You are on page 1of 14

Histone Deacetylase (HDAC) Inhibition as a Novel Treatment

for Diabetes Mellitus

Dan P Christensen,1* Mattias Dahllöf,1* Morten Lundh,1* Daniel N Rasmussen,1* Mette D Nielsen,1*
Nils Billestrup,2 Lars G Grunnet,1† and Thomas Mandrup-Poulsen1,3,4,5†
1
Center for Medical Research Methodology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;
2
Cellular and Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;
3
Department for Diabetes Inflammation, Hagedorn Research Institute, Gentofte, Denmark; and 4Department of Medicine and
Surgery, Karolinska Institute, Stockholm, Sweden; 5‡Institute of Biomedical Sciences, the Panum Institute, University of Copenhagen,
Copenhagen, Denmark

Both common forms of diabetes have an inflammatory pathogenesis in which immune and metabolic factors converge on
interleukin-1β as a key mediator of insulin resistance and β-cell failure. In addition to improving insulin resistance and preventing
β-cell inflammatory damage, there is evidence of genetic association between diabetes and histone deacetylases (HDACs); and
HDAC inhibitors (HDACi) promote β-cell development, proliferation, differentiation and function and positively affect late diabetic
microvascular complications. Here we review this evidence and propose that there is a strong rationale for preclinical studies and
clinical trials with the aim of testing the utility of HDACi as a novel therapy for diabetes.
© 2011 The Feinstein Institute for Medical Research, www.feinsteininstitute.org
Online address: http://www.molmed.org
doi: 10.2119/molmed.2011.00021

INTRODUCTION is intact. Preserving a functional β-cell (1). Thus, T1D is believed to be an auto-
Diabetes mellitus designates a group of mass is therefore the primary target of immune disease where variations in
chronic diseases where absolute or rela- novel treatments aimed at curing and pre- mainly immune-regulatory genes predis-
tive lack of insulin leads to aberrancies in venting diabetes mellitus. pose individuals to immune-mediated de-
substrate metabolism, causing acute and Type 1 diabetes mellitus (T1D) and struction of the β cells by a T-cell–driven
long-term complications. Although the Type 2 diabetes mellitus (T2D) constitute chronic inflammatory process in the islets
clinical and diagnostic hallmark is ele- the two main forms of diabetes. It has (insulitis) (2,3). In contrast, genome-wide
vated blood glucose, the metabolic distur- been estimated that 250 million individu- association scans (GWAS) have suggested
bances in diabetes mellitus are not limited als are currently afflicted by diabetes T2D to be predominately a disease of the
to glucose but encompass most processes worldwide and the prevalence is doubling β cell, where variations in genes affecting
of the intermediary metabolism of nutri- every 10 years. Whereas T1D is associated β-cell function and/or mass impair β-cell
ents, including proteins and lipids. Insulin with absolute insulin deficiency as a con- compensation to increased insulin de-
resistance is a component of most types of sequence of selective destruction of β cells, mands (4). In both diseases, there are
diabetes mellitus, but with the exception T2D is associated with a relative lack of strong gene–environment interactions that
of rare mutations in the insulin signaling insulin most commonly due to failure of trigger the pathogenetic process (in T1D,
cascade, the disease does not occur if in- the β cells to compensate for insulin re- hitherto unidentified viral and nutritional
sulin release from the pancreatic β cells in sistance caused by obesity. Of note, T1D etiologies, and in T2D, mainly lifestyle-
the islets of Langerhans (hereafter islets) and T2D are genetically distinct diseases related factors).
With respect to pathogenesis, the strict
dichotomy between T1D and T2D is most
*DPC, MD, ML, DNR, and MDN contributed equally to this work. likely an oversimplification. There is in-

LGG and TM-P contributed equally to this work. creasing recognition that T1D and T2D

Current affiliation. may represent extremes of a continuous
Address correspondence and reprint requests to Thomas Mandrup-Poulsen, Center for spectrum with a dominating β-cell defect
Medical Research Methodology, Department of Biomedical Sciences, The Panum Insti- at one end and dominating insulin resist-
tute, University of Copenhagen, 3 Blegdamsvej, DK-2200 Copenhagen N, Denmark. ance at the other (5). However, when dis-
Phone: +45-30-33-03-87; Fax: +45-35-32-75-37; E-mail: tmpo@sund.ku.dk. regarding diabetes caused by rare muta-
Submitted January 13, 2011; Accepted for publication January 24, 2011; Epub tions in insulin signaling, insulin
(www.molmed.org) ahead of print January 25, 2011. resistance is neither necessary nor suffi-

3 7 8 | C H R I S T E N S E N E T A L . | M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1
REVIEW ARTICLE

cient to cause diabetes, whereas β-cell high glucose concentrations (15) and the ment of an expanding range of diseases.
dysfunction is both a necessary and suffi- adipocytokine leptin (16) and by While transcriptional control over onco-
cient cause. This notion is supported by macrophages exposed to free fatty acids gene networks in cancer was the original
studies demonstrating progressive reduc- and islet amyloid polypeptide (10,11); the target of HDAC inhibition, neurodegener-
tion in β-cell function and mass in T2D naturally occurring IL-1 inhibitor, IL-1 re- ative and other inflammatory diseases are
(6–8). Autoimmune islet inflammation ceptor antagonist (IL-Ra), protects against now increasingly being evaluated as
and β-cell destruction are long-recognized high glucose–induced human β-cell toxic- novel indications, as illustrated by the re-
causes of T1D, although it is debated if ity (glucotoxicity) in vitro (15) and dia- views in this issue of Molecular Medicine.
the molecular effector mechanisms in- betes as well as β-cell dysfunction in- Acetylation is now recognized to regu-
volve predominantly classical cytotoxic duced by a high-fat diet (lipotoxicity) in late the master transcription factor in the
T-cell–mediated or predominantly in- an animal model (17); elevated IL-1β lev- inflammation nuclear factor (NF)-κB (re-
flammatory cytokine-mediated β-cell els contribute to the risk of developing viewed in [24]). Because the activation of
killing or both. Several mechanisms lead- T2D (18); and IL-1Ra treatment improves NFκB is a critical event in IL-1β–induced
ing to β-cell destruction in T2D have been β-cell function in patients with T2D for β-cell death (25,26), these findings led to
proposed: glucolipotoxicity, membrane up to 39 weeks after 13 weeks of treat- the investigation and demonstration of
disruption caused by islet amyloid ment (19,20). the protective effects of HDAC inhibition
polypeptide deposition and, more re- In summary, antiinflammatory treat- in β cells exposed to toxicity-mediating
cently, inflammation in the islets (9). Re- ment, and in particular inhibition of IL- cytokines (27).
cently, a unifying hypothesis was pro- 1β–induced toxicity, has therapeutic po- In this article, we review the potential
posed by the observation that all these tential in the treatment of both T1D and of inhibiting the classical HDACs as a
stimuli lead to the induction of inflamma- T2D. However, antiinflammatory biolog- novel treatment for diabetes. This review
tory mediators in the pancreatic islets ics are costly and require parenteral ad- includes a short overview of genetic as-
(10,11) that cause β-cell destruction by ac- ministration either via the subcutaneous sociations between HDACs and the etiol-
tivating pathways in β cells similar to or intravenous route. There is thus an ogy of diabetes followed by a discussion
those in T1D. unmet need to develop safe, inexpensive of the potential for HDACi as an oral
Thus, despite their different genetic and patient-convenient (oral) antiinflam- therapy with respect to modulation of
background, the immune and metabolic matory drugs that mimic the beneficial the immune system, insulin resistance,
pathogeneses of T1D and T2D, respec- effects of IL-1 blockade. β-cell development, differentiation and
tively, seem to converge on common ex- function, and pathogenetic events rele-
tracellular inflammatory stressors in the ANTIINFLAMMATORY PROPERTIES OF vant for β-cell failure and destruction
islets and intracellular signaling induced HISTONE DEACETYLASE INHIBITORS and islet graft rejection. Of note, HDACi
by these stressors (reviewed in [12]). As outlined in the current issue of also hold promise with respect to treat-
Within the last years, several publications Molecular Medicine, histone deacetylase in- ment of late diabetic complications such
reported that the inflammatory cytokine hibitors (HDACi) show promising antiin- as diabetic nephropathy (28,29) and reti-
interleukin (IL)-1β can act as a common flammatory properties, as demonstrated nal ischemia playing a central role in dia-
extracellular inflammatory stressor. In the in an increasing number of animal and betic retinopathy (30). HDACi and late
mid-1980s, IL-1β secreted from activated cellular models of inflammatory diseases diabetic complications will not be dis-
mononuclear cells was found to be selec- (21). As indicated by their name, the mo- cussed further here, and readers are re-
tively toxic to β cells and was found to lecular function of histone deacetylases ferred to the aforementioned references.
both inhibit β-cell function and induce (HDACs) was thought to be restricted to
β-cell death after prolonged exposure (re- histone deacetylation, but recent advances HDACs IN THE ETIOLOGY OF DIABETES
viewed in [13]). IL-1β has since received in phylogenetic analysis suggested that As mentioned above, the etiology of
much attention as an important mediator HDACs regulate the activity of a wide diabetes is complex and multifactorial
of the immune-induced β-cell destruction range of nonhistone proteins (22). This with contributions from many genes and
underlying T1D. Moreover, a number of was substantiated in a recent study (23) unknown environmental factors. Al-
observations in the last decade argue by the finding of 3,600 acetylation sites though GWAS point to T1D and T2D as
strongly for an important role of IL-1β in (of which only 61 were on histones) on being genetically distinct (31,32), at least
the pathogenesis of T2D as well. Thus, 1,750 proteins including exclusively cyto- two GWAS studies have found signifi-
mice deficient in caspase-1 and thereby plasmic proteins. Thus, the impact of cant linkage between the chromosomal
unable to process pro-IL-1β to mature bi- acetylation in terms of posttranslational region 6q21, where HDAC2 is located,
ologically active IL-1β are more insulin regulation is comparable to that of phos- and both T1D and T2D (33,34), indicat-
sensitive than wild-type animals (14); phorylation. A growing number of ing that HDAC2 could play a role in
IL-1β is secreted by β cells exposed to HDACi are being developed for the treat- both diseases.

