1
|
Hammam N, El-Husseiny PN, Al-Adle SS, Samy N, Elsaid NY, El-Essawi DF, Mohamed EF, Fawzy SM, El Bakry SA, Nassr M, Nasef SI, El-Saadany HM, Elwan S, Gamal NM, Moshrif A, Hammam O, El Shereef RR, Ismail F, Tharwat S, Mosa DM, Elazeem MIA, Abdelaleem EA, Gheita TA. Clinical implications of seropositive and seronegative autoantibody status in rheumatoid arthritis patients: A comparative multicentre observational study. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2024; 5:57-65. [PMID: 38571932 PMCID: PMC10985701 DOI: 10.1515/rir-2024-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 11/20/2023] [Indexed: 04/05/2024]
Abstract
Background and Objectives Rheumatoid factor (RF) and anti-cyclic citrullinated protein (anti-CCP) have been used to improve the diagnosis and prognosis of rheumatoid arthritis (RA). However, their association with RA disease phenotypes, individually and in combination, is not well studied. The aim of the study was to compare patients' and disease characteristics, activity and severity in double seronegative (DNRA), single seropositive RF, single seropositive anti-CCP and double seropositive (DPRA) patients. Methods Adults subjects with RA from Egyptian College of Rheumatology (ECR) database who had RF and anti-CCP results available were included. Demographic, clinical features, disease activity score 28 (DAS28), Health Assessment Questionnaire (HAQ) and laboratory data were collected and compared among different RA groups. Results 5268 RA patients with mean age of 44.9±11.6 years, and 4477 (85%) were females. 2900 (55%) had DPRA, 892 (16.9%) had single positive RF, 597 (11.3%) had single positive anti-CCP while 879 (16.7%) had DNRA. Patients with DPRA had significantly high percentage of metabolic syndrome (19.3%, P < 0.001), and functional impairment using HAQ (P = 0.01). Older age (RRR [relative risk ratio]: 1.03, 95%CI: 1.0, 1.0, P = 0.029), greater DAS28 (RRR: 1.51, 95%CI: 1.2, 1.9, P < 0.001), higher steroid use (RRR: 2.4, 95%CI: 1.36, 4.25, P = 0.002) were at higher risk of DPRA while longer disease duration (RRR: 1.08, 95%CI: 1.01, 1.16, P = 0.017) and fibromyalgia syndrome (RRR: 2.54, 95%CI: 1.10, 5.88, P = 0.028) were associated with higher odds of single positive RF status. Conclusion Dual antibody-positive status has higher disease activity and severity, and higher chance of development of metabolic syndrome; highlighting the implicated role of inflammation, atherogenesis and cardiovascular disease risk in RA.
Collapse
Affiliation(s)
- Nevin Hammam
- Rheumatology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | | | - Suzan S. Al-Adle
- Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nermeen Samy
- Internal Medicine Department, Rheumatology Unit, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Nora Y. Elsaid
- Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Dina F. El-Essawi
- Health Radiation Research Department, Internal Medicine Unit, Rheumatology Clinic, (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Eman F. Mohamed
- Internal Medicine Department, Rheumatology Unit, Faculty of Medicine (Girls), Al-Azhar University, Cairo, Egypt
| | - Samar M. Fawzy
- Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Samah A. El Bakry
- Internal Medicine Department, Rheumatology Unit, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
| | - Maha Nassr
- Rheumatology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Samah I. Nasef
- Rheumatology Department, Faculty of Medicine, Suez-Canal University, Ismailia, Egypt
| | - Hanan M. El-Saadany
- Rheumatology Department, Faculty of Medicine, Tanta University, Gharbia, Egypt
| | - Shereen Elwan
- Rheumatology Department, Faculty of Medicine, Tanta University, Gharbia, Egypt
| | - Nada M. Gamal
- Rheumatology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Abdelhfeez Moshrif
- Rheumatology Department, Faculty of Medicine, Al-Azhar University, Assiut, Egypt
| | - Osman Hammam
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, New Valley University, New Valley, Egypt
| | | | - Faten Ismail
- Rheumatology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - Samar Tharwat
- Internal Medicine Department, Rheumatology Unit, Faculty of Medicine, Mansoura University, Dakahlia, Egypt
| | - Doaa Mosad Mosa
- Department of Rheumatology and Rehabilitation, Mansoura University Hospitals, Mansoura University Faculty of Medicine, Mansoura, Egypt
| | - Mervat I Abd Elazeem
- Rheumatology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Enas A. Abdelaleem
- Rheumatology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Tamer A. Gheita
- Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - on behalf of The Egyptian College of Rheumatology (ECR) Rheumatoid Arthritis (ECR-RA) Study Group
- Rheumatology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
- Rheumatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
- Internal Medicine Department, Rheumatology Unit, Faculty of Medicine, Ain-Shams University, Cairo, Egypt
- Health Radiation Research Department, Internal Medicine Unit, Rheumatology Clinic, (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
- Internal Medicine Department, Rheumatology Unit, Faculty of Medicine (Girls), Al-Azhar University, Cairo, Egypt
- Rheumatology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
- Rheumatology Department, Faculty of Medicine, Suez-Canal University, Ismailia, Egypt
- Rheumatology Department, Faculty of Medicine, Tanta University, Gharbia, Egypt
- Rheumatology Department, Faculty of Medicine, Al-Azhar University, Assiut, Egypt
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, New Valley University, New Valley, Egypt
- Rheumatology Department, Faculty of Medicine, Minia University, Minia, Egypt
- Internal Medicine Department, Rheumatology Unit, Faculty of Medicine, Mansoura University, Dakahlia, Egypt
- Department of Rheumatology and Rehabilitation, Mansoura University Hospitals, Mansoura University Faculty of Medicine, Mansoura, Egypt
- Rheumatology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
2
|
Kim AR, Ji E, Lee S, Hong S, Kim DH, Song JM, Kang DH, Song JK. Association of citrullination with the progression of aortic stenosis. Sci Rep 2023; 13:8919. [PMID: 37264066 DOI: 10.1038/s41598-023-36153-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 05/30/2023] [Indexed: 06/03/2023] Open
Abstract
Despite its clinical importance, biomarkers of disease activity in aortic stenosis (AS) are lacking. We investigated the association between anti-cyclic citrullinated peptide (CCP) antibodies and AS. All 678 patients who underwent echocardiography and anti-CCP antibody testing were analysed. Anti-CCP antibody status was categorized as negative, low-positive, and high-positive. In addition, aortic valve (AV) tissues were obtained from the patients with and without AS to analyze the presence of citrullinated proteins. At baseline, 241 (35.5%) subjects with AV degeneration had a higher rate of anti-CCP antibody positivity (42.7% versus 34.6%, p = 0.035) than those without AV degeneration. Out of the 331 (48.8%) subjects who underwent echocardiographic follow-up, progression of AS was observed in 34 (10.3%) patients, with a higher incidence in the high-positive group compared to the low-positive or negative group (19.0% vs. 11.3% vs. 8.4%, respectively; p = 0.041). On multivariable analysis, high anti-CCP antibody positivity was independently associated with progression to AS (odds ratio: 2.312; 95% confidence interval: 1.006-5.310; p = 0.048). Furthermore, immunohistochemistry and Western blotting revealed increased citrullination in diseased AV compared to normal AV tissue. This study demonstrated that a high positive anti-CCP antibody result is associated with AV degeneration and may be an independent factor for AS progression.
Collapse
Affiliation(s)
- Ah-Ram Kim
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eunhye Ji
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43 Gil, Songpa-gu, Seoul, 05505, Korea
| | - Sahmin Lee
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43 Gil, Songpa-gu, Seoul, 05505, Korea.
| | - Seokchan Hong
- Division of Rheumatology, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dae-Hee Kim
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jong-Min Song
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Duk-Hyun Kang
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae-Kwan Song
- Division of Cardiology, Heart Institute, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
3
|
Identification of essential genes and immune cell infiltration in rheumatoid arthritis by bioinformatics analysis. Sci Rep 2023; 13:2032. [PMID: 36739468 PMCID: PMC9899220 DOI: 10.1038/s41598-023-29153-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease that can lead to severe joint damage and disability. And early diagnosis and treatment of RA can avert or substantially slow the progression of joint damage in up to 90% of patients, thereby preventing irreversible disability. Previous research indicated that 50% of the risk for the development of RA is attributable to genetic factors, but the pathogenesis is not well understood. Thus, it is urgent to identify biomarkers to arrest RA before joints are irreversibly damaged. Here, we first use the Robust Rank Aggregation method (RRA) to identify the differentially expressed genes (DEGs) between RA and normal samples by integrating four public RA patients' mRNA expression data. Subsequently, these DEGs were used as the input for the weighted gene co-expression network analysis (WGCNA) approach to identify RA-related modules. The function enrichment analysis suggested that the RA-related modules were significantly enriched in immune-related actions. Then the hub genes were defined as the candidate genes. Our analysis showed that the expression levels of candidate genes were significantly associated with the RA immune microenvironment. And the results indicated that the expression of the candidate genes can use as predictors for RA. We hope that our method can provide a more convenient approach for the early diagnosis of RA.
Collapse
|
4
|
Romão VC, Fonseca JE. Disease mechanisms in preclinical rheumatoid arthritis: A narrative review. Front Med (Lausanne) 2022; 9:689711. [PMID: 36059838 PMCID: PMC9437632 DOI: 10.3389/fmed.2022.689711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/04/2022] [Indexed: 11/20/2022] Open
Abstract
In the last decades, the concept of preclinical rheumatoid arthritis (RA) has become established. In fact, the discovery that disease mechanisms start years before the onset of clinical RA has been one of the major recent insights in the understanding of RA pathogenesis. In accordance with the complex nature of the disease, preclinical events extend over several sequential phases. In a genetically predisposed host, environmental factors will further increase susceptibility for incident RA. In the initial steps of preclinical disease, immune disturbance mechanisms take place outside the joint compartment, namely in mucosal surfaces, such as the lung, gums or gut. Herein, the persistent immunologic response to altered antigens will lead to breach of tolerance and trigger autoimmunity. In a second phase, the immune response matures and is amplified at a systemic level, with epitope spreading and widening of the autoantibody repertoire. Finally, the synovial and bone compartment are targeted by specific autoantibodies against modified antigens, initiating a local inflammatory response that will eventually culminate in clinically evident synovitis. In this review, we discuss the elaborate disease mechanisms in place during preclinical RA, providing a broad perspective in the light of current evidence.
