Role of Hdac Inhibitors in Diabetes Mellitus: Sciencedirect

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

G Model

RETRAM 3233 No. of Pages 6

Current Research in Translational Medicine xxx (2019) xxx–xxx

Available online at

ScienceDirect
www.sciencedirect.com

General review

Role of HDAC inhibitors in diabetes mellitus


Rashita Makkara , Tapan Behla,* , Sandeep Arorab
a
Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
b
Department of Pharmacy, Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India

A R T I C L E I N F O A B S T R A C T

Article history: Objective: The role of histone deacetylases has come out as an emerging remedy in control and treatment
Received 13 April 2019 of various metabolic disorders and cancers. This review highlights their intricate role in diabetes mellitus
Accepted 21 August 2019 as well as its associated complications.
Available online xxx
Key findings: Through recent studies and reports the role of various epigenetic markers in treatment of
diabetes mellitus has been revealed. HDAC enzyme regulates the structure of chromatin and transcripts
Keywords: genes in the nucleus synthesizing various proteins that control metabolic activities in the body. It mainly
Histone deacetylases
acts by removing an acetyl group from its precursor protein thereby modulating gene expression and
Insulin resistance
Acetylation
regulates the metabolic enzyme acetylation in mitochondria and cytosol.
Transcription Summary: The present review focus on the intrinsic role of HDAC inhibitors as an emerging remedy for
β-cell destruction diabetes and its complications demonstrating their use in preventing resistance of β-cells towards
Gene expression insulin, destruction of β-cells and provide protection against cytokine mediated attack on pancreatic
cells.
© 2019 Elsevier Masson SAS. All rights reserved.

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Diabetes mellitus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Role of HDACs in etiology of diabetes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
HDAC inhibitors and insulin signaling . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
HDAC inhibitors and activity of β-CELLS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
HDACs as treatment approach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
Declaration of Competing Interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 00

Introduction making it an essential hallmark of the disease [1]. The WHO last
reported the prevalence of diabetes in October 2018 [2]. The
Diabetes mellitus allocates a group of chronic diseases where number of people affected with the disease has reached to 422
aberrancies in substrate metabolism occur in response to relative million from 108 million in 1980 and the numbers are supposed to
or absolute lack of insulin with pronounced long term complica- double every ten years. The prevalence of disease has also reached
tions. The disease may be due to inherited or acquired deficiency of upto 8.5% from 4.7% among the individuals above 18 years in 1980.
insulin synthesis or its inability to bind to receptor and produce The low and middle income countries are more exposed to
effective results. Various body systems particularly the nerves and diabetes and it has become one of the major causes of blindness,
blood are highly damaged due to increased levels of glucose stroke, lower limb amputations, kidney failure and heart attacks.
Diabetes has also been stamped to be the 7th leading cause of
death in 2016 by WHO [3,4]. The long recognized causes of type I
diabetes include destruction of β-cell and autoimmune islet
* Corresponding author.
E-mail addresses: tapanbehl31@gmail.com, tapan.behl@chitkara.edu.in (T. Behl) inflammation though it is still debated whether molecular
. mechanisms involve predominantly inflammatory cytokine

http://dx.doi.org/10.1016/j.retram.2019.08.001
2452-3186/© 2019 Elsevier Masson SAS. All rights reserved.

Please cite this article in press as: R. Makkar, et al., Role of HDAC inhibitors in diabetes mellitus, Curr Res Transl Med (2019), https://doi.org/
10.1016/j.retram.2019.08.001
G Model
RETRAM 3233 No. of Pages 6

