Transcriptional therapy with the histone deacetylase inhibitor trichostatin A ameliorates experimental autoimmune encephalomyelitis

https://doi.org/10.1016/j.jneuroim.2005.02.022Get rights and content

Abstract

We demonstrate that the histone deacetylase (HDAC) inhibitor drug trichostatin A (TSA) reduces spinal cord inflammation, demyelination, neuronal and axonal loss and ameliorates disability in the relapsing phase of experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis (MS). TSA up-regulates antioxidant, anti-excitotoxicity and pro-neuronal growth and differentiation mRNAs. TSA also inhibits caspase activation and down-regulates gene targets of the pro-apoptotic E2F transcription factor pathway. In splenocytes, TSA reduces chemotactic, pro-Th1 and pro-proliferative mRNAs. A transcriptional imbalance in MS may contribute to immune dysregulation and neurodegeneration, and we identify HDAC inhibition as a transcriptional intervention to ameliorate this imbalance.

Introduction

Multiple sclerosis (MS) is a Th1 cytokine-driven inflammatory, demyelinating and neurodegenerative disease of the central nervous system (CNS) (Peterson et al., 2001). Neuronal or axonal injury may account for disability in MS and can occur in the absence of overt inflammation (Trapp et al., 1999). Mechanisms that have been implicated in MS pathogenesis include oxidative stress, excitotoxicity, autoimmunity and hormonal imbalance. Therapies that target these distinct pathophysiologic events have been shown to be effective in experimental autoimmune encephalomyelitis (EAE), a Th1 cytokine-driven model of MS. Based on these findings, some are being tested in humans (Youssef et al., 2002). However, drugs with preferential effects on the chronic relapsing (i.e., post-inflammation peak) phase of EAE, in which axonal loss correlates with disability (Wujek et al., 2002), have yet to be identified (Steinman, 1999).

Histone deacetylase (HDAC) enzymes repress genes via condensation of the nucleosome and interactions with transcription factors. For instance, HDACs bind repressor element 1 silencing transcription factor (REST), which is known to counteract neuronal differentiation traits (Ballas et al., 2001), and Sp1, another transcription factor that mediates neuronal antioxidant pathways (Ryu et al., 2003a, Ryu et al., 2003b). Compounds that inhibit HDAC enzymes range from nonspecific agents such as the short-chain fatty acid sodium phenylbutyrate (SPB), to highly specific hydroxamic acids such as trichostatin A (TSA). These structurally diverse agents induce histone hyperacetylation with concomitant up-regulation of neuronal maturation and antioxidant gene expression, indicating REST (Ballas et al., 2001, Hakimi et al., 2002) and Sp1 (Ryu et al., 2003a, Ryu et al., 2003b) transcription factor derepression.

E2F1 is a transcription factor that promotes immune cell proliferation (Wu et al., 2001). E2F1 activity is triggered by activation of T cells via the IL-2 receptor (IL-2R), and thus holds special relevance for MS and EAE, diseases driven by activated Th1 T cells. In fact, we have shown that peripheral blood mononuclear cells (PBMCs) from MS patients exhibit an activated E2F pathway signature (Iglesias et al., 2004), suggesting E2F as a potential therapeutic target in this disease. Accordingly, E2f1-deficient mice have less severe EAE and their splenocytes have decreased γ-IFN and elevated IL-4 responses to antigenic stimulation in vitro (Iglesias et al., 2004), suggesting a contribution by E2F1 to Th1 differentiation. E2F1 overexpression, in addition, contributes to neuronal apoptosis (Hou et al., 2000), further supporting our rationale for targeting the E2F pathway in MS.

We have previously shown that HDAC mRNAs are elevated in activated immune cells (Dangond et al., 1998), suggesting that they may serve as markers of activation and potential targets of treatment. We have also shown that TSA blocks immune cell proliferation and suppresses the pro-Th1 factor γ-IFN (Dangond and Gullans, 1998), in accordance with a growing literature suggesting that HDAC inhibitors cause a shift to a Th2 phenotype (Saemann et al., 2000). For instance, HDAC inhibitors down-regulate the CD28 costimulatory molecule (Moreira et al., 2003), the IL-2 receptor (Moreira et al., 2003) and IL-12 (p35 and p40) (Saemann et al., 2000) and up-regulate IFN-α and -β (Shuttleworth et al., 1983). Since MS is associated with transcriptional dysregulation (Iglesias et al., 2004), we tested whether the transcriptional drug TSA is clinically beneficial for EAE and whether it has neuroimmunoprotective effects.

Section snippets

EAE induction

For disease induction, we injected 6- to 8-week-old C57BL/6 female mice (Jackson Laboratory, Ann Harbor, ME) subcutaneously with 150 μg myelin oligodendrocyte glycoprotein (MOG)35–55 peptide in PBS and CFA containing 0.4 mg of Mycobacterium tuberculosis (H37Ra, Difco, Detroit, MI), and i.p. on days 1 and 3 with 200 ng Pertussis (List Biological, Campbell, CA), as described (Iglesias et al., 2004).