Collapse
Affiliation(s)
- Vasco C. Romão
- Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon Academic Medical Centre and European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ERN-ReCONNET), Lisbon, Portugal
- Rheumatology Research Unit, Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| | - João Eurico Fonseca
- Rheumatology Research Unit, Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
5
|
Shi L, Zhao Y, Feng C, Miao F, Dong L, Wang T, Stalin A, Zhang J, Tu J, Liu K, Sun W, Wu J. Therapeutic effects of shaogan fuzi decoction in rheumatoid arthritis: Network pharmacology and experimental validation. Front Pharmacol 2022; 13:967164. [PMID: 36059943 PMCID: PMC9428562 DOI: 10.3389/fphar.2022.967164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Shaogan Fuzi Decoction (SGFD), one of the classical prescriptions of Chinese Medicine, has a long history in the treatment of rheumatoid arthritis (RA), but definitive studies on its efficacy and mechanism of action are lacking. This study aims to elucidate the pharmacodynamic role of SGFD against RA and the potential mechanisms based on a combination of network pharmacology and experimental verification. The RA model in rats was induced by intradermal injection of bovine type Ⅱ collagen and incomplete Freund’s adjuvant at the tail root. SGFD was administered once a day by oral gavage for 4 weeks. After SGFD administration, rat’s arthritis index (AI) score and paw swelling decreased to some extent, and synovial inflammation, vascular hyperplasia, and cartilage destruction of the ankle joint were improved. Simultaneously, thymus and spleen index and serum levels of C-reactive protein (CRP) were lowered. Network pharmacology revealed that quercetin, kaempferol, naringenin, formononetin isorhamnetin and licochalcone A were the potentialiy active components, and IL6, TP53, TNF, PTGS2, MAPK3 and IL-1β were potential key targets for SGFD in the treatment of RA. Ingredients-targets molecular docking showed that the components had the high binding activity to these target proteins. The mechanism of SGFD for RA involves various biological functions and is closely correlated with TNF signaling pathway, Osteoclast differentiation, T cell receptor signaling pathway, mitogen-activated protein kinase (MAPK) signaling pathway, NF-κB signaling pathway, toll-like receptor signaling pathway, and so on. Western blot and ELISA showed that the expression of toll-like receptor 4 (TLR4), nuclear factor kappa-B (NF-κB) p65, phosphorylated c-Jun N-terminal kinase (p-JNK), p-p38, phosphorylated extracellular regulated kinase (p-ERK) and TNF-α was significantly upregulated in the synovium of RA rats, and the levels of serum inflammatory factors were significantly increased. SGFD inhibits the activation of the TLR4/NF-κB/MAPK pathway and the expression/production of pro-inflammatory cytokines. In summary, SGFD could improve the symptoms and inflammatory response in collagen-induced arthritis (CIA) rat model. The mechanism might be related to the regulation of TLR4/MAPKs/NF-κB signaling pathway and the reduction of inflammatory factor release, which partially confirms the results predicted by network pharmacology.
Collapse
Affiliation(s)
- Lu Shi
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yiying Zhao
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chenran Feng
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Feng Miao
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Linlin Dong
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Tianquan Wang
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingru Tu
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Kexin Liu
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wenyan Sun
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Wenyan Sun, ; Jiarui Wu,
| | - Jiarui Wu
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Moura RA, Fonseca JE. B Cells on the Stage of Inflammation in Juvenile Idiopathic Arthritis: Leading or Supporting Actors in Disease Pathogenesis? Front Med (Lausanne) 2022; 9:851532. [PMID: 35449805 PMCID: PMC9017649 DOI: 10.3389/fmed.2022.851532] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Juvenile idiopathic arthritis (JIA) is a term that collectively refers to a group of chronic childhood arthritides, which together constitute the most common rheumatic condition in children. The International League of Associations for Rheumatology (ILAR) criteria define seven categories of JIA: oligoarticular, polyarticular rheumatoid factor (RF) negative (RF-), polyarticular RF positive (RF+), systemic, enthesitis-related arthritis, psoriatic arthritis, and undifferentiated arthritis. The ILAR classification includes persistent and extended oligoarthritis as subcategories of oligoarticular JIA, but not as distinct categories. JIA is characterized by a chronic inflammatory process affecting the synovia that begins before the age of 16 and persists at least 6 weeks. If not treated, JIA can cause significant disability and loss of quality of life. Treatment of JIA is adjusted according to the severity of the disease as combinations of non-steroidal anti-inflammatory drugs (NSAIDs), synthetic and/ or biological disease modifying anti-rheumatic drugs (DMARDs). Although the disease etiology is unknown, disturbances in innate and adaptive immune responses have been implicated in JIA development. B cells may have important roles in JIA pathogenesis through autoantibody production, antigen presentation, cytokine release and/ or T cell activation. The study of B cells has not been extensively explored in JIA, but evidence from the literature suggests that B cells might have indeed a relevant role in JIA pathophysiology. The detection of autoantibodies such as antinuclear antibodies (ANA), RF and anti-citrullinated protein antibodies (ACPA) in JIA patients supports a breakdown in B cell tolerance. Furthermore, alterations in B cell subpopulations have been documented in peripheral blood and synovial fluid from JIA patients. In fact, altered B cell homeostasis, B cell differentiation and B cell hyperactivity have been described in JIA. Of note, B cell depletion therapy with rituximab has been shown to be an effective and well-tolerated treatment in children with JIA, which further supports B cell intervention in disease development.
Collapse
Affiliation(s)
- Rita A Moura
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João Eurico Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon Academic Medical Centre, Lisbon, Portugal
| |
Collapse
|
7
|
Tang X, Liu Z, Yang Z, Xu S, Wang M, Chen X, Wen Z, Huang R. The Effect of Chinese Medicine Compound in the Treatment of Rheumatoid Arthritis on the Level of Rheumatoid Factor and Anti-Cyclic Citrullinated Peptide Antibodies: A Systematic Review and Meta-Analysis. Front Pharmacol 2021; 12:686360. [PMID: 34276376 PMCID: PMC8278104 DOI: 10.3389/fphar.2021.686360] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/17/2021] [Indexed: 01/19/2023] Open
Abstract
Objectives: To evaluate the current evidence whether Chinese medicine compound (CMC) can reduce the serum levels of rheumatoid factor (RF) and anti-cyclic citrullinated peptide antibodies (anti-CCP). Methods: We comprehensively searched PubMed, Embase, the Cochrane Library, China National Knowledge Infrastructure (CNKI), the Database for Chinese Technical Periodicals (VIP), and Wanfang data. We then performed a systematic review and meta-analysis of all randomized controlled trials (RCTs) assessing the CMC therapy methods. This study is registered with PROSPERO, number CRD42020216284. Results: In total, 65 studies were eligible for inclusion, including 6099 patients. The result of the meta-analysis showed that compared with common Western medicine therapy, CMC monotherapy or combined with Western medicine was able to reduce serum RF (SMD= -0.85, 95%CI -1.04 to -0.67) and anti-CCP (SMD= -0.56, 95%CI -0.79 to -0.32) levels to some extent. In the efficacy meta-analysis, a greater number of CMC-treated patients achieved the efficacy criteria after a period of treatment, where the relative risk (RR) was 1.20 [1.08, 1.33] for achieving ACR20, 1.57 [1.38, 1.78] for ACR50, and 2.21 [1.72, 2.84] for ACR70. At the same time, there was a statistically significant difference in the effective rate of the patient's TCM symptoms (RR = 1.22, 95%CI 1.19-1.26). Conclusions: Through this meta-analysis and systematic review, we found that CMC for the treatment of RA is effective in reducing RF and anti-CCP levels and might have better clinical efficacy than Western medicine monotherapy. Some active components are responsible for this efficacy and worth further exploring.
Collapse
Affiliation(s)
- Xuan Tang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Zehao Liu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Zhihua Yang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Shengmei Xu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| | - Maojie Wang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Xiumin Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zehuai Wen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
| | - Runyue Huang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
8
|
Hernández-Breijo B, Plasencia-Rodríguez C, Navarro-Compán V, García-Hoz C, Nieto-Gañán I, Sobrino C, Bachiller-Corral J, Díaz-Almirón M, Martínez-Feito A, Jurado T, Lapuente-Suanzes P, Bonilla G, Pijoán-Moratalla C, Roy G, Vázquez-Díaz M, Balsa A, Villar LM, Pascual-Salcedo D, Rodríguez-Martín E. Remission Induced by TNF Inhibitors Plus Methotrexate is Associated With Changes in Peripheral Naïve B Cells in Patients With Rheumatoid Arthritis. Front Med (Lausanne) 2021; 8:683990. [PMID: 34222289 PMCID: PMC8245775 DOI: 10.3389/fmed.2021.683990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/24/2021] [Indexed: 12/30/2022] Open
Abstract
Biological therapies, such as TNF inhibitors (TNFi), are increasing remission (REM) rates in rheumatoid arthritis (RA) patients, although these are still limited. The aim of our study was to analyze changes in the profile of peripheral blood mononuclear cells (PBMC) in patients with RA treated with TNFi in relation to the clinical response. This is a prospective and observational study including 78 RA patients starting the first TNFi. PBMC were analyzed by flow cytometry both at baseline and at 6 months. Disease activity at the same time points was assessed by DAS28, establishing DAS28 ≤ 2.6 as the criteria for REM. Logistic regression models were employed to analyze the association between the changes in PBMC and REM. After 6 months of TNFi treatment, 37% patients achieved REM by DAS28. Patients who achieved REM showed a reduction in the percentage of naive B cells, but only when patients had received concomitant methotrexate (MTX) (OR: 0.59; 95% CI: 0.39–0.91). However, no association was found for patients who did not receive concomitant MTX (OR: 0.85; 95% CI: 0.63–1.16). In conclusion, PBMC, mainly the B-cell subsets, are modified in RA patients with TNFi who achieve clinical REM. A significant decrease in naive B-cell percentage is associated with achieving REM after 6 months of TNFi treatment in patients who received concomitant therapy with MTX.
