GET THE APP

Isoform-Selective HDAC Inhibition in Autoimmune Disease
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2014) Volume 0, Issue 0

Isoform-Selective HDAC Inhibition in Autoimmune Disease

Nicole L Regna1* and Christopher M Reillya2
1Department of Biomedical Sciences & Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
2Edward Via College of Osteopathic Medicine, Blacksburg, VA, 24060, USA
*Corresponding Author: Nicole L Regna, Center for Molecular Medicine and Infectious Diseases (CMMID), Virginia-Maryland Regional College of Veterinary Medicine, 1410 Prices Fork Road, Blacksburg, VA, 24061, USA, Tel: (434) 609-2703, Fax: (540) 231-3426 Email:

Abstract

Histone deacetylases are a class of enzymes that play an important role in protein modification and cellular function. Ongoing research suggests that HDAC inhibitors may be efficacious in the treatment of a wide range of diseases from cancer to autoimmune disease. HDACi therapy has shown promising results both in vitro and in vivo for the treatment of autoimmune disease. To date, 18 isoforms of HDACs have been identified, which exist in four different classes: class I (HDAC1, 2, 3, and 8), class II (HDAC4, 5, 6, 7, 9, and 10) class III (sirtuins1-7), and class IV (HDAC11). The mechanism of action through which HDACs function remains to be fully elucidated. However, the use of isoform-selective HDAC inhibitors has been helpful in determining the physiological role of individual HDACs as well as in decreasing the toxicity of HDACi therapy. This review will focus on isoform-selective HDACs and how they may be effective for the treatment of autoimmune disease.

Keywords: HDAC; Autoimmune disease; SLE

Introduction

Regulation of the immune system is dependent upon both genetic and epigenetic factors. Epigenetics control gene packaging and expression through heritable and stable changes without altering the DNA sequence [1,2]. These changes can be reversible dependent upon environmental factors and thus may provide the link between the environment and genetics that results in autoimmune disease [1]. Epigenetic changes in cellular function include changes in DNA methylation, microRNA (miRNA), and protein acetylation [3]. Proper cellular function requires acetylation of both histone and nonhistone proteins [4]. Abnormal histone acetyl transferase (HAT) and histone deacetylase (HDAC) expression and activity has been associated with a number of autoimmune and inflammatory diseases and may therefore be a potential target to therapeutically modulate disease [5-12].

HATs add an acetyl group to histone proteins allowing for transcriptional activities. Conversely, histone deacetylases (HDACs) are a group of enzymes that catalyze the removal of acetyl groups from lysine residues on histones thereby restricting chromatin availability for gene transcription [13,14]. Traditionally, HDACs were thought to function solely through epigenetic regulation of histone proteins; however, HDACs have more recently been shown to regulate acetylation of over 50 nonhistone proteins and may be more accurately described as lysine deacetylases (KDACs) [15,16]. Of particular interest is the ability of HDACs to regulate transcription factors, signaling molecules, and structural proteins thereby exhibiting an immunomodulatory effect [17].

HDACs have been implicated in immune cell regulation and may therefore be efficacious in the treatment of autoimmune disease [18,19]. Due to the large number of HDACs that are targeted, pan-HDAC inhibitors have been associated with deleterious side effects during clinical trials including fatigue, nausea, thrombocytopenia, and electrocardiograph abnormalities [20,21]. For this reason, a more targeted approach is warranted if HDAC inhibitors are to be used in the treatment of autoimmune disease. This review will discuss the potential use of isoform-selective HDAC inhibitors as therapeutics for autoimmune disease. Isoform-selective HDAC inhibitors may allow researchers to determine not only the biological functions of particular HDACs, but also provide a more specific target for potential therapeutics without adversely affecting normal physiological functions.

HDACs and Autoimmunity

There are 18 known mammalian HDACs, which are grouped into classes I-IV. The classical HDACs consist of HDACs 1-11, which are grouped into classes I, II, and IV [22]. Class III HDACs are comprised of 7 members called seven mammalian silent information regulator two proteins (sirtuins or Sirt) which differ from classical HDACs in that they require NAD+ as a cofactor and are not dependent upon Zn2+ as a catalytic mechanism [23,24]. HDACs are found in both the nucleus and cytoplasm, with some shuttling between the two and others confined to a specific compartment [25].

A nuclear localization signal (NLS) allows HDACs to localize within the nucleus and therefore exert their function on nuclear proteins. HDAC1 and 2 lack a nuclear export signal (NES) and are unable to leave the nucleus [22]. HDAC3 has both a NLS and a NES; however, it is almost always found within the nucleus [22,26]. Conversely, class II HDACs, particularly HDACs 4, 5, 7, 9, and 10, are known to travel back in forth between the nucleus and the cytoplasm and are thought to play an important role in the function of both nuclear and cytoplasmic proteins [22]. HDAC6 is predominantly found within the cytoplasm and mainly influences cytosolic proteins [27]. Similarly to class II HDACs, HDAC11 (class IV), can be found in both the nucleus and the cytoplasm and has been demonstrated to colocalize with HDAC6 in the cytoplasm [28]. Due to the specificity of HDACs, selective therapeutic targeting may allow for modulation of specific histones or other non-nuclear proteins.

Autoimmunity is characterized by an abnormal immune response during which the body perceives a normal substance as foreign leading to autoantibody production and inflammation [29]. Studies of monozygotic twins discordant for systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and dermatomyositis; suggest a role of non-genetic factors in disease pathogenesis [30,31]. HDAC inhibitors have been shown to modulate a number of key regulators of the immune system including B cells, T cells, and APCs [5,32-36]. During autoimmune disease it is thought that HDAC activity is upregulated leading to increased nuclear translocation and binding of the transcription factors, particularly STAT3 and NF-κB, which promote gene expression of pro-inflammatory genes [37]. HDAC inhibitors have proven to have an anti-inflammatory effect, which may be helpful in the treatment of autoimmune disease during which prolonged inflammation results in tissue destruction and organ failure [36,38].

Due to the anti-proliferative effect that HDAC inhibitors exhibit; they may be effective agents of immunosuppression for the treatment of autoimmune disease. Treatment with pan-HDAC inhibitors including ITF2357, SAHA, and TSA have shown efficacy in treating autoimmune diseases including SLE, RA, and inflammatory bowel disease (IBD) in murine models [34,36,39-43]. We have shown that treatment with ITF2357 is able to reduce disease in lupus-prone mice while increasing the number of Treg cells and decreasing the number of CD4+ T cells [34]. SLE is thought to involve aberrant B and T cell regulation [6,44-47]. Studies showing the regulatory effect of HDAC inhibitors on both B and T cell populations make selective HDACi therapy of particular interest in the treatment of SLE [34,40,48].

