3-Novel Histone Post-Translational Modifications in Diabetes and Complications of Diabetes The Underlying Mechanisms and Implications

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Biomedicine & Pharmacotherapy 156 (2022) 113984

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

Novel histone post-translational modifications in diabetes and


complications of diabetes: The underlying mechanisms and implications
Dongze Li a, b, c, 1, Li Zhang d, 1, Yanqiu He a, b, c, 1, Tingting Zhou a, b, c, 1, Xi Cheng a, b, c, 1,
Wei Huang 1, a, b, c, *, Yong Xu 1, a, b, c, *
a
Department of Endocrinology and Metabolism, Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical
University, Luzhou 646000, Sichuan, China
b
Sichuan Clinical Research Center for Nephropathy, Luzhou 646000, Sichuan, China
c
Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
d
Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China

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

Keywords: Diabetes is a group of global epidemic diseases with high prevalence and morbidity. The pathophysiological
Diabetes changes of diabetes can be affected by genes, environmental factors, and several socioeconomic factors. Histone
Epigenetics post-translational modification is the central epigenetic mechanism regulated by genetic and environmental
Acylation
factors. Histone methylation and acetylation have been thoroughly studied in various pathological processes of
Histone modification
Metabolic memory
diabetes, such as the metabolic memory phenomenon, inflammation, and endothelial dysfunction. In recent
years, an increasing number of novel histone acylation modifications have been identified by mass spectrometry.
The level of any novel histone modification depends on the relative concentration of its respective acyl-CoA,
which can be generated through various intermediate metabolic pathways. Although studies have shown that
these novel modifications and metabolic regulation of histone acylation have a close connection with diabetes,
investigations on this connection have lagged. Moreover, the locations of acylation modification sites and how
enzymatic reactions regulate diabetes and its complications remain largely unknown. This review summarizes
the relationship between novel histone modifications and diabetes, mainly focusing on β-hydroxybutyrylation,
propionylation, butyrylation, malonylation, and succinylation. We aim to highlight the typical characteristics of
novel acylation modifications and provide a new perspective for further research on the pathogenesis and
treatment of diabetes and its complications.

1. Introduction derived from Diabetes Control and Complications Trial (DCCT) and
Epidemiology of Diabetes Interventions and Complications Study (EDIC)
It is estimated that 537 million adults worldwide are living with [1] demonstrates a “metabolic memory” phenomenon: early exposure to
diabetes according to the latest International Diabetes Federation (IDF) hyperglycemia results in persistently induced damage even after the
report. Chronic complications of diabetes caused by long-term chronic hyperglycemic state is under control, which indicates that epigenetics is
hyperglycemia, often affecting the cardiovascular system, nervous sys­ a critical factor as a bridge between genes and the environment.
tem, and urinary system, are the main source of the health and economic The basic goal of epigenetics is to identify heritable changes in gene
burdens of patients. The pathogenesis of diabetes is complex, involving expression or cellular phenotype with no accompanying alterations in
genetic susceptibility and environmental factors. Emerging evidence the DNA sequence. Epigenetic mechanisms principally include histone

Abbreviations: HPTMs, Histones Post-translational Modifications; CoA, coenzyme A; Kac, lysine acetylation; Kme, lysine methylation; Kpr, lysine propionylation;
Kbu, lysine butyrylation; Ksucc, lysine succinylation; Kma, lysine malonylation; Kbhb, lysine β-hydroxybutyrylation; Kbz, lysine benzoylation; HDAC, histone
deacetylase; KD, ketogenic diet; BHB, β-hydroxybutyrate; DKD, diabetic kidney disease; SCFAs, short chain fatty acids; TNF-α, tumor necrosis factor alpha; MCP-1,
macrophage chemoattractant protein-1; IL, interleukin; VEGF, vascular endothelial growth factor; MMP-2, matrix metalloproteinase-2.
* Corresponding authors at: Department of Endocrinology and Metabolism, Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital
of Southwest Medical University, Luzhou 646000, Sichuan, China.
E-mail addresses: huangwei1212520@163.com (W. Huang), xywyll@swmu.edu.cn (Y. Xu).
1
These authors have contributed equally to this work and share first authorship.

https://doi.org/10.1016/j.biopha.2022.113984

Available online 7 November 2022


0753-3322/© 2022 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
D. Li et al. Biomedicine & Pharmacotherapy 156 (2022) 113984