Collapse
Affiliation(s)
- Borja Hernández-Breijo
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain
| | - Chamaida Plasencia-Rodríguez
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain.,Rheumatology, La Paz University Hospital, Madrid, Spain
| | - Victoria Navarro-Compán
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain.,Rheumatology, La Paz University Hospital, Madrid, Spain
| | - Carlota García-Hoz
- Immunology, Ramón y Cajal Institute for Health Research, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Israel Nieto-Gañán
- Immunology, Ramón y Cajal Institute for Health Research, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Cristina Sobrino
- Rheumatology, Ramón y Cajal Institute for Health Research, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Javier Bachiller-Corral
- Rheumatology, Ramón y Cajal Institute for Health Research, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Mariana Díaz-Almirón
- Biostatistics Unit, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain
| | - Ana Martínez-Feito
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain.,Immunology, La Paz University Hospital, Madrid, Spain
| | - Teresa Jurado
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain
| | - Paloma Lapuente-Suanzes
- Immunology, Ramón y Cajal Institute for Health Research, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Gema Bonilla
- Rheumatology, La Paz University Hospital, Madrid, Spain
| | - Cristina Pijoán-Moratalla
- Rheumatology, Ramón y Cajal Institute for Health Research, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Garbiñe Roy
- Immunology, Ramón y Cajal Institute for Health Research, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Mónica Vázquez-Díaz
- Rheumatology, Ramón y Cajal Institute for Health Research, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alejandro Balsa
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain.,Rheumatology, La Paz University Hospital, Madrid, Spain
| | - Luisa M Villar
- Immunology, Ramón y Cajal Institute for Health Research, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Dora Pascual-Salcedo
- Immuno-Rheumatology Research Group, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain
| | - Eulalia Rodríguez-Martín
- Immunology, Ramón y Cajal Institute for Health Research, Hospital Universitario Ramón y Cajal, Madrid, Spain
| |
Collapse
|
9
|
Moura RA, Fonseca JE. JAK Inhibitors and Modulation of B Cell Immune Responses in Rheumatoid Arthritis. Front Med (Lausanne) 2021; 7:607725. [PMID: 33614673 PMCID: PMC7892604 DOI: 10.3389/fmed.2020.607725] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic immune-mediated inflammatory disease that can lead to joint destruction, functional disability and substantial comorbidity due to the involvement of multiple organs and systems. B cells have several important roles in RA pathogenesis, namely through autoantibody production, antigen presentation, T cell activation, cytokine release and ectopic lymphoid neogenesis. The success of B cell depletion therapy with rituximab, a monoclonal antibody directed against CD20 expressed by B cells, has further supported B cell intervention in RA development. Despite the efficacy of synthetic and biologic disease modifying anti-rheumatic drugs (DMARDs) in the treatment of RA, few patients reach sustained remission and refractory disease is a concern that needs critical evaluation and close monitoring. Janus kinase (JAK) inhibitors or JAKi are a new class of oral medications recently approved for the treatment of RA. JAK inhibitors suppress the activity of one or more of the JAK family of tyrosine kinases, thus interfering with the JAK-Signal Transducer and Activator of Transcription (STAT) signaling pathway. To date, there are five JAK inhibitors (tofacitinib, baricitinib, upadacitinib, peficitinib and filgotinib) approved in the USA, Europe and/ or Japan for RA treatment. Evidence from the literature indicates that JAK inhibitors interfere with B cell functions. In this review, the main results obtained in clinical trials, pharmacokinetic, in vitro and in vivo studies concerning the effects of JAK inhibitors on B cell immune responses in RA are summarized.
Collapse
Affiliation(s)
- Rita A Moura
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - João Eurico Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Rheumatology Department, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte (CHULN), Lisbon Academic Medical Centre, Lisbon, Portugal
| |
Collapse
|
10
|
Tsujimura S, Tanaka Y. Potential of B-cell-targeting therapy in overcoming multidrug resistance and tissue invasiveness associated with P-glycoprotein expressing-B cell compartments. Immunol Med 2020; 44:142-151. [PMID: 33017281 DOI: 10.1080/25785826.2020.1825276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune mediated inflammatory disease characterized by progressive joint damage and extra-articular organ manifestations. Among the effector pathways and cells involved in the development of RA, activated B cells play a pivotal role in the pathological process of RA. P-glycoprotein (P-gp), a member of ATP-binding cassette transporters, is induced on the cell membrane by certain stimuli. P-gp transports various drugs from the cytoplasm to the cell exterior, resulting in the development of drug resistance. P-gp expression on B cells appears in patients with RA as the disease activity increases, and treatment of these patients' results in modification of over-expression of P-gp on activated B cells. Evidence suggests that P-gp expressing-activated B cells play important roles in the pathogenesis and treatment resistance in RA through the efflux of intracellular drugs and progression of infiltration in inflammatory lesions. Therapies designed to target activated B cells might overcome refractory RA. Identification of the subsets of peripheral activated B cells that express P-gp in RA patients might help the selection of suitable treatment strategy.
Collapse
Affiliation(s)
- Shizuyo Tsujimura
- The First Department of Internal Medicine, University of Occupational & Environmental Health, School of Medicine, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational & Environmental Health, School of Medicine, Kitakyushu, Japan
| |
Collapse
|
11
|
Rodríguez-Martín E, Nieto-Gañán I, Hernández-Breijo B, Sobrino C, García-Hoz C, Bachiller J, Martínez-Feito A, Navarro-Compán V, Lapuente-Suanzes P, Bonilla G, Pascual-Salcedo D, Roy G, Jurado T, Nozal P, Vázquez-Díaz M, Balsa A, Villar LM, Plasencia-Rodríguez C. Blood Lymphocyte Subsets for Early Identification of Non-Remission to TNF Inhibitors in Rheumatoid Arthritis. Front Immunol 2020; 11:1913. [PMID: 32973793 PMCID: PMC7481468 DOI: 10.3389/fimmu.2020.01913] [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: 02/24/2020] [Accepted: 07/16/2020] [Indexed: 12/21/2022] Open
Abstract
Background: TNF inhibitors (TNFis) are widely used for the treatment of rheumatoid arthritis (RA), although the response rates to this therapy in patients with RA remains heterogeneous and < 50% achieve remission (REM). Objective: To analyze baseline peripheral blood leukocytes profiles in order to search for biomarkers identifying patients who will most likely not achieve REM under TNFi treatment. Methods: A prospective bi-center pilot study including 98 RA patients treated with TNFis and followed-up during 6 months. Patients were classified according to DAS28 as follows: those who achieved REM (DAS28 ≤ 2.6) and those who did not (DAS28 > 2.6) at 6 months after starting TNFis. These rates were also assessed by simplified disease activity index (SDAI ≤ 3.3 and SDAI > 3.3, respectively). Peripheral blood immune cells were studied by flow cytometry before treatment initiation. Results: At 6 months, 61 or 80% of patients did not achieve REM by DAS28 or SDAI, respectively. Basal leukocyte profiles differed between REM vs. non-REM patients. Non-REM patients showed lower percentages of total and naïve B cells at baseline than REM subjects. A B lymphocyte/CD4+ lymphocyte ratio (BL/CD4 ratio) <0.2 clearly associated with a higher probability of non-REM status based on DAS28 at 6 months (OR = 9.2, p = 0.006). These data were confirmed when patient response was evaluated by SDAI index. Conclusion: Our results strongly suggest that BL/CD4 ratio could be considered as a useful biomarker for the early identification of non-remitters to TNFi in clinical practice.
Collapse
Affiliation(s)
| | - Israel Nieto-Gañán
- Department of Immunology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | - Cristina Sobrino
- Department of Rheumatology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Carlota García-Hoz
- Department of Immunology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Javier Bachiller
- Department of Rheumatology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Ana Martínez-Feito
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | | | | | - Gema Bonilla
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | - Dora Pascual-Salcedo
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | - Garbiñe Roy
- Department of Immunology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Teresa Jurado
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | - Pilar Nozal
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | - Mónica Vázquez-Díaz
- Department of Rheumatology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Alejandro Balsa
- Immuno-Rheumatology Research Group, IdiPaz Hospital Universitario La Paz, Madrid, Spain
| | - Luisa M Villar
- Department of Immunology, IRYCIS, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | |
Collapse
|
12
|
Autoantibodies as Diagnostic Markers and Mediator of Joint Inflammation in Arthritis. Mediators Inflamm 2019; 2019:6363086. [PMID: 31772505 PMCID: PMC6854956 DOI: 10.1155/2019/6363086] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/14/2019] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid arthritis is a systemic, polygenic, and multifactorial syndrome characterized by erosive polyarthritis, damage to joint architecture, and presence of autoantibodies against several self-structures in the serum and synovial fluid. These autoantibodies (anticitrullinated protein/peptide antibodies (ACPAs), rheumatoid factors (RF), anticollagen type II antibodies, antiglucose-6 phosphate isomerase antibodies, anticarbamylated protein antibodies, and antiacetylated protein antibodies) have different characteristics, diagnostic/prognostic value, and pathological significance in RA patients. Some of these antibodies are present in the patients' serum several years before the onset of clinical disease. Various genetic and environmental factors are associated with autoantibody production against different autoantigenic targets. Both the activating and inhibitory FcγRs and the activation of different complement cascades contribute to the downstream effector functions in the antibody-mediated disease pathology. Interplay between several molecules (cytokines, chemokines, proteases, and inflammatory mediators) culminates in causing damage to the articular cartilage and bones. In addition, autoantibodies are proven to be useful disease markers for RA, and different diagnostic tools are being developed for early diagnosis of the clinical disease. Recently, a direct link was proposed between the presence of autoantibodies and bone erosion as well as in the induction of pain. In this review, the diagnostic value of autoantibodies, their synthesis and function as a mediator of joint inflammation, and the significance of IgG-Fc glycosylation are discussed.
Collapse
|
13
|
de Brito Rocha S, Baldo DC, Andrade LEC. Clinical and pathophysiologic relevance of autoantibodies in rheumatoid arthritis. Adv Rheumatol 2019; 59:2. [PMID: 30657101 DOI: 10.1186/s42358-018-0042-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 11/29/2018] [Indexed: 12/20/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune/inflammatory disease affecting 0.5 to 1% of adults worldwide and frequently leads to joint destruction and disability. Early diagnosis and early and effective therapy may prevent joint damage and lead to better long-term results. Therefore, reliable biomarkers and outcome measures are needed. Refinement of the understanding of molecular pathways involved in disease pathogenesis have been achieved by combining knowledge on RA-associated genes, environmental factors and the presence of serological elements. The presence of autoantibodies is a distinctive feature of RA. Rheumatoid Factor and Anti-Citrullinated Protein Antibodies are the two most remarkable autoantibodies in RA and provide different clinical and pathophysiological information. They precede the onset of disease symptoms and predict a more severe disease course, indicating a pathogenetic role in RA. Therefore, they promote a more accurate prognosis and contribute for a better disease management. Several RA-associated autoantibody systems have been identified: Anti-Carbamylated Antibodies, Anti-BRAF, Anti-Acetylated, Anti-PAD4 antibodies and others. Hopefully, the characterization of a comprehensive array of novel autoantibody systems in RA will provide unique pathogenic insights of relevance for the development of diagnostic and prognostic approaches compatible with an effective personalized medicine.