2 R. Makkar et al. / Current Research in Translational Medicine xxx (2019) xxx–xxx

mediated β-cell destruction or classical cytotoxic T-cell mediated removing acetyl group from histone lysine residues and restores
killing or both. A number of mechanisms explaining destruction of the persisting positive charge on terminal tails of Nitrogen
β-cells have been proposed like glucolipotoxicity, islet amyloid therefore increasing their likeliness for DNA. The tails displace
polypeptide deposition mediated disruption of membrane and transcription factors and forms a repressive structure of chromatin
inflammation of islets. Later, a unified hypothesis was proposed hence suppress transcription [13]. The mammalian HDACs can be
that explained the cause of β-cell destruction by induction of divided into two families and four classes. The first family includes
inflammatory mediators in pancreatic islets that further activate category of HDACs that are classically Zinc ion dependent while
pathways in β-cells resembling those in type 1 diabetes (T1D) [5,6]. the other family is nicotinamide adenine dinucleotide (NAD)
The inventory processes for developing novel treatment dependent HDACs.
approaches has undergone a revolutionary turn which has aided The Zn dependent family of HDACs includes:
study and targeting of newer protein molecules with exceptional Class I HDACs: It comprises of HDAC isoforms 1, 2, 3 and 8.
outcomes. A drug developer is no less than an artist as targeting These HDACs are localized and found exclusively in the nucleus
specific proteins with respect to disease pathology to achieve and are related to Rpd3 yeast enzyme with homology shared in
higher degree of effective treatment with minimal possible side catalytic pocket.
effects is not a job of left hand. The recent trend of drug Class II HDACs: It is advanced into two categories which is Class
development has turned to epigenetic modifications in gene IIa (HDAC 4, 5, 7 and 9) and Class IIb (HDAC 6 and 10). It covers two
expressions of the diseased patient which aims at targeting homology regions, in Nitrogen terminal regulatory area and
proteins involved in pathogenesis of the disease. The manner in Carbon terminal area of catalysis. The HDACs circulate between
which DNA is packaged profoundly influences the transcription of nucleus and cytosol with certain recruitment of protein complexes
genes in eukaryotic cells. The stable and potentially reversible and deacetylation sites occur.
alteration in gene expressions of an individual without any Class IV HDACs: They comprise only one member i.e. HDAC 11. It
permanent change in DNA sequence with inheritance property is found in both nucleus as well as cytoplasm.
is called epigenetic modifications [7]. Waddington originally The NAD+ dependent family of HDACs include:
defined epigenetics as a firm reason for programmed changes Class III HDACs: They are also called the silent information
during development of embryo due to interactions between genes regulator 2. The HDAC enzymes are dependent on NAD+ under
and their products bringing out a phenotypic change. The bridge presence of which acetyl groups are removed from residues of
between environment and genetics is epigenome while determin- acetylated lysine. This class includes homology of yeast Sir 2, which
ing the final phenotype on modification of gene expressions in presence of cofactor NAD acts as a vital protein for silencing
without any alteration in DNA sequence is epigenetic code. By certain genes and catalyze their action. They are also called
targeting the specific gene directly, the phenotype of the disease Sirtuins. Till now seven members have been noted in the family of
can be changed through epigenetic modifications in response to protein sirtuin localized in mitochondria, nucleus and cytoplasm of
pathological and environmental conditions like diet, exercise, the cell [14].
toxins, inflammation, oxidative stress and metabolic changes. The The deacetylation action of HDAC 6 is higher on 5th and 8th
epigenetic modifications show a phenomenal role in genomic lysine residue of histone 4 while HDAC 1 and 3 have been tested for
imprinting, stable inheritance of gene expression, embryonic their efficacy to deacetylate the lysine residues containing four
development, cell differentiation and identity, inactivation of X core histones on varied levels [15,16] (Table 1).
chromosome, immune cell functioning, and responses to environ- HDAC inhibitors through hyperacetylation of non histone and
mental signals at cellular levels [8]. One such emerging epigenetic nucleosomal histone proteins regulate expression of genes. Most of
modification that is being investigated recently is Histone the HDACs are large multiprotein complex regulated and do not
modification. It has managed to obtain exceptional results in activate enzymatically without interacting with other proteins.
treatment of severe life threatening diseases including cancer and NuRD/Mi2, coREST, Sin3, and N-CoR and SMRT, the nuclear
metabolic disorders [9]. Gene transcription processes in cells of receptor corepressors are some of the multiprotein complexes
eukaryotes depend highly upon the chromatin confirmation. required in the activation of HDAC proteins. The SMRT and N-coR
Nucleosome is the basic unit of chromatin constituting four core nuclear receptor complexes enzymatically activate HDAC3 where-
dimers of histones namely H2A, H2B, H3 and H4 which are doubly as the other complexes i.e. the Sin 3, CoREST and NuRD/Mi2
folded around the DNA in the form of an octamer. The covalent complexes stimulate the activity of HDAC1and HDAC2 deacetylase.
modifications undergo in the basic N-terminal tail of histone Class IIa HDACs recruit HDAC3 comprising N-CoR/SMRT active
proteins which recruit factors responsible for transcription via complexes and are inactive HDACs. HDACs and its inhibitors
changes in structure causing packaging of chromatin and regulate predominantly control cell proliferation, differentiation, apoptosis
gene expression [10]. The remodeling of chromatin through and also modulate cell cycle by regulating cellular reactions [17,18].
acetylation, phosphorylation, methylation and various other HDACs have multiple actions as they target numerous cellular
posttranslational modifications in histones is the fundamental pathways and produce resultant responses.
phenomenon in eukaryotic cell biology highlighting diverse HDACs bind to nuclear receptors and regulate signaling of
approaches for treatment in response to physiological and hormones by modulating their release. This indicates its use in
pathological conditions. The histone acetyl transferase (HAT) management of diabetes mellitus as HADCs stimulate release of
and histone deacetylase (HDAC) as their name suggests acetylate insulin.
and deacetylate histone proteins respectively [11,12]. The HAT The α-tubulin proteins are expressed in the cytosol of the cells.
enzyme undergoes posttranslational histone acetylation by HDAC 6 is highly specific and binds to α-tubulin and actin proteins
transferring an acetyl group from acetyl coenzyme A to the which control the stability and functioning of microtubules. The
e-NH3 groups of internal residues of histone proteins. Acetylation acetylation state of actin is highly modulated by HDAC6 thereby
forms decondensed chromatin called ‘euchromatin’ by neutraliz- blocking its fibrillar action and affects actin dependent motility.
ing the positive charge on tail of N-terminal residues of lysine. This Angiogenesis is also prevented by use of HDAC inhibitors as
decreases the propensity of positive terminal residues towards angiogenic markers like VEGF and HIF-1α are targeted by HDACs
DNA activation, increasing access of DNA towards transcription The role of HDACs in modulating inflammation by suppressing
factors and RNA polymerase by unfolding nucleosomes and causes outburst of inflammatory mediators has already been discussed in
gene activation. In contrast to HAT, the enzyme HDAC acts by the article.