post-transcriptional modifications (HPTMs), DNA methylation, and not decrease, and the expression of SOD2 continued to be abnormal
small non-coding regulatory RNAs [2]. Histones can bind tightly to DNA [19].
to form nucleosome structures, including core histones H2A, H2B, H3, These studies emphasize the key role of epigenetic histone modifi­
H4, and a linker histone H1 [3]. HPTM refers to a covalent process in cations in the pathogenesis of diabetes. Along with methylation and
which different acylation modifications are added to one or several acetylation, other histone modification processes are involved in the
amino acid residues on the tails of histones, which alters the loose or progression of diabetes and complications of diabetes. However, our
tight binding state between histones and DNA and effectively regulates knowledge of novel HPTMs in diabetes remains limited due to the lack of
the transcriptional activity of the target gene. The most prominent of thorough investigation of this relatively new field of study. A strong
HPTMs are acetylation (Kac) and methylation (Kme). In recent years, understanding of these variations in histone lysine acetylation and
benefiting from the development of anti-acylated antibodies and the use methylation and corresponding regulatory elements may aid in identi­
of ultrasensitive mass spectrometry (MS), types of novel histone lysine fying new histone modification markers and significant therapeutic
acylations have been identified and characterized, such as lysine pro­ targets for diabetes.
pionylation (Kpr), butyrylation (Kbu) [4], succinylation (Ksucc) [5],
crotonylation (Kcr) [6], malonylation (Kma) [7], 2-hydroxyisobutyla­ 3. Novel histone modifications in diabetes and complications of
tion (Khib) [8], glutarylation (Kglu) [9], β-hydroxybutyrylation diabetes
(Kbhb) [10], benzoylation (Kbz) [11], and, most recently, lactylation
(Kla) [12]. These external modifications constitute the basic elements of 3.1. β-hydroxybutyrylation
the histone code. Acyl-CoA is not only a specific donor of these histone
modifications but also a metabolic intermediate in many metabolic Due to a rising emphasis on the comprehensive treatment of diabetes,
pathways. In addition to some endogenous acyl-CoA (such as acetyl-CoA new dietary replacement strategies have attracted increasing attention
and succinyl-CoA) produced by glucose and lipid metabolism in healthy from researchers. The ketogenic diet (KD) is a high-fat, adequate-pro­
or diabetic states, acyl-CoA synthetase (ACS), also known as acyl-CoA tein, and low-carbohydrate diet. As a dietary treatment, KD can reduce
synthetase short chain member 2 (ACSS2), can promote the produc­ postprandial blood glucose levels, promote insulin sensitivity, reduce
tion of acyl-CoA (such as acetyl-CoA, propionyl-CoA, butyryl-CoA, and body weight, and even reverse diabetic nephropathy [20–22]. With KD,
β-hydroxybutyryl-CoA). the body relies more on burning fat for energy and increases the pro­
Many studies have shown that the development of diabetes and the duction of ketone bodies, namely β-hydroxybutyrate (BHB), acetoace­
occurrence of the “metabolic memory” phenomenon is closely related to tate, and acetone. KD has primarily been used to increase serum BHB
classic histone modifications [13], and recent studies also have hinted at levels, and it can also be enriched after prolonged exercise, fasting, and
a relationship between diabetes and some novel HPTMs, including diabetic ketoacidosis [23]. Physiologically elevated BHB levels have
β-hydroxybutyrylation, propionylation, butyrylation, malonylation, and been shown to play a potential protective role in diabetic patients. It has
succinylation. been reported that BHB may reduce oxidative stress in type 2 diabetic
In this review, we briefly outline the relationship between diabetes rats, thereby preventing diabetic cardiomyopathy and diabetic cardiac
and classic HPTMs and summarize the latest advances of the above- death [24]. The underlying epigenetic molecular mechanisms of BHB as
mentioned novel HPTMs in diabetes and complications of diabetes. a beneficial factor for diabetes were discovered recently.
Moreover, we detail the traits of these novel HPTMs, including their In a landmark 2016 study, Kbhb was first identified and character­
acylation donors, sites, and corresponding enzymes, and we briefly ized by Yingming Zhao’s team in a mouse model of streptozotocin (STZ)
analyze the less common HPTMs. We also provide a fresh perspective on induced ketoacidosis using high mass accuracy tandem mass spec­
the pathogenesis of diabetes and its complications and suggest possible trometry proteomics. Physiological and pathological changes in BHB
strategies for diabetes prevention and anti-diabetic therapies based on concentration under conditions such as starvation and diabetic ketoa­
novel HPTMs pathways. cidosis can give rise to many Kbhb histone markers in mouse liver
(Fig. 1). Based on these findings, it is reasonable to assume that the KD
2. General overview of histone modifications in diabetes and could also cause these changes. Kbhb is mainly enriched in active gene
complications of diabetes promoters related to genes up-regulated in the starvation-responsive
metabolic pathway [10]. In addition to indirectly participating in
Due to the inherent complexity of the pathogenesis of diabetes and HPTMs as an HDAC inhibitor [25], BHB can also directly regulate the
its complications, much effort has been put towards determining the complex process of gene transcription as an acyl donor, which greatly
underlying etiologies. In recent decades, accumulating evidence has expands the regulatory range of BHB in the field of epigenetics.
shown that HPTMs play a significant role in modulating gene expression Additionally, recent studies have revealed that Kbhb ameliorates
under the diabetic state and causing the phenomenon of “metabolic diabetic complications by regulating the expression of certain genes.
memory.” Some researchers have pointed out that BHB treatment upregulated the
Acetylation and methylation are the two classic and most thoroughly generation of vascular endothelial growth factor (VEGF) and promoted
studied histone modifications related to diabetes. A large volume of data endothelial repair after vascular injury in diabetic rats [26]. During this
has confirmed that these histone modifications are intimately related process, BHB caused a significant increase in total protein Kbhb and
with the “metabolic memory” phenomenon [14], inflammatory H3K9bhb content in the aorta of diabetic rats. However, whether
response [15], endothelial dysfunction [16], and other pathological H3K9bhb was the initiating factor promoting VEGF gene expression and
processes of diabetes. For example, compared with non-diabetic pa­ which enzymes were involved in this process were not further elabo­
tients, increased acetylation of histone H3 binding to TNF-α promoters rated in this report. A recent study linked Kbhb to another salutary
in diabetic blood monocytes has been found to promote transcription of biological impact. Luo et al. found that BHB could significantly upre­
this inflammatory gene in a hyperglycemic environment. In contrast, gulate the expression of MMP-2 by increasing the content of H3K9bhb in
overexpression of histone deacetylation (HDAC) can reverse this process the MMP-2 promoter in diabetic rats [27], which reduced the patho­
[17]. Another study in diabetic mice shows that histone H3K4 methyl­ logical changes of glomeruli and decreased the renal fibrosis index in a
transferase SET7/9 in monocytes can significantly inhibit concentration-dependent manner. The above research can provide an
TNF-α-induced inflammation [18]. In a study of diabetic retinopathy, essential experimental basis for the clinical application of BHB in
the levels of H4K20me3 and H3K9ac in the promoter and enhancer of treating diabetic kidney disease (DKD). The expression of VEGF not only
superoxide dismutase 2 (SOD2) were reported to increase due to hy­ contributes to diabetic angiopathy but also contributes to the damage
perglycemia, but after the termination of hyperglycemia, their levels did repair of diabetic retinopathy and nephropathy. Similarly, MMPs have