Selective HDAC inhibitors are able to provide a more targeted approach to treating autoimmune disease and reduce the risk of complications from unwanted side effects. Pan-HDAC and class I-selective inhibitors, currently undergoing clinical trials, alter physiological functions that require protein deacetylation [20,49]. There are currently two HDAC inhibitors, SAHA (pan-HDACi) and FK228 (selective-class I HDACi), approved by the FDA for the treatment of cutaneous T cell lymphoma (CTCL). Both of these drugs have also been tested for their efficacy in the treatment of autoimmune diseases. SAHA has shown efficacy in the treatment of lupus-prone mice; however, long term treatment resulted in unwanted side effects including possible drug toxicity [41]. SLE is a chronic disease requiring long-term treatment and these results indicate that inhibition of class I and II HDACs by a pan-HDACi may not be optimal [41].

Currently undergoing phase III clinical trials are panobinostat (LBH589) and Valproic acid (VPA) [50,51]. LBH589 is being tested for its use as a CTCL therapeutic and a number of other cancers [50]. VPA is currently in phase III clinical trials for the treatment of cervical and ovarian cancer, but it has recently shown therapeutic potential in the treatment of autoimmune disease [51,52]. HDAC inhibitors currently undergoing phase II clinical trials include Mocetinostat (MGCD0103), Entinostat (MS-275), Belinostat (PXD101), and Givinostat (ITF2357) for the treatment of various cancers [53]. CUDC-101, ACY-1215, CHR-2845, and CG200745 have begun phase I clinical trials for the treatments of cancer [54,55]. Dokamanovic et al. provide a more extensive review of specific HDAC inhibitors currently undergoing clinical trials [15].

Due to the ubiquitous nature of HDACs, not only are the cellular pathways involved with autoimmunity affected, but HDAC inhibition also disrupts the pathways involved with normal cellular function [24]. Furthermore, pan-HDAC inhibitors can be cytotoxic, and it may prove important in clinical treatment for HDAC inhibition to be more selective [56]. The mechanisms through which HDAC inhibitors regulate the immune response are not fully understood. Currently ongoing studies of HDAC inhibitors both in vivo and in vitro are working to determine the mechanism of both pan-and isoform-selective HDAC inhibitors.

Selective Class I Inhibitors

Class I HDACs (HDACs 1, 2, 3, and 8) play an important role in cell survival and proliferation [57]. While insight has been gained about the function of HDACs through various knockout mouse studies, gene deletion of HDACs 1, 2, and 3, has proven to be embryonic lethal in mice [24]. HDAC1 has been demonstrated to be overexpressed in SLE, RA, multiple sclerosis (MS), and juvenile idiopathic arthritis (JIA) [5]. Furthermore, HDAC3 and HDAC7 have also been shown to play a role in immune regulation during SLE, suggesting the potential importance of targeting these HDACs for treatment of disease [5].

In regard to class I HDACs it is interesting to note that HDAC2 is able to regulate the binding ability of p53, which controls transcription. HDAC2 has been shown to increase p53 binding activity and consequently increase cellular proliferation [58]. HDAC2 was demonstrated to be involved with an anti-apoptotic function following HDAC2 knockdown in cancer cells [59]. Furthermore, p53 activation has been linked to inhibition of autoimmune disease. Studies suggest that p53 expression is able to induce Treg differentiation leading to suppression of the autoimmune response [60]. P53 activation is further thought to inhibit autoimmune disease through downregulation of STAT1 resulting in decreased proinflammatory cytokine production [61]. Autoimmune diseases have been shown to be more severe on a p53-deficient background in mice [60,62]. The studies explain why targeting HDAC2 may be a viable approach for treating autoimmune diseases such as lupus in which Treg cell function may be important to modulate the immune response.

MS-275 is a benzamide-derived selective class I inhibitor currently undergoing Phase I-II clinical trials that has shown promising anti-rheumatic activities including prevention of bone erosion and delayed onset of collagen-induced arthritis (CIA) [24,63]. Studies have demonstrated MS-275 treatment suppressed LPS-induced pro-inflammatory cytokine production in monocytic cells. Treatment led to phase arrest at G0/G1 without increasing apoptosis [64]. Treatment with MS-275 after the onset of arthritis in rodents has been demonstrated to halt disease progression suggesting its potential as a therapeutic. Furthermore, MS-275 may have potential as a therapeutic in the treatment of other inflammatory autoimmune diseases based off of its anti-inflammatory effect in vitro and in the CIA induced mouse model [63,64]. Following treatment with MS-275, E11 cells and monocytic cells had decreased LPS-induced NF-κB nuclear translocation, decreased production of IL-6, IL-18, NO, VEGF, MMP-2, and MMP-9 [64]. Furthermore, MS-275 has been shown to decrease sera production of pro-inflammatory cytokines IL-6 and IL-1β, which are overproduced during a number of autoimmune diseases including RA, SLE, autoimmune encephalomyelitis, and IBD [65-71].

The exact mechanism through which MS-275 treatment results in anti-rheumatic and anti-inflammatory effects remains to be elucidated. One proposed mechanism suggests MS-275 increases the stability of histone acetylation associated with the c-Fos promoter which plays an important role in cellular functions including proliferation, differentiation and survival [72]. MS-275 has been shown to increase acetylation of NF-κB p65 leading to decreased nuclear translocation and inhibition of gene transcription [64]. NF-κB activation and nuclear translocation is required for c-Fos expression [72]. These studies suggest that inhibition of NF-κB nuclear accumulation by MS-275 treatment, results in decreased cellular proliferation of osteoclasts induced by c-Fos expression [72,73]. Furthermore, MS-275 has been shown to decrease the chaperone activity of Hsp90 [74]. The ability of MS-275 to inhibit Hsp90 is of particular interest in the treatment of SLE, which has been found to have elevated hsp90 sera levels [75]. Furthermore, use of an Hsp90 inhibitor in lupus-prone mice has shown therapeutic potential [76].

VPA is a selective class I HDACi, effective against HDACs 1-5 and HDAC 7, that has been used as a treatment for seizures and mental disorders [77]. More recently VPA has been tested for its efficacy in the treatment of autoimmune disease using the Fas-deficient MRL/MPJ-Faslpr/J (MRL/lpr-/-) mouse model. MRL/lpr-/- mice injected intraperitoneally with 500 mg/kg VPA for 8 weeks had decreased lymphoid organ weight and cellularity, decreased DN T cells in the spleen, lymph nodes, and blood, and a reduced number of WBCs, particularly lymphocytes, in the peripheral blood compared to vehicle-treated control mice. VPA treatment was found to induce caspase- dependent and independent apoptosis in PBMCs in vitro [52]. Furthermore, treatment of glomerulosclerosis in the adriamycin nephropathy mouse model with VPA reduced proteinuria in early phase renal disease [78]. VPA has been demonstrated to inhibit TNF-α, NF-κB, and IL-6 pathways, which have been shown to be dysregulated during many autoimmune diseases [78]. The mechanism of action for VPA in the treatment of autoimmune diseases has yet to be identified. However, treatment of ADR nephropathy with VPA was found to increase glomerular H3K9 acetylation and decrease glomerular apoptosis [78].