Please cite this article in press as: R. Makkar, et al., Role of HDAC inhibitors in diabetes mellitus, Curr Res Transl Med (2019), https://doi.org/
10.1016/j.retram.2019.08.001
G Model
RETRAM 3233 No. of Pages 6

R. Makkar et al. / Current Research in Translational Medicine xxx (2019) xxx–xxx 3

Table 1
An overview of HDACs.

CLASS NAME LOCATION Co - FACTOR EXPRESSING TISSUES USES


CLASS I HDAC 1 Nucleus Zn2+ DEPENDENT Widespread Inflammation, Insulin resistance
HDAC 2
HDAC 3 Nucleus/Cytoplasm
HDAC 8 Nucleus
CLASS II (a) HDAC 4 Nucleus/Cytoplasm Heart, Brain Hyperglycemia, Insulin resistance,
HDAC 5 Smooth muscle Decreased insulin production
HDAC 7 Placenta, Pancreas, Heart
HDAC 9 Smooth muscle, Brain
CLASS II (b) HDAC 6 Kidney, Liver, Heart Glucose intolerance, Hyperglycemia
HDAC 10 Spleen, Kidney, Liver
+
CLASS III SIRT 1 Nucleus, Cytoplasm NAD DEPENDENT White adipose tissue Enhanced insulin signaling
SIRT 2 Cytoplasm Testes, Heart, Brain Impaired insulin signaling
SIRT 3 Mitochondria Brown adipose tissue Enhanced insulin signaling
SIRT 4 Islets of pancreas Impaired insulin signaling
SIRT 5 Liver Enhanced insulin signaling
SIRT 6 Nucleus Impaired insulin signaling
SIRT 7 Enhanced insulin signaling
CLASS IV HDAC 11 Nucleus Zn2+ DEPENDENT Heart, Smooth muscle, Brain, Inflammation
Kidney

