Sei sulla pagina 1di 9

Acta Pharmaceutica Sinica B 2012;2(4):387–395

Institute of Materia Medica, Chinese Academy of Medical Sciences


Chinese Pharmaceutical Association

Acta Pharmaceutica Sinica B

www.elsevier.com/locate/apsb
www.sciencedirect.com

REVIEW

Histone deacetylases and their inhibitors: molecular


mechanisms and therapeutic implications in
diabetes mellitus
Xiaojie Wanga, Xinbing Weia, Qi Pangb, Fan Yia,n

a
Department of Pharmacology, School of Medicine, Shandong University, Jinan 250012, China
b
Department of Neurosurgery, Provincial Hospital Affiliated to Shandong University, Jinan 250021, China

Received 10 April 2012; revised 16 May 2012; accepted 7 June 2012

KEY WORDS Abstract Epigenetic mechanisms such as DNA methylation, histone modification and microRNA
Histone deacetylases;
changes have been shown to be important for the regulation of cellular functions. Among them,
Diabetes mellitus; histone deacetylases (HDACs) are enzymes that balance the acetylation activities of histone
Histone acetyltrans- acetyltransferases in chromatin remodeling and play essential roles in gene transcription to regulate
ferases; cell proliferation, migration and death. Recent studies indicate that HDACs are promising drug
HDAC proteins; targets for a wide range of diseases including cancer, neurodegenerative and psychiatric disorders,
HDAC inhibitors cardiovascular dysfunction, autoimmunity and diabetes mellitus. This review highlights the role of
HDACs in diabetes mellitus and outlines several important cellular and molecular mechanisms by
which HDACs regulate glucose homeostasis and can be targeted for the treatment of diabetic
microvascular complications. It is hoped that our understanding of the role of HDACs in diabetes
mellitus will lead to the development of better diagnostic tools and the design of more potent and
specific drugs targeting selective HDAC proteins for the treatment of the disease.

& 2012 Institute of Materia Medica, Chinese Academy of Medical Sciences and Chinese Pharmaceutical
Association. Production and hosting by Elsevier B.V. All rights reserved.

n
Corresponding author. Tel./fax: þ86 0531 88382616.
E-mail address: fanyi@sdu.edu.cn (Fan Yi).

2211-3835 & 2012 Institute of Materia Medica, Chinese Academy of Medical Sciences and Chinese Pharmaceutical Association. Production and
hosting by Elsevier B.V. All rights reserved.

Peer review under the responsibility of Institute of Materia Medica, Chinese Academy of Medical Sciences and Chinese Pharmaceutical
Association.
http://dx.doi.org/10.1016/j.apsb.2012.06.005
388 Xiaojie Wang et al.

1. Introduction homology with the yeast Sir2 protein10,11. Because of this,


class III HDACs are also called ‘sirtuins’ (SIRTs). SIRTs 1–7
Histone deacetylases (HDACs) are enzymes that balance the target a wide range of cellular proteins in the nucleus,
activities of histone acetyltransferases (HATs) in chromatin cytoplasm and mitochondria for post-translational modifica-
remodeling and thereby play an essential role in gene tran- tion by acetylation or ADP-ribosylation. Among them, SIRT1
scription to regulate cell proliferation, migration and apopto- and SIRT2 can shuttle between the nucleus and cyto-
sis, immune pathways and angiogenesis. In the last few years, plasm12,13. SIRT1, the founding member of the class, couples
numerous studies have demonstrated the role of HDACs in protein deacetylation with NADþ hydrolysis and links cellular
cancer initiation and progression giving rise to the view that energy and redox state to multiple signaling and survival
HDACs are potentially important drug targets in the treat- pathways which positively regulate transcription factors such
ment of cancer. Recent studies also indicate that targeting as NF-kB, p53 and forkhead box (FOX) proteins. In addition,
HDACs is a promising therapeutic strategy for a number of SIRT1 may play a role in tumor initiation and progression as
other diseases including neurodegenerative disorders, cardio- well as in the development of drug resistance by blocking
vascular dysfunction, autoimmunity and diabetes mellitus. senescence and apoptosis or promoting cell growth and
This review highlights the role of HDACs in the regulation angiogenesis. SIRT2 is mainly localized to the cytoplasm
of glucose metabolism, insulin resistance, insulin expression and affects a-tubulin acetylation status.
and secretion and the therapeutic potential of HDAC inhibi- SIRTs 3, 4 and 5 are mainly localized in the mitochondria14.
tors for the treatment of diabetes mellitus. It is hoped that SIRT3 helps to maintain energetic cell homeostasis by posi-
clarification of the pathological role of HDAC-mediated tively regulating the activity of acetylcoenzyme A synthetase 2
signaling pathways will aid in the development of diagnostic and deacetylating complex I of the respiratory chain involved
tools and in the design of more potent and specific drugs that in ATP production. SIRT4 has ADP ribosylase activity which
target individual HDAC proteins. inactivates glutamate dehydrogenase and, in turn, inhibits
insulin secretion in pancreatic b-cells. SIRT5 deacetylates and
thereby activates carbamoyl phosphate synthetase 1 to pro-
2. The HDAC family: classification, location and substrates mote ammonia detoxification. SIRTs 6 and 7 are predomi-
nantly found in the nucleus where they are involved in DNA
HDACs are a family of enzymes which compete with HATs to repair and ribosomal RNA transcription, respectively.
control the acetylation of lysine residues making up the To date, sirtuins have emerged as potential therapeutic
histones. The balance of acetylation and deacetylation then targets for the treatment of cancer and of metabolic, cardio-
determines the post-translational acetylation status of histone vascular and neurodegenerative diseases. This review focuses
and other non-histone proteins1. on the Zn2þ-dependent HDACs because they are the targets of
To date, 18 HDACs have been identified in mammals made small molecule HDAC inhibitors that have shown efficacy in
up of four classes according to their sequence identity and animal models of diabetes mellitus. The classification, loca-
catalytic activity2. Class I HDACs (HDAC1, 2, 3 and 8) share tion, substrates, main biological functions and inhibitors of
sequence homology with the yeast Rpd3 protein and contain a HDACs are shown in Table 1.
nuclear localization signal (NLS) but, with the exception of
HDAC3, no nuclear export signal (NES). Accordingly, they
are mainly located in the nucleus and regulate the expression of 3. HDAC inhibitors
many genes3, such as thioredoxin binding protein-2 (TBP-2),
myocyte enhancer factor-2 (MEF2), NF-kB and GATA4. HDAC inhibitors are chemical compounds that inhibit Zn2þ-
Class II HDACs (HDAC4–7, 9 and 10) are larger proteins dependent HDAC enzymes and for which some 490 clinical
than class I because they contain additional regulatory trials have been carried out over the last 10 years. Their efficacy
domains. They contain both an NLS and NES and shuttle against malignant cells and their potent anticancer activity in
between the cytoplasm and nucleus depending on their pre-clinical studies have been demonstrated. They also display
phosphorylation status. Class II HDACs are further subdi- protective effects in animal models of various neurological and
vided into class IIa (HDAC4, 5, 7, 9) and IIb (HDAC6, 10) cardiovascular diseases and of diabetes mellitus15–17.
with class IIa having relatively low enzymatic activity com- HDAC inhibitors can be structurally grouped into hydro-
pared to class I possibly to allow them to efficiently process xamates, cyclic peptides, aliphatic acids, benzamides, boronic
restricted sets of specific, unknown natural substrates. Class acid-based compounds, benzofuranone and sulfonamide con-
IIb HDACs have primarily non-epigenetic functions involving taining molecules and a/b peptide structures18. Most of them
the regulation of protein folding and turnover5,6. possess a stereotypical three-part structure consisting of a
HDAC11 is the sole member of class IV. It is localized in zinc-binding ‘‘warhead’’ group which docks in the active site, a
the nucleus and has a catalytic domain in the N-terminal linker, and a surface recognition domain which interacts with
region. HDAC11 has been demonstrated to regulate the residues near the entrance of the active site. Among them,
balance between immune activation and immune tolerance in vorinostat (suberoylanilide hydroxamic acid) and depsipeptide
CD4þ T-cells7 and to play an essential role in oligodendrocyte (Romidepsin, FK-228) were approved by the FDA in 2006
differentiation8 and the regulation of OX40 ligand expression and 2009, respectively. Vorinostat is structurally related to
in Hodgkin lymphoma9. trichostatin A (TSA) which is indicated for the treatment of
Classes I, II and IV HDACs possess a Zn2þ-dependent relapsed and refractory cutaneous T-cell lymphoma (CTCL).
mechanism of action whereas the structurally unrelated class The cyclic peptides are the most structurally complex group of
III HDACs comprise a family of nicotinamide adenine HDAC inhibitors of which depsipeptide is one of the most
dinucleotide (NAD)-dependent deacetylases sharing sequence important members.
Histone deacetylases and their inhibitors: molecular mechanisms and therapeutic implications in diabetes mellitus 389