Collapse
Affiliation(s)
- Sara de Brito Rocha
- Rheumatology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo, Disciplina de Reumatologia, Rua Botucatu 740, 3o andar, São Paulo, SP, ZIP:04023-062, Brazil.
| | - Danielle Cristiane Baldo
- Rheumatology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo, Disciplina de Reumatologia, Rua Botucatu 740, 3o andar, São Paulo, SP, ZIP:04023-062, Brazil
| | - Luis Eduardo Coelho Andrade
- Rheumatology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo, Disciplina de Reumatologia, Rua Botucatu 740, 3o andar, São Paulo, SP, ZIP:04023-062, Brazil
| |
Collapse
|
14
|
Role of CXCL13 and CCL20 in the recruitment of B cells to inflammatory foci in chronic arthritis. Arthritis Res Ther 2018; 20:114. [PMID: 29880013 PMCID: PMC5992813 DOI: 10.1186/s13075-018-1611-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/29/2018] [Indexed: 12/19/2022] Open
Abstract
Background B cells exert their pathogenic action in rheumatoid arthritis (RA) locally in the synovium. This study was undertaken to elucidate the chemokines responsible for the recruitment of B cells in the inflamed synovium, taking into account that the rich chemokine milieu present in the synovial tissue can fine-tune modulate discrete chemokine receptors. Methods Expression levels of chemokine receptors from the CC and CXC family, as well as CD27, were assessed by flow cytometry in CD20+ mononuclear cells isolated from the peripheral blood (PB) and synovial fluid (SF) of RA and psoriatic arthritis patients. Transwell experiments were used to study migration of B cells in response to a chemokine or in the presence of multiple chemokines. Results B cells from the SF of arthritis patients showed a significant increase in the surface expression of CCR1, CCR2, CCR4, CCR5 and CXCR4 with respect to PB. Conversely, SF B cells expressed consistently lower amounts of CXCR5, CXCR7 and CCR6, independent of CD27 expression. Analysis of permeabilized B cells suggested internalization of CXCR5 and CCR6 in SF B cells. In Transwell experiments, CCL20 and CXCL13, ligands of CCR6 and CXCR5, respectively, caused a significantly higher migration of B cells from PB than of those from SF of RA patients. Together, these two chemokines synergistically increased B-cell migration from PB, but not from SF. Conclusions These results suggest that CXCL13 and CCL20 might play major roles in RA pathogenesis by acting singly on their selective receptors and synergistically in the accumulation of B cells within the inflamed synovium. Electronic supplementary material The online version of this article (10.1186/s13075-018-1611-2) contains supplementary material, which is available to authorized users.
Collapse
|
15
|
Thalayasingam N, Nair N, Skelton AJ, Massey J, Anderson AE, Clark AD, Diboll J, Lendrem DW, Reynard LN, Cordell HJ, Eyre S, Isaacs JD, Barton A, Pratt AG. CD4+ and B Lymphocyte Expression Quantitative Traits at Rheumatoid Arthritis Risk Loci in Patients With Untreated Early Arthritis: Implications for Causal Gene Identification. Arthritis Rheumatol 2018; 70:361-370. [PMID: 29193869 PMCID: PMC5888199 DOI: 10.1002/art.40393] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 11/22/2017] [Indexed: 12/04/2022]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is a genetically complex disease of immune dysregulation. This study sought to gain further insight into the genetic risk mechanisms of RA by conducting an expression quantitative trait locus (eQTL) analysis of confirmed genetic risk loci in CD4+ T cells and B cells from carefully phenotyped patients with early arthritis who were naive to therapeutic immunomodulation. METHODS RNA and DNA were isolated from purified B and/or CD4+ T cells obtained from the peripheral blood of 344 patients with early arthritis. Genotyping and global gene expression measurements were carried out using Illumina BeadChip microarrays. Variants in linkage disequilibrium (LD) with non-HLA RA single-nucleotide polymorphisms (defined as r2 ≥ 0.8) were analyzed, seeking evidence of cis- or trans-eQTLs according to whether the associated probes were or were not within 4 Mb of these LD blocks. RESULTS Genes subject to cis-eQTL effects that were common to both CD4+ and B lymphocytes at RA risk loci were FADS1, FADS2, BLK, FCRL3, ORMDL3, PPIL3, and GSDMB. In contrast, those acting on METTL21B, JAZF1, IKZF3, and PADI4 were unique to CD4+ lymphocytes, with the latter candidate risk gene being identified for the first time in this cell subset. B lymphocyte-specific eQTLs for SYNGR1 and CD83 were also found. At the 8p23 BLK-FAM167A locus, adjacent genes were subject to eQTLs whose activity differed markedly between cell types; in particular, the FAM167A effect displayed striking B lymphocyte specificity. No trans-eQTLs approached experiment-wide significance, and linear modeling did not identify a significant influence of biologic covariates on cis-eQTL effect sizes. CONCLUSION These findings further refine the understanding of candidate causal genes in RA pathogenesis, thus providing an important platform from which downstream functional studies, directed toward particular cell types, may be prioritized.
Collapse
Affiliation(s)
- Nishanthi Thalayasingam
- NIHR Newcastle Biomedical Research CentreNewcastle upon Tyne Hospitals NHS Foundation Trust, and Newcastle UniversityNewcastle upon TyneUK
| | - Nisha Nair
- Arthritis Research UK Centre for Genetics and GenomicsCentre for Musculoskeletal ResearchInstitute of Inflammation and RepairUniversity of Manchesterand NIHR Manchester Musculoskeletal Biomedical Research UnitCentral Manchester NHS Foundation TrustManchesterUK
| | - Andrew J. Skelton
- NIHR Newcastle Biomedical Research CentreNewcastle upon Tyne Hospitals NHS Foundation Trust, and Newcastle UniversityNewcastle upon TyneUK
| | - Jonathan Massey
- Arthritis Research UK Centre for Genetics and GenomicsCentre for Musculoskeletal ResearchInstitute of Inflammation and RepairUniversity of Manchesterand NIHR Manchester Musculoskeletal Biomedical Research UnitCentral Manchester NHS Foundation TrustManchesterUK
| | - Amy E. Anderson
- NIHR Newcastle Biomedical Research CentreNewcastle upon Tyne Hospitals NHS Foundation Trust, and Newcastle UniversityNewcastle upon TyneUK
| | - Alexander D. Clark
- NIHR Newcastle Biomedical Research CentreNewcastle upon Tyne Hospitals NHS Foundation Trust, and Newcastle UniversityNewcastle upon TyneUK
| | - Julie Diboll
- NIHR Newcastle Biomedical Research CentreNewcastle upon Tyne Hospitals NHS Foundation Trust, and Newcastle UniversityNewcastle upon TyneUK
| | - Dennis W. Lendrem
- NIHR Newcastle Biomedical Research CentreNewcastle upon Tyne Hospitals NHS Foundation Trust, and Newcastle UniversityNewcastle upon TyneUK
| | - Louise N. Reynard
- NIHR Newcastle Biomedical Research CentreNewcastle upon Tyne Hospitals NHS Foundation Trust, and Newcastle UniversityNewcastle upon TyneUK
| | | | - Stephen Eyre
- Arthritis Research UK Centre for Genetics and GenomicsCentre for Musculoskeletal ResearchInstitute of Inflammation and RepairUniversity of Manchesterand NIHR Manchester Musculoskeletal Biomedical Research UnitCentral Manchester NHS Foundation TrustManchesterUK
| | - John D. Isaacs
- NIHR Newcastle Biomedical Research CentreNewcastle upon Tyne Hospitals NHS Foundation Trust, and Newcastle UniversityNewcastle upon TyneUK
| | - Anne Barton
- Arthritis Research UK Centre for Genetics and GenomicsCentre for Musculoskeletal ResearchInstitute of Inflammation and RepairUniversity of Manchesterand NIHR Manchester Musculoskeletal Biomedical Research UnitCentral Manchester NHS Foundation TrustManchesterUK
| | - Arthur G. Pratt
- NIHR Newcastle Biomedical Research CentreNewcastle upon Tyne Hospitals NHS Foundation Trust, and Newcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
16
|
Nasonov EL, Mazurov VI, Zonova EV, Knyazeva LA, Marusenko IM, Nesmeyanova OB, Plaksina TV, Shapovalova YS, Ilivanova EP, Krechikova DG, Petrochenkova NA, Reshetko OV, Denisov LN, Gordeev IG, Davydova AF, Eremina NA, Zemerova EV, Ivanova TB, Kastanayan AA, Pokrovskaya TG, Smakotina SA, Smolyarchuk EA, Artemyeva AV, Ivanov RA, Usacheva YV, Chernyaeva EV. THE EFFICACY AND SAFETY OF RITUXIMAB BIOSIMILAR (ACELLBIA®) IN RHEUMATOID ARTHRITIS AS THE FIRST BIOLOGICAL AGENT: RESULTS OF PHASE III (ALTERRA) CLINICAL TRIAL. RHEUMATOLOGY SCIENCE AND PRACTICE 2017. [DOI: 10.14412/1995-4484-2017-351-359] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The Russian biotechnological company «BIOCAD» has designed a chimeric monoclonal antibody against CD20 (BCD-020, Acellbia®) that is a biosimilar of rituximab (RTM; MabThera®, F. Hoffmann-La Roche Ltd., Switzerland). In recent years, there has been evidence that RTM can be used at lower doses than those given in the standard recommendations and instructions for the use of this drug. This serves as the basis for the BCD-020-4/ALTERRA (ALTErnative Rituximab regimen in Rheumatoid Arthritis) trial, the objective of which was to investigate the efficiency and safety of using Acellbia® (at a dose of 600 mg twice at a 2-week interval) as the first biological agent (BA) for methotrexate (MTX)-resistant active rheumatoid arthritis (RA). The investigation enrolled 159 patients aged 18 to 80 years with active RA. After 24 weeks 65.7 and 29.4% of patients achieved 20% improvement by the American College of Rheumatology (ACR) criteria in the Acellbia® + MTX and placebo (PL) + MTX groups, respectively (p<0.0001). The differences in the ACR20 response rate in the two groups were 36.3% (95% CI, 19.27–53.28%). There were significant differences between the groups in the ACR50 response rates: 28.4% and 5.9% (p=0.001) and in the ACR70 ones: 12.8% and only 2.0%, respectively (p=0.036). Analysis of all recorded adverse events (AE) frequency showed no significant differences between the patients in the study and control groups and demonstrates its equivalence with that of RTM (MabThera®); all the AE were expectable. It is noted that antibodies to RTM with binding and neutralizing activities had no impact on the efficiency and safety of therapy.