HDACs suppress tumor growth by binding to p53, p21 and converge on common extracellular inflammatory stressors despite
cyclin proteins and alter cell cycle of the body. their dissimilar genetic background and these stressors induce
intracellular signaling in islet cells. The most commonly reported
Diabetes mellitus intracellular stressors is IL-1β which is secreted from activated
mononuclear cells that prohibit functioning of β-cell and cause cell
The main source of energy for most of the cellular functions is death on prolonged exposure due to their selective β-cell toxicity
glucose. An increased level of circulating glucose in body post meal [22,23].
stimulates pancreatic β cells to release insulin. Insulin plays an The molecular performance of HDAC was assumed to be limited
important role in glucose homeostasis. It undergoes autophos- to deacetylation as indicated by their name but recent advances in
phorylation on binding with transmembrane receptors and phylogenetic analysis suggested their role in regulating perfor-
phosphorylates the intercellular protein substrates including mance in a broader range of non histone proteins. In terms of
insulin receptor substrate – 1 and 2 (IRS-1 and IRS- 2). The posttranslational mechanisms the impact of acetylation is no less
phosphorylated IRS subsequently activates phosphoinositide 3- than phosphorylation increasing demand of HDACi as a treatment
kinase (PI3K/AKT) pathway. These phosphorylated products approach for an expanding range of diseases. The original goal of
stimulate activation of several enzymes and proteins and translo- HDAC inhibition was transcriptional control over oncogene net-
cate glucose transporter 4 (GLUT 4) to cell membrane from works in cancer cells but their indications as novel treatment in
intracellular cell vesicles. An array of metabolic events is other diseases including neurodegenerative and inflammatory
demonstrated by another important pathway in insulin signaling diseases is growing exceptionally. The master transcription factor
called mitogen activated protein kinase (MAPK) pathway [19]. Over nuclear factor (NF-kB) in inflammation is now recognized to be
the last century drastic behavioural and lifestyle changes have regulated by acetylation. In the current review the prospects of
produced conditions of insulin resistance where amidst the inhibition of HDAC as a novel approach for treatment of diabetes
presence of abundant amount of insulin, it is no longer able to will be reviewed. It also incorporates an outline of genetic
bind to its receptor causing prohibition of any further series of associations relating to etiology of diabetes and HDACs and
events [20]. evaluating its potential as an oral therapy in treatment of disease
Diabetes can be categorized into two principal forms: and its associated complications depending upon the pathological
Type I diabetes: formerly known as insulin dependent diabetes, events [24,25].
is a condition where pancreas fails to synthesize insulin hence
disturbing homeostasis of glucose in body. It is frequently observed Role of HDACs in etiology of diabetes
in children and adolescents and is termed juvenile diabetes.
Type II diabetes: also known as non-insulin dependent A combination of environmental and genetics factors contrib-
diabetes, is a condition in which the body fails to respond to ute in the etiology of diabetes making it complex. Though T1D and
pre-existing insulin. About 90% of diabetic cases worldwide are T2D are considered to be genetically distinct, an association
accounted to be T2D and predominantly targets adults [21]. between the 6q21 chromosomal region has been identified by
Mutations in one or more genes by genetic-environmental genome wide association scans (GWAS) studies where HDAC 2 is
interactions trigger onset of diabetes making it a complex disease. also located signifying the latter’s role in both the diabetes. The
Both the types of diabetes are genetically distinct where type I epigenetic and environmental factors significantly contribute in
diabetes is believed to happen due to variations in immune the etiology of the disease making it a polygenetic disorder.
regulatory genes highlighting its autoimmune nature and exposing Simmons and Pinney reported that an unfavorable fetal milieu due
individuals to T-cells driven β-cell destruction and chronic to exposure of fetus to intra uterine growth retardation (IUGR)
inflammation whereas in contrast genome wide association scans affects the development of β-cells due to modification in muscular
(GWAS) suggested type II diabetes as a disease where alterations in transport of glucose by glucose transporter 4 (GLUT 4) and varying
genes evoke changes on β-cell function and impairs its compensa- key regulatory genes like pancreatic and duodenal homeobox
tion to increased demands of insulin [1]. The immune and factor 1 (Pdx1) thereby advancing to T2D. Loss of acetylation of
metabolic pathogenesis of both types of diabetes seems to histone recruits HDAC1 which complexes Pdx1 a proximal

Please cite this article in press as: R. Makkar, et al., Role of HDAC inhibitors in diabetes mellitus, Curr Res Transl Med (2019), https://doi.org/
10.1016/j.retram.2019.08.001
G Model
RETRAM 3233 No. of Pages 6