MGCD0103 is a selective class I HDAC (HDACs 1, 2, and 3) inhibitor that has also shown selectivity for HDAC11 and is currently undergoing phase I/II clinical trials [79]. MGCD0103 has been demonstrated to have antiproliferative activity in Hodgkin lymphoma cell lines and B-cell chronic lymphocytic leukemia [79-81]. Previous studies indicate that MGCD10103 increases caspase-dependent apoptosis while inhibiting autophagy through the activation of the PI3K/AKT/mTOR pathway [81,82]. Furthermore, MGD10103 increased NF-κB activation and resulted in increased TNF-α expression and production [80]. For these reasons, MGCD0103 may not be optimal for treatment for autoimmune diseases, including SLE and RA, which are characterized by increased PI3K/AKT/mTOR signaling and NF-κB activation [83-85].

Selective HDAC3 inhibition has also been explored for its use in treating inflammatory autoimmune disease. HDAC3 expression has been shown to be elevated in PBMCs from MS patients when compared to healthy controls [86]. MI192 is a selective HDAC3i that has been shown to regulate cytokine production from PBMCs. IL-6 production by PBMCs was decreased in a dose-dependent manner following treatment with MI192; however, the mechanism remains to be elucidated [87]. Studies indicate that overexpression of HDAC3 causes apoptotic-resistant autoreactive lymphocytes that contribute to autoimmune disease [86]. These data suggest that HDAC3 inhibition may be beneficial in the treatment of autoimmunity.

Romidepsin (Depsipeptide, FK288) is a selective HDACi of HDACs 1, 2, 3, and 4 currently undergoing clinical trials for the treatment of T cell lymphoma [88]. Treatment of autoantibody-mediated arthritis (AMA) mice with FK228 reduced inflammation, joint swelling, and bone destruction. Pro-inflammatory cytokines IL-1β and TNF-α were reduced following treatment with FK228 [89]. TNF-α is known to play an important role in the pathogenesis of a number of autoimmune diseases including SLE, RA, and Crohn’s disease and anti-TNF-α therapies have proven to be an effective clinical treatment for people with these diseases [38,90-93]. The molecular mechanism through which FK288 reduces inflammation has yet to be determined.

Class IIa HDAC Inhibitors

Class II HDACS are not as ubiquitous as class I, but they are still thought to be essential for regulatory functions of the cell. Class IIa HDACs include HDACs 4, 5, 7, and 9 [24]. Expression of class IIa HDACs is thought to be somewhat tissue specific with increased expression in the brain, muscle, and T lymphocytes [94]. While deletion of HDAC7 is embryonic lethal in mice, deletion of HDACs 4, 5, and 9 produce viable mice, but with defects in cellular hypertrophy, stress response, cardiovascular function, and bone development [24]. Studies suggest a role for class IIa HDACs (HDAC4, 5, and 7) in pro-inflammatory gene expression [95]. Given the pro-inflammatory environment associated with many autoimmune diseases, class IIa HDACs could serve as promising targets for autoimmune therapies. While class IIa HDACs are able to move back and forth between the nucleus and the cytoplasm, they are currently thought to have limited deacetylase function; rather functioning through the recruitment of HDAC3 [56,96].

HDAC9 has been found to be overexpressed in T cells from lupus patients and SLE murine models. HDAC9 deficient MRL/lpr mice had prolonged survival and decreased lymphproliferation, autoantibody production, inflammation, and kidney disease [97]. Furthermore, HDAC9 deficient mice have decreased colitis following dextran sodium sulfate (DSS) treatment compared to wild type (WT) mice [48]. HDAC9 deficiency increased site specific lysine histone acetylation of H3K9, H3K14, and H3K18 localized to IL-4, roquin, and PPAR-γ, respectively in MRL/lpr mice [97]. These results indicate that inhibition of HDAC9 may be able to decrease the inflammatory response through hyperacetylation and stabilization of IL-4 and PPAR-γ.

Inhibition of HDAC9 has also been associated with an increased Treg suppressive function, and studies have shown that HDAC9 is exported from the nucleus upon Treg activation [12,48]. These studies suggest that when located within the nucleus HDAC9 suppresses Foxp3 function and HDAC9 nuclear exportation is required for an effective Treg response [12,48]. SiRNA knockdown of HDAC9 in WT Tregs resulted in increased Foxp3 expression and enhanced Treg suppressive function in vitro. HDAC9 knockdown in Treg cells caused increased HSP70 expression; however, when HDAC9-/- Tregs were treated with triptolide (an HSP70 inhibitor) suppressive function was decreased to levels comparable by WT Tregs [48]. Similarly to HDAC9 inhibition, knockdown of HDAC7 increased Treg suppressive ability [98]. These data suggest the potential of inhibiting HDACs 7 and 9 with isoform-selective inhibitors to decrease autoimmune disease. Tregs function to suppress the proliferation of immune cell subsets and regulate cytokine production and the response to self-Ags. Furthermore, the maintenance of self-tolerance. Treg deficiency has been associated with a number of autoimmune diseases including SLE, MS, RA, and IBD [99-103]. Deletion of Tregs in animals has been demonstrated to cause autoimmunity [104-107].

Class IIb HDAC Inhibitors

Class IIb HDACs (HDAC6 and 10) are found in both the nucleus and the cytoplasm [24]. The role of HDAC10 has yet to be determined; however, HDAC6 has been shown to regulate acetylation of cytoplasmic and nuclear proteins as wells as deacetylase-independent functions. HDAC6 is thought to play an integral role in a number of cellular functions including regulation of the cytoskeleton, cell migration, and degradation of misfolded proteins through deacetylation of α-tubulin, HSP90, and cortacin [108-110].

