serum levels were an important determinant of DKK1 serum levels. The aim of the present study was to investigate it in patients with AS. We recruited 71 patients diagnosed with AS. Levels of C-reactive protein (CRP), DKK1, PTH, 25OH-vitamin D, and bone turnover markers (intact N-propeptide of type I collagen, P1NP, and C-terminal telopeptide of type I collagen, CTX) were measured and compared to healthy controls (HC). Dual X-ray absorptiometry at lumbar spine and proximal femoral site was used for bone mineral density (BMD) assessment and spine X-rays were also performed. PTH serum levels were found to be significantly higher in AS patients than in HC (33.8 ± 14.11 vs 24.8 ± 13 pg/ml, p = 0.002), while mean DKK1 serum levels were lower than in HC (23.3 ± 13.1 vs 29.8 ± 15.9 pmol/l, p = 0.009). A positive correlation between DKK1 and PTH (correlation coefficient + 0.25, p = 0.03) was observed; it remained significant in a multivariate analysis. In patients with longer disease duration, DKK1 was also positively correlated with CTX (coefficient 0.42, p = 0.01), and PTH was higher in those patients with low BMD (Z-score ≤ - 1) at any site (p = 0.04). Also in AS, PTH is an important determinant of DKK1 serum levels and should be evaluated in studies on DKK1. PTH might have a role in bone involvement in AS, also through the Wnt pathway.
Collapse
Affiliation(s)
- Giovanni Orsolini
- University of Verona, Rheumatology Unit, Piazzale L. Scuro 10, 37134, Verona, Italy.
| | - Giovanni Adami
- University of Verona, Rheumatology Unit, Piazzale L. Scuro 10, 37134, Verona, Italy
| | - Maurizio Rossini
- University of Verona, Rheumatology Unit, Piazzale L. Scuro 10, 37134, Verona, Italy
| | - Francesco Ghellere
- University of Verona, Rheumatology Unit, Piazzale L. Scuro 10, 37134, Verona, Italy
| | - Cristian Caimmi
- University of Verona, Rheumatology Unit, Piazzale L. Scuro 10, 37134, Verona, Italy
| | - Angelo Fassio
- University of Verona, Rheumatology Unit, Piazzale L. Scuro 10, 37134, Verona, Italy
| | - Luca Idolazzi
- University of Verona, Rheumatology Unit, Piazzale L. Scuro 10, 37134, Verona, Italy
| | - Davide Gatti
- University of Verona, Rheumatology Unit, Piazzale L. Scuro 10, 37134, Verona, Italy
| | - Ombretta Viapiana
- University of Verona, Rheumatology Unit, Piazzale L. Scuro 10, 37134, Verona, Italy
| |
Collapse
|
36
|
Huang X, Post JN, Zhong L, Leijten J, Larsson S, Karperien M, Struglics A. Dickkopf-related protein 1 and gremlin 1 show different response than frizzled-related protein in human synovial fluid following knee injury and in patients with osteoarthritis. Osteoarthritis Cartilage 2018. [PMID: 29526783 DOI: 10.1016/j.joca.2018.02.904] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To explore the involvement of the wingless-type MMTV integration site (WNT) and bone morphogenetic protein (BMP) antagonists dickkopf-related protein 1 (DKK1), frizzled-related protein (FRZB) and gremlin 1 (GREM1) in knee injury and osteoarthritis (OA). DESIGN The antagonists were immunoassayed in synovial fluid from a cross-sectional cohort of nine knee healthy reference subjects, patients with recent (0-77 days, n = 158) or old (1-37 years, n = 50) knee injuries, and OA (n = 22). Cartilage (ARGS-aggrecan, cartilage oligomeric matrix protein and C2C type II collagen) and other biomarkers were assessed in synovial fluid in a subset of samples. Statistical analysis was by Kendall's tau (τ) correlation, Mann-Whitney U test, and linear regression analysis. RESULTS Compared to references, median concentration of GREM1 (but not DKK1 and FRZB) was elevated 1.5-fold immediately after injury, and FRZB was reduced 1000-folds in OA. All three antagonists decreased with increasing time after injury as well as with increasing age, but the temporal change after injury was less accentuated for FRZB (peaked 8-22 days after injury) compared to that of DKK1 and GREM1 (peaked immediately after injury). In the recent injury group, there was a correlation between GREM1 and DKK1 (τ = 0.172); FRZB concentrations correlated with concentrations of cartilage biomarkers (τ between 0.257 and 0.369), while DKK1 and GREM1 were inversely correlated (τ between -0.177 and -0.217) with these markers. CONCLUSIONS Our results indicate separate roles for the antagonists, where DKK1 and GREM1 had similarities in response to injury and in OA, with a different response for FRZB.
