全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Rheumatoid Factor Positivity Is Associated with Increased Joint Destruction and Upregulation of Matrix Metalloproteinase 9 and Cathepsin K Gene Expression in the Peripheral Blood in Rheumatoid Arthritic Patients Treated with Methotrexate

DOI: 10.1155/2013/457876

Full-Text   Cite this paper   Add to My Lib

Abstract:

We evaluated changes in gene expression of mTOR, p21, caspase-3, ULK1, TNFα, matrix metalloproteinase (MMP)-9, and cathepsin K in the whole blood of rheumatoid arthritic (RA) patients treated with methotrexate (MTX) in relation to their rheumatoid factor status, clinical, immunological, and radiological parameters, and therapeutic response after a 24-month follow-up. The study group consisted of 35 control subjects and 33 RA patients without previous history of MTX treatment. Gene expression was measured using real-time RT-PCR. Decreased disease activity in patients at the end of the study was associated with significant downregulation of TNFα expression. Downregulation of mTOR was observed in seronegative patients, while no significant changes in the expression of p21, ULK1, or caspase-3 were noted in any RA patients at the end of the study. The increase in erosion numbers observed in the seropositive patients at the end of the follow-up was accompanied by upregulation of MMP-9 and cathepsin K, while seronegative patients demonstrated an absence of significant changes in MMP-9 and cathepsin K expression and no increase in the erosion score. Our results suggest that increased expression of MMP-9 and cathepsin K genes in the peripheral blood might indicate higher bone tissue destruction activity in RA patients treated with methotrexate. The clinical study registration number is 0120.0810610. 1. Introduction Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by synovial hyperplasia, mononuclear cell infiltration, bone erosion, and joint destruction. Early diagnosis and immediate aggressive treatment are required for the amelioration of progressive joint damage and patient disability [1, 2]. Methotrexate (MTX) is the most conventional disease-modifying antirheumatic drug (DMARD) for RA, with the best efficacy and the fewest adverse effects [3, 4]. However, only approximately 30% of patients respond to MTX treatment [5, 6]. The identification of patients who are less responsive to MTX could avoid delays in adjusting their treatment and prevent future irreversible joint damage [7]. Rheumatoid factor (RF) is a part of the 2010 American College of Rheumatology (ACR) classification criteria for RA [8]. RF is an autoantibody directed against the Fc portion of IgG and is associated with disease persistence and progressive joint destruction [9–11]. However, the data related to RF status in treatment response to MTX is inconsistent, as some studies reported no association between RF positivity and treatment efficacy [12–21], while others

References

[1]  E. L. Matteson, C. M. Weyand, J. W. Fulbright, T. J. H. Christianson, R. L. McClelland, and J. J. Goronzy, “How aggressive should initial therapy for rheumatoid arthritis be? Factors associated with response to ‘non-aggressive’ DMARD treatment and perspective from a 2-yr open label trial,” Rheumatology, vol. 43, no. 5, pp. 619–625, 2004.
[2]  D. L. Scott, “Evidence for early disease-modifying drugs in rheumatiod arthritis,” Arthritis Research and Therapy, vol. 6, no. 1, pp. 15–18, 2004.
[3]  M. E. Weinblatt, “Efficacy of methotrexate in rheumatoid arthritis,” British Journal of Rheumatology, vol. 34, supplement 2, pp. 43–48, 1995.
[4]  W. Katchamart, J. Trudeau, V. Phumethum, and C. Bombardier, “Efficacy and toxicity of methotrexate (MTX) monotherapy versus MTX combination therapy with non-biological disease-modifying antirheumatic drugs in rheumatoid arthritis: a systematic review and meta-analysis,” Annals of the Rheumatic Diseases, vol. 68, no. 7, pp. 1105–1112, 2009.
[5]  F. C. Breedveld, M. H. Weisman, A. F. Kavanaugh et al., “The PREMIER study: a multicenter, randomized, double-blind clinical trial of combination therapy with adalimumab plus methotrexate versus methotrexate alone or adalimumab alone in patients with early, aggressive rheumatoid arthritis who had not had previous methotrexate treatment,” Arthritis & Rheumatism, vol. 54, no. 1, pp. 26–37, 2006.