Collapse
Affiliation(s)
- E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology
I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia
| | - V. I. Mazurov
- I.I. Mechnikov North-Western State Medical University, Ministry of Health of Russia
| | | | | | - I. M. Marusenko
- V.A. Baranov Republican Hospital, Ministry of Health of the Republic of Karelia
| | | | - T. V. Plaksina
- N.A. Semashko Nizhny Novgorod Regional Clinical Hospital
| | | | | | | | | | | | | | | | - A. F. Davydova
- Professor S.V. Ochapovsky Territorial Clinical Hospital One
| | - N. A. Eremina
- Railway Clinical Hospital at the Gorky Station, OAO «RZhD»
| | | | | | | | | | - S. A. Smakotina
- Kemerovo State Medical Academy, Ministry of Health of Russia
| | - E. A. Smolyarchuk
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia
| | | | | | | | | |
Collapse
|
17
|
Abstract
Rheumatoid arthritis (RA) is a chronic immunoinflammatory (autoimmune) disease manifested by progressive joint destruction, systemic inflammation of the internal organs, and a wide range of comorbidities associated with chronic inflammation and frequently with adverse drug reactions. However, despite the major advances in the early diagnosis and treatment of RA, which have led to the radical improvement of prognosis in many patients, the problem of pharmacotherapy for RA is far from being solved. This is determined by a lack of sensitive and specific diagnostic and prognostic biomarkers in the early stage of the disease and, most importantly, by the heterogeneity of immunopathogenesis mechanisms in both at the onset of RA and during its progression, which make the personalization of therapy difficult in the patients. Selective block of inflammatory mediators with innovative medicines is frequently associated with primary inefficiency, secondary drug resistance, the development of generalized immunosuppression, the paradoxical activation of an autoimmune process, and the aggravation of comorbidities. At the same time, it is difficult to search for new RA pharmacotherapy targets since the nature of immunopathological disorders in patients can be substantially different from the inflammatory process that takes place when simulating arthritis in laboratory animals. The paper discusses the novel drugs that are used in rheumatology to treat RA or tested in different phases of preclinical or clinical trials, such as tumor necrosis factor-α inhibitors, interleukin-6 (IL-6), IL-17, anti-B cell therapy, bispecific antibodies, blockers of JAK (and other signaling molecules), bioelectronic vagus nerve activation, dendritic cell-based immunotherapy, and other therapies, as well as approaches to secondary prevention of RA in patients with undifferentiated arthritis and clinically suspect arthralgia, who are at high risk for RA. Decoding the mechanisms underlying the pathogenesis of RA and a chronic inflammatory process as a whole has created preconditions for the design of novel medications for the prevention and treatment of this disease, the introduction of which into clinical practice should lead to a radical improvement of prognosis in this disease.
Collapse
Affiliation(s)
- E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology
I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia
| |
Collapse
|
18
|
Moura RA, Quaresma C, Vieira AR, Gonçalves MJ, Polido-Pereira J, Romão VC, Martins N, Canhão H, Fonseca JE. B-cell phenotype and IgD-CD27- memory B cells are affected by TNF-inhibitors and tocilizumab treatment in rheumatoid arthritis. PLoS One 2017; 12:e0182927. [PMID: 28886017 PMCID: PMC5590747 DOI: 10.1371/journal.pone.0182927] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 07/18/2017] [Indexed: 01/22/2023] Open
Abstract
Background The use of TNF-inhibitors and/or the IL-6 receptor antagonist, tocilizumab, in rheumatoid arthritis (RA) have pleiotropic effects that also involve circulating B-cells. The main goal of this study was to assess the effect of TNF-inhibitors and tocilizumab on B-cell phenotype and gene expression in RA. Methods Blood samples were collected from untreated early RA (ERA) patients, established RA patients under methotrexate treatment, established RA patients before and after treatment with TNF-inhibitors and tocilizumab, and healthy donors. B-cell subpopulations were characterized by flow cytometry and B-cell gene expression was analyzed by real-time PCR on isolated B-cells. Serum levels of BAFF, CXCL13 and sCD23 were determined by ELISA. Results The frequency of total CD19+ B cells in circulation was similar between controls and all RA groups, irrespective of treatment, but double negative (DN) IgD-CD27- memory B cells were significantly increased in ERA and established RA when compared to controls. Treatment with TNF-inhibitors and tocilizumab restored the frequency of IgD-CD27- B-cells to normal levels, but did not affect other B cell subpopulations. TACI, CD95, CD5, HLA-DR and TLR9 expression on B-cells significantly increased after treatment with either TNF-inhibitors and/ or tocilizumab, but no significant changes were observed in BAFF-R, BCMA, CD69, CD86, CXCR5, CD23, CD38 and IgM expression on B-cells when comparing baseline with post-treatment follow-ups. Alterations in B-cell gene expression of BAFF-R, TACI, TLR9, FcγRIIB, BCL-2, BLIMP-1 and β2M were found in ERA and established RA patients, but no significant differences were observed after TNF-inhibitors and tocilizumab treatment when comparing baseline and follow-ups. Serum levels of CXCL13, sCD23 and BAFF were not significantly affected by treatment with TNF-inhibitors and tocilizumab. Conclusions In RA patients, the use of TNF-inhibitors and/ or tocilizumab treatment affects B-cell phenotype and IgD-CD27- memory B cells in circulation, but not B-cell gene expression levels.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Arthritis, Rheumatoid/diagnosis
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- B-Lymphocyte Subsets/drug effects
- B-Lymphocyte Subsets/immunology
- B-Lymphocyte Subsets/metabolism
- Biomarkers
- Chemokine CXCL13/blood
- Follow-Up Studies
- Gene Expression Profiling
- Gene Expression Regulation/drug effects
- Humans
- Immunoglobulin D/metabolism
- Immunologic Memory
- Immunophenotyping
- Lymphocyte Count
- Methotrexate/pharmacology
- Methotrexate/therapeutic use
- Phenotype
- Receptors, CXCR5/metabolism
- Receptors, IgE/blood
- Treatment Outcome
- Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
Collapse
Affiliation(s)
- Rita A. Moura
- Rheumatology Research Unit, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- * E-mail:
| | - Cláudia Quaresma
- Rheumatology Research Unit, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ana R. Vieira
- Rheumatology Research Unit, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria J. Gonçalves
- Rheumatology Research Unit, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Rheumatology Department, Centro Hospitalar de Lisboa Norte, EPE, Hospital de Santa Maria, Lisbon Academic Medical Centre, Lisbon, Portugal
| | - Joaquim Polido-Pereira
- Rheumatology Research Unit, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Rheumatology Department, Centro Hospitalar de Lisboa Norte, EPE, Hospital de Santa Maria, Lisbon Academic Medical Centre, Lisbon, Portugal
| | - Vasco C. Romão
- Rheumatology Research Unit, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Rheumatology Department, Centro Hospitalar de Lisboa Norte, EPE, Hospital de Santa Maria, Lisbon Academic Medical Centre, Lisbon, Portugal
| | - Nádia Martins
- Rheumatology Department, Centro Hospitalar de Lisboa Norte, EPE, Hospital de Santa Maria, Lisbon Academic Medical Centre, Lisbon, Portugal
| | - Helena Canhão
- Rheumatology Research Unit, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Rheumatology Department, Centro Hospitalar de Lisboa Norte, EPE, Hospital de Santa Maria, Lisbon Academic Medical Centre, Lisbon, Portugal
| | - João E. Fonseca
- Rheumatology Research Unit, Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Rheumatology Department, Centro Hospitalar de Lisboa Norte, EPE, Hospital de Santa Maria, Lisbon Academic Medical Centre, Lisbon, Portugal
| |
Collapse
|
19
|
Mina-Osorio P. Stem Cell Therapy in the Treatment of Rheumatic Diseases and Application in the Treatment of Systemic Lupus Erythematosus. NEXT-GENERATION THERAPIES AND TECHNOLOGIES FOR IMMUNE-MEDIATED INFLAMMATORY DISEASES 2017. [PMCID: PMC7123283 DOI: 10.1007/978-3-319-42252-7_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Current systemic therapies help to improve the symptoms and quality of life for patients with severe life-threatening rheumatic diseases but provide no curative treatment. For the past two decades, preclinical and clinical studies of stem cell transplantation (SCT) have demonstrated tremendous therapeutic potential for patients with autoimmune rheumatic diseases. Herein, the current advances on stem cell therapies, both in animal models and clinical studies, are discussed, with particular attention on systemic lupus erythematosus (SLE). Despite extensive research and promising data, our knowledge on mechanisms of action for SCT, its administration route and timing, the optimal dose of cells, the cells’ fate and distribution in vivo, and the safety and efficacy of the treatments remains limited. Further research on stem cell biology is required to ensure that therapeutic safety and efficacy, as observed in animal models, can be successfully translated in clinical trials. Current understanding, limitations, and future directions for SCT with respect to rheumatic diseases are also discussed.