M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1 | C H R I S T E N S E N E T A L . | 3 7 9
HDACs AS THERAPEUTIC TARGETS IN DIABETES MELLITUS

Although T1D and T2D are clearly


polygenetic disorders, the concordance
rate in twin studies is far from 100%
(35,36), indicating a significant etiologic
contribution from environmental
and/or epigenetic factors. Fetal expo-
sure to intrauterine growth retardation
(IUGR) contributes to the development
of T2D, as reviewed by Pinney and Sim-
mons (37). An adverse fetal milieu af-
fects β-cell development by modifying
key regulatory genes such as pancreatic
and duodenal homeobox factor 1 (Pdx1) (38)
as well as muscular glucose transport
through glucose transporter 4 (Glut4) (39).
Interestingly, the reduced expression of
Pdx1 after IUGR is mediated by loss of
histone acetylation through the recruit-
ment of HDAC1 in complex with the
corepressor Sin3A to the proximal pro-
moter of Pdx1 (Figure 1). Thereby, a self- Figure 1. Model of changes in acetylation levels in pancreatic β cells of IUGR. (A) Under
propagating epigenetic cycle is induced normal conditions, the Pdx1 proximal promoter is associated with acetylated nucleo-
somes (Ac, green squares). (B) During IUGR, a complex containing HDAC1 and Sin3A in-
in which the HDAC1/Sin3 complex re-
duces a progressive loss of histone acetylation in islets from fetal and 2-week-old mice.
cruits a histone demethylase leading to
This then leads to repression of Pdx1 expression. (C) This effect is reversed by HDAC inhibi-
loss of histone 3 lysine 4 trimethylation
tion. Simplified from Park et al. (38).
(H3K4me3), further repressing Pdx1
transcription. This effect was reversed
by HDAC inhibition in the neonatal ani- also to the more infrequent forms of histone acetylation as an important
mal but not in the adult animal, where monogenic autosomal diabetes termed pathogenetic factor in diabetes.
H3K9 dimethylation and extensive “maturity-onset diabetes of the young”
DNA methylation locked the Pdx1 pro- (MODY). MODY comprises at least INNATE AND ADAPTIVE IMMUNE
moter in its transcriptionally inactive seven distinct subtypes on the basis of SYSTEMS AND HDACi IN DIABETES
state (38). the mutated genes in question (31). With As reviewed in other sections of this
Prenatal nutritional restriction leading the exception of glucokinase and in- issue of Molecular Medicine, HDAC inhibi-
to IUGR also leads to HDAC1- and sulin, these genes all encode transcrip- tion modifies innate and adaptive
HDAC4-mediated loss of histone acety- tion factors—namely hepatocyte nuclear immune responses (42–45). The specific
lation of the Glut4 promoter in adult factor (HNF)-1α, -1β and -4α, involved impact of HDACi on the immune system
muscle tissue, thereby inhibiting Glut4 in insulin transcription and hepatic glu- in relation to T1D and T2D is under-
transcription (39). The effective meta- coneogenesis, and pancreatic and duo- investigated. However, histone H3 is hy-
bolic repression of this important regula- denal homeobox 1 (PDX1) and neuro- peracetylated in the promoters of tumor
tor of peripheral glucose uptake and genic differentiation 1 (NeuroD1), necrosis factor-α (TNFα) and the inflam-
insulin resistance may contribute impor- involved in pancreatic development and matory-associated enzyme cyclooxyge-
tantly to the T2D phenotype. Of note, insulin production. These transcription nase (COX)-2 in monocytes isolated from
chromatin remodeling may already be factors all associate with histone acetyl- patients with T1D or T2D (46), suggesting
induced by current T2D treatments, transferases (HATs) and HDACs, sug- a potential importance of the activity of
since incretin hormones such as gesting an important role of histone HATs and HDACs in the expression of
glucagon-like peptide 1 and glucose- acetylation in their normal function. Un- proinflammatory genes in monocytes
dependent insulinotropic peptide 1 in- derlining this, some of the MODY muta- from patients suffering from diabetes
crease in vitro global acetylation of his- tions directly affect the ability of the (46). In vitro, increased histone acetylation
tone H3, leading to increased association transcription factors to interact with is induced by high glucose concentrations
with transcription factors (40). HAT/HDACs (41). and the HDAC inhibitor trichostatin A
Histone acetylation and HDACs are In summary, these findings all point to (TSA) in monocytes from diabetics (46),
not only relevant to T1D and T2D but inappropriate chromatin remodeling and and the production of the inflammatory

3 8 0 | C H R I S T E N S E N E T A L . | M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1
REVIEW ARTICLE

cytokines IL-1β and TNFα was induced


by high glucose concentrations through
activation of NFκB (47), suggesting that
hyperacetylation is a consequence of hy-
perglycemia or other metabolic aberran-
cies of diabetes rather than a cause of dia-
betes. Further, NFκB activity was
enhanced by HAT overexpression and
TSA and accordingly reversed by overex-
pression of HDAC1, -2, -3, -4, -5, and -6
(46). Taken together, these data suggest
that HDACi treatment of patients suffer-
ing from diabetes could have an undesir-
able effect on cytokine production by
monocytes. However, since effects of
HDACi are highly concentration depen- Figure 2. Effects of HDACs and HDACi on insulin signaling. For details, see the text.
dent, this potential adverse effect may
not be seen if lower HDACi concentra- Figure 2, insulin signals via binding to creased acetylation of IRS-1 and reduced
tions are used, since lower concentrations the insulin receptor leading to receptor insulin receptor–mediated tyrosine phos-
are generally associated with antiinflam- autophosphorylation and phosphoryla- phorylation of IRS-1. Accordingly, inhibi-
matory responses. In the above-men- tion of members of the insulin receptor tion of HDAC2 with TSA or RNAi-medi-
tioned study by Miao et al. (46), TSA was substrate (IRS) family (see i, Figure 2). ated knockdown inhibited deacetylation
used in a concentration of 300 nmol/L Upon phosphorylation, IRSs bind (and of IRS-1 and partially restored insulin
and was found to increase expression of activate) phosphatidylinositol 3-kinase signaling (55).
TNFα and COX-2. Similar results were (PI3K), which in turn leads to phosphory- Both translocation and expression of
reported from another study using 500 lation of the protein kinase Akt (see ii, GLUT4 are important for glucose uptake.
nmol/L TSA (48). Lower concentrations Figure 2). Among other effects, Akt in- Thus, overexpression of GLUT4 increases
of TSA (1–10 nmol/L) were not reported duces translocation of the glucose trans- basal and insulin-stimulated glucose dis-
to have this effect while still causing his- porter (GLUT4) from intracellular vesi- posal in mice (56,57). Transcription of
tone hyperacetylation (48). In contrast to cles to the plasma membrane, mediating GLUT4 is mainly under the regulation of
the effects of TSA, the HDAC inhibitor glucose uptake (see iii, Figure 2) (re- the GLUT4 enhancer factor (GEF) and
ITF2357 was shown to reduce the inflam- viewed in [50]). Obstruction of insulin the myocyte enhancer factor (MEF)-2,
matory response of peripheral blood signaling leading to insulin resistance both of which bind to transcriptional ele-
mononuclear cells (PBMCs) by lowering may occur at several levels in this path- ments in the GLUT4 promoter (58).
the release of TNFα, secretion of IL-1β way. As mentioned above, insulin resist- Through complex formation with GEF
and synthesis of interferon γ (IFNγ) (49). ance is a feature of both T1D and T2D— and MEF2, HDAC5 functions as a tran-
In summary, the effects of HDAC inhi- in the former case suspected to be scriptional repressor of GLUT4 by his-
bition on the immune system specifically secondary to deficient insulin secretion in tone deactylation and compacting of the
with respect to diabetes are not clarified, lean and underweight subjects (51), but chromatin structure (59–61). The forma-
and further studies are needed to un- also increasingly associated with over- tion of this inhibitory complex is regu-
ravel the dose-response relationships for weight of T1D subjects. In addition to lated by phosphorylation of HDAC5 by
different HDACi on cytokine production obesity, aging and genetic predisposition AMPK and CaMK, which induces the re-
from monocytes. Studies from other in- are proposed to enhance risk of develop- lease of HDAC5 from the complex
flammatory diseases have to our knowl- ing insulin resistance (52,53). (59,62). This allows recruitment of, for ex-
edge not reported monocyte activation as HDACs have been suggested to play a ample, peroxisome proliferator–activated
an adverse effect, lending optimism to regulatory role in physiological insulin receptor-γ coactivator 1 (PGC-1), which
future safe utility of HDACi in treating signaling (see Figure 2). Thus, HDACi in- functions as a transcriptional coactivator
diabetes. crease GLUT4 translocation and augment allowing GLUT4 transcription (see Fig-
basal and insulin-induced glucose up- ure 2, iv) (63,64). The HDAC inhibitor
INSULIN RESISTANCE AND HDAC take in skeletal muscle (54). IRS-1 binds TSA upregulates PGC-1 expression in
INHIBITION to HDAC2 in liver cells from the ob/ob skeletal muscle (65). This observation is
Insulin action is critical for cellular glu- mouse, a model of insulin resistance (55). clinically relevant, since decreased my-
cose uptake in most cells. As simplified in This result was associated with de- ocyte PGC-1α expression in patients with

M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1 | C H R I S T E N S E N E T A L . | 3 8 1
HDACs AS THERAPEUTIC TARGETS IN DIABETES MELLITUS