Table 1 The HDAC family: classification, localization and substrates.

HDAC Location Substrates Biological functions Main HDAC


inhibitor
Class Member

I HDAC1 Nucleus Histones, MEF2, GATA, YY1, NF-kB, Cell survival and proliferation Valproic acid,
DNMT1–SHP, ATM, MyoD, p53, AR, vorinostat, TSA,
BRCA1, MECP2, pRb RGFP1-36
HDAC2 Nucleus Histones, HOP, NF-kB, GATA2, Insulin resistance; cell (HDAC3)
BRCA1, pRb, MECP IRS-1 proliferation
HDAC3 Nucleus Histones, HDAC4, 5, 7–9, SHP, Cell survival and
GATA1, NF-kB, pRb proliferation
HDAC8 Nucleus HSP70 Cell proliferation
IIa HDAC4 Nucleocytoplasmic Histones, SRF, Runx2, p53, p21, GATA, Regulation of skeletogenesis TSA, vorinostat,
traffic FOXO, HIF-1a, SUV39H1, HP1, HDAC3, and gluconeogenesis phenylbutyrate
MEF2, CaM, 14-3-3
HDAC5 Nucleocytoplasmic Histones, HDAC3, YY1, MEF2, Cardiovascular growth and
traffic Runx2, CaM, 14-3-3 function; cardiac myocytes
and endothelial cell function;
gluconeogenesis
HDAC7 Nucleocytoplasmic Histones, HDAC3, MEF2, PML, Thymocyte differentiation–
traffic Runx2, CaM, 14-3-3, HIF-1a endothelial function–
gluconeogenesis
HDAC9 Nucleus Histones, HDAC3, MEF2, CaM, Thymocyte differentiation;
14-3-3 cardiovascular growth and
function
IIb HDAC6 Cytoplasm a-tubulin, Cortactin, HSP90, HDAC11, Cell motility–control of Vorinostat,
SHP, PP1, Runx2, LcoR cytoskeletal dynamics TSA, tubacin
HDAC10 Nucleocytoplasmic LcoR, PP1 Homologous recombination (HDAC6)
traffic
III SIRT1 Nucleus Histones, p53, p73, p300, NF-kB–FOXO, Aging; redox control–cell Nicotinamide
PTEN, NICD, MEF2, HIFs, SREBP-1c, b- survival–autoimmune system (SIRT1),
catenin, PGC1a, Bmal, NF-kB, Per2, Ku70, regulation suramin (SIRT1
XPA, SMAD7, Cortactin, Ku70, IRS-2, and SIRT2)
APE1, PCAF, TIP60, p300, AceCS1,
PPARg, ER-a, AR, LXR
SIRT2 Nucleus Histones, a-tubulin Cell survival–cell migration
and invasion
SIRT3 Mitochondria Histones, Ku70, IDH2, HMGCS2, GDH, Redox control
AceCS, SdhA, SOD2, LCAD
SIRT4 Mitochondria GDH Enegry metabolism
SIRT5 Mitochondria Cytochrome c, CPS1 Urea cycle
SIRT6 Nucleus Histone H3, TNF-a Metabolic regulation
SIRT7 Nucleus p53 Apoptosis
IV HDAC11 Nucleus Histones, HDAC6, Cdt1 Immunomodulators–DNA Sodium
replication butyrate, TSA,
vorinostat