Collapse
Affiliation(s)
- X Huang
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.
| | - J N Post
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.
| | - L Zhong
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.
| | - J Leijten
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.
| | - S Larsson
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Lund, Sweden.
| | - M Karperien
- Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands.
| | - A Struglics
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Lund, Sweden.
| |
Collapse
|
37
|
Luchetti MM, Ciccia F, Avellini C, Benfaremo D, Guggino G, Farinelli A, Ciferri M, Rossini M, Svegliati S, Spadoni T, Bolognini L, Fava G, Mosca P, Gesuita R, Skrami E, Triolo G, Gabrielli A. Sclerostin and Antisclerostin Antibody Serum Levels Predict the Presence of Axial Spondyloarthritis in Patients with Inflammatory Bowel Disease. J Rheumatol 2018; 45:630-637. [PMID: 29419466 DOI: 10.3899/jrheum.170833] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The early diagnosis of inflammatory bowel disease (IBD)-associated spondyloarthritis (SpA/IBD) in patients affected by IBD represents a major topic in clinical practice; in particular, to date there are no available serum biomarkers revealing the presence of joint inflammation in these patients. Sclerostin (SOST), an antagonist of the Wnt/β-catenin pathway, and antisclerostin-immunoglobulin G (anti-SOST-IgG) have been recently studied in patients with ankylosing spondylitis (AS) as a putative marker of disease activity. METHODS SOST and anti-SOST-IgG serum levels were assayed in 125 patients with IBD, 85 with axial or peripheral SpA, and in control groups (patients with AS and rheumatoid arthritis, and healthy individuals). The diagnostic performance in discriminating the presence of SpA/IBD was assessed for both candidate biomarkers. RESULTS Patients affected by SpA/IBD with axial involvement displayed significantly lower levels of SOST and higher levels of anti-SOST-IgG compared to patients with only peripheral arthritis, IBD, and controls. Moreover, SOST and anti-SOST-IgG serum levels were inversely correlated and were associated with the duration of articular symptoms. Both biomarkers showed good accuracy in predicting the presence of axial SpA in patients with IBD. CONCLUSION We demonstrated that in patients with IBD, SOST and anti-SOST-IgG might represent novel biomarkers to assess the presence of axial joint involvement. Moreover, the development of anti-SOST-IgG and the subsequent decrease of SOST serum levels could play a role in the pathogenesis of SpA/IBD.
Collapse
Affiliation(s)
- Michele Maria Luchetti
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy.
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica.
