Next Article in Journal
What Do Single-Cell Models Already Know About Perturbations?
Previous Article in Journal
Role of Genetic and Epigenetic Biomarkers in Treatment-Resistant Depression: A Literature Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

From Metabolic to Epigenetic Memory: The Impact of Hyperglycemia-Induced Epigenetic Signature on Kidney Disease Progression and Complications

by
Sara Cannito
1,*,
Ida Giardino
1,
Maria D’Apolito
2,
Alessandra Ranaldi
2,
Francesca Scaltrito
3,
Massimo Pettoello-Mantovani
4,5,6 and
Annamaria Piscazzi
1
1
Laboratory Medicine, Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
2
Medical Genetics, Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
3
Residency Course in Pediatrics, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
4
Italian Academy of Pediatrics, 20126 Milan, Italy
5
Department of Pediatrics, Institute for Scientific Research «Casa Sollievo», University of Foggia, 71122 Foggia, Italy
6
European Pediatric Association, Union of National European Pediatric Societies and Associations, 10115 Berlin, Germany
*
Author to whom correspondence should be addressed.
Genes 2025, 16(12), 1442; https://doi.org/10.3390/genes16121442
Submission received: 20 October 2025 / Revised: 8 November 2025 / Accepted: 10 November 2025 / Published: 2 December 2025
(This article belongs to the Section Epigenomics)

Abstract

Chronic kidney disease is a significant global health burden and a leading cause of cardiovascular morbidity and mortality. Diabetes mellitus is the primary cause of kidney disease, driving the progression of both micro- and macrovascular complications. Sustained hyperglycemia initiates a cascade of deleterious molecular and cellular events, including mitochondrial dysfunction, inflammation, oxidative stress, and dysregulated apoptosis and autophagy, which collectively contribute to the progression of renal injury. Beyond these well-established mechanisms, a compelling body of evidence highlights the pivotal role of epigenetic alterations (such as DNA methylation, histone post-translational modifications, and non-coding RNAs) in mediated long-term kidney damage. The interplay between transcriptional and epigenetic regulation underlies the phenomenon of the “metabolic memory”, wherein cellular dysfunction persists even after glycemic control is achieved. This review synthesizes the current knowledge on mechanisms sustaining metabolic and epigenetic memory, with a particular focus on the epigenetic machinery that establishes and maintains these signals, a concept increasingly termed “epigenetic memory.” Given their reversible nature, epigenetic determinants are emerging as promising biomarkers and a compelling therapeutic avenue. Targeting these “epifactors” offers a novel strategy to halt progression to end-stage renal disease, thereby paving the way for precision medicine approaches in diabetes-related renal disease.

1. Introduction

Kidney diseases, encompassing both acute kidney injury (AKI) and chronic kidney disease (CKD), represent a major global health burden and recognized as leading contributors to cardiovascular morbidity and mortality [1,2]. CKD is frequently associated with a higher incidence of comorbid conditions, such as hypertension, diabetes mellitus, glomerulonephritis, chronic pyelonephritis, autoimmune diseases, and recurrent urinary tract infections, whose progression is associated with the development of renal impairment and eventual kidney failure [1,3]. Globally, in 2017, the Global Burden Disease, Injuries, and Risk Factors (GBD) study showed that impaired fasting plasma glucose and high glucose pressure account for 57.6% and 43.2% of the age-standardized rate of CKD, respectively [4]. Both AKI and CKD arise from complex systemic interactions and cellular disturbances, involving hemodynamic alterations, inflammatory responses, and maladaptive repair mechanisms that perpetuate the renal injury. CKD progression, in particular, is driven by endothelial dysfunction, vascular rarefaction, inflammation, and progressive nephron loss [5]. Recent guideline frameworks have reinterpreted CKD as a highly prevalent condition with a continuum of severity, no longer viewed solely as a specialist disease but as a public health priority requiring early detection, preventive strategies, and multidisciplinary management [6]. According to the Kidney Disease Outcomes Quality Initiative (KDOQI) and Kidney Disease: Improving Global Outcomes (KDIGO) guidelines, CKD is defined by the presence of kidney damage or a reduction in glomerular filtration rate (GFR) for at least three months, regardless of etiology. Decreased kidney function is typically assessed through estimated GFR (eGFR), derived from serum creatinine–based equations. CKD often progresses silently, with patients remaining asymptomatic until substantial functional decline occurs. In a subset of individuals, disease progression culminates in end-stage renal disease (ESRD), characterized by severely reduced kidney function or dependence on dialysis. Despite advances in therapeutic strategies, the coexistence of diabetes remains a critical determinant of adverse cardiovascular and renal outcomes. Diabetic kidney disease (DKD) is a major microvascular complication of both type 1 and type 2 diabetes and a leading cause of ESRD worldwide [7]. DKD is characterized by a constellation of structural and functional changes, including podocyte effacement, mesangial expansion, excessive deposition of extracellular matrix (ECM), and tubular epithelial-to-mesenchymal transition (EMT) [8]. Alongside hypertension, sustained high glucose levels are the principal pathogenic driver of DKD [9]. Chronic hyperglycemia, orchestrates a complex and multifactorial cascade involving mitochondrial dysfunction, oxidative stress, inflammation, and dysregulated apoptosis and autophagy—processes that collectively promote cellular injury and fibrosis [10,11,12]. While the classical biochemical pathways underlying diabetic nephropathy have been extensively characterized, emerging evidence highlights the pivotal role of epigenetic mechanisms—including DNA methylation, histone post-translational modifications, and non-coding RNAs—in initiating and perpetuating renal dysfunction and its complications [13]. Epigenetic dysregulation affects multiple renal cell types, including mesangial cells, podocytes, tubular epithelial cells, and glomerular endothelial cells [14]. These alterations, which do not change the DNA sequence itself, modify gene expression patterns, leading to cellular damage and fibrosis, typical hallmarks of DKD. These environmentally responsive mechanisms may mediate the sustained expression of genes and phenotypes associated with DKD. These alterations, which do not modify the DNA sequence itself, alter gene expression patterns, driving cellular damage and fibrotic remodeling—hallmarks of DKD. Importantly, these environmentally responsive mechanisms may sustain the expression of pathogenic genes and phenotypes associated with DKD even after restoration of normoglycemia. The interplay between transcriptional and epigenetic factors underlies the phenomenon known as “metabolic memory,” a term coined by Michael Brownlee to describe the long-term deleterious effects of prior hyperglycemic exposure that persist despite subsequent glycemic control [15].
This review aims to summarize the current understanding of the molecular bases underlining the metabolic memory, emphasizing on epigenetic machinery mechanisms responsible for its establishment and maintenance—collectively referred to as “epigenetic memory”. Given the reversible nature of epigenetic modifications, “epifactors” hold promise as both biomarkers and therapeutic targets for hyperglycemia-related CKD. Modulating epigenetic regulators may open new avenues for halting disease progression, preventing ESRD, and advancing precision medicine approaches in DKD.

