Open access peer-reviewed chapter

Cytokines in Rheumatoid Arthritis (RA)

Written By

Selim Nalbant and Ahmet Merih Birlik

Submitted: 14 May 2016 Reviewed: 20 September 2016 Published: 22 February 2017

DOI: 10.5772/65893

From the Edited Volume

New Developments in the Pathogenesis of Rheumatoid Arthritis

Edited by Lazaros I. Sakkas

Chapter metrics overview

2,544 Chapter Downloads

View Full Metrics

Abstract

Cytokines are cell molecules that are secreted by immune cells and aid cell to cell communication in immune responses and stimulate the movement of cells towards sites of inflammation, infection and trauma. So, the cytokines are the main part of the immune network to provide the communication in rheumatoid arthritis (RA) too. In RA, cytokines may be classified into four groups: pro-inflammatory cytokines, inflammatory cytokines in joints, anti-inflammatory cytokines and natural cytokine antagonists. After the initial stimuli have occurred, cytokines play a role in communication between the parts of immune system in every step of the pathophysiology process of RA. The differentiation of narve T cells into Th17 cells results in inflammation (synovitis) in joints. B cells further the pathogenic process through antigen presentation and autoantibody and cytokine production. The release of cytokines, especially tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1, causes synovial inflammation. In addition to their articular effects, pro-inflammatory cytokines promote the development of systemic effects (anemia, cardiovascular disease, fatigue and depression). So, cytokines are the main molecules contributing to all facets of the disease.

Keywords

  • cytokines
  • effector cells
  • rheumatoid arthritis
  • anti-cytokine treatment

1. Cytokine and rheumatoid arthritis

Rheumatoid arthritis (RA) is a progressive inflammatory disease, which is characterized by symmetrical polyarthritis. As an inflammatory disease, RA is characterized by increased levels in pro-inflammatory cytokines. In this complex cytokine environment, apart from arthritis, systemic manifestations also occur. Genetic and environmental factors are contributory to this complex nature of RA process. T cells, B cells and their cytokines play key roles in the pathophysiology of RA. Today’s modern RA treatment basically targets these cytokines. While we do this, we are generally treat the “normal and abnormal cytokine response” at the same time, because we do not know the main etiology of this abnormal cytokine response. To better manage RA, we should understand the role of cytokines and the relation between the effector cells [16].

The term “cytokine” is derived from a combination of two Greek words–“cyto” meaning cell and “kine” meaning move. Cytokines are cell molecules that are secreted by immune cells and aid cell to cell communication in cases of inflammation, infection and injury. So, the cytokines are vital part of the immune network to provide communication [7].

Advertisement

2. What is the origin of this complex cytokine response?

RA is an autoimmune disease. The term autoimmunity usually means a lot information that defines the way but not the origin of the process , and this stands for RA too. On the other hand, autoimmunity also means autoantibody production. In RA, there are two autoantibodies, rheumatoid factor and anti-cyclic citrullinated peptide antibodies, which contribute to inflammation. In the end, chronic inflammatory arthritis and the organ damage occur [8, 9].

The very early events of RA pathogenesis get into motion by breaking T-cell and/or B-cell tolerance and/or ignorance. However, this mechanism was poorly defined. After, shaping of this autoimmune background, some subsequent events intervene to perpetuate the process of synovial inflammation. What directs this process to the joints is unknown and it probably includes biomechanical factors, neuro-immuno-endocrinological interactions and altered articular microvascular microenvironment. Several factors have been proposed to have an association with the susceptibility and severity of rheumatoid arthritis [110].

These factors are as follows:

  1. Genetic loci:

    • HLA-DR4 alleles

    • PTPN22 (protein tyrosine phosphatase, nonreceptor type 22)

    • PADI4 (peptidyl arginine deiminase, type IV)

    • CTLA4 (cytotoxic T-lymphocyte antigen 4)

    • FcγRs (Fc receptors for IgG)

    • Various cytokine and cytokine-receptor loci.

  2. Environmental factors (smoking, stress, hormonal factors, etc.)

  3. Infectious organisms

The earliest event in RA pathogenesis perhaps is activation of the innate immune response. In this first step, cytokines play a role in communication between the parts of immune system. In second step, antigen-presenting cells present arthritis-associated antigens to T cells. This step is the starting point of the cytokine effect, which augments the inflammation and stimulate the other systems (such as lipid mediators, nitric oxide, RANKL-RANK signaling, etc.) that cause joint destruction and the organ damage [1113].