[6]  P. Emery, F. C. Breedveld, S. Hall et al., “Comparison of methotrexate monotherapy with a combination of methotrexate and etanercept in active, early, moderate to severe rheumatoid arthritis (COMET): a randomised, double-blind, parallel treatment trial,” The Lancet, vol. 372, no. 9636, pp. 375–382, 2008.
[7]  J. K. de Vries-Bouwstra, Y. P. M. Goekoop-Ruiterman, K. N. Verpoort et al., “Progression of joint damage in early rheumatoid arthritis: association with HLA-DRB1, rheumatoid factor, and anti-citrullinated protein antibodies in relation to different treatment strategies,” Arthritis & Rheumatism, vol. 58, no. 5, pp. 1293–1298, 2008.
[8]  D. Aletaha, T. Neogi, A. J. Silman, et al., “2010 rheumatoid arthritis classification criteria: an American College of Rheumatology/European League against Rheumatism collaborative initiative,” Arthritis & Rheumatism, vol. 62, no. 9, pp. 2569–2581, 2010.
[9]  T. M?tt?nen, L. Paimela, M. Leirisalo-Repo, H. Kautiainen, J. Ilonen, and P. Hannonen, “Only high disease activity and positive rheumatoid factor indicate poor prognosis in patients with early rheumatoid arthritis treated with “sawtooth” strategy,” Annals of the Rheumatic Diseases, vol. 57, no. 9, pp. 533–539, 1998.
[10]  O. Vittecoq, S. Pouplin, K. Krzanowska et al., “Rheumatoid factor is the strongest predictor of radiological progression of rheumatoid arthritis in a three-year prospective study in community-recruited patients,” Rheumatology, vol. 42, no. 8, pp. 939–946, 2003.
[11]  M. Bukhari, M. Lunt, B. J. Harrison, D. G. I. Scott, D. P. M. Symmons, and A. J. Silman, “Rheumatoid factor is the major predictor of increasing severity of radiographic erosions in rheumatoid arthritis: results from the norfolk arthritis register study, a large inception cohort,” Arthritis & Rheumatism, vol. 46, no. 4, pp. 906–912, 2002.
[12]  S. L. Hider, A. J. Silman, W. Thomson, M. Lunt, D. Bunn, and D. P. M. Symmons, “Can clinical factors at presentation be used to predict outcome of treatment with methotrexate in patients with early inflammatory polyarthritis?” Annals of the Rheumatic Diseases, vol. 68, no. 1, pp. 57–62, 2009.
[13]  S. Saevarsdottir, H. Wallin, M. Seddighzadeh et al., “Predictors of response to methotrexate in early DMARD na?ve rheumatoid arthritis: results from the initial open-label phase of the SWEFOT trial,” Annals of the Rheumatic Diseases, vol. 70, no. 3, pp. 469–475, 2011.
[14]  B. Hodkinson, E. Musenge, M. Ally, P. W. A. Meyer, R. Anderson, and M. Tikly, “Response to traditional disease-modifying anti-rheumatic drugs in indigent South Africans with early rheumatoid arthritis,” Clinical Rheumatology, vol. 31, no. 4, pp. 613–619, 2012.
[15]  K. Forslind, I. Hafstr?m, M. Ahlmén, and B. Svensson, “Sex: a major predictor of remission in early rheumatoid arthritis?” Annals of the Rheumatic Diseases, vol. 66, no. 1, pp. 46–52, 2007.
[16]  T. M?tt?nen, P. Hannonen, M. Korpela et al., “Delay to institution of therapy and induction of remission using single-drug or combination-disease-modifying antirheumatic drug therapy in early rheumatoid arthritis,” Arthritis & Rheumatism, vol. 46, no. 4, pp. 894–898, 2002.
[17]  B. Combe, A. Cantagrel, P. Goupille et al., “Predictive factors of 5-year health assessment questionnaire disability in early rheumatoid arthritis,” Journal of Rheumatology, vol. 30, no. 11, pp. 2344–2349, 2003.
[18]  D. Aletaha and J. S. Smolen, “The rheumatoid arthritis patient in the clinic: comparing more than 1300 consecutive DMARD courses,” Rheumatology, vol. 41, no. 12, pp. 1367–1374, 2002.