AceCS, Acetylcoenzyme A synthetase; AR, Androgen receptor; ATM, Ataxia-telangiectasia-mutated; APE, Apurinic/apyrimidinic
endonuclease-1; BRCA, Breast cancer; Bmal, Brain and muscle aryl hydrocarbon receptor nuclear translocator (ARNT)-like; CaM,
Calmodulin; CPS1, Carbamoyl phosphate synthetase 1; DNMT, DNA methyltransferase; ER-a, estrogen receptor-a; FOXO, Forkhead
box O; GATA, GATA binding protein; GDH, Glutamate dehydrogenase; HP1, Heterochromatin protein 1; HMGCS, Hydroxymethyl-
glutaryl-CoA synthase; HOP, Homeodomain only protein; HIF, Hypoxia-inducible factor; HSP, Heat shock protein; IDH, Isocitrate
dehydrogenase; IRS, Insulin receptor substrate; LCAD, Long chain acyl coenzyme A dehydrogenase; LcoR, ligand-dependent receptor
co-repressor; LXR, Liver X receptor; MECP, Methyl-CpG-binding domain protein; MEF, myocyte enhancer factor; NICD, Notch1
intracellular domain; NF-kB, Nuclear transcription factor-kappa B; PCAF, p300/CBP-associated factor; Per-2, Period circadian protein
homolog-2; PTEN, Phosphatase and tensin homolog; PGC, Peroxisome proliferator-activated receptor gamma coactivator; PP1, Protein
phosphatase; PPAR, Peroxisome proliferator-activated receptor; pRb, Retinoblastoma protein; PML, Promyelocytic leukaemia protein;
Runx, Runt-related transcription factor; SdhA, Succinate dehydrogenase complex subunit A; SHP, Src homology region 2-domain-
containing phosphatase; SOD, Superoxide dismutase; SUV39H1, Histone-lysine N-methyltransferase; SREBP, Sterol regulatory element-
binding protein; TNF, tumor necrosis factor.

Although HDAC enzymes are promising drug targets selective (pan-HDAC inhibitors). Thus the effects of HDAC
because of their importance in cell cycle regulation, prolifera- inhibitors are often studied by examining changes in bulk
tion, differentiation, metabolism, protein trafficking and DNA histone acetylation or the therapeutic effects observed in a
repair, currently available HDAC inhibitors are mostly non- given experimental model or in clinical trails. Consequently, it
390 Xiaojie Wang et al.

is important to develop more specific inhibitors of individual phosphatase (G-6-Pase), the rate-controlling gluconeogenetic
HDAC isoforms that are critically involved in particular enzymes32. It does this by phosphorylating foxhead box O1
processes to improve their efficacy and reduce toxicity. (FOXO1), thus repressing the transcription of the PEPCK and
Recent studies have provided some promising results for G-6-Pase genes33. In contrast, glucagon is known to increase
more selective HDAC inhibitors. For instance, compounds hepatic gluconeogenesis by inducing the expression of PEPCK
containing aryl substitutions on the benzamide warhead have and G-6-Pase, the regulation of which requires co-activators
been found to be highly selective for HDACs 1 and 219–21. such as peroxisome proliferator-activated receptor g co-acti-
SNDX-275 (Syndax Pharmaceutical Inc.) is a synthetic benza- vator 1a (PGC-1a) and cAMP response element-binding
mide derivative with activity against HDACs 1, 2 and 3 (class I) (CREB) protein34.
in the mM range. Similarly, a series of apicidin-based alkyl Recent studies indicate that the regulation of glucose
ketones appear to be selective for HDACs 1, 2 and 322–24. homeostasis is associated with epigenetic mechanisms. Post-
MGCD0103 (Methylgene Inc.) is an isoform-selective amino- translational modifications of histones by acetylation, phos-
phenylbenzamide that inhibits HDACs of classes I and IV with phorylation, methylation and ubiquitination play important
almost no effect on those of class II. Tubacin and TSA exhibit roles in gene transcription35. In particular, acetylation of
greater selectivity for HDAC625–28. histone and non-histone proteins provides a key mechanism
for controlling cellular signaling and gene expression. The
HDACs regulate this acetylation of histone and transcrip-
4. HDAC and glucose homeostasis tional factors which are involved in glucose homeostasis and
play a central role in the regulation of glucose metabolism.
Glucose homeostasis is maintained through tight regulation of The functions of HDACs in the regulation of glucose home-
glucose production in the liver and glucose uptake in the ostasis are illustrated in Fig. 1.
peripheral tissues particularly skeletal muscle and adipose
tissue. Insulin and glucagon play critical roles in this regula-
tion29. In the fed state, insulin signals lower blood glucose by 4.1. HDAC and insulin resistance
facilitating glucose uptake mainly into peripheral tissues and
by inhibiting endogenous glucose production in the liver30. As previously stated, insulin signals play an important role in
Conversely, in the fasted state, glucagon signals up-regulate regulating blood glucose by promoting glucose uptake in
gluconeogenesis and the breakdown of glycogen in the liver. peripheral tissues and decreasing glucose production and
In terms of mechanism, insulin activates the serine/threo- stimulating glycogen synthesis in the liver. Once insulin binds
nine kinase Akt which, in turn, activates the glucose trans- to its receptor, phosphate groups are added to tyrosines on
porter 4 (GLUT4) to translocate from intracellular vesicles to particular proteins in the cell including insulin receptor
the plasma membrane where it facilitates glucose uptake31. substrate (IRS) 1. This then activates PI3K which, in turn,
The activation of Akt is also known to suppress the genes for phosphorylates the protein kinase Akt which is important for
phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6- the stimulation of GLUT4 expression.