T2D is associated with elevated fasting rather, inhibition of insulin metabolism DNA damage correlated with inefficient
insulin concentrations (reviewed in [66]). in the liver was suggested to be the cause repair of DNA double-strand breaks (73);
As described above, HDAC1 and (reviewed in [68]). It remains to be exam- and HDAC4 inhibits cell cycle progres-
HDAC4 are also inhibitors of GLUT4 ex- ined if these side effects of VPA are asso- sion and has been suggested as a neuro-
pression, further underlining an impor- ciated with its HDAC inhibitor function, protective enzyme (74).
tant regulatory role of HDACs in glucose its effects on the central nervous system A plethora of transcription factors
uptake and insulin resistance. HDACs or other actions of the drug. The fact that have to be minutely orchestrated at the
may thus be a target for treatment of in- VPA is a branched-chain fatty acid may expressional level to mediate the forma-
sulin resistance in muscular tissue, since account for these side effects (68). To our tion of the fully differentiated tissue. The
compensatory GLUT4 transcription may knowledge, hyperinsulinemia, insulin re- pancreatic development network of tran-
reverse the resistant state (56,67). sistance and obesity have not been asso- scription factors, their interaction and
To summarize, insulin signaling is reg- ciated with other HDACi in clinical use. temporal control are reviewed elsewhere
ulated in a complex and not fully under- In conclusion, there is a preclinical ra- (75,76). Here, only a few essential tran-
stood manner, and defects causing in- tionale to conduct clinical trials with scription factors linked to HDACs will
sulin resistance occur on many levels, HDACi other than VPA to investigate the be mentioned. The transcription factor
including at the level of histone and non- therapeutic potential of HDAC inhibition Pdx1 is synthesized in the entire early
histone protein deacetylation. On the in the therapy of insulin resistance. pancreatic rudiment that comprises the
basis of preclinical evidence, inhibition of pancreatic buds (77), and Pdx1 plays a
various HDACs is a promising novel PANCREATIC AND ENDOCRINE CELL central role in the early development of
therapeutic principle to correct the DEVELOPMENT AND HDAC INHIBITION the pancreas, since deletion of Pdx1 re-
insulin-resistant state. The pancreas consists of mainly two sults in complete pancreatic agenesis
Clinical support for this notion, how- types of tissue: (a) the exocrine tissue (78,79). The Pdx1-expressing progenitor
ever, is lacking. Valproate (VPA) used in composed of acinar cells secreting diges- cells differentiate into endo- and exocrine
the long-term treatment of, for example, tive enzymes into the duodenum and (b) cells. It is generally believed that the en-
epilepsy and bipolar disorders is associ- the endocrine tissue that produces hor- docrine differentiation from the Pdx1-ex-
ated with weight gain and hyperinsu- mones such as glucagon (α cells), insulin pressing progenitor cells is initiated by
linemia. However, the causal interaction (β cells), somatostatin (δ cells), pancre- the expression of neurogenin 3 (Ngn3),
and the role of insulin resistance herein atic polypeptide (PP-cells) and ghrelin since Ngn3-deficient mice fail to generate
are not clarified. The development of in- (ε cells). The endocrine tissue, represent- endocrine cells (80), and recently, line-
sulin resistance is suggested in many ing approximately 1% of the fully devel- age-tracing experiments have provided
studies, mainly on the basis of the occur- oped pancreas is organized into ~106 direct evidence that Ngn3-expressing
rence of hyperinsulinemia and on esti- islets that develop concomitantly with cells are islet progenitors (81). Further,
mations of the homeostasis model the ongoing pancreatic morphogenesis. the expression of Pax4 has been linked to
assessment–insulin resistance (HOMA- Characterization of the mechanisms reg- the specific development of the β/δ-cell
IR) index (reviewed in [68]). In a study ulating the development of the endocrine lineage in rodents (82,83).
that more directly measured insulin re- pancreas and especially the insulin- The understanding of the biology of
sistance using a modified frequently producing β cells has undergone an im- HDACs in pancreatic development is
sampled intravenous glucose tolerance mense development in particular with incomplete. HDACs are expressed and
test with tolbutamide, Verrotti et al. (69) the mapping of the network of transcrip- developmentally regulated in the pan-
reported increased insulin resistance tion factors that constitute the decision- creas (82). As described above, HDAC1
only in epileptic patients who gained makers of pancreatic cell fate during is involved in silencing of Pdx1 in a
weight and not in those who remained morphogenesis, proliferation and differ- model of IUGR, leading to failure in β-
lean following 1 year of VPA treatment. entiation. In nonpancreatic tissues, cell development and β-cell dysfunction
Hyperinsulinemia occurred both in VPA- HDACs are not redundant, and it is gen- (38). Additionally, treatment of rat em-
treated patients with epilepsy who erally accepted that individual HDACs bryonic explants with HDACi ex vivo
gained weight as well as in VPA-treated are required for specific functions during enhances and maintains the expression
patients who remained lean (70), and embryogenesis and postnatal life. For profile of the proendocrine marker
fasting hyperinsulinemia in VPA-treated example, HDAC1 is essential for un- Ngn3 (82). As Ngn3 is believed to initi-
patients was not associated with in- restricted cell proliferation by suppress- ate endocrine differentiation from Pdx1-
creased fasting serum proinsulin or ing the expression of cell cycle inhibitors, expressing progenitor cells (80), HDACi
C-peptide concentrations (71). Together, a function unique to HDAC1 (72); dele- may lead to an increased pool of en-
these data do not imply insulin resist- tion of Hdac3 was found to lead to early docrine progenitor cells without modi-
ance as the cause of hyperinsulinemia; embryonic death and apoptosis due to fying the proliferation/apoptosis bal-

3 8 2 | C H R I S T E N S E N E T A L . | M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1
REVIEW ARTICLE

ance (82). Furthermore, HDAC1 associ- sists of three peptide chains (A, B and C).
ates with the sex-determining region The hormone is processed by prohor-
Y-box (SOX)-6, leading to an inhibitory mone convertases 1 and 2, which excise
effect of SOX6 on β-cell proliferation the central part of the protein (the con-
(84) (Figure 3). necting [C]-peptide), leaving the A and B
In zebrafish embryos with HDAC1 chains linked by two disulfide bonds. In-
loss-of-function or HDAC1 knockdown, sulin is finally processed by carboxypep-
the exocrine pancreas failed to form cor- tidase E to produce the mature form that
rectly, whereas no marked effects were is stored as homohexamers in secretory
found on insulin expression (85,86), since vesicles and released in response to in-
ectopic clusters of insulin-expressing creased blood glucose and other stimuli
cells were observed outside the normal (reviewed in [94]). As depicted in Figure 4,
aggregation of endocrine insulin-express- glucose induces both release (see i in fig-
ing cells (85). However, the effect of ure) and transcription of insulin (see ii in
HDAC1 inhibition on endocrine pan- Figure 3. Overview of the roles of HDACs in figure), with the latter depending on at
creas formation is debated (86). pancreatic endocrine cell development. least three β-cell–specific transcription
Different HDACi have distinct effects factors: Pdx1, NeuroD1 and V-maf mus-
on endocrine lineage development. Thus, and thereby different HDACi and the culoaponeurotic fibrosarcoma oncogene
TSA enhances, while VPA suppresses, impact of concentration of HDACi on homologue A (MafA) (95).
β/δ-cell lineage differentiation. In con- the effects observed. The use of more
trast, both inhibitors promote the α/PP- specific HDACi along with careful titra- In Vitro Studies
cell lineage, illustrating the specific series tion studies should allow clarification of The expression of insulin from β cells
of events that control pancreatic develop- these questions. is regulated by acetylation. Thus, at high
ment (82). However, these observations glucose levels, Pdx1 associates with the
cannot be construed to assign specific β-CELL FUNCTION AND HDAC histone acetyltransferase p300, leading to
functions of certain HDAC subtypes in INHIBITION increased acetylation of histone H4 in the
pancreatic development (87,88), since dif- The most important function of the insulin promoter. These events appear to
ferent HDACi have distinct structures pancreatic β cell is to release insulin in re- be necessary for preproinsulin transcrip-
and thereby possibly distinct functions sponse to nutrients, hormones and other tion induced by glucose (96–100). Con-
independent of their inhibitory action on humoral mediators as well as to neuronal versely, at low glucose levels where in-
HDAC activity and since the action of signals to maintain glucose homeostasis sulin production is shut off, the
many HDACi vary with concentration. and lipid and protein metabolism. Insulin acetylation of histone H4 at the insulin
HDACi also promote differentiation of is a peptide hormone synthesized as a promoter is abolished, correlating with
embryonic stem (ES) cells into insulin- longer precursor (proinsulin) that con- recruitment of HDAC1 and -2 to the in-
producing cells, a property of consider-
able importance for β-cell replacement
therapy. TSA inhibits ES cell differentia-
tion, while sodium butyrate (NaB) stimu-
lates early events of pancreatic specifica-
tion in ES cells (89–91). In concordance
with the studies in ES cells, TSA im-
proved the transdifferentiation of bone
marrow stem cells into insulin-producing
cells (92). The inclusion of NaB in early
stages of the differentiation protocol led
to differentiation of human ES cells into
islet-like clusters expressing insulin as
well as glucagon and somatostatin (93).
In summary, HDACi have a potential
to differentiate stem cells into insulin-
producing cells. However, further stud-
ies are needed to clarify the differential Figure 4. Simplified overview of the effect of HDACs and HATs on glucose-induced insulin
importance of various HDAC subtypes expression. NeuD1, NeuroD1.