During SLE, the number and function of Treg cells is diminished [111,112]. Pan-HDAC inhibitors have been shown to increase the number and suppressive effects of Tregs, but treatment with class-I specific HDAC inhibitors, such as MS-275, have been unable to produce the same result suggesting a role of class II HDACs [12,109]. Treatment with a specific HDAC6i leads to increased Treg function. Furthermore, Tregs from HDAC6 deficient mice have been demonstrated to have increased suppressive Treg function [109]. Treg cells from HDAC6-/- mice had a Treg effector/memory phenotype with decreased expression of CD44 and CD62L, but increased expression of CD103 [109]. Regulatory effector-memory T cells (TREM) are Treg cells capable of activation, expansion, and memory that function to control the immune response in inflamed tissues [113]. Furthermore, Tregs isolated from HDAC6 deficient mice had increased function in vitro suppressive of CFSE-labeled WT conventional T (Tcon) cells [109]. Similarly, treatment with the HDAC6 specific inhibitors tubacin and tubastatin A, resulted in increased suppression of in vitro proliferation of Tcon cells by Treg cells. Although Tubastatin A and tubacin inhibit HDAC6, tubacin is more selective for HDAC6 and may have greater efficacy at lower doses [114].

Crohn’s diseases and ulcerative colitis are two forms of IBD and are modeled by the DSS model of colitis. Similarly to other autoimmune diseases, IBD requires a genetic susceptibility coupled with environmental factors leading to an inflammatory response [115]. Studies have demonstrated treatment with tubacin is able to prevent weight loss and diarrhea in the DSS model of colitis in a Treg dependent fashion [109].

Another selective HDAC6i, ACY-738, has minimal reactivity against other class II HDACs and 100-fold less selectivity against class I HDACs [116]. ACY-738 was tested for its efficacy in the treatment of SLE in NZB/W mice. We found that HDAC6 inhibition with ACY-738 was able to decrease a number of hallmarks of SLE disease including splenomegaly, immune complex-mediated glomerulonephritis, and sera anti-dsDNA levels. ACY-738 treatment altered BM B cell differentiation by increasing the percentage of cells in the late pro-B cell and early pre-B cell fractions while decreasing the accumulation of cells in the late pre-B fraction F. Furthermore, ACY-738 also increased the percentage of Treg cells with a concomitant decrease in SLE-associated markers of disease (unpublished data). Studies have shown that treatment with ACY-738 (1 µM) increased the suppressive function of Tregs alone and in combination with a sirtuin1 inhibitor, Ex-527 [117].

Class IV HDAC Inhibitors

HDAC11 is the most recently identified member of HDAC proteins and is the sole member of class IV [24]. The role of HDAC11 in normal cell function still remains to be fully elucidated and no isoform-selective HDACi has yet been developed [118]. However, HDAC11 has been identified as a potential molecular target for the treatment of autoimmune disease due to its role as a negative transcriptional regulator of IL10 [119]. Overexpression of HDAC11 in a mouse macrophage cell line prevented an increase in IL10 mRNA expression following LPS-stimulation. Furthermore, knocking down HDAC11 using shRNA in human APCs resulted in an increase in expression of IL10 mRNA following immune stimulation. Given the role IL-10 plays in the induction of tolerance, these results suggest targeting HDAC11 in the treatment of autoimmune disease may be beneficial. IL-10 is an anti-inflammatory cytokine with wide-ranging effects from B cell stimulation to limiting the immune response and action of pro-inflammatory cytokines. Dysregulation of IL-10 production contributes to an increased risk for autoimmune diseases including SLE, IBD, and allergic asthma [120]. Specifically during SLE, high sera levels of IL-10 correlate with disease activity [121].

HDAC11 has also been identified as a potential target for regulating APC- mediated immune activation. Primary mouse macrophages overexpressing HDAC11 showed enhanced production of IL-2 and IFN-γ following clonotypic T cell encounter. Conversely, clonotypic T cells that were introduced to APCs with knocked down HDAC11 had reduced IL-2 and IFN-γ production [119].

Summary

Previous studies suggest a complex mechanism of action for HDAC inhibitors; the use of isoform-selective HDAC inhibitors will be helpful in determining the specific roles of individual HDACs. Questions remain about the long-term safety of HDAC inhibitor use for the treatment of chronic diseases. The identification of aberrant HDAC specific isoforms to each autoimmune disease may be important in reducing toxicity. Isoform-selective HDAC inhibition has the potential to correct aberrant immune regulation by altering the function of components of the inflammatory cascades without the deleterious side effects associated with traditional pan-HDAC inhibitors (Table 1).

Compound Isoform-Specificity Protein/enzyme/gene Cellular response Disease Reference
MS-275 HDACs 1,2,3,9 ↑Foxp3
↓nuclear NFκB p65, VEGF
↑IL-10
↓IL-1β, IFN-γ, IL-17, IL-18, TNF-α, IL-18, NO
RA [64,122]
MI192 HDAC3 ND ↓TNF, IL-6, IFN-γ RA [87]
MGCD0103 HDACs 1,2,3,11 Jak/STAT
↑NF-κB activation, TNFSF4, TNFSF9, TNF
TNFRSF8
↑TNF-α ND [79,80]
Valproic acid HDACs 1,2,3,8 PI3K/Akt, mTOR, NF-κB ↓TNF-α ALPS, SLE, IBD [52,78]
FK228 HDACs 1,2,3,4 ND ↓IL-1β, TNF-α AMA, RA, diabetes [20,88,89]
ACY-738 HDAC6 Foxp3 ↑TGF-β
↓IL-1β
SLE [117]
Unpublished data
Tubacin HDAC6 α-tubulin, HSp90 Foxp3
↑CTLA-4, PD-1, GITR
↓IL-2, IFN-γ
↑IL-10
RA, IBD [109,123]
Tubastatin A HDAC6 α-tubulin, Foxp3 ↓TNF-α, IL-6 RA, IBD [114,124]
ND: No data

Table 1: Isoform-Selective HDAC Inhibitors and Immune Regulation.