Figure 1 The role of HDACs in the regulation of glucose metabolism. (A) HDACs down-regulate the expression of GLUT4 and
promote insulin resistance in adipocytes and muscle cells. (B) HDAC1 reduces the expression of PDX1 leading to the decreased
expression of insulin while SIRT1 up-regulates UCP2 which promotes the secretion of insulin. (C) HDACs are involved in the regulation
of gluconeogenesis in hepatocytes in several ways: in the stimulation of glucagon, class IIa HDACs are dephosphorylated and
translocated from the cytoplasm to the nucleus; class IIa HDACs recruit HDAC3 which can deacetylate FOXO1 and FOXO3 thereby
enhancing FOXO DNA-binding to promote gluconeogenetic gene expression; ER stress inhibits STAT3-dependent suppression of
hepatic gluconeogenic enzymes via HDAC-dependent STAT3 deacetylation.
Histone deacetylases and their inhibitors: molecular mechanisms and therapeutic implications in diabetes mellitus 391

It has been shown that HDACs play a regulatory role in In contrast, a report from Moynihan et al.54 has demon-
insulin signalling. Recent reports indicate that HDAC2 can strated that SIRT1 improves glucose-induced pancreatic insu-
bind to IRS-1 in liver cells of the db/db mouse, leading to lin secretion. In their study, transgenic mice selectively
decreased acetylation and reduced insulin receptor-mediated overexpressing SIRT1 in pancreatic b-cells (BESTO mice)
tyrosine phosphorylation of IRS-1. Accordingly, the HDAC showed improved glucose tolerance and enhanced insulin
inhibitor TSA or gene silencing of HDAC2 enhance acetyla- secretion in response to glucose. Furthermore, microarray
tion of IRS-1 and partially attenuate insulin resistance36. analyses of b-cell lines revealed that SIRT1 regulated genes,
GLUT4 is important for glucose uptake as indicated by the including uncoupling protein 2 (UCP2), are involved in insulin
fact that insulin-dependent glucose homeostasis is sensitive to secretion. These results indicate for the first time that SIRT1 is
changes in GLUT4 expression and translocation37–39. The important in b-cell function in vivo and has potential as a
GLUT4 gene promoter is governed by two domains namely target in treating type 2 diabetes.
the MEF2 binding domain and Domain I. When MEF2 binds
to the MEF2 binding domain and GLUT4 enhancer factor 4.3. HDAC and gluconeogenesis
(GEF) binds to Domain I, transcriptional activity is the most
advanced40–43. It has been suggested that class II HDACs Gluconeogenesis is another important process in glucose home-
down-regulate MEF2-dependent gene transcription. Some ostasis that is reportedly regulated by HDACs. This is through
studies also report that activation of HDAC5 decreases the control of the activity of FOXO transcription factors which
GLUT4 expression in cardiac tissue and that a decrease in are critical for mediating the expression of gluconeogenetic rate-
HDAC5 activity correlates with an increase in GLUT4 limiting genes including those expressing G-6-Pase and PEPCK.
expression during exercise44. Chromatin immunoprecipitation HDAC1 induces the expression of hepatocyte nuclear factor 4a
(ChIP) assays also demonstrate that HDAC5 is associated (HNF4a) and the dephosphorylation of FOXO1 in hepatocytes
with the GLUT4 promoter in preadipocytes45 where, through leading to the induction of PEPCK expression and gluconeo-
analysis by in vitro transcription assays, it has been shown that genesis in the liver55. Moreover, some studies have found that
HDAC5 specifically represses transcriptional activation of the class IIa HDACs regulate glucagon-induced FOXO activation
GLUT4 promoter. After HDAC5 undergoes complex forma- and that AMPK and salt-inducible kinases (SIKs) 1 and 2 can
tion with GEF and MEF2, controlled by its phosphorylation phosphorylate class IIa HDACs (HDAC4, 5 and 7) which are
via AMP-activated protein kinase (AMPK) and calmodulin- then excluded from the nucleus. Under stimulation from the
dependent protein kinase (CaMK), HDAC5 deacetylates fasting hormone, glucagon, class IIa HDACs are rapidly
histone and compacts the chromatin structure leading to dephosphorylated and translocated from the cytoplasm to the
repression of GLUT4. nucleus where they recruit and form a complex with HDAC3
Recent studies also provide strong evidence of the impor- which has the ability to deacetylate FOXO1 and FOXO3a. This
tance of HDAC6 in glucocorticoid receptor-mediated effects on enhances FOXO DNA-binding which regulates gluconeoge-
glucose metabolism and its potential as a therapeutic target for netic gene promoters such as that of G-6-Pase. The recruitment
the prevention of glucocorticoid-induced diabetes. For example, of HDAC3 also results in the acute transcriptional induction of
impaired dexamethasone-induced hepatic glucocorticoid recep- gluconeogenetic genes via deacetylation and activation of
tor translocation has been found in HDAC6 knockout mice. In FOXO family transcription factors. Loss of class IIa HDACs
fact, dexamethasone-induced expression of some hepatic genes inhibits FOXO targeted gluconeogenetic genes and lowers
is markedly attenuated in these mice and significant improve- blood glucose leading to increased glycogen storage. In mouse
ments in dexamethasone-induced whole-body glucose intoler- models of type 2 diabetes, inhibition of class IIa HDACs has
ance and insulin resistance is observed indicating that HDAC6 been further shown to ameliorate hyperglycemia, indicating
is an essential regulator of insulin signaling46. that class IIa HDACs regulate G-6-Pase expression and
subsequently affect gluconeogenesis56.
HDACs have also been shown to be involved in signal
4.2. HDAC and insulin expression and secretion transducer and activator of transcription 3 (STAT3)-mediated
gluconeogenesis57. STAT3 can be acetylated by CREB pro-
HDACs have been demonstrated to down-regulate pancreatic tein/p300 and deacetylated by HDACs58. In the liver, STAT3
insulin formation and secretion47,48. Insulin gene expression is plays an important role in the suppression of gluconeogenic
up-regulated at higher glucose concentrations and is asso- enzyme expression. The study by Kimura et al.57 reveals that
ciated with several transcription factors including duodenal endoplasmic reticulum (ER) stress inhibits STAT3-dependent
homeobox 1 (PDX1)48, V-maf musculoaponeurotic fibrosar- suppression of hepatic gluconeogenic enzymes via Janus
coma oncogene homologue A (MafA)49,50 and neurogenic kinase 2 (JAK2) dephosphorylation and HDAC-dependent
differentiation 1 (NeuroD1)51. Expression and release of STAT3 deacetylation demonstrating the importance of
insulin is regulated by the acetylation status of histone H447. HDACs in mediating the increase in hepatic glucose produc-
The transcription factor PDX1, specific to pancreatic b-cells, tion that occurs in obesity and diabetes. However, the
interacts with the p300 transcriptional co-activator to enhance particular HDAC isoforms involved in the deacetylation of
proinsulin expression48. Some studies indicate that HDAC1 is STAT-3 remains unknown.
involved in silencing PDX1 leading to failure in b-cell devel-
opment and to b-cell dysfunction52. Although some studies
also report that transcription factors such as NeuroD153 and 5. HDAC inhibition in the treatment of diabetic complications
MafA49,50 are associated with acetylation states that lead to
increased binding activity, whether HDACs actually regulate The evidence reviewed here indicates that HDACs are
this association remains unclear. involved in the pathogenesis of diabetes mellitus through
392 Xiaojie Wang et al.