2. The Metabolic Memory

There is a complex interplay of molecular pathways activated by chronic hyperglycemia during the progression of diabetic kidney disease (DKD). Sustained exposure to elevated glucose levels triggers a cascade of intricate and deleterious molecular and cellular events that ultimately culminate in widespread cellular and tissue injury. The detrimental effects of glucotoxicity are mediated through multiple, interconnected mechanisms, including the accumulation of advanced glycation end products (AGEs), excessive generation of reactive oxygen species (ROS), and profound bioenergetic dysfunctions. These processes ultimately lead to epigenetic remodeling and the establishment of inflammatory memory [16] (Figure 1).
Collectively, these ROS-associated pathways contribute to glomerular cell dysfunction, renal inflammation, and fibrosis by promoting DNA damage, mitochondrial impairment, lipid peroxidation, and abnormal protein modifications [17].
Chronic hyperglycemia accelerates the formation of AGEs, which are closely linked to cardiovascular and microvascular complications [15,18]. The AGEs binding to their receptor (RAGE) activates multiple intracellular signaling pathways—including MAPK, ERKs, JNK, NF-κB, TGF-α, and NOX-1—which amplify oxidative stress, inflammation, and apoptosis [18,19,20,21]. Interestingly, transient or prolonged hyperglycemic insults can leave a persistent molecular fingerprint that continues to drive diabetic complications, like DKD, even after the normoglycemia is restored [22,23,24,25,26]. The first experimental evidence supporting this phenomenon came in vitro studies demonstrating self-perpetuating changes in the gene expression induced by transient hyperglycemia. Human endothelial cells exposed to high glucose for 14 days, followed by a 7-day normoglycemic period, continued to overexpress fibronectin mRNA—an important mediator of diabetic retinopathy—for several weeks after glucose normalization [27]. These observations early mechanistic insight into what is also known as “legacy effect” or “glycemic memory”.

2.1. Clinical Evidence from Landmark Trials

The concept of metabolic memory is strongly supported by longitudinal follow-up data from two landmark clinical trials: the Diabetes Control and Complications Trial (DCCT) and the UK Prospective Diabetes Study (UKPDS). In the DCCT, patients with type 1 diabetes who received intensive glucose control exhibited a significant reduction in microvascular complications compared with those receiving conventional therapy. Remarkably, during the subsequent Epidemiology of Diabetes Interventions and Complications (EDIC) follow-up study, glycemic levels between the two groups converged. Crucially, however, the initial benefit of intensive therapy on reducing the risk of microvascular complications (retinopathy, nephropathy, and neuropathy), not only persisted, but often widened over the subsequent two decades [28]. A similar effect was observed in the follow-up of the UKPDS cohort of type 2 diabetic patients. The early intensive glucose management conferred a lasting reduction in myocardial infarction and all-cause mortality, even after glycemic differences dissipated. Together, these findings demonstrated that early and tight glycemic control establishes a durable protective imprint, underscoring the need for early intervention to prevent the establishment of this pathogenic memory [28].

