Next Article in Journal
Brain Macro-Structural Alterations in Aging Rats: A Longitudinal Lifetime Approach
Next Article in Special Issue
Methylglyoxal-Modified Albumin Effects on Endothelial Arginase Enzyme and Vascular Function
Previous Article in Journal
The Crosstalk between Microbiome and Mitochondrial Homeostasis in Neurodegeneration
Previous Article in Special Issue
New Treatment for Type 2 Diabetes Mellitus Using a Novel Bipyrazole Compound
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diabetes and Its Cardiovascular Complications: Potential Role of the Acetyltransferase p300

by
Nadia Di Pietrantonio
1,*,
Pamela Di Tomo
1,
Domitilla Mandatori
1,
Gloria Formoso
2 and
Assunta Pandolfi
1
1
Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology-CAST, University G. D’Annunzio of Chieti-Pescara, 66100 Chieti, Italy
2
Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology-CAST, University G. D’Annunzio of Chieti-Pescara, 66100 Chieti, Italy
*
Author to whom correspondence should be addressed.
Cells 2023, 12(3), 431; https://doi.org/10.3390/cells12030431
Submission received: 21 December 2022 / Revised: 17 January 2023 / Accepted: 24 January 2023 / Published: 28 January 2023

Abstract

:
Diabetes has been shown to accelerate vascular senescence, which is associated with chronic inflammation and oxidative stress, both implicated in the development of endothelial dysfunction. This condition represents the initial alteration linking diabetes to related cardiovascular (CV) complications. Recently, it has been hypothesised that the acetyltransferase, p300, may contribute to establishing an early vascular senescent phenotype, playing a relevant role in diabetes-associated inflammation and oxidative stress, which drive endothelial dysfunction. Specifically, p300 can modulate vascular inflammation through epigenetic mechanisms and transcription factors acetylation. Indeed, it regulates the inflammatory pathway by interacting with nuclear factor kappa-light-chain-enhancer of activated B cells p65 subunit (NF-κB p65) or by inducing its acetylation, suggesting a crucial role of p300 as a bridge between NF-κB p65 and the transcriptional machinery. Additionally, p300-mediated epigenetic modifications could be upstream of the activation of inflammatory cytokines, and they may induce oxidative stress by affecting the production of reactive oxygen species (ROS). Because several in vitro and in vivo studies shed light on the potential use of acetyltransferase inhibitors, a better understanding of the mechanisms underlying the role of p300 in diabetic vascular dysfunction could help in finding new strategies for the clinical management of CV diseases related to diabetes.

Graphical Abstract

1. Cardiovascular Disease and Diabetes

Cardiovascular diseases (CVDs) are the main causes of death and disability among patients with diabetes. This was confirmed by a meta-analysis of 102 prospective studies, showing that diabetes confers a two-fold excess risk in adverse vascular outcomes, independently of other risk factors. CV complications associated with diabetes can be classified as microvascular, such as nephropathy and retinopathy, which are major causes of kidney disease and blindness, respectively, or macrovascular complications, including ischaemic heart disease, heart failure, stroke, coronary artery disease and peripheral artery disease [1,2,3]. Specifically, macrovascular damage in diabetes is characterised by atherosclerotic disease affecting coronary, cerebral and peripheral arteries [4].
Diabetes-associated endothelial dysfunction is one of the most critical initiating triggers of vascular complications and, therefore, an important predictor of the onset of CVD.
This is a very complex process orchestrated by several factors, such as hyperglycaemia, dyslipidaemia, free fatty acids and insulin resistance, which impair the normal functionality of the endothelium by promoting inflammation, oxidative stress, thrombosis, arterial stiffness and the impaired regulation of arterial tone and flow [4]. All these factors contribute to generating an imbalance between vasodilating and vasoconstricting factors, favouring proinflammatory and prothrombotic effects that promote atherosclerosis. In detail, hyperglycaemia can induce a series of cellular events that increase the production of ROS, such as superoxide anion, which induces oxidative stress and, in turn, leads to a reduced NO bioavailability [5].
In addition to impaired vasodilation, diabetes is also associated with increased circulating levels of endothelium-derived adhesion molecules, plasminogen activator inhibitor-1 and high triglyceride levels, which denote a proinflammatory endothelial phenotype. Inflammation is also driven by the increase in advanced glycation end products (AGEs), which result from the overproduction of mitochondrial superoxide anion. Diabetes is also characterised by high circulating levels of free fatty acids, which may compromise endothelial function through the activation of protein kinase C and the dysregulation of insulin pathway transduction.
All these processes exacerbate vascular inflammation and are characterised by the increased expression of several cytokines, such as Vascular cell adhesion molecule 1 (VCAM-1), E-selectin, Intercellular adhesion molecule-1 (ICAM-1), Interleukin-6 (IL-6), IL-1 and IL-8 and Monocyte chemoattractant protein-1 (MCP-1), facilitating the adhesion of monocytes, neutrophils and macrophages to the endothelium and, consequently, the formation of atherosclerotic plaque [6,7,8,9,10,11,12,13,14].
Although diabetes management has improved, the prevention and control of cardiovascular complications represent the main target of diabetes therapy [15], and the mechanisms by which hyperglycaemia contributes to CV system alteration need further elucidation.
Diabetic patients present dysfunction of multiple organ systems similar to that observed in chronological ageing, suggesting that this metabolic disease might induce an early ageing state associated with early vascular senescence, implicated in the development of endothelial dysfunction [16,17,18,19,20,21,22,23]. Therefore, a deep understanding of the molecular mechanisms underlying diabetic premature vascular senescence could lead to identifying new potential targets in order to control the progression of vascular damage in diabetes.

2. Cardiovascular Senescence and Diabetes

Cellular senescence is an irreversible arrest of the cell cycle, which has the purpose of repairing or eliminating damaged cells by restoring tissue homeostasis [24,25,26]. However, senescent cells can significantly increase, leading to biological ageing and the development of diseases. The process of senescence is triggered by various intrinsic and extrinsic stimuli, including genomic instability, epigenetic alterations, telomere modifications, mitochondrial dysfunction, stem cell exhaustion, mitogenic signals, oncogenic activation, radiation, oxidative stress, inflammation, tissue damage signals and nutrient deprivation [27]. All these stimuli lead to the upregulation of p53/p21, proteins with a central role in DNA repair and cell cycle regulation, p16/retinoblastoma protein (Rb) pathway activation and markers associated with activation of the DNA damage response (DDR), such as p38 mitogen-activated protein kinase (MAPK) and phosphorylated histone 2AX (cH2AX) [28,29].
Age increases the susceptibility to a wide variety of diseases, which can be considered as degenerative pathologies because they lead to the tissues’ normal functionality loss. Examples include neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, cardiovascular disease, musculoskeletal decrements and cancer [30,31]. Furthermore, evidence has shown that the senescence-associated secretory phenotype (SASP) can promote malignant phenotypes in culture and tumour growth in vivo [32]. Moreover, the accumulation of senescent cells has been also associated with the accelerated degeneration of bones and joints and skeletal muscle frailty [33].
Senescence has also been shown to be key in the pathophysiology of CVDs, such as atherosclerosis, myocardial infarction and cardiac fibrosis. Indeed, it was found that cardiac cells exhibit a SASP, which induces the release of soluble signalling factors, proteases and extracellular matrix (ECM) components in the surrounding environment that contribute to cardiovascular damage initiation and progression [34,35].
Numerous studies have shown that cardiovascular ageing is largely characterised by cardiomyocyte hypertrophy, increased senescence, cardiac fibroblast and endothelial dysfunction and decreased cardiac function. In particular, senescent cardiomyocytes show augmented cellular stressors, such as inflammation, ROS levels, DNA damage, endoplasmic reticulum stress, mitochondrial dysfunction, telomere shortening and SASP. All these factors determine contractile dysfunction loss and hypertrophic growth, which negatively affect myocardial function [35,36,37,38,39]. Of note, it was observed that patients who develop ventricular arrhythmias after acute myocardial infarction exhibit cardiomyocytes with a senescent phenotype characterised by increased telomere shortening [40]. Moreover, human appendages from patients with atrial fibrillation (AF) and in sinus rhythm show a higher expression of senescence markers such as p53 and p16 compared to controls, which correlate with prothrombotic and inflammatory protein expression, providing evidence of a strong correlation between AF progression and human atrial senescence [41].
In addition, an ageing heart may undergo fibrotic remodelling with senescent fibroblasts showing changes in the expression of genes regulating inflammation, extracellular matrix organization and angiogenesis. This leads to a higher proinflammatory response, autophagy and contractile dysfunction [42,43].
Senescence could also alter myofibroblast functionality: several studies have shown that senescent myofibroblasts, positive to senescence-associated beta-galactosidase (SA-β-gal), accumulate in the perivascular fibrotic areas of transverse aortic constriction-treated mice [44,45] and in the heart after myocardial infarction [45]. This was accompanied by the upregulation of the senescence regulator, p53, which was significantly upregulated in the infarcted heart or hypoxia-treated fibroblasts [45].
The dysregulation of p53 also appears to be involved in other age-related pathologies, such as hypertrophy cardiopathy. Specifically, it was found that p53 upregulation in pathological hypertrophy leads to the ubiquitylation and proteasomal degradation of Hypoxia-inducible factor 1α (HIF1α) through the E3 ubiquitin-protein ligase, MDM2, promoting heart failure [46,47,48].
Several pieces of evidence have demonstrated that senescence plays a key role in promoting vascular EC dysfunction through the dysregulation of the cell cycle, oxidative stress, altered calcium signalling and vascular inflammation (Figure 1). Senescent EC usually shows a flatter and enlarged phenotype with a polypoid nucleus. These changes could affect cytoskeleton structure, angiogenesis, proliferation and cell migration [18,20,49,50,51]. Of note, senescent EC shows increased endothelin-1 (ET-1), which promotes inflammation, the impairment of vascular relaxation and collagen accumulation [34,52,53,54]. At the same time inflammation, oxidative stress and reduced NO availability represent the main drivers of EC senescence, as reported in a recent study showing that prolonged Tumour Necrosis Factor α (TNF-α) exposure induces EC premature senescence, which is then blunted by the inhibition of NF-κB activation [20,55,56,57].
Diabetes, which is strictly associated with inflammation, oxidative stress and impaired endothelial function, has also been shown to accelerate vascular senescence and, consequently, favour the occurrence of CVD [19,20,21,22,58].
Several in vitro studies demonstrated that high-glucose-induced senescence in EC is accompanied by a reduction in NO synthesis [59,60], and interestingly, the upregulated expression of p22phox, a nicotinamide adenine dinucleotide phosphate (NADPH) oxidase component, which increases superoxide production, seems to be involved [61]. Moreover, high blood glucose levels also promote the accumulation of advanced glycation end products (AGEs) and, consequently, an increase in glycated proteins [62]. The latter has been associated with premature EC senescence and a reduction in NO bioavailability despite a three-fold increase in endothelial nitric oxide synthase (eNOS) expression [63].
Of note, animal models, such as Zucker diabetic rats and streptozotocin diabetic mice, show vascular cell senescence associated with an increase in SA β-Gal activity and p16INK4a and p53 expression [64,65,66,67,68,69,70].
In addition, several in vitro studies have highlighted the key role of the deacetylase Sirtuin 1 (SIRT1) in p53 activation through the regulation of acetylation. Indeed, SIRT1 leads to the inactivation of p53-driven DNA damage responses, which plays a major role in contributing to endothelial senescence [19,71,72,73,74]. Notably, the link between SIRT1 and p53 appears to be crucial in the phenomenon known as endothelial glycaemic memory, which, as better described in the following paragraph, contributes to the occurrence of CVD in diabetic patients, even when glycaemic control has been achieved [75,76].