regulating glucose homeostasis via a number of pathways. In of HDAC activity suppresses the EMT induced by TGF-b1 in
addition, many large scale clinical studies of diabetes have human renal epithelial cells58. HDAC inhibitors can induce
demonstrated that early intensive glycemic control reduces the the expression of DNA binding/differentiation 2 (Id2) and
incidence and progression of micro and macrovasular compli- bone-morphogenic protein (BMP)-7 to inhibit TGF-b1 signal-
cations. However, epidemiological and prospective data reveal ling64. Further studies demonstrate that the HDAC inhibitor,
that the stressors in the diabetic vasculature persist beyond the TSA, prevents TGF-b1-induced apoptosis by inhibiting TGF-
point when glycemic control is achieved. In fact, the detri- b1-induced extracellular signal-regulated kinase (ERK) activa-
mental effects that result from the development and progres- tion. Furthermore, recent studies report that TSA decreases
sion of the complications of hyperglycemia can persist for mRNA and protein expression of the components of ECM
years. Fortunately, in addition to regulate glucose metabolism and prevents the EMT in streptozotocin (STZ)-induced
by increasing insulin secretion, improving insulin resistance diabetic kidneys. In addition, it has been found that HDAC2
and controlling gluconeogenesis, HDAC inhibitors also show activity is significantly increased in the kidney of STZ-induced
promise in the treatment of diabetic complications including diabetic rats and db/db mice as well as in TGF-b1-treated
diabetic nephropathy (DN)59,60 and diabetic retinopathy normal rat kidney tubular epithelial cells60.
(DR)61,62. The efficacy of HDAC inhibitors in treating these HDAC2 knockdown reduces fibronectin and a-smooth
conditions appears to be governed by multiple actions on a muscle actin expression but increases E-cadherin expression
variety of pathophysiological processes as shown in Fig. 2. suggesting that it plays an important role in the accumulation
of ECM and the EMT in diabetic kidney. Very recently, a
study has revealed that long-term treatment with vorinostat
5.1. HDAC and diabetic nephropathy improves albuminuria and mesangial matrix accumulation in
diabetic mice through an endothelial nitric oxide synthase
DN is one of the major microvascular complications of (eNOS)-dependent mechanism65. This finding provides further
diabetes and the most common cause of end-stage renal evidence to support the potential utility of HDAC inhibitors
disease (ESRD)63. Although the pathogenesis of DN is multi- in the treatment of cardiorenal diseases. Our recent studies
factorial, recent clinical trials and experimental studies have also show that, among the Zn2þ-dependent HDACs, class II
highlighted the importance of HDAC-mediated epigenetic HDACs 4 and 5 are significantly increased both in vivo and
processes in its development. in vitro in kidneys exposed, to high glucose concentrations
HDAC inhibitors have been reported to have an antifibrotic which are associated with NADPH oxidase-mediated oxida-
effect in the kidney64. Epithelial-to-mesenchymal transition tive stress (unpublished data).
(EMT) of renal tubular epithelial cells and excessive accumu- One of the earliest features of DN is renal enlargement.
lation of extracellular matrix (ECM) in the kidney contribute Epidermal growth factor (EGF) plays a pivotal role in the
to the renal fibrosis in DN. It has been reported that inhibition development of diabetic nephromegaly and transactivation of

Figure 2 Summary of the mechanisms by which HDACs induce diabetic nephropathy and retinopathy.
Histone deacetylases and their inhibitors: molecular mechanisms and therapeutic implications in diabetes mellitus 393

its receptor has been implicated in the pathogenesis of later- Foundation of China (81170772, 81070918, 81171062 and
stage disease. Vorinostat has been found to attenuate early 30901551); the Shandong Natural Science Fund for Distin-
renal enlargement in experimental diabetes, an effect probably guished Young Scholars to Yi F (JQ201121); the Independent
mediated, at least in part, by down-regulation of the EGFR Innovation Foundation of Shandong University (IIFS-
and attenuation of EGF-mediated tubular epithelial cell DU2010JC17); and the Nature Science Foundation of Shan-
proliferation66. dong Province (ZR2010HM112).