2.2. Oxidative Stress and Mitochondrial Dysfunction

From an experimental standpoint, memory reflects a sustained vascular stress signature characterized by increased ROS production, endothelial dysfunction, endoplasmic reticulum stress, and persistent activation of pro-atherogenic pathways [29]. Extensive evidence links oxidative stress and diabetic complications across multiple tissues [15,30,31]. Among the various sources of ROS, mitochondrial-derived ROS (mtROS) play a particularly critical role in diabetic vascular and renal pathology [24,32,33,34]. mtROS influence endothelial function, nitric oxide bioavailability, and redox-sensitive signaling cascades [34,35]. Ceriello and colleagues demonstrated that metabolic memory originates at the level of glycated mitochondrial proteins, further involving non-enzymatic glycation of cellular proteins, lipids, and nucleic acids [22]. Consistent with these observations, inhibition of oxidative stress through antioxidant therapy has been shown to attenuate diabetic complications [15,30,32]. Normalization of mtROS levels alleviates key hyperglycemia-induced pathways, including the polyol pathway, protein kinase C (PKC) activation, and AGEs accumulation [32]. Consequently, reducing mtROS production can “erase” the hyperglycemia-induced metabolic imprint, confirming the pivotal role of mitochondrial dysfunction in metabolic memory [23].
Glycation of mitochondrial components can trigger a deleterious cycle of mitochondrial DNA (mtDNA) damage and bioenergetic decline, perpetuating ROS overproduction and cellular injury [36]. Kowluru and colleagues reported that hyperglycemia alters mitochondrial dynamics, reducing the fusion protein Mitofusin 2 (Mfn2) and increasing the fission protein Dynamin-Related Protein 1 (Drp1), resulting in mitochondrial fragmentation. Restoration of normoglycemia failed to reverse these alterations, suggesting the persistence of dysfunctional mitochondria. Therapeutic modulation of mitochondrial fusion has been proposed as a means to restore mitochondrial quality control after high-glucose exposure [37].
Compromised mitochondrial integrity promotes cellular senescence and impairs mitophagy, as shown in retinal endothelial cells in diabetic retinopathy models [37,38,39]. Given the kidney’s high energy demand, similar mechanisms are pivotal in CKD. Chronic oxidative and nephrotoxic insults disrupt mitochondrial biogenesis, dynamics, and clearance, driving progressive renal dysfunction [40]. Moreover, mitochondrial impairment contributes to systemic metabolic disturbances, including inflammation, endothelial dysfunction, and insulin resistance [41,42,43]. Targeting mitochondrial quality control and metabolic memory regulators represents a promising therapeutic approach for limiting CKD progression and related complications [26,44,45]. Pharmacological agents such as mitochondrial division inhibitor 1 (Mdivi-1) and leflunomide, have shown efficacy in preserving mitochondrial structure and function in endothelial cells [37,46]. Notably, leflunomide administered during the hyperglycemia prevented mitochondrial damage, while post-hyperglycemia treatment restored mitochondrial dynamics, reactivated the mitophagy machinery, reduced ROS production, and inhibited apoptosis [37].

2.3. Impact of Glucose Fluctuations

An additional aspect of metabolic memory relates to glycemic variability. Intermittent glucose spikes can exert more deleterious effects than sustained hyperglycemia. Several studies have demonstrated that glucose oscillations elicit greater oxidative stress and endothelial dysfunction than sustained hyperglycemia, and that these detrimental effects persist even after glucose levels return to normal. Such intermittent glucose fluctuations promote a long-lasting pro-inflammatory and dysfunctional endothelial phenotype, driven by exacerbated oxidative stress and persistent epigenetic reprogramming [23,47]. Clinically, high glycemic variability is now recognized as an independent predictor of diabetes-related complications and mortality, beyond glycated haemoglobin (HbA1c) levels [48]. Experimental evidence indicates that transient hyperglycemia can induce epigenetic modifications of histone tails (e.g., methylation and acetylation) that persist after return to normoglycemia [49,50]. These stable epigenetic marks are increasingly recognized as key mediators of long-lasting detrimental effects of hyperglycemia in CKD pathophysiology and in the transition from AKI to CKD [51,52,53].

2.4. Therapeutic Implications

Understanding the mechanisms governing metabolic memory underscores the clinical importance of early, intensive glycemic control to rapidly restore metabolic homeostasis. Complementary strategies attenuating oxidative stress and inhibiting non-enzymatic glycation may further enhance long-term outcomes. Integrating such approach could form the foundation of more effective interventions aimed at preventing the long-term sequelae of hyperglycemia and limiting the progression of DKD.

3. Epigenetic Regulation of the Gene Expression

Understanding the hierarchical organization of DNA within chromatin provides the fundamental basis for elucidating the mechanisms of epigenetic regulation. Epigenetic mechanisms refer to potentially reversible and heritable modifications in chromatin structure and gene expression that drive tissue-specific transcription programs and maintain cellular identity, without altering the underlying DNA sequence [54]. Through coordinated biochemical modifications of DNA and histone or non-histone proteins, together with the action of chromatin remodelers and regulatory complexes, specific genes can be switched “on” or “off”, in response to environmental and cellular signals.