References

  1. Brooks WH, Le Dantec C, Pers JO, Youinou P, Renaudineau Y (2010) Epigenetics and autoimmunity. J Autoimmun 34: J207-219.
  2. Meda F, Folci M, Baccarelli A, Selmi C (2011) The epigenetics of autoimmunity. Cell Mol Immunol 8: 226-236.
  3. Portela A, Esteller M (2010) Epigenetic modifications and human disease. Nat Biotechnol 28: 1057-1068.
  4. Batta K, Das C, Gadad S, Shandilya J, Kundu TK (2007) Reversible acetylation of non histone proteins: role in cellular function and disease. Subcell Biochem 41: 193-212.
  5. Woan KV, Sahakian E, Sotomayor EM, Seto E, Villagra A (2012) Modulation of antigen-presenting cells by HDAC inhibitors: implications in autoimmunity and cancer. Immunol Cell Biol 90: 55-65.
  6. Reilly CM, Regna N, Mishra N (2011) HDAC inhibition in lupus models. Mol Med 17: 417-425.
  7. Leoni F, Fossati G, Lewis EC, Lee JK, Porro G, et al. (2005) The histone deacetylase inhibitor ITF2357 reduces production of pro-inflammatory cytokines in vitro and systemic inflammation in vivo. Mol Med 11: 1-15.
  8. Kawabata T, Nishida K, Takasugi K, Ogawa H, Sada K, et al. (2010) Increased activity and expression of histone deacetylase 1 in relation to tumor necrosis factor-alpha in synovial tissue of rheumatoid arthritis. Arthritis Res Ther 12: R133.
  9. Faraco G, Cavone L, Chiarugi A (2011) The therapeutic potential of HDAC inhibitors in the treatment of multiple sclerosis. Mol Med 17: 442-447.
  10. Huber LC, Brock M, Hemmatazad H, Giger OT, Moritz F, et al. (2007) Histone deacetylase/acetylase activity in total synovial tissue derived from rheumatoid arthritis and osteoarthritis patients. Arthritis Rheum 56: 1087-1093.
  11. Perl A (2010) Pathogenic mechanisms in systemic lupus erythematosus. Autoimmunity 43: 1-6.
  12. de Zoeten EF, Wang L, Sai H, Dillmann WH, Hancock WW (2010) Inhibition of HDAC9 increases T regulatory cell function and prevents colitis in mice. Gastroenterology 138: 583-594.
  13. Dokmanovic M, Clarke C, Marks PA (2007) Histone deacetylase inhibitors: overview and perspectives. Mol Cancer Res 5: 981-989.
  14. Glozak MA, Sengupta N, Zhang X, Seto E (2005) Acetylation and deacetylation of non-histone proteins. Gene 363: 15-23.
  15. Spange S, Wagner T, Heinzel T, Krämer OH (2009) Acetylation of non-histone proteins modulates cellular signalling at multiple levels. Int J Biochem Cell Biol 41: 185-198.
  16. Shakespear MR, Halili MA, Irvine KM, Fairlie DP, Sweet MJ (2011) Histone deacetylases as regulators of inflammation and immunity. Trends Immunol 32: 335-343.
  17. Akimova T, Beier UH, Liu Y, Wang L, Hancock WW (2012) Histone/protein deacetylases and T-cell immune responses. Blood 119: 2443-2451.
  18. Gryder BE, Sodji QH, Oyelere AK (2012) Targeted cancer therapy: giving histone deacetylase inhibitors all they need to succeed. Future Med Chem 4: 505-524.
  19. Tan J, Cang S, Ma Y, Petrillo RL, Liu D (2010) Novel histone deacetylase inhibitors in clinical trials as anti-cancer agents. J Hematol Oncol 3: 5.
  20. de Ruijter AJ, van Gennip AH, Caron HN, Kemp S, van Kuilenburg AB (2003) Histone deacetylases (HDACs): characterization of the classical HDAC family. Biochem J 370: 737-749.
  21. Saunders LR, Verdin E (2007) Sirtuins: critical regulators at the crossroads between cancer and aging. Oncogene 26: 5489-5504.
  22. Witt O, Deubzer HE, Milde T, Oehme I (2009) HDAC family: What are the cancer relevant targets? Cancer Lett 277: 8-21.
  23. Delcuve GP, Khan DH, Davie JR (2012) Roles of histone deacetylases in epigenetic regulation: emerging paradigms from studies with inhibitors. Clin Epigenetics 4: 5.
  24. Yang WM, Tsai SC, Wen YD, Fejer G, Seto E (2002) Functional domains of histone deacetylase-3. J Biol Chem 277: 9447-9454.
  25. Hubbert C, Guardiola A, Shao R, Kawaguchi Y, Ito A, et al. (2002) HDAC6 is a microtubule-associated deacetylase. Nature 417: 455-458.
  26. Gao L, Cueto MA, Asselbergs F, Atadja P (2002) Cloning and functional characterization of HDAC11, a novel member of the human histone deacetylase family. J Biol Chem 277: 25748-25755.
  27. Davidson A, Diamond B (2001) Autoimmune diseases. N Engl J Med 345: 340-350.
  28. Javierre BM, Fernandez AF, Richter J, Al-Shahrour F, Martin-Subero JI, et al. (2010) Changes in the pattern of DNA methylation associate with twin discordance in systemic lupus erythematosus. Genome Res 20: 170-179.
  29. Ballestar E (2010) Epigenetics lessons from twins: prospects for autoimmune disease. Clin Rev Allergy Immunol 39: 30-41.
  30. Simmons JK, Patel J, Michalowski A, Zhang S, Wei BR, et al. (2014) TORC and class I HDAC inhibitors synergize to suppress mature B cell neoplasms. Mol Oncol 8: 261-272.
  31. Akimova T, Ge G, Golovina T, Mikheeva T, Wang L, et al. (2010) Histone/protein deacetylase inhibitors increase suppressive functions of human FOXP3+ Tregs. Clin Immunol 136: 348-363.
  32. Regna NL, Chafin CB, Hammond SE, Puthiyaveetil AG, Caudell DL, et al. (2014) Class I and II histone deacetylase inhibition by ITF2357 reduces SLE pathogenesis in vivo. Clin Immunol 151: 29-42.
  33. Dovey OM, Foster CT, Conte N, Edwards SA, Edwards JM, et al. (2013) Histone deacetylase 1 and 2 are essential for normal T-cell development and genomic stability in mice. Blood 121: 1335-1344.
  34. Glauben R, Siegmund B (2011) Inhibition of histone deacetylases in inflammatory bowel diseases. Mol Med 17: 426-433.
  35. Wang L, de Zoeten EF, Greene MI, Hancock WW (2009) Immunomodulatory effects of deacetylase inhibitors: therapeutic targeting of FOXP3+ regulatory T cells. Nat Rev Drug Discov 8: 969-981.
  36. Zhu LJ, Yang X, Yu XQ (2010) Anti-TNF-alpha therapies in systemic lupus erythematosus. J Biomed Biotechnol 2010: 465898.
