Elsevier

Pathophysiology

Volume 12, Issue 3, October 2005, Pages 167-181
Pathophysiology

Animal models of rheumatoid arthritis and their relevance to human disease

https://doi.org/10.1016/j.pathophys.2005.07.011Get rights and content

Abstract

Rodent models of rheumatoid arthritis (RA) are useful tools to study the pathogenic process of RA. Among the most widely used models of RA are the streptococcal cell wall (SCW) arthritis model and the collagen-induced arthritis (CIA). Both innate and adaptive immune mechanisms are involved in these rodent models. While no models perfectly duplicate the condition of human RA, they are easily reproducible, well defined and have proven useful for development of new therapies for arthritis, as exemplified by cytokine blockade therapies. Besides SCW and CIA models, there are numerous others including transgenic models such as K/BxN, induced models such as adjuvant-induced and pristane models, and spontaneous models in certain mouse strains, that have been used to help understand some of the underlying mechanisms. This review provides an update and analysis of RA models in mice and rats. The array of models has provided rheumatologists and immunologists a means to understand the multifactorial disease in humans, to identify new drug targets, and to test new therapies.

Introduction

Rheumatoid arthritis (RA) is a systemic inflammatory disorder that affects approximately 1% of the population worldwide. Even after decades of research, our understanding of the pathogenesis of the disease and the underlying mechanisms remains rudimentary. Clues to the etiology of RA reside in the preclinical stage where the initial triggering and induction events leading to disease take place. Unfortunately, this phase of human RA is not readily accessible to investigation and therefore remains speculative. The spectrum and disease progression of RA is governed by multiple factors including immune, genetic and environmental factors [1], [2]. In patients with clinically evident disease, RA is readily diagnosed by joint involvement and indices of immune activation and inflammation, however it remains a clinical challenge for Rheumatologists to effectively treat and manage the disease. Multiple components of immunity and inflammation play a role in established disease including T and B lymphocytes, neutrophils and monocytes, and vascular endothelium. Treatment thus typically requires multi-drug therapy. Therefore, a better understanding of the immune and genetic mechanisms during the early phases of RA is critical to the development of novel therapies that may one day lead to a cure of the disease.

The significance of understanding the mechanism of RA is exemplified by the recently developed anti-TNF blockade therapy, which has raised the bar for therapeutic effectiveness in the management of patients. Yet, the relative resistance of RA to various immunotherapeutic strategies remains an enigma and therefore a challenge to both basic scientists and Rheumatologists [2], [3], [4]. Rodent models of RA serve as valuable tools to investigate the underlying mechanisms at early, intermediate and late stages of RA [5], [6], [7]. With the recent advances in molecular biology, immunology, bioinformatics, and drug designing techniques, the possibility of developing novel therapies for RA and other inflammatory diseases is all the more promising.

The concern that no two arthritis models are alike and none is exactly like human RA, makes it all the more important to understand these models given the clinical observation that no two RA patients are exactly alike, and the etiologic stimulus (or stimuli) is unknown. This review encompasses various rodent models of RA that are used to understand the pathogenesis and pathophysiology of RA, and for the evaluation of therapies. Special emphasis has been placed on streptococcal cell wall (SCW) arthritis in rats and collagen-induced arthritis (CIA) in mice, two widely used animal models. The numerous models of RA available to-date have contributed to our understanding of the role of MHC and non-MHC genes, cell lineages involved, cell surface receptors, cytokines and chemokines, and vascular adhesion molecules that are involved in immunologic and inflammatory process leading to arthritis. The contributions and limitations of animal models are discussed in relation to human RA.