M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1 | C H R I S T E N S E N E T A L . | 3 8 3
HDACs AS THERAPEUTIC TARGETS IN DIABETES MELLITUS

sulin promoter by Pdx1 (97,101). Neu- (SAHA) and ITF2357 but was slightly in- ment is able to derepress the functional
roD1 also interacts with p300 and is hibited by TSA (27,108). inhibition of residual β-cell mass, it
acetylated by the p300-associated factor would have significant therapeutic po-
(PCAF). This acetylation increases the Clinical Studies tential, not only with respect to treatment
binding of the transcription factor to the As described above, several studies in- of patients with T2D, but also in newly
insulin promoter, leading to enhanced in- vestigated long-term metabolic effects of diagnosed patients with T1D.
sulin gene expression (102). In β cells, treatment with VPA. In general, no
MafA protein is constitutively phospho- changes were reported with respect to β-CELL DESTRUCTION AND HDACi
rylated by glycogen synthase kinase fasting serum insulin and C-peptide in To the best of our knowledge, investi-
(GSK-3), leading to ubiquitination and VPA-treated patients, whereas a single gations of the role of HDACi on the
proteosomal degradation (103). How- study found increased postprandial C- pathogenetic events that lead to β-cell
ever, phosphorylation of MafA is also re- peptide and proinsulin levels (reviewed destruction have been restricted to ex-
quired for binding of the insulin pro- in [71]). Although VPA stimulates insulin aminations on cytokine-induced β-cell
moter and transactivating properties release from isolated islets in vitro (107), death. As described above, the proin-
(104,105). In a non–β-cell system, phos- a more recent report argues against a di- flammatory cytokine IL-1β causes β-cell
phorylated MafA recruits PCAF, the ef- rect stimulatory effect of VPA on insulin apoptosis and is implicated in the patho-
fect of which is not only associated with release in vivo and suggests that the ele- genesis of both T1D and T2D. Further-
increased transcriptional activity but also vated insulin concentrations are a conse- more, two other proinflammatory cy-
with reduced ubiquitination and degra- quence of decreased hepatic degradation tokines, namely TNFα and IFNγ, have
dation of MafA (106). In β cells, the deg- of insulin, as mentioned above (71). been shown to potentiate the toxic ef-
radation of MafA is delayed by exposure Again, it is unclear whether the effects of fects of IL-1β (113–116). In T1D in partic-
to high concentrations of glucose, even VPA are caused by its function as an ular, there is a pronounced selective
though MafA is still phosphorylated HDAC inhibitor, its actions as a fatty destruction of the β cells, and in accor-
(103). This may suggest that high concen- acid derivative or other putative mecha- dance with a role of proinflammatory
trations of glucose allow interaction be- nisms, and more research is needed to cytokines as mediators of the β-cell de-
tween MafA and PCAF (or another shed light on the effects of other HDACi struction, the toxic effects of cytokines
HAT), thereby stabilizing MafA and in- on insulin secretion in vivo. are selective for β cells (117). This selec-
creasing insulin transcription through In summary, insulin production de- tivity is further supported by studies
opening of the chromatin structure in the pends on acetylation and is inhibited by showing that maturation of the β cell
insulin promoter. However, further stud- deacetylation. Taken together, studies ex- makes it prone to the toxic effects of
ies are needed to clarify the putative ef- amining the effects of HDACi on insulin IL-1β (118). On the basis of animal stud-
fect of PCAF on MafA stability and activ- expression indicate that HDACi treat- ies, IL-1β plays an important role in de-
ity in β cells. ment increases insulin expression at low struction of transplanted islet grafts
Taken together, the above-mentioned glucose levels, whereas insulin release is (119–123), and blocking cytokines in
studies suggest that acetylation favors less affected (see Figure 4). As discussed clinical islet transplantation has also
insulin expression and that HDAC activ- later, HDACi protects against β-cell in- been suggested as a future intervention
ity accordingly decreases insulin expres- hibitory and cytotoxic effects of cy- to prevent graft destruction (124).
sion. The HDACi TSA and NaB increase tokines. In addition to a direct induction We originally reported a protective ef-
histone H4 acetylation and enhance in- of apoptosis, cytokines also induce a fect of HDACi treatment against cytokine-
sulin expression at low glucose levels β-cell dedifferentiation associated with induced β-cell death (27), an observation
(3 mmol/L), supporting a repressive role decreased expression and/or activity of now confirmed by us and others
of HDACs on preproinsulin transcrip- Pdx1, NeuroD1 and MafA (109–111). It is (108,125,126). Accordingly, several HDACi
tion (97). Of note, TSA and NaB did not likely that a part of the protective effects (ITF2357, TSA and SAHA) protect against
potentiate acetylation of H4 after expo- of HDACi against cytokine-induced toxi- cytokine-induced reduction in accumu-
sure to high concentrations of glucose city are consequences of a maintained lated insulin secretion (27,108). Of note,
(30 mmol/L) (97). A stimulatory effect of β-cell differentiation. ITF2357 was not only found to prevent
VPA on insulin release has also been re- Recent studies suggest that a 40% re- cytokine-mediated reduction of accumu-
ported in human islets incubated in low duction of the β-cell mass is sufficient to lated insulin release but increased insulin
glucose concentrations (2.8 mmol/L) precipitate clinical symptoms of T1D release over and above that of control
(107). In contrast, accumulated insulin (112) and that a significant proportion of islets (108). Because cytokines at low con-
release from rat islets incubated in 11 insulinopenia is likely to be caused by re- centrations stimulate insulin secretion
mmol/L glucose was unaffected by versible functional inhibition of β cells (127), we interpret these observations to
suberoylanilide hydroxamic acid due to inflammation. If HDACi treat- suggest that ITF2357 selectively prevents

3 8 4 | C H R I S T E N S E N E T A L . | M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1
REVIEW ARTICLE

proapoptotic cytokine signaling while of more than 2,000 genes, many of which ER stress and β-cell death (137). Upon ac-
sparing the cytokine-mediated regulatory are related to pathways signaling apo- tivation of the unfolded protein re-
signaling that stimulates insulin secretion ptosis, cell cycle regulation and endo- sponse, several protective cellular com-
(108). However, a reversal of cytokine-in- plasmic reticulum (ER) stress, but also pensatory mechanisms are initiated to
duced reduction of insulin release to lev- pathways involved in maintenance of stabilize ER homeostasis (for example,
els over and above the control level has differentiation, cell metabolism (for ex- production of chaperones, such as
not been seen with other HDACi (27), and ample, through the Krebs cycle) and al- Hsp70) (138,139). Accordingly, expres-
further studies are needed to substantiate ternative splicing (130). NFκB has re- sion of Hsp70 is also induced by cy-
these observations. ceived much attention for its role in tokines, and overexpression of Hsp70
Cytokines inhibit both first- and cytokine-induced β-cell death and plays protects against cytokine-induced β-cell
second-phase insulin release through de- an essential role in mediating the pro- death (140). In non–β cells, Hsp70 forms
creased expression of insulin (128) and apoptotic effects of cytokines (25,26). complexes with HDAC1, -2 and -3 (141),
proteins that are important for insulin se- HDACi reduce cytokine-induced NFκB but whether these complexes are also
cretion (129). Interestingly, the cytokine- activity and decrease expression of found in β cells and whether HDACs af-
mediated reduction in acute glucose- NFκB-dependent genes (for example, in- fect Hsp70 activity has not been exam-
stimulated insulin secretion, which ducible nitric oxide synthase [iNOS] and ined. Furthermore, TSA increases the ex-
mainly depends on release of preformed inhibitor of kappa B [IκB]) pression of the chaperone BiP (an Hsp70
insulin vesicles and less on de novo in- (27,108,125,126). On the basis of results class protein) in non–β cells (142). In
sulin transcription, translation and pro- from an electrophoretic mobility shift β-cells, overexpression of BiP protects
cessing, is unaffected by HDACi (27). assay showing no effects of HDACi on against in vitro cytotoxic effects of the
Taken together, these data indicate that cytokine-induced NFκB binding to syn- fatty acid palmitate (143) but not of cy-
in addition to antiapoptotic effects of thetic oligonucleotides, HDACi were tokines (144). Whether HDACi modu-
HDACi, these compounds preferentially suggested to modulate the chromatin lates BiP expression in β cells and
protect preproinsulin transcription, structure of NFκB-dependent genes, re- whether BiP is part of the protective
and/or proinsulin translation and pro- sulting in decreased NFκB transactivation mechanism require further investigation.
cessing or expression of genes involved by unknown coactivators (27). In non–β Although the unfolded protein re-
in non–glucose-induced signaling of in- cells, NFκB interacts with HDAC1, -2 and sponse is a protective ER response, pro-
sulin secretion from the inhibitory effects -3 (131,132), but whether these interac- longed unfolded protein response leads
of cytokines (27), with little effect on in- tions also take place in β cells and what to β-cell death by mechanisms that are
sulin granule formation, translocation, the impact is of the interplay on the pro- not fully clarified. The transcription fac-
docking and exocytosis, although this tective effect of HDAC inhibition on tor C/EBP homologous protein (CHOP)
needs to be investigated in more detail. cytokine-mediated β-cell toxicity are is induced upon ER Ca2+ depletion.
The β cell expresses all classical unknown. CHOP may induce apoptosis through
HDACs, albeit at different levels, and In the β cell, IFNγ signal transduction several mechanisms including activation
they are differently regulated by cy- proceeds through the JAK/STAT pathway of the intrinsic apoptotic pathway (137).
tokines (108). On the basis of relative ex- by activation of STAT1. Purified β cells In non–β cells, CHOP interacts with
pressions and regulation by cytokines, from STAT1 knockout mice are protected HDAC1, -5 and -6, and TSA has been
important roles of especially HDAC1, -2, against apoptosis induced by IFNγ and shown to repress degradation of CHOP
-6 and -11 have been suggested (108). IL-1β, suggesting an important role of (145), although other investigators have
However, it remains to be experimentally STAT1 in cytokine-induced β-cell death shown that TSA does not affect the pro-
investigated on which specific HDAC (133). In non–β cells, IFNγ-induced JAK tein level of CHOP (142). Further, the im-
family member(s) cytokine-induced pro- activation and STAT1 activity depend on portance of CHOP and ER stress in
apoptotic signaling depends. Studies that HDAC1, -2 and -3 activity (134). Whether cytokine-induced β-cell death is debated,
include molecular approaches and/or IFNγ-induced STAT1 activity is inhibited since neither knockdown of CHOP nor
more selective inhibitors of individual after HDACi treatment in β cells has to overexpression of BiP protect against
HDAC members are needed to elucidate our knowledge not been investigated. cytokine-induced β-cell death (144). Fur-
this question. IL-1β in combination with IFNγ re- ther, a role of ER stress in the pathogene-
duces the expression of the sarcoplasmic/ sis of T1D in humans is also questioned,
MECHANISMS OF HDACI-MEDIATED endoplasmic reticulum Ca2+-ATPase since CHOP expression was not consis-
β-CELL PROTECTION AGAINST (SERCA)-2 pump, resulting in depletion tently demonstrated in eight pancreatic
INFLAMMATORY ATTACK of the ER Ca2+ stores (135,136). Ca2+ de- autopsies of T1D patients (146).
Exposure of islets to the cytokines pletion hinders proper protein folding, Another mechanism by which cy-
IL-1β and IFNγ modifies the expression leading to the unfolded protein response, tokines induce apoptosis is through di-