Glycaemic Memory and Endothelial Senescence

The findings of the Diabetes Control and Complications Trial (DCCT), the follow-up observational Epidemiology of Diabetes Interventions and Complications (EDIC) and 10-year UKPDS follow-up studies in patients with type 1 (T1D) or type 2 (T2D) diabetes mellitus suggest that early exposure to hyperglycaemia prompts individuals to the development of diabetic complications—a phenomenon referred to as glycaemic memory [75,76,77]. According to that, EC exposed to a hyperglycaemic environment keep showing an altered phenotype even when an ideal glycaemic control has been achieved, contributing to the development of vascular complications in diabetic patients [6,78,79,80,81].
Notably, post-translational modification, such as the acetylation of p53, has been demonstrated to be involved in the “glycaemic memory” correlated with endothelial senescence. In detail, a study conducted by Zhang and colleagues showed that transient hyperglycaemic stress induces a persistent premature cellular senescence in EC, despite the subsequent restoration of normoglycaemia. This phenomenon was mediated by the deacetylase, SIRT1, and the acetyltransferase, p300, which modulate p53 activity. Specifically, three days of high-glucose incubation followed by three days of normal glucose induced a persistent downregulation of SIRT1 and the upregulation of acetyltransferase p300, which in turn drives the hyperacetylation and activation of p53, contributing to the maintenance of p53/p21-mediated senescent “memory” [73].
In this regard, our recent study contributed to strengthening the concept of the glycaemic memory of endothelial senescence. Thoroughly, we observed that EC isolated from the umbilical cords of gestational diabetic (GD-HUVECs) women compared to EC isolated from control women revealed an impaired antioxidant enzymatic defence, as well as increased basal and stimulated ROS levels associated with decreased nuclear localization of nuclear factor erythroid 2–related factor 2 (Nrf2)—an essential transcriptional factor regulating the antioxidant defence gene expression [82]. Furthermore, we found that GD-HUVEC, exposed to in vivo chronic hyperglycaemia, shows reduced SIRT1 activity, together with an increase in p300-mediated p53 acetylation compared to control cells. Finally, in agreement with the data from Zhang and colleagues, we demonstrated that p300 silencing reduced both the high-glucose-increased protein levels of p300 and acetylated p53 in control cells and their elevated basal levels in GD-cells, thus, indicating the possible involvement of SIRT1/P300/P53/P21 pathway in the early senescent GD-HUVEC phenotype [80].
Furthermore, several studies highlighted the role of p300 in establishing glycaemic memory also through epigenetic modifications, such as histone acetylation [79,83,84,85,86]. In the DCCT and EDIC trials, chromatin and DNA analysis performed in T1D patients revealed that monocytes from patients who developed complications during the subsequent EDIC follow-up study showed significant enrichment of histone 3 lysine 9 acetylation (H3K9ac), a gene-associated activation mark, located at key inflammatory loci [87,88]. In support of this evidence, it has been observed that prolonged exposure to specific inflammatory stimuli, such as TNF-α or lipopolysaccharides, determines an increase in histone 3 lysine 27 acetylation (H3K27ac) mediated by p300 at the gene enhancers imprinting a memory, which induce these cells to respond faster and stronger to a second inflammatory stimulation [89].
Therefore, we believe that a deep discernment of p300 function in inflammation and oxidative stress, the two main drivers of diabetes-related endothelial dysfunction, is urgently needed to support new pre-clinical and clinical studies.

3. Protein Lysine Acetylation by p300 in Diabetes

Several studies have pointed out the key role of acetylation in diabetes [90], which is a reversible reaction consisting of the transfer of an acetyl group from acetyl-coenzyme A (acetyl-CoA) to the ε-amino group of lysine residues [91,92,93]. Histone lysine acetylation is an epigenetic modification that is able to regulate gene transcriptional activity by inducing chromatin relaxation [91]. However, lysine acetylation also occurs in non-histone proteins, which modulate cellular processes, including gene transcription, cell cycle, cell division, DNA damage repair, signalling transduction, protein folding, protein aggregation and autophagy [94,95,96].
Acetylation is mainly controlled by histone acetyltransferases (HATs) and histone deacetylases (HDACs). HATs are classified into the following families: general control non-depressible 5 (GCN5), CREB-binding protein (CBP)/p300 and Moz, Ybf2/Sas3, Sas2 and Tip60 (MYST families). While the NAD+-dependent SIRT family is the most common type of HDAC [97].
The acetyltransferase, p300, regulates several cellular processes, including proliferation, migration, differentiation, senescence and apoptosis [98,99,100]. It drives histone acetylation, playing an important role as an epigenetic regulator through chromatin remodelling. In detail, p300 transfers the acetyl group from acetyl-CoA to histone lysine residues, leading to chromatin relaxation, which makes DNA more accessible for gene transcription [101,102]. It also functions as acetyltransferase for non-histone targets, such as transcription factors, enhancing their DNA-binding activity [73,80,103,104].
Importantly, p300 is involved in regulating numerous transcription factors, including NF-κB [105]. The N- and C-terminal domains of both CBP and p300 functionally interact with the region of p65 containing the transcriptional activation domain, playing an important role in the cytokine-induced expression of various immune and inflammatory genes and ECM proteins [106,107,108]. p300 also regulates myocyte enhancing factor 2 (MEF2) and GATA binding protein 4 (GATA4) [109]. Furthermore, p300 may control cellular processes by regulating protein–protein interactions as a coactivator. It is the case of the hypoxia gene transcription that requires the binding of p300 to hypoxia-inducible-factor-1alpha (HIF-1alpha) and some inflammatory gene promoters, which are bound by the p65 interacting with p300 [110,111,112].

4. Role of p300 in Diabetic Cardiovascular Complications

Numerous studies have documented the physiological role of p300 in development and heart diseases, such as accelerated cardiac hypertrophy, cardiomyopathy, matrix remodelling or fibrogenesis and heart failure [113]. The involvement of p300 acetyltransferase activity in mammalian heart development was highlighted by studies showing that mice with a mutation of p300, causing the lack of acetyltransferase domain, are characterised by embryonic lethality [114] and cardiomyopathy-associated cardiac dysfunction [115].
Recently, lysine acetylation has emerged as a major mechanism underlying diabetic vascular complications [116,117,118,119,120]. Indeed, supporting this idea, it has been demonstrated that acetyltransferase p300 may accelerate cardiac ageing by affecting ECM remodelling. This could be explained by the fact that p300 plays an essential role in profibrogenic cytokine transforming growth factor beta (TGF-β)-induced synthesis of type I collagen. In particular, the interaction of p300 with phospho-Smad2/3 turned out to be essential for matrix protein collagen synthesis and secretion by fibroblasts [121].
Additionally, p300 was significantly elevated in myofibroblast-like cells derived from the primary culture of mouse cardiac EC in response to profibrogenic cytokine TGF-β. On the contrary, the dissociation of p300 from the transcription initiation complex at the collagen gene promoter blunts profibrogenic signalling-induced type I collagen synthesis [122]. Interestingly, small acetyltransferase inhibitors suppress p300 activity in fibroblasts, reducing TGF-β-associated H3K9ac, myofibroblast differentiation and matrix protein synthesis [123]. Furthermore, p300 is known to control the transcription of a variety of genes involved in cardiac hypertrophy.
Gusterson and colleagues demonstrated that inhibition of p300 activity arrests cardiomyocyte hypertrophy induced by phenylephrine. On the other hand, p300 overexpression promotes this adverse cardiac phenotype [124,125]. It was also shown that phenylephrine induces the expression of p300, the acetylation of GATA4 and its binding to the ET-1 promoter in cardiomyocytes [126].
Notably, p300 levels are significantly elevated also in angiotensin (Ang) II-induced hypertensive myocardial tissues and are associated with increased H3K9ac and cardiac hypertrophy and fibrogenesis [127]. Moreover, p300-driven H3K9ac also appears to be involved in cardiomyopathy associated with loss-of-function mutations of Short-chain enoyl-CoA hydratase (ECHS1)—a key mitochondrial enzyme for fatty acid β-oxidation [128].
In vascular smooth muscle cells (VSMCs), it was also observed that p300 modulates the pro-atherogenic effect of 12(S)-Hydroxyeicosatetraenoic acid (12(S)-HETE) by mediating IL-6 and MCP-1 expression through p300-driven H3K9/14ac [129].
Recently, several studies pointed out the importance of p300 as a regulator of diabetes-associated CVD [36,85,113,130,131,132,133]. In fact, the expression of p300 was found to be remarkably elevated in the hearts of diabetic rats compared with normal controls. Accordingly, an increase in hypertrophy in neonatal rat cardiomyocytes exposed to high levels of glucose was observed [134]. Among the factors contributing to diabetic cardiopathy, there is also the accumulation of ECM that is responsible for the increase in fibrosis, in which p300 plays a key role by activating TGF-β via the acetylation of Smad2 [135]. Diabetes favours the production of ECM proteins also through the activation of NF-κB and the activator protein 1 (AP-1) signalling pathway, which appears to be regulated by p300 in diabetic rats [136]. This was also supported by an in vitro study demonstrating that p300 regulates the expression of vasoactive factors and ECM proteins in EC exposed to high glucose through histone acetylation [137].
As mentioned above, the tight interconnection between p300 and SIRT1 may regulate accelerated vascular ageing associated with hyperglycaemia. In this regard, EC exposed to high glucose and tissues from diabetic animals showed downregulation of SIRT1, which, in turn, leads to the reduction in mitochondrial antioxidant enzymes in a p300- and Forkhead box O1 (FOXO1)-mediated pathway [138].
The essential role of p300 and SIRT1 in high-glucose-induced vascular senescence was further confirmed in a cellular model of hyperglycaemia, evidencing the direct role of p300 in the activation of p53 and, consequently, p21 [73,80]. Additionally, p300 seems also to be involved in vascular tone regulation, as suggested by the fact that p300-mediated hyperacetylation of lysine residues in VSMCs has been associated with impairing VSMCs-dependent vasodilation in advanced T2D [130].