Advertisement

3. How can we classify this cytokine network in RA

Actually to make classification of cytokines for rheumatoid arthritis is not feasible. Because many of them have some pleotropic effect at the same time. However, we may basically, classify cytokines four groups in the pathogenesis of rheumatoid arthritis. Actually, this classification is not a real one, it is just for the establishment of understanding the whole mechanism [10] ( Table 1 ).

1. Pro-inflammatory cytokines: IL-1 and TNF-alpha cytokine role in IL-1
Increased synovial fibroblast cytokine, chemokine, MMP and PG release
Increased monocyte cytokine, reactive oxygen intermediate and PG release
Osteoclast activation
Endothelial cell adhesion molecule expression
Acute-phase protein production
Cardiovascular disease promotion
HPA axis dysregulation (fatigue and depression) [2, 14, 15]
TNF-alpha
Increased monocyte activation, cytokine release, prostaglandin release
T-cell clonal regulation
Increased endothelial cell adhesion molecule expression, cytokine release
Acute-phase protein production and fatigue-depression [2, 16]
2. Inflammatory cytokines in joints: IL-1 and TNF-alpha, IL-6, IL-15, IL-16, IL-17, IL-18, IFN-gamma, granulocyte macrophage-colony stimulating factor
IL-6
Osteoclast and B-cell activation
T-cell proliferation and differentiation
Acute-phase protein and hepcidin (anemia) production [17, 18]
IL-15
Structural similarities to IL-2,4 produced primarily by macrophages
Regulation of synovial inflammation [19-21]
IL-16
Suppression of IFN-γ, TNF-α and IL-1β expression
Anti-inflammatory effect by regulation of Tregs [22]
IL-17
Increase local chemokine production
Augmentation of immune response (increase IL-6 production)
Cartilage damage
Promotes the effect of IL-1b, TNF-a and IFN-g [20, 23, 24]
IL-18
Increase the production of pro-inflammatory cytokines, chemokines, adhesion molecules and RANKL which are the main molecules of joint destruction
Increase the production of fibroblast-like synoviocytes and chondrocytes [25, 26]
IL-21
Activate TH17 cells
Induces osteoclastogenesis
Plasma levels shows correlation with DAS28 [2]
IFN-gamma
Immune modulation (both protection and activation) [27, 28]
Granulocyte macrophage-colony stimulating factor (GM-CSF)
Promotes existing RA [29]
3. Anti-inflammatory cytokines
IL-10
Inhibit Th1 cell activity by suppressing IFN-γ expression
Direct inhibitory effect on the macrophage activity in the synovium
Elevated levels in the synovial fluid
Dominant suppressive cytokine effect
Protection against cartilage destruction combination with IL-4 [2, 7]
IL-4
Increased level in synovial fluid during only synovial inflammation
Preventing collagen type I breakdown in RA [3033]
IL-13
Synergistic or inhibitory roles during the arthritis with IL-10, IL-21R, galectin-3 and TGFβ [34]
IL-20
Regulates osteoclast differentiation [35]
4-Natural cytokine antagonists
IL-1 receptor antagonist (IL-1ra)
Low levels of IL-1 receptor antagonist (IL-1ra) causes erosive disease in patients and [36]
?Soluble type 2 IL-1 receptor
Cause competitive inhibition by binding interleukin-1α (IL1A), interleukin-1β (IL1B) and interleukin 1 receptor antagonist (IL1Ra), and acts as a pseudo receptor activity that inhibits the activity of its ligands [37, 38]
?Soluble TNF receptor (sTNF-RI)
It is not well-known; possible effect is to cause cleavage of TNF alpha [39]
IL-18 binding protein
Protect against the joint destructive effect by binding IL-18 in RA [40]

Table 1.

Actions of cytokines that play major roles in RA pathobiology.