| | - Francesco Ciccia
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Chiara Avellini
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Devis Benfaremo
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Giuliana Guggino
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Alessia Farinelli
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Monia Ciferri
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Matteo Rossini
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Silvia Svegliati
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Tatiana Spadoni
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Laura Bolognini
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Giammarco Fava
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Piergiorgio Mosca
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Rosaria Gesuita
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Edlira Skrami
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Giovanni Triolo
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| | - Armando Gabrielli
- From the Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G.Salesi," Ancona; Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo, Palermo, Italy
- M.M. Luchetti, MD, Assistant Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; F. Ciccia, MD, Assistant Professor, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; C. Avellini, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; D. Benfaremo, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; G. Guggino, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; A. Farinelli, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Ciferri, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; M. Rossini, MD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; S. Svegliati, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; T. Spadoni, PhD, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica; L. Bolognini, MD, Dipartimento Gastroenterologico e dei Trapianti, Polo Didattico Ospedaliero "Umberto I-G.M. Lancisi-G. Salesi;" G. Fava, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; P. Mosca, MD, Dipartimento Biomedico di Medicina Interna e Specialistica, Sezione di Reumatologia, Università degli Studi di Palermo; R. Gesuita, MD, Associate Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; E. Skrami, MD, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; G. Triolo, MD, Full Professor, Centro di Epidemiologia e Biostatistica, Università Politecnica delle Marche; A. Gabrielli, MD, Full Professor, Dipartimento Scienze Cliniche e Molecolari, Clinica Medica
| |
Collapse
|
38
|
Ma Y, Zhang X, Wang M, Xia Q, Yang J, Wu M, Han R, Chen M, Hu X, Yuan Y, Liu R, Jiang G, Pan G, Zou Y, Xu S, Pan F. The serum level of Dickkopf-1 in patients with rheumatoid arthritis: A systematic review and meta-analysis. Int Immunopharmacol 2018; 59:227-232. [PMID: 29665496 DOI: 10.1016/j.intimp.2018.04.019] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/10/2018] [Accepted: 04/10/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Dickkopf-1 (DKK-1) is an endogenous inhibitor of canonical Wnt pathway that was implicated in the pathogenesis of rheumatoid arthritis (RA), but the serum levels of DKK-1 in RA were inconsistent among studies. Therefore, we conducted a meta-analysis to systematically evaluate the relationship between serum DKK-1 levels and RA. METHODS PubMed, Web of Science and Cochrane Library were comprehensively retrieved till 1 January 2018 for pertinent studies. The pooled standard mean differences (SMDs) of serum DKK-1 levels were calculated according to the random effects model. RESULTS Nine original studies containing 1305 RA patients and 504 healthy controls were included in the meta-analysis. The pooled SMD of serum DKK-1 between RA patients and healthy controls was 0.79 (95% CI = 0.11 to 1.48, Z = 2.28 and P = 0.023), indicating a significantly higher serum level of DKK-1 in RA patients. CONCLUSION Serum level of DKK-1 is elevated in patients with RA compared to healthy controls, suggesting an important role of DKK-1 in the pathogenesis and treatment of RA.
Collapse
Affiliation(s)
- Yubo Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Xu Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Mengmeng Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Qing Xia
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Jiajia Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Meng Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Renfang Han
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Mengya Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Xingxing Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Yaping Yuan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Rui Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Guangming Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Guixia Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Yanfeng Zou
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China
| | - Shengqian Xu
- Department of Rheumatism and Immunity, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Faming Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China; The Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, 81 Meishan Road, Hefei, Anhui 230032, China.