[19]  M. Hoekstra, A. E. van Ede, C. J. Haagsma et al., “Factors associated with toxicity, final dose, and efficacy of methotrexate in patients with rheumatoid arthritis,” Annals of the Rheumatic Diseases, vol. 62, no. 5, pp. 423–426, 2003.
[20]  H. Maradit-Kremers, P. J. Nicola, C. S. Crowson, W. M. O'Fallon, and S. E. Gabriel, “Patient, disease, and therapy-related factors that influence discontinuation of disease-modifying antirheumatic drugs: a population-based incidence cohort of patients with rheumatoid arthritis,” Journal of Rheumatology, vol. 33, no. 2, pp. 248–255, 2006.
[21]  L. M. da Mota, L. L. dos Santos Neto, J. F. de Carvalho, et al., “The presence of anti-citrullinated protein antibodies (ACPA) and rheumatoid factor on patients with rheumatoid arthritis (RA) does not interfere with the chance of clinical remission in a follow-up of 3 years,” Rheumatology International, vol. 32, no. 12, pp. 3807–3812, 2012.
[22]  J. A. M. Wessels, S. M. van der Kooij, S. le Cessie et al., “A clinical pharmacogenetic model to predict the efficacy of methotrexate monotherapy in recent-onset rheumatoid arthritis,” Arthritis & Rheumatism, vol. 56, no. 6, pp. 1765–1775, 2007.
[23]  L. Gossec, M. Dougados, P. Goupille et al., “Prognostic factors for remission in early rheumatoid arthritis: a multiparameter prospective study,” Annals of the Rheumatic Diseases, vol. 63, no. 6, pp. 675–680, 2004.
[24]  D. T. Felson, “Epidemiology of the rheumatic diseases,” in Arthritis and Allied Conditions. Textbook of Rheumatology, W. J. Koopman, Ed., pp. 3–38, Lippincott, Williams & Wilkins, Philadelphia, Pa, USA, 14th edition, 2001.
[25]  T. C. T. M. van der Pouw Kraan, F. A. van Gaalen, T. W. J. Huizinga, E. Pieterman, F. C. Breedveld, and C. L. Verweij, “Discovery of distinctive gene expression profiles in rheumatoid synovium using cDNA microarray technology: evidence for the existence of multiple pathways of tissue destruction and repair,” Genes and Immunity, vol. 4, no. 3, pp. 187–196, 2003.
[26]  J. Shou, C. M. Bull, L. Li et al., “Identification of blood biomarkers of rheumatoid arthritis by transcript profiling of peripheral blood mononuclear cells from the rat collagen-induced arthritis model,” Arthritis Research and Therapy, vol. 8, no. 1, article R28, 2006.
[27]  H. Moncrieffe, A. Hinks, S. Ursu et al., “Generation of novel pharmacogenomic candidates in response to methotrexate in juvenile idiopathic arthritis: correlation between gene expression and genotype,” Pharmacogenetics and Genomics, vol. 20, no. 11, pp. 665–676, 2010.
[28]  K. Andreas, T. H?upl, C. Lübke, et al., “Antirheumatic drug response signatures in human chondrocytes: potential molecular targets to stimulate cartilage regeneration,” Arthritis Research & Therapy, vol. 11, no. 1, article R15, 2009.
[29]  J. Gu, E. M?rker-Hermann, D. Baeten et al., “A 588-gene microarray analysis of the peripheral blood mononuclear cells of spondyloarthropathy patients,” Rheumatology, vol. 41, no. 7, pp. 759–766, 2002.
[30]  N. J. Olsen, T. Sokka, C. L. Seehorn et al., “A gene expression signature for recent onset rheumatoid arthritis in peripheral blood mononuclear cells,” Annals of the Rheumatic Diseases, vol. 63, no. 11, pp. 1387–1392, 2004.
[31]  T. C. T. M. van der Pouw Kraan, C. A. Wijbrandts, L. G. M. van Baarsen et al., “Rheumatoid arthritis subtypes identified by genomic profiling of peripheral blood cells: assignment of a type I interferon signature in a subpopulation of patients,” Annals of the Rheumatic Diseases, vol. 66, no. 8, pp. 1008–1014, 2007.