M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1 | C H R I S T E N S E N E T A L . | 3 8 5
HDACs AS THERAPEUTIC TARGETS IN DIABETES MELLITUS

rect activation of the intrinsic apoptotic


pathway. Thus, cytokines induce activa-
tion of proapoptotic Bcl-2 proteins, and
inhibition of antiapoptotic Bcl-2 proteins
causes release of cytochrome c from the
mitochondria, followed by activation of
caspase-9 and subsequently caspase-3
activation (147–152). Overexpression of
antiapoptotic Bcl-2 proteins protects
against cytokine-induced β-cell death,
supporting an important role of the Bcl-2
proteins (153,154). Several links between
Bcl-2 proteins and HDACi have been Figure 5. Proinflammatory cytokines induce β-cell apoptosis through regulation of a range
found primary in models of cancer of pro- (red) and antiapoptotic (blue) genes and proteins. The protective effect of HDAC
where high concentrations of HDACi are inhibition likely involves NFκB and possibly other death pathways.
used to induce apoptosis in cancer cells.
In transformed cells, HDACi operates and pathogenesis of T1D and T2D. As tively affecting late diabetic microvascu-
via the proapoptotic Bcl-2 proteins Bim summarized in Table 1 and in Figure 6, lar complications. Taken together, this ev-
(155), Bid (156) and Bax (157), which are there is evidence of genetic association idence provides a strong rationale for
upregulated, processed or translocated between diabetes and HDACs, as there is continuing preclinical studies and initiat-
to the mitochondrial membrane, respec- of HDACi promoting β-cell development, ing clinical trials, with the aim of testing
tively, while expression of the antiapop- proliferation, differentiation and function; the clinical utility of HDACi in diabetes.
totic Bcl-2 protein Bcl-XL is downregu- preventing β-cell inflammatory damage; However, there is still much to be
lated (158). The effects of lower HDACi improving insulin resistance; and posi- learned about the mechanisms of action
concentrations used in inflammatory
diseases on the regulation of the Bcl-2
Table 1. Evidence in favor of HDACs as therapeutic targets in diabetes mellitus.
protein family and an effect of HDACi
on cytokine-induced activation of the in- Genetic associations
trinsic apoptotic pathway in β cells are Genetic association with the 6q21 region harboring HDAC2 with T1D and T2D
yet to be examined. MODY gene mutations directly affect the ability of the MODY proteins to interact with
As summarized in Figure 5, cytokine- HAT/HDACs
induced β-cell apoptosis depends on HDACs and β-cell development and function
HDAC activity, since it is prevented by HDAC1 associates with SOX6 to inhibit β-cell proliferation
HDACi treatment. Although NFκB sig- HDAC1 silences Pdx1 in IUGR, leading to failure in β-cell development and β-cell
naling has been identified as an HDACi dysfunction and reversible in the neonatal state by HDAC inhibition
target, the exact molecular mechanisms HDACi enhances and maintains expression of the proendocrine transcription factor Ngn3
by which HDACi prevents cytokine-in- HDACi stimulates early events of pancreatic specification in embryonic stem cells
duced β-cell death are not clarified, and HDACi improves transdifferentiation of bone marrow–derived stem cells into insulin-
producing cells
effects of HDACi on other key players in
HDACi enhances insulin expression
cytokine-induced signaling such as
JAK/STAT1, Bcl-2 proteins and proteins HDACs and inflammatory pathogenesis of T1D and T2D
involved in ER stress have not yet been HDACi reduces the peripheral blood mononuclear cell production of IL-1β, TNFα and IFNγ
HDACi protects against cytokine-induced reduction in accumulated insulin secretion
investigated. Finally, studies in animal
HDACi protects β cells against cytokine-induced apoptosis by targeting NFκB
models and phase II clinical trials are
needed to shed light on the translational HDACs and insulin resistance
significance of the promising in vitro ef- IUGR promotes HDAC1- and -4–mediated histone deacetylation of the Glut4 promoter in
adult muscle tissue, thereby repressing its transcription
fects of HDACi on cytokine-induced
HDAC5 functions as a transcriptional repressor of GLUT4
β-cell toxicity.
HDACi upregulates PGC-1 expression, GLUT4 translocation and insulin-induced glucose
uptake in skeletal muscle
CONCLUSIONS AND PERSPECTIVES HDAC2 inhibition hyperacetylates IRS-1 and partially restores insulin signaling
The evidence reviewed here indicates
HDACs and late diabetic complications
that HDACs are involved in several bio-
HDACi in part protects against diabetic nephropathy and retinopathy
logical pathways relevant for the etiology

3 8 6 | C H R I S T E N S E N E T A L . | M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1
REVIEW ARTICLE

5. Wilkin TJ. (2007) Changing perspectives in dia-


betes: their impact on its classification. Diabetolo-
gia 50:1587–92.
6. Butler AE, et al. (2003) Beta-cell deficit and
increased beta-cell apoptosis in humans with
type 2 diabetes. Diabetes 52:102–10.
7. Sakuraba H, et al. (2002) Reduced beta-cell mass
and expression of oxidative stress-related DNA
damage in the islet of Japanese type II diabetic
patients. Diabetologia 45:85–96.
8. Yoon KH, et al. (2003) Selective beta-cell loss and
alpha-cell expansion in patients with type 2 dia-
betes mellitus in Korea. J. Clin. Endocrinol. Metab.
88:2300–8.
9. Ehses JA, Ellingsgaard H, Boni-Schnetzler M,
Donath MY. (2009) Pancreatic islet inflammation
in type 2 diabetes: from alpha and beta cell com-
pensation to dysfunction. Arch. Physiol. Biochem.
115:240–7.
10. Masters SL, et al. (2010) Activation of the NLRP3
inflammasome by islet amyloid polypeptide pro-
vides a mechanism for enhanced IL-1beta in type
Figure 6. Overview of the therapeutic potential of HDACi in diabetes. 2 diabetes. Nat. Immunol. 11:897–904.
11. Mandrup-Poulsen T. (2010) IAPP boosts islet
and the differential importance of the 11 velopment to pave the way for future- macrophage IL-1 in type 2 diabetes. Nat. Im-
munol. 11:881–3.
classic HDACs, the sirtuins and the generation HDACi with higher speci-
12. Donath MY, Storling J, Berchtold LA, Billestrup
HATs controlling the acetylation balance ficity and safety for the treatment of dia- N, Mandrup-Poulsen T. (2008) Cytokines and
in diabetes. An important task will be to betes and other inflammatory diseases. beta-cell biology: from concept to clinical transla-
define the relative importance of acety- tion. Endocr. Rev. 29:334–50.
lated histones, transcription factors and ACKNOWLEDGMENTS 13. Mandrup-Poulsen T. (1996) The role of inter-
leukin-1 in the pathogenesis of IDDM. Diabetolo-
other nuclear, cytosolic and compart- This review was written with financial
gia 39:1005–29.
mentalized proteins in the β-cell acety- support from the Novo Nordisk Founda- 14. Stienstra R, et al. (2010) The inflammasome-medi-
lome, and, eventually, the interaction be- tion, the Juvenile Diabetes Research Foun- ated caspase-1 activation controls adipocyte dif-
tween acetylation and other dation International grant 26-2008-893, the ferentiation and insulin sensitivity. Cell Metab.
12:593–605.
posttranslational protein modifications University of Copenhagen and the Novo
15. Maedler K, et al. (2002) Glucose-induced beta cell
needs to be interpreted into the lan- Nordisk TRAP Program. production of IL-1beta contributes to glucotoxicity
guage now known as the histone or pro- in human pancreatic islets. J. Clin. Invest. 110:851–60.
tein code. Lastly, the enigma of how DISCLOSURE 16. Maedler K, et al. (2004) Leptin modulates beta
HDAC inhibition, an apparently nonspe- The authors declare that they have no cell expression of IL-1 receptor antagonist and re-
lease of IL-1beta in human islets. Proc. Natl. Acad.
cific treatment, can exert therapeutic competing interests as defined by Molec-
Sci. U. S. A. 101:8138–43.
benefit to so many diverse disorders ular Medicine, or other interests that 17. Sauter NS, Schulthess FT, Galasso R, Castellani
needs to be unraveled. Does HDACi might be perceived to influence the re- LW, Maedler K. (2008) The antiinflammatory cy-
only reset a disturbed protein acetyla- sults and discussion reported in this tokine interleukin-1 receptor antagonist protects
tion balance as a result of cell stress paper. from high-fat diet-induced hyperglycemia. En-
docrinology 149:2208–18.
without affecting cell homeostasis? Why
18. Spranger J, et al. (2003) Inflammatory cytokines
does HDACi-mediated hyperacetylation REFERENCES and the risk to develop type 2 diabetes: results of
mainly associated with increased gene 1. Raj SM, et al. (2009) No association of multiple the prospective population-based European Pro-
type 2 diabetes loci with type 1 diabetes. Dia-
transcription counteract inflammatory spective Investigation into Cancer and Nutrition
betologia 52:2109–16. (EPIC)-Potsdam Study. Diabetes 52:812–7.
gene expression changes? Is this a conse-
2. Barrett JC, et al. (2009) Genome-wide association 19. Larsen CM, et al. (2007) Interleukin-1-receptor
quence of expressional upregulation of study and meta-analysis find that over 40 loci af- antagonist in type 2 diabetes mellitus. N. Engl. J.
genes encoding antiapoptotic proteins or fect risk of type 1 diabetes. Nat. Genet. 41:703–7. Med. 356:1517–26.
microRNAs? How can this be reconciled 3. Willcox A, Richardson SJ, Bone AJ, Foulis AK, 20. Larsen CM, et al. (2009) Sustained effects of inter-
with an inhibitory effect of HDACi on Morgan NG. (2009) Analysis of islet inflamma- leukin-1 receptor antagonist treatment in type 2
tion in human type 1 diabetes. Clin. Exp. Im- diabetes. Diabetes Care 32:1663–8.
NFκB transcriptional activity? These
munol. 155:173–81. 21. Halili MA, Andrews MR, Sweet MJ, Fairlie DP.
basic research questions and many more 4. Prokopenko I, McCarthy MI, Lindgren CM. (2009) Histone deacetylase inhibitors in inflam-
will have to be addressed in parallel (2008) Type 2 diabetes: new genes, new under- matory disease. Curr. Top. Med. Chem. 9:309–19.
with further preclinical and clinical de- standing. Trends Genet. 24:613–21. 22. Gregoretti IV, Lee YM, Goodson HV. (2004) Mo-