| |
Collapse
|
39
|
Transcriptional signature associated with early rheumatoid arthritis and healthy individuals at high risk to develop the disease. PLoS One 2018; 13:e0194205. [PMID: 29584756 PMCID: PMC5870959 DOI: 10.1371/journal.pone.0194205] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 02/27/2018] [Indexed: 12/17/2022] Open
Abstract
Background Little is known regarding the mechanisms underlying the loss of tolerance in the early and preclinical stages of autoimmune diseases. The aim of this work was to identify the transcriptional profile and signaling pathways associated to non-treated early rheumatoid arthritis (RA) and subjects at high risk. Several biomarker candidates for early RA are proposed. Methods Whole blood total RNA was obtained from non-treated early RA patients with <1 year of evolution as well as from healthy first-degree relatives of patients with RA (FDR) classified as ACCP+ and ACCP- according to their antibodies serum levels against cyclic citrullinated peptides. Complementary RNA (cRNA) was synthetized and hybridized to high-density microarrays. Data was analyzed in Genespring Software and functional categories were assigned to a specific transcriptome identified in subjects with RA and FDR ACCP positive. Specific signaling pathways for genes associated to RA were identified. Gene expression was evaluated by qPCR. Receiver operating characteristic (ROC) analysis was used to evaluate these genes as biomarkers. Results A characteristic transcriptome of 551 induced genes and 4,402 repressed genes were identified in early RA patients. Bioinformatics analysis of the data identified a specific transcriptome in RA patients. Moreover, some overlapped transcriptional profiles between patients with RA and ACCP+ were identified, suggesting an up-regulated distinctive transcriptome from the preclinical stages up to progression to an early RA state. A total of 203 pathways have up-regulated genes that are shared between RA and ACCP+. Some of these genes show potential to be used as progression biomarkers for early RA with area under the curve of ROC > 0.92. These genes come from several functional categories associated to inflammation, Wnt signaling and type I interferon pathways. Conclusion The presence of a specific transcriptome in whole blood of RA patients suggests the activation of a specific inflammatory transcriptional signature in early RA development. The set of overexpressed genes in early RA patients that are shared with ACCP+ subjects but not with ACCP- subjects, can represent a transcriptional signature involved with the transition of a preclinical to a clinical RA stage. Some of these particular up-regulated and down-regulated genes are related to inflammatory processes and could be considered as biomarker candidates for disease progression in subjects at risk to develop RA.
Collapse
|
40
|
Early sclerostin expression explains bone formation inhibition before arthritis onset in the rat adjuvant-induced arthritis model. Sci Rep 2018; 8:3492. [PMID: 29472591 PMCID: PMC5823923 DOI: 10.1038/s41598-018-21886-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 02/13/2018] [Indexed: 12/20/2022] Open
Abstract
Periarticular bone loss in rheumatoid arthritis (RA) is considered to be mainly related to synovial inflammation. However, strong bone loss has also described at the time of arthritis onset. Recently, a paradoxical exacerbation of joint damage was described when blocking sclerostin in various arthritis models. Thus, we aimed to determine kinetics of bone loss and its mechanisms in the adjuvant induced arthritis (AIA) rat model of RA. AIA was induced (n = 35) or not (n = 35) at day 0. In addition to well-known arthritis at day 12, we showed with 3D-imaging and histomorphometry that bone microstructural alterations occurred early from day 8 post-induction, characterized by cortical porosity and trabecular bone loss. Active osteoclastic surfaces were increased from day 8 with RANKL upregulation. More surprisingly SOST and DKK1 were overexpressed from day 6 and followed by a dramatic decrease in bone formation from day 8. At the time of arthritis onset, SOST and DKK1 returned to control values, but frizzled related protein 1 (SFRP1), proinflammatory cytokines, and MMPs started to increase. Bone alterations before arthritis onset reinforce the hypothesis of an early bone involvement in arthritis. Kinetics of osteocyte markers expression should be considered to refine Wnt inhibitor treatment strategies.
Collapse
|
41
|
Dickkopf 1 protein circulating levels as a possible biomarker of functional disability and chronic damage in patients with rheumatoid arthritis. Clin Rheumatol 2017; 37:795-801. [PMID: 29282619 DOI: 10.1007/s10067-017-3957-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/09/2017] [Accepted: 12/14/2017] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease characterized by joint destruction, deformity, lower functionality, and decrease in life expectancy. Wingless signaling pathway (Wnt) has been recently involved in bone homeostasis. Studies suggest that overexpression of the pathway inhibitors, like the Dickkopf 1 protein (DKK1), has been implicated in bone destruction. The objective of this study is to compare circulating levels of DKK1 in different groups of patients with disease activity (remission, low, moderate, high activity,) and functionality status. Three hundred seventy-nine patients with RA were evaluated between March 2015 and November 2016. Disease activity was evaluated by disease activity score 28 with C-reactive protein (DAS28CPR), simplified and clinical disease activity scores (SDAI, CDAI), routine assessment of patient index data 3 (RAPID3), functional status using Multidimensional Health Assessment Questionnaire (MD-HAQ), and the Steinbrocker functional classification. DKK1 levels were measured by ELISA. The mean age was 60.7 ± 13.9 years. Disease duration was 13.2 ± 10.9 years. Higher levels of DKK1 were not associated with disease activity by CDAI (p = 0.70), SDAI (p = 0.84), DAS28CRP (p = 0.80), or RAPID3 (p = 0.70). Interestingly higher levels of DKK1 were significantly associated to lower functional status evaluating by the Steinbrocker classification (p = 0,013), severe disability by MD-HAQ (p = 0,004), and variables associated with joint destruction like osteoporosis, higher titles of rheumatoid factor, smoking, and increased hospital admissions related to RA. Higher levels of DKK1 were found in patients with lower functional status. This association was not found in patients with greater disease activity by CDAI, SDAI, DAS28, and RAPID3. This could be explained by more structural damage; DKK1 could be used as a biomarker of joint destruction in RA.
