Review
Glucocorticoids in the control of neuroinflammation

https://doi.org/10.1016/j.mce.2007.03.007Get rights and content

Abstract

Glucocorticoids are a class of steroid hormones that are endowed with profound anti-inflammatory and immunosuppressive activities. Endogenous glucocorticoids are key players in the modulation of the immune system and establish an endocrine basis of many inflammatory diseases. In addition, synthetic glucocorticoids are amongst the most commonly prescribed drugs worldwide for the treatment of autoimmune disorders. In this review we summarize our present knowledge on the mechanisms by which glucocorticoids impact on multiple sclerosis (MS), a highly prevalent neuroinflammatory disease, and its animal model experimental autoimmune encephalomyelitis (EAE). In spite of the new methodologies that have become available during recent years, we are still far from a comprehensive picture of the mechanism by which glucocorticoids control neuroinflammation.

Introduction

Glucocorticoids (GCs) are a class of steroid hormones that are commonly used to treat acute and chronic inflammatory disorders (Tuckermann et al., 2005). They exert most of their functions by binding to the glucocorticoid receptor (GR), which controls gene expression and signalling cascades through genomic as well as non-genomic mechanisms. By this means, GCs modulate the survival, differentiation, migration and various effector functions of leukocytes and other cell types and thereby impact both, innate and adaptive immunity. Whilst GCs administered at pharmacological concentrations are considered purely immunosuppressive, fluctuations in the levels of endogenous GCs, e.g. during stress, result in more complex immunomodulatory effects (Elenkov and Chrousos, 1999).

One major application of GCs is the treatment of neuroinflammatory disorders, in particular multiple sclerosis (MS), the most prevalent chronic autoimmune disease of the central nervous system (CNS) in the Western world (Noseworthy et al., 2000). MS is characterized by infiltrating autoreactive T-cells in the CNS that initiate an inflammatory cascade involving leukocytes and humoral components (Sospedra and Martin, 2005). This causes demyelination and axonal loss, which eventually leads to severe functional deficits. While GCs have no positive effect on the long-term prognosis of MS, optic neuritis and other acute relapses are widely treated with high doses of methylprednisolone (Milligan et al., 1987). Still, such a therapy may lead to severe complications or incomplete recovery. Furthermore, it is known that the course of MS is influenced by the level of endogenous GCs (Elenkov and Chrousos, 1999). Women in the third trimester of pregnancy and Cushing syndrome patients often experience remission of the disease. This is explained by a polarization of the immune system towards TH2 dominated responses as a consequence of increased cortisol synthesis. Thus, fluctuations in GC production impact the course of neuroinflammatory diseases such as MS.

Whilst the importance of GCs for the control of neuroinflammation is doubtless, the mechanism at work is still unclear. Presently the pro-apoptotic function of GCs, their ability to impact migration and extravasation through the blood–brain barrier (BBB) and various influences on the effector functions of immune cells are discussed (Reichardt et al., 2006). While clinical studies have made important contributions to solve these questions, new insight has mainly been achieved using experimental autoimmune encephalomyelitis (EAE), a widely recognized animal model for MS that reflects many hallmarks of the human disease (Gold et al., 2006). It will be important in the future to continue along this line to improve our understanding of the mechanism of GCs in the control of neuroinflammation.

Section snippets

Molecular basis of glucocorticoid actions

The GR, a member of the nuclear receptor superfamily, is involved in the control of a plethora of physiological processes ranging from energy homeostasis and cognitive functions to the modulation of the immune system (Mangelsdorf et al., 1995). In the absence of GCs, the GR is sequestered in a multimeric complex in the cytoplasm. Once GCs have entered the cell, the GR is released and translocates into the nucleus (Beato et al., 1995). After binding to DNA, it drives transcription from so-called

Multiple sclerosis and its animal model experimental autoimmune encephalomyelitis