Collapse
|
20
|
DAĞLI AF, KARATAŞ A, ORHAN C, TUZCU M, ÖZGEN M, ŞAHİN K, KOCA SS. Antiinflammatory and antioxidant effects of gemcitabinein collagen-induced arthritis model. Turk J Med Sci 2017; 47:1037-1044. [DOI: 10.3906/sag-1606-80] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 12/13/2016] [Indexed: 11/03/2022] Open
|
21
|
Investigation of the human FCRL1, 2, and 4 gene expressions in patients with rheumatoid arthritis. Rheumatol Int 2016; 36:1149-56. [DOI: 10.1007/s00296-016-3495-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 05/07/2016] [Indexed: 10/21/2022]
|
22
|
Bashi T, Shovman O, Fridkin M, Volkov A, Barshack I, Blank M, Shoenfeld Y. Novel therapeutic compound tuftsin-phosphorylcholine attenuates collagen-induced arthritis. Clin Exp Immunol 2016; 184:19-28. [PMID: 26618631 PMCID: PMC4778098 DOI: 10.1111/cei.12745] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 11/26/2015] [Accepted: 11/26/2015] [Indexed: 12/31/2022] Open
Abstract
Treatment with helminthes and helminthes ova improved the clinical symptoms of several autoimmune diseases in patients and in animal models. Phosphorylcholine (PC) proved to be the immunomodulatory molecule. We aimed to decipher the tolerogenic potential of tuftsin-PC (TPC), a novel helminth-based compound in collagen-induced arthritis (CIA) a mouse model of rheumatoid arthritis (RA). CIA DBA/1 mice were treated with TPC subcutaneously (5 µg/0.1 ml) or orally (250 µg/0.1 ml), starting prior to disease induction. The control groups were treated with PBS. Collagen antibodies were tested by enzyme-linked immunosorbent assay (ELISA), cytokine protein levels by ELISA kits and regulatory T (Treg ) and regulatory B (Breg ) cell phenotypes by fluorescence-activated cell sorter (FACS). TPC-treated mice had a significantly lower arthritis score of 1.5 in comparison with control mice 11.8 (P < 0.0001) in both subcutaneous and orally treated groups at day 31. Moreover, histology analysis demonstrated highly inflamed joints in control mice, whereas TPC-treated mice maintained normal joint structure. Furthermore, TPC decreased the titres of circulating collagen II antibodies in mice sera (P < 0.0001), enhanced expression of IL-10 (P < 0.0001) and inhibited production of tumour necrosis factor (TNF)-α, interleukin (IL)-17 and IL-1β (P < 0.0001). TPC significantly expanded the CD4(+) CD25(+) forkhead box protein 3 (FoxP3(+) ) Treg cells and CD19(+) IL-10(+) CD5(high) CD1d(high) T cell immunoglobulin mucin-1 (TIM-1(+) ) Breg cell phenotypes (P < 0.0001) in treated mice. Our data indicate that treatment with TPC attenuates CIA in mice demonstrated by low arthritic score and normal joints histology. TPC treatment reduced proinflammatory cytokines and increased anti-inflammatory cytokine expression, as well as expansion of Treg and Breg cells. Our results may lead to a new approach for a natural therapy for early rheumatoid arthritis onset.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Antibodies/blood
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- B-Lymphocytes, Regulatory/drug effects
- B-Lymphocytes, Regulatory/immunology
- B-Lymphocytes, Regulatory/pathology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Collagen Type II/blood
- Collagen Type II/immunology
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Gene Expression
- Hepatitis A Virus Cellular Receptor 1
- Immunophenotyping
- Injections, Subcutaneous
- Interleukin-10/genetics
- Interleukin-10/immunology
- Interleukin-17/genetics
- Interleukin-17/immunology
- Interleukin-1beta/genetics
- Interleukin-1beta/immunology
- Joints/drug effects
- Joints/immunology
- Joints/pathology
- Male
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Inbred DBA
- Phosphorylcholine/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Tuftsin/pharmacology
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- T. Bashi
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel‐Aviv UniversityTel‐AvivIsrael
| | - O. Shovman
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel‐Aviv UniversityTel‐AvivIsrael
| | - M. Fridkin
- Department of Organic Chemistry, The Weizmann Institute of Sciences, Rehovot, IsraelTel‐AvivIsrael
| | - A. Volkov
- Institute of Pathology, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel‐Aviv UniversityTel‐AvivIsrael
| | - I. Barshack
- Institute of Pathology, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel‐Aviv UniversityTel‐AvivIsrael
| | - M. Blank
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel‐Aviv UniversityTel‐AvivIsrael
| | - Y. Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, affiliated to the Sackler Faculty of Medicine Tel‐Aviv UniversityTel‐AvivIsrael
| |
Collapse
|
23
|
Zampeli E, Vlachoyiannopoulos PG, Tzioufas AG. Treatment of rheumatoid arthritis: Unraveling the conundrum. J Autoimmun 2015; 65:1-18. [PMID: 26515757 DOI: 10.1016/j.jaut.2015.10.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 10/09/2015] [Indexed: 11/28/2022]
Abstract
Rheumatoid arthritis (RA) is a heterogeneous disease with a complex and yet not fully understood pathophysiology, where numerous different cell-types contribute to a destructive process of the joints. This complexity results into a considerable interpatient variability in clinical course and severity, which may additionally involve genetics and/or environmental factors. After three decades of focused efforts scientists have now achieved to apply in clinical practice, for patients with RA, the "treat to target" approach with initiation of aggressive therapy soon after diagnosis and escalation of the therapy in pursuit of clinical remission. In addition to the conventional synthetic disease modifying anti-rheumatic drugs, biologics have greatly improved the management of RA, demonstrating efficacy and safety in alleviating symptoms, inhibiting bone erosion, and preventing loss of function. Nonetheless, despite the plethora of therapeutic options and their combinations, unmet therapeutic needs in RA remain, as current therapies sometimes fail or produce only partial responses and/or develop unwanted side-effects. Unfortunately the mechanisms of 'nonresponse' remain unknown and most probable lie in the unrevealed heterogeneity of the RA pathophysiology. In this review, through the effort of unraveling the complex pathophysiological pathways, we will depict drugs used throughout the years for the treatment of RA, the current and future biological therapies and their molecular or cellular targets and finally will suggest therapeutic algorithms for RA management. With multiple biologic options, there is still a need for strong predictive biomarkers to determine which drug is most likely to be effective, safe, and durable in a given individual. The fact that available biologics are not effective in all patients attests to the heterogeneity of RA, yet over the long term, as research and treatment become more aggressive, efficacy, toxicity, and costs must be balanced within the therapeutic equation to enhance the quality of life in patients with RA.
Collapse
Affiliation(s)
- Evangelia Zampeli
- Department of Pathophysiology, School of Medicine, University of Athens, Athens, Greece
| | | | - Athanasios G Tzioufas
- Department of Pathophysiology, School of Medicine, University of Athens, Athens, Greece.
| |
Collapse
|
24
|
Jeffery LE, Raza K, Hewison M. Vitamin D in rheumatoid arthritis-towards clinical application. Nat Rev Rheumatol 2015; 12:201-10. [PMID: 26481434 DOI: 10.1038/nrrheum.2015.140] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In addition to its well-documented involvement in mineral homeostasis, vitamin D seems to have broad effects on human health that go beyond the skeletal system. Prominent among these so-called nonclassical effects of vitamin D are its immunomodulatory properties. In vitro studies have shown anti-inflammatory effects of 1,25-dihydroxyvitamin D (1,25(OH)2D), the active form of vitamin D. In addition, epidemiological analysis of patients with established inflammatory disease identified associations between vitamin D deficiency (low serum concentrations of inactive 25-hydroxyvitamin D, abbreviated to 25(OH)D) and inflammatory conditions, including rheumatoid arthritis (RA). The association of vitamin D deficiency with RA severity supports the hypothesis of a role for vitamin D in the initiation or progression of the disease, or possibly both. However, whether 25(OH)D status is a cause or consequence of RA is still incompletely understood and requires further analysis in prospective vitamin D supplementation trials. The characterization of factors that promote the transition from preclinical to clinical phases of RA has become a major focus of research, with the aim to facilitate earlier diagnosis and treatment, and improve therapeutic outcomes. In this Review, we aim to describe the current knowledge of vitamin D and the immune system specifically in RA, and discuss the potential benefits that vitamin D might have on slowing RA progression.
Collapse
Affiliation(s)
- Louisa E Jeffery
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, The University of Birmingham, Birmingham B15 2TH, UK
| | - Karim Raza
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, The University of Birmingham, Birmingham B15 2TH, UK
| | - Martin Hewison
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, The University of Birmingham, Birmingham B15 2TH, UK
| |
Collapse
|
25
|
Chighizola CB, Favalli EG, Meroni PL. Novel mechanisms of action of the biologicals in rheumatic diseases. Clin Rev Allergy Immunol 2015; 47:6-16. [PMID: 23345026 DOI: 10.1007/s12016-013-8359-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Biological drugs targeting pro-inflammatory or co-stimulatory molecules or depleting lymphocyte subsets made a revolution in rheumatoid arthritis (RA) treatment. Their comparable efficacy in clinical trials raised the point of the heterogeneity of RA pathogenesis, suggesting that we are dealing with a syndrome rather than with a single disease. Several tumor necrosis factor-alpha (TNF-α) blockers are available, and a burning question is whether they are biosimilar or not. The evidence of diverse biological effects in vitro is in line with the fact that a lack of efficacy to one TNF-α agent does not imply a non-response to another one. As proteins, biologicals are potentially immunogenic. It has been recently raised that anti-drug antibodies (ADA) may affect their bioavailability and eventually the clinical efficacy through local formation of immune complexes and directly by preventing the interaction between the drug and TNF-α. Regular monitoring of drug and ADA levels appears the best way to tailor anti-TNF-α therapies. Owing to the pleiotropic characteristics of the target, anti-TNF-α blockers may affect several mechanisms beyond rheumatoid synovitis. As TNF-α plays a pivotal role in the induction of early atherosclerosis, treatment with TNF-inhibitors may modulate cholesterol handling, in particular, cholesterol efflux from macrophages. Side effects are a major issue because of the systemic TNF-α blocking action. The efficacy of an anti-C5 monoclonal antibody fused to a peptide targeting inflamed synovia in experimental arthritis opened the way for new strategies: Homing to the synovium of molecules neutralizing TNF would allow to maximize the therapeutic action avoiding the side effects.
Collapse
|
26
|
Armas-González E, Díaz-Martín A, Domínguez-Luis MJ, Arce-Franco MT, Herrera-García A, Hernández-Hernández MV, Bustabad S, Usategui A, Pablos JL, Cañete JD, Díaz-González F. Differential Antigen-presenting B Cell Phenotypes from Synovial Microenvironment of Patients with Rheumatoid and Psoriatic Arthritis. J Rheumatol 2015; 42:1825-34. [PMID: 26178284 DOI: 10.3899/jrheum.141577] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2015] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To study the qualitative and quantitative phenotypic changes that occur in molecules involved in antigen presentation and costimulation in synovial B cells from rheumatoid arthritis (RA) and psoriatic arthritis (PsA). METHODS The presence of HLA-DR, CD86, and CD40 in CD20+ cells was studied in RA synovium biopsies using immunohistochemistry and immunofluorescence. Expression was assessed by flow cytometry of the Class II molecules CD40, CD86, CD23, and CD27 on B cells from the synovial fluid (SF), with respect to peripheral blood, from 13 patients with RA and 15 patients with PsA. Expression of interferon-induced protein with tetratricopeptide repeats 4 (IFIT4) in immune-selected CD20+ cells from patients with RA was assessed by quantitative realtime PCR. RESULTS Infiltrating synovial RA, B cells expressed HLA-DR, CD40, and CD86. Increased expression of CD86, HLA-DR, and HLA-DQ in B cells from SF was found in patients with RA and PsA. HLA-DP was also elevated in rheumatoid SF B cells; conversely, a significantly lower expression was observed in SF from patients with PsA. CD40 expression was increased in SF B cells from PsA, but not in patients with RA. Interestingly, CD20 surface expression level was significantly lower in SF B cells (CD19+, CD138-) from RA, but not in patients with PsA. CD27 upregulation and CD23 downregulation were observed in synovial B cells in both pathologies. Finally, a 4-fold increase in IFIT4 mRNA content was shown in B cells from SF in patients with RA. CONCLUSION Synovial B cells from patients with RA and patients with PsA express different antigen-presenting cell phenotypes, suggesting that this cell type plays a dissimilar role in the pathogenesis of each disease.