M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1 | C H R I S T E N S E N E T A L . | 3 8 7
HDACs AS THERAPEUTIC TARGETS IN DIABETES MELLITUS

lecular evolution of the histone deacetylase fam- M, Devaskar SU. (2008) Histone code modifica- stimulated whole body glucose disposal in con-
ily: functional implications of phylogenetic anal- tions repress glucose transporter 4 expression in scious mice. J. Clin. Invest. 95:429–32.
ysis. J. Mol. Biol. 338:17–31. the intrauterine growth-restricted offspring. J. Biol. 57. Leturque A, Loizeau M, Vaulont S, Salminen M,
23. Choudhary C, et al. (2009) Lysine acetylation tar- Chem. 283:13611–26. Girard J. (1996) Improvement of insulin action in
gets protein complexes and co-regulates major 40. Kim SJ, Nian C, McIntosh CH. (2009) Glucose- diabetic transgenic mice selectively overexpress-
cellular functions. Science 325:834–40. dependent insulinotropic polypeptide and ing GLUT4 in skeletal muscle. Diabetes 45:23–7.
24. Mankan AK, Lawless MW, Gray SG, Kelleher D, glucagon-like peptide-1 modulate beta-cell chro- 58. Thai MV, Guruswamy S, Cao KT, Pessin JE,
McManus R. (2009) NF-kappaB regulation: the matin structure. J. Biol. Chem. 284:12896–904. Olson AL. (1998) Myocyte enhancer factor 2
nuclear response. J. Cell. Mol. Med. 13:631–43. 41. Gray SG, De Meyts P. (2005) Role of histone and (MEF2)-binding site is required for GLUT4 gene
25. Giannoukakis N, Rudert WA, Trucco M, Robbins transcription factor acetylation in diabetes patho- expression in transgenic mice: regulation of
PD. (2000) Protection of human islets from the ef- genesis. Diabetes Metab. Res. Rev. 21:416–33. MEF2 DNA binding activity in insulin-deficient
fects of interleukin-1beta by adenoviral gene 42. Camelo S, et al. (2005) Transcriptional therapy diabetes. J. Biol. Chem. 273:14285–92.
transfer of an Ikappa B repressor. J. Biol. Chem. with the histone deacetylase inhibitor trichostatin 59. McGee SL, et al. (2008) AMP-activated protein ki-
275:36509–13. A ameliorates experimental autoimmune en- nase regulates GLUT4 transcription by phospho-
26. Heimberg H, et al. (2001) Inhibition of cytokine- cephalomyelitis. J. Neuroimmunol. 164:10–21. rylating histone deacetylase 5. Diabetes 57:860–7.
induced NF-kappaB activation by adenovirus- 43. Lin HS, et al. (2007) Anti-rheumatic activities of 60. Sparling DP, Griesel BA, Weems J, Olson AL.
mediated expression of a NF-kappaB super- histone deacetylase (HDAC) inhibitors in vivo in (2008) GLUT4 enhancer factor (GEF) interacts
repressor prevents beta-cell apoptosis. Diabetes collagen-induced arthritis in rodents. Br. J. Phar- with MEF2A and HDAC5 to regulate the GLUT4
50:2219–24. macol. 150:862–72. promoter in adipocytes. J. Biol. Chem.
27. Larsen L, et al. (2007) Inhibition of histone 44. Suuronen T, Huuskonen J, Pihlaja R, Kyrylenko 283:7429–37.
deacetylases prevents cytokine-induced toxicity S, Salminen A. (2003) Regulation of microglial in- 61. McKinsey TA, Zhang CL, Olson EN. (2001) Con-
in beta cells. Diabetologia 50:779–89. flammatory response by histone deacetylase in- trol of muscle development by dueling HATs
28. Lee HB, Noh H, Seo JY, Yu MR, Ha H. (2007) His- hibitors. J. Neurochem. 87:407–16. and HDACs. Curr. Opin. Genet. Dev. 11:497–504.
tone deacetylase inhibitors: a novel class of ther- 45. Suh HS, Choi S, Khattar P, Choi N, Lee SC. (2010) 62. McKinsey TA, Zhang CL, Olson EN. (2000) Acti-
apeutic agents in diabetic nephropathy. Kidney Histone deacetylase inhibitors suppress the ex- vation of the myocyte enhancer factor-2 transcrip-
Int. Suppl. S61–6. pression of inflammatory and innate immune re- tion factor by calcium/calmodulin-dependent
29. Villeneuve LM, Natarajan R. (2010) The role of sponse genes in human microglia and astrocytes. protein kinase-stimulated binding of 14–3–3 to hi-
epigenetics in the pathology of diabetic compli- J. Neuroimmune Pharmacol. 5:521–32. stone deacetylase 5. Proc. Natl. Acad. Sci. U. S. A.
cations. Am. J. Physiol. Renal Physiol. 299:F14–25. 46. Miao F, Gonzalo IG, Lanting L, Natarajan R. 97:14400–5.
30. Crosson CE, Mani SK, Husain S, Alsarraf O, (2004) In vivo chromatin remodeling events lead- 63. McGee SL, Hargreaves M. (2004) Exercise and
Menick DR. (2010) Inhibition of histone deacety- ing to inflammatory gene transcription under di- myocyte enhancer factor 2 regulation in human
lase protects the retina from ischemic injury. In- abetic conditions. J. Biol. Chem. 279:18091–7. skeletal muscle. Diabetes 53:1208–14.
vest. Ophthalmol. Vis. Sci. 51:3639–45. 47. Shanmugam N, Reddy MA, Guha M, Natarajan 64. Michael LF, et al. (2001) Restoration of insulin-
31. Bonnefond A, Froguel P, Vaxillaire M. (2010) The R. (2003) High glucose-induced expression of sensitive glucose transporter (GLUT4) gene ex-
emerging genetics of type 2 diabetes. Trends Mol. proinflammatory cytokine and chemokine genes pression in muscle cells by the transcriptional
Med. 16:407–16. in monocytic cells. Diabetes 52:1256–64. coactivator PGC-1. Proc. Natl. Acad. Sci. U. S. A.
32. Pociot F, et al. (2010) Genetics of type 1 diabetes: 48. Halili MA, et al. (2010) Differential effects of se- 98:3820–5.
what’s next? Diabetes 59:1561–71. lective HDAC inhibitors on macrophage inflam- 65. Crunkhorn S, et al. (2007) Peroxisome proliferator
33. Nerup J, Pociot F. (2001) A genomewide scan for matory responses to the Toll-like receptor 4 ago- activator receptor gamma coactivator-1 expres-
type 1-diabetes susceptibility in Scandinavian nist LPS. J. Leukoc. Biol. 87:1103–14. sion is reduced in obesity: potential pathogenic
families: identification of new loci with evidence 49. Leoni F, et al. (2005) The histone deacetylase in- role of saturated fatty acids and p38 mitogen-ac-
of interactions. Am. J. Hum. Genet. 69:1301–13. hibitor ITF2357 reduces production of pro- tivated protein kinase activation. J. Biol. Chem.
34. Xiang K, et al. (2004) Genome-wide search for inflammatory cytokines in vitro and systemic in- 282:15439–50.
type 2 diabetes/impaired glucose homeostasis flammation in vivo. Mol. Med. 11:1–15. 66. Bonen A. (2009) PGC-1alpha-induced improve-
susceptibility genes in the Chinese: significant 50. Choi K, Kim YB. (2010) Molecular mechanism of ments in skeletal muscle metabolism and insulin
linkage to chromosome 6q21-q23 and chromo- insulin resistance in obesity and type 2 diabetes. sensitivity. Appl. Physiol. Nutr. Metab. 34:307–14.
some 1q21-q24. Diabetes 53:228–34. Korean J. Intern. Med. 25:119–29. 67. McGee SL, Hargreaves M. (2010) Histone modifi-
35. Redondo MJ, Fain PR, Eisenbarth GS. (2001) Ge- 51. Greenbaum CJ. (2002) Insulin resistance in type 1 cations and skeletal muscle metabolic gene ex-
netics of type 1A diabetes. Recent Prog. Horm. Res. diabetes. Diabetes Metab. Res. Rev. 18:192–200. pression. Clin. Exp. Pharmacol. Physiol. 37:392–6.
56:69–89. 52. Scheen AJ. (2005) Diabetes mellitus in the el- 68. Verrotti A, D’Egidio C, Mohn A, Coppola G,
36. Poulsen P, Kyvik KO, Vaag A, Beck-Nielsen H. derly: insulin resistance and/or impaired insulin Chiarelli F. (2010) Weight gain following treatment
(1999) Heritability of type II (non-insulin-depen- secretion? Diabetes Metab. 31:5S27–25S34. with valproic acid: pathogenetic mechanisms and
dent) diabetes mellitus and abnormal glucose 53. Mercado MM, McLenithan JC, Silver KD, clinical implications. Obes. Rev. 12:e32–43.
tolerance: a population-based twin study. Dia- Shuldiner AR. (2002) Genetics of insulin resist- 69. Verrotti A, et al. (2002) Insulin resistance in
betologia 42:139–45. ance. Curr. Diab. Rep. 2:83–95. epileptic girls who gain weight after therapy
37. Pinney SE, Simmons RA. (2010) Epigenetic 54. Takigawa-Imamura H, et al. (2003) Stimulation of with valproic acid. J. Child Neurol. 17:265–8.
mechanisms in the development of type 2 dia- glucose uptake in muscle cells by prolonged 70. Pylvanen V, et al. (2002) Serum insulin and leptin
betes. Trends Endocrinol. Metab. 21:223–29. treatment with scriptide, a histone deacetylase levels in valproate-associated obesity. Epilepsia
38. Park JH, Stoffers DA, Nicholls RD, Simmons RA. inhibitor. Biosci. Biotechnol. Biochem. 67:1499–1506. 43:514–17.
(2008) Development of type 2 diabetes following 55. Kaiser C, James SR. (2004) Acetylation of insulin 71. Pylvanen V, Pakarinen A, Knip M, Isojarvi J.
intrauterine growth retardation in rats is associ- receptor substrate-1 is permissive for tyrosine (2006) Characterization of insulin secretion in
ated with progressive epigenetic silencing of phosphorylation. BMC Biol 2:23. valproate-treated patients with epilepsy. Epilepsia
Pdx1. J. Clin. Invest. 118:2316–24. 56. Ren JM, et al. (1995) Overexpression of Glut4 47:1460–4.
39. Raychaudhuri N, Raychaudhuri S, Thamotharan protein in muscle increases basal and insulin- 72. Lagger G, et al. (2002) Essential function of histone