Collapse
|
42
|
Klavdianou K, Liossis SN, Daoussis D. Dkk1: A key molecule in joint remodelling and fibrosis. Mediterr J Rheumatol 2017; 28:174-182. [PMID: 32185280 PMCID: PMC7045998 DOI: 10.31138/mjr.28.4.174] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 07/20/2017] [Accepted: 08/25/2017] [Indexed: 12/17/2022] Open
Abstract
Dickkopf-1 (Dkk-1) is a Wnt signaling pathway inhibitor that has been shown to play an important role in joint remodeling, in experimental models of arthritis and in humans. Recent data suggest that this molecule is involved in the fibrotic process as well.
Collapse
Affiliation(s)
- Kalliopi Klavdianou
- Department of Rheumatology, University of Patras Medical School, Patras University Hospital, Patras, Greece
| | - Stamatis-Nick Liossis
- Department of Rheumatology, University of Patras Medical School, Patras University Hospital, Patras, Greece
| | - Dimitrios Daoussis
- Department of Rheumatology, University of Patras Medical School, Patras University Hospital, Patras, Greece
| |
Collapse
|
43
|
T-Cell Subsets in Rheumatoid Arthritis Patients on Long-Term Anti-TNF or IL-6 Receptor Blocker Therapy. Mediators Inflamm 2017; 2017:6894374. [PMID: 29209104 PMCID: PMC5676470 DOI: 10.1155/2017/6894374] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/22/2017] [Accepted: 07/19/2017] [Indexed: 12/28/2022] Open
Abstract
Data on the impact of biological therapies on the T-cell phenotype in rheumatoid arthritis are limited. Here, we prospectively measured the percentages of 15 circulating T-cell subtypes using flow cytometry. We obtained transversal and longitudinal data in 30 anti-TNF responders, 19 secondary anti-TNF nonresponders, and 43 IL-6R antagonist responders, before, 8 weeks and at least 6 months after biological therapy. Untreated RA patients and healthy controls were also included. The important findings are the following: (1) the proportion of regulatory T-cells (Tregs) which are decreased in untreated RA patients becomes normal in all long-term-treated groups; (2) in anti-TNF responders as well as in nonresponders, the frequencies of naïve CD4+ and CD8+ cells are lower, whereas those of proinflammatory Th1, Th2, and Th17 cells and HLA-DR+-activated cells are higher than those in untreated RA or healthy controls; (3) in IL-6R responders, Th1 proportion is decreased, while that of Th2 and Th17 is increased as compared to that in anti-TNF-treated patients and controls; (4) pending confirmation, a CD4CD69 ratio < 2.43 at baseline, could be useful to predict a good therapeutic response to anti-TNF therapy. This study provides comprehensive information regarding the long-term impacts of those biological therapies on the ecotaxis of T-cells in RA. The ClinicalTrials.gov registration number of our study is NCT03266822.