2
D. Li et al. Biomedicine & Pharmacotherapy 156 (2022) 113984

Fig. 1. Sources and regulatory processes of histone malonylation, β-hydroxybutyrylation, and succinylation in diabetes.
Glucose, fatty acids, and amino acids are transported to mitochondria for oxidative decomposition, where they are catalyzed by acyl-COA synthases to generate the
intermediates malonyl-CoA, BHB-CoA, and succinyl-CoA. These three types of acyl-CoA can be transported to the nucleus and catalyzed by the corresponding
acyltransferase or deacylase to regulate Kma, Kbhb, and Ksucc in histone lysine residues. These three novel HPTMs are involved in neural tube defects, vascular
endothelial dysfunction, glomerulosclerosis, and diabetic obesity, respectively. BHB-CoA, β-hydroxybutyryl CoA; TCA cycle, Tricarboxylic acid cycle; SIRT, Sirtuin;
CBP, CREB-binding proteins; GCN5, general control non-depressible 5; NTDs, neural tube defects; DIO, diet-induced obesity.

been found to be involved in the pathogenesis of retinopathy [28–30]. it can enhance glucose-stimulated insulin secretion and β-cell mass by
Therefore, it is not clear whether elevated Kbhb levels are tissue-specific inhibiting apoptosis [38]. The above findings support the crucial regu­
drivers of these genes. Accordingly, we were unable to identify the latory role of SCFAs produced in the gut in maintaining glucose ho­
maximum beneficial target organs for the up-regulation of VEGF and meostasis and attenuating microvascular complications caused by
MMP2 by Kbhb. The concentration-dependent regulatory role of Kbhb hyperglycemia. Besides that, microbial signals also calibrate the tran­
may need to be further elucidated in a wider range of genes. A recent scriptional program of host cells through short-chain lysine acylations
proteomics report on BHB quantified the distribution of modification directly on target histones.
sites and functional enrichment of Kbhb after BHB treatment in mouse In 2007, Yue’s group first reported two novel lysine modifications,
embryonic fibroblasts. The results showed that BHB-induced Kbhb Kpr and Kbu [4]. Kbu and Kpr are respectively derived from butyrate
proteins were mainly involved in glycolysis/gluconeogenesis, pyruvate and propionate, and they are catalyzed by ACSS2 to form small active
metabolism, and citrate cycle pathways, and 26 % of the Kbhb protein molecules similar to acetyl-CoA to mediate histone acylation processes
was located in the nucleus [31]. Single or multiple Kbhb sites in histones (Fig. 2). Histone Kbu and Kpr may have the possibility of extensive
may play an indispensable role in the occurrence and development of pathophysiological regulation similar to histone acetylation. Mass
diabetes through their transcriptional regulation of intermediate en­ spectrometry reveals they can be catalyzed in vitro by two well-known
zymes in glucose metabolism. acetyltransferases, p300 and CREB-binding proteins (CBP). Kpr and
Kbu are highly dynamic and preferentially enriched in the promoters of
3.2. Butyrylation and propionylation active genes. They can be detected in eukaryotic cells such as yeast cells,
mouse liver, and leukemia cell line U937, reflecting their high conser­
The microbial community can metabolize more than two dozen vation [39,40]. It is exciting that these two short-chain lysine acylations
short-chain fatty acids (SCFAs), including acetic, propionic, and butyric were discovered on histones in the livers of pre-diabetic mice with
acids, which are crucial bacterial fermentation products of high-fiber high-fat diet-induced obesity (DIO) in 2017 [41]. Specifically, compre­
foods [32]. Research in recent decades has highlighted the importance hensive proteomic analysis of histone samples extracted from known
of the composition and abundance of the gut microbiota in the physio­ and unknown histone marks indicated 15 Kpr marks and 12 Kbu marks
logical regulation of bacterial hosts, such as inhibiting intestinal in­ among the 45 newly discovered histone lysine acylation marks. The
flammatory responses and combating oxidative stress [33,34]. The detection of these histone marks had never been done before in diabetic
process of nutrient degradation by the symbiotic microbial community animal models. Furthermore, antibody affinity enrichment showed that
is also crucial for maintaining the host’s glucose homeostasis. One study Kpr and Kbu marks appeared on histones with a high presence rate.
found that butyrate synthesized by gut microbiota can improve glucose Recent studies have confirmed that intraperitoneal injection of sodium
tolerance and reduce glycated hemoglobin A1c levels to alleviate dia­ butyrate can up-regulate renal histone Pan-butyrylation (Pan-Kbu) and
betes [35,36]. Intake of sodium butyrate can inhibit high H3K9 butyrylation (H3K9bu) in DKD mice, and the proinflammatory
glucose-induced oxidative stress and inflammatory response in mesan­ factors MCP-1 and IL-6 and the fibrosis indicators COV-IV and TGF-β are
gial cells, reduce renal fibrosis, and reduce apoptosis to strengthen renal significantly down-regulated. Additionally, the anti-inflammatory and
function in diabetic rats [37]. Propionate also has positive effects on the anti-fibrotic effects of sodium butyrate are reversed after inhibition of
function of pancreatic β cells, and in vivo and in vitro analyses show that Kbu with A485, a histone modifying enzyme p300 inhibitor [42]. Due to

3
D. Li et al. Biomedicine & Pharmacotherapy 156 (2022) 113984

Fig. 2. Sources and regulatory processes of histone propionylation and butyrylation in diabetes.
Microorganisms such as bacteria grown on the surface of intestinal epithelial cells can digest dietary fiber derived from food by fermenting it into more than 20 kinds
of short-chain fatty acids (SCFAs), including propionate and butyrate. These SCFAs can be transported into cells by cell membrane channel monocarboxylate
transporters (MCTs), where they are catalyzed by ACSS2 to generate propionyl-CoA and butyryl-CoA. Meanwhile, the metabolic cycle of mitochondria can also serve
as a secondary source of propionyl-CoA and butyryl-CoA. As histone modification substrates, propionyl-CoA and butyryl-CoA are involved in the occurrence and
development of diabetes, diabetes-related kidney disease, and atherosclerosis under the catalysis of corresponding acyltransferases and deacylases. IR, insulin
resistance; ASCVD, atherosclerotic cardiovascular disease; KAT6, lysine acetyltransferase 6; PCAF, p300/CBP-associated factor; MOZ, monocytic leukemia zinc finger
protein; MOF, male absent on the first; HBO1, histone acetyltransferase binding to ORC1; GCN5, general control non-depressible 5; SIRT, Sirtuin.

the nonspecific nature of enzyme regulation of A485 and the fact that opening the door for subsequent functional studies related to histone
sodium butyrate is also an important inhibitor of HDAC, this report lacks modifications. Ksucc is a reversible, dynamic, abundant, and evolu­
experimental results related to the effect of BHB on acetylation and other tionarily conserved histone mark in eukaryotic cells. It has modification
histone modifications, and attention should be paid to the dynamic sites on histones H2A, H2B, H3, and H4. Ksucc may have a significant
changes in the abundance of other butyrylation modification sites in the effect on chromatin structure and function because it can produce mass
DKD model. Recent studies have identified a semisynthetic derivative shifts of up to + 100 Da, which is greater than the mass shifts produced
(LTK-14A) that has inhibitory effects on p300 and specifically inhibits by Kme or Kac. And the addition of succinylation substrates to lysine
Kbu without affecting the function of Kac [43]. This finding may residues can cause two charge shifts, which change the positively
accelerate our investigation of the ambiguous causal relationship be­ charged side chains to negatively charged side chains. These drastic
tween diabetes and Kbu. charge reversals and mass changes can obviously change the binding
Short-chain histone acylations may provide new insights into the ability of histone to DNA, which may be highly correlated with gene
epigenetic roles of gut microbiota metabolites involved in the patho­ expression regulation.
genesis or treatment of diabetes, so further studies need to explore the Current studies on the write-read-erase mechanism of Ksucc found
specific modification enzymes, modification sites, and effect on the that SIRT3, SIRT5, and SIRT7 in the deacetylase family have potent
expression of Kbu and Kpr target genes. desuccinylase activity [47–49]. Among them, SIRT5 is the most thor­
oughly studied desuccinylase. The levels of succinylation on mito­
3.3. Succinylation chondria, cytoplasm, and histones in the liver cells of SIRT5 gene
knockout mice were significantly increased. Another study found that
In most cases, intracellular succinyl-CoA originates from α-ketoglu­ overexpression of SIRT5 considerably reduced the levels of apoptosis
tarate, succinyl-carnitine, and succinate in the mitochondrial tricar­ factor Caspase-3 and MDA, upregulated the activation of the
boxylic acid (TCA) cycle. Moreover, carnitine shuttling during fatty acid anti-apoptosis factor B-cell lymphoma 2 (Bcl-2) in pancreatic β cells
β-oxidation also increases the level of succinyl-CoA [44,45]. The above damaged by high blood sugar levels, and improved the insulin secretion
two processes are closely related to the glycolipid metabolism abnor­ function [50]. These data indicate that SIRT5-mediated Ksucc may be
mality of diabetes. The connection between them reminds us that the involved in easing pancreatic β cell apoptosis and promoting the
high sugar environment and the metabolic imbalance of aerobic expression of genes related to insulin secretion, pointing to its potential
oxidation of glucose caused by diabetes can increase the level of role in maintaining glucose homeostasis. In a recent paper, Nie’s
succinyl-CoA, which participates in the development of diabetes. research team reported 30 previously unknown modification sites
Succinyl-CoA can cross the mitochondrial membrane and produce identified by HPLC-MS/MS analysis of histone samples extracted from
Ksucc in cytoplasmic proteins and nuclear histones [46]. In 2011, Zhang the liver of a pre-diabetic C57BL/6J mouse model induced by a high-fat
et al. discovered and characterized 69 succinylated lysine sites [5], diet for eight weeks. These included nine Ksucc sites, along with Kpr,

4
D. Li et al. Biomedicine & Pharmacotherapy 156 (2022) 113984

Kbu, and Kma sites [41], indicating that the three newly discovered higher malonyl-CoA concentrations in people with type 2 diabetes, two
histone acylations in the pre-diabetic mouse model may have some studies reported that compared with wild-type mice, higher levels of
unrevealed association with diabetes and obesity. Since the procedure in glucose and fatty acid oxidation were found in ACC2-/- mutant mice fed
this study used pan-antibody and did not involve affinity enrichment a high-fat and high-carbohydrate diet, and systemic insulin resistance
analysis for histone modifications of interest, it is reasonable to assume and hyperglycemia were improved in mice by ACC knockout or use of
that these newly discovered histone markers, including histone succi­ ACC2 selective inhibitors [55,56]. Another study found that in mice
nylation, have a high stoichiometric ratio in DIO mouse livers. with ACC1 and ACC2 double-knockout or phosphorylated inhibition,
Increasing evidence suggests that succinylation is a widespread malonyl-CoA could not be transformed by acetyl-CoA, resulting in
protein modification, and it is vital in many diseases, such as cancer, increased fat production, decreased fatty acid oxidation, increased levels
neonatal cardiac energy metabolism, Alzheimer’s disease [51], and of insulin resistance, and abnormal glucose tolerance [57]. These results
cardiovascular disease [52]. However, since current studies on the reveal an intrinsic link between metabolic pathways using malonyl-CoA
regulatory mechanism of succinylation generally focus on the enzyme as intermediates and the establishment of glucose homeostasis.
activity regulating the TCA cycle and the relationship between meta­ Kma is a novel modification based on malonyl-CoA. It was first
bolism and the four aforementioned diseases, little is known about most identified in 2011 by Peng et al. in Escherichia coli and HeLa cells using
other biological functions, even less in the field of diabetes. Therefore, it specific malonyl-lysine antibody, high-performance liquid
is necessary to investigate the lysine succinylation targets of histones in chromatography-tandem mass spectrometry (HPLC/MS/MS), and pro­
diabetic models besides H3K79, H4K31, H4K77, H2BK46, and H2BK85 tein sequence-alignment [7]. Functional enrichment analysis showed
(Table 1) and determine the factors involved in their upstream and that Kma was significantly enriched in glucose and fatty acid metabolic
downstream regulation. We require more clear evidence based on new pathways [58]. Kma has a large molecular size, can cause a mass shift of
molecular mechanisms of succinylation and desuccinylation to meet the + 86.00039 Da in histones, and is an evolutionally conserved modifi­
needs of future discovery and development of therapies and drugs for cation that is widespread in many species, including humans, fruit flies,
diabetes. zebrafish, and mice. Therefore, it is reasonable to predict that Kma af­
fects the binding between DNA and histones in many species. With the
3.4. Malonylation deepening of research on Kma, its sites on histones and function in
diabetes gradually surfaced. In the absence of antibody enrichment of
Malonyl-coA is an essential intermediate in fatty acid synthesis and core histones, seven malonylation sites were found in the DIO mouse
β-oxidation, and it is an inhibitor of fatty acid oxidation [53]. Two model of prediabetes, among which were H3K23, H3K56, H3K79,
distinct isoforms of acetyl-CoA carboxylase (ACC), ACC1 and ACC2, and H2BK108, H2BK116, and H2BK120 [41]. However, it is not clear which
malonyl-CoA decarboxylase (MCD) are the key catalytic enzymes of genes are regulated by modifications at these sites. A recent work
intracellular lipid metabolism, as they respectively mediate the syn­ documented that when mouse NE4C cells were cultured with glucose at
thesis and degradation of malonyl-coA [54]. In addition to finding different concentrations, the level of Kma was upregulated with the

Table 1
Summary of histone modifications in diabetes.
Histone Models Sites Regulatory Specific functions Ref.
modification enzymes

Kbhb ①Liver from prolonged fasting or H3:K9, K18 Writers: P300/CBP ①it can antagonize aortic endothelial injury by [10],
STZ-induced T1DM ketoacidosis Erasers: SIRT1–3, promoting generation of VEGF; [26],
mouse; HDAC1–3 ②It can upregulate MMP-2 and reduce diabetic [27]
②Kidney from STZ-induced diabetic Readers: not identified glomerular fibrosis.
SD rat
Kma ①Liver from high-fat diet-induced H2A: K74, K75, K95, Writers: Not available It may influence the regulation of genes and [7],
prediabetic mice; K99, K118, K119, K124 Erasers: SIRT2, SIRT3, pathways related to neurological complications [41],
②Brain from neural tube defect H2B:K12, K15, K34, SIRT5 caused by diabetes. [59]
(NTD) embryos of female diabetic K46, K57, K108, K116, Readers: not identified
mice; K120,
③High glucose induced NE4C cell H3:K4, K23, K37, K56,
K79, K119
H4:K5, K8, K12, K16,
K44, K59, K77, K79,
K91
Kpr Liver from high-fat diet-induced H1:K65 Writers: P300/CBP, Not identified [4],[41]
prediabetic mice H2B:K5, K12, K15, GCN5, PCAF, KAT6,
K16, K20, K108, K116 MOF, MOZ, HBO1
H3:K9, K18, K23, K27 Erasers: SIRT1–3, SIRT5
H4:K5, K12, K16 Readers: Bromodomain,
YEATS
Kbu ①Liver from high-fat diet-induced H2A:K9 Writers: P300/CBP, ①It may promote the transcriptional expression of [4],
prediabetic mice; H2B:K5, K12, K15, GCN5, PCAF, KAT6, Ins1 and Ins2 genes; [41],
②Kidney from DKD mice induced by K16, K20, H3:K18, K27 MOF, ②It may decrease the transcriptional expression of [42]
high-fat diet and STZ; H3:K9 HBO1 MCP-1, IL-6, TGF-β genes;
③High glucose induced SV- H4:K5, K8 Erasers: SIRT1–3
40MES13 cell Readers: Bromodomain,
YEATS
Ksucc Liver from high-fat diet-induced H1:K171 Writers: P300/CBP, Not identified [5],
prediabetic mice H2B:K46, K85, K108, GCN5 [41],
K116, K120 Erasers: SIRT3, SIRT5, [45]
H3: K23, K27, K56, SIRT7
K79, K122 Readers: YEATS
H4: K31, K77, K79, (GAS41)
K91

5
D. Li et al. Biomedicine & Pharmacotherapy 156 (2022) 113984

increase of glucose intervention concentration [59], suggesting a posi­ one of the known acetyl-CoA synthetases involved in important meta­
tive correlation between glucose concentration and the level of histone bolic processes in the body. A future research direction should be to
malonylation, which may be related to diabetic neuropathy mediated by verify whether ACSS2 can affect the synthesis of other types of acetyl-
high glucose. Neural tube defects (NTDs) are common congenital mal­ CoA, and to investigate how to make ACSS2 selectively regulate one
formations in the fetuses of diabetic women. Western blotting has novel histone modification without affecting its other functions. Some
detected increased histone malonylation levels of embryos with NTDs in studies have found that acyl-CoA synthetases, acyltransferases, and
STZ-induced female diabetic mice. These findings remind us that histone deacylases are involved in the pathogenesis of diabetes complications,
malonylation may also participate in neurological complications of but the regulation they mediate does not target specific modifications.
diabetes, and the enzymes regulating malonylation may be suitable Therefore, while studying one of these novel HPTMs, it is necessary to
targets for treating such complications. detect the dynamic changes of other modifications such as histone
SIRT5 is the primary demalonylase and it can mildly reduce Kma. acetylation. We still need to discover and utilize specific regulatory
There are only a few known modification sites of Kma, such as H3K4, enzymes to determine whether novel HPTMs could be beneficial for
H3K23, H3K56, H4K8, and H4K77. Key future directions in the study of diabetes control. A further complication is that we do not yet know
Kma should be to identify the many other “write-erase” enzymes whether different HPTMs cooperate with or antagonize each other.
mediating this modification, determine its specific or nonspecific sites, Therefore, the functions and underlying mechanisms of two or more
determine the levels of Kma in critical locations on histones associated novel combinations of HPTMs need to be investigated in the same
with diabetes, and assess the role of Kma in diabetes. experiment. Moreover, we should continue to investigate whether, like
Kme and Kac, other novel HPTMs found in diabetes and complications of
3.5. Benzoylation diabetes mediate the persistent epigenetic change called "metabolic
memory" that may be transferred to offspring.
Kbz is a novel histone modification added to lysine residues by the Above all, this review discusses how some novel HPTMs—repre­
food additive sodium benzoate and endogenous intestinal metabolite sented by β-hydroxybutyrylation, malonylation, propionylation, butyr­
benzoyl-CoA as substrates. It was discovered in 2018 as a functionally ylation, and succinylation—are closely or potentially related to the
conserved modification that exists in Drosophila, mouse, and human cell pathophysiology of diabetes and complications of diabetes. With the
lines. Because Kbz is the only known type of HPTM with an aromatic development of new site-specific antibodies in the future, the accuracy
ring structure and its mass migration can be as high as + 104 Da, the and detection range of modified omics technology will see great im­
probability of it regulating neighboring genes is greatly increased. provements. The catalog of histone modifications will continue to
Notably, intracellular benzoyl-CoA and global Kbz levels significantly expand, and future work will need to focus on establishing a complete
increased even when sodium benzoate was ingested at concentrations map of the associations between histone modification and diabetes and
below the maximum allowable addition concentration (~0.1 %) in foods exploring the precise functions and mechanisms of these associations.
set by the US Food and Drug Administration (FDA). KEGG functional Such work will enable the development of new prevention and treat­
enrichment analysis hinted that Kbz might regulate insulin secretion ment targets for diabetes and its complications.
[11]. However, no further experiments in vivo or in vitro have been
conducted to verify this possible relationship. Due to the unusual Funding
structure and prevalence of sodium benzoate in the food industry, it will
be fascinating to further investigate the role of Kbz in the regulation of This work was supported by the National Natural Science Foundation
genes related to insulin secretion, which could contribute to our un­ of China (NO.81970676) and the Luzhou-Southwest Medical University
derstanding of the pathological process of diabetes and the development cooperation project (NO.2021LZXNYD-P02).
of dietary strategies.
Author contributions
4. Conclusions and prospects
All authors conceived the manuscript structure and contributed to
With the advancement of proteomic technology, a mounting number
the writing and editing.
of histone acylation reactions have been discovered and confirmed to be
involved in the progress of diabetes and associated complications.
Indeed, almost all of the novel histone modifications discussed in this
Declaration of Competing Interest
review are highly enriched in the histones of diabetic models. To some
extent, they also reveal the common pathologic mechanisms between
The authors declare that they have no known competing financial
diabetes, obesity, and other metabolic diseases. Nevertheless, the re­
interests or personal relationships that could have appeared to influence
lationships between novel histone modifications and diabetes are still
the work reported in this paper.
understood on only a superficial level, and the associated molecular
mechanisms have not been thoroughly studied. Because only a small
Data availability
amount of evidence suggests that these novel HPTMs may be beneficial
for part of diabetes complications, it is not clear whether novel HPTMs
Data will be made available on request.
have pathological effects on diabetic metabolic homeostasis in terms of
genetics and pharmacology, so more extensive and in-depth patho­
physiological studies are urgently needed. References
Since most HPTMs are dynamic and reversible, intervention of up­ [1] Epidemiology of Diabetes Interventions and Complications (EDIC) Research Group.
stream or downstream targets in the formation of these modifications Epidemiology of Diabetes Interventions and Complications (EDIC). Design,
could give rise to new diabetes treatments. Moreover, these HPTMs implementation, and preliminary results of a long-term follow-up of the Diabetes
Control and Complications Trial cohort. Diabetes Care. 1999 Jan;22(1):99–111.
share the same acyl-CoA synthetases, acyltransferases, and deacylases,
[2] L. Zhang, Q. Lu, C. Chang, Epigenetics in health and disease, Adv. Exp. Med. Biol.
which can be taken advantage of to quickly find their regulatory en­ 1253 (2020) 3–55.
zymes and corresponding substrates and conduct in-depth research on [3] S. Brahma, S. Henikoff, Epigenome regulation by dynamic nucleosome
their biological functions. For example, we can continue to explore unwrapping, Trends Biochem. Sci. 45 (1) (2020) 13–26.
[4] Y. Chen, R. Sprung, Y. Tang, H. Ball, et al., Lysine propionylation and butyrylation
whether the classic acyltransferase P300/CBP can also catalyze the are novel post-translational modifications in histones, Mol. Cell Proteom. 6 (5)
addition of novel histone modifications identified in the future. ACSS2 is (2007) 812–819.

6
D. Li et al. Biomedicine & Pharmacotherapy 156 (2022) 113984

[5] Z. Zhang, M. Tan, Z. Xie, L. Dai, Y. Chen, Y. Zhao, Identification of lysine [33] D. Parada Venegas, M.K. De la Fuente, et al., Short chain fatty acids (SCFAs)-
succinylation as a new post-translational modification, Nat. Chem. Biol. 7 (1) mediated gut epithelial and immune regulation and its relevance for inflammatory
(2011) 58–63. bowel diseases, Front. Immunol. 10 (2019) 277.
[6] M. Tan, H. Luo, S. Lee, F. Jin, et al., Identification of 67 histone marks and histone [34] W. Huang, Y. Man, C. Gao, L. Zhou, et al., Short-chain fatty acids ameliorate
lysine crotonylation as a new type of histone modification, Cell 146 (6) (2011) diabetic nephropathy via GPR43-mediated inhibition of oxidative stress and NF-κB
1016–1028. signaling, Oxid. Med. Cell. Longev. 2020 (2020), 4074832.
[7] C. Peng, Z. Lu, Z. Xie, Z. Cheng, et al., The first identification of lysine malonylation [35] Z. Gao, J. Yin, J. Zhang, R.E. Ward, R.J. Martin, M. Lefevre, W.T. Cefalu, J. Ye,
substrates and its regulatory enzyme, Mol. Cell Proteom. 10 (12) (2011) M111. Butyrate improves insulin sensitivity and increases energy expenditure in mice,
[8] L. Dai, C. Peng, E. Montellier, et al., Lysine 2-hydroxyisobutyrylation is a widely Diabetes 58 (7) (2009) 1509–1517.
distributed active histone mark, Nat. Chem. Biol. 10 (5) (2014) 365–370. [36] A. Gonzalez, R. Krieg, et al., Sodium butyrate ameliorates insulin resistance and
[9] M. Tan, C. Peng, K.A. Anderson, P. Chhoy, et al., Lysine glutarylation is a protein renal failure in CKD rats by modulating intestinal permeability and mucin
posttranslational modification regulated by SIRT5, Cell Metab. 19 (4) (2014) expression, Nephrol. Dial. Transpl. 34 (5) (2019) 783–794.
605–617. [37] S. Khan, G. Jena, Sodium butyrate reduces insulin-resistance, fat accumulation and
[10] Z. Xie, D. Zhang, D. Chung, et al., Metabolic regulation of gene expression by dyslipidemia in type-2 diabetic rat: a comparative study with metformin, Chem.
histone lysine β-hydroxybutyrylation, Mol. Cell 62 (2) (2016) 194–206. Biol. Interact. 254 (2016) 124–134.
[11] H. Huang, D. Zhang, Y. Wang, M. Perez-Neut, Z. Han, Y.G. Zheng, Q. Hao, Y. Zhao, [38] A. Pingitore, E.S. Chambers, T. Hill, I.R. Maldonado, et al., The diet-derived short
Lysine benzoylation is a histone mark regulated by SIRT2, Nat. Commun. 9 (1) chain fatty acid propionate improves beta-cell function in humans and stimulates
(2018) 3374. insulin secretion from human islets in vitro, Diabetes Obes. Metab. 19 (2) (2017)
[12] D. Zhang, Z. Tang, H. Huang, et al., Metabolic regulation of gene expression by 257–265.
histone lactylation, Nature 574 (7779) (2019) 575–580. [39] K. Zhang, Y. Chen, Z. Zhang, Y. Zhao, Identification and verification of lysine
[13] C. Ling, T. Rönn, Epigenetics in human obesity and type 2 diabetes, Cell Metab. 29 propionylation and butyrylation in yeast core histones using PTMap software,
(5) (2019) 1028–1044. J. Proteome Res. 8 (2) (2009) 900–906.
[14] Q. Zhong, R.A. Kowluru, Role of histone acetylation in the development of diabetic [40] B. Liu, Y. Lin, A. Darwanto, X. Song, G. Xu, K. Zhang, Identification and
retinopathy and the metabolic memory phenomenon, J. Cell Biochem. 110 (6) characterization of propionylation at histone H3 lysine 23 in mammalian cells,
(2010) 1306–1313. J. Biol. Chem. 284 (47) (2009) 32288–32295.
[15] Z.A. Qu, X.J. Ma, S.B. Huang, et al., SIRT2 inhibits oxidative stress and [41] L. Nie, L. Shuai, M. Zhu, P. Liu, Z.F. Xie, S. Jiang, H.W. Jiang, J. Li, Y. Zhao, J.Y. Li,
inflammatory response in diabetic osteoarthritis, Eur. Rev. Med Pharmacol. Sci. 24 M. Tan, The landscape of histone modifications in a high-fat diet-induced obese
(6) (2020) 2855–2864. (DIO) mouse model, Mol. Cell Proteom. 16 (7) (2017) 1324–1334.
[16] Y. Liao, L. Gou, L. Chen, et al., NADPH oxidase 4 and endothelial nitric oxide [42] T. Zhou, H. Xu, X. Cheng, Y. He, Q. Ren, D. Li, Y. Xie, C. Gao, Y. Zhang, X. Sun,
synthase contribute to endothelial dysfunction mediated by histone methylations Y. Xu, W. Huang, Sodium butyrate attenuates diabetic kidney disease partially via
in metabolic memory, Free Radic. Biol. Med. 115 (2018) 383–394. histone butyrylation modification, Mediat. Inflamm. 2022 (2022), 7643322.
[17] F. Miao, I.G. Gonzalo, L. Lanting, R. Natarajan, In vivo chromatin remodeling [43] A. Bhattacharya, S. Chatterjee, U. Bhaduri, A.K. Singh, M. Vasudevan, et al.,
events leading to inflammatory gene transcription under diabetic conditions, Butyrylation meets adipogenesis-probed by a p300-catalyzed acylation-specific
J. Biol. Chem. 279 (17) (2004) 18091–18097. small molecule inhibitor: implication in anti-obesity therapy, J. Med. Chem. 65
[18] Y. Li, M.A. Reddy, et al., Role of the histone H3 lysine 4 methyltransferase, SET7/9, (18) (2022) 12273–12291.
in the regulation of NF-kappaB-dependent inflammatory genes. Relevance to [44] J. Smestad, L. Erber, Y. Chen, L.J. Maher 3rd, Chromatin succinylation correlates
diabetes and inflammation, J. Biol. Chem. 283 (39) (2008) 26771–26781 [doi: with active gene expression and is perturbed by defective TCA cycle metabolism,
10.1074/jbc.M802800200]. iScience 2 (2018) 63–75.
[19] Q. Zhong, R.A. Kowluru, Epigenetic changes in mitochondrial superoxide [45] K. Kurmi, S. Hitosugi, E.K. Wiese, et al., Carnitine palmitoyltransferase 1A has a
dismutase in the retina and the development of diabetic retinopathy, Diabetes 60 lysine succinyltransferase activity, Cell Rep. 22 (6) (2018) 1365–1373.