3 8 8 | C H R I S T E N S E N E T A L . | M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1
REVIEW ARTICLE

deacetylase 1 in proliferation control and CDK in- 90. Lee JH, Hart SR, Skalnik DG. (2004) Histone 106. Rocques N, et al. (2007) GSK-3-mediated phos-
hibitor repression. EMBO J. 21:2672–81. deacetylase activity is required for embryonic phorylation enhances Maf-transforming activ-
73. Bhaskara S, et al. (2008) Deletion of histone deacety- stem cell differentiation. Genesis 38:32–8. ity. Mol. Cell 28:584–97.
lase 3 reveals critical roles in S phase progression 91. Li L, et al. (2008) Combination of GLP-1 and 107. Luef GJ, Lechleitner M, Bauer G, Trinka E,
and DNA damage control. Mol. Cell 30:61–72. sodium butyrate promote differentiation of pan- Hengster P. (2003) Valproic acid modulates islet
74. Majdzadeh N, et al. (2008) HDAC4 inhibits cell- creatic progenitor cells into insulin-producing cell insulin secretion: a possible mechanism of
cycle progression and protects neurons from cell cells. Tissue Cell 40:437–45. weight gain in epilepsy patients. Epilepsy Res.
death. Dev. Neurobiol. 68:1076–92. 92. Tayaramma T, Ma B, Rohde M, Mayer H. (2006) 55:53–8.
75. Collombat P, Hecksher-Sorensen J, Serup P, Man- Chromatin-remodeling factors allow differentia- 108. Lundh M, et al. (2010) Lysine deacetylases are
souri A. (2006) Specifying pancreatic endocrine tion of bone marrow cells into insulin-producing produced in pancreatic beta cells and are differ-
cell fates. Mech. Dev. 123:501–12. cells. Stem Cells 24:2858–67. entially regulated by proinflammatory cy-
76. Chakrabarti SK, Mirmira RG. (2003) Transcrip- 93. Jiang J, et al. (2007) Generation of insulin- tokines. Diabetologia 53:2569–78.
tion factors direct the development and function producing islet-like clusters from human em- 109. Eizirik DL, Mandrup-Poulsen T. (2001) A choice
of pancreatic beta cells. Trends Endocrinol. Metab. bryonic stem cells. Stem Cells 25:1940–53. of death: the signal-transduction of immune-
14:78–84. 94. Weiss MA. (2009) Proinsulin and the genetics of mediated beta-cell apoptosis. Diabetologia
77. Ohlsson H, Karlsson K, Edlund T. (1993) IPF1, a diabetes mellitus. J. Biol. Chem. 284:19159–63. 44:2115–33.
homeodomain-containing transactivator of the 95. Aramata S, Han SI, Yasuda K, Kataoka K. (2005) 110. Maier B, et al. (2010) The unique hypusine mod-
insulin gene. EMBO J. 12:4251–9. Synergistic activation of the insulin gene pro- ification of eIF5A promotes islet beta cell in-
78. Jonsson J, Carlsson L, Edlund T, Edlund H. moter by the beta-cell enriched transcription flammation and dysfunction in mice. J. Clin. In-
(1994) Insulin-promoter-factor 1 is required for factors MafA, Beta2, and Pdx1. Biochim. Biophys. vest. 120:2156–70.
pancreas development in mice. Nature 371:606–9. Acta. 1730:41–6. 111. Oetjen E, et al. (2007) Inhibition of MafA tran-
79. Offield MF, et al. (1996) PDX-1 is required for 96. Mosley AL, Corbett JA, Ozcan S. (2004) Glucose scriptional activity and human insulin gene
pancreatic outgrowth and differentiation of the
regulation of insulin gene expression requires transcription by interleukin-1beta and mitogen-
rostral duodenum. Development 122:983–95.
the recruitment of p300 by the beta-cell-specific activated protein kinase kinase kinase in pan-
80. Gradwohl G, Dierich A, LeMeur M, Guillemot F.
transcription factor Pdx-1. Mol. Endocrinol. creatic islet beta cells. Diabetologia 50:1678–87.
(2000) Neurogenin3 is required for the develop-
18:2279–90. 112. Atkinson MA, Gianani R. (2009) The pancreas
ment of the four endocrine cell lineages of the
97. Mosley AL, Ozcan S. (2003) Glucose regulates in human type 1 diabetes: providing new an-
pancreas. Proc. Natl. Acad. Sci. U. S. A. 97:1607–11.
insulin gene transcription by hyperacetylation swers to age-old questions. Curr. Opin. En-
81. Gu G, Dubauskaite J, Melton DA. (2002) Direct
of histone h4. J. Biol. Chem. 278:19660–6. docrinol. Diabetes Obes. 16:279–85.
evidence for the pancreatic lineage: NGN3+ cells
98. Evans-Molina C, et al. (2007) Glucose regulation 113. Mandrup-Poulsen T, Bendtzen K, Dinarello CA,
are islet progenitors and are distinct from duct
of insulin gene transcription and pre-mRNA Nerup J. (1987) Human tumor necrosis factor
progenitors. Development 129:2447–57.
processing in human islets. Diabetes 56:827–35. potentiates human interleukin 1-mediated rat
82. Haumaitre C, Lenoir O, Scharfmann R. (2008) Hi-
99. Qiu Y, Guo M, Huang S, Stein R. (2002) Insulin pancreatic beta-cell cytotoxicity. J. Immunol.
stone deacetylase inhibitors modify pancreatic
gene transcription is mediated by interactions 139:4077–82.
cell fate determination and amplify endocrine
between the p300 coactivator and PDX-1, 114. Pukel C, Baquerizo H, Rabinovitch A. (1988)
progenitors. Mol. Cell. Biol. 28:6373–83.
BETA2, and E47. Mol. Cell. Biol. 22:412–20. Destruction of rat islet cell monolayers by cy-
83. Sosa-Pineda B, Chowdhury K, Torres M, Oliver
100. Qiu Y, Sharma A, Stein R. (1998) p300 mediates tokines: synergistic interactions of interferon-
G, Gruss P. (1997) The Pax4 gene is essential for
transcriptional stimulation by the basic helix- gamma, tumor necrosis factor, lymphotoxin,
differentiation of insulin-producing beta cells in
loop-helix activators of the insulin gene. Mol. and interleukin 1. Diabetes 37:133–6.
the mammalian pancreas. Nature 386:399–402.
84. Iguchi H, et al. (2007) SOX6 suppresses cyclin D1 Cell. Biol. 18:2957–64. 115. Cetkovic-Cvrlje M, Eizirik DL. (1994) TNF-
promoter activity by interacting with beta- 101. Mosley AL, Ozcan S. (2004) The pancreatic duo- alpha and IFN-gamma potentiate the deleteri-
catenin and histone deacetylase 1, and its down- denal homeobox-1 protein (Pdx-1) interacts with ous effects of IL-1 beta on mouse pancreatic
regulation induces pancreatic beta-cell prolifera- histone deacetylases Hdac-1 and Hdac-2 on low islets mainly via generation of nitric oxide. Cy-
tion. J. Biol. Chem. 282:19052–61. levels of glucose. J. Biol. Chem. 279:54241–7. tokine 6:399–406.
85. Noel ES, et al. (2008) Organ-specific requirements 102. Qiu Y, Guo M, Huang S, Stein R. (2004) Acetyla- 116. Rabinovitch A, et al. (1994) Human pancreatic
for Hdac1 in liver and pancreas formation. Dev. tion of the BETA2 transcription factor by p300- islet beta-cell destruction by cytokines is inde-
Biol. 322:237–50. associated factor is important in insulin gene pendent of nitric oxide production. J. Clin. En-
86. Farooq M, et al. (2008) Histone deacetylase 3 expression. J. Biol. Chem. 279:9796–802. docrinol. Metab. 79:1058–62.
(hdac3) is specifically required for liver develop- 103. Han SI, Aramata S, Yasuda K, Kataoka K. (2007) 117. Mandrup-Poulsen T, et al. (1987) Ultrastructural
ment in zebrafish. Dev. Biol. 317:336–53. MafA stability in pancreatic beta cells is regulated studies of time-course and cellular specificity of
87. Bradner JE, et al. (2010) Chemical genetic strategy by glucose and is dependent on its constitutive interleukin-1 mediated islet cytotoxicity. Acta
identifies histone deacetylase 1 (HDAC1) and phosphorylation at multiple sites by glycogen Pathol. Microbiol. Immunol. Scand. C. 95:55–63.
HDAC2 as therapeutic targets in sickle cell dis- synthase kinase 3. Mol. Cell. Biol. 27:6593–605. 118. Nielsen K, et al. (1999) Beta-cell maturation
ease. Proc. Natl. Acad. Sci. U. S. A. 107:12617–22. 104. Zhao L, Cissell MA, Henderson E, Colbran R, leads to in vitro sensitivity to cytotoxins. Dia-
88. Matalon S, et al. (2010) The histone deacetylase in- Stein R. (2000) The RIPE3b1 activator of the in- betes 48:2324–32.
hibitor ITF2357 decreases surface CXCR4 and CCR5 sulin gene is composed of a protein(s) of ap- 119. Drage M, et al. (2002) Nondepleting anti-CD4
expression on CD4(+) T-cells and monocytes and is proximately 43 kDa, whose DNA binding activ- and soluble interleukin-1 receptor prevent auto-
superior to valproic acid for latent HIV-1 expression ity is inhibited by protein phosphatase immune destruction of syngeneic islet grafts in
in vitro. J. Acquir. Immune Defic. Syndr. 54:1–9. treatment. J. Biol. Chem. 275:10532–7. diabetic NOD mice. Transplantation 74:611–19.
89. Goicoa S, Alvarez S, Ricordi C, Inverardi L, 105. Guo S, et al. (2009) The stability and transactiva- 120. Gysemans C, et al. (2003) Prevention of primary
Dominguez-Bendala J. (2006) Sodium butyrate ac- tion potential of the mammalian MafA tran- non-function of islet xenografts in autoimmune
tivates genes of early pancreatic development in scription factor are regulated by serine 65 phos- diabetic NOD mice by anti-inflammatory
embryonic stem cells. Cloning Stem Cells 8:140–9. phorylation. J. Biol. Chem. 284:759–65. agents. Diabetologia 46:1115–23.