Collapse
|
44
|
Heinlen L, Humphrey MB. Skeletal complications of rheumatoid arthritis. Osteoporos Int 2017; 28:2801-2812. [PMID: 28779302 DOI: 10.1007/s00198-017-4170-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 07/20/2017] [Indexed: 12/27/2022]
Abstract
Rheumatoid arthritis (RA) is associated with local and systemic inflammation that induces many changes in the skeletal health. Locally, periarticular bone loss and juxta-articular bone erosions may occur while joint ankylosis, generalized bone loss, osteoporosis, and fractures may develop secondary to inflammation. The aim of this narrative review is to summarize the clinical evidence for abnormal skeletal health in RA, the effects of disease modifying anti-rheumatic drugs (DMARDS) on bone health, and the effects of drugs for the prevention or treatment of osteoporosis in the RA population.
Collapse
Affiliation(s)
- L Heinlen
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - M B Humphrey
- University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
45
|
Wehmeyer C, Pap T, Buckley CD, Naylor AJ. The role of stromal cells in inflammatory bone loss. Clin Exp Immunol 2017; 189:1-11. [PMID: 28419440 PMCID: PMC5461090 DOI: 10.1111/cei.12979] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2017] [Indexed: 12/26/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation, local and systemic bone loss and a lack of compensatory bone repair. Fibroblast-like synoviocytes (FLS) are the most abundant cells of the stroma and a key population in autoimmune diseases such as RA. An increasing body of evidence suggests that these cells play not only an important role in chronic inflammation and synovial hyperplasia, but also impact bone remodelling. Under inflammatory conditions FLS release inflammatory cytokines, regulate bone destruction and formation and communicate with immune cells to control bone homeostasis. Other stromal cells, such as osteoblasts and terminally differentiated osteoblasts, termed osteocytes, are also involved in the regulation of bone homeostasis and are dysregulated during inflammation. This review highlights our current understanding of how stromal cells influence the balance between bone formation and bone destruction. Increasing our understanding of these processes is critical to enable the development of novel therapeutic strategies with which to treat bone loss in RA.
Collapse
Affiliation(s)
- C. Wehmeyer
- Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth HospitalBirminghamUK
| | - T. Pap
- Institute of Experimental Musculoskeletal Medicine, University Hospital MuensterMuensterGermany
| | - C. D. Buckley
- Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth HospitalBirminghamUK
| | - A. J. Naylor
- Institute of Inflammation and Ageing (IIA), University of Birmingham, Queen Elizabeth HospitalBirminghamUK
| |
Collapse
|
46
|
Wang X, Ma C, Li P, Zhao F, Bi L. Effects of iguratimod on the levels of circulating regulators of bone remodeling and bone remodeling markers in patients with rheumatoid arthritis. Clin Rheumatol 2017; 36:1369-1377. [PMID: 28474138 DOI: 10.1007/s10067-017-3668-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/17/2017] [Accepted: 04/25/2017] [Indexed: 12/24/2022]
Abstract
This study aims to investigate the effect of iguratimod, a novel disease-modifying antirheumatic drug, alone or combined with methotrexate (MTX), on the serum levels of regulators of bone remodeling (receptor activator of nuclear factor-κB ligand (RANKL), osteoprotegerin (OPG), and Dickkopf-1 (DKK-1)) and bone remodeling markers (C-telopeptide of type I collagen (CTX-I) and procollagen type I N-terminal propeptide (PINP)) in patients with rheumatoid arthritis (RA). Patients with RA were treated with iguratimod, MTX, or their combination for 12 months. Serum samples were collected before treatment and 6 and 12 months afterwards. RANKL, OPG, DKK-1, CTX-I, and PINP levels were measured, and radiographic progression was assessed. The serum RANKL levels decreased after treatment for 6 and 12 months with iguratimod (median: baseline 565.00 pmol/L vs. 6 months 411.00 pmol/L vs. 12 months 212.00 pmol/L), MTX (median: baseline 562.50 pmol/L vs. 6 months 399.50 pmol/L vs. 12 months 163.50 pmol/L), and their combination (median: baseline 971.00 pmol/L vs. 6 months 272.50 pmol/L vs. 12 months 241.50 pmol/L). Combination therapy showed greater effects 6 months post-treatment compared to single-drug therapy. PINP levels increased significantly 12 months post-treatment with all therapies, but only the combination therapy led to decreased CTX-I levels. OPG and DKK-1 levels showed no significant changes. The three treatments showed no significant differences in radiographic progression. Iguratimod could stimulate bone formation and regulate the RANKL/RANK/OPG system rather than DKK-1levels. Its effects are comparable to those of MTX, and combination therapy showed stronger effects.