(4) (2011) 1304–1313. [46] B.T. Weinert, C. Schölz, et al., Lysine succinylation is a frequently occurring
[20] X. Yuan, J. Wang, S. Yang, et al., Effect of the ketogenic diet on glycemic control, modification in prokaryotes and eukaryotes and extensively overlaps with
insulin resistance, and lipid metabolism in patients with T2DM: a systematic acetylation, Cell Rep. 4 (4) (2013) 842–851.
review and meta-analysis, Nutr. Diabetes 10 (1) (2020) 38. [47] M.J. Rardin, W. He, Y. Nishida, J.C. Newman, et al., SIRT5 regulates the
[21] W.S. Yancy Jr, M.K. Olsen, et al., A low-carbohydrate, ketogenic diet versus a low- mitochondrial lysine succinylome and metabolic networks, Cell Metab. 18 (6)
fat diet to treat obesity and hyperlipidemia: a randomized, controlled trial, Ann. (2013) 920–933.
Intern. Med. 140 (10) (2004) 769–777. [48] J. Du, Y. Zhou, X. Su, J.J. Yu, et al., Sirt5 is a NAD-dependent protein lysine
[22] M.M. Poplawski, J.W. Mastaitis, F. Isoda, F. Grosjean, F. Zheng, C.V. Mobbs, demalonylase and desuccinylase, Science 334 (6057) (2011) 806–809.
Reversal of diabetic nephropathy by a ketogenic diet, PLOS One 6 (4) (2011), [49] H.B. Yu, S.T. Cheng, F. Ren, et al., SIRT7 restricts HBV transcription and replication
e18604. through catalyzing desuccinylation of histone H3 associated with cccDNA
[23] A. Dąbek, M. Wojtala, et al., Modulation of cellular biochemistry, epigenetics and minichromosome, Clin. Sci. 135 (12) (2021) 1505–1522.
metabolomics by ketone bodies. implications of the ketogenic diet in the [50] Y. Wang, Q. Liu, Y. Huan, R. Li, C. Li, S. Sun, N. Guo, M. Yang, S. Liu, Z. Shen,
physiology of the organism and pathological states, Nutrients 12 (3) (2020) 788. Sirtuin 5 overexpression attenuates glucolipotoxicity-induced pancreatic β cells
[24] H. Qi, L. Gu, D. Xu, K. Liu, et al., β-hydroxybutyrate inhibits cardiac microvascular apoptosis and dysfunction, Exp. Cell Res. 371 (1) (2018) 205–213.
collagen 4 accumulation by attenuating oxidative stress in streptozotocin-induced [51] Y. Yang, V. Tapias, D. Acosta, H. Xu, et al., Altered succinylation of mitochondrial
diabetic rats and high glucose treated cells, Eur. J. Pharmacol. 899 (2021), 174012. proteins, APP and tau in Alzheimer’s disease, Nat. Commun. 13 (1) (2022) 159.
[25] T. Shimazu, M.D. Hirschey, J. Newman, et al., Suppression of oxidative stress by [52] L. Yang, S. Miao, J. Zhang, P. Wang, G. Liu, J. Wang, The growing landscape of
β-hydroxybutyrate, an endogenous histone deacetylase inhibitor, Science 339 succinylation links metabolism and heart disease, Epigenomics 13 (4) (2021)
(6116) (2013) 211–214. 319–333.
[26] X. Wu, D. Miao, Z. Liu, K. Liu, B. Zhang, J. Li, Y. Li, J. Qi, β-hydroxybutyrate [53] D.W. Foster, Malonyl-CoA: the regulator of fatty acid synthesis and oxidation,
antagonizes aortic endothelial injury by promoting generation of VEGF in diabetic J. Clin. Investig. 122 (6) (2012) 1958–1959, https://doi.org/10.1172/jci63967.
rats, Tissue Cell 64 (2020), 101345. [54] D. Saggerson, Malonyl-CoA, a key signaling molecule in mammalian cells, Annu
[27] W. Luo, Y. Yu, H. Wang, K. Liu, Y. Wang, M. Huang, C. Xuan, Y. Li, J. Qi, Up- Rev. Nutr. 28 (2008) 253–272.
regulation of MMP-2 by histone H3K9 β-hydroxybutyrylation to antagonize [55] H. akagi, T. Ikehara, Y. Kashiwagi, et al., ACC2 deletion enhances IMCL reduction
glomerulosclerosis in diabetic rat, Acta Diabetol. 57 (12) (2020) 1501–1509. along with acetyl-CoA metabolism and improves insulin sensitivity in male mice,
[28] J. Song, B.B. Huang, J.X. Ong, N. Konopek, A.A. Fawzi, Hemodynamic effects of Endocrinology 159 (8) (2018) 3007–3019.
anti-vascular endothelial growth factor injections on optical coherence [56] H. Takagi, K. Tanimoto, A. Shimazaki, et al., A novel acetyl-CoA carboxylase 2
tomography angiography in diabetic macular edema eyes, Transl. Vis. Sci. Technol. selective inhibitor improves whole-body insulin resistance and hyperglycemia in
11 (10) (2022) 5. diabetic mice through target-dependent pathways, J. Pharmacol. Exp. Ther. 372
[29] B. Yang, X.H. Zhao, G.B. Ma, Role of serum β2-microglobulin, glycosylated (3) (2020) 256–263.
hemoglobin, and vascular endothelial growth factor levels in diabetic nephropathy, [57] M.D. Fullerton, S. Galic, K. Marcinko, et al., Single phosphorylation sites in Acc1
World J. Clin. Cases 10 (23) (2022) 8205–8211. and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of
[30] M. Caban, K. Owczarek, U. Lewandowska, The role of metalloproteinases and their metformin, Nat. Med. 19 (12) (2013) 1649–1654.
tissue inhibitors on ocular diseases: focusing on potential mechanisms, Int J. Mol. [58] Y. Du, T. Cai, T. Li, P. Xue, B. Zhou, X. He, et al., Lysine malonylation is elevated in
Sci. 23 (8) (2022) 4256. type 2 diabetic mouse models and enriched in metabolic associated proteins, Mol.
[31] W. Hou, G. Liu, X. Ren, X. Liu, L. He, H. Huang, Quantitative proteomics analysis Cell Proteom. 14 (1) (2015) 227–236.
expands the roles of lysine β-Hydroxybutyrylation pathway in response to [59] Q. Zhang, T. Cai, Z. Xiao, D. Li, C. Wan, X. Cui, B. Bai, Identification of histone
environmental β-Hydroxybutyrate, Oxid. Med. Cell. Longev. 2022 (2022), malonylation in the human fetal brain and implications for diabetes-induced
4592170. neural tube defects, Mol. Genet. Genom. Med. 8 (9) (2020), e1403.
[32] H.D. Holscher, Dietary fiber and prebiotics and the gastrointestinal microbiota, Gut
Microbes 8 (2) (2017) 172–184.

You might also like