Section snippets

Animal models of rheumatoid arthritis

Rodent models of RA have been developed in both rats and mice. Other species have also been used over the years, however rodent models are most common, due to cost, homogeneity of the genetic background, and in mice, the capacity to use genetically modified strains. Most RA in animals is produced by treatment with an inducing agent, and even “spontaneous” models can be considered induced since they develop by the introduction or deletion of specific genes in animals with the proper immunologic

Mouse models of rheumatoid arthritis

Numerous approaches have been developed to induce arthritis in mice by immunization or injection. Genetically modified mouse strains have provided other, usually spontaneous, models of arthritis and are reviewed below. Collagen-induced arthritis is the best known induced model and is perhaps the easiest to appreciate conceptually, as it involves immunization with a known cartilage component. This model is discussed in more detail below. Antigen-induced arthritis (AIA) is another model, induced

Understanding mechanisms of arthritis from animal models

The immune mechanisms in the pathogenesis of RA have been difficult to understand given the inability to identify an initiating event. The putative mechanisms have evolved as our immunologic knowledge has expanded, and have varied with the trends in immunology. Throughout these developments in understanding human RA, animal models have played a key role in defining mechanisms and establishing some common features. They have also often challenged our cherished beliefs in what factors are

Concluding remarks

In this review we have attempted to cover several important animal models of RA that are widely used or provide important insights. In spite of decades of research, RA still remains a disease of unknown etiology and a complex disease without a single treatment plan. Of the many available animal models, CIA, SCW and more recently the K/BxN model have proven very useful in understanding some of the mechanisms involved. The spontaneous models of RA also appear to be useful, however, lack of a

Acknowledgements

We thank Dr. Shu Man Fu, Division of Rheumatology, University of Virginia Health System at Charlottesville; Dr. Robert Wolf, Rheumatology, Veterans Affairs Hospital, Shreveport, Louisiana; and Dr. Mathew Grisham, Department of Cellular and Molecular Physiology, LSUHSC, Louisiana, for fruitful discussions on this topic. Additional thanks are due to National Institutes of Health for grant support (NIH R01-AR-46976).

References (140)

  • Z. Szekanecz et al.

    Chemokines and chemokine receptors in rheumatoid arthritis

    Semin. Immunol.

    (2003)
  • D. Kontoyiannis et al.

    Impaired on/off regulation of TNF biosynthesis in mice lacking TNF AU-rich elements: implications for joint and gut-associated immunopathologies

    Immunity

    (1999)
  • B. Sadlack et al.

    Ulcerative colitis-like disease in mice with a disrupted interleukin-2 gene

    Cell

    (1993)
  • G.S. Firestein

    Evolving concepts of rheumatoid arthritis

    Nature

    (2003)
  • B. Bresnihan

    Pathogenesis of joint damage in rheumatoid arthritis

    J. Rheumatol.

    (1999)
  • M. Feldmann et al.

    Biological insights from clinical trials with anti-TNF therapy

    Springer Semin. Immunopathol.

    (1998)
  • R.O. Williams

    Rodent models of arthritis: relevance for human disease

    Clin. Exp. Immunol.

    (1998)
  • R.O. Williams

    Collagen-induced arthritis as a model for rheumatoid arthritis

    Methods Mol. Med.

    (2004)
  • R.O. Williams

    Models of rheumatoid arthritis

    Ernst Schering Res. Found. Workshop

    (2005)
  • D. Kyburz et al.

    The KRN mouse model of inflammatory arthritis

    Springer Semin. Immunopathol.

    (2003)
  • R. Horai et al.

    Development of chronic inflammatory arthropathy resembling rheumatoid arthritis in interleukin 1 receptor antagonist-deficient mice

    J. Exp. Med.

    (2000)
  • J. Keffer et al.

    Transgenic mice expressing human tumour necrosis factor: a predictive genetic model of arthritis

    EMBO J.

    (1991)
  • H.C. Stoerk et al.

    Chronic polyarthritis in rats injected with spleen in adjuvants

    Am. J. Pathol.

    (1954)
  • C.M. Pearson et al.

    Studies of polyarthritis and other lesions induced in rats given adjuvants

    Arthritis Rheum.

    (1959)
  • Billingham MEJ

    Adjuvant arthritis: the first model

  • B. Knight et al.

    Induction of adjuvant arthritis in mice

    Clin. Exp. Immunol.

    (1992)
  • P.H. Wooley

    Animal models of rheumatoid arthritis

    Curr. Opin. Rheumatol.

    (1991)
  • G.S. Geis

    Update on clinical developments with celecoxib, a new specific COX-2 inhibitor: what can we expect?

    Scand. J. Rheumatol. Suppl.