4.1. Inflammation

Diabetes is associated with augmented inflammation, which has been proven by a consistent body of evidence as one of the main drivers of atherosclerotic CVD [4,139] and is characterised by the increased activity of inflammasomes and increased levels of proinflammatory cytokines [140,141].
The role of p300 appears to be crucial in the regulation of NF-κB—one of the main mediators of the inflammatory pathway [142]. Lan and colleagues demonstrated in a T2D nephropathy mouse model that p300 leads to the activation of the NF-κB signalling pathway and the production of proinflammatory cytokines by a direct interaction with the subunit, p65 [103,143]. In diabetic mice, it was also shown that p300-driven H3K9/14ac modulates c-Jun N-terminal kinase (JNK)-downstream genes, such as connective tissue growth factor (CTGF), plasminogen activator inhibitor-1 (PAI-1) and fibronectin 1, and that the inhibition of p300 by a curcumin analogue (C66) was able to prevent renal injury and dysfunction in diabetic mice [144].
Several in vitro studies supported the key role of p300 in vascular inflammation associated with hyperglycaemia. Chen and colleagues demonstrated that glucose exposure increased p300 expression, histone acetylation and p300 binding to ET-1 and to fibronectin promoters in EC. This suggests that glucose-induced p300 upregulation regulates the gene expression of vasoactive factors and ECM proteins in EC through epigenetic mechanisms [137].
Moreover, in vitro studies have shown that the p300-driven acetylation of H3K9 recruits NF-κB to the promoters of proinflammatory genes, such as IL-6, IL-8 and cyclooxygenase-2 (COX-2) [145,146].
It was also found that high glucose increases the p300-driven acetylation of histones H3K14, H3K18, H3K23, H4K5 and H4K16 in retinal glial cells. These changes are positively correlated with the induction of the proinflammatory molecules ICAM-1, Inducible Nitric Oxide Synthase (iNOS) and Vascular Endothelial Growth Factor (VEGF). Of note, both the acetylation and expression of the inflammatory proteins can be inhibited by modifying HAT/HDAC activity [117].
The regulating Forkhead box O3a (FOXO3a) acetylation by p300 is another mechanism involved in diabetes-induced inflammation: diabetes-induced FOXO3a acetylation prevents its binding to gene target promoters, increasing NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome activation. This mechanism was then reverted by C646, a p300 inhibitor [147].

4.2. Oxidative Stress

Diabetes enhances oxidative stress and compromises antioxidant defences, as evidenced by increased superoxide generation and reduced Superoxide dismutase (SOD) activity catalase (CAT) and glutathione (GLT) [148]. Chronic hyperglycaemia also diminishes endothelial NO bioavailability, resulting in vascular homeostasis impairment, which, in turn, leads to a prothrombotic and proinflammatory state [149].
It is well known that eNOS plays a central role in vascular homeostasis by regulating the synthesis of NO and that the impairment of enzyme activity is involved in the pathogenesis of diabetic endothelial damage [150]. In this regard, histone acetylation appears to be highly implicated in the regulation of eNOS gene expression in EC; H3K9ac and H4K12ac at the eNOS promoter are functionally relevant to its expression, as confirmed by the treatment of cells with trichostatin A, a histone deacetylase inhibitor, which was associated with the increased acetylation of histones H3 and H4 at the eNOS proximal promoter [151].
In support of these results, the involvement of p300 in the regulation of eNOS was demonstrated. In detail, Chen and colleagues have shown that the acute stimulation of endothelial eNOS mRNA transcription by laminar shear stress is dependent on the NF-kB subunits, p50 and p65, which in turn bind to a shear stress response element (SSRE) in the human eNOS promoter. This is mediated by the activation of p300, which acetylates p65 and the histones, H3 and H4, in proximity to the human eNOS SSRE, promoting an opened chromatin structure and allowing gene transcription [152].
A recent study indicated a key role of histone acetyltransferase p300 in diabetes-accelerated renal damage. Interestingly, the authors observed that kidneys of diabetic C57BL/6J mice treated with C646, a pharmacological inhibitor of p300, showed significant downregulation of H3K27ac and diabetes-induced expression of NADPH oxidase (NOX), known to be the main trigger of ROS production [153].
SIRT1, antagonizing p300, has been shown to inhibit transcriptional factors, such as NF-κB, MAPK, Matrix metallopeptidase 9(MMP-9), FOXO3a and p53. On the other hand, it increases Cardiac sarcoplasmic reticulum Ca2+-ATPase2a (SERCA2a), extracellular signal-regulated protein kinase (ERK1/2), eNOS activity, proliferator-activated receptor-gamma coactivator α (PGC-1α) and AMP-activated protein Kinase (AMPK) [154]. The overexpression of SIRT1 was also able to decrease superoxide production and increase antioxidant enzyme SOD activity in diabetic ischaemic reperfused heart tissue [155]. Importantly, as mentioned above, SIRT1 plays a major role also in the regulation of eNOS activity by reducing eNOS acetylation and increasing its activation [156].
In VSMCs, HDAC1/2 and p300 proteins were found to bind the promoter’s sites of active NOX1/4/5 genes. H3K27 acetylation was increased at the promoters of NOX genes in high-glucose-exposed VSMCs [157].

5. Histone Acetyltransferase Inhibitors: Future Prospective

Considering the possibility of interfering with epigenetic modifications [131], Histone acetyltransferase inhibitors (HATis) could be explored as new potential drugs targeting the epigenetic mechanisms involved in vascular damage associated with diabetes [158].
Some compounds, such as metformin, already used in clinical practice and established as a first-line drug in the treatment of T2D patients, have shown some epigenetic effects [159]. Metformin may inhibit HATs through the activation of AMPK [160]. Furthermore, a recent study demonstrates that metformin can restrain phenylephrine-induced hypertrophic responses by inhibiting p300 activity in cardiomyocytes [161].
Interestingly, the activity of some pharmacological inhibitors targeting epigenetic modifiers for the treatment of cancer, such as JQ1 [162], has also been investigated in experimental models of diabetes complications. Notably, it was able to inhibit An-gII-regulated genes in vitro, as well as AngII-induced hypertension in mice [86].
To date, studies on the potential effect of drugs with proven cardio-renal benefits in diabetes, such as glucagon-like peptide 1 agonists (GLP1-as) and sodium-glucose co-transporter-2 inhibitors (SGLT2is), on p300 regulation are lacking. However, a recent study showed that liraglutide, a GLP1-a that has been reported to exert protective effects against myocardial ischaemia–reperfusion injury, displays a beneficial effect on renal ischaemia–reperfusion injury by decreasing histone acetyltransferase activity [163].
Regarding SGLT2i, the new drugs, β-hydroxybutyrate and dapagliflozin, have shown some effects on oxidative stress and cardioprotection, respectively, through epigenetic mechanisms [164,165]. Therefore, this evidence supports a further investigation to better define the effect of these drugs on the regulation of p300 expression and/or activity.
Among natural compounds, there is garcinol—a potent p300 inhibitor—that was able to mitigate high-glucose-induced inflammation in retinal cells, suggesting its potential use in the prevention of diabetic retinopathy [117,166].
A recent study demonstrated that epigallocatechin gallate (EGCG), the major bioactive polyphenol present in green tea, attenuates vascular inflammation via a repressive epigenetic effect on the NF-κB signalling pathway. In detail, this compound was able to block the recruitment of p65, p300 and HDAC1-3 in the promoters of IL-6, C-reactive Protein (CRP), ICAM-1, vascular cell adhesion molecule 1 (VCAM-1), IL-1α and IL-1β [142].
Curcumin has been considered a potential epidrug for diabetes because of its hypoglycaemic, hypolipidaemic and epigenetic effects in various rodent models [167]. Interestingly, a study conducted by Jung-Mi Yun and colleagues suggests that curcumin could improve glucose metabolism and prevent diabetic complications by activating HDAC-2 and inhibiting p300, which, in turn, reduce NF-κB signalling and vascular inflammation [168]. Furthermore, curcumin, through the modulation of the PKC-α and MAPK pathways, decreased the mRNA expression of transcriptional coactivator p300, the accumulation of ECM proteins and controlled oxidative stress and apoptosis in the heart of diabetic rats; this resulted in the attenuation of cardiomyocyte hypertrophy, myocardial fibrosis and left ventricular dysfunction [169].
Unfortunately, important evidence gaps remain before implementing the use of these substances in a clinical setting. These are mainly due to the lack of specificity in chromatin modifiers, implicating the unspecific and undesirable modulation of transcriptional programs with wide effects on cellular pathways. In addition, the safety of these approaches remains to be proven.

6. Conclusions

In summary, the evidence analysed and reported in this review supports the idea that p300 may be a relevant actor in diabetes-induced vascular inflammation and oxidative stress, which promote endothelial senescence, driving atherosclerotic cardiovascular disease.
As shown in Figure 2, it may modulate vascular alterations through both epigenetic mechanisms and transcription factor acetylation. Specifically, p300 may regulate the activation of the inflammatory signalling pathway by interacting directly with NF-κB or by inducing its acetylation. This suggests a crucial role of p300 as an important bridge between NF-κB p65 and the transcriptional machinery. Recently, the role of p300 as an epigenetic modifier that is able to regulate the upstream activation of inflammatory cytokines and oxidative stress in the setting of CV complication in diabetes has also emerged. Indeed, recent evidence unveiled a new function of p300 driving histone acetylation in establishing the so-called glycaemic memory, which turned out to be a key process in establishing permanent vascular damage.
Lastly, several in vitro and in vivo pre-clinical studies shed light on the potential use of acetyltransferase inhibitors, which, following further clinical validation, could be promising in the treatment of diabetes complications. Therefore, it would be very interesting to further study p300 as a potential therapeutic target that may contribute to the clinical management of diabetic vascular complications.

Author Contributions

N.D.P. was the lead researcher of this paper, and A.P. supervised all work performed. All authors (N.D.P., P.D.T., D.M., G.F. and A.P.) were involved in the conception and design of the article, as well as the drafting of the manuscript. The selection for study inclusion and data regarding each article was extracted by N.D.P. All authors (N.D.P., P.D.T., D.M., G.F. and A.P.) critically reviewed the submitted manuscript to ensure the accuracy of the study’s inclusion and interpretation. All authors (N.D.P., P.D.T., D.M., G.F. and A.P.) are accountable for every aspect of the work and ensure that the studies and points made in this manuscript are accurate and are completely factually based. All authors have read and agreed to the published version of the manuscript.