Collapse
Affiliation(s)
- Xuetong Wang
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, China
| | - Cuili Ma
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, China
| | - Ping Li
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, China
| | - Feng Zhao
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, China
| | - Liqi Bi
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, China.
| |
Collapse
|
47
|
Serum Sclerostin Levels in Patients with Ankylosing Spondylitis and Rheumatoid Arthritis: A Systematic Review and Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9295313. [PMID: 28553652 PMCID: PMC5434265 DOI: 10.1155/2017/9295313] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/23/2017] [Accepted: 04/11/2017] [Indexed: 12/17/2022]
Abstract
Objective. Current studies of serum sclerostin levels in AS and RA patients are inconsistent. This meta-analysis was performed to identify the association of serum sclerostin level with AS and RA patients. Methods. Embase, PubMed, MEDLINE, and Cochrane Library databases (up to 25 January 2017) were used to collect all relevant published articles. Studies were pooled and standard mean difference (SMD) with 95% confidence interval (CI) was calculated. All data analyses were performed using RevMan 5.3. Results. Totally eight studies of AS including 420 AS patients and 317 healthy controls (HC) and three studies of RA including 145 RA patients and 127 HC were finally included in this meta-analysis. The results revealed that the serum sclerostin levels in both AS patients (SMD = −0.14; 95% CI = [−0.39,0.11]; P = 0.28) and RA patients (SMD = −0.10; 95% CI = [−0.34,0.15]; P = 0.43) were not significantly different from those in HC. Conclusion. The difference of serum sclerostin levels in AS and RA patients was not significantly different from HC, indicating that the sclerostin may not associate with the development of AS and RA.
Collapse
|
48
|
Jacobsen CM. Application of anti-Sclerostin therapy in non-osteoporosis disease models. Bone 2017; 96:18-23. [PMID: 27780792 PMCID: PMC5328800 DOI: 10.1016/j.bone.2016.10.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/17/2016] [Accepted: 10/20/2016] [Indexed: 12/29/2022]
Abstract
Sclerostin, a known inhibitor of the low density lipoprotein related protein 5 and 6 (LRP5 and LRP6) cell surface signaling receptors, is integral in the maintenance of normal bone mass and strength. Patients with loss of function mutations in SOST or missense mutations in LRP5 that prevent Sclerostin from binding and inhibiting the receptor, have significantly increased bone mass. This observation leads to the development of Sclerostin neutralizing therapies to increase bone mass and strength. Anti-Sclerostin therapy has been shown to be effective at increasing bone density and strength in animal models and patients with osteoporosis. Loss of function of Sost or treatment with a Sclerostin neutralizing antibody improves bone properties in animal models of Osteoporosis Pseudoglioma syndrome (OPPG), likely due to action through the LRP6 receptor, which suggests patients may benefit from these therapies. Sclerostin antibody is effective at improving bone properties in mouse models of Osteogenesis Imperfecta, a genetic disorder of low bone mass and fragility due to type I collagen mutations, in as little as two weeks after initiation of therapy. However, these improvements are due to increases in bone quantity as the quality (brittleness) of bone remains unaffected. Similarly, Sclerostin antibody treatment improves bone density in animal models of other diseases. Sclerostin neutralizing therapies are likely to benefit many patients with genetic disorders of bone, as well as other forms of metabolic bone disease.