5.2. HDAC and diabetic retinopathy References

DR presenting as macular edema and proliferative retinopathy 1. Verdin E, Dequiedt F, Kasler HG. Class II histone deacetylases:
is considered the most sight-threatening ocular complication versatile regulators. Trends Genet 2003;19:286–93.
in diabetic patients but its exact pathogenesis remains unclear. 2. Shakespear MR, Halili MA, Irvine KM, Fairlie DP, Sweet MJ.
A report from Zhong and Kowluru62 indicates that, in STZ- Histone deacetylases as regulators of inflammation and immunity.
Trends Immunol 2011;32:335–43.
induced diabetic rats, the expressions of HDACs 1, 2 and 8 are
3. Yang XJ, Seto E. Collaborative spirit of histone deacetylases in
significantly increased in retinal tissue and cells derived from
regulating chromatin structure and gene expression. Curr Opin
it. As a result, the activity of HAT is compromised and the Genet Dev 2003;13:143–53.
acetylation of histone H3 is decreased62. 4. Kato T, Shimono Y, Hasegawa M, Jijiwa M, Enomoto A, Asai N,
Retinal ischemia plays an important role in DR and may et al. Characterization of the HDAC1 complex that regulates the
involve necrotic and apoptotic processes. A variety of cellular sensitivity of cancer cells to oxidative stress. Cancer Res 2009;
events are also thought to contribute to the degenerative response 69:3597–604.
in diabetic patients. It has been reported that TSA administration 5. Seidel C, Schnekenburger M, Dicato M, Diederich M. Histone
produces a significant anti-inflammatory effect in the retina deacetylase modulators provided by Mother Nature. Genes Nutr
associated with the suppression of retinal tumor necrosis 2012. http://dx.doi.org/10.1007/s12263-012-0283-9.
6. Spiegel S, Milstien S, Grant S. Endogenous modulators and
factor-a (TNF-a) expression and inhibition of MMPs. Valproic
pharmacological inhibitors of histone deacetylases in cancer
acid (VPA) can also protect the retina from ischemia through the
therapy. Oncogene 2012;31:537–51.
endoplasmic reticulum (ER) stress pathway. VPA significantly 7. Villagra A, Cheng F, Wang HW, Suarez I, Glozak M, Maurin M,
increases the expression of glucose-regulated protein (GRP) 78, et al. The histone deacetylase HDAC11 regulates the expression
which may result from the hyperacetylation of histone H3. of interleukin 10 and immune tolerance. Nat Immunol 2009;10:
GRP78 forms a complex with caspase-7 and -12 to prevent the 92–100.
release of caspase-12 from the ER, thereby inhibiting ER stress- 8. Liu H, Hu Q, D’Ercole AJ, Ye P. Histone deacetylase 11 regulates
induced caspase activation67. In this way ER stress induced oligodendrocyte-specific gene expression and cell development in
apoptosis can be ameliorated by treatment with VPA68. OL-1 oligodendroglia cells. Glia 2009;57:1–12.
9. Buglio D, Khaskhely NM, Voo KS, Martinez-Valdez H, Liu YJ,
Younes A. HDAC11 plays an essential role in regulating OX40
6. Conclusions and perspectives ligand expression in Hodgkin lymphoma. Blood 2011;117:2910–7.
10. Haigis MC, Sinclair DA. Mammalian sirtuins: biological insights
This review provides evidence that supports the role of HDACs and disease relevance. Annu Rev Pathol 2010;5:253–95.
in diabetes mellitus and outlines several important cellular and 11. Houtkooper RH, Canto C, Wanders RJ, Auwerx J. The secret life
of NADþ: an old metabolite controlling new metabolic signaling
molecular mechanisms for the regulation of glucose homeostasis.
pathways. Endocr Rev 2010;31:194–223.
It also describes the targeting of HDACs for the treatment of 12. North BJ, Verdin E. Interphase nucleo-cytoplasmic shuttling and
diabetic complications through the regulation of insulin resis- localization of SIRT2 during mitosis. PloS One 2007;2:e784.
tance and secretion. Due to space limitations, the possible roles 13. Tanno M, Sakamoto J, Miura T, Shimamoto K, Horio Y.
of HDACs in inflammation and the immunoresponse in diabetes Nucleocytoplasmic shuttling of the NADþ-dependent histone
mellitus are not included but have been comprehensively deacetylase SIRT1. J Biol Chem 2007;282:6823–32.
reviewed elsewhere69. Taken together, the evidence provides a 14. Michishita E, Park JY, Burneskis JM, Barrett JC, Horikawa I.
strong rationale to continue preclinical studies and initiate Evolutionarily conserved and nonconserved cellular localizations
clinical trials of HDAC inhibitors. However, prolonged broad- and functions of human SIRT proteins. Mol Biol Cell 2005;
spectrum HDAC inhibition using the currently available pan- 16:4623–35.
15. Kim HJ, Rowe M, Ren M, Hong JS, Chen PS, Chuang DM.
HDAC inhibitors involves risk not only because these substances
Histone deacetylase inhibitors exhibit anti-inflammatory and
are associated with adverse side effects, but also because little is neuroprotective effects in a rat permanent ischemic model of
known about the function of individual HDACs in diabetes stroke: multiple mechanisms of action. J Pharmacol Exp Ther
mellitus. In further researching the clinical use of HDAC 2007;321:892–901.
inhibitors, it will be important to elucidate the role of individual 16. Wang Z, Leng Y, Tsai LK, Leeds P, Chuang DM. Valproic acid
HDACs within the body and to develop HDAC inhibitors with attenuates blood–brain barrier disruption in a rat model of
improved specificity to provide an effective therapeutic strategy transient focal cerebral ischemia: the roles of HDAC and MMP-
for the treatment of diabetes mellitus. 9 inhibition. J Cereb Blood Flow Metab 2011;31:52–7.
17. Ren M, Leng Y, Jeong M, Leeds PR, Chuang DM. Valproic acid
reduces brain damage induced by transient focal cerebral ischemia
Acknowledgments
in rats: potential roles of histone deacetylase inhibition and heat
shock protein induction. J Neurochem 2004;89:1358–67.
The authors wish to thank the following for financial support 18. Marks PA. Histone deacetylase inhibitors: a chemical genetics
for this study: the National 973 Basic Research Program approach to understanding cellular functions. Biochim Biophys
of China (2012CB517700); the National Nature Science Acta 2010;1799:717–25.
394 Xiaojie Wang et al.