[32]  C. S. Haas, C. J. Creighton, X. Pi et al., “Identification of genes modulated in rheumatoid arthritis using complementary DNA microarray analysis of lymphoblastoid B cell lines from disease-discordant monozygotic twins,” Arthritis & Rheumatism, vol. 54, no. 7, pp. 2047–2060, 2006.
[33]  Z. Tu, L. Wang, M. Xu, X. Zhou, T. Chen, and F. Sun, “Further understanding human disease genes by comparing with housekeeping genes and other genes,” BMC Genomics, vol. 7, article 31, 2006.
[34]  D. Grcevic, Z. Jajic, N. Kovacic et al., “Peripheral blood expression profiles of bone morphogenetic proteins, tumor necrosis factor-superfamily molecules, and transcription factor Runx2 could be used as markers of the form of arthritis, disease activity, and therapeutic responsiveness,” Journal of Rheumatology, vol. 37, no. 2, pp. 246–256, 2010.
[35]  A. I. Catrina, A. K. Ulfgren, S. Lindblad, L. Grondal, and L. Klareskog, “Low levels of apoptosis and high FLIP expression in early rheumatoid arthritis synovium,” Annals of the Rheumatic Diseases, vol. 61, no. 10, pp. 934–936, 2002.
[36]  K. Raza, D. Scheel-Toellner, C.-Y. Lee et al., “Synovial fluid leukocyte apoptosis is inhibited in patients with very early rheumatoid arthritis,” Arthritis Research and Therapy, vol. 8, no. 4, article R120, 2006.
[37]  K. Eguchi, “Apoptosis in autoimmune diseases,” Internal Medicine, vol. 40, no. 4, pp. 275–284, 2001.
[38]  N. Hay and N. Sonenberg, “Upstream and downstream of mTOR,” Genes and Development, vol. 18, no. 16, pp. 1926–1945, 2004.
[39]  N. Yoshimura, Y. Ohmoto, H. Yasui et al., “The direct effect of FK506 and rapamycin on interleukin 1(β) and immunoglobulin production in vitro,” Transplantation, vol. 57, no. 12, pp. 1815–1818, 1994.
[40]  A. D. Foey, M. Feldmann, and F. M. Brennan, “CD40 ligation induces macrophage IL-10 and TNF-α production: differential use of the PI3K and p42/44 MAPK-pathways,” Cytokine, vol. 16, no. 4, pp. 131–142, 2001.
[41]  R. P. Carlson, D. A. Hartman, L. A. Tomchek et al., “Rapamycin, a potential disease-modifying antiarthritic drug,” Journal of Pharmacology and Experimental Therapeutics, vol. 266, no. 2, pp. 1125–1138, 1993.
[42]  J. J. Lum, R. J. DeBerardinis, and C. B. Thompson, “Autophagy in metazoans: cell survival in the land of plenty,” Nature Reviews Molecular Cell Biology, vol. 6, no. 6, pp. 439–448, 2005.
[43]  A. Lleo, P. Invernizzi, C. Selmi et al., “Autophagy: highlighting a novel player in the autoimmunity scenario,” Journal of Autoimmunity, vol. 29, no. 2-3, pp. 61–68, 2007.
[44]  Y.-J. Shin, S.-H. Han, D.-S. Kim et al., “Autophagy induction and CHOP under-expression promotes survival of fibroblasts from rheumatoid arthritis patients under endoplasmic reticulum stress,” Arthritis Research and Therapy, vol. 12, no. 1, article R19, 2010.
[45]  F. Grassi, S. Cristino, S. Toneguzzi, A. Piacentini, A. Facchini, and G. Lisignoli, “CXCL12 chemokine up-regulates bone resorption and MMP-9 release by human osteoclasts: CXCL12 levels are increased in synovial and bone tissue of rheumatoid arthritis patients,” Journal of Cellular Physiology, vol. 199, no. 2, pp. 244–251, 2004.
[46]  M. Kaneko, T. Tomita, T. Nakase et al., “Expression of proteinases and inflammatory cytokines in subchondral bone regions in the destructive joint of rhematoid arthritis,” Rheumatology, vol. 40, no. 3, pp. 247–255, 2001.