Collapse
Affiliation(s)
- Christina M Jacobsen
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, Boston Children's Hospital, Boston, MA, United States; Division of Endocrinology, Boston Children's Hospital, Boston, MA, United States; Division of Genetics, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
49
|
Boman A, Kokkonen H, Ärlestig L, Berglin E, Rantapää-Dahlqvist S. Receptor activator of nuclear factor kappa-B ligand (RANKL) but not sclerostin or gene polymorphisms is related to joint destruction in early rheumatoid arthritis. Clin Rheumatol 2017; 36:1005-1012. [PMID: 28190118 PMCID: PMC5400786 DOI: 10.1007/s10067-017-3570-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 01/22/2017] [Accepted: 01/31/2017] [Indexed: 01/25/2023]
Abstract
The aim of this study was to analyze relationships between receptor activator of nuclear factor kappa-B (RANKL), sclerostin and their gene polymorphisms with radiological progression in patients with early rheumatoid arthritis (RA). Patients with early RA (n = 407, symptomatic <1 year) (ARA criteria) examined radiologically at inclusion and after 24 months were consecutively included. Disease activity score and C-reactive protein were regularly recorded. Sclerostin, RANKL, and anti-CCP2 antibodies were analyzed in plasma at baseline using ELISAs. Data on gene polymorphism for sclerostin and RANKL were extracted from Immunochip analysis. Sex- and age-matched controls (n = 71) were identified from the Medical Biobank of Northern Sweden. The concentration of RANKL was significantly higher in patients compared with controls, median (IQR) 0.56 (0.9) nmol/L and 0.20 (0.25) nmol/L (p < 0.001), and in anti-CCP2-positive patients compared with sero-negative individuals. Sclerostin was significantly increased in female patients 0.59 (0.47–0.65) ng/mL compared with female controls 0.49 (0.4–0.65) ng/mL (p < 0.02). RANKL concentration was related to the Larsen score at baseline (p < 0.01), after 24 months (p < 0.001), and to radiological progression at 24 months (p < 0.001). Positivity of RANKL and anti-CCP2 yielded significant risk for progression with negativity for both as reference. No single nucleotide polymorphism encoding TNFSF11 or SOST was associated with increased concentrations of the factors. The concentration of RANKL was related to the Larsen score at baseline, at 24 months, and radiological progression at 24 months particularly in anti-CCP2-positive patients, while the concentration of sclerostin was unrelated to radiological findings.
Collapse
Affiliation(s)
- Antonia Boman
- Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, SE-901 85, Umeå, Sweden
| | - Heidi Kokkonen
- Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, SE-901 85, Umeå, Sweden
| | - Lisbeth Ärlestig
- Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, SE-901 85, Umeå, Sweden
| | - Ewa Berglin
- Department of Public Health and Clinical Medicine/Rheumatology, Umeå University, SE-901 85, Umeå, Sweden
| | | |
Collapse
|
50
|
Courbon G, Cleret D, Linossier MT, Vico L, Marotte H. Early Subchondral Bone Loss at Arthritis Onset Predicted Late Arthritis Severity in a Rat Arthritis Model. J Cell Physiol 2017; 232:1318-1325. [DOI: 10.1002/jcp.25601] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/14/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Guillaume Courbon
- SAINBIOSE, INSERM U1059, LBTO; Saint-Etienne France
- University of Lyon; Saint-Etienne France
| | - Damien Cleret
- SAINBIOSE, INSERM U1059, LBTO; Saint-Etienne France
- University of Lyon; Saint-Etienne France
| | | | - Laurence Vico
- SAINBIOSE, INSERM U1059, LBTO; Saint-Etienne France
- University of Lyon; Saint-Etienne France
| | - Hubert Marotte
- SAINBIOSE, INSERM U1059, LBTO; Saint-Etienne France
- University of Lyon; Saint-Etienne France
- Rheumatology Department; University Hospital of Saint-Etienne; Saint-Etienne France
| |
Collapse
|