  37. Mishra N, Reilly CM, Brown DR, Ruiz P, Gilkeson GS (2003) Histone deacetylase inhibitors modulate renal disease in the MRL-lpr/lpr mouse. J Clin Invest 111: 539-552.
  38. Reilly CM, Thomas M, Gogal R Jr, Olgun S, Santo A, et al. (2008) The histone deacetylase inhibitor trichostatin A upregulates regulatory T cells and modulates autoimmunity in NZB/W F1 mice. J Autoimmun 31: 123-130.
  39. Reilly CM, Mishra N, Miller JM, Joshi D, Ruiz P, et al. (2004) Modulation of renal disease in MRL/lpr mice by suberoylanilide hydroxamic acid. J Immunol 173: 4171-4178.
  40. Joosten LA, Leoni F, Meghji S, Mascagni P (2011) Inhibition of HDAC activity by ITF2357 ameliorates joint inflammation and prevents cartilage and bone destruction in experimental arthritis. Mol Med 17: 391-396.
  41. Nasu Y, Nishida K, Miyazawa S, Komiyama T, Kadota Y, et al. (2008) Trichostatin A, a histone deacetylase inhibitor, suppresses synovial inflammation and subsequent cartilage destruction in a collagen antibody-induced arthritis mouse model. Osteoarthritis Cartilage 16: 723-732.
  42. Chan VS, Tsang HH, Tam RC, Lu L, Lau CS (2013) B-cell-targeted therapies in systemic lupus erythematosus. Cell Mol Immunol 10: 133-142.
  43. Anolik J, Sanz I (2004) B cells in human and murine systemic lupus erythematosus. Curr Opin Rheumatol 16: 505-512.
  44. Morimoto C, Reinherz EL, Schlossman SF, Schur PH, Mills JA, et al. (1980) Alterations in immunoregulatory T cell subsets in active systemic lupus erythematosus. J Clin Invest 66: 1171-1174.
  45. Crispín JC, Oukka M, Bayliss G, Cohen RA, Van Beek CA, et al. (2008) Expanded double negative T cells in patients with systemic lupus erythematosus produce IL-17 and infiltrate the kidneys. J Immunol 181: 8761-8766.
  46. Tao R, de Zoeten EF, Ozkaynak E, Chen C, Wang L, et al. (2007) Deacetylase inhibition promotes the generation and function of regulatory T cells. Nat Med 13: 1299-1307.
  47. Kim HJ, Bae SC (2011) Histone deacetylase inhibitors: molecular mechanisms of action and clinical trials as anti-cancer drugs. Am J Transl Res 3: 166-179.
  48. Ellis L, Pan Y, Smyth GK, George DJ, McCormack C, et al. (2008) Histone deacetylase inhibitor panobinostat induces clinical responses with associated alterations in gene expression profiles in cutaneous T-cell lymphoma. Clin Cancer Res 14: 4500-4510.
  49. Coronel J, Cetina L, Pacheco I, Trejo-Becerril C, Gonzalez-Fierro A, et al. (2011) A double-blind, placebo-controlled, randomized phase III trial of chemotherapy plus epigenetic therapy with hydralazine valproate for advanced cervical cancer. Preliminary results. Med Oncol 28: S540-546.
  50. Dowdell KC, Pesnicak L, Hoffmann V, Steadman K, Remaley AT, et al. (2009) Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, diminishes lymphoproliferation in the Fas -deficient MRL/lpr(-/-) murine model of autoimmune lymphoproliferative syndrome (ALPS). Exp Hematol 37: 487-494.
  51. Wagner JM, Hackanson B, Lübbert M, Jung M (2010) Histone deacetylase (HDAC) inhibitors in recent clinical trials for cancer therapy. Clin Epigenetics 1: 117-136.
  52. Ganesan A, Nolan L, Crabb SJ, Packham G (2009) Epigenetic therapy: histone acetylation, DNA methylation and anti-cancer drug discovery. Curr Cancer Drug Targets 9: 963-981.
  53. Bannerman B, Xu L, Jones M, Tsu C, Yu J, et al. (2011) Preclinical evaluation of the antitumor activity of bortezomib in combination with vitamin C or with epigallocatechin gallate, a component of green tea. Cancer Chemother Pharmacol 68: 1145-1154.
  54. Hancock WW (2011) Rationale for HDAC inhibitor therapy in autoimmunity and transplantation. Handb Exp Pharmacol 206: 103-123.
  55. Reichert N, Choukrallah MA, Matthias P (2012) Multiple roles of class I HDACs in proliferation, differentiation, and development. Cell Mol Life Sci 69: 2173-2187.
  56. Harms KL, Chen X (2007) Histone deacetylase 2 modulates p53 transcriptional activities through regulation of p53-DNA binding activity. Cancer Res 67: 3145-3152.
  57. Huang BH, Laban M, Leung CH, Lee L, Lee CK, et al. (2005) Inhibition of histone deacetylase 2 increases apoptosis and p21Cip1/WAF1 expression, independent of histone deacetylase 1. Cell Death Differ 12: 395-404.
  58. Kawashima H, Takatori H, Suzuki K, Iwata A, Yokota M, et al. (2013) Tumor suppressor p53 inhibits systemic autoimmune diseases by inducing regulatory T cells. J Immunol 191: 3614-3623.
  59. Zheng SJ, Lamhamedi-Cherradi SE, Wang P, Xu L, Chen YH (2005) Tumor suppressor p53 inhibits autoimmune inflammation and macrophage function. Diabetes 54: 1423-1428.
  60. Komarova EA, Krivokrysenko V, Wang K, Neznanov N, Chernov MV, et al. (2005) p53 is a suppressor of inflammatory response in mice. FASEB J 19: 1030-1032.
  61. Lin HS, Hu CY, Chan HY, Liew YY, Huang HP, et al. (2007) Anti-rheumatic activities of histone deacetylase (HDAC) inhibitors in vivo in collagen-induced arthritis in rodents. Br J Pharmacol 150: 862-872.
  62. Choo QY, Ho PC, Tanaka Y, Lin HS (2010) Histone deacetylase inhibitors MS-275 and SAHA induced growth arrest and suppressed lipopolysaccharide-stimulated NF-kappaB p65 nuclear accumulation in human rheumatoid arthritis synovial fibroblastic E11 cells. Rheumatology (Oxford) 49: 1447-1460.
  63. Tackey E, Lipsky PE, Illei GG (2004) Rationale for interleukin-6 blockade in systemic lupus erythematosus. Lupus 13: 339-343.
  64. Ohshima S, Saeki Y, Mima T, Sasai M, Nishioka K, et al. (1998) Interleukin 6 plays a key role in the development of antigen-induced arthritis. Proc Natl Acad Sci U S A 95: 8222-8226.
  65. Rogler G, Andus T (1998) Cytokines in inflammatory bowel disease. World J Surg 22: 382-389.