Funding

This research was founded by the PON-MISE Sustainable Growth Funding-DD27 September 2018 (prog. n. F/180021/01-04/X43). Domitilla Mandatori was supported by PON R&I “Ricerca e innovazione” 2014–2020.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Einarson, T.R.; Acs, A.; Ludwig, C.; Panton, U.H. Prevalence of cardiovascular disease in type 2 diabetes: A systematic literature review of scientific evidence from across the world in 2007–2017. Cardiovasc. Diabetol. 2018, 17, 83. [Google Scholar] [CrossRef] [Green Version]
  2. The Emerging Risk Factors Collaboration; Sarwar, N.; Gao, P.; Seshasai, S.R.; Gobin, R.; Kaptoge, S.; Di Angelantonio, E.; Ingelsson, E.; Lawlor, D.A.; Selvin, E.; et al. Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: A collaborative meta-analysis of 102 prospective studies. Lancet 2010, 375, 2215–2222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Grant, P.J.; Cosentino, F. The 2019 ESC Guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD: New features and the ‘Ten Commandments’ of the 2019 Guidelines are discussed by Professor Peter, J. Grant and Professor Francesco Cosentino, the Task Force chairmen. Eur. Heart J. 2019, 40, 3215–3217. [Google Scholar] [PubMed]
  4. Paneni, F.; Beckman, J.; Creager, M.A.; Cosentino, F. Diabetes and vascular disease: Pathophysiology, clinical consequences, and medical therapy: Part I. Eur. Hear. J. 2013, 34, 2436–2443. [Google Scholar] [CrossRef]
  5. Pandolfi, A.; De Filippis, E. Chronic hyperglicemia and nitric oxide bioavailability play a pivotal role in pro-atherogenic vascular modifications. Genes Nutr. 2007, 2, 195–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Di Fulvio, P.; Pandolfi, A.; Formoso, G.; Di Silvestre, S.; Di Tomo, P.; Giardinelli, A.; De Marco, A.; Di Pietro, N.; Taraborrelli, M.; Sancilio, S.; et al. Features of endothelial dysfunction in umbilical cord vessels of women with gestational diabetes. Nutr. Metab. Cardiovasc. Dis. 2014, 24, 1337–1345. [Google Scholar] [CrossRef] [PubMed]
  7. Gimbrone, M.A.; Garcia-Cardena, G., Jr. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016, 118, 620–636. [Google Scholar] [CrossRef] [Green Version]
  8. Henry, R.M.; Ferreira, I.; Kostense, P.J.; Dekker, J.M.; Nijpels, G.; Heine, R.J.; Kamp, O.; Bouter, L.M.; Stehouwer, C.D. Type 2 diabetes is associated with impaired endothelium-dependent, flow-mediated dilation, but impaired glucose metabolism is not: The Hoorn Study. Atherosclerosis 2004, 174, 49–56. [Google Scholar] [CrossRef] [Green Version]
  9. Higashi, Y.; Noma, K.; Yoshizumi, M.; Kihara, Y. Endothelial Function and Oxidative Stress in Cardiovascular Diseases. Circ. J. 2009, 73, 411–418. [Google Scholar] [CrossRef] [Green Version]
  10. Maruhashi, T.; Soga, J.; Fujimura, N.; Idei, N.; Mikami, S.; Iwamoto, Y.; Kajikawa, M.; Matsumoto, T.; Hidaka, T.; Kihara, Y.; et al. Relationship between flow-mediated vasodilation and cardiovascular risk factors in a large community-based study. Heart 2013, 99, 1837–1842. [Google Scholar] [CrossRef]
  11. Shi, Y.; Vanhoutte, P. Macro- and microvascular endothelial dysfunction in diabetes. J. Diabetes 2017, 9, 434–449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Ucci, M.; Di Tomo, P.; Tritschler, F.; Cordone, V.G.P.; Lanuti, P.; Bologna, G.; Di Silvestre, S.; Di Pietro, N.; Pipino, C.; Mandatori, D.; et al. Anti-inflammatory Role of Carotenoids in Endothelial Cells Derived from Umbilical Cord of Women Affected by Gestational Diabetes Mellitus. Oxidative Med. Cell. Longev. 2019, 2019, 8184656. [Google Scholar] [CrossRef] [PubMed]
  13. Vanhoutte, P.M.; Shimokawa, H.; Feletou, M.; Tang, E.H.C. Endothelial dysfunction and vascular disease—A 30th anniversary update. Acta Physiol. 2015, 219, 22–96. [Google Scholar] [CrossRef]
  14. Williams, S.B.; Cusco, J.A.; Roddy, M.-A.; Johnstone, M.T.; Creager, M.A. Impaired nitric oxide-mediated vasodilation in patients with non-insulin-dependent diabetes mellitus. J. Am. Coll. Cardiol. 1996, 27, 567–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Davies, M.J.; Aroda, V.R.; Collins, B.S.; Gabbay, R.A.; Green, J.; Maruthur, N.M.; Rosas, S.E.; Del Prato, S.; Mathieu, C.; Mingrone, G.; et al. Management of hyperglycaemia in type 2 diabetes, 2022. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia 2022, 65, 1925–1966. [Google Scholar] [CrossRef]
  16. Costantino, S.; Paneni, F.; Cosentino, F. Ageing, metabolism and cardiovascular disease. J. Physiol. 2016, 594, 2061–2073. [Google Scholar] [CrossRef]
  17. Kovacic, J.C.; Moreno, P.; Nabel, E.G.; Hachinski, V.; Fuster, V. Cellular Senescence, Vascular Disease, and Aging. Circulation 2011, 123, 1900–1910. [Google Scholar] [CrossRef] [Green Version]
  18. Shimizu, I.; Minamino, T. Cellular senescence in cardiac diseases. J. Cardiol. 2019, 74, 313–319. [Google Scholar] [CrossRef] [Green Version]
  19. Burton, D.G.A.; Faragher, R.G.A. Obesity and type-2 diabetes as inducers of premature cellular senescence and ageing. Biogerontology 2018, 19, 447–459. [Google Scholar] [CrossRef] [Green Version]
  20. Jia, G.; Aroor, A.R.; Jia, C.; Sowers, J.R. Endothelial cell senescence in aging-related vascular dysfunction. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2018, 1865, 1802–1809. [Google Scholar] [CrossRef]
  21. Katsuumi, G.; Shimizu, I.; Yoshida, Y.; Minamino, T. Vascular Senescence in Cardiovascular and Metabolic Diseases. Front. Cardiovasc. Med. 2018, 5, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Mooradian, A.D. Tissue specificity of premature aging in diabetes mellitus. The role of cellular replicative capacity. J. Am. Geriatr. Soc. 1988, 36, 831–839. [Google Scholar] [CrossRef] [PubMed]
  23. Palmer, A.K.; Gustafson, B.; Kirkland, J.L.; Smith, U. Cellular senescence: At the nexus between ageing and diabetes. Diabetologia 2019, 62, 1835–1841. [Google Scholar] [CrossRef] [Green Version]
  24. Burton, D.G.; Faragher, R. Cellular senescence: From growth arrest to immunogenic conversion. Age 2015, 37, 27. [Google Scholar] [CrossRef]
  25. Sapieha, P.; Mallette, F. Cellular Senescence in Postmitotic Cells: Beyond Growth Arrest. Trends Cell Biol. 2018, 28, 595–607. [Google Scholar] [CrossRef] [PubMed]
  26. Van Deursen, J.M. The role of senescent cells in ageing. Nature 2014, 509, 439–446. [Google Scholar] [CrossRef] [Green Version]
  27. Kumari, R.; Jat, P. Mechanisms of Cellular Senescence: Cell Cycle Arrest and Senescence Associated Secretory Phenotype. Front. Cell Dev. Biol. 2021, 9, 645593. [Google Scholar] [CrossRef]
  28. Freund, A.; Patil, C.; Campisi, J. p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. EMBO J. 2011, 30, 1536–1548. [Google Scholar] [CrossRef] [Green Version]
  29. Rufini, A.; Tucci, P.; Celardo, I.; Melino, G. Senescence and aging: The critical roles of p53. Oncogene 2013, 32, 5129–5143. [Google Scholar] [CrossRef]
  30. Campisi, J.; Andersen, J.K.; Kapahi, P.; Melov, S. Cellular senescence: A link between cancer and age-related degenerative disease? Semin. Cancer Biol. 2011, 21, 354–359. [Google Scholar] [CrossRef]
  31. Liu, R.M. Aging, Cellular Senescence, and Alzheimer’s Disease. Int. J. Mol. Sci. 2022, 23, 1989. [Google Scholar] [CrossRef] [PubMed]
  32. Krtolica, A.; Parrinello, S.; Lockett, S.; Desprez, P.-Y.; Campisi, J. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging. Proc. Natl. Acad. Sci. USA 2001, 98, 12072–12077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Wan, M.; Gray-Gaillard, E.; Elisseeff, J. Cellular senescence in musculoskeletal homeostasis, diseases, and regeneration. Bone Res. 2021, 9, 41. [Google Scholar] [CrossRef] [PubMed]
  34. Chen, M.S.; Lee, R.; Garbern, J. Senescence mechanisms and targets in the heart. Cardiovasc. Res. 2022, 118, 1173–1187. [Google Scholar] [CrossRef] [PubMed]
  35. Tang, X.; Li, P.; Chen, H. Cardiomyocyte Senescence and Cellular Communications Within Myocardial Microenvironments. Front. Endocrinol. 2020, 11, 280. [Google Scholar] [CrossRef]
  36. Ghosh, A.K. Acetyltransferase p300 Is a Putative Epidrug Target for Amelioration of Cellular Aging-Related Cardiovascular Disease. Cells 2021, 10, 2839. [Google Scholar] [CrossRef] [PubMed]
  37. Campisi, J. Cellular senescence: Putting the paradoxes in perspective. Curr. Opin. Genet. Dev. 2011, 21, 107–112. [Google Scholar] [CrossRef] [Green Version]
  38. Gude, N.A.; Broughton, K.M.; Firouzi, F.; Sussman, M.A. Cardiac ageing: Extrinsic and intrinsic factors in cellular renewal and senescence. Nat. Rev. Cardiol. 2018, 15, 523–542. [Google Scholar] [CrossRef]
  39. Masoud, S.; McDonald, F.; Bister, D.; Kotecki, C.; Bootman, M.D.; Rietdorf, K. Examining Cardiomyocyte Dysfunction Using Acute Chemical Induction of an Ageing Phenotype. Int. J. Mol. Sci. 2019, 21, 197. [Google Scholar] [CrossRef] [Green Version]
  40. Sawhney, V.; Campbell, N.G.; Brouilette, S.W.; Coppen, S.R.; Harbo, M.; Baker, V.; Ikebe, C.; Shintani, Y.; Hunter, R.J.; Dhinoja, M.; et al. Telomere shortening and telomerase activity in ischaemic cardiomyopathy patients—Potential markers of ventricular arrhythmia. Int. J. Cardiol. 2016, 207, 157–163. [Google Scholar] [CrossRef]