4 R. Makkar et al. / Current Research in Translational Medicine xxx (2019) xxx–xxx

promoter to the Sin3A corepressor hence reducing the expression regulators that control translocation of GLUT 4 by binding to its
of Pdx1after IUGR. The HDAC1/Sin3 complex formed propagates translational elements [31]. The obstruction in release of insulin
epigenetic cycle and recruits histone demethylase which inhibits may occur at several stages. There are two mechanisms that
the transcription of Pdx1 thereby causing loss of histone 3 lysine 4 control the signaling of insulin in β-cells through HDACs, firstly
trimethylation (H3K4me3). Inhibition of HDACs reverses this effect through epigenetic control while secondly through IRS2 expres-
by methylation and dimethylation of H3K9 which lodges the sion and activity regulation via protein acetylation [32]. The role of
promoter Pdx1 in its transcriptionally inactive state [26]. The IRS2 has been reported to be very crucial in proliferation of β-cells
transcription of Glut4 is inhibited as a result of lack of histone of pancreas hence identifying and employing a precise isoform of
acetylation in adult muscle tissue of Glut 4 promoter as mediated HDAC can be a valuable approach as therapy forT2D [33]. The
by HDAC1 and HDAC4 thereby repressing the peripheral uptake of HDAC1 located in nucleus acts via acetylation of histone in the
glucose and producing insulin resistance thus contributing to T2D. promoter region of IRS2 while the cytoplasm localized HDAC
The remodeling of chromatin in incretin hormones such as glucose acetylates the lysine residues in IRS2 and leads to modification of
dependent insulinotropic peptide 1 and glucagon-like peptide 1 proteins [34]. The expression of different isoforms of HDACs
has already been induced as a therapeutic approach in treatment of differed in the brain with altered activity in kidneys of diabetic
T2D which has led to greater association with the transcription patients. The differentiation of β-cells of pancreas was reported to
factors due to in vitro global acetylation of H3 histones [27,28]. be controlled by HDAC 4, 5 and 9. An increased expression of
HDACs and histone acetylation are also useful in treatment of rare HDAC1 was encountered from pancreatic cells isolated from
kinds of autosomal diabetes namely “maturity onset diabetes of children with T1D with decrease in HDAC2 and HDAC3 expres-
the young” (MODY) and its seven dissimilar subtypes based on the sions. The secretion of insulin is suppressed by histone acetylation
mutations in genes apart from their clinical relevance in treatment and promoted expression of Tcf7l2 with increased articulation of
of T1D and T2D. The genes encode nearly all transcription factors HDAC7 in pancreatic cells of T2D patients. The acetylation of lysine
which are involved in the production of insulin and development in SREBP 1a and p53 inhibits ubiquitination while acetylation of
of pancreas except for insulin and glucokinase. Some of these lysine in PGC1α and FOXO promotes autophosphorylation. The
include hepatocyte nuclear factor (HNF)- 1α, -1β and -4α which phosphorylation of tyrosine in neuronal cells seems to diminish on
produces hepatic gluconeogenesis and transcription of insulin, IRS2 post translational modification due to acetylation of lysine in
neurogenic differentiation 1 (NeuroD1) and Pdx1. Histone acetyl IRS2 with no reports β cells of pancreas [35,36]. The patients
transferases (HATs) and HDACs by remodeling chromatin interact receiving HDAC inhibitors as therapy for treatment of cancer have
with the transcription factors and regulate normal functioning been reported to fabricate hyperglycemia as its consequence
suggesting their important role in pathogenesis of diabetes [29]. indicating its role in signaling of insulin in some organs with
impairment of tolerance to glucose. Hence through these findings
HDAC inhibitors and insulin signaling the need to pick more selective HDAC inhibitors can be
suggested necessary to provide protective effect for insulin
The signaling of insulin is vital in maintaining the glucose signaling [37,1] (Fig. 2).
homeostasis in body as a decline in its signaling inhibits hepatic
steatosis in liver and containment of gluconeogenesis with HDAC inhibitors and activity of β-CELLS
reduced uptake of glucose in muscles. Insulin signaling also
regulates the mass of β-cells which is directly involved in onset and The factors mediating transcription play crucial role in
progression of diabetes [30]. The Fig. 1 below describes the development of pancreas by differentiation of endocrine and
pathway for insulin signaling. On binding to insulin receptor, the exocrine. The etiology of type-1 diabetes is highlighted by the
autophosphorylation and phosphorylation of insulin receptor understanding of mechanism of apoptosis of β-cells induced by
substrate (IRS) family members occurs causing release and action inflammatory cytokines. The molecules with capability to suppress
of insulin. The IRS proteins post phosphorylation bind and activate this apoptotic processes can be efficiently employed as therapy for
phosphatidylinositol 3-kinase which further stimulate protein preventing β-cells from hypersensitive reactions and immune
kinase Akt phosphorylation ultimately translocating glucose responses. Various studies have reported the protective actions of
transporter (GLUT 4) to the plasma membrane from intracellular HDAC inhibitors in preventing destruction of β-cells [38]. The
vesicles and mediating uptake of glucose. The myocyte enhancer determination of precise isoform of HDAC which is responsible for
factor (MEF)-2 and GLUT 4 enhancer factor (GEF) are chief survival of β-cells is fundamental to its therapeutic application as

Fig. 1. signaling pathways associated with HDACs.

Please cite this article in press as: R. Makkar, et al., Role of HDAC inhibitors in diabetes mellitus, Curr Res Transl Med (2019), https://doi.org/
10.1016/j.retram.2019.08.001
G Model
RETRAM 3233 No. of Pages 6

R. Makkar et al. / Current Research in Translational Medicine xxx (2019) xxx–xxx 5

proteins accumulate on HDACi treatment modifying their func-


tions [41,42]. The targets in pancreas that can are involved in
etiology of DM are also under investigation for contributing better
in expression and regulation of Ngn3. Hence through various
studies the role of HDACs in proliferation of β-cells has been stated
evident as they have direct effects on fate regulation of endocrine
cells [1] (Fig. 3).