[47]  D. Br?mme and F. Lecaille, “Cathepsin K inhibitors for osteoporosis and potential off-target effects,” Expert Opinion on Investigational Drugs, vol. 18, no. 5, pp. 585–600, 2009.
[48]  K. S. Kim, H. M. Choi, Y.-A. Lee et al., “Expression levels and association of gelatinases MMP-2 and MMP-9 and collagenases MMP-1 and MMP-13 with VEGF in synovial fluid of patients with arthritis,” Rheumatology International, vol. 31, no. 4, pp. 543–547, 2011.
[49]  P. S. Burrage, K. S. Mix, and C. E. Brinckerhoff, “Matrix metalloproteinases: role in arthritis,” Frontiers in Bioscience, vol. 11, no. 1, pp. 529–543, 2006.
[50]  M. Skoumal, G. Haberhauer, G. Kolarz et al., “The imbalance between osteoprotegerin and cathepsin K in the serum of patients with longstanding rheumatoid arthritis,” Rheumatology International, vol. 28, no. 7, pp. 637–641, 2008.
[51]  V. J. Richardson, “Divergent and synergistic regulation of matrix metalloprotease production by cytokines in combination with C-C chemokines,” International Journal of Immunopathology and Pharmacology, vol. 23, no. 3, pp. 715–726, 2010.
[52]  J.-L. Huang, S.-Y. Wu, X.-J. Xie, M.-X. Wang, S. Zhu, and J.-R. Gu, “Inhibiting effects of Leflunomide metabolite on overexpression of CD147, MMP-2 and MMP-9 in PMA differentiated THP-1 cells,” European Journal of Pharmacology, vol. 670, no. 1, pp. 304–310, 2011.
[53]  B. Vandooren, E. Kruithof, D. T. Y. Yu et al., “Involvement of matrix metalloproteinases and their inhibitors in peripheral synovitis and down-regulation by tumor necrosis factor α blockade in spondylarthropathy,” Arthritis & Rheumatism, vol. 50, no. 9, pp. 2942–2953, 2004.
[54]  I. Matsushita, M. Uzuki, H. Matsuno, E. Sugiyama, and T. Kimura, “Rheumatoid nodulosis during methotrexate therapy in a patient with rheumatoid arthritis,” Modern Rheumatology, vol. 16, no. 6, pp. 401–403, 2006.
[55]  F. C. Arnett, S. M. Edworthy, D. A. Bloch et al., “The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis,” Arthritis & Rheumatism, vol. 31, no. 3, pp. 315–324, 1988.
[56]  M. L. L. Prevoo, M. A. van 't Hof, H. H. Kuper, M. A. van Leeuwen, L. B. A. van de Putte, and P. L. C. M. van Riel, “Modified disease activity scores that include twenty-eight-joint counts: development and validation in a prospective longitudinal study of patients with rheumatoid arthritis,” Arthritis & Rheumatism, vol. 38, no. 1, pp. 44–48, 1995.
[57]  M. L. L. Prevoo, A. M. van Gestel, M. A. van 't Hof, M. H. van Rijswijk, L. B. A. van de Putte, and P. L. C. M. van Riel, “Remission in a prospective study of patients with rheumatoid arthritis. American Rheumatism Association preliminary remission criteria in relation to the disease activity score,” British Journal of Rheumatology, vol. 35, no. 11, pp. 1101–1105, 1996.
[58]  O. Steinbrocker, C. H. Traeger, and R. C. Batterman, “Therapeutic criteria in rheumatoid arthritis,” Journal of the American Medical Association, vol. 140, no. 8, pp. 659–662, 1949.
[59]  D. M. van der Heijde, P. L. van Riel, I. H. Nuver-Zwart, F. W. Gribnau, and L. B. van de Putte, “Effects of hydroxychloroquine and sulphasalazine on progression of joint damage in rheumatoid arthritis,” The Lancet, vol. 1, no. 8646, pp. 1036–1038, 1989.
[60]  E. V. Tchetina, A. R. Poole, E. M. Zaitseva, et al., “Differences in Mammalian target of rapamycin gene expression in the peripheral blood and articular cartilages of osteoarthritic patients and disease activity,” Arthritis, vol. 2013, Article ID 461486, 14 pages, 2013.