  41. Jesel, L.; Abbas, M.; Park, S.-H.; Matsushita, K.; Kindo, M.; Hasan, H.; Auger, C.; Sato, C.; Ohlmann, P.; Mazzucotelli, J.-P.; et al. Atrial Fibrillation Progression Is Associated with Cell Senescence Burden as Determined by p53 and p16 Expression. J. Clin. Med. 2019, 9, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Nicin, L.; Wagner, J.U.G.; Luxán, G.; Dimmeler, S. Fibroblast-mediated intercellular crosstalk in the healthy and diseased heart. FEBS Lett. 2021, 596, 638–654. [Google Scholar] [CrossRef] [PubMed]
  43. Vidal, R.; Wagner, J.U.G.; Braeuning, C.; Fischer, C.; Patrick, R.; Tombor, L.S.; Muhly-Reinholz, M.; John, D.; Kliem, M.; Conrad, T.; et al. Transcriptional heterogeneity of fibroblasts is a hallmark of the aging heart. J. Clin. Investig. 2019, 4, e131092. [Google Scholar] [CrossRef] [PubMed]
  44. Meyer, K.; Hodwin, B.; Ramanujam, D.; Engelhardt, S.; Sarikas, A. Essential Role for Premature Senescence of Myofibroblasts in Myocardial Fibrosis. J. Am. Coll. Cardiol. 2016, 67, 2018–2028. [Google Scholar] [CrossRef] [PubMed]
  45. Hu, C.; Zhang, X.; Teng, T.; Ma, Z.-G.; Tang, Q.-Z. Cellular Senescence in Cardiovascular Diseases: A Systematic Review. Aging Dis. 2022, 13, 103–128. [Google Scholar] [CrossRef] [PubMed]
  46. Caturano, A.; Vetrano, E.; Galiero, R.; Salvatore, T.; Docimo, G.; Epifani, R.; Alfano, M.; Sardu, C.; Marfella, R.; Rinaldi, L.; et al. Cardiac Hypertrophy: From Pathophysiological Mechanisms to Heart Failure Development. Rev. Cardiovasc. Med. 2022, 23, 165. [Google Scholar] [CrossRef]
  47. Ravi, R.; Mookerjee, B.; Bhujwalla, Z.M.; Sutter, C.H.; Artemov, D.; Zeng, Q.; Dillehay, L.E.; Madan, A.; Semenza, G.L.; Bedi, A. Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1α. Genes Dev. 2000, 14, 34–44. [Google Scholar] [CrossRef]
  48. Sano, M.; Minamino, T.; Toko, H.; Miyauchi, H.; Orimo, M.; Qin, Y.; Akazawa, H.; Tateno, K.; Kayama, Y.; Harada, M.; et al. p53-induced inhibition of Hif-1 causes cardiac dysfunction during pressure overload. Nature 2007, 446, 444–448. [Google Scholar] [CrossRef]
  49. Matsushita, H.; Chang, E.; Glassford, A.J.; Cooke, J.P.; Chiu, C.-P.; Tsao, P.S. eNOS Activity Is Reduced in Senescent Human Endothelial Cells. Circ. Res. 2001, 89, 793–798. [Google Scholar] [CrossRef]
  50. Rossman, M.J.; Kaplon, R.E.; Hill, S.D.; McNamara, M.N.; Santos-Parker, J.R.; Pierce, G.L.; Seals, U.R.; Donato, A.J. Endothelial cell senescence with aging in healthy humans: Prevention by habitual exercise and relation to vascular endothelial function. Am. J. Physiol. Circ. Physiol. 2017, 313, H890–H895. [Google Scholar] [CrossRef]
  51. Uryga, A.K.; Bennett, M. Ageing induced vascular smooth muscle cell senescence in atherosclerosis. J. Physiol. 2016, 594, 2115–2124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Ishibazawa, A.; Nagaoka, T.; Takahashi, T.; Yamamoto, K.; Kamiya, A.; Ando, J.; Yoshida, A. Effects of Shear Stress on the Gene Expressions of Endothelial Nitric Oxide Synthase, Endothelin-1, and Thrombomodulin in Human Retinal Microvascular Endothelial Cells. Investig. Opthalmology Vis. Sci. 2011, 52, 8496–8504. [Google Scholar] [CrossRef] [PubMed]
  53. Olmos, G.; Martínez-Miguel, P.; Alcalde-Estevez, E.; Medrano, D.; Sosa, P.; Rodríguez-Mañas, L.; Naves-Diaz, M.; Rodríguez-Puyol, D.; Ruiz-Torres, M.P.; López-Ongil, S. Hyperphosphatemia induces senescence in human endothelial cells by increasing endothelin-1 production. Aging Cell 2017, 16, 1300–1312. [Google Scholar] [CrossRef] [PubMed]
  54. Wang, X.; Guo, Z.; Ding, Z.; Khaidakov, M.; Lin, J.; Xu, Z.; Sharma, S.G.; Jiwani, S.; Mehta, J.L. Endothelin-1 upregulation mediates aging-related cardiac fibrosis. J. Mol. Cell. Cardiol. 2015, 80, 101–109. [Google Scholar] [CrossRef] [PubMed]
  55. Abbas, M.; Jesel, L.; Auger, C.; Amoura, L.K.; Messas, N.; Manin, G.; Rumig, C.; León-González, A.J.; Ribeiro, T.P.; Silva, G.C.; et al. Endothelial Microparticles From Acute Coronary Syndrome Patients Induce Premature Coronary Artery Endothelial Cell Aging and Thrombogenicity: Role of the ang II/AT1 receptor/NADPH oxidase-mediated activation of MAPKs and PI3-Kinase pathways. Circulation 2017, 135, 280–296. [Google Scholar] [CrossRef]
  56. Brodsky, S.V.; Gealekman, O.; Chen, J.; Zhang, F.; Togashi, N.; Crabtree, M.; Gross, S.S.; Nasjletti, A.; Goligorsky, M.S. Prevention and Reversal of Premature Endothelial Cell Senescence and Vasculopathy in Obesity-Induced Diabetes by Ebselen. Circ. Res. 2004, 94, 377–384. [Google Scholar] [CrossRef] [Green Version]
  57. Khan, S.Y.; Awad, E.M.; Oszwald, A.; Mayr, M.; Yin, X.; Waltenberger, B.; Stuppner, H.; Lipovac, M.; Uhrin, P.; Breuss, J.M. Premature senescence of endothelial cells upon chronic exposure to TNFα can be prevented by N-acetyl cysteine and plumericin. Sci. Rep. 2017, 7, 39501. [Google Scholar] [CrossRef] [Green Version]
  58. Imanishi, T.; Hano, T.; Sawamura, T.; Nishio, I. Oxidized Low-Density Lipoprotein Induces Endothelial Progenitor Cell Senescence, Leading to Cellular Dysfunction. Clin. Exp. Pharmacol. Physiol. 2004, 31, 407–413. [Google Scholar] [CrossRef]
  59. Matsui-Hirai, H.; Hayashi, T.; Yamamoto, S.; Ina, K.; Maeda, M.; Kotani, H.; Iguchi, A.; Ignarro, L.J.; Hattori, Y. Dose-Dependent Modulatory Effects of Insulin on Glucose-Induced Endothelial Senescence In Vitro and In Vivo: A Relationship between Telomeres and Nitric Oxide. Experiment 2011, 337, 591–599. [Google Scholar] [CrossRef] [Green Version]
  60. Zhong, W.; Zou, G.; Gu, J.; Zhang, J. L-arginine attenuates high glucose-accelerated senescence in human umbilical vein endothelial cells. Diabetes Res. Clin. Pr. 2010, 89, 38–45. [Google Scholar] [CrossRef]
  61. Maeda, M.; Hayashi, T.; Mizuno, N.; Hattori, Y.; Kuzuya, M. Intermittent High Glucose Implements Stress-Induced Senescence in Human Vascular Endothelial Cells: Role of Superoxide Production by NADPH Oxidase. PLoS ONE 2015, 10, e0123169. [Google Scholar] [CrossRef]
  62. Singh, V.P.; Bali, A.; Singh, N.; Jaggi, A.S. Advanced Glycation End Products and Diabetic Complications. Korean J. Physiol. Pharmacol. 2014, 18, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Chen, J.; Brodsky, S.V.; Goligorsky, D.M.; Hampel, D.J.; Li, H.; Gross, S.S.; Goligorsky, M.S. Glycated Collagen I Induces Premature Senescence-Like Phenotypic Changes in Endothelial Cells. Circ. Res. 2002, 90, 1290–1298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Chen, J.; Park, H.-C.; Patschan, S.; Brodsky, S.V.; Gealikman, O.; Kuo, M.-C.; Li, H.; Addabbo, F.; Zhang, F.; Nasjletti, A.; et al. Premature vascular senescence in metabolic syndrome: Could it be prevented and reversed by a selenorganic antioxidant and peroxynitrite scavenger ebselen? Drug Discov. Today: Ther. Strat. 2007, 4, 93–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Gu, J.; Wang, S.; Guo, H.; Tan, Y.; Liang, Y.; Feng, A.; Liu, Q.; Damodaran, C.; Zhang, Z.; Keller, B.B.; et al. Inhibition of p53 prevents diabetic cardiomyopathy by preventing early-stage apoptosis and cell senescence, reduced glycolysis, and impaired angiogenesis. Cell Death Dis. 2018, 9, 82. [Google Scholar] [CrossRef] [Green Version]
  66. Hayashi, T.; Kotani, H.; Yamaguchi, T.; Taguchi, K.; Iida, M.; Ina, K.; Maeda, M.; Kuzuya, M.; Hattori, Y.; Ignarro, L.J. Endothelial cellular senescence is inhibited by liver X receptor activation with an additional mechanism for its atheroprotection in diabetes. Proc. Natl. Acad. Sci. USA 2014, 111, 1168–1173. [Google Scholar] [CrossRef] [Green Version]
  67. Orimo, M.; Minamino, T.; Miyauchi, H.; Tateno, K.; Okada, S.; Moriya, J.; Komuro, I. Protective Role of SIRT1 in Diabetic Vascular Dysfunction. Arter. Thromb. Vasc. Biol. 2009, 29, 889–894. [Google Scholar] [CrossRef] [Green Version]
  68. Prattichizzo, F.; De Nigris, V.; Mancuso, E.; Spiga, R.; Giuliani, A.; Matacchione, G.; Lazzarini, R.; Marcheselli, F.; Recchioni, R.; Testa, R.; et al. Short-term sustained hyperglycaemia fosters an archetypal senescence-associated secretory phenotype in endothelial cells and macrophages. Redox Biol. 2017, 15, 170–181. [Google Scholar] [CrossRef]
  69. Shosha, E.; Xu, Z.; Narayanan, S.P.; Lemtalsi, T.; Fouda, A.Y.; Rojas, M.; Xing, J.; Fulton, D.; Caldwell, R. Mechanisms of Diabetes-Induced Endothelial Cell Senescence: Role of Arginase 1. Int. J. Mol. Sci. 2018, 19, 1215. [Google Scholar] [CrossRef] [Green Version]
  70. Yokoi, T.; Fukuo, K.; Yasuda, O.; Hotta, M.; Miyazaki, J.; Takemura, Y.; Kawamoto, H.; Ichijo, H.; Ogihara, T. Apoptosis Signal-Regulating Kinase 1 Mediates Cellular Senescence Induced by High Glucose in Endothelial Cells. Diabetes 2006, 55, 1660–1665. [Google Scholar] [CrossRef]
  71. Li, Y.; Peng, Z.; Wang, C.; Li, L.; Leng, Y.; Chen, R.; Yuan, H.; Zhou, S.; Zhang, Z.; Chen, A.F.; et al. Novel role of PKR in palmitate-induced Sirt1 inactivation and endothelial cell senescence. Am. J. Physiol. Circ. Physiol. 2018, 315, H571–H580. [Google Scholar] [CrossRef]
  72. Ota, H.; Akishita, M.; Eto, M.; Iijima, K.; Kaneki, M.; Ouchi, Y. Sirt1 modulates premature senescence-like phenotype in human endothelial cells. J. Mol. Cell. Cardiol. 2007, 43, 571–579. [Google Scholar] [CrossRef] [PubMed]