19. McKinsey TA. Isoform-selective HDAC inhibitors: closing in on 39. Gibbs EM, Stock JL, McCoid SC, Stukenbrok HA, Pessin JE,
translational medicine for the heart. J Mol Cell Cardiol 2011; Stevenson RW, et al. Glycemic improvement in diabetic db/db
51:491–6. mice by over-expression of the human insulin-regulatable glucose
20. Methot JL, Chakravarty PK, Chenard M, Close J, Cruz JC, transporter (GLUT4). J Clin Invest 1995;95:1512–8.
Dahlberg WK, et al. Exploration of the internal cavity of histone 40. Lu J, McKinsey TA, Nicol RL, Olson EN. Signal-dependent
deacetylase (HDAC) with selective HDAC1/HDAC2 inhibitors activation of the MEF2 transcription factor by dissociation from
(SHI-1:2). Bioorg Med Chem Lett 2008;18:973–8. histone deacetylases. Proc Natl Acad Sci USA 2000;97:4070–5.
21. Vannini A, Volpari C, Filocamo G, Casavola EC, Brunetti M, 41. Miska EA, Karlsson C, Langley E, Nielsen SJ, Pines J, Kouzar-
Renzoni D, et al. Crystal structure of a eukaryotic zinc-dependent ides T. HDAC4 deacetylase associates with and represses the
histone deacetylase, human HDAC8, complexed with a hydro- MEF2 transcription factor. EMBO J 1999;18:5099–107.
xamic acid inhibitor. Proc Natl Acad Sci USA 2004;101:15064–9. 42. Sparrow DB, Miska EA, Langley E, Reynaud-Deonauth S,
22. Furumai R, Matsuyama A, Kobashi N, Lee KH, Nishiyama M, Kotecha S, Towers N, et al. MEF-2 function is modified by a
Nakajima H, et al. FK228 (depsipeptide) as a natural prodrug that novel co-repressor, MITR. EMBO J 1999;18:5085–98.
inhibits class I histone deacetylases. Cancer Res 2002;62:4916–21. 43. Youn HD, Grozinger CM, Liu JO. Calcium regulates transcrip-
23. Khan N, Jeffers M, Kumar S, Hackett C, Boldog F, Khramtsov tional repression of myocyte enhancer factor 2 by histone
N, et al. Determination of the class and isoform selectivity of deacetylase 4. J Biol Chem 2000;275:22563–7.
small-molecule histone deacetylase inhibitors. Biochem J 2008;409: 44. Czubryt MP, McAnally J, Fishman GI, Olson EN. Regulation of
581–9. peroxisome proliferator-activated receptor gamma coactivator 1
24. Maulucci N, Chini MG, Micco SD, Izzo I, Cafaro E, Russo A, alpha (PGC-1 alpha ) and mitochondrial function by MEF2 and
et al. Molecular insights into azumamide histone deacetylases HDAC5. Proc Natl Acad Sci USA 2003;100:1711–6.
inhibitory activity. J Am Chem Soc 2007;129:3007–12. 45. Weems J, Olson AL. Class II histone deacetylases limit GLUT4
25. Chen Y, Lopez-Sanchez M, Savoy DN, Billadeau DD, Dow GS, gene expression during adipocyte differentiation. J Biol Chem
Kozikowski AP. A series of potent and selective, triazolylphenyl- 2011;286:460–8.
based histone deacetylases inhibitors with activity against pan- 46. Winkler R, Benz V, Clemenz M, Bloch M, Foryst-Ludwig A,
creatic cancer cells and Plasmodium falciparum. J Med Chem Wardat S, et al. Histone deacetylase 6 (HDAC6) is an essential
2008;51:3437–48. modifier of glucocorticoid-induced hepatic gluconeogenesis. Dia-
26. Kozikowski AP, Tapadar S, Luchini DN, Kim KH, Billadeau betes 2012;61:513–23.
DD. Use of the nitrile oxide cycloaddition (NOC) reaction for 47. Mosley AL, Ozcan S. Glucose regulates insulin gene transcription
molecular probe generation: a new class of enzyme selective by hyperacetylation of histone H4. J Biol Chem 2003;278:19660–6.
histone deacetylase inhibitors (HDACIs) showing picomolar 48. Mosley AL, Ozcan S. The pancreatic duodenal homeobox-1
activity at HDAC6. J Med Chem 2008;51:4370–3. protein (Pdx-1) interacts with histone deacetylases Hdac-1 and
27. Schafer S, Saunders L, Schlimme S, Valkov V, Wagner JM, Kratz Hdac-2 on low levels of glucose. J Biol Chem 2004;279:54241–7.
F, et al. Pyridylalanine-containing hydroxamic acids as selective 49. Zhao L, Cissell MA, Henderson E, Colbran R, Stein R. The
HDAC6 inhibitors. ChemMedChem 2009;4:283–90. RIPE3b1 activator of the insulin gene is composed of a protein(s)
28. Smil DV, Manku S, Chantigny YA, Leit S, Wahhab A, Yan TP, of approximately 43 kDa, whose DNA binding activity is inhibited
et al. Novel HDAC6 isoform selective chiral small molecule by protein phosphatase treatment. J Biol Chem 2000;275:10532–7.
histone deacetylase inhibitors. Bioorg Med Chem Lett 2009; 50. Guo S, Burnette R, Zhao L, Vanderford NL, Poitout V, Hagman
19:688–92. DK, et al. The stability and transactivation potential of the
29. Biddinger SB, Kahn CR. From mice to men: insights into the mammalian MafA transcription factor are regulated by serine
insulin resistance syndromes. Annu Rev Physiol 2006;68:123–58. 65 phosphorylation. J Biol Chem 2009;284:759–65.
30. Saltiel AR, Kahn CR. Insulin signalling and the regulation of 51. Kitamura YI, Kitamura T, Kruse JP, Raum JC, Stein R, Gu W,
glucose and lipid metabolism. Nature 2001;414:799–806. et al. FOXO1 protects against pancreatic beta-cell failure through
31. Choi K, Kim YB. Molecular mechanism of insulin resistance in NeuroD and MafA induction. Cell Metab 2005;2:153–63.
obesity and type 2 diabetes. Kor J Intern Med 2010;25:119–29. 52. Park JH, Stoffers DA, Nicholls RD, Simmons RA. Development
32. Viollet B, Guigas B, Leclerc J, Hebrard S, Lantier L, Mounier R, of type 2 diabetes following intrauterine growth retardation in rats
et al. AMP-activated protein kinase in the regulation of hepatic is associated with progressive epigenetic silencing of Pdx1. J Clin
energy metabolism: from physiology to therapeutic perspectives. Invest 2008;118:2316–24.
Acta Physiol (Oxf) 2009;196:81–98. 53. Qiu Y, Guo M, Huang S, Stein R. Acetylation of the BETA2
33. Puigserver P, Rhee J, Donovan J, Walkey CJ, Yoon JC, Oriente transcription factor by p300-associated factor is important in
F, et al. Insulin-regulated hepatic gluconeogenesis through insulin gene expression. J Biol Chem 2004;279:9796–802.
FOXO1-PGC-1alpha interaction. Nature 2003;423:550–5. 54. Moynihan KA, Grimm AA, Plueger MM, Bernal-Mizrachi E,
34. Yoon JC, Puigserver P, Chen G, Donovan J, Wu Z, Rhee J, et al. Ford E, Cras-Meneur C, et al. Increased dosage of mammalian
Control of hepatic gluconeogenesis through the transcriptional Sir2 in pancreatic beta cells enhances glucose-stimulated insulin
coactivator PGC-1. Nature 2001;413:131–8. secretion in mice. Cell Metab 2005;2:105–17.
35. Berger SL. Histone modifications in transcriptional regulation. 55. Oiso H, Furukawa N, Suefuji M, Shimoda S, Ito A, Furumai R,
Curr Opin Genet Dev 2002;12:142–8. et al. The role of class I histone deacetylase (HDAC) on gluconeo-
36. Sun C, Zhou J. Trichostatin A improves insulin stimulated glucose genesis in liver. Biochem Biophys Res Commun 2011;404:166–72.
utilization and insulin signaling transduction through the repres- 56. Mihaylova MM, Vasquez DS, Ravnskjaer K, Denechaud PD, Yu
sion of HDAC2. Biochem Pharmacol 2008;76:120–7. RT, Alvarez JG, et al. Class IIa histone deacetylases are hormone-
37. Brozinick JT Jr., McCoid SC, Reynolds TH, Nardone NA, activated regulators of FOXO and mammalian glucose home-
Hargrove DM, Stevenson RW, et al. GLUT4 over-expression in ostasis. Cell 2011;145:607–21.
db/db mice dose-dependently ameliorates diabetes but is not a 57. Kimura K, Yamada T, Matsumoto M, Kido Y, Hosooka T,
lifelong cure. Diabetes 2001;50:593–600. Asahara S, et al. Endoplasmic reticulum stress inhibits STAT3-
38. Christ-Roberts CY, Pratipanawatr T, Pratipanawatr W, Berria R, dependent suppression of hepatic gluconeogenesis via depho-
Belfort R, Kashyap S, et al. Exercise training increases glycogen sphorylation and deacetylation. Diabetes 2012;61:61–73.
synthase activity and GLUT4 expression but not insulin signaling 58. Yuan ZL, Guan YJ, Chatterjee D, Chin YE. Stat3 dimerization
in overweight nondiabetic and type 2 diabetic subjects. Metabo- regulated by reversible acetylation of a single lysine residue.
lism 2004;53:1233–42. Science 2005;307:269–73.
Histone deacetylases and their inhibitors: molecular mechanisms and therapeutic implications in diabetes mellitus 395