  66. Samoilova EB, Horton JL, Hilliard B, Liu TS, Chen Y (1998) IL-6-deficient mice are resistant to experimental autoimmune encephalomyelitis: roles of IL-6 in the activation and differentiation of autoreactive T cells. J Immunol 161: 6480-6486.
  67. Kay J, Calabrese L (2004) The role of interleukin-1 in the pathogenesis of rheumatoid arthritis. Rheumatology (Oxford) 43 Suppl 3: iii2-2iii9.
  68. Coccia M, Harrison OJ, Schiering C, Asquith MJ, Becher B, et al. (2012) IL-1beta mediates chronic intestinal inflammation by promoting the accumulation of IL-17A secreting innate lymphoid cells and CD4(+) Th17 cells. J Exp Med 209: 1595-1609.
  69. Voronov E, Dayan M, Zinger H, Gayvoronsky L, Lin JP, et al. (2006) IL-1 beta-deficient mice are resistant to induction of experimental SLE. Eur Cytokine Netw 17: 109-116.
  70. Kim HN, Lee JH, Jin WJ, Ko S, Jung K, et al. (2012) MS-275, a benzamide histone deacetylase inhibitor, prevents osteoclastogenesis by down-regulating c-Fos expression and suppresses bone loss in mice. Eur J Pharmacol 691: 69-76.
  71. Yamashita T, Yao Z, Li F, Zhang Q, Badell IR, et al. (2007) NF-kappaB p50 and p52 regulate receptor activator of NF-kappaB ligand (RANKL) and tumor necrosis factor-induced osteoclast precursor differentiation by activating c-Fos and NFATc1. J Biol Chem 282: 18245-18253.
  72. Nishioka C, Ikezoe T, Yang J, Takeuchi S, Koeffler HP, et al. (2008) MS-275, a novel histone deacetylase inhibitor with selectivity against HDAC1, induces degradation of FLT3 via inhibition of chaperone function of heat shock protein 90 in AML cells. Leuk Res 32: 1382-1392.
  73. Shukla HD, Pitha PM (2012) Role of hsp90 in systemic lupus erythematosus and its clinical relevance. Autoimmune Dis 2012: 728605.
  74. Shimp SK 3rd, Chafin CB, Regna NL, Hammond SE, Read MA, et al. (2012) Heat shock protein 90 inhibition by 17-DMAG lessens disease in the MRL/lpr mouse model of systemic lupus erythematosus. Cell Mol Immunol 9: 255-266.
  75. Gurvich N, Tsygankova OM, Meinkoth JL, Klein PS (2004) Histone deacetylase is a target of valproic acid-mediated cellular differentiation. Cancer Res 64: 1079-1086.
  76. Van Beneden K, Geers C, Pauwels M, Mannaerts I, Verbeelen D, et al. (2011) Valproic acid attenuates proteinuria and kidney injury. J Am Soc Nephrol 22: 1863-1875.
  77. Siu LL, Pili R, Duran I, Messersmith WA, Chen EX, et al. (2008) Phase I study of MGCD0103 given as a three-times-per-week oral dose in patients with advanced solid tumors. J Clin Oncol 26: 1940-1947.
  78. Buglio D, Mamidipudi V, Khaskhely NM, Brady H, Heise C, et al. (2010) The class-I HDAC inhibitor MGCD0103 induces apoptosis in Hodgkin lymphoma cell lines and synergizes with proteasome inhibitors by an HDAC6-independent mechanism. Br J Haematol 151: 387-396.
  79. El-Khoury V, Pierson S, Szwarcbart E, Brons NH, Roland O, et al. (2014) Disruption of autophagy by the histone deacetylase inhibitor MGCD0103 and its therapeutic implication in B-cell chronic lymphocytic leukemia. Leukemia .
  80. El-Khoury V, Moussay E, Janji B, Palissot V, Aouali N, et al. (2010) The histone deacetylase inhibitor MGCD0103 induces apoptosis in B-cell chronic lymphocytic leukemia cells through a mitochondria-mediated caspase activation cascade. Mol Cancer Ther 9: 1349-1360.
  81. Stylianou K, Petrakis I, Mavroeidi V, Stratakis S, Vardaki E, et al. (2011) The PI3K/Akt/mTOR pathway is activated in murine lupus nephritis and downregulated by rapamycin. Nephrol Dial Transplant 26: 498-508.
  82. Kim TH, Choi SJ, Lee YH, Song GG, Ji JD (2012) Combined therapeutic application of mTOR inhibitor and vitamin D(3) for inflammatory bone destruction of rheumatoid arthritis. Med Hypotheses 79: 757-760.
  83. Foster JG, Blunt MD, Carter E, Ward SG (2012) Inhibition of PI3K signaling spurs new therapeutic opportunities in inflammatory/autoimmune diseases and hematological malignancies. Pharmacol Rev 64: 1027-1054.
  84. Zhang F, Shi Y, Wang L, Sriram S (2011) Role of HDAC3 on p53 expression and apoptosis in T cells of patients with multiple sclerosis. PLoS One 6: e16795.
  85. Gillespie J, Savic S, Wong C, Hempshall A, Inman M, et al. (2012) Histone deacetylases are dysregulated in rheumatoid arthritis and a novel histone deacetylase 3-selective inhibitor reduces interleukin-6 production by peripheral blood mononuclear cells from rheumatoid arthritis patients. Arthritis Rheum 64: 418-422.
  86. Furumai R, Matsuyama A, Kobashi N, Lee KH, Nishiyama M, et al. (2002) FK228 (depsipeptide) as a natural prodrug that inhibits class I histone deacetylases. Cancer Res 62: 4916-4921.
  87. Nishida K, Komiyama T, Miyazawa S, Shen ZN, Furumatsu T, et al. (2004) Histone deacetylase inhibitor suppression of autoantibody-mediated arthritis in mice via regulation of p16INK4a and p21(WAF1/Cip1) expression. Arthritis Rheum 50: 3365-3376.
  88. Aringer M, Houssiau F, Gordon C, Graninger WB, Voll RE, et al. (2009) Adverse events and efficacy of TNF-alpha blockade with infliximab in patients with systemic lupus erythematosus: long-term follow-up of 13 patients. Rheumatology (Oxford) 48: 1451-1454.
  89. Aringer M, Smolen JS (2008) The role of tumor necrosis factor-alpha in systemic lupus erythematosus. Arthritis Res Ther 10: 202.
  90. Reimund JM, Ratajczyk J, Sola B, Justum AM, Muller CD (2007) Anti-tumor necrosis factor-alpha (TNF-alpha) treatment strategies in Crohn's disease. Recent Pat Inflamm Allergy Drug Discov 1: 21-34.
  91. Seymour HE, Worsley A, Smith JM, Thomas SH (2001) Anti-TNF agents for rheumatoid arthritis. Br J Clin Pharmacol 51: 201-208.
  92. Haberland M, Montgomery RL, Olson EN (2009) The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat Rev Genet 10: 32-42.