3.1. Chromatin Structure and DNA Methylation

Histone proteins constitute the core components of chromatin, whose fundamental unit is the nucleosome—147 base pairs of DNA wrapped around a histone octamer, consisting of two copies for each H2A, H2B, H3, and H4, while H1 serves as a linker histone (Figure 2A). In higher eukaryotes, principal epigenetic modifications include DNA methylation, post-translational modifications (PTMs), chromatin remodeling, histone variant exchange, and regulation by non-coding RNAs.
Among these, DNA methylation is the most extensively studied mechanism. It involves the covalent addition of a methyl group (-CH3) to the 5′ carbon of cytosine residues, generating 5-methylcytosines (5mC), primarily within CpG islands—short DNA regions (<500 bp) with high cytosine-guanosine content (>55%), commonly clustered around gene promoters (Figure 2B). This modification is catalyzed by DNA methyltransferases (DNMTs), which establish and maintain methylation patterns critical for normal tissues development and differentiation [55,56] (Figure 2B).
DNMTs are classified into two main functional groups: those responsible for de novo methylation (DNMT3A, DNMT3B) and those that maintain methylation during DNA replication (DNMT1). DNMT3L, although catalytically inactive, acts as a regulatory cofactor [57,58,59]. All of these enzymes catalyze the reaction through the s-adenosyl-l-methionine (SAM) as methyl groups donor. Interestingly, DNMT expression is tissue-specific; for instance, DNMT3A primarily regulates hematopoietic differentiation, whereas DNMT3B contributes to cartilage homeostasis and ossification [60]. Understanding the context-dependent functions of DNMTs is crucial for elucidating their role in disease pathogenesis and exploring therapeutic targeting strategies.
DNA methylation marks are reversible through passive demethylation (failure to maintain methylation during replication) or active demethylation mediated by Ten-Eleven Translocation (TET) enzyme family (TET1, TET2, and TET3). These enzymes iteratively oxidize 5mC to 5-hydroxymethylcytosine and further intermediates, which are then excised and removed via the base excision repair pathway, restoring the unmethylated state [61,62,63] (Figure 2B).

3.2. Histone Post-Translational Modifications

Histone PTMs represent another essential layer of epigenetic regulation, controlling chromatin accessibility and thereby influencing transcriptional activity. The amino-terminal tails of histones are subject to diverse PTMs, including acetylation, methylation, phosphorylation, ubiquitylation, SUMOylation, crotonylation, lactylation, and others [64,65,66,67]. These modifications can either promote chromatin relaxation and gene activation or induce compaction and transcriptional repression (Figure 2C). For example, acetylation of lysine residues neutralized their positive charge, reducing the electrostatic interaction between histones and DNA, thereby loosening chromatin and facilitating transcription. Conversely, removal of acetyl groups enhances chromatin condensation into transcriptionally inactive heterochromatin [68].
Histone methylation is another key modification that can either activate or repress gene expression, depending on the residue and degree of methylation. Lysine methylation is catalyzed by lysine methyltransferases (KMTs) and reversed by lysine demethylases (KDMs), which have been implicated in neurological disorders, inflammation, cancer, and metabolic disease [69,70,71]. Canonical methylation sites include H3K4, H3K9, H3K27, H3K36, H3K79, and H4K20, each carrying out distinct regulatory effects on chromatin structure and transcription [72].

3.3. Polycomb Repressive Complexes and Chromatin Remodeling

The stable maintenance of repressive chromatin states during development relies on recruitment of silencing complexes, such as methyl-CpG-binding proteins (e.g., MeCP2) or the Polycomb Repressive Complex 2 (PRC2) [73,74]. PRC2 catalyzes mono-, di-, and trimethylation of histone H3 at lysine 27 (H3K27me1/2/3), promoting chromatin compaction and transcriptional repression. Although the mechanisms governing PRC2 recruitment of specific genomic loci remain incompletely understood, its function is central to the propagation of silenced gene states [74,75].
Methylated histone residues are recognized by chromodomain-containing proteins, which translate histone marks into functional outcomes by assembling transcriptional activators or repressor [76]. For instance, the Polycomb complex is recognized by CBX chromodomain, leading to the monoubiquitination of histone H2A at lysine 119 (H2AK119ub) [77].

3.4. Histone Acetylation and Deacetylation

Histone acetylation is dynamically regulated by histone acetyl-transferases (HATs) and histone deacetylases (HDACs). HATs catalyze the transfer of an acetyl group (CH3CO–) from acetyl-CoA to the ε-amino group of lysine residues, a process recognized by bromodomain-containing proteins that facilitate transcriptional activation. Conversely, HDACs remove the acetyl groups, restoring positive charges on histones and promoting chromatin condensation and transcriptional repression [78].
Eighteen HDACs have been identified in humans, classified into four groups:
  • Class I (HDAC1, 2, 3, 8);
  • Class II (HDAC4, 5, 6, 7, 9, 10);
  • Class III or Sirtuins (SIRT1-7);
  • Class IV (HDAC11).
While classes I, II, and IV share structural and functional similarities, Sirtuins employ a unique NAD+-dependent deacetylation mechanism, producing nicotinamide and 2′-O-acetyl-ADP-ribose as metabolic products [78] (Table 1).

3.5. Interplay Between Epigenetic Mechanisms

Although mediated by distinct enzymatic machinery, DNA methylation and histone PTMs are functionally interconnected. Histone modifications have been shown to induce DNA methylation, a process especially observed during the early development also able to stabilize DNMTs [79,80]. Once established, methylated CpGs recruit methyl-CpG-binding proteins, such as MeCP2 and associated histone-modifying enzymes such as HDACs and KMTs, forming co-repressor complexes that reinforce gene silencing [79,81]. Moreover, the establishment of specific histone modifications can require rearrangements. For instance, before PRC2 can methylate H3K27, any pre-existing acetylation at this residue must be removed; this requires HDACs, which render the lysine’s group accessible for subsequent PRC2-mediated methylation, highlighting the dynamic interplay among these modifications [82].