59. Lee HB, Noh H, Seo JY, Yu MR, Ha H. Histone deacetylase 65. Advani A, Huang Q, Thai K, Advani SL, White KE, Kelly DJ,
inhibitors: a novel class of therapeutic agents in diabetic nephro- et al. Long-term administration of the histone deacetylase inhi-
pathy. Kidney Int Suppl 2007:61–6. bitor vorinostat attenuates renal injury in experimental diabetes
60. Noh H, Oh EY, Seo JY, Yu MR, Kim YO, Ha H, et al. Histone through an endothelial nitric oxide synthase-dependent mechan-
deacetylase-2 is a key regulator of diabetes- and transforming ism. Am J Pathol 2011;178:2205–14.
growth factor-beta1-induced renal injury. Am J Physiol Renal 66. Gilbert RE, Huang Q, Thai K, Advani SL, Lee K, Yuen DA,
Physiol 2009;297:729–39. et al. Histone deacetylase inhibition attenuates diabetes-associated
61. Crosson CE, Mani SK, Husain S, Alsarraf O, Menick DR. kidney growth: potential role for epigenetic modification of the
Inhibition of histone deacetylase protects the retina from ischemic epidermal growth factor receptor. Kidney Int 2011;79:1312–21.
injury. Invest Ophthalmol Vis Sci 2010;51:3639–45. 67. Rao RV, Ellerby HM, Bredesen DE. Coupling endoplasmic
62. Zhong Q, Kowluru RA. Role of histone acetylation in the reticulum stress to the cell death program. Cell Death Differ
development of diabetic retinopathy and the metabolic memory
2004;11:372–80.
phenomenon. J Cell Biochem 2010;110:1306–13.
68. Zhang Z, Tong N, Gong Y, Qiu Q, Yin L, Lv X, et al. Valproate
63. Collins AJ, Kasiske B, Herzog C, Chavers B, Foley R, Gilbertson D,
protects the retina from endoplasmic reticulum stress-induced
et al. Excerpts from the United States Renal Data System 2006
apoptosis after ischemia-reperfusion injury. Neurosci Lett 2011;
Annual Data Report. Am J Kidney Dis 2007;49(1 suppl. 1)6–7.
S1-296. 504:88–92.
64. Yoshikawa M, Hishikawa K, Marumo T, Fujita T. Inhibition of 69. Christensen DP, Dahllof M, Lundh M, Rasmussen DN, Nielsen
histone deacetylase activity suppresses epithelial-to-mesenchymal MD, Billestrup N, et al. Histone deacetylase (HDAC) inhibition
transition induced by TGF-beta1 in human renal epithelial cells. as a novel treatment for diabetes mellitus. Mol Med 2011;17:
J Am Soc Nephrol 2007;18:58–65. 378–90.

Potrebbero piacerti anche