Collapse
Affiliation(s)
- Estefanía Armas-González
- From the Departamento de Farmacología, and Departamento de Medicina, Facultad de Medicina, and Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Investigaciones Biomédicas, Universidad de La Laguna; Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife; Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre, Madrid; Servicio de Reumatología, Hospital Clinic, Barcelona, Spain.E. Armas-González, PhD; A. Díaz-Martín, PhD, Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, and Servicio de Reumatología, Hospital Universitario de Canarias; M.J. Domínguez-Luis, PhD, Centro para la Investigación Biomédica de las Islas Canarias, and Instituto de Investigaciones Biomédicas, Universidad de la Laguna; M.T. Arce-Franco, PhD; A. Herrera-García, PhD; M.V. Hernández-Hernández, MD; S. Bustabad, MD, Servicio de Reumatología, Hospital Universitario de Canarias; A. Usategui, MD, Servicio de Reumatología, Hospital 12 de Octubre; J.L. Pablos, MD, Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre; J.D. Cañete, MD, Servicio de Reumatología, Hospital Clinic; F. Díaz-González, MD, Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna
| | - Ana Díaz-Martín
- From the Departamento de Farmacología, and Departamento de Medicina, Facultad de Medicina, and Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Investigaciones Biomédicas, Universidad de La Laguna; Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife; Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre, Madrid; Servicio de Reumatología, Hospital Clinic, Barcelona, Spain.E. Armas-González, PhD; A. Díaz-Martín, PhD, Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, and Servicio de Reumatología, Hospital Universitario de Canarias; M.J. Domínguez-Luis, PhD, Centro para la Investigación Biomédica de las Islas Canarias, and Instituto de Investigaciones Biomédicas, Universidad de la Laguna; M.T. Arce-Franco, PhD; A. Herrera-García, PhD; M.V. Hernández-Hernández, MD; S. Bustabad, MD, Servicio de Reumatología, Hospital Universitario de Canarias; A. Usategui, MD, Servicio de Reumatología, Hospital 12 de Octubre; J.L. Pablos, MD, Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre; J.D. Cañete, MD, Servicio de Reumatología, Hospital Clinic; F. Díaz-González, MD, Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna
| | - María Jesús Domínguez-Luis
- From the Departamento de Farmacología, and Departamento de Medicina, Facultad de Medicina, and Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Investigaciones Biomédicas, Universidad de La Laguna; Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife; Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre, Madrid; Servicio de Reumatología, Hospital Clinic, Barcelona, Spain.E. Armas-González, PhD; A. Díaz-Martín, PhD, Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, and Servicio de Reumatología, Hospital Universitario de Canarias; M.J. Domínguez-Luis, PhD, Centro para la Investigación Biomédica de las Islas Canarias, and Instituto de Investigaciones Biomédicas, Universidad de la Laguna; M.T. Arce-Franco, PhD; A. Herrera-García, PhD; M.V. Hernández-Hernández, MD; S. Bustabad, MD, Servicio de Reumatología, Hospital Universitario de Canarias; A. Usategui, MD, Servicio de Reumatología, Hospital 12 de Octubre; J.L. Pablos, MD, Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre; J.D. Cañete, MD, Servicio de Reumatología, Hospital Clinic; F. Díaz-González, MD, Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna
| | - María Teresa Arce-Franco
- From the Departamento de Farmacología, and Departamento de Medicina, Facultad de Medicina, and Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Investigaciones Biomédicas, Universidad de La Laguna; Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife; Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre, Madrid; Servicio de Reumatología, Hospital Clinic, Barcelona, Spain.E. Armas-González, PhD; A. Díaz-Martín, PhD, Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, and Servicio de Reumatología, Hospital Universitario de Canarias; M.J. Domínguez-Luis, PhD, Centro para la Investigación Biomédica de las Islas Canarias, and Instituto de Investigaciones Biomédicas, Universidad de la Laguna; M.T. Arce-Franco, PhD; A. Herrera-García, PhD; M.V. Hernández-Hernández, MD; S. Bustabad, MD, Servicio de Reumatología, Hospital Universitario de Canarias; A. Usategui, MD, Servicio de Reumatología, Hospital 12 de Octubre; J.L. Pablos, MD, Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre; J.D. Cañete, MD, Servicio de Reumatología, Hospital Clinic; F. Díaz-González, MD, Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna
| | - Ada Herrera-García
- From the Departamento de Farmacología, and Departamento de Medicina, Facultad de Medicina, and Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Investigaciones Biomédicas, Universidad de La Laguna; Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife; Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre, Madrid; Servicio de Reumatología, Hospital Clinic, Barcelona, Spain.E. Armas-González, PhD; A. Díaz-Martín, PhD, Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, and Servicio de Reumatología, Hospital Universitario de Canarias; M.J. Domínguez-Luis, PhD, Centro para la Investigación Biomédica de las Islas Canarias, and Instituto de Investigaciones Biomédicas, Universidad de la Laguna; M.T. Arce-Franco, PhD; A. Herrera-García, PhD; M.V. Hernández-Hernández, MD; S. Bustabad, MD, Servicio de Reumatología, Hospital Universitario de Canarias; A. Usategui, MD, Servicio de Reumatología, Hospital 12 de Octubre; J.L. Pablos, MD, Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre; J.D. Cañete, MD, Servicio de Reumatología, Hospital Clinic; F. Díaz-González, MD, Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna
| | - María Vanesa Hernández-Hernández
- From the Departamento de Farmacología, and Departamento de Medicina, Facultad de Medicina, and Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Investigaciones Biomédicas, Universidad de La Laguna; Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife; Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre, Madrid; Servicio de Reumatología, Hospital Clinic, Barcelona, Spain.E. Armas-González, PhD; A. Díaz-Martín, PhD, Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, and Servicio de Reumatología, Hospital Universitario de Canarias; M.J. Domínguez-Luis, PhD, Centro para la Investigación Biomédica de las Islas Canarias, and Instituto de Investigaciones Biomédicas, Universidad de la Laguna; M.T. Arce-Franco, PhD; A. Herrera-García, PhD; M.V. Hernández-Hernández, MD; S. Bustabad, MD, Servicio de Reumatología, Hospital Universitario de Canarias; A. Usategui, MD, Servicio de Reumatología, Hospital 12 de Octubre; J.L. Pablos, MD, Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre; J.D. Cañete, MD, Servicio de Reumatología, Hospital Clinic; F. Díaz-González, MD, Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna
| | - Sagrario Bustabad
- From the Departamento de Farmacología, and Departamento de Medicina, Facultad de Medicina, and Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Investigaciones Biomédicas, Universidad de La Laguna; Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife; Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre, Madrid; Servicio de Reumatología, Hospital Clinic, Barcelona, Spain.E. Armas-González, PhD; A. Díaz-Martín, PhD, Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, and Servicio de Reumatología, Hospital Universitario de Canarias; M.J. Domínguez-Luis, PhD, Centro para la Investigación Biomédica de las Islas Canarias, and Instituto de Investigaciones Biomédicas, Universidad de la Laguna; M.T. Arce-Franco, PhD; A. Herrera-García, PhD; M.V. Hernández-Hernández, MD; S. Bustabad, MD, Servicio de Reumatología, Hospital Universitario de Canarias; A. Usategui, MD, Servicio de Reumatología, Hospital 12 de Octubre; J.L. Pablos, MD, Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre; J.D. Cañete, MD, Servicio de Reumatología, Hospital Clinic; F. Díaz-González, MD, Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna
| | - Alicia Usategui
- From the Departamento de Farmacología, and Departamento de Medicina, Facultad de Medicina, and Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Investigaciones Biomédicas, Universidad de La Laguna; Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife; Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre, Madrid; Servicio de Reumatología, Hospital Clinic, Barcelona, Spain.E. Armas-González, PhD; A. Díaz-Martín, PhD, Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, and Servicio de Reumatología, Hospital Universitario de Canarias; M.J. Domínguez-Luis, PhD, Centro para la Investigación Biomédica de las Islas Canarias, and Instituto de Investigaciones Biomédicas, Universidad de la Laguna; M.T. Arce-Franco, PhD; A. Herrera-García, PhD; M.V. Hernández-Hernández, MD; S. Bustabad, MD, Servicio de Reumatología, Hospital Universitario de Canarias; A. Usategui, MD, Servicio de Reumatología, Hospital 12 de Octubre; J.L. Pablos, MD, Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre; J.D. Cañete, MD, Servicio de Reumatología, Hospital Clinic; F. Díaz-González, MD, Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna
| | - José L Pablos
- From the Departamento de Farmacología, and Departamento de Medicina, Facultad de Medicina, and Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Investigaciones Biomédicas, Universidad de La Laguna; Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife; Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre, Madrid; Servicio de Reumatología, Hospital Clinic, Barcelona, Spain.E. Armas-González, PhD; A. Díaz-Martín, PhD, Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, and Servicio de Reumatología, Hospital Universitario de Canarias; M.J. Domínguez-Luis, PhD, Centro para la Investigación Biomédica de las Islas Canarias, and Instituto de Investigaciones Biomédicas, Universidad de la Laguna; M.T. Arce-Franco, PhD; A. Herrera-García, PhD; M.V. Hernández-Hernández, MD; S. Bustabad, MD, Servicio de Reumatología, Hospital Universitario de Canarias; A. Usategui, MD, Servicio de Reumatología, Hospital 12 de Octubre; J.L. Pablos, MD, Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre; J.D. Cañete, MD, Servicio de Reumatología, Hospital Clinic; F. Díaz-González, MD, Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna
| | - Juan D Cañete
- From the Departamento de Farmacología, and Departamento de Medicina, Facultad de Medicina, and Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Investigaciones Biomédicas, Universidad de La Laguna; Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife; Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre, Madrid; Servicio de Reumatología, Hospital Clinic, Barcelona, Spain.E. Armas-González, PhD; A. Díaz-Martín, PhD, Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, and Servicio de Reumatología, Hospital Universitario de Canarias; M.J. Domínguez-Luis, PhD, Centro para la Investigación Biomédica de las Islas Canarias, and Instituto de Investigaciones Biomédicas, Universidad de la Laguna; M.T. Arce-Franco, PhD; A. Herrera-García, PhD; M.V. Hernández-Hernández, MD; S. Bustabad, MD, Servicio de Reumatología, Hospital Universitario de Canarias; A. Usategui, MD, Servicio de Reumatología, Hospital 12 de Octubre; J.L. Pablos, MD, Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre; J.D. Cañete, MD, Servicio de Reumatología, Hospital Clinic; F. Díaz-González, MD, Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna
| | - Federico Díaz-González
- From the Departamento de Farmacología, and Departamento de Medicina, Facultad de Medicina, and Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Investigaciones Biomédicas, Universidad de La Laguna; Servicio de Reumatología, Hospital Universitario de Canarias, Tenerife; Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre, Madrid; Servicio de Reumatología, Hospital Clinic, Barcelona, Spain.E. Armas-González, PhD; A. Díaz-Martín, PhD, Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, and Servicio de Reumatología, Hospital Universitario de Canarias; M.J. Domínguez-Luis, PhD, Centro para la Investigación Biomédica de las Islas Canarias, and Instituto de Investigaciones Biomédicas, Universidad de la Laguna; M.T. Arce-Franco, PhD; A. Herrera-García, PhD; M.V. Hernández-Hernández, MD; S. Bustabad, MD, Servicio de Reumatología, Hospital Universitario de Canarias; A. Usategui, MD, Servicio de Reumatología, Hospital 12 de Octubre; J.L. Pablos, MD, Servicio de Reumatología, and Instituto de Investigación, Hospital 12 de Octubre; J.D. Cañete, MD, Servicio de Reumatología, Hospital Clinic; F. Díaz-González, MD, Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna.