3.1. Pro-inflammatory cytokines

Interleukin (IL)-1 and tumor necrosis factor (TNF)-alpha are the main pro-inflammatory cytokines involved in RA. The influx and/or local activation of mononuclear cells and the formation of new blood vessels are main findings in synovial membrane. Differentiation of naive T cells into Th17 cells contributes to synovitis. B cells further the pathogenic process through antigen presentation and autoantibody and cytokine production. Enzymes secreted by synoviocytes and chondrocytes degrade cartilage. The release of cytokines, especially TNF-alpha and IL-1, have multiple detrimental effects on cartilage and bone. Pro-inflammatory cytokines act locally but also have systemic effects, such as production of acute-phase proteins, anemia of chronic disease, cardiovascular disease, osteoporosis, etc. [17, 11, 12].

3.2. Inflammatory cytokines in joints

These cytokines are basically cytokine found in higher levels in the joints of patients with RA than in the serum. Most are pro-inflammatory cytokines. They cause mostly local joint destruction and also systemic effects of the disease and include IL-1, TNF-alpha, IL-6, IL-15, IL-16, IL-17, IL-18, interferon (IFN)-γ, granulocyte macrophage-colony stimulating factor) [1012].

3.3. Anti-inflammatory cytokines: (IL-4, IL-10, IL-11, IL-13 and IL-20)

In case of an inflammatory state, such as RA, the immune system utilizes anti-inflammatory cytokines to restrict the inflammatory reaction. In RA synovitis, there is an imbalance between pro-inflammatory and anti-inflammatory cytokines due to insufficient local concentrations of anti-inflammatory cytokines, IL-10, IL-11 and IL-13 to mediate counter-regulatory activity against the dominant pro-inflammatory cytokines. This is almost valid for the T-cell-derived cytokines, which are IL-2 and IL-4. They are also absent, which may impair Treg-cell generation and favor TH1-cell or TH17-cell immune responses [1, 2, 10].

3.4. Natural cytokine antagonists

Immune system is the most complex system, which has a self-limiting or self-controlling mechanism. The known molecules for this purpose are IL-1 receptor antagonist (IL-1ra), soluble type 2 IL-1 receptor, soluble TNF receptor (sTNF-RI) and IL-18 binding protein. However, current information about them is not entirely known. It is suggested that TNFR1 and the type II IL-1R have a regulatory role in sequestering soluble TNF and IL-1 away from their cell-bound receptors. Their detected levels in synovial tissues and fluid are insufficient to counteract the inflammatory cytokines and bring cytokine homeostasis. We currently use these natural anti-inflammatory molecules for therapeutic reasons [16].

Advertisement

4. Conclusion

As a result, cytokine effects do not occur with a single cytokine signalling cascade. There are many factors effecting the cytokine response. These are not only to control the equilibrium between inflammatory and anti-inflammatory cytokines but also the pleotropic and individual effects of cytokines. Here, we describe the topics of cytokine network only. It will be crucial to select cytokine targets based not on one single inflammatory pathway but rather on a biosystematic approach to pathogenesis. Implicit in this will be the recognition of pivotal checkpoints that facilitate the progression from autoimmunity to chronic inflammation [1, 1113].

As it was seen, cytokines are the main molecules at all these stages. However, they are not only the origin of the all cascade but also the last point which is occurring the damage. So, this fact makes the cytokine as a target molecule to treat.