HDACs as treatment approach

Various HDACi have recently been approved by FDA e.g.,


Panobinostat is under phase IV clinical trials and has also been
approved by FDA in 2015 for its use in management of multiple
myeloma and cutaneous T-cell lymphoma (CTCL)
Some of the HDACi under clinical trials include:
Valproic acid is under phase III clinical trials for its use in
cervical and ovarian cancer. Tacedinaline, another HDACi is under
Fig. 2. Mechanism of Insulin Signaling. phase III trials for its use in management of multiple myeloma and
lung cancer. Mocetinostat is under phase II clinical trials for its use
broad spectrum HDAC inhibitors may have toxic effects and fail to in management of Follicular and Hodgkin lymphoma and acute
restore significant β-cell functions like secretion of insulin as myeloid leukemia. Rocilinostat has been recently developed and is
stimulated by levels of glucose [39]. Genetic and chemical under phase I trials for its use in treatment of multiple myelomas.
inhibition of HDAC3 has been found to suppress the apoptosis Many other HDAC inhibitors drugs like Mocetinostat, Abexinostat,
of β-cell line of rats. The selective HDAC 1, 2 and 3 inhibitors Practinostat are under phase II clinical trials and are being studied
embarked better protective effects in β-cell line as compared to for their beneficial effects in treatment of Follicular and Hodgkin
broad spectrum HDAC inhibitors like SAHA indicating additional lymphoma and acute myeloid leukemia, Sarcoma and lymphoma
side toxicities induced by inhibition of various isoforms of HDAC. and Recurrent or metastatic prostate cancer respectively.
MS-275 a selective HDAC inhibitor he genetic knock down of Some of the HDACs used in management of diabetes mellitus
HDAC3 stimulated secretion of insulin in response to glucose levels include:
and restores it [40]. The development of endocrine progenitors of Vorinostat (SAHA) was the first HDAC inhibitor to be approved
NGN3+ was enhanced on treatment with HDACi. The epithelial cells by FDA in 2006 for its use in treatment of cutaneous T-cell
in pancreas contain precursors for PDX1+ which augments lymphoma. Apart from its use in myeloma, SAHA is also being
development of various cell types of pancreas besides NGN3+ examined for its therapeutic role in management of diabetes
cells. On enhancing proliferation of precursors of PDX1+ by FGF 10 mellitus as its is reported to increase expression of insulin even in
or mesenchyme the progenitors of endocrine NGN3+ are intensi- low concentrations of glucose. Sodium butyrate (NaB) and
fied. On initiating treatment with HDACi no increase in the Trichostatin A (TSA) are other HDACi that are simultaneously
production of PDX1+ precursors was noticed indicating role of being examined for their use in diabetes mellitus. TSA is believed to
HDACi and FGF 10 in enhancing the endocrine progenitors NGN3+ increase insulin release by stimulating expression of β-cells and
in alternate ways except for poorly growing cells of NGN3+ making stimulating GLP-1 proteins. TSA also improved the transdiffer-
it independent of proliferation of cells. Also the relationship entiation of bone marrow stem cells into insulin-producing cells
between the β-cell signaling of transforming growth factor in Sodium butyrate (NaB) stimulates early events of pancreatic
differentiation of pancreatic cells is being investigated. A specification in embryonic stem (ES) cells by promoting their
possibility of HDACi to inhibit the promoter NGN3+ cells directly differentiation into insulin-producing cells.
as mechanism of action is being explored for its therapeutic Valproate has been used commonly in treatment of bipolar
efficiency. The presence of non histone substrates of proteins in disorders and other CNS disorders. A typical side effect associated
HDACi regulates expression of genes and also controls death and with Valproate was hyperinsulinemia. It was found that the
proliferation of cells. The acetylated forms of these substrate function is attributed to inhibition of HDACs by Valproate. Hence,

Fig. 3. HDAC inhibitors and β-cell destruction.