3.6. Non-Coding RNAs and microRNA-Mediated Regulation

Non-coding RNAs, particularly microRNAs (miRNAs), have emerged as pivotal epigenetic regulators and biomarkers with significant diagnostic and prognostic potential across a variety of diseases [40,83]. For instance, several circulating miRNAs are dysregulated in the blood of cancer patients’, serving as potential biomarkers for early cancer diagnosis [40]. Given their established role in carcinogenesis, panels of miRNAs have been proposed as valuable tools for cancer screening and classification. For example, a specific five-miRNA signature—comprising miR-486-5p, miR-451, miR-92a, miR-25, and miR-16—has been identified for the early detection of gastric cancer [40].
Beyond their role in oncology, the prognostic and diagnostic relevance of miRNAs is increasingly recognized in a wide spectrum of other pathological conditions, including renal and metabolic diseases [84]. Altered expression profiles of circulating miRNAs are emerging as promising non-invasive biomarkers for predicting the progression of CKD and assessing the prognosis of metabolic disorders such as diabetes and its associated complications, including diabetic nephropathy and cardiovascular disease [85,86].
At the molecular level, miRNAs are small (18–25 nucleotide), endogenous, single-stranded RNA molecules that act as post-transcriptional regulators of gene expression. They originate from hairpin-loop precursors that undergo a series of tightly regulated processing steps. Most miRNAs are transcribed by RNA polymerase II as long primary transcripts (pri-miRNAs), which are cleaved in the nucleus by the Drosha–Pasha (DGCR8) complex to produce precursor miRNAs (pre-miRNAs). These pre-miRNAs are subsequently exported to the cytoplasm, where the RNase III enzyme Dicer further processes them into short miRNA duplexes. One strand of this duplex, known as the guide strand, is incorporated into the Argonaute-containing RNA-induced silencing complex (RISC). Through this complex, miRNAs exert their regulatory function by binding to complementary sequences in target mRNAs, leading to either mRNA degradation or translational repression, depending on the degree of sequence complementarity [87].

3.7. Epigenetics and Disease Implications

Aberrant epigenetic regulation contributes to multiple pathological conditions, including cancer, fibrosis, autoimmune disorders, inflammatory diseases, and metabolic disorders [88,89]. Persistent epigenetic marks can sustain maladaptive gene expression long after the initial trigger, contributing to the progressive nature of CKD and other metabolic complications. Importantly, the reversible nature of epigenetic modifications offers promising opportunities for therapeutic intervention and prognostic innovation, particularly in preventing or mitigating the metabolic memory phenomenon in diabetic and renal disease.

4. The Role of the Epigenetic Memory in Hyperglycemia-Related CKD Progression

Epigenetics serves as a crucial mediator between genetic predisposition and environmental or lifestyle factors—such as diet, exercise, oxidative stress, toxins, metabolic alterations, and inflammation—thereby influencing disease onset and progression. In the context of renal disease, oxidative stress, inflammation, and metabolic dysfunction are tightly linked to extensive epigenetic remodeling, which heightens the risk of developing ESRD and related complications. This association is particularly pronounced in the presence of hyperglycemia or diabetes. DKD represents a paradigmatic example in which sustained hyperglycemia induces profound epigenetic alterations—such as DNA methylation and histone modifications—that reprogram pathological gene expression [9,90]. Notably, histone modifications observed in the proximal tubular cells of diabetic mice persist even after normalization of glycemia, underscoring the long-lasting nature of these epigenetic changes [91,92].
The investigation of epigenetic processes contributing to metabolic memory of CKD progression and its cardiovascular complications is an emerging and rapidly expanding field. Multiple biochemical and signaling pathways are disrupted in CKD, including those governing inflammation, immune modulation, EMT, and metabolic homeostasis, key drivers of renal fibrosis and vasculopathy, including accelerated atherosclerosis and vascular calcification [93,94].

4.1. Histone Modifications and the Activation of Pro-Inflammatory Signaling

Epigenetic modulation involves alterations to DNA accessibility, determined in part by post-translational modifications of core histones. These histone modifications are intrinsically linked to the development and progression of DKD, contributing to localized oxidative stress, fibrosis, and inflammation within the renal environment [50]. Specifically, certain histone modifications can activate pro-inflammatory signaling pathways, thereby initiating sustained inflammation and structural damage within the kidney parenchyma [95]. In diabetic mouse models, reduced Suv39h1 levels led to decreased H3K9me3 deposition at inflammatory gene promoters, promoting gene derepression and persistent vascular inflammation [96]. Hyperglycemia enhances activating histone marks (H3K9ac, H3K4me1, and H3K4me3) while diminishing repressive marks (H3K27me3) at promoters such as TXNIP, a key regulator of oxidative stress and inflammation [97]. Preclinical evidence from DKD models also indicates that genes such as plasminogen activator inhibitor 1 (Pai1) and cyclin-dependent kinase inhibitor 1A (CDKN1A) are epigenetically regulated by TGF-β or high-glucose conditions in mesangial cells. Both Pai1 and CDKN1A are pro- fibrotic genes that contribute to DKD pathogenesis by promoting fibrosis, hypertrophy, tubular dysfunction, and glomerulosclerosis. Increased expression of these genes has been associated with enhanced deposition of activating histone marks (H3K9ac and H3K14ac) and the recruitment of the histone acetyltransferases p300 and CREB-binding protein (CBP) at Smad and Sp1 promoters [98].
Hyperglycemia also induces sustained DNMT1 overexpression and histone methylation changes, even after glucose normalization, establishing an epigenetic “memory” [23,99]. Short-term hyperglycemic exposure can trigger long-lasting H3K4 monomethylation at the NF-κB p65 promoter, maintaining pro-inflammatory gene expression and vascular dysfunction through persistent VCAM-1 and CCL-2 expression [23]. These effects are prevented by suppressing mitochondrial superoxide production. Similarly, decreased H3K9me2/3 at the NF-κB promoter sustains its expression, promoting inflammation [50]. Furthermore, hyperglycemia-induced PTMs at nitric oxide synthase (NOX4) and endothelial nitric oxide synthase (eNOS) promoters perpetuate ROS production and vascular damage [100]. Epigenetic enzymes such as histone methyltransferases (HMTs), histone demethylases (HDMs), and DNMTs interact antagonistically to maintain inflammatory gene expression [23,101,102]. The HMT EZH2, a PRC2 complex component, contributes to H3K27me3 deposition on the HIC1 gene in glucose-stimulated tubular endothelial cells, repressing SIRT1, a key antioxidant factor, and promoting ROS accumulation [102].