| |
Collapse
|
27
|
Abstract
PURPOSE The function of regulatory B lymphocytes is known to be abnormal in inflammatory diseases. However, a recent study indicates that IL-10+ B cells seem to be expanded in rheumatoid arthritis (RA). Therefore, the state of IL-10+ B cells in the peripheral blood from RA patients and healthy controls were investigated. MATERIALS AND METHODS CD19+ cells in peripheral blood mononuclear cells were purified from blood samples of RA patients and age and gender-matched healthy controls, and stimulated with CD40 ligand and CpG for 48 hours. Then, intracellular IL-10 in CD19+ cells was analyzed using flow cytometry. RESULTS There was no significant difference in the proportion of IL-10+ B cells between 10 RA patients and 10 healthy controls (RA, 0.300±0.07 vs. healthy control 0.459±0.07, p=0.114). The proportion of induced IL-10+ B cells to total B cells in RA patients was significantly higher than those in controls (RA, 4.44±3.44% vs. healthy control 2.44±1.64%, p=0.033). However, the proportion of IL-10+ B cells to total B cells correlated negatively with disease activity in RA patients (r=-0.398, p=0.040). Erythrocyte sedimentation rate or C-reactive protein or medication was not associated with the proportion of IL-10+ B cells. CONCLUSION The proportion of induced IL-10+ B cell increased in RA patients compared to healthy control, however, negatively correlated with disease activity in RA.
Collapse
Affiliation(s)
- Jinhyun Kim
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Hyun Ji Lee
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - In Seol Yoo
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Seong Wook Kang
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Korea
| | - Jae Ho Lee
- Department of Paediatrics, Chungnam National University School of Medicine, Daejeon, Korea.
| |
Collapse
|
28
|
Stem Cell Therapy in Autoimmune Rheumatic Diseases: a Comprehensive Review. Clin Rev Allergy Immunol 2014; 47:244-57. [DOI: 10.1007/s12016-014-8445-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
29
|
Ishikawa LLW, Shoenfeld Y, Sartori A. Immunomodulation in human and experimental arthritis: including vitamin D, helminths and heat-shock proteins. Lupus 2014; 23:577-87. [DOI: 10.1177/0961203314527369] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that is mainly directed to the joints, affecting the synovial membrane, the cartilage and also the bone. This disease affects 1% to 2% of the world population and is associated with significant morbidity and increased mortality. RA experimental models have allowed a great deal of information to be translated to the corresponding human disease. This review summarizes some of the most relevant findings targeting immunomodulation in arthritis. Some general guidelines to choose an adequate experimental model and also our experience with arthritis are supplied.
Collapse
Affiliation(s)
- LLW Ishikawa
- Department of Microbiology and Immunology, Biosciences Institute, Univ. Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | - Y Shoenfeld
- The Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
| | - A Sartori
- Department of Microbiology and Immunology, Biosciences Institute, Univ. Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
30
|
CD20+ B cell depletion in systemic autoimmune diseases: common mechanism of inhibition or disease-specific effect on humoral immunity? BIOMED RESEARCH INTERNATIONAL 2014; 2014:973609. [PMID: 24791010 PMCID: PMC3985174 DOI: 10.1155/2014/973609] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 03/03/2014] [Indexed: 11/21/2022]
Abstract
Autoimmunity remains a complex physiologic deviation, enabled and perpetuated by a variety of interplayers and pathways. Simplistic approaches, targeting either isolated end-effectors of more centrally placed interactors of these mechanisms, are continuously tried in an effort to comprehend and halt cascades with potential disabling and deleterious effects in the affected individuals. This review focuses on theoretical and clinically proved effects of rituximab-induced CD20+ B cell depletion on different systemic autoimmune diseases and extrapolates on pathogenetic mechanisms that may account for different interindividual or interdisease responses.
Collapse
|
31
|
Paula FS, Alves JD. Non-tumor necrosis factor-based biologic therapies for rheumatoid arthritis: present, future, and insights into pathogenesis. Biologics 2013; 8:1-12. [PMID: 24353404 PMCID: PMC3861294 DOI: 10.2147/btt.s35475] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The way rheumatoid arthritis is treated has changed dramatically with the introduction of anti-tumor necrosis factor (anti-TNF) biologics. Nevertheless, many patients still have less than adequate control of their disease activity even with these therapeutic regimens, and current knowledge fails to explain all the data already gathered. There is now a wide range of drugs from different classes of biologic disease-modifying anti-rheumatic drugs available (and soon this number will increase significantly), that provides the opportunity to address each patient as a particular case and thereby optimize medical intervention. Currently available biologics for the treatment of rheumatoid arthritis apart from anti-TNF-based therapies are reviewed, along with an analysis of the new insights they provide into the pathogenesis of the disease and a discussion of future prospects in the area.
Collapse
Affiliation(s)
- Filipe Seguro Paula
- Immunomediated Systemic Diseases Unit, Department of Medicine 4, Fernando Fonseca Hospital, Amadora, Portugal
| | - José Delgado Alves
- Immunomediated Systemic Diseases Unit, Department of Medicine 4, Fernando Fonseca Hospital, Amadora, Portugal ; Center for the Study of Chronic Diseases, Department of Pharmacology, Faculty of Medical Sciences, Lisbon, Portugal
| |
Collapse
|
32
|
Morris-Rosenfeld S, Lipinski MJ, McNamara CA. Understanding the role of B cells in atherosclerosis: potential clinical implications. Expert Rev Clin Immunol 2013; 10:77-89. [PMID: 24308836 DOI: 10.1586/1744666x.2014.857602] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Atherosclerosis is a progressive inflammatory disease of the medium to large arteries that is the largest contributor to cardiovascular disease. B-cell subsets have been shown in animal models of atherosclerosis to have both atherogenic and atheroprotective properties. In this review, we highlight the research that developed our understanding of the role of B cells in atherosclerosis both in humans and mice. From this we discuss the potential clinical impact B cells could have both as diagnostic biomarkers and as targets for immunotherapy. Finally, we recognize the inherent difficulty in translating findings from animal models into humans given the differences in both cardiovascular disease and the immune system between mice and humans, making the case for greater efforts at addressing the role of B cells in human atherosclerosis.
Collapse
Affiliation(s)
- Samuel Morris-Rosenfeld
- Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA and Department of Medicine, Cardiovascular Division at the University of Virginia, Charlottesville, VA, USA
| | | | | |
Collapse
|
33
|
Klementiev B, Enevoldsen MN, Li S, Carlsson R, Liu Y, Issazadeh-Navikas S, Bock E, Berezin V. Antiinflammatory properties of a peptide derived from interleukin-4. Cytokine 2013; 64:112-21. [PMID: 23972727 DOI: 10.1016/j.cyto.2013.07.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 06/19/2013] [Accepted: 07/20/2013] [Indexed: 12/23/2022]
Abstract
Interleukin-4 (IL-4) is a potent antiinflammatory cytokine. However its use in the clinic is hampered by side effects. We here describe the identification of a novel synthetic peptide, termed Ph8, derived from α-helix C of IL-4, which interacts with IL-4 receptor α (IL-4Rα). Employing various cultured genetically engineered cell lines and primary lymphocytes, surface plasmon resonance, qPCR, ELISA and immunoblotting techniques we found that Ph8 bound IL-4Rα and mimicked the anti-inflammatory effects of IL-4 by inhibiting TNF-α production by macrophages in vitro. It induced phosphorylation of STAT6 65kD but inhibited phosphorylation of STAT6 110 kD induced by IL-4 in a B-cell line that expressed the type I receptor. It also inhibited the IL-4-stimulated expression of a STAT6-inducible reporter gene in cells that expressed the type II receptor. Ph8 inhibited the proliferation of Th1/2 cells and downregulated the production of IFN-γ in stimulated Th1 cells. Moreover, Ph8 did not induce any shift in Th1/Th2 profile. This is a favorable effect and it is indicating that Ph8 could block general T cell activation and inflammatory responses without further inducing the side effects generally associated with IL-4 signaling. These data collectively show that Ph8 is only a partial agonist of IL-4 mimicking its desirable properties. In agreement, Ph8 treatment of rats with collagen-induced arthritis, a Th1- and antibody- mediated disease of joint, delayed the manifestation of chronic inflammation and reduced acute inflammation in carrageenan-induced edema. Our findings indicate that Ph8 is a promising potential drug candidate for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Boris Klementiev
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Selmi C, Gershwin ME. The long-term marriage between autoimmunity and internal medicine: a homage to Manuel Carlos Dias. Clin Rev Allergy Immunol 2013; 43:207-10. [PMID: 22826113 DOI: 10.1007/s12016-012-8333-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Our understanding of autoimmune diseases results from the perfect combination of basic and clinical scientific research, and the figure that is closest to the proposed autoimmunology specialist is certainly the internist. The role of B cells in rheumatoid arthritis, the immunological mechanisms to fibrosis or to tissue specific damage, the classification of Bechet's syndrome, the clinical outcomes of antiphospholid syndrome, and new biomarkers for vascular complications in systemic sclerosis constitute, among others, are ideal examples of this combination. For these reason, this issue includes comprehensive reviews in all these areas and is dedicated to Dr. Manuel Carlos Dias and his career in the perfectioning and teaching of the clinical skills necessary to manage autoimmune disease. We are convinced that these discussions are likely of interest to basic scientists and clinicians alike for the proposed translational applications.
Collapse
Affiliation(s)
- Carlo Selmi
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA 95616, USA.
| | | |
Collapse
|