HDAC inhibitor dose

TSA (Biomol, Plymouth Meeting, PA and Wako, Richmond, VA) in PBS (9):DMSO(1) vehicle was

Clinical effects of TSA

Treatment of mice with TSA (7.5 mg/kg/day i.p.) on days 4–40 post-MOG injection resulted in a decrease in EAE disability measures during the chronic relapsing phase (Fig. 1a–b). The mean peak of the remission phase had a statistical significant reduction (P < 0.04) in TSA-treated mice. The disease index measured on day 30 and even on day 40, at the end-stage of the chronic relapsing phase, was also lower in TSA-treated mice than in vehicle-treated controls. TSA had no effects on the time of

Discussion

Herein, we show that the specific HDAC inhibitor TSA harnesses neuronal survival and anti-inflammatory pathways in EAE, resulting in clinical amelioration during the chronic relapsing phase. Several lines of evidence point to the potential use of HDAC inhibitors as transcriptional modulators in MS. We have previously shown that HDAC mRNAs are elevated in proliferating immune cells after stimulation with mitogens (Dangond et al., 1998), and HDAC inhibitors inhibit this proliferation (Dangond and

Acknowledgments

This work was supported by NIH 1KO8-CA80084 and R21-NS41623 (F.D.) and NIH NS045242, NS045806 and the Veterans Administration (R.J.F.). Thanks to the BWH GATC Center for technical support.

References (59)

  • H.S. Kim et al.

    Sodium butyrate suppresses interferon-gamma-, but not lipopolysaccharide-mediated induction of nitric oxide and tumor necrosis factor-alpha in microglia

    J. Neuroimmunol.

    (2004)
  • Y. Konishi et al.

    Cdc2 phosphorylation of BAD links the cell cycle to the cell death machinery

    Mol. Cell

    (2002)
  • K.A. Longo et al.

    Wnt signaling protects 3T3-L1 preadipocytes from apoptosis through induction of insulin-like growth factors

    J. Biol. Chem.

    (2002)
  • Y. Ma et al.

    Identification of novel E2F1-regulated genes by microarray

    Arch. Biochem. Biophys.

    (2002)
  • M. Polanczyk et al.

    The protective effect of 17beta-estradiol on experimental autoimmune encephalomyelitis is mediated through estrogen receptor-alpha

    Am. J. Pathol.

    (2003)
  • H. Sies et al.

    Glutathione peroxidase protects against peroxynitrite-mediated oxidations. A new function for selenoproteins as peroxynitrite reductase

    J. Biol. Chem.

    (1997)
  • L. Steinman

    Assessment of animal models for MS and demyelinating disease in the design of rational therapy

    Neuron

    (1999)
  • B. Beltran et al.

    Oxidative stress and S-nitrosylation of proteins in cells

    Br. J. Pharmacol.

    (2000)
  • J.G. Chang et al.

    Treatment of spinal muscular atrophy by sodium butyrate

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

    (2001)
  • S.P. Cregan et al.

    Apoptosis-inducing factor is involved in the regulation of caspase-independent neuronal cell death

    J. Cell Biol.

    (2002)
  • F. Dangond et al.

    Molecular signature of late-stage human ALS revealed by expression profiling of post-mortem spinal cord gray matter

    Physiol. Genomics

    (2004)
  • S. Dasgupta et al.

    Sodium phenylacetate inhibits adoptive transfer of experimental allergic encephalomyelitis in SJL/J mice at multiple steps

    J. Immunol.

    (2003)
  • R.J. Ferrante et al.

    Histone deacetylase inhibition by sodium butyrate chemotherapy ameliorates the neurodegenerative phenotype in Huntington's disease mice

    J. Neurosci.

    (2003)
  • A.R. Glabinski et al.

    Chemokine upregulation follows cytokine expression in chronic relapsing experimental autoimmune encephalomyelitis

    Scand. J. Immunol.

    (2003)
  • S.D. Gore et al.

    Modifying histones to tame cancer: clinical development of sodium phenylbutyrate and other histone deacetylase inhibitors

    Expert Opin. Investig. Drugs

    (2000)
  • J. Guy et al.

    Antioxidant enzymes reduce loss of blood–brain barrier integrity in experimental optic neuritis

    Arch. Ophthalmol.

    (1989)
  • M.A. Hakimi et al.

    A core-BRAF35 complex containing histone deacetylase mediates repression of neuronal-specific genes

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

    (2002)
  • S. Her et al.

    Regulation of the rat phenylethanolamine N-methyltransferase gene by transcription factors Sp1 and MAZ

    Mol. Pharmacol.

    (2003)
  • E. Hockly et al.

    Suberoylanilide hydroxamic acid, a histone deacetylase inhibitor, ameliorates motor deficits in a mouse model of Huntington's disease

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

    (2003)
  • Cited by (0)

    1

    These authors contributed equally to the work.

    View full text