MS was first described in 1868 by the French neurologist Jean Martin Charcot who observed an accumulation of inflammatory cells in the CNS of patients suffering from periodic episodes of neurological dysfunction (Hafler, 2004). In 1933, Thomas Rivers showed that symptoms similar to the ones seen in MS patients could be induced by repeated injection of rabbit brain homogenate into primates without a need of infectious agents (Rivers et al., 1933). This marks the beginning of the long history of

Induction of apoptosis by glucocorticoids

The phenomenon of GC-induced T cell apoptosis has been recognized almost 30 years ago but the exact mechanism is not yet fully understood (Screpanti et al., 1989). The presence of the GR, DNA-binding dependent transcription and de novo gene expression is required but none of the identified target genes such as the pro-apoptotic proteins Bim and PUMA has turned out to be essential (Erlacher et al., 2005, Reichardt et al., 1998, Wang et al., 2006). Signal transduction events initiated in T cells

Side effects of glucocorticoid therapy

Despite the unsurpassed efficacy of GCs in the treatment of MS, such therapies are accompanied by multiple side effects. These comprise mood disorders, gastrointestinal pain and headache, while chronic administration may additionally lead to osteoporosis, diabetes and an increased susceptibility to infectious diseases (Pozzilli et al., 2004). Importantly, these side effects depend on the duration, dose and combination with other therapeutic agents and complicate MS therapy to a not yet fully

Conclusions and future directions

Despite the long history of GC research and the wide usage of these drugs in the clinical management of MS there are still many open questions. Firstly, which molecular mechanisms underlie the effects of GCs in MS and how could this knowledge be exploited to develop new drugs with an improved therapeutic profile? Also the cellular basis of GC effects continues to attract attention, e.g. which cell-types are the predominant targets of GCs in MS therapy and which of these effects are most

Acknowledgements

This work was supported by grants form the DFG (RE1631/1-3) and the Gemeinnützige Hertie-Stiftung (1.01.1/06/010).

References (84)

  • P.A. McCombe et al.

    Corticosteroid treatment of experimental autoimmune encephalomyelitis in the Lewis rat results in loss of V beta 8.2+ and myelin basic protein-reactive cells from the spinal cord, with increased total T-cell apoptosis but reduced apoptosis of V beta 8.2+ cells

    J. Neuroimmunol.

    (1996)
  • D.M. Mirowska-Guzel et al.

    High dose of intravenously given glucocorticosteroids decrease IL-8 production by monocytes in multiple sclerosis patients treated during relapse

    J. Neuroimmunol.

    (2006)
  • J. Navarro et al.

    Circulating dendritic cells subsets and regulatory T-cells at multiple sclerosis relapse: differential short-term changes on corticosteroids therapy

    J. Neuroimmunol.

    (2006)
  • C. Paul et al.

    Inhibition of blood–brain barrier disruption in experimental allergic encephalomyelitis by short-term therapy with dexamethasone or cyclosporin A

    Int. J. Immunopharmacol.

    (1995)
  • M.P. Pender et al.

    Apoptosis of alpha beta T lymphocytes in the nervous system in experimental autoimmune encephalomyelitis: its possible implications for recovery and acquired tolerance

    J. Autoimmun.

    (1992)
  • C. Pozzilli et al.

    Corticosteroids treatment

    J. Neurol. Sci.

    (2004)
  • H.M. Reichardt et al.

    DNA binding of the glucocorticoid receptor is not essential for survival

    Cell

    (1998)
  • I.A. Romero et al.

    Changes in cytoskeletal and tight junctional proteins correlate with decreased permeability induced by dexamethasone in cultured rat brain endothelial cells

    Neurosci. Lett.

    (2003)
  • T. Smith et al.

    Apoptosis of T cells and macrophages in the central nervous system of intact and adrenalectomized Lewis rats during experimental allergic encephalomyelitis

    J. Autoimmun.

    (1996)
  • I.H. Song et al.

    Non-genomic glucocorticoid effects to provide the basis for new drug developments

    Mol. Cell. Endocrinol.

    (2006)
  • T.L. Sorensen et al.