4.2. DNA Methylation Status Connected to Renal Disease Progression

Likewise histone methylation, altered DNA methylation patterns influence the expression of cytokines and chemokines. Hyperglycemia induces dramatic changes in DNA methylation status that coincide with the activation of pathways associated with renal disease progression [13]. Mechanistically, high glucose exposure leads to the accumulation of ROS, which not only causes direct damage to glomerular cells but also alters gene expression via epigenetic changes [13]. Emerging evidence indicates that oxidative stress, a known driver of tubular damage, directly modulates DNA methylation patterns by altering the function of key methylation enzymes, specifically DNMTs and TET proteins [103]. This interplay provides a direct causal mechanism linking environmental stress (oxidative damage) to sustained epigenetic outcomes that drive disease. Changes in DNA methylation were specifically observed in the promoter regions of genes related to oxidative stress, inflammation, and renal fibrosis in both glomerular and tubular cells under diabetic conditions [13].
Studies focusing on advanced CKD patients, particularly those undergoing dialysis, have established a strong relationship between global DNA methylation status in circulating immune cells and poor clinical outcomes [104,105]. Global DNA hypermethylation correlates with systemic inflammation and poor clinical outcomes [106,107]. In contrast to systemic methylation markers, highly localized, tissue-specific methylation changes offer critical information for predicting organ-specific progression. Gluck et al. identified 471 methylation probes associated with renal failure, enriched within kidney regulatory regions, including the epidermal growth factor (EGF) gene locus, capable of predicting renal function decline in DKD [108,109]. Additional evidence indicates that diabetes-related DNA methylation changes in genes regulating oxidative stress, inflammation, and fibrosis occur in glomerular and tubular cells [92]. Similarly, hyperglycemia-induced DNA methylation alterations parallel the activation of pathways driving renal disease progression. Furthermore, extensive alterations in histone methylation have been observed in diabetic kidney tissue, with numerous sites exhibiting both hypermethylation and hypomethylation compared to non-diabetic controls [110]. Complementary evidence from a study published in Diabetic Medicine identified distinct DNA methylation patterns in mitochondrial protein-coding genes associated with kidney disease in type 1 diabetes. This study highlighted 51 genes, including TAMM41 and COX6A1, exhibiting both genetic and epigenetic associations with DKD [111]. Moreover, differential methylation of genes essential for mitochondrial metabolism—such as PMPCB, TSFM, and AUH—further implicates epigenetic dysregulation in the pathogenesis of DKD and progression to end-stage renal failure [111].

4.3. The Role of Sirtuins in the Hyperglycemia-Induced Epigenetic Memory

Among epigenetic regulators, the NAD+-dependent histone deacetylases known as Sirtuins have drawn substantial attention. In particular, SIRT1 regulates mitochondrial energy metabolism and redox balance [112]. Experimental studies demonstrate that SIRT1 activation stimulates the LKB1/AMPK/ROS pathway, suppressing ROS production and inflammation [113]. Conversely, hyperglycemia-induced the Poly (ADP-ribose) polymerase (PARP) activation inhibits SIRT1, forming a self-perpetuating cycle of oxidative stress. Wang and colleagues elucidated a molecular mechanism underlying the protective role of SIRT1. Their study revealed that miRNA-155 expression is markedly elevated in the serum and kidney tissues of diabetic mice, as well as in podocytes exposed to high-glucose conditions. This upregulation enhances the production of inflammatory mediators in podocytes and appears to occur via suppression of SIRT1 mRNA expression [114].
Additionally, SIRT1 also deacetylates endothelial eNOS, maintaining endothelial integrity and preventing vascular senescence and macrophage foam cell formation [115]. Downregulation of SIRT1 elevates miR-34a levels, which enhances p66shc-mediated ROS production and endothelial dysfunction [116]. Overexpression of SIRT1 activates the NRF2/ARE antioxidant pathway, increasing expression of HO-1 and SOD, and reducing AGE-induced ROS in mesangial cells [117].
Other Sirtuins, including SIRT3 and SIRT7, are critical for mitochondrial dynamics and renal protection. SIRT3 mitigates oxidative stress and metabolic dysfunction in proximal tubular cells [118,119]. Reduced SIRT3 expression correlates with increased mitochondrial mtROS and cardiovascular risk [120,121]. The mtROS accumulation activates PARP, leading to endothelial dysfunction—a key event in diabetic complications [122]. SIRT7, responsible for H3K18 deacetylation, plays a protective role against hyperglycemia-induced endothelial-to-mesenchymal transition (EndMT) and podocytes apoptosis [122,123,124,125]. Its downregulation after transient hyperglycemia perpetuates inflammation via the ELK1/DAPK3 axis and enhances vascular injury [126,127]. Notably, miR-20b directly targets SIRT7, and its overexpression induces podocytes apoptosis through caspase-3 activation, whereas SIRT7 restoration mitigates this damage [124].