Please cite this article in press as: R. Makkar, et al., Role of HDAC inhibitors in diabetes mellitus, Curr Res Transl Med (2019), https://doi.org/
10.1016/j.retram.2019.08.001
G Model
RETRAM 3233 No. of Pages 6

6 R. Makkar et al. / Current Research in Translational Medicine xxx (2019) xxx–xxx

valproate is also under study for evaluation of its effect in [18] Grunstein M. Histone acetylation in chromatin structure and transcription.
management of diabetes mellitus. Nature 1997;389:349–52.
[19] Sharma S, Taliyan R. Histone deacetylase inhibitors: future therapeutics for
Various other HDACi like Romidepsin, Panabiostat have been insulin resistance and type 2 diabetes. Pharmacol Res 2016;113:320–6.
approved by FDA for management of lymphomas and their role in [20] Ye J. Improving insulin sensitivity with HDAC inhibitor. Diabetes
diabetes is yet under investigation. 2013;62:685–7.
[21] Baynes HW. Classification, pathophysiology, diagnosis and management of
diabetes mellitus. J Diabetes Metab 2015;6(5):1–9.
Conclusion [22] DMICC. Genetic basis of type 1 and type2 diabetes, obesity, and their
complications. Advances and emerging opportunities in diabetes research: a
Strategic Planning report of the DMICC. 2014.
HDACs are indulged in multiple biological system pathways and [23] Pablo A. Recent advances in understanding/managing type 2 diabetes mellitus.
contribute majorly in pathogenesis and development of diabetes F1000Research 2017;6.
mellitus even through genetic basis. HDACs promote development, [24] Sharma S, Taliyan R. Histone deacetylase inhibitors: future therapeutics for
insulin resistance and type 2 diabetes. Pharmacol Res 2016;113:320–6.
proliferation and differentiation of β-cells and regulate their
[25] Gray SG, De Meyts P. Role of histone and transcription factor acetylation in
functioning by preventing inflammatory cell damage, improve diabetes pathogenesis. Diabetes Metab Res Rev 2005;21:416–33.
resistance of insulin and prevent microvascular complication of [26] Pirola L, Balcerczyk A, Okabe J, El-Osta A. Epigenetic phenomena linked to
diabetes at later stages. Altogether it can be summoned that HDACi diabetic complications. Nat Rev Endocrinol 2010;6(12):665.
[27] Sathishkumar C, Prabu P, Balakumar M, Lenin R, Prabhu D, Anjana RM, et al.
can be initiated as promising therapy for preventing and treating Augmentation of histone deacetylase 3 (HDAC3) epigenetic signature at the
diabetes and its clinical utility is under examination as there is still interface of proinflammation and insulin resistance in patients with type 2
a lot to be learned about its mechanism. diabetes. Clin Epigenetics 2016;8(1):125.
[28] Patel MM, Patel BM. Repurposing of sodium valproate in colon cancer
associated with diabetes mellitus: role of HDAC inhibition. Eur J Pharm Sci
Declaration of Competing Interest 2018;121:188–99.
[29] Hull EE, Montgomery MR, Leyva KJ. HDAC inhibitors as epigenetic regulators of
the immune system: impacts on cancer therapy and inflammatory diseases.
The author(s) declare no conflict of interest. Biomed Res Int 2016.
[30] Rafehi H, Kaspi A, Ziemann M, Okabe J, Karagiannis TC, El-Osta A. Systems
References approach to the pharmacological actions of HDAC inhibitors reveals EP300
activities and convergent mechanisms of regulation in diabetes. Epigenetics
2017;12(11):991–1003.
[1] Christensen DP, Dahllöf M, Lundh M, Rasmussen DN, Nielsen MD, Billestrup N,
[31] Anuradha R, Saraswati M, Kumar KG, Rani SH. Apoptosis of beta cells in
et al. T. HDAC inhibition as a novel treatment for diabetes mellitus. 2011.
diabetes mellitus. DNA Cell Biol 2014;33(11):743–8.
[2] Kaiser AB, Zhang N, Van der Pluijm W. Global prevalence of type 2 diabetes
[32] Remsberg JR, Ediger BN, Ho WY, Damle M, Li Z, Teng C, et al. Deletion of histone
over the next ten years (2018–2028). 2019.
deacetylase 3 in adult beta cells improves glucose tolerance via increased
[3] Chen L, Magliano DJ, Zimmet PZ. The worldwide epidemiology of type 2
insulin secretion. Mol Metab 2017;6(1):30–7.
diabetes mellitus: present and future perspectives. Nat Rev Endocrinol 2014.