References

  1. 1. Feldmann, M., Brennan, F. M. & Maini, R. N. Rheumatoid arthritis. Cell 1996; 85: 307–310.
  2. 2. Feldmann, M., Brennan, F. M. & Maini, R. N. Role of cytokines in rheumatoid arthritis. Annu. Rev. Immunol. 1996; 14: 397–440.
  3. 3. Firestein, G. S. Evolving concepts of rheumatoid arthritis. Nature 2003; 423: 356–361.
  4. 4. McInnes, I. B. & Liew, F. Y. Cytokine networks—towards new therapies for rheumatoid arthritis. Nat. Clin. Pract. Rheumatol 2005; 1: 31–39.
  5. 5. Venkatesha, S. H., Dudics, S., Acharya, B. & Kamal D. Moudgil. Cytokine-modulating strategies and newer cytokine targets for arthritis therapy. Int. J. Mol. Sci. 2015; 16: 887–906.
  6. 6. Ponchel, F., Goéb, V., Parmar, R., El-Sherbiny, Y., Boissinot, M., El Jawhari, J., Burska, A., Vital, E. M., Harrison, S., Conaghan, P. G., Hensor, E & Emery, P. An immunological biomarker to predict MTX response in early RA. Ann. Rheum. Dis. 2014; 73(11): 2047–2053.
  7. 7. Kasama, T., Isozaki, T., Takahashi, R. & Miwa, Y. Clinical effects of tocilizumab on cytokines and immunological factors in patients with rheumatoid arthritis. Int. Immunopharmacol. 2016; 35: 301–306.
  8. 8. Burmester, G. R. et al. Emerging cell and cytokine targets in rheumatoid arthritis. Nat. Rev. Rheumatol. 2014; 10: 77–88.
  9. 9. van der Helm-van Mil, A. H., Wesoly, J. Z. & Huizinga, T. W. Understanding the genetic contribution to rheumatoid arthritis. Curr. Opin. Rheumatol. 2005; 17: 299–304.
  10. 10. Choy, E. Understanding the dynamics: pathways involved in the pathogenesis of rheumatoid arthritis. Rheumatology 2012; 51: 3–11.
  11. 11. Jimenez-Boj, E. et al. Interaction between synovial inflammatory tissue and bone marrow in rheumatoid arthritis. J. Immunol. 2005; 175: 2579–2588.
  12. 12. Chabaud, M. et al. Human interleukin-17: a T cell-derived proinflammatory cytokine produced by the rheumatoid synovium. Arthritis Rheum. 1999; 42: 963–970.
  13. 13. Joosten, L. A. et al. IL-32, a proinflammatory cytokine in rheumatoid arthritis. Proc. Natl. Acad. Sci. 2006; 103: 3298–3303.
  14. 14. Dayer, J. M. & Bresnihan, B. Targeting interleukin-1 in the treatment of rheumatoid arthritis. Arthritis Rheum. 2002; 46: 574–578.
  15. 15. Horai, R. et al. Development of chronic inflammatory arthropathy resembling rheumatoid arthritis in interleukin 1 receptor antagonist-deficient mice. J. Exp. Med. 2000; 191: 313–320.
  16. 16. Redlich, K. et al. Osteoclasts are essential for TNF-α-mediated joint destruction. J. Clin. Invest. 2002; 110: 1419–1427.
  17. 17. Nishimoto, N. & Kishimoto, T. Interleukin 6: from bench to bedside. Nat. Clin. Pract. Rheumatol. 2006; 2: 619–626.
  18. 18. Alonzi, T. et al. Interleukin 6 is required for the development of collagen-induced arthritis. J. Exp. Med. 1998; 187: 461–468.
  19. 19. McInnes, I. B., Leung, B. P., Sturrock, R. D., Field, M. & Liew, F. Y. Interleukin-15 mediates T celldependent regulation of tumor necrosis factor-α production in rheumatoid arthritis. Nat. Med. 1997;3: 189–195.
  20. 20. Ferrari-Lacraz, S. et al. Targeting IL-15 receptor bearing cells with an antagonist mutant IL-15/Fc protein prevents disease development and progression in murine collagen-induced arthritis. J. Immunol. 2004; 173: 5818–5826.
  21. 21. Baslund, B. et al. Targeting interleukin-15 in patients with rheumatoid arthritis: a proof-of-concept study. Arthritis Rheum. 2005; 52: 2686–2692.
  22. 22. Suzuki, M., Konya, C., Goronzy, J. J. & Weyand, C. M. Inhibitory CD8+ T cells in autoimmune disease. Hum. Immunol. 2008; 69(11): 781–789.
  23. 23. Lubberts, E., Koenders, M. I. & van den Berg, W. B. The role of T-cell interleukin-17 in conducting destructive arthritis: lessons from animal models. Arthritis Res. Ther. 2005; 7: 29–37.
  24. 24. Weaver, C. T., Hatton, R. D., Mangan, P. R. & Harrington, L. E. IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annu. Rev. Immunol. 2007; 25: 821–852.
  25. 25. Gracie, J. A. et al. A proinflammatory role for IL-18 in rheumatoid arthritis. J. Clin. Invest. 1999; 104: 1393–1401.
  26. 26. McInnes, I. B., Liew, F. Y. & Gracie, J. A. Interleukin-18: a therapeutic target in rheumatoid arthritis? Arthritis Res. Ther. 2004; 7: 38–41.
  27. 27. Manoury-Schwartz, B. et al. High susceptibility to collagen-induced arthritis in mice lacking IFN-γ receptors. J. Immunol. 1997; 158: 5501–5506.
  28. 28. Vermeire, K. et al. Accelerated collagen-induced arthritis in IFN-γ receptor-deficient mice. J. Immunol. 1997; 158: 5507–5513.
  29. 29. Di Franco, M., Gerardi, M. C., Lucchino, B. & Conti, F. Mavrilimumab: an evidence based review of its potential in the treatment of rheumatoid arthritis. Core Evid. 2014; 9: 41–48.
  30. 30. Krabben A, Wilson AG, de Rooy DP, Zhernakova A, Brouwer E, Lindqvist E, Saxne T, Stoeken G, van Nies JA, Knevel R, Huizinga TW, Toes R, Gregersen PK, van der Helm-van Mil AH. Association of genetic variants in the IL4 and IL4R genes with the severity of joint damage in rheumatoid arthritis: a study in seven cohorts. Arthritis Rheum. 2013; 65: 3051–7.
  31. 31. Schulze-Koops, H. & Kalden, J. R. The balance of Th1/Th2 cytokines in rheumatoid arthritis. Best Pract. Res. Clin. Rheumatol. 2001; 15: 677–691.
  32. 32. Raza, K. et al. Early rheumatoid arthritis is characterized by a distinct and transientsynovial fluid cytokine profile of T cell and stromal cell origin. Arthritis Res. Ther. 2005; 7: R784–R795.
  33. 33. Nalbant, S., Koc, B., Top, C., Kucukardali, Y., Baykal, Y., Danaci, M. & Kocer, I. Hypersensitivity vasculitis and cytokines. Rheumatol. Int. 2002; 22(6): 244–248.
  34. 34. Steven J. Van Dyken & Richard M. Locksley. Interleukin-4-and interleukin-13-mediated alternatively activated macrophages: roles in homeostasis and disease. Annu. Rev. Immunol. 2013; 31: 317–343.
  35. 35. Hsu, Y. H. & Chang, M. S. IL-20 in rheumatoid arthritis. Drug Discov Today. 2015; 19. pii:S1359-6446(15)00312-8. doi:10.1016/j.drudis.2015.08.002 [Epub ahead of print]
  36. 36. Cantagrel, A., Navaux, F., Loubet-Lescoulié, P., Nourhashemi, F., Enault, G., Abbal, M., Constantin, A., Laroche, M. & Maziéres, B. Interleukin-1β, interleukin-1 receptor antagonist, interleukin-4, and interleukin-10 gene polymorphisms: relationship to occurrence and severity of rheumatoid arthritis. Arthritis Rheum. 1999; 42(6): 1093–1100.
  37. 37. McMahan, C. J., Slack, J. L., Mosley, B., Cosman, D., Lupton, S. D., Brunton, L.L., Grubin, C. E., Wignall, J. M., Jenkins, N. A. & Brannan, C. I. A novel IL-1 receptor, cloned from B cells by mammalian expression, is expressed in many cell types. EMBO J. 1991; 10(10): 2821–2832.
  38. 38. Dripps, D. J., Verderber, E., Ng, R. K., Thompson, R. C. & Eisenberg, S. P. Interleukin-1 receptor antagonist binds to the type II interleukin-1 receptor on B cells and neutrophils. J. Biol. Chem. 1991; 266(30): 20311–20315.
  39. 39. Galvani, V. Soluble tumor necrosis factor receptor I (sTNFRI) as a prognostic factor in melanoma patients in Slovene population Article Pflügers Archiv. Eur. J. Physiol. 2000; 440(5 Suppl): R61–R63.
  40. 40. Bresnihan, B., Roux-Lombard, P., Murphy, E., Kane, D., FitzGerald, O., Dayer, J. M. Serum interleukin 18 and interleukin 18 binding protein in rheumatoid arthritis. Ann. Rheum. Dis. 2002; 61: 726–729.

Written By

Selim Nalbant and Ahmet Merih Birlik

Submitted: 14 May 2016 Reviewed: 20 September 2016 Published: 22 February 2017