Trends in Immunology
Volume 26, Issue 11, November 2005, Pages 572-579
Journal home page for Trends in Immunology

Autoimmunity special issue
SLE: translating lessons from model systems to human disease

https://doi.org/10.1016/j.it.2005.08.013Get rights and content

Systemic lupus erythematosus (SLE, lupus) results from immune-mediated damage to multiple organs. Its pathogenesis should be viewed as a series of steps, beginning with impaired immune regulation that permits self-reactive T–B-cell activation, which results in the production of autoantibodies. Activated T and B cells then infiltrate tissues, which along with autoantibody and immune complex deposition, triggering local events that ultimately cause organ damage. Although improved understanding of early autoimmune events might open up avenues for disease prevention, future investigations must focus on the mechanisms of end-organ damage in model systems and how to translate this knowledge into human disease. Understanding the mechanisms of each pathogenetic step would provide a rational basis for the development of disease stage-specific diagnostic markers and treatments.

Section snippets

The challenge

Systemic lupus erythematosus (SLE) is the most heterogeneous autoimmune disease that affects multiple organs [1]. The disease progresses through four broad stages, that is, the presence of autoantibodies against a variety of ubiquitous self-antigens, deposition of autoantibodies and immune complexes in tissues, development of tissue inflammation and finally, tissue damage and fibrosis. Although there has been a marked improvement in five-year survival from <50% in the 1950s to >90% in the

Heterogeneity of SLE: different animal models represent its various stages and subsets

Although clinical criteria have helped clinicians in making the diagnosis of SLE, the marked heterogeneity in disease expression has posed a difficulty in clearly defining the disease and formulating mechanistic investigations. Consequently, many investigators have turned toward animal models, which develop a homogeneous disease recapitulating the serological and histopathological features of SLE [4]. Examples of such models include the (NZBxNZW)F1 (BWF1), MRL-MpJ and NZM.2410 mouse strains.

Up

Tracing the steps of the pathogenesis of SLE

The clinical syndrome, known as human lupus, might actually encompass several diseases that have similar clinical features, yet with different mechanisms of pathogenesis. Here, we envision the natural course and pathogenesis of SLE as a series of steps that are narrated in the following working roadmap (Figure 3). It is hoped that a better understanding of this roadmap will facilitate the development of treatment strategies that can block the progression of disease at each step in most patients.

Step 1: insufficient immune regulation in SLE – harnessing the capacity of inhibitory or suppressor T cells to suppress lupus

Otherwise healthy, non-autoimmune mice can be induced to develop antibodies to DNA and mild nephritis by in vivo stimulation of the Th cells that are capable of promoting autoantibody production [9]. These animals, however, completely recover from such an episode of autoimmunity, despite persistent exposure to autoreactive Th cells. Recovery from disease in these mice correlates temporally with the appearance of certain CD8+ T, CD4+CD25+ T and natural killer T (NKT) cells that are capable of

Step 2: activation of autoreactive T and B cells

Strikingly, impairments in almost every step of the immune response occur in SLE. As narrated in the following substeps, vigorous attempts are underway to characterize these impairments in animal models and to translate this knowledge into human disease.

Step 3: autoantibodies can cause SLE lesions

Although patients with SLE have autoantibodies against several different specificities, they are usually restricted to a recurring set of autoantigens. Such restricted polyclonality might be related to a unique case of molecular mimicry, whereby variable regions of autoantibodies contain shared T-cell epitopes [56]. Thus, T cells stimulated by a peptide derived from one autoantibody can stimulate several different B cells that express the shared epitope 20, 56. The importance of these

Step 4: tissue inflammation and disease activity

Although ample evidence supports a pathogenic role for autoantibodies, it is unclear how they cause the myriad lesions of lupus. Multiple mechanisms have been proposed. For example, certain murine anti-DNA antibodies interact with antigens in glomerular basement membrane or hippocampal neurons, thus initiating death or dysfunction of local cells, such as podocytes or neurons, respectively 59, 60. The autoantibody and immune complex deposition triggers the activation of the complement cascade,

Step 5: tissue fibrosis and organ damage

Results of repeated kidney biopsies in SLE patients and longitudinal analysis of renal pathology in BWF1 mice suggest that lupus nephritis progresses generally from focal glomerular infiltration to diffuse proliferative glomerulonephritis to glomerulosclerosis and eventually to tubulo-interstitial fibrosis [65]. Some models and patients, however, have only localized renal inflammation 8, 9, 22, whereas others have marked inflammation but not much renal fibrosis, and still others have severe

Synthesis

Parallel developments in human disease observations and animal model investigations have helped in tracing some pathogenetic steps that lead to the manifestations of lupus. Some observations made in animal models are already being translated into human clinical trials. However, past experience has taught us that a rush to clinical trials must not occur without a full realization that the biological basis by which an intervention might suppress disease in an animal model might not be

Acknowledgements

I am supported by grants from the NIH/NIAMS and NIDDK (AR47322, DK69282 and AR50797). I thank Robert Kimberly sincerely for suggestions.

References (66)

  • R.W. Hoffman

    T cells in the pathogenesis of systemic lupus erythematosus

    Clin. Immunol.

    (2004)
  • L.A. Lee

    Transient autoimmunity related to maternal autoantibodies: neonatal lupus

    Autoimmun. Rev.