M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1 | C H R I S T E N S E N E T A L . | 3 8 9
HDACs AS THERAPEUTIC TARGETS IN DIABETES MELLITUS

121. Sandberg JO, Eizirik DL, Sandler S. (1997) IL-1 135. Cardozo AK, et al. (2005) Cytokines downregu- an early event contributing to β-cell apoptosis.
receptor antagonist inhibits recurrence of dis- late the sarcoendoplasmic reticulum pump Cell Death Differ. 18:328–37.
ease after syngeneic pancreatic islet transplanta- Ca2+ ATPase 2b and deplete endoplasmic retic- 151. Gurzov EN, et al. (2010) p53 up-regulated modu-
tion to spontaneously diabetic non-obese dia- ulum Ca2+, leading to induction of endoplas- lator of apoptosis (PUMA) activation contributes
betic (NOD) mice. Clin. Exp. Immunol. 108:314–7. mic reticulum stress in pancreatic beta-cells. Di- to pancreatic beta-cell apoptosis induced by
122. Sandberg JO, Eizirik DL, Sandler S, Tracey DE, abetes 54:452–61. proinflammatory cytokines and endoplasmic
Andersson A. (1993) Treatment with an inter- 136. Oyadomari S, et al. (2001) Nitric oxide-induced reticulum stress. J. Biol. Chem. 285:19910–20.
leukin-1 receptor antagonist protein prolongs apoptosis in pancreatic beta cells is mediated by 152. Gurzov EN, et al. (2009) Signaling by IL-
mouse islet allograft survival. Diabetes 42:1845–51. the endoplasmic reticulum stress pathway. Proc. 1beta+IFN-gamma and ER stress converge on
123. Tellez N, et al. (2007) Adenoviral overproduc- Natl. Acad. Sci. U. S. A. 98:10845–50. DP5/Hrk activation: a novel mechanism for
tion of interleukin-1 receptor antagonist in- 137. Eizirik DL, Cardozo AK, Cnop M. (2008) The pancreatic beta-cell apoptosis. Cell Death Differ.
creases beta cell replication and mass in syn- role for endoplasmic reticulum stress in dia- 16:1539–50.
geneically transplanted islets, and improves betes mellitus. Endocr. Rev. 29:42–61. 153. Rabinovitch A, et al. (1999) Transfection of
metabolic outcome. Diabetologia 50:602–11. 138. Eizirik DL, Cnop M. (2010) ER stress in pancre- human pancreatic islets with an anti-apoptotic
124. Amoli MM, Larijani B. (2006) Would blockage atic beta cells: the thin red line between adapta- gene (bcl-2) protects beta-cells from cytokine-in-
of cytokines improve the outcome of pancreatic tion and failure. Sci Signal 3:pe7. duced destruction. Diabetes 48:1223–9.
islet transplantation? Med. Hypotheses 66:816–9. 139. Weber SM, Chambers KT, Bensch KG, Scarim AL, 154. Holohan C, Szegezdi E, Ritter T, O’Brien T,
125. Susick L, Veluthakal R, Suresh MV, Hadden T, Corbett JA. (2004) PPARgamma ligands induce Samali A. (2008) Cytokine-induced beta-cell
Kowluru A. (2008) Regulatory roles for histone ER stress in pancreatic beta-cells: ER stress activa- apoptosis is NO-dependent, mitochondria-
deacetylation in IL-1beta-induced nitric oxide tion results in attenuation of cytokine signaling. mediated and inhibited by BCL-XL. J. Cell Mol.
release in pancreatic beta-cells. J. Cell. Mol. Med. Am. J. Physiol. Endocrinol. Metab. 287:E1171–7. Med. 12:591–606.
12:1571–1583. 140. Mokhtari D, et al. (2009) Increased Hsp70 ex- 155. Chen S, Dai Y, Pei XY, Grant S. (2009) Bim up-
126. Susick L, Senanayake T, Veluthakal R, Woster pression attenuates cytokine-induced cell death regulation by histone deacetylase inhibitors me-
PM, Kowluru A. (2009) A novel histone deacety- in islets of Langerhans from Shb knockout mice. diates interactions with the Bcl-2 antagonist
lase inhibitor prevents IL-1beta induced meta- Biochem. Biophys. Res. Commun. 387:553–7. ABT-737: evidence for distinct roles for Bcl-2,
bolic dysfunction in pancreatic beta-cells. J. Cell. 141. Johnson CA, White DA, Lavender JS, O’Neill Bcl-xL, and Mcl-1. Mol. Cell. Biol. 29:6149–69.
Mol. Med. 13:1877–85. LP, Turner BM. (2002) Human class I histone 156. Ruefli AA, et al. (2001) The histone deacetylase
127. Spinas GA, et al. (1986) Low concentrations of deacetylase complexes show enhanced catalytic inhibitor and chemotherapeutic agent suberoy-
interleukin-1 stimulate and high concentrations activity in the presence of ATP and co-immuno- lanilide hydroxamic acid (SAHA) induces a
inhibit insulin release from isolated rat islets of precipitate with the ATP-dependent chaperone cell-death pathway characterized by cleavage of
Langerhans. Acta. Endocrinol. (Copenh.) 113:551–8. protein Hsp70. J. Biol. Chem. 277:9590–7. Bid and production of reactive oxygen species.
128. Sandler S, Andersson A, Hellerstrom C. (1987) 142. Shi Y, Gerritsma D, Bowes AJ, Capretta A, Proc. Natl. Acad. Sci. U. S. A. 98:10833–8.
Inhibitory effects of interleukin 1 on insulin se- Werstuck GH. (2007) Induction of GRP78 by val- 157. Sutheesophon K, et al. (2006) Histone deacety-
cretion, insulin biosynthesis, and oxidative me- proic acid is dependent upon histone deacetylase lase inhibitor depsipeptide (FK228) induces
tabolism of isolated rat pancreatic islets. En- inhibition. Bioorg. Med. Chem. Lett. 17:4491–4. apoptosis in leukemic cells by facilitating mito-
docrinology 121:1424–31. 143. Laybutt DR, et al. (2007) Endoplasmic reticulum chondrial translocation of Bax, which is en-
129. Ohara-Imaizumi M, Cardozo AK, Kikuta T, stress contributes to beta cell apoptosis in type hanced by the proteasome inhibitor bortezomib.
Eizirik DL, Nagamatsu S. (2004) The cytokine 2 diabetes. Diabetologia 50:752–63. Acta. Haematol. 115:78–90.
interleukin-1beta reduces the docking and fu- 144. Akerfeldt MC, et al. (2008) Cytokine-induced 158. Cao XX, Mohuiddin I, Ece F, McConkey DJ,
sion of insulin granules in pancreatic beta-cells, beta-cell death is independent of endoplasmic Smythe WR. (2001) Histone deacetylase in-
preferentially decreasing the first phase of exo- reticulum stress signaling. Diabetes 57:3034–44. hibitor downregulation of bcl-xl gene expres-
cytosis. J. Biol. Chem. 279:41271–4. 145. Ohoka N, Hattori T, Kitagawa M, Onozaki K, sion leads to apoptotic cell death in mesothe-
130. Ortis F, et al. (2010) Cytokines interleukin-1beta Hayashi H. (2007) Critical and functional regu- lioma. Am. J. Respir. Cell Mol. Biol. 25:562–68.
and tumor necrosis factor-alpha regulate differ- lation of CHOP (C/EBP homologous protein)
ent transcriptional and alternative splicing net- through the N-terminal portion. J. Biol. Chem.
works in primary beta-cells. Diabetes 59:358–74. 282:35687–94.
131. Ashburner BP, Westerheide SD, Baldwin AS Jr. 146. Huang CJ, et al. (2007) High expression rates of
(2001) The p65 (RelA) subunit of NF-kappaB in- human islet amyloid polypeptide induce endo-
teracts with the histone deacetylase (HDAC) core- plasmic reticulum stress mediated beta-cell apo-
pressors HDAC1 and HDAC2 to negatively regu- ptosis, a characteristic of humans with type 2
late gene expression. Mol. Cell. Biol. 21:7065–77. but not type 1 diabetes. Diabetes 56:2016–27.
132. Kiernan R, et al. (2003) Post-activation turn-off of 147. Grunnet LG, et al. (2009) Proinflammatory cy-
NF-kappa B-dependent transcription is regulated tokines activate the intrinsic apoptotic pathway
by acetylation of p65. J. Biol. Chem. 278:2758–66. in beta-cells. Diabetes 58:1807–15.
133. Gysemans CA, et al. (2005) Disruption of the 148. Chang I, et al. (2004) Role of calcium in pancre-
gamma-interferon signaling pathway at the atic islet cell death by IFN-gamma/TNF-alpha.
level of signal transducer and activator of tran- J. Immunol. 172:7008–14.
scription-1 prevents immune destruction of 149. McKenzie MD, et al. (2008) Proapoptotic BH3-
beta-cells. Diabetes 54:2396–403. only protein Bid is essential for death receptor-
134. Klampfer L, Huang J, Swaby LA, Augenlicht L. induced apoptosis of pancreatic beta-cells. Dia-
(2004) Requirement of histone deacetylase ac- betes 57:1284–92.
tivity for signaling by STAT1. J. Biol. Chem. 150. Allagnat F, et al. (2011) Mcl-1 downregulation
279:30358–68. by pro-inflammatory cytokines and palmitate is

3 9 0 | C H R I S T E N S E N E T A L . | M O L M E D 1 7 ( 5 - 6 ) 3 7 8 - 3 9 0 , M AY- J U N E 2 0 1 1

You might also like