  73. Zhang, E.; Guo, Q.; Gao, H.; Xu, R.; Teng, S.; Wu, Y. Metformin and Resveratrol Inhibited High Glucose-Induced Metabolic Memory of Endothelial Senescence through SIRT1/p300/p53/p21 Pathway. PLoS ONE 2015, 10, e0143814. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, T.; Tian, F.; Wang, J.; Zhou, S.; Dong, X.; Guo, K.; Jing, J.; Zhou, Y.; Chen, Y. Donepezil attenuates high glucose-accelerated senescence in human umbilical vein endothelial cells through SIRT1 activation. Cell Stress Chaperones 2015, 20, 787–792. [Google Scholar] [CrossRef] [PubMed]
  75. Nathan, D.M.; Cleary, P.A.; Backlund, J.-Y.C.; Genuth, S.M.; Lachin, J.; Orchard, T.; Raskin, P.; Zinman, B.; Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study Research Group. Intensive Diabetes Treatment and Cardiovascular Disease in Patients with Type 1 Diabetes. N. Engl. J. Med. 2005, 353, 2643–2653. [Google Scholar] [CrossRef] [PubMed]
  76. The Writing Team for the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications Research Group. Sustained effect of intensive treatment of type 1 diabetes mellitus on development and progression of diabetic nephropathy: The Epidemiology of Diabetes Interventions and Complications (EDIC) study. JAMA 2003, 290, 2159–2167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Holman, R.R.; Paul, S.K.; Bethel, M.A.; Matthews, D.R.; Neil, H.A. 10-year follow-up of intensive glucose control in type 2 diabetes. N. Engl. J. Med. 2008, 359, 1577–1589. [Google Scholar] [CrossRef] [Green Version]
  78. Ceriello, A.; Ihnat, M.; Thorpe, J. Clinical review 2: The “metabolic memory”: Is more than just tight glucose control necessary to prevent diabetic complications? J. Clin. Endocrinol. Metab. 2009, 94, 410–415. [Google Scholar] [CrossRef] [Green Version]
  79. Coco, C.; Sgarra, L.; Potenza, M.A.; Nacci, C.; Pasculli, B.; Barbano, R.; Parrella, P.; Montagnani, M. Can Epigenetics of Endothelial Dysfunction Represent the Key to Precision Medicine in Type 2 Diabetes Mellitus? Int. J. Mol. Sci. 2019, 20, 2949. [Google Scholar] [CrossRef] [Green Version]
  80. Di Tomo, P.; Alessio, N.; Falone, S.; Pietrangelo, L.; Lanuti, P.; Cordone, V.; Santini, S.J.; Di Pietrantonio, N.; Marchisio, M.; Protasi, F.; et al. Endothelial cells from umbilical cord of women affected by gestational diabetes: A suitable in vitro model to study mechanisms of early vascular senescence in diabetes. FASEB J. 2021, 35, e21662. [Google Scholar] [CrossRef]
  81. Natarajan, R. Epigenetic Mechanisms in Diabetic Vascular Complications and Metabolic Memory: The 2020 Edwin Bierman Award Lecture. Diabetes 2021, 70, 328–337. [Google Scholar] [CrossRef]
  82. Chen, B.; Lu, Y.; Chen, Y.; Cheng, J. The role of Nrf2 in oxidative stress-induced endothelial injuries. J. Endocrinol. 2015, 225, R83–R99. [Google Scholar] [CrossRef] [Green Version]
  83. Cesselli, D.; Beltrami, A. Stem cell senescence in diabetes: Forgetting the sweet old memories. Diabetes 2014, 63, 1841–1843. [Google Scholar] [CrossRef] [Green Version]
  84. Christ, A.; Günther, P.; Lauterbach, M.A.; Duewell, P.; Biswas, D.; Pelka, K.; Scholz, C.J.; Oosting, M.; Haendler, K.; Baßler, K.; et al. Western Diet Triggers NLRP3-Dependent Innate Immune Reprogramming. Cell 2018, 172, 162–175.e14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Diedisheim, M.; Carcarino, E.; Vandiedonck, C.; Roussel, R.; Gautier, J.-F.; Venteclef, N. Regulation of inflammation in diabetes: From genetics to epigenomics evidence. Mol. Metab. 2020, 41, 101041. [Google Scholar] [CrossRef] [PubMed]
  86. Kato, M.; Natarajan, R. Epigenetics and epigenomics in diabetic kidney disease and metabolic memory. Nat. Rev. Nephrol. 2019, 15, 327–345. [Google Scholar] [CrossRef] [PubMed]
  87. Chen, Z.; Miao, F.; Paterson, A.D.; Lachin, J.M.; Zhang, L.; Schones, D.E.; Wu, X.; Wang, J.; Tompkins, J.D.; Genuth, S.; et al. Epigenomic profiling reveals an association between persistence of DNA methylation and metabolic memory in the DCCT/EDIC type 1 diabetes cohort. Proc. Natl. Acad. Sci. USA 2016, 113, E3002–E3011. [Google Scholar] [CrossRef] [Green Version]
  88. Miao, F.; Chen, Z.; Genuth, S.; Paterson, A.; Zhang, L.; Wu, X.; Li, S.M.; Cleary, P.; Riggs, A.; Harlan, D.M.; et al. Evaluating the Role of Epigenetic Histone Modifications in the Metabolic Memory of Type 1 Diabetes. Diabetes 2014, 63, 1748–1762. [Google Scholar] [CrossRef] [Green Version]
  89. Park, S.H.; Kang, K.; Giannopoulou, E.; Qiao, Y.; Kang, K.; Kim, G.; Park-Min, K.-H.; Ivashkiv, L.B. Type I interferons and the cytokine TNF cooperatively reprogram the macrophage epigenome to promote inflammatory activation. Nat. Immunol. 2017, 18, 1104–1116. [Google Scholar] [CrossRef] [Green Version]
  90. Chatterjee, B.; Thakur, S.S. Investigation of post-translational modifications in type 2 diabetes. Clin. Proteom. 2018, 15, 32. [Google Scholar] [CrossRef]
  91. Verdin, E.; Ott, M. 50 years of protein acetylation: From gene regulation to epigenetics, metabolism and beyond. Nat. Rev. Mol. Cell Biol. 2014, 16, 258–264. [Google Scholar] [CrossRef] [PubMed]
  92. Xiong, Y.; Guan, K.-L. Mechanistic insights into the regulation of metabolic enzymes by acetylation. J. Cell Biol. 2012, 198, 155–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Zhang, J.; Sprung, R.; Pei, J.; Tan, X.; Kim, S.; Zhu, H.; Liu, C.-F.; Grishin, N.V.; Zhao, Y. Lysine Acetylation Is a Highly Abundant and Evolutionarily Conserved Modification in Escherichia Coli. Mol. Cell. Proteom. 2009, 8, 215–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Narita, T.; Weinert, B.T.; Choudhary, C. Functions and mechanisms of non-histone protein acetylation. Nat. Rev. Mol. Cell Biol. 2018, 20, 156–174. [Google Scholar] [CrossRef] [PubMed]
  95. Spange, S.; Wagner, T.; Heinzel, T.; Krämer, O.H. Acetylation of non-histone proteins modulates cellular signalling at multiple levels. Int. J. Biochem. Cell Biol. 2009, 41, 185–198. [Google Scholar] [CrossRef] [PubMed]
  96. Zhao, S.; Xu, W.; Jiang, W.; Yu, W.; Lin, Y.; Zhang, T.; Yao, J.; Zhou, L.; Zeng, Y.; Li, H.; et al. Regulation of Cellular Metabolism by Protein Lysine Acetylation. Science 2010, 327, 1000–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Berndsen, C.E.; Denu, J. Catalysis and substrate selection by histone/protein lysine acetyltransferases. Curr. Opin. Struct. Biol. 2008, 18, 682–689. [Google Scholar] [CrossRef] [Green Version]
  98. Chan, H.M.; La Thangue, N. p300/CBP proteins: HATs for transcriptional bridges and scaffolds. J. Cell Sci. 2001, 114, 2363–2373. [Google Scholar] [CrossRef]
  99. Dancy, B.M.; Cole, P. Protein lysine acetylation by p300/CBP. Chem. Rev. 2015, 115, 2419–2452. [Google Scholar] [CrossRef]
  100. Dyson, H.J.; Wright, P. Role of Intrinsic Protein Disorder in the Function and Interactions of the Transcriptional Coactivators CREB-binding Protein (CBP) and p300. J. Biol. Chem. 2016, 291, 6714–6722. [Google Scholar] [CrossRef]
  101. He, Z.-X.; Wei, B.-F.; Zhang, X.; Gong, Y.-P.; Ma, L.-Y.; Zhao, W. Current development of CBP/p300 inhibitors in the last decade. Eur. J. Med. Chem. 2020, 209, 112861. [Google Scholar] [CrossRef] [PubMed]
  102. Mohammed, S.A.; Albiero, M.; Ambrosini, S.; Gorica, M.E.; Karsai, G.; Caravaggi, C.M.; Masi, S.; Camici, G.G.; Wenzl, F.A.; Calderone, V.; et al. The BET Protein Inhibitor Apabetalone Rescues Diabetes-Induced Impairment of Angiogenic Response by Epigenetic Regulation of Thrombospondin-1. Antioxidants Redox Signal. 2022, 36, 667–684. [Google Scholar] [CrossRef] [PubMed]
  103. Lan, F.; Hu, Y.; Tang, D.; Cai, J.; Zhang, Q. Transcription coactivator p300 promotes inflammation by enhancing p65 subunit activation in type 2 diabetes nephropathy. Int. J. Clin. Exp. Pathol. 2019, 12, 1826–1834. [Google Scholar] [PubMed]
  104. Qiu, Y.; Zhao, Y.; Becker, M.; John, S.; Parekh, B.S.; Huang, S.; Hendarwanto, A.; Martinez, E.D.; Chen, Y.; Lu, H.; et al. HDAC1 Acetylation Is Linked to Progressive Modulation of Steroid Receptor-Induced Gene Transcription. Mol. Cell 2006, 22, 669–679. [Google Scholar] [CrossRef]
  105. Chen, L.-F.; Greene, W.C. Regulation of distinct biological activities of the NF-?B transcription factor complex by acetylation. J. Mol. Med. 2003, 81, 549–557. [Google Scholar] [CrossRef]
  106. Chiu, J.; Khan, Z.A.; Farhangkhoee, H.; Chakrabarti, S. Curcumin prevents diabetes-associated abnormalities in the kidneys by inhibiting p300 and nuclear factor-κB. Nutrition 2009, 25, 964–972. [Google Scholar] [CrossRef]
  107. Gerritsen, M.E.; Williams, A.J.; Neish, A.S.; Moore, S.; Shi, Y.; Collins, T. CREB-binding protein/p300 are transcriptional coactivators of p65. Proc. Natl. Acad. Sci. USA 1997, 94, 2927–2932. [Google Scholar] [CrossRef] [Green Version]
  108. Zhong, H.; May, M.J.; Jimi, E.; Ghosh, S. The Phosphorylation Status of Nuclear NF-ΚB Determines Its Association with CBP/p300 or HDAC-1. Mol. Cell 2002, 9, 625–636. [Google Scholar] [CrossRef]
  109. Giordano, A.; Avantaggiati, M.L. p300 and CBP: Partners for life and death. J. Cell Physiol. 1999, 181, 218–230. [Google Scholar] [CrossRef]