    (1999)
  • T. Goodson et al.

    The energy cost of adjuvant-induced arthritis in rats

    Arthritis Rheum.

    (2003)
  • M.M. Griffiths

    Immunogenetics of collagen-induced arthritis in rats

    Int. Rev. Immunol.

    (1988)
  • R.L. Wilder

    Streptococcal cell-wall-induced arthritis in rats: an overview

    Int. J. Tissue React.

    (1988)
  • R. Eisenberg et al.

    Measurement of bacterial cell wall in tissues by solid-phase radioimmunoassay: correlation of distribution and persistence with experimental arthritis in rats

    Infect. Immun.

    (1982)
  • R.L. Wilder

    Streptococcal Cell Wall Arthritis. Current Protocols in Immunology, vol. 3

    (1998)
  • A.E. Calogero et al.

    Neurotransmitter-induced hypothalamic–pituitary–adrenal axis responsiveness is defective in inflammatory disease-susceptible Lewis rats: in vivo and in vitro studies suggesting globally defective hypothalamic secretion of corticotropin-releasing hormone

    Neuroendocrinology

    (1992)
  • R.L. Wilder et al.

    Regulation of susceptibility to bacterial cell wall-induced arthritis in rats

    Arthritis Rheum.

    (1985)
  • X. Zhang et al.

    Characterisation of Eubacterium cell wall: peptidoglycan structure determines arthritogenicity

    Ann. Rheum. Dis.

    (2001)
  • S.A. Stimpson et al.

    Arthropathic properties of cell wall polymers from normal flora bacteria

    Infect. Immun.

    (1986)
  • D. Kimpel et al.

    Streptococcal cell wall induced arthritis: leukocyte activation in extra-articular lymphoid tissue

    Inflammation

    (2003)
  • D. Kimpel et al.

    Splenectomy attenuates streptococcal cell wall-induced arthritis and alters leukocyte activation

    Arthritis Rheum.

    (2003)
  • J.H. Schwab et al.

    Superantigen can reactivate bacterial cell wall-induced arthritis

    J. Immunol.

    (1993)
  • S.A. Stimpson et al.

    Lipopolysaccharide induces recurrence of arthritis in rat joints previously injured by peptidoglycan-polysaccharide

    J. Exp. Med.

    (1987)
  • M.F. van den Broek et al.

    Flare-up reaction of streptococcal cell wall induced arthritis in Lewis and F344 rats: the role of T lymphocytes

    Clin. Exp. Immunol.

    (1990)
  • S.N. Lichtman et al.

    Reactivation of arthritis induced by small bowel bacterial overgrowth in rats: role of cytokines, bacteria, and bacterial polymers

    Infect. Immun.

    (1995)
  • I. Rioja et al.

    Gene expression profiles in the rat streptococcal cell wall-induced arthritis model identified using microarray analysis

    Arthritis Res. Ther.

    (2005)
  • S.S. Makarov et al.

    Suppression of experimental arthritis by gene transfer of interleukin 1 receptor antagonist cDNA

    Proc. Natl. Acad. Sci. U.S.A.

    (1996)
  • V.A. Adarichev et al.

    Gene expression profiling in murine autoimmune arthritis during the initiation and progression of joint inflammation

    Arthritis Res. Ther.

    (2005)
  • M. Andersson et al.

    Analysis of type II collagen-reactive T cells in the mouse. I. Different regulation of autoreactive vs. non-autoreactive anti-type II collagen T cells in the DBA/1 mouse

    Eur. J. Immunol.

    (1990)
  • J.M. Stuart et al.

    Type II collagen-induced arthritis in rats. Passive transfer with serum and evidence that IgG anticollagen antibodies can cause arthritis

    J. Exp. Med.

    (1982)
  • L.K. Myers et al.

    T cell epitopes of type II collagen that regulate murine collagen-induced arthritis

    J. Immunol.

    (1993)
  • K. Terato et al.

    Collagen-induced arthritis in mice. Localization of an arthritogenic determinant to a fragment of the type II collagen molecule

    J. Exp. Med.

    (1985)
  • Cited by (197)

    View all citing articles on Scopus
    View full text