    (2005)
  • C. Kowal

    Cognition and immunity; antibody impairs memory

    Immunity

    (2004)
  • D.A. De Albuquerque

    An ACE inhibitor reduces Th2 cytokines and TGF-β1 and TGF-β2 isoforms in murine lupus nephritis

    Kidney Int.

    (2004)
  • L. Daniel

    Tubular lesions and tubular cell adhesion molecules for the prognosis of lupus nephritis

    Kidney Int.

    (2001)
  • Malaviya, A.N. et al. (1988) Systemic lupus erythematosus in northern India: a review of 329 cases. J. Assoc....
  • K.M. Uramoto

    Trends in the incidence and mortality of systemic lupus erythematosus, 1950–1992

    Arthritis Rheum.

    (1999)
  • B.S. Andrews

    Spontaneous murine lupus-like syndromes. Clinical and immunopathological manifestations in several strains

    J. Exp. Med.

    (1978)
  • A. Teubner

    Prevalence of circulating autoantibodies in healthy individuals

    Med. Klin.

    (2002)
  • R.R. Singh

    Induction of autoantibody production is limited in nonautoimmune mice

    J. Immunol.

    (2002)
  • R.R. Singh

    Differential contribution of IL-4 and STAT6 vs STAT4 to the development of lupus nephritis

    J. Immunol.

    (2003)
  • R.L. Brey

    Animal models for nervous system disease in systemic lupus erythematosus

    Ann. N. Y. Acad. Sci.

    (1997)
  • J.Q. Yang

    Repeated α-galactosylceramide administration results in expansion of NK T cells and alleviates inflammatory dermatitis in MRL-lpr/lpr mice

    J. Immunol.

    (2003)
  • J.Q. Yang

    CD1d deficiency exacerbates inflammatory dermatitis in MRL-lpr/lpr mice

    Eur. J. Immunol.

    (2004)
  • G. Filaci

    Impairment of CD8+ T suppressor cell function in patients with active systemic lupus erythematosus

    J. Immunol.

    (2001)
  • G.C. Fan et al.

    Vaccination with minigenes encoding V(H)-derived major histocompatibility complex class I-binding epitopes activates cytotoxic T cells that ablate autoantibody-producing B cells and inhibit lupus

    J. Exp. Med.

    (2002)
  • H.K. Kang

    Very low-dose tolerance with nucleosomal peptides controls lupus and induces potent regulatory T cell subsets

    J. Immunol.

    (2005)
  • J.Q. Yang

    Immunoregulatory role of CD1d in the hydrocarbon oil-induced model of lupus nephritis

    J. Immunol.

    (2003)
  • A.K. Singh

    The natural killer T cell ligand α-galactosylceramide prevents or promotes pristane-induced lupus in mice

    Eur. J. Immunol.

    (2005)
  • R.R. Singh et al.

    CD1d-reactive NKT cells inhibit autoreactive B cells

    Arthritis Rheum.

    (2004)
  • S. Kojo

    Dysfunction of T cell receptor AV24AJ18+, BV11+ double-negative regulatory natural killer T cells in autoimmune diseases

    Arthritis Rheum.

    (2001)
  • Y. Oishi

    Selective reduction and recovery of invariant Vα24JαQ T cell receptor T cells in correlation with disease activity in patients with systemic lupus erythematosus

    J. Rheumatol.

    (2001)
  • L. Bennett

    Interferon and granulopoiesis signatures in systemic lupus erythematosus blood

    J. Exp. Med.

    (2003)
  • Cited by (50)

    • Animal models in lupus

      2018, Dubois' Lupus Erythematosus and Related Syndromes
    • Culture promotes transfer of thyroid epithelial cell hyperplasia and proliferation by reducing regulatory T cell numbers

      2013, Cellular Immunology
      Citation Excerpt :

      IFN-γ inhibits TEC H/P both in vivo and in vitro [6], and inhibits TEC (thyroid epithelial cell) proliferation by upregulation of the cyclin-dependent kinase inhibitors p18 and p21 and downregulation of cyclin D [7]. Uncontrolled proliferation, hyperplasia and fibrosis of epithelial cells occurs in several autoimmune diseases including systemic lupus erythematosus, systemic sclerosis, rheumatoid arthritis, and autoimmune thyroiditis [8–10]. Thyroid autoimmunity and thyroid hyperplasia are very common [8,11,12] and can be associated with an increased risk of thyroid cancer [8,11,13].

    • Animal Models of SLE

      2013, Dubois' Lupus Erythematosus and Related Syndromes: Eighth Edition
    • Immune Tolerance Defects in Lupus

      2013, Dubois' Lupus Erythematosus and Related Syndromes: Eighth Edition
    • Administration of adenovirus encoding anti-CD20 antibody gene induces B-cell deletion and alleviates lupus in the BWF1 mouse model

      2011, International Immunopharmacology
      Citation Excerpt :

      Systemic lupus erythematosus (SLE) is a complex disease characterized by numerous autoantibodies and clinical involvement in multiple organ systems [1–3].

    • Definition of human autoimmunity - autoantibodies versus autoimmune disease

      2010, Autoimmunity Reviews
      Citation Excerpt :

      The deposit of circulating IC can cause glomerulonephritis, vasculitis and even arthritis. Tissue damage by IC deposits is a common mechanism in SLE [47], this is in part attributed to the presence of IgG against ubiquitous and abundant antigens. One of the most intriguing problems in the pathogenesis of AD is the presence of antibodies against intracellular antigens and their specific role in the immunological attack [48].

    View all citing articles on Scopus
    View full text