  93. Aung HT, Schroder K, Himes SR, Brion K, van Zuylen W, et al. (2006) LPS regulates proinflammatory gene expression in macrophages by altering histone deacetylase expression. FASEB J 20: 1315-1327.
  94. Fischle W, Dequiedt F, Hendzel MJ, Guenther MG, Lazar MA, et al. (2002) Enzymatic activity associated with class II HDACs is dependent on a multiprotein complex containing HDAC3 and SMRT/N-CoR. Mol Cell 9: 45-57.
  95. Yan K, Cao Q, Reilly CM, Young NL, Garcia BA, et al. (2011) Histone deacetylase 9 deficiency protects against effector T cell-mediated systemic autoimmunity. J Biol Chem 286: 28833-28843.
  96. Tao R, Hancock WW (2008) Resistance of Foxp3+ regulatory T cells to Nur77-induced apoptosis promotes allograft survival. PLoS One 3: e2321.
  97. Kuhn A, Beissert S, Krammer PH (2009) CD4(+)CD25 (+) regulatory T cells in human lupus erythematosus. Arch Dermatol Res 301: 71-81.
  98. Crispín JC, Liossis SN, Kis-Toth K, Lieberman LA, Kyttaris VC, et al. (2010) Pathogenesis of human systemic lupus erythematosus: recent advances. Trends Mol Med 16: 47-57.
  99. Cao D, Malmstrom V, Baecher-Allan C, Hafler D, Klareskog L, et al. (2003) Isolation and functional characterization of regulatory CD25brightCD4+ T cells from the target organ of patients with rheumatoid arthritis. Eur J Immunol 33: 215-223.
  100. Viglietta V, Baecher-Allan C, Weiner HL, Hafler DA (2004) Loss of functional suppression by CD4+CD25+ regulatory T cells in patients with multiple sclerosis. J Exp Med 199: 971-979.
  101. Boden EK, Snapper SB (2008) Regulatory T cells in inflammatory bowel disease. Curr Opin Gastroenterol 24: 733-741.
  102. Liu MF, Lin LH, Weng CT, Weng MY (2008) Decreased CD4+CD25+bright T cells in peripheral blood of patients with primary Sjogren's syndrome. Lupus 17: 34-39.
  103. Wahl SM, Chen W (2005) Transforming growth factor-beta-induced regulatory T cells referee inflammatory and autoimmune diseases. Arthritis Res Ther 7: 62-68.
  104. Wenzel J, Henze S, Brahler S, Bieber T, Tuting T (2005) The expression of human leukocyte antigen-DR and CD25 on circulating T cells in cutaneous lupus erythematosus and correlation with disease activity. Exp Dermatol 14: 454-459.
  105. Hayashi T, Hasegawa K, Adachi C (2005) Elimination of CD4(+)CD25(+) T cell accelerates the development of glomerulonephritis during the preactive phase in autoimmune-prone female NZB x NZW F mice. Int J Exp Pathol 86: 289-296.
  106. Dallavalle S, Pisano C, Zunino F (2012) Development and therapeutic impact of HDAC6-selective inhibitors. Biochem Pharmacol 84: 756-765.
  107. de Zoeten EF, Wang L, Butler K, Beier UH, Akimova T, et al. (2011) Histone deacetylase 6 and heat shock protein 90 control the functions of Foxp3(+) T-regulatory cells. Mol Cell Biol 31: 2066-2078.
  108. Valenzuela-Fernández A, Cabrero JR, Serrador JM, Sánchez-Madrid F (2008) HDAC6: a key regulator of cytoskeleton, cell migration and cell-cell interactions. Trends Cell Biol 18: 291-297.
  109. Horwitz DA (2008) Regulatory T cells in systemic lupus erythematosus: past, present and future. Arthritis Res Ther 10: 227.
  110. La Cava A (2008) T-regulatory cells in systemic lupus erythematosus. Lupus 17: 421-425.
  111. Kleinewietfeld M, Puentes F, Borsellino G, Battistini L, Rötzschke O, et al. (2005) CCR6 expression defines regulatory effector/memory-like cells within the CD25(+)CD4+ T-cell subset. Blood 105: 2877-2886.
  112. Butler KV, Kalin J, Brochier C, Vistoli G, Langley B, et al. (2010) Rational design and simple chemistry yield a superior, neuroprotective HDAC6 inhibitor, tubastatin A. J Am Chem Soc 132: 10842-10846.
  113. Zhang YZ, Li YY (2014) Inflammatory bowel disease: pathogenesis. World J Gastroenterol 20: 91-99.
  114. Jochems J, Boulden J, Lee BG, Blendy JA, Jarpe M, et al. (2013) Antidepressant-Like Properties of Novel HDAC6 Selective Inhibitors with Improved Brain Bioavailability. Neuropsychopharmacology 39: 389-400.
  115. Beier UH, Wang L, Han R, Akimova T, Liu Y, et al. (2012) Histone deacetylases 6 and 9 and sirtuin-1 control Foxp3+ regulatory T cell function through shared and isoform-specific mechanisms. Sci Sigal 5: ra45.
  116. Hancock WW, Akimova T, Beier UH, Liu Y, Wang L (2012) HDAC inhibitor therapy in autoimmunity and transplantation. Ann Rheum Dis 71 Suppl 2: i46-54.
  117. Villagra A, Cheng F, Wang HW, Suarez I, Glozak M, et al. (2009) The histone deacetylase HDAC11 regulates the expression of interleukin 10 and immune tolerance. Nat Immunol 10: 92-100.
  118. Iyer SS, Cheng G (2012) Role of interleukin 10 transcriptional regulation in inflammation and autoimmune disease. Crit Rev Immunol 32: 23-63.
  119. Park YB, Lee SK, Kim DS, Lee J, Lee CH, et al. (1998) Elevated interleukin-10 levels correlated with disease activity in systemic lupus erythematosus. Clin Exp Rheumatol 16: 283-288.
  120. Zhang ZY, Zhang Z, Schluesener HJ (2010) MS-275, an histone deacetylase inhibitor, reduces the inflammatory reaction in rat experimental autoimmune neuritis. Neuroscience 169: 370-377.
  121. Namdar M, Perez G, Ngo L, Marks PA (2010) Selective inhibition of histone deacetylase 6 (HDAC6) induces DNA damage and sensitizes transformed cells to anticancer agents. Proc Natl Acad Sci U S A 107: 20003-20008.
  122. Vishwakarma S, Iyer LR, Muley M, Singh PK, Shastry A, et al. (2013) Tubastatin, a selective histone deacetylase 6 inhibitor shows anti-inflammatory and anti-rheumatic effects. Int Immunopharmacol 16: 72-78.
Citation: Regna NL, Reillya CM (2014) Isoform-Selective HDAC Inhibition in Autoimmune Disease. J Clin Cell Immunol 5:207.

Copyright: © 2014 Regna NL, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top