4.4. MicroRNAs and the Epigenetic Memory

MiRNAs have emerged as pivotal epigenetic regulators in hyperglycemia-induced kidney disease, fine-tuning gene expression programs that control fibrosis, inflammation, and apoptosis [128,129]. Yao et al. demonstrated that transient hyperglycemia could induce long-lasting inflammation through the NF-κB/miR-27a-3p/NRF2/ROS/TGF-β/EndMT feedback loop, and the activation of NRF2 can reverse these effects. NRF2, a master regulator of antioxidant defense, controls the expression of genes such as SOD2 and GPX, thereby maintaining cellular redox balance [130]. Its deregulation throughout the stages of CKD reflects an initial compensatory activation followed by functional exhaustion, likely mediated by epigenetic memory mechanisms [131,132]. Aberrant miRNA expression contributes, not only to DKD, but also to other vascular complications, such as atherosclerosis, retinopathy, and microvascular dysfunction [133,134]. Endothelial cells miRNA profiling revealed that miR-130b-3p, miR-221-3p, and miR-140-5p are glucose-responsive miRNAs that target genes involved in apoptosis and angiogenesis [135]. Persistent upregulation of miR-125b activates NF-κB signaling, while reduced miR-146a-5p enhances pro-inflammatory gene transcription [136]. Similarly, miR-214 downregulates PTEN, promoting Akt/mTORC activation and renal injury [137], whereas miR-22 suppresses PTEN and autophagy, thereby driving fibrotic responses [138]. Under hyperglycemic conditions, miR-21 also suppresses PTEN while activating Akt and TORC1 pathways; its overexpression, commonly observed in DKD, contributes to renal cell hypertrophy, fibrosis, and EMT [139]. Dysregulation of this and other miRNAs, including members of the miR-200 family, constitutes a central mechanism in DKD progression. The increased TGF-β expression observed in diabetic nephropathy is mediated by miR-192, which in turn regulates miR-200, further amplifying glomerular fibrogenesis and hypertrophy [140]. The crucial role of miR-126 in endothelial protection and angiogenesis has positioned it as a major focus of research in both the pathophysiology of diabetes and its vascular complications [141]. Notably, patients with CKD exhibit reduced levels of the atheroprotective, endothelium-derived miR-126, and its downregulation has been specifically associated with both micro- and macrovascular complications in type 1 diabetes [142]. In addition, circulating miRNA profiles are significantly altered in response to hyperglycemia. In patients with type 1 diabetes, miR-125b-5p and miR-365a-3p levels shown a positive correlation with HbA1c levels, whereas miR-5190 and miR-770-5p display negative correlations [143]. Furthermore, extracellular vesicles enriched with miR-15 and miR-16 derived from diabetic patients have been shown to preserve a “metabolic memory” signal that perpetuates endothelial CaMK2a activation and cardiac dysfunction [144].
The ongoing elucidation of epigenetically regulated networks in CKD is providing deeper insights into disease pathogenesis and potential therapeutic strategies. Sustained endothelial dysfunction—driven by oxidative stress, inflammation, EndMT, and disrupted metabolic homeostasis—is maintained by epigenetic “memory” mechanisms that stabilize pathological phenotypes [145] (Figure 3). Consequently, effective therapeutic approaches for CKD should extend beyond glycemic control to specifically target molecular and epigenetic memory pathways, offering new opportunities to halt or even reverse renal functional decline.

5. Epigenetic Players as Therapeutic Targets and Biomarkers for CKD Patients’ Stratification

Epigenetic molecules act cooperatively to preserve the integrity of the epigenetic machinery and maintain cell identity. They can be categorized functionally as: (i) Writers, which add chemical marks (e.g., methyl or acetyl groups) to DNA or histones, including DNMTs, HATs, and HMTs; (ii) Readers, which recognize and bind specific epigenetic marks, such as bromodomains (acetyl-lysine readers) and chromodomains (methyl-lysine readers); (iii) Erasers, which remove post-translational modifications, including HDACs and HDMs [146,147].
The therapeutic targeting of these regulators has emerged as a promising pharmacological avenue. Several epigenetic modulators—collectively known as epidrugs—have already achieved clinical success in oncology. DNMT inhibitors (DNMTi; e.g., azacytidine, decitabine) and HDAC inhibitors (HDACi; e.g., vorinostat, panobinostat) have been approved for the treatment of myelodysplastic syndromes, leukemias, and other hematologic malignancies [147,148,149,150]. The current landscape of FDA-approved epidrugs includes a wide range of compounds (Table 2), and numerous inhibitors (e.g., ivaltinostat, AR-42, abexinostat, bisthianostat, valproic acid) are undergoing clinical evaluations.
Combination strategies integrating epidrugs with traditional chemotherapeutics are being explored to curb both solid and hematologic tumor progression [151,152,153,154]. Beyond oncology, epigenetic modulators are being investigated for a variety of conditions, including cardiovascular, neurodevelopmental, and neurodegenerative disorders [155,156]. Originally designed for cancer, these compounds also exhibit potential for mitigating chronic inflammation, oxidative stress, and fibrotic remodeling—key mechanisms underpinning metabolic memory and CKD progression [157,158,159,160]. Because epigenetic marks are reversible, targeting the enzymes that write, read, and erase them, including through miRNA mimics or inhibitors, offers a promising therapeutic strategy for managing CKD and its complications, and supports the development of precision medicine approaches for high-risk patients.