  110. Bedford, D.C.; Kasper, L.H.; Fukuyama, T.; Brindle, P.K. Target gene context influences the transcriptional requirement for the KAT3 family of CBP and p300 histone acetyltransferases. Epigenetics 2010, 5, 9–15. [Google Scholar] [CrossRef]
  111. Chen, F.; Li, X.; Aquadro, E.; Haigh, S.; Zhou, J.; Stepp, D.W.; Weintraub, N.L.; Barman, S.A.; Fulton, D.J. Inhibition of histone deacetylase reduces transcription of NADPH oxidases and ROS production and ameliorates pulmonary arterial hypertension. Free. Radic. Biol. Med. 2016, 99, 167–178. [Google Scholar] [CrossRef] [Green Version]
  112. McKinsey, T.A. Targeting Inflammation in Heart Failure with Histone Deacetylase Inhibitors. Mol. Med. 2011, 17, 434–441. [Google Scholar] [CrossRef] [PubMed]
  113. Ghosh, A.K. p300 in Cardiac Development and Accelerated Cardiac Aging. Aging Dis. 2020, 11, 916–926. [Google Scholar] [CrossRef] [PubMed]
  114. Shikama, N.; Lutz, W.; Kretzschmar, R.; Sauter, N.; Roth, J.; Marino, S.; Wittwer, J.; Scheidweiler, A.; Eckner, R. Essential function of p300 acetyltransferase activity in heart, lung and small intestine formation. EMBO J. 2003, 22, 5175–5185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Nakagawa, Y.; Kuwahara, K.; Takemura, G.; Akao, M.; Kato, M.; Arai, Y.; Takano, M.; Harada, M.; Murakami, M.; Nakanishi, M.; et al. p300 Plays a Critical Role in Maintaining Cardiac Mitochondrial Function and Cell Survival in Postnatal Hearts. Circ. Res. 2009, 105, 746–754. [Google Scholar] [CrossRef]
  116. Fukushima, A.; Lopaschuk, G.D. Acetylation control of cardiac fatty acid β-oxidation and energy metabolism in obesity, diabetes, and heart failure. Biochim. et Biophys. Acta (BBA)—Mol. Basis Dis. 2016, 1862, 2211–2220. [Google Scholar] [CrossRef]
  117. Kadiyala, C.S.R.; Zheng, L.; Du, Y.; Yohannes, E.; Kao, H.-Y.; Miyagi, M.; Kern, T.S. Acetylation of Retinal Histones in Diabetes Increases Inflammatory Proteins. J. Biol. Chem. 2012, 287, 25869–25880. [Google Scholar] [CrossRef] [Green Version]
  118. Kaisaki, P.J.; Otto, G.W.; McGouran, J.F.; Toubal, A.; Argoud, K.; Waller-Evans, H.; Finlay, C.; Caldérari, S.; Bihoreau, M.-T.; Kessler, B.M.; et al. Genetic Control of Differential Acetylation in Diabetic Rats. PLoS ONE 2014, 9, e94555. [Google Scholar] [CrossRef] [Green Version]
  119. Kumar, S.; Kim, Y.-R.; Vikram, A.; Naqvi, A.; Li, Q.; Kassan, M.; Kumar, V.; Bachschmid, M.M.; Jacobs, J.S.; Kumar, A.; et al. Sirtuin1-regulated lysine acetylation of p66Shc governs diabetes-induced vascular oxidative stress and endothelial dysfunction. Proc. Natl. Acad. Sci. USA 2017, 114, 1714–1719. [Google Scholar] [CrossRef] [Green Version]
  120. Yang, M.; Zhang, Y.; Ren, J. Acetylation in cardiovascular diseases: Molecular mechanisms and clinical implications. Biochim. et Biophys. Acta (BBA)—Mol. Basis Dis. 2020, 1866, 165836. [Google Scholar] [CrossRef]
  121. Ghosh, A.K.; Yuan, W.; Mori, Y.; Varga, J. Smad-dependent stimulation of type I collagen gene expression in human skin fibroblasts by TGF-β involves functional cooperation with p300/CBP transcriptional coactivators. Oncogene 2000, 19, 3546–3555. [Google Scholar] [CrossRef] [Green Version]
  122. Ghosh, A.K.; Quaggin, S.E.; Vaughan, D.E. Molecular basis of organ fibrosis: Potential therapeutic approaches. Exp. Biol. Med. 2013, 238, 461–481. [Google Scholar] [CrossRef]
  123. Rai, R.; Verma, S.K.; Kim, D.; Ramirez, V.; Lux, E.; Li, C.; Sahoo, S.; Wilsbacher, L.D.; Vaughan, D.E.; Quaggin, S.E.; et al. A novel acetyltransferase p300 inhibitor ameliorates hypertension-associated cardio-renal fibrosis. Epigenetics 2017, 12, 1004–1013. [Google Scholar] [CrossRef] [Green Version]
  124. Gusterson, R.J.; Jazrawi, E.; Adcock, I.M.; Latchman, D.S. The Transcriptional Co-activators CREB-binding Protein (CBP) and p300 Play a Critical Role in Cardiac Hypertrophy That Is Dependent on Their Histone Acetyltransferase Activity. J. Biol. Chem. 2003, 278, 6838–6847. [Google Scholar] [CrossRef] [Green Version]
  125. Peng, C.; Zhang, W.; Zhao, W.; Zhu, J.; Huang, X.; Tian, J. Alcohol-induced histone H3K9 hyperacetylation and cardiac hypertrophy are reversed by a histone acetylases inhibitor anacardic acid in developing murine hearts. Biochimie 2015, 113, 1–9. [Google Scholar] [CrossRef]
  126. Yanazume, T.; Hasegawa, K.; Morimoto, T.; Kawamura, T.; Wada, H.; Matsumori, A.; Kawase, Y.; Hirai, M.; Kita, T. Cardiac p300 Is Involved in Myocyte Growth with Decompensated Heart Failure. Mol. Cell. Biol. 2003, 23, 3593–3606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Rai, R.; Sun, T.; Ramirez, V.; Lux, E.; Eren, M.; Vaughan, D.E.; Ghosh, A.K. Acetyltransferase p300 inhibitor reverses hypertension-induced cardiac fibrosis. J. Cell. Mol. Med. 2019, 23, 3026–3031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Gan, L.; Pei, L. Epigenetic Regulation of Heart-ECHS. JACC: Basic Transl. Sci. 2022, 7, 363–365. [Google Scholar] [CrossRef] [PubMed]
  129. Reddy, M.A.; Sahar, S.; Villeneuve, L.M.; Lanting, L.; Natarajan, R. Role of Src Tyrosine Kinase in the Atherogenic Effects of the 12/15-Lipoxygenase Pathway in Vascular Smooth Muscle Cells. Arter. Thromb. Vasc. Biol. 2009, 29, 387–393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Carrillo-Sepulveda, M.A.; Maddie, N.; Johnson, C.M.; Burke, C.; Lutz, O.; Yakoub, B.; Kramer, B.; Persand, D. Vascular hyperacetylation is associated with vascular smooth muscle dysfunction in a rat model of non-obese type 2 diabetes. Mol. Med. 2022, 28, 30. [Google Scholar] [CrossRef]
  131. Costantino, S.; Mohammed, S.A.; Ambrosini, S.; Paneni, F. The vascular epigenome in patients with obesity and type 2 diabetes: Opportunities for personalized therapies. Vasc. Biol. 2020, 2, H19–H28. [Google Scholar] [CrossRef]
  132. Feng, B.; Chen, S.; Chiu, J.; George, B.; Chakrabarti, S. Regulation of cardiomyocyte hypertrophy in diabetes at the transcriptional level. Am. J. Physiol. Metab. 2008, 294, E1119–E1126. [Google Scholar] [CrossRef] [Green Version]
  133. Mortuza, R.; Chen, S.; Feng, B.; Sen, S.; Chakrabarti, S. High Glucose Induced Alteration of SIRTs in Endothelial Cells Causes Rapid Aging in a p300 and FOXO Regulated Pathway. PLoS ONE 2013, 8, e54514. [Google Scholar] [CrossRef]
  134. Duan, Y.; Zhou, B.; Su, H.; Liu, Y.; Du, C. miR-150 regulates high glucose-induced cardiomyocyte hypertrophy by targeting the transcriptional co-activator p300. Exp. Cell Res. 2013, 319, 173–184. [Google Scholar] [CrossRef]
  135. Bugyei-Twum, A.; Advani, A.; Advani, S.L.; Zhang, Y.; Thai, K.; Kelly, D.J.; Connelly, K.A. High glucose induces Smad activation via the transcriptional coregulator p300 and contributes to cardiac fibrosis and hypertrophy. Cardiovasc. Diabetol. 2014, 13, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Chen, S.; Khan, Z.A.; Cukiernik, M.; Chakrabarti, S. Differential activation of NF-κB and AP-1 in increased fibronectin synthesis in target organs of diabetic complications. Am. J. Physiol. Metab. 2003, 284, E1089–E1097. [Google Scholar] [CrossRef]
  137. Chen, S.; Feng, B.; George, B.; Chakrabarti, R.; Chen, M.; Chakrabarti, S. Transcriptional coactivator p300 regulates glucose-induced gene expression in endothelial cells. Am. J. Physiol. Metab. 2010, 298, E127–E137. [Google Scholar] [CrossRef] [Green Version]
  138. Mortuza, R.; Feng, B.; Chakrabarti, S. SIRT 1 reduction causes renal and retinal injury in diabetes through endothelin 1 and transforming growth factor β1. J. Cell. Mol. Med. 2015, 19, 1857–1867. [Google Scholar] [CrossRef]
  139. Poznyak, A.; Grechko, A.V.; Poggio, P.; Myasoedova, V.A.; Alfieri, V.; Orekhov, A.N. The Diabetes Mellitus–Atherosclerosis Connection: The Role of Lipid and Glucose Metabolism and Chronic Inflammation. Int. J. Mol. Sci. 2020, 21, 1835. [Google Scholar] [CrossRef] [Green Version]
  140. Lee, H.-M.; Kim, J.-J.; Kim, H.J.; Shong, M.; Ku, B.J.; Jo, E.-K. Upregulated NLRP3 inflammasome activation in patients with type 2 diabetes. Diabetes 2013, 62, 194–204. [Google Scholar] [CrossRef]
  141. Tabit, C.E.; Shenouda, S.M.; Holbrook, M.; Fetterman, J.L.; Kiani, S.; Frame, A.A.; Kluge, M.A.; Held, A.; Dohadwala, M.M.; Gokce, N.; et al. Protein kinase C-beta contributes to impaired endothelial insulin signaling in humans with diabetes mellitus. Circulation 2013, 127, 86–95. [Google Scholar] [CrossRef]
  142. Liu, D.; Perkins, J.T.; Hennig, B. EGCG prevents PCB-126-induced endothelial cell inflammation via epigenetic modifications of NF-κB target genes in human endothelial cells. J. Nutr. Biochem. 2015, 28, 164–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Mezzano, S.; Aros, C.; Droguett, A.; Burgos, M.E.; Ardiles, L.; Flores, C.; Schneider, H.; Ruiz-Ortega, M.; Egido, J. NF- B activation and overexpression of regulated genes in human diabetic nephropathy. Nephrol. Dial. Transplant. 2004, 19, 2505–2512. [Google Scholar] [CrossRef] [Green Version]
  144. Wang, Y.; Wang, Y.; Luo, M.; Wu, H.; Kong, L.; Xin, Y.; Cui, W.; Zhao, Y.; Wang, J.; Liang, G.; et al. Novel curcumin analog C66 prevents diabetic nephropathy via JNK pathway with the involvement of p300/CBP-mediated histone acetylation. Biochim. et Biophys. Acta (BBA)—Mol. Basis Dis. 2015, 1852, 34–46. [Google Scholar] [CrossRef] [Green Version]