[61]  K. J. Livak, “Comparative Ct method,” User Bulletin no. 2: ABI PRISM 7700 Sequence Detection System, PE Applied Biosystems, Foster City, Calif, USA, 1997.
[62]  C. L. Verweij and S. Vosslamber, “New insight in the mechanism of action of rituximab: the interferon signature towards personalized medicine,” Discovery Medicine, vol. 12, no. 64, pp. 229–236, 2011.
[63]  Z. Brkic, E. D. Olthof, H. A. Drexhage, and M. A. Versnel, “Monocyte gene expression signatures in rheumatic diseases: biomarkers for disease activity and tools for diagnosis and classification,” The Open Rheumatology Journal, vol. 3, pp. 13–17, 2010.
[64]  A. H. Gerards, S. de Lathouder, E. R. de Groot, B. A. C. Dijkmans, and L. A. Aarden, “Inhibition of cytokine production by methotrexate. Studies in healthy volunteers and patients with rheumatoid arthritis,” Rheumatology, vol. 42, no. 10, pp. 1189–1196, 2003.
[65]  M.-E. Miranda-Carús, A. Balsa, M. Benito-Miguel, C. Pérez De Ayala, and E. Martín-Mola, “Rheumatoid arthritis synovial fluid fibroblasts express TRAIL-R2 (DR5) that is functionally active,” Arthritis & Rheumatism, vol. 50, no. 9, pp. 2786–2793, 2004.
[66]  I. Vázquez, E. Graell, J. Gratacós et al., “Prognostic markers of clinical remission in early rheumatoid arthritis after two years of DMARDs in a clinical setting,” Clinical and Experimental Rheumatology, vol. 25, no. 2, pp. 231–238, 2007.
[67]  M. H. Y. Ma, F. Ibrahim, D. Walker et al., “Remission in early rheumatoid arthritis: predicting treatment response,” Journal of Rheumatology, vol. 39, no. 3, pp. 470–475, 2012.
[68]  J. S. Smolen, D. M. F. M. van der Heijde, E. W. St Clair et al., “Predictors of joint damage in patients with early rheumatoid arthritis treated with high-dose methotrexate with or without concomitant infliximab: results from the ASPIRE trial,” Arthritis & Rheumatism, vol. 54, no. 3, pp. 702–710, 2006.
[69]  P. Emery, M. C. Genovese, A. F. Kavanaugh, S. B. Cohen, J. L. Perez, and E. H. Sasso, “Adalimumab plus methotrexate results in less frequent and less severe radiographic progression than methotrexate alone at all levels of clinical response in early rheumatoid arthritis,” Annals of the Rheumatic Diseases, vol. 65, supplement 2, p. 88, 2006.
[70]  K. Forslind, M. Ahlmén, K. Eberhardt, I. Hafstr?m, and B. Svensson, “Prediction of radiological outcome in early rheumatoid arthritis in clinical practice: role of antibodies to citrullinated peptides (anti-CCP),” Annals of the Rheumatic Diseases, vol. 63, no. 9, pp. 1090–1095, 2004.
[71]  S. Mori, J. Hirose, and K. Yonemura, “Contribution of HLA-DRB1*04 alleles and anti-cyclic citrullinated antibodies to development of resistance to disease-modifying antirheumatic drugs in early rheumatoid arthritis,” Clinical Rheumatology, vol. 29, no. 12, pp. 1357–1366, 2010.
[72]  N. Yoshimura, Y. Ohmoto, H. Yasui et al., “The direct effect of FK506 and rapamycin on interleukin 1(β) and immunoglobulin production in vitro,” Transplantation, vol. 57, no. 12, pp. 1815–1818, 1994.
[73]  K. Migita, K. Eguchi, T. Aoyagi et al., “The effects of the immunosuppressant rapamycin on the growth of rheumatoid arthritis (RA) synovial fibroblast,” Clinical and Experimental Immunology, vol. 104, no. 1, pp. 86–91, 1996.
[74]  T. Sugatani and K. A. Hruska, “Akt1/Akt2 and mammalian target of rapamycin/Bim play critical roles in osteoclast differentiation and survival, respectively, whereas Akt is dispensable for cell survival in isolated osteoclast precursors,” Journal of Biological Chemistry, vol. 280, no. 5, pp. 3583–3589, 2005.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133