    Multiple sclerosis: a study of CXCL10 and CXCR3 co-localization in the inflamed central nervous system

    J. Neuroimmunol.

    (2002)
  • H.Y. Wang et al.

    Immune parameters associated with early treatment effects of high-dose intravenous methylprednisolone in multiple sclerosis

    J. Neurol. Sci.

    (2003)
  • J.C. Webster et al.

    Mouse glucocorticoid receptor phosphorylation status influences multiple functions of the receptor protein

    J. Biol. Chem.

    (1997)
  • R.A. Adams et al.

    The fibrin-derived γ377–395 peptide inhibits microglia activation and suppresses relapsing paralysis in central nervous system autoimmune disease

    J. Exp. Med.

    (2007)
  • W.Y. Almawi et al.

    Regulation of cytokine and cytokine receptor expression by glucocorticoids

    J. Leukoc. Biol.

    (1996)
  • B. Bartholome et al.

    Membrane glucocorticoid receptors (mGCR) are expressed in normal human peripheral blood mononuclear cells and up-regulated after in vitro stimulation and in patients with rheumatoid arthritis

    FASEB J.

    (2004)
  • R.W. Beck et al.

    A randomized, controlled trial of corticosteroids in the treatment of acute optic neuritis

    N. Engl. J. Med.

    (1992)
  • A. Ben-Nun et al.

    The rapid isolation of clonable antigen-specific T lymphocyte lines capable of mediating autoimmune encephalomyelitis

    Eur. J. Immunol.

    (1981)
  • C. Bjartmar et al.

    Axonal and neuronal degeneration in multiple sclerosis: mechanisms and functional consequences

    Curr. Opin. Neurol.

    (2001)
  • J.R. Chan et al.

    Glucocorticoids and progestins signal the initiation and enhance the rate of myelin formation

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

    (1998)
  • X. Chen et al.

    Differential response of murine CD4+ CD25+ and CD4+ CD25− T cells to dexamethasone-induced cell death

    Eur. J. Immunol.

    (2004)
  • X. Chen et al.

    Glucocorticoid amplifies IL-2-dependent expansion of functional FoxP3(+)CD4(+)CD25(+) T regulatory cells in vivo and enhances their capacity to suppress EAE

    Eur. J. Immunol.

    (2006)
  • J. Correale et al.

    Steroid hormone regulation of cytokine secretion by proteolipid protein-specific CD4+ T cell clones isolated from multiple sclerosis patients and normal control subjects

    J. Immunol.

    (1998)
  • R. Diem et al.

    Methylprednisolone increases neuronal apoptosis during autoimmune CNS inflammation by inhibition of an endogenous neuroprotective pathway

    J. Neurosci.

    (2003)
  • R. Diem et al.

    Combined therapy with methylprednisolone and erythropoietin in a model of multiple sclerosis

    Brain

    (2005)
  • I. Elovaara et al.

    Effect of high-dose methylprednisolone treatment on CCR5 expression on blood cells in MS exacerbation

    Acta Neurol. Scand.

    (2006)
  • I. Elovaara et al.

    Methylprednisolone reduces adhesion molecules in blood and cerebrospinal fluid in patients with MS

    Neurology

    (1998)
  • I. Elovaara et al.

    Adhesion molecules in multiple sclerosis: relation to subtypes of disease and methylprednisolone therapy

    Arch. Neurol.

    (2000)
  • B. Engelhardt

    Molecular mechanisms involved in T cell migration across the blood–brain barrier

    J. Neural. Transm.

    (2006)
  • P. Fontoura et al.

    Emerging therapeutic targets in multiple sclerosis

    Curr. Opin. Neurol.

    (2006)
  • C. Förster et al.

    Occludin as direct target for glucocorticoid-induced improvement of blood–brain barrier properties in a murine in vitro system

    J. Physiol.

    (2005)
  • C. Förster et al.

    Glucocorticoid effects on mouse microvascular endothelial barrier permeability are brain specific

    J. Physiol.

    (2006)
  • Cited by (0)

    View full text