[33] Khan S, Jena G. Valproic acid improves glucose homeostasis by increasing
[4] Frier BM. Hypoglycaemia in diabetes mellitusA and clinical implications. Nat
beta-cell proliferation, function, and reducing its apoptosis through HDAC
Rev Endocrinol 2014;10(12):711.
inhibition in juvenile diabetic rat. J Biochem Mol Toxicol 2016;30(9):438–46.
[5] Adeghate E, Schattner P, Dunn E. An update on the etiology and epidemiology
[34] McGee-Lawrence ME, Pierce JL, Yu K, Culpepper NR, Bradley EW, Westendorf
of diabetes mellitus. Ann N Y Acad Sci 2006;1084(1):1–29.
JJ. Loss of Hdac3 in osteoprogenitors increases bone expression of
[6] Alonso-Magdalena P, Quesada I, Nadal A. Endocrine disruptors in the etiology
osteoprotegerin, improving systemic insulin sensitivity. J Cell Physiol
of type 2 diabetes mellitus. Nat Rev Endocrinol 2011;7(6):346.
2018;233(4):2671–80.
[7] Portela A, Esteller M. Epigenetic modifications and human disease. Nat
[35] Lundh M, Galbo T, Poulsen SS, Mandrup-Poulsen T. Histone deacetylase 3
Biotechnol 2010;28(10):1057.
inhibition improves glycaemia and insulin secretion in obese diabetic rats.
[8] Kanwal R, Gupta S. Epigenetic modifications in cancer. Clin Genet 2012;81
Diabetes Obes Metab 2015;17(7):703–7.
(4):303–11.
[36] Kawada Y, Asahara SI, Sugiura Y, Sato A, Furubayashi A, Kawamura M, et al.
[9] Kelly TK, De Carvalho DD, Jones PA. Epigenetic modifications as therapeutic
Histone deacetylase regulates insulin signaling via two pathways in pancreatic
targets. Nat Biotechnol 2010;28(10):1069.
β cells. PLoS One 2017;12(9)e0184435.
[10] Abel T, Zukin RS. Epigenetic targets of HDAC inhibition in neurodegenerative
[37] Sun C, Zhou J. Trichostatin A improves insulin stimulated glucose utilization
and psychiatric disorders. Curr Opin Pharmacol 2008;8(1):57–64.
and insulin signaling transduction through the repression of HDAC2. Biochem
[11] Shen Y, Wei W, Zhou DX. Histone acetylation enzymes coordinate metabolism
Pharmacol 2008;76(1):120–7.
and gene expression. Trends Plant Sci 2015;20(10):614–21.
[38] Sabir S, Saleem A, Akhtar MF, Saleem M, Raza M. Increasing beta cell mass to
[12] Allis CD, Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev
treat diabetes mellitus. Adv Clin Exp Med 2018.
Genet 2016;17:487–500.
[39] Larsen L, Tonnesen M, Ronn SG, Størling J, Jørgensen S, Mascagni P, et al.
[13] Schütz LF, Park MH, Choudhury M. HDACs in diabetes: a new era of epigenetic
Inhibition of histone deacetylases prevents cytokine-induced toxicity in beta
drug. Nutritional and therapeutic interventions for diabetes and metabolic
cells. Diabetologia 2007;50(4):779–89.
syndrome. second edition . p. 475–86.
[40] Vestergaard AL, Bang-Berthelsen CH, Fløyel T, Stahl JL, Christen L, Sotudeh FT,
[14] Yang XJ, Seto E. HATs and HDACs: from structure, function and regulation to
et al. MicroRNAs and histone deacetylase inhibition-mediated protection
novel strategies for therapy and prevention. Oncogene 2007;26:5310–8.
against inflammatory β-cell damage. PLoS One 2018;13(9)e0203713.
[15] Sengupta N, Seto E. Regulation of histone deacetylase activities. J Cell Biochem
[41] Khan S, Jena GB. Protective role of sodium butyrate, a HDAC inhibitor on beta-
2004;93:57–67.
cell proliferation, function and glucose homeostasis through modulation of
[16] Kelly RD, Cowley SM. The physiological roles of histone deacetylase (HDAC) 1
p38/ERK MAPK and apoptotic pathways: study in juvenile diabetic rat. Chem
and 2: complex co-stars with multiple leading parts. Biochem Soc Trans
Biol Interact 2014;213:1–12.
2013;41:741–9.
[42] Chou DHC, Holson EB, Wagner FF, Tang AJ, Maglathlin RL, Lewis TA, et al.
[17] Mathias RA, Guise AJ, Cristea IM. Post-translational modifications regulate
Inhibition of histone deacetylase 3 protects beta cells from cytokine-induced
class IIa histone deacetylase (HDAC) function in health and disease. Mol Cell
apoptosis. Chem Biol 2012;19(6):669–73.
Proteomics 2015;14:456–70.

Please cite this article in press as: R. Makkar, et al., Role of HDAC inhibitors in diabetes mellitus, Curr Res Transl Med (2019), https://doi.org/
10.1016/j.retram.2019.08.001

You might also like