5.1. Epigenetic Biomarkers in CKD and Diabetes

Early biomarker identification is critical for timely diagnosis, intervention, and prevention of progression to ESRD. Epigenome-wide association studies (EWAS) in individuals newly diagnosed with diabetes have uncovered blood-based epigenetic biomarkers capable of: (i) early risk stratification, (ii) predicting CKD development, and (iii) monitoring disease progression. To elucidate the relationship between metabolic memory and epigenetic modifications, several genome-wide studies based on samples from the DCCT/EDIC cohort have been conducted [161]:
(1)
In a first study, patients exhibited elevated levels of the active chromatin mark H3K9ac in monocytes, correlated with baseline hyperglycemia and the upregulation of genes linked to inflammation and vascular complications. H3K9ac thus emerged as a potential biomarker for metabolic memory [162].
(2)
In a second study, DNA methylation profiling at two time points revealed 12 persistently differentially methylated loci, notably TXNIP, which was hypomethylated in patients with more complications [163]. Other validations confirmed a strong inverse correlation between TXNIP methylation and glycemic control, suggesting that TXNIP hypomethylation reflects chronic hyperglycemia and may predict renal and vascular damage [164].
(3)
In a third study, longitudinal analysis (18-year follow-up) showed that specific HbA1c-associated CpGs predicted DKD risk, again highlighting TXNIP as a key biomarker [165,166].
These findings, corroborated by studies across tissues such as kidney, nerves, and retina, suggest TXNIP as a pivotal player in metabolic memory and a target for intervention [97,167,168].
A longitudinal study from Lund University followed 752 newly diagnosed type 2 diabetes patients, identifying over 400 differentially methylated sites associated with cardiovascular events, 87 of which were used to build a methylation risk score for cardiovascular risk prediction [169]. Similar EWAS have identified differential methylation in genes regulating mitochondrial function, fibrosis, and inflammation, molecular hallmarks of persistent renal injury [14,111,170,171,172].
A methylation score proposed by Marchiori et al., derived from 37 methylation sites, predicted CKD incidence during an 11.5-years follow-up [173]. Moreover, aberrant DNA methylation patterns in key genes (mTOR, RPTOR, IRS2, GRK5, LCAT, SLC27A3, SLC1A5) have been linked to DKD progression [174]. Elevated DNMT1 expression in DKD mononuclear cells induces mTOR-related methylation changes and inflammation, reversible through DNMT1 inhibition with 5-aza-2′-deoxycytidine (5-aza), offering a new therapeutic perspective [175].
Different compounds have been proven to act on the key mechanisms of hyperglycemic memory, such as epigenetic modifications, inflammation, and senescence, but none have been approved for CKD and complications. The DNMT1 inhibitor SGI-1027 disrupts p21 methylation, attenuating senescence and fibrosis in DKD models [45,176]. Histone methylation also contributes to metabolic memory. Downregulation of the demethylase JMJD3 (also called KDM6B) correlates with neointimal hyperplasia in CKD-related vascular lesions [177]. JMJD3 loss promotes EndMT, vascular remodeling, and nitric oxide deficiency. Additionally, EZH2 upregulation, with concurrent loss of JMJD3, represses antioxidant genes (SOD1/2, JunD), exacerbating oxidative stress [178]. In fact, its upregulation is linked to tubular cell injury, podocytes dedifferentiation, fibroblast proliferation, inflammatory cytokines/chemokines production and infiltration of inflammatory cells [179]; moreover, depleting EZH2 from podocytes culture in high-glucose conditions derepressed the endogenous antioxidant TXNIP, abrogating ROS accumulation [180]. These findings make EZH2 as a legitimate therapeutic target; however, no current clinical trials of EZH2 inhibitors are available for treating hyperglycemia-induced CKD.
Aberrant activity of BET (bromodomain and extra-terminal domain) proteins—particularly BRD2, BRD3, and BRD4—also sustains vascular and renal injury. BET inhibitors (BETi) such as JQ1 and Apabetalone (RVX-208) have shown preclinical efficacy in DKD and diabetic cardiomyopathy. The selective pharmacological inhibition of BRD4 with JQ1 restored mitochondrial functions via the PTEN-induced PINK1/Parkin mitophagy pathway [181,182]. Besides, the phase 3 BETonMACE trial (ClinicalTrials.gov ID: NCT02586155) tested Apabetalone in type 2 diabetes, with or without CKD, focusing on cardiovascular complications. While it did not significantly reduce major adverse cardiac events overall, it markedly reduced heart failure hospitalizations, particularly among CKD patients’ subgroup [