  145. Morinobu, A.; Kanno, Y.; O’Shea, J.J. Discrete Roles for Histone Acetylation in Human T Helper 1 Cell-specific Gene Expression. J. Biol. Chem. 2004, 279, 40640–40646. [Google Scholar] [CrossRef] [Green Version]
  146. Yang, S.-R.; Valvo, S.; Yao, H.; Kode, A.; Rajendrasozhan, S.; Edirisinghe, I.; Caito, S.; Adenuga, D.; Henry, R.; Fromm, G.; et al. IKKα Causes Chromatin Modification on Pro-Inflammatory Genes by Cigarette Smoke in Mouse Lung. Am. J. Respir. Cell Mol. Biol. 2008, 38, 689–698. [Google Scholar] [CrossRef] [Green Version]
  147. Gupta, P.; Sharma, G.; Lahiri, A.; Barthwal, M.K. FOXO3a acetylation regulates PINK1, mitophagy, inflammasome activation in murine palmitate-conditioned and diabetic macrophages. J. Leukoc. Biol. 2021, 111, 611–627. [Google Scholar] [CrossRef]
  148. Yaribeygi, H.; Sathyapalan, T.; Atkin, S.L.; Sahebkar, A. Molecular Mechanisms Linking Oxidative Stress and Diabetes Mellitus. Oxidative Med. Cell. Longev. 2020, 2020, 8609213. [Google Scholar] [CrossRef] [Green Version]
  149. Cyr, A.R.; Huckaby, L.V.; Shiva, S.S.; Zuckerbraun, B.S. Nitric Oxide and Endothelial Dysfunction. Crit. Care Clin. 2020, 36, 307–321. [Google Scholar] [CrossRef]
  150. Meza, C.A.; La Favor, J.D.; Kim, D.-H.; Hickner, R.C. Endothelial Dysfunction: Is There a Hyperglycemia-Induced Imbalance of NOX and NOS? Int. J. Mol. Sci. 2019, 20, 3775. [Google Scholar] [CrossRef]
  151. Fish, J.E.; Matouk, C.C.; Rachlis, A.; Lin, S.; Tai, S.C.; D’Abreo, C.; Marsden, P.A. The Expression of Endothelial Nitric-oxide Synthase Is Controlled by a Cell-specific Histone Code. J. Biol. Chem. 2005, 280, 24824–24838. [Google Scholar] [CrossRef] [Green Version]
  152. Chen, W.; Bacanamwo, M.; Harrison, D.G. Activation of p300 Histone Acetyltransferase Activity Is an Early Endothelial Response to Laminar Shear Stress and Is Essential for Stimulation of Endothelial Nitric-oxide Synthase mRNA Transcription. J. Biol. Chem. 2008, 283, 16293–16298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Lazar, A.-G.; Vlad, M.-L.; Manea, A.; Simionescu, M.; Manea, S.-A. Activated Histone Acetyltransferase p300/CBP-Related Signalling Pathways Mediate Up-Regulation of NADPH Oxidase, Inflammation, and Fibrosis in Diabetic Kidney. Antioxidants 2021, 10, 1356. [Google Scholar] [CrossRef]
  154. Karbasforooshan, H.; Karimi, G. The role of SIRT1 in diabetic cardiomyopathy. Biomed. Pharmacother. 2017, 90, 386–392. [Google Scholar] [CrossRef]
  155. Koka, S.; Aluri, H.S.; Xi, L.; Lesnefsky, E.J.; Kukreja, R.C.; Pinti, M.V.; Fink, G.K.; Hathaway, Q.A.; Durr, A.J.; Kunovac, A.; et al. Chronic inhibition of phosphodiesterase 5 with tadalafil attenuates mitochondrial dysfunction in type 2 diabetic hearts: Potential role of NO/SIRT1/PGC-1α signaling. Am. J. Physiol. Circ. Physiol. 2014, 306, H1558–H1568. [Google Scholar] [CrossRef] [Green Version]
  156. Ding, M.; Lei, J.; Han, H.; Li, W.; Qu, Y.; Fu, E.; Fu, F.; Wang, X. SIRT1 protects against myocardial ischemia–reperfusion injury via activating eNOS in diabetic rats. Cardiovasc. Diabetol. 2015, 14, 143. [Google Scholar] [CrossRef] [Green Version]
  157. Manea, S.-A.; Antonescu, M.-L.; Fenyo, I.M.; Raicu, M.; Simionescu, M.; Manea, A. Epigenetic regulation of vascular NADPH oxidase expression and reactive oxygen species production by histone deacetylase-dependent mechanisms in experimental diabetes. Redox Biol. 2018, 16, 332–343. [Google Scholar] [CrossRef]
  158. Sommese, L.; Zullo, A.; Mancini, F.P.; Fabbricini, R.; Soricelli, A.; Napoli, C. Clinical relevance of epigenetics in the onset and management of type 2 diabetes mellitus. Epigenetics 2017, 12, 401–415. [Google Scholar] [CrossRef]
  159. Napoli, C.; Benincasa, G.; Schiano, C.; Salvatore, M. Differential epigenetic factors in the prediction of cardiovascular risk in diabetic patients. Eur. Hear. J.—Cardiovasc. Pharmacother. 2019, 6, 239–247. [Google Scholar] [CrossRef] [Green Version]
  160. Bridgeman, S.C.; Ellison, G.C.; Melton, P.E.; Newsholme, P.; Mamotte, C.D.S. Epigenetic effects of metformin: From molecular mechanisms to clinical implications. Diabetes Obes. Metab. 2018, 20, 1553–1562. [Google Scholar] [CrossRef]
  161. Sunagawa, Y.; Shimizu, K.; Katayama, A.; Funamoto, M.; Shimizu, K.; Nurmila, S.; Shimizu, S.; Miyazaki, Y.; Katanasaka, Y.; Hasegawa, K.; et al. Metformin suppresses phenylephrine-induced hypertrophic responses by inhibiting p300-HAT activity in cardiomyocytes. J. Pharmacol. Sci. 2021, 147, 169–175. [Google Scholar] [CrossRef]
  162. Ghasemi, S. Cancer’s epigenetic drugs: Where are they in the cancer medicines? Pharm. J. 2020, 20, 367–379. [Google Scholar] [CrossRef]
  163. Li, Y.; Xu, B.; Yang, J.; Wang, L.; Tan, X.; Hu, X.; Sun, L.; Chen, S.; Zhu, L.; Chen, X.; et al. Liraglutide protects against lethal renal ischemia-reperfusion injury by inhibiting high-mobility group box 1 nuclear-cytoplasmic translocation and release. Pharmacol. Res. 2021, 173, 105867. [Google Scholar] [CrossRef]
  164. Shimazu, T.; Hirschey, M.D.; Newman, J.; He, W.; Shirakawa, K.; Le Moan, N.; Grueter, C.A.; Lim, H.; Saunders, L.R.; Stevens, R.D.; et al. Suppression of Oxidative Stress by β-Hydroxybutyrate, an Endogenous Histone Deacetylase Inhibitor. Science 2013, 339, 211–214. [Google Scholar] [CrossRef] [Green Version]
  165. Solini, A.; Seghieri, M.; Giannini, L.; Biancalana, E.; Parolini, F.; Rossi, C.; Dardano, A.; Taddei, S.; Ghiadoni, L.; Bruno, R.M. The Effects of Dapagliflozin on Systemic and Renal Vascular Function Display an Epigenetic Signature. J. Clin. Endocrinol. Metab. 2019, 104, 4253–4263. [Google Scholar] [CrossRef]
  166. Balasubramanyam, K.; Altaf, M.; Varier, R.A.; Swaminathan, V.; Ravindran, A.; Sadhale, P.P.; Kundu, T.K. Polyisoprenylated Benzophenone, Garcinol, a Natural Histone Acetyltransferase Inhibitor, Represses Chromatin Transcription and Alters Global Gene Expression. J. Biol. Chem. 2004, 279, 33716–33726. [Google Scholar] [CrossRef] [Green Version]
  167. Zhang, D.-W.; Fu, M.; Gao, S.-H.; Liu, J.-L. Curcumin and Diabetes: A Systematic Review. Evid. Based Complement. Altern. Med. 2013, 2013, 636053. [Google Scholar] [CrossRef] [Green Version]
  168. Yun, J.-M.; Jialal, I.; Devaraj, S. Epigenetic regulation of high glucose-induced proinflammatory cytokine production in monocytes by curcumin. J. Nutr. Biochem. 2011, 22, 450–458. [Google Scholar] [CrossRef] [Green Version]
  169. Soetikno, V.; Sari, F.R.; Sukumaran, V.; Lakshmanan, A.P.; Mito, S.; Harima, M.; Thandavarayan, R.A.; Suzuki, K.; Nagata, M.; Takagi, R.; et al. Curcumin prevents diabetic cardiomyopathy in streptozotocin-induced diabetic rats: Possible involvement of PKC–MAPK signaling pathway. Eur. J. Pharm. Sci. 2012, 47, 604–614. [Google Scholar] [CrossRef]
Figure 1. Endothelial cell senescence: Senescence is triggered by various stimuli, including changes in telomeric structure, oxidative stress, mitochondrial dysfunction, inflammation and DNA damage, which contribute to cell cycle arrest through the activation of the p53/p21CIP1 or p16INK4A/hosphor-Rb tumour suppressor pathways. In addition, senescent cells exhibit activation of the senescence-associated secretory phenotype (SASP) characterised by the increased lysosomal activity and accumulation of β-galactosidase. The SASP leads to the upregulation and release of growth factors, cytokines and proteases that can exert detrimental effects.
Figure 1. Endothelial cell senescence: Senescence is triggered by various stimuli, including changes in telomeric structure, oxidative stress, mitochondrial dysfunction, inflammation and DNA damage, which contribute to cell cycle arrest through the activation of the p53/p21CIP1 or p16INK4A/hosphor-Rb tumour suppressor pathways. In addition, senescent cells exhibit activation of the senescence-associated secretory phenotype (SASP) characterised by the increased lysosomal activity and accumulation of β-galactosidase. The SASP leads to the upregulation and release of growth factors, cytokines and proteases that can exert detrimental effects.
Cells 12 00431 g001
Figure 2. p300 mechanisms underlying vascular inflammation and oxidative stress and the role of some HAT inhibitors.
Figure 2. p300 mechanisms underlying vascular inflammation and oxidative stress and the role of some HAT inhibitors.
Cells 12 00431 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Di Pietrantonio, N.; Di Tomo, P.; Mandatori, D.; Formoso, G.; Pandolfi, A. Diabetes and Its Cardiovascular Complications: Potential Role of the Acetyltransferase p300. Cells 2023, 12, 431. https://doi.org/10.3390/cells12030431

AMA Style

Di Pietrantonio N, Di Tomo P, Mandatori D, Formoso G, Pandolfi A. Diabetes and Its Cardiovascular Complications: Potential Role of the Acetyltransferase p300. Cells. 2023; 12(3):431. https://doi.org/10.3390/cells12030431

Chicago/Turabian Style

Di Pietrantonio, Nadia, Pamela Di Tomo, Domitilla Mandatori, Gloria Formoso, and Assunta Pandolfi. 2023. "Diabetes and Its Cardiovascular Complications: Potential Role of the Acetyltransferase p300" Cells 12, no. 3: 431. https://doi.org/10.3390/cells12030431

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop