Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Chemokines in rheumatic diseases: pathogenic role and therapeutic implications

Abstract

Chemokines, a family of small secreted chemotactic cytokines, and their G protein-coupled seven transmembrane spanning receptors control the migratory patterns, positioning and cellular interactions of immune cells. The levels of chemokines and their receptors are increased in the blood and within inflamed tissue of patients with rheumatic diseases, such as rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis, vasculitis or idiopathic inflammatory myopathies. Chemokine ligand–receptor interactions control the recruitment of leukocytes into tissue, which are central to the pathogenesis of these rheumatic diseases. Although the blockade of various chemokines and chemokine receptors has yielded promising results in preclinical animal models of rheumatic diseases, human clinical trials have, in general, been disappointing. However, there have been glimmers of hope from several early-phase clinical trials that suggest that sufficiently blocking the relevant chemokine pathway might in fact have clinical benefits in rheumatic diseases. Hence, the chemokine system remains a promising therapeutic target for rheumatic diseases and requires further study.

Key points

  • Chemokines are a large family of secreted chemotactic cytokines that control the recruitment of immune cells into tissue and their cellular interactions once in tissue.

  • Chemokine receptors are G protein-coupled seven transmembrane spanning proteins that are expressed on immune cells and regulate their migration and cell–cell interactions.

  • Concentrations of chemokines are increased in the blood and tissues of patients with rheumatic diseases, suggesting their involvement in the pathogenesis of these diseases and highlighting them as therapeutic targets.

  • Preclinical animal models of rheumatic diseases show the important functional roles of the chemokine system in the pathogenesis of these diseases.

  • Unfortunately, the majority of clinical trials testing the efficacy of chemokine and chemokine receptor inhibitors have failed to show meaningful clinical benefit.

  • However, several clinical studies have shown promise and suggest that targeting the relevant chemokine system and ensuring complete inhibition at all times might be needed for therapeutic benefit.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Chemokines and chemokine receptors in RA.
Fig. 2: Chemokines and chemokine receptors in systemic lupus erythematosus.
Fig. 3: Chemokines and chemokine receptors in systemic sclerosis.
Fig. 4: Chemokines and chemokine receptors in giant cell arteritis.
Fig. 5: Chemokines and chemokine receptors in idiopathic inflammatory myositis.

Similar content being viewed by others

References

  1. Griffith, J. W., Sokol, C. L. & Luster, A. D. Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu. Rev. Immunol. 32, 659–702 (2014).

    CAS  PubMed  Google Scholar 

  2. Szekanecz, Z. & Koch, A. E. Successes and failures of chemokine-pathway targeting in rheumatoid arthritis. Nat. Rev. Rheumatol. 12, 5–13 (2016).

    CAS  PubMed  Google Scholar 

  3. Reynolds, J. A. et al. Cytokine profiling in active and quiescent SLE reveals distinct patient subpopulations. Arthritis Res. Ther. 20, 173 (2018).

    PubMed  PubMed Central  Google Scholar 

  4. Clark, K. E. et al. Multiplex cytokine analysis of dermal interstitial blister fluid defines local disease mechanisms in systemic sclerosis. Arthritis Res. Ther. 17, 73 (2015).

    PubMed  PubMed Central  Google Scholar 

  5. Berti, A. et al. Brief report: circulating cytokine profiles and antineutrophil cytoplasmic antibody specificity in patients with antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis Rheumatol. 70, 1114–1121 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Reed, A. M. et al. Changes in novel biomarkers of disease activity in juvenile and adult dermatomyositis are sensitive biomarkers of disease course. Arthritis Rheum. 64, 4078–4086 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. McInnes, I. B. & Schett, G. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med. 365, 2205–2219 (2011).

    CAS  PubMed  Google Scholar 

  8. Komano, Y. et al. Incidence and risk factors for serious infection in patients with rheumatoid arthritis treated with tumor necrosis factor inhibitors: a report from the Registry of Japanese Rheumatoid Arthritis Patients for Longterm Safety. J. Rheumatol. 38, 1258–1264 (2011).

    CAS  PubMed  Google Scholar 

  9. Ogata, A., Kato, Y., Higa, S. & Yoshizaki, K. IL-6 inhibitor for the treatment of rheumatoid arthritis: a comprehensive review. Mod. Rheumatol. 29, 258–267 (2019).

    CAS  PubMed  Google Scholar 

  10. Chang, M. H. & Nigrovic, P. A. Antibody-dependent and -independent mechanisms of inflammatory arthritis. JCI Insight 4, 125278 (2019).

    PubMed  Google Scholar 

  11. Monach, P. A., Mathis, D. & Benoist, C. The K/BxN arthritis model. Curr. Protoc. Immunol. 81, 15.22.1–15.22.12 (2008).

    Google Scholar 

  12. Brand, D. D., Latham, K. A. & Rosloniec, E. F. Collagen-induced arthritis. Nat. Protoc. 2, 1269–1275 (2007).

    CAS  PubMed  Google Scholar 

  13. Khachigian, L. M. Collagen antibody-induced arthritis. Nat. Protoc. 1, 2512–2516 (2006).

    CAS  PubMed  Google Scholar 

  14. Matsumoto, I., Staub, A., Benoist, C. & Mathis, D. Arthritis provoked by linked T and B cell recognition of a glycolytic enzyme. Science 286, 1732–1735 (1999).

    CAS  PubMed  Google Scholar 

  15. Kim, N. D., Chou, R. C., Seung, E., Tager, A. M. & Luster, A. D. A unique requirement for the leukotriene B4 receptor BLT1 for neutrophil recruitment in inflammatory arthritis. J. Exp. Med. 203, 829–835 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Nanki, T. et al. Pathogenic role of the CXCL16-CXCR6 pathway in rheumatoid arthritis. Arthritis Rheum. 52, 3004–3014 (2005).

    CAS  PubMed  Google Scholar 

  17. Meeuwisse, C. M. et al. Identification of CXCL13 as a marker for rheumatoid arthritis outcome using an in silico model of the rheumatic joint. Arthritis Rheum. 63, 1265–1273 (2011).

    CAS  PubMed  Google Scholar 

  18. Endo, H., Akahoshi, T., Takagishi, K., Kashiwazaki, S. & Matsushima, K. Elevation of interleukin-8 (IL-8) levels in joint fluids of patients with rheumatoid arthritis and the induction by IL-8 of leukocyte infiltration and synovitis in rabbit joints. Lymphokine Cytokine Res. 10, 245–252 (1991).

    CAS  PubMed  Google Scholar 

  19. Watanabe, K. et al. Pathogenic role of CXCR7 in rheumatoid arthritis. Arthritis Rheum. 62, 3211–3220 (2010).

    CAS  PubMed  Google Scholar 

  20. Yellin, M. et al. A phase II, randomized, double-blind, placebo-controlled study evaluating the efficacy and safety of MDX-1100, a fully human anti-CXCL10 monoclonal antibody, in combination with methotrexate in patients with rheumatoid arthritis. Arthritis Rheum. 64, 1730–1739 (2012).

    CAS  PubMed  Google Scholar 

  21. Nanki, T. et al. Migration of CX3CR1-positive T cells producing type 1 cytokines and cytotoxic molecules into the synovium of patients with rheumatoid arthritis. Arthritis Rheum. 46, 2878–2883 (2002).

    CAS  PubMed  Google Scholar 

  22. Koch, A. E. et al. Epithelial neutrophil activating peptide-78: a novel chemotactic cytokine for neutrophils in arthritis. J. Clin. Invest. 94, 1012–1018 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Pandya, J. M. et al. Blood chemokine profile in untreated early rheumatoid arthritis: CXCL10 as a disease activity marker. Arthritis Res. Ther. 19, 20 (2017).

    PubMed  PubMed Central  Google Scholar 

  24. Haringman, J. J., Smeets, T. J., Reinders-Blankert, P. & Tak, P. P. Chemokine and chemokine receptor expression in paired peripheral blood mononuclear cells and synovial tissue of patients with rheumatoid arthritis, osteoarthritis, and reactive arthritis. Ann. Rheum. Dis. 65, 294–300 (2006).

    CAS  PubMed  Google Scholar 

  25. Chou, R. C. et al. Lipid-cytokine-chemokine cascade drives neutrophil recruitment in a murine model of inflammatory arthritis. Immunity 33, 266–278 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Isozaki, T. et al. Evidence that CXCL16 is a potent mediator of angiogenesis and is involved in endothelial progenitor cell chemotaxis: studies in mice with K/BxN serum-induced arthritis. Arthritis Rheum. 65, 1736–1746 (2013).

    CAS  PubMed  Google Scholar 

  27. Zheng, B. et al. CXCL13 neutralization reduces the severity of collagen-induced arthritis. Arthritis Rheum. 52, 620–626 (2005).

    CAS  PubMed  Google Scholar 

  28. Jacobs, J. P. et al. Deficiency of CXCR2, but not other chemokine receptors, attenuates autoantibody-mediated arthritis in a murine model. Arthritis Rheum. 62, 1921–1932 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Yokoyama, W. et al. Abrogation of CC chemokine receptor 9 ameliorates collagen-induced arthritis of mice. Arthritis Res. Ther. 16, 445 (2014).

    PubMed  PubMed Central  Google Scholar 

  30. Barnes, D. A. et al. Polyclonal antibody directed against human RANTES ameliorates disease in the Lewis rat adjuvant-induced arthritis model. J. Clin. Invest. 101, 2910–2919 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Garcia-Vicuna, R. et al. CC and CXC chemokine receptors mediate migration, proliferation, and matrix metalloproteinase production by fibroblast-like synoviocytes from rheumatoid arthritis patients. Arthritis Rheum. 50, 3866–3877 (2004).

    CAS  PubMed  Google Scholar 

  32. Haringman, J. J., Ludikhuize, J. & Tak, P. P. Chemokines in joint disease: the key to inflammation? Ann. Rheum. Dis. 63, 1186–1194 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Takayasu, A. et al. CCL18 activates fibroblast-like synoviocytes in patients with rheumatoid arthritis. J. Rheumatol. 40, 1026–1028 (2013).

    CAS  PubMed  Google Scholar 

  34. Pickens, S. R. et al. Characterization of CCL19 and CCL21 in rheumatoid arthritis. Arthritis Rheum. 63, 914–922 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Nanki, T. et al. Inhibition of fractalkine ameliorates murine collagen-induced arthritis. J. Immunol. 173, 7010–7016 (2004).

    CAS  PubMed  Google Scholar 

  36. Shi, K. et al. Lymphoid chemokine B cell-attracting chemokine-1 (CXCL13) is expressed in germinal center of ectopic lymphoid follicles within the synovium of chronic arthritis patients. J. Immunol. 166, 650–655 (2001).

    CAS  PubMed  Google Scholar 

  37. Rump, L., Mattey, D. L., Kehoe, O. & Middleton, J. An initial investigation into endothelial CC chemokine expression in the human rheumatoid synovium. Cytokine 97, 133–140 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Miyabe, Y. et al. Complement C5a receptor is the key initiator of neutrophil adhesion igniting immune complex-induced arthritis. Sci. Immunol. 2, eaaj2195 (2017).

    PubMed  PubMed Central  Google Scholar 

  39. Koch, A. E. et al. Synovial tissue macrophage as a source of the chemotactic cytokine IL-8. J. Immunol. 147, 2187–2195 (1991).

    CAS  PubMed  Google Scholar 

  40. Li, J. L. et al. Neutrophils self-regulate immune complex-mediated cutaneous inflammation through CXCL2. J. Invest. Dermatol. 136, 416–424 (2016).

    CAS  PubMed  Google Scholar 

  41. Lee, J. H. et al. Pathogenic roles of CXCL10 signaling through CXCR3 and TLR4 in macrophages and T cells: relevance for arthritis. Arthritis Res. Ther. 19, 163 (2017).

    PubMed  PubMed Central  Google Scholar 

  42. Armas-Gonzalez, E. et al. Role of CXCL13 and CCL20 in the recruitment of B cells to inflammatory foci in chronic arthritis. Arthritis Res. Ther. 20, 114 (2018).

    PubMed  PubMed Central  Google Scholar 

  43. Moschovakis, G. L. et al. T cell specific CXCR5 deficiency prevents rheumatoid arthritis. Sci. Rep. 7, 8933 (2017).

    PubMed  PubMed Central  Google Scholar 

  44. Kraan, M. C. et al. The development of clinical signs of rheumatoid synovial inflammation is associated with increased synthesis of the chemokine CXCL8 (interleukin-8). Arthritis Res. 3, 65–71 (2001).

    CAS  PubMed  Google Scholar 

  45. Nanki, T. et al. Chemokine receptor expression and functional effects of chemokines on B cells: implication in the pathogenesis of rheumatoid arthritis. Arthritis Res. Ther. 11, R149 (2009).

    PubMed  PubMed Central  Google Scholar 

  46. Matsumoto, N. et al. A novel alpha9 integrin ligand, XCL1/lymphotactin, is involved in the development of murine models of autoimmune diseases. J. Immunol. 199, 82–90 (2017).

    CAS  PubMed  Google Scholar 

  47. Patterson, A. M. et al. Differential binding of chemokines to macrophages and neutrophils in the human inflamed synovium. Arthritis Res. 4, 209–214 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Ruth, J. H. et al. Selective lymphocyte chemokine receptor expression in the rheumatoid joint. Arthritis Rheum. 44, 2750–2760 (2001).

    CAS  PubMed  Google Scholar 

  49. Wengner, A. M. et al. CXCR5- and CCR7-dependent lymphoid neogenesis in a murine model of chronic antigen-induced arthritis. Arthritis Rheum. 56, 3271–3283 (2007).

    CAS  PubMed  Google Scholar 

  50. Manzo, A. et al. Mature antigen-experienced T helper cells synthesize and secrete the B cell chemoattractant CXCL13 in the inflammatory environment of the rheumatoid joint. Arthritis Rheum. 58, 3377–3387 (2008).

    CAS  PubMed  Google Scholar 

  51. Ruth, J. H. et al. CXCL16-mediated cell recruitment to rheumatoid arthritis synovial tissue and murine lymph nodes is dependent upon the MAPK pathway. Arthritis Rheum. 54, 765–778 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Petit, I., Jin, D. & Rafii, S. The SDF-1-CXCR4 signaling pathway: a molecular hub modulating neo-angiogenesis. Trends Immunol. 28, 299–307 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Ko, T. M. et al. CXCL10/IP-10 is a biomarker and mediator for Kawasaki disease. Circ. Res. 116, 876–883 (2015).

    CAS  PubMed  Google Scholar 

  54. Wang, C. R., Liu, M. F., Huang, Y. H. & Chen, H. C. Up-regulation of XCR1 expression in rheumatoid joints. Rheumatology 43, 569–573 (2004).

    CAS  PubMed  Google Scholar 

  55. Pickens, S. R. et al. Role of the CCL21 and CCR7 pathways in rheumatoid arthritis angiogenesis. Arthritis Rheum. 64, 2471–2481 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Chen, Z. et al. Characterising the expression and function of CCL28 and its corresponding receptor, CCR10, in RA pathogenesis. Ann. Rheum. Dis. 74, 1898–1906 (2015).

    CAS  PubMed  Google Scholar 

  57. Smith, E. et al. Duffy antigen receptor for chemokines and CXCL5 are essential for the recruitment of neutrophils in a multicellular model of rheumatoid arthritis synovium. Arthritis Rheum. 58, 1968–1973 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Patterson, A. M., Siddall, H., Chamberlain, G., Gardner, L. & Middleton, J. Expression of the Duffy antigen/receptor for chemokines (DARC) by the inflamed synovial endothelium. J. Pathol. 197, 108–116 (2002).

    CAS  PubMed  Google Scholar 

  59. Baldwin, H. M. et al. Elevated ACKR2 expression is a common feature of inflammatory arthropathies. Rheumatology 56, 1607–1617 (2017).

    CAS  PubMed  Google Scholar 

  60. Miyabe, Y., Miyabe, C., Mani, V., Mempel, T. R. & Luster, A. D. Atypical complement receptor C5aR2 transports C5a to initiate neutrophil adhesion and inflammation. Sci. Immunol. 4, eaav5951 (2019).

    CAS  PubMed  Google Scholar 

  61. Klimatcheva, E. et al. CXCL13 antibody for the treatment of autoimmune disorders. BMC Immunol. 16, 6 (2015).

    PubMed  PubMed Central  Google Scholar 

  62. Ogata, H., Takeya, M., Yoshimura, T., Takagi, K. & Takahashi, K. The role of monocyte chemoattractant protein-1 (MCP-1) in the pathogenesis of collagen-induced arthritis in rats. J. Pathol. 182, 106–114 (1997).

    CAS  PubMed  Google Scholar 

  63. Angelini, A. et al. Directed evolution of broadly crossreactive chemokine-blocking antibodies efficacious in arthritis. Nat. Commun. 9, 1461 (2018).

    PubMed  PubMed Central  Google Scholar 

  64. Kim, B. et al. JN-2, a C-X-C motif chemokine receptor 3 antagonist, ameliorates arthritis progression in an animal model. Eur. J. Pharmacol. 823, 1–10 (2018).

    CAS  PubMed  Google Scholar 

  65. Talbot, J. et al. CCR2 expression in neutrophils plays a critical role in their migration into the joints in rheumatoid arthritis. Arthritis Rheumatol. 67, 1751–1759 (2015).

    CAS  PubMed  Google Scholar 

  66. Amat, M. et al. Pharmacological blockade of CCR1 ameliorates murine arthritis and alters cytokine networks in vivo. Br. J. Pharmacol. 149, 666–675 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Brodmerkel, C. M. et al. Discovery and pharmacological characterization of a novel rodent-active CCR2 antagonist, INCB3344. J. Immunol. 175, 5370–5378 (2005).

    CAS  PubMed  Google Scholar 

  68. Vierboom, M. P. et al. Inhibition of the development of collagen-induced arthritis in rhesus monkeys by a small molecular weight antagonist of CCR5. Arthritis Rheum. 52, 627–636 (2005).

    CAS  PubMed  Google Scholar 

  69. Bonelli, M. et al. CCR6 controls autoimmune but not innate immunity-driven experimental arthritis. J. Cell Mol. Med. 22, 5278–5285 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Moschovakis, G. L. et al. The chemokine receptor CCR7 is a promising target for rheumatoid arthritis therapy. Cell Mol. Immunol. 16, 791–799 (2018).

    PubMed  Google Scholar 

  71. Min, S. H. et al. Pharmacological targeting reveals distinct roles for CXCR2/CXCR1 and CCR2 in a mouse model of arthritis. Biochem. Biophys. Res. Commun. 391, 1080–1086 (2010).

    CAS  PubMed  Google Scholar 

  72. Slauenwhite, D., Gebremeskel, S., Doucette, C. D., Hoskin, D. W. & Johnston, B. Regulation of cytokine polarization and T cell recruitment to inflamed paws in mouse collagen-induced arthritis by the chemokine receptor CXCR6. Arthritis Rheumatol. 66, 3001–3012 (2014).

    CAS  PubMed  Google Scholar 

  73. Haringman, J. J. et al. A randomized controlled trial with an anti-CCL2 (anti-monocyte chemotactic protein 1) monoclonal antibody in patients with rheumatoid arthritis. Arthritis Rheum. 54, 2387–2392 (2006).

    CAS  PubMed  Google Scholar 

  74. Haringman, J. J., Kraan, M. C., Smeets, T. J., Zwinderman, K. H. & Tak, P. P. Chemokine blockade and chronic inflammatory disease: proof of concept in patients with rheumatoid arthritis. Ann. Rheum. Dis. 62, 715–721 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Tak, P. P. et al. Chemokine receptor CCR1 antagonist CCX354-C treatment for rheumatoid arthritis: CARAT-2, a randomised, placebo controlled clinical trial. Ann. Rheum. Dis. 72, 337–344 (2013).

    CAS  PubMed  Google Scholar 

  76. Vergunst, C. E. et al. MLN3897 plus methotrexate in patients with rheumatoid arthritis: safety, efficacy, pharmacokinetics, and pharmacodynamics of an oral CCR1 antagonist in a phase IIa, double-blind, placebo-controlled, randomized, proof-of-concept study. Arthritis Rheum. 60, 3572–3581 (2009).

    CAS  PubMed  Google Scholar 

  77. Vergunst, C. E. et al. Modulation of CCR2 in rheumatoid arthritis: a double-blind, randomized, placebo-controlled clinical trial. Arthritis Rheum. 58, 1931–1939 (2008).

    CAS  PubMed  Google Scholar 

  78. van Kuijk, A. W. et al. CCR5 blockade in rheumatoid arthritis: a randomised, double-blind, placebo-controlled clinical trial. Ann. Rheum. Dis. 69, 2013–2016 (2010).

    PubMed  Google Scholar 

  79. Gerlag, D. M. et al. Preclinical and clinical investigation of a CCR5 antagonist, AZD5672, in patients with rheumatoid arthritis receiving methotrexate. Arthritis Rheum. 62, 3154–3160 (2010).

    CAS  PubMed  Google Scholar 

  80. Fleishaker, D. L. et al. Maraviroc, a chemokine receptor-5 antagonist, fails to demonstrate efficacy in the treatment of patients with rheumatoid arthritis in a randomized, double-blind placebo-controlled trial. Arthritis Res. Ther. 14, R11 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Tanaka, Y. et al. Safety, pharmacokinetics, and efficacy of E6011, an antifractalkine monoclonal antibody, in a first-in-patient phase 1/2 study on rheumatoid arthritis. Mod. Rheumatol. 28, 58–65 (2018).

    CAS  PubMed  Google Scholar 

  82. Lebre, M. C. et al. Why CCR2 and CCR5 blockade failed and why CCR1 blockade might still be effective in the treatment of rheumatoid arthritis. PLOS ONE 6, e21772 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Dairaghi, D. J. et al. Pharmacokinetic and pharmacodynamic evaluation of the novel CCR1 antagonist CCX354 in healthy human subjects: implications for selection of clinical dose. Clin. Pharmacol. Ther. 89, 726–734 (2011).

    CAS  PubMed  Google Scholar 

  84. Tsokos, G. C. Systemic lupus erythematosus. N. Engl. J. Med. 365, 2110–2121 (2011).

    CAS  PubMed  Google Scholar 

  85. Celhar, T. & Fairhurst, A. M. Modelling clinical systemic lupus erythematosus: similarities, differences and success stories. Rheumatology 56, i88–i99 (2017).

    CAS  PubMed  Google Scholar 

  86. Theofilopoulos, A. N. & Dixon, F. J. Murine models of systemic lupus erythematosus. Adv. Immunol. 37, 269–390 (1985).

    CAS  PubMed  Google Scholar 

  87. Perry, D., Sang, A., Yin, Y., Zheng, Y. Y. & Morel, L. Murine models of systemic lupus erythematosus. J. Biomed. Biotechnol. 2011, 271694 (2011).

    PubMed  PubMed Central  Google Scholar 

  88. Watson, M. L. et al. Genetic analysis of MRL-lpr mice: relationship of the Fas apoptosis gene to disease manifestations and renal disease-modifying loci. J. Exp. Med. 176, 1645–1656 (1992).

    CAS  PubMed  Google Scholar 

  89. Andrews, B. S. et al. Spontaneous murine lupus-like syndromes. Clinical and immunopathological manifestations in several strains. J. Exp. Med. 148, 1198–1215 (1978).

    CAS  PubMed  Google Scholar 

  90. Fang, C., Luo, T. & Lin, L. The correlational research among serum CXCL13 levels, circulating plasmablasts and memory B cells in patients with systemic lupus erythematosus: a STROBE-compliant article. Med. 96, e8675 (2017).

    CAS  Google Scholar 

  91. Worthmann, K. et al. Pathogenetic role of glomerular CXCL13 expression in lupus nephritis. Clin. Exp. Immunol. 178, 20–27 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Sun, X. et al. CXCL12/CXCR4/CXCR7 chemokine axis and cancer progression. Cancer Metastasis Rev. 29, 709–722 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Hrycek, E., Franek, A., Blaszczak, E., Dworak, J. & Hrycek, A. Serum levels of selected chemokines in systemic lupus erythematosus patients. Rheumatol. Int. 33, 2423–2427 (2013).

    CAS  PubMed  Google Scholar 

  94. Steinmetz, O. M. et al. CXCR3 mediates renal Th1 and Th17 immune response in murine lupus nephritis. J. Immunol. 183, 4693–4704 (2009).

    CAS  PubMed  Google Scholar 

  95. Ferreira, G. A., Teixeira, A. L. & Sato, E. I. Atorvastatin therapy reduces interferon-regulated chemokine CXCL9 plasma levels in patients with systemic lupus erythematosus. Lupus 19, 927–934 (2010).

    CAS  PubMed  Google Scholar 

  96. Balabanian, K. et al. Role of the chemokine stromal cell-derived factor 1 in autoantibody production and nephritis in murine lupus. J. Immunol. 170, 3392–3400 (2003).

    CAS  PubMed  Google Scholar 

  97. Menke, J. et al. CXCL9, but not CXCL10, promotes CXCR3-dependent immune-mediated kidney disease. J. Am. Soc. Nephrol. 19, 1177–1189 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Devarapu, S. K. et al. Reprint of “Dual blockade of the pro-inflammatory chemokine CCL2 and the homeostatic chemokine CXCL12 is as effective as high dose cyclophosphamide in murine proliferative lupus nephritis”. Clin. Immunol. 185, 119–127 (2017).

    CAS  PubMed  Google Scholar 

  99. Liao, X., Pirapakaran, T. & Luo, X. M. Chemokines and chemokine receptors in the development of lupus nephritis. Mediators. Inflamm. 2016, 6012715 (2016).

    PubMed  PubMed Central  Google Scholar 

  100. Furuichi, K. et al. Distinct expression of CCR1 and CCR5 in glomerular and interstitial lesions of human glomerular diseases. Am. J. Nephrol. 20, 291–299 (2000).

    CAS  PubMed  Google Scholar 

  101. Yoshimoto, S. et al. Elevated levels of fractalkine expression and accumulation of CD16+ monocytes in glomeruli of active lupus nephritis. Am. J. Kidney Dis. 50, 47–58 (2007).

    CAS  PubMed  Google Scholar 

  102. Nakatani, K. et al. Fractalkine expression and CD16+ monocyte accumulation in glomerular lesions: association with their severity and diversity in lupus models. Am. J. Physiol. Ren. Physiol. 299, F207–F216 (2010).

    CAS  Google Scholar 

  103. Biajoux, V. et al. Expression of CXCL12 receptors in B cells from Mexican Mestizos patients with systemic lupus erythematosus. J. Transl. Med. 10, 251 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Wu, X., Guo, J., Ding, R., Lv, B. & Bi, L. CXCL13 blockade attenuates lupus nephritis of MRL/lpr mice. Acta Histochem. 117, 732–737 (2015).

    CAS  PubMed  Google Scholar 

  105. Inoue, A. et al. Antagonist of fractalkine (CX3CL1) delays the initiation and ameliorates the progression of lupus nephritis in MRL/lpr mice. Arthritis Rheum. 52, 1522–1533 (2005).

    CAS  PubMed  Google Scholar 

  106. Bignon, A. et al. CCR1 inhibition ameliorates the progression of lupus nephritis in NZB/W mice. J. Immunol. 192, 886–896 (2014).

    CAS  PubMed  Google Scholar 

  107. Cheng, Q. et al. CXCR4-CXCL12 interaction is important for plasma cell homing and survival in NZB/W mice. Eur. J. Immunol. 48, 1020–1029 (2018).

    CAS  PubMed  Google Scholar 

  108. Ble, A. et al. Antiproteinuric effect of chemokine C-C motif ligand 2 inhibition in subjects with acute proliferative lupus nephritis. Am. J. Nephrol. 34, 367–372 (2011).

    CAS  PubMed  Google Scholar 

  109. Volkmann, E. R. & Varga, J. Emerging targets of disease-modifying therapy for systemic sclerosis. Nat. Rev. Rheumatol. 15, 208–224 (2019).

    PubMed  Google Scholar 

  110. Denton, C. P. & Ong, V. H. Targeted therapies for systemic sclerosis. Nat. Rev. Rheumatol. 9, 451–464 (2013).

    CAS  PubMed  Google Scholar 

  111. Yamamoto, T. Animal model of systemic sclerosis. J. Dermatol. 37, 26–41 (2010).

    CAS  PubMed  Google Scholar 

  112. Carulli, M. T., Handler, C., Coghlan, J. G., Black, C. M. & Denton, C. P. Can CCL2 serum levels be used in risk stratification or to monitor treatment response in systemic sclerosis? Ann. Rheum. Dis. 67, 105–109 (2008).

    CAS  PubMed  Google Scholar 

  113. Torok, K. S. et al. Peripheral blood cytokine and chemokine profiles in juvenile localized scleroderma: T-helper cell-associated cytokine profiles. Semin. Arthritis Rheum. 45, 284–293 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Distler, J. H., Akhmetshina, A., Schett, G. & Distler, O. Monocyte chemoattractant proteins in the pathogenesis of systemic sclerosis. Rheumatology 48, 98–103 (2009).

    CAS  PubMed  Google Scholar 

  115. Carulli, M. T. et al. Chemokine receptor CCR2 expression by systemic sclerosis fibroblasts: evidence for autocrine regulation of myofibroblast differentiation. Arthritis Rheum. 52, 3772–3782 (2005).

    CAS  PubMed  Google Scholar 

  116. Carvalheiro, T. et al. Increased frequencies of circulating CXCL10-, CXCL8- and CCL4-producing monocytes and Siglec-3-expressing myeloid dendritic cells in systemic sclerosis patients. Inflamm. Res. 67, 169–177 (2018).

    CAS  PubMed  Google Scholar 

  117. McCoy, S. S. et al. Scleroderma keratinocytes promote fibroblast activation independent of transforming growth factor beta. Rheumatology 56, 1970–1981 (2017).

    CAS  PubMed  Google Scholar 

  118. Hoffmann-Vold, A. M. et al. High level of chemokine CCL18 is associated with pulmonary function deterioration, lung fibrosis progression, and reduced survival in systemic sclerosis. Chest 150, 299–306 (2016).

    PubMed  Google Scholar 

  119. Lim, J. Y., Ryu, D. B., Lee, S. E., Park, G. & Min, C. K. Mesenchymal stem cells (MSCs) attenuate cutaneous sclerodermatous graft-versus-host disease (Scl-GVHD) through inhibition of immune cell infiltration in a mouse model. J. Invest. Dermatol. 137, 1895–1904 (2017).

    CAS  PubMed  Google Scholar 

  120. Tsou, P. S. et al. Scleroderma dermal microvascular endothelial cells exhibit defective response to pro-angiogenic chemokines. Rheumatology 55, 745–754 (2016).

    CAS  PubMed  Google Scholar 

  121. Rabquer, B. J. et al. Dysregulated expression of MIG/CXCL9, IP-10/CXCL10 and CXCL16 and their receptors in systemic sclerosis. Arthritis Res. Ther. 13, R18 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. van Bon, L. et al. Proteome-wide analysis and CXCL4 as a biomarker in systemic sclerosis. N. Engl. J. Med. 370, 433–443 (2014).

    PubMed  Google Scholar 

  123. Cossu, M. et al. Earliest phase of systemic sclerosis typified by increased levels of inflammatory proteins in the serum. Arthritis Rheumatol. 69, 2359–2369 (2017).

    CAS  PubMed  Google Scholar 

  124. Benyamine, A. et al. Increased serum levels of fractalkine and mobilisation of CD34+CD45– endothelial progenitor cells in systemic sclerosis. Arthritis Res. Ther. 19, 60 (2017).

    PubMed  PubMed Central  Google Scholar 

  125. Yamamoto, T. & Nishioka, K. Role of monocyte chemoattractant protein-1 and its receptor, CCR-2, in the pathogenesis of bleomycin-induced scleroderma. J. Invest. Dermatol. 121, 510–516 (2003).

    CAS  PubMed  Google Scholar 

  126. Arai, M. et al. Chemokine receptors CCR2 and CX3CR1 regulate skin fibrosis in the mouse model of cytokine-induced systemic sclerosis. J. Dermatol. Sci. 69, 250–258 (2013).

    CAS  PubMed  Google Scholar 

  127. Watts, R. A. & Scott, D. G. Recent developments in the classification and assessment of vasculitis. Best Pract. Res. Clin. Rheumatol. 23, 429–443 (2009).

    PubMed  Google Scholar 

  128. Mogi, M. & Liu, S. Animal models of vasculitis. Methods Mol. Biol. 1868, 223–232 (2018).

    CAS  PubMed  Google Scholar 

  129. Miyabe, C. et al. Am80, a retinoic acid receptor agonist, ameliorates murine vasculitis through the suppression of neutrophil migration and activation. Arthritis Rheum. 65, 503–512 (2013).

    CAS  PubMed  Google Scholar 

  130. Dallos, T. et al. CCL17/thymus and activation-related chemokine in Churg-Strauss syndrome. Arthritis Rheum. 62, 3496–3503 (2010).

    CAS  PubMed  Google Scholar 

  131. Brix, S. R. et al. CC chemokine ligand 18 in ANCA-associated crescentic GN. J Am. Soc. Nephrol. 26, 2105–2117 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Eriksson, P., Andersson, C., Cassel, P., Nystrom, S. & Ernerudh, J. Increase in Th17-associated CCL20 and decrease in Th2-associated CCL22 plasma chemokines in active ANCA-associated vasculitis. Scand. J. Rheumatol. 44, 80–83 (2015).

    CAS  PubMed  Google Scholar 

  133. Matsunawa, M. et al. Elevated serum levels of soluble CX3CL1 in patients with microscopic polyangiitis. Clin. Exp. Rheumatol. 27, 72–78 (2009).

    CAS  PubMed  Google Scholar 

  134. Blaschke, S., Brandt, P., Wessels, J. T. & Muller, G. A. Expression and function of the C-class chemokine lymphotactin (XCL1) in Wegener’s granulomatosis. J. Rheumatol. 36, 2491–2500 (2009).

    CAS  PubMed  Google Scholar 

  135. Savioli, B., Abdulahad, W. H., Brouwer, E., Kallenberg, C. G. M. & de Souza, A. W. S. Are cytokines and chemokines suitable biomarkers for Takayasu arteritis? Autoimmun. Rev. 16, 1071–1078 (2017).

    CAS  PubMed  Google Scholar 

  136. Dhawan, V., Mahajan, N. & Jain, S. Role of C-C chemokines in Takayasu’s arteritis disease. Int. J. Cardiol. 112, 105–111 (2006).

    PubMed  Google Scholar 

  137. Samson, M. et al. Involvement and prognosis value of CD8+ T cells in giant cell arteritis. J. Autoimmun. 72, 73–83 (2016).

    CAS  PubMed  Google Scholar 

  138. van Sleen, Y. et al. Involvement of monocyte subsets in the immunopathology of giant cell arteritis. Sci. Rep. 7, 6553 (2017).

    PubMed  PubMed Central  Google Scholar 

  139. Feng, S., Yadav, S. K., Gao, F. & Yi, Q. Plasma levels of monokine induced by interferon-gamma/chemokine (C-X-X motif) ligand 9, thymus and activation-regulated chemokine/chemokine (C-C motif) ligand 17 in children with Kawasaki disease. BMC Pediatr. 15, 109 (2015).

    PubMed  PubMed Central  Google Scholar 

  140. Stock, A. T., Hansen, J. A., Sleeman, M. A., McKenzie, B. S. & Wicks, I. P. GM-CSF primes cardiac inflammation in a mouse model of Kawasaki disease. J. Exp. Med. 213, 1983–1998 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Suzuki, C. et al. Non-receptor type, proline-rich protein tyrosine kinase 2 (Pyk2) is a possible therapeutic target for Kawasaki disease. Clin. Immunol. 179, 17–24 (2017).

    CAS  PubMed  Google Scholar 

  142. Samson, M. et al. Recent advances in our understanding of giant cell arteritis pathogenesis. Autoimmun. Rev. 16, 833–844 (2017).

    CAS  PubMed  Google Scholar 

  143. Martinez, H. G. et al. Important role of CCR2 in a murine model of coronary vasculitis. BMC Immunol. 13, 56 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Miyabe, C. et al. Dectin-2-induced CCL2 production in tissue-resident macrophages ignites cardiac arteritis. J. Clin. Invest. 130, 3610–3624 (2019).

    PubMed  Google Scholar 

  145. Jayne, D. R. W. et al. Randomized trial of C5a receptor inhibitor avacopan in ANCA-associated vasculitis. J. Am. Soc. Nephrol. 28, 2756–2767 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Sadik, C. D., Miyabe, Y., Sezin, T. & Luster, A. D. The critical role of C5a as an initiator of neutrophil-mediated autoimmune inflammation of the joint and skin. Semin. Immunol. 37, 21–29 (2018).

    CAS  PubMed  Google Scholar 

  147. Dalakas, M. C. Inflammatory muscle diseases. N. Engl. J. Med. 373, 393–394 (2015).

    CAS  PubMed  Google Scholar 

  148. Zhu, Z. et al. Altered chemokine receptor expression in the peripheral blood lymphocytes in polymyositis and dermatomyositis. Cytokine 99, 316–321 (2017).

    CAS  PubMed  Google Scholar 

  149. Malmstrom, V., Venalis, P. & Albrecht, I. T cells in myositis. Arthritis Res. Ther. 14, 230 (2012).

    PubMed  PubMed Central  Google Scholar 

  150. Rosenberg, N. L. & Kotzin, B. L. Aberrant expression of class II MHC antigens by skeletal muscle endothelial cells in experimental autoimmune myositis. J. Immunol. 142, 4289–4294 (1989).

    CAS  PubMed  Google Scholar 

  151. Sugihara, T. et al. A new murine model to define the critical pathologic and therapeutic mediators of polymyositis. Arthritis Rheum. 56, 1304–1314 (2007).

    CAS  PubMed  Google Scholar 

  152. Rosenberg, N. L., Ringel, S. P. & Kotzin, B. L. Experimental autoimmune myositis in SJL/J mice. Clin. Exp. Immunol. 68, 117–129 (1987).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. De Paepe, B., Creus, K. K. & De Bleecker, J. L. Role of cytokines and chemokines in idiopathic inflammatory myopathies. Curr. Opin. Rheumatol. 21, 610–616 (2009).

    PubMed  Google Scholar 

  154. Gono, T. et al. Cytokine profiles in polymyositis and dermatomyositis complicated by rapidly progressive or chronic interstitial lung disease. Rheumatology 53, 2196–2203 (2014).

    CAS  PubMed  Google Scholar 

  155. De Paepe, B., Creus, K. K. & De Bleecker, J. L. Chemokines in idiopathic inflammatory myopathies. Front. Biosci. 13, 2548–2577 (2008).

    PubMed  Google Scholar 

  156. Hak, A. E., de Paepe, B., de Bleecker, J. L., Tak, P. P. & de Visser, M. Dermatomyositis and polymyositis: new treatment targets on the horizon. Neth. J. Med. 69, 410–421 (2011).

    CAS  PubMed  Google Scholar 

  157. Suzuki, F. et al. Serum level of soluble CX3CL1/fractalkine is elevated in patients with polymyositis and dermatomyositis, which is correlated with disease activity. Arthritis Res. Ther. 14, R48 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Suzuki, F. et al. Inhibition of CX3CL1 (fractalkine) improves experimental autoimmune myositis in SJL/J mice. J. Immunol. 175, 6987–6996 (2005).

    CAS  PubMed  Google Scholar 

  159. Kim, J. et al. Therapeutic effect of anti-C-X-C motif chemokine 10 (CXCL10) antibody on C protein-induced myositis mouse. Arthritis Res. Ther. 16, R126 (2014).

    PubMed  PubMed Central  Google Scholar 

  160. De Paepe, B., Creus, K. K. & De Bleecker, J. L. Chemokine profile of different inflammatory myopathies reflects humoral versus cytotoxic immune responses. Ann. NY Acad. Sci. 1109, 441–453 (2007).

    PubMed  Google Scholar 

Download references

Acknowledgements

The work of Y.M. is supported by the Japanese Society for the Promotion of Science (JSPS) Kakenhi grant number JP19K08895, the Takeda Science Foundation, the Maruyama Memorial Research Foundation and the Medical Research Encouragement Prize of the Japan Medical Association. The work of A.D.L. is supported by grants from the National Institutes of Health and the Rheumatology Research Foundation.

Author information

Authors and Affiliations

Authors

Contributions

Y.M. researched data for this article. Y.M. and A.D.L. provided substantial contributions to discussions of content. Y.M., J.L., C.M. and A.D.L. wrote this article and reviewed and/or edited the manuscript before submission.

Corresponding author

Correspondence to Andrew D. Luster.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Rheumatology thanks A. Proudfoot, P. Proost and P.P. Tak for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Miyabe, Y., Lian, J., Miyabe, C. et al. Chemokines in rheumatic diseases: pathogenic role and therapeutic implications. Nat Rev Rheumatol 15, 731–746 (2019). https://doi.org/10.1038/s41584-019-0323-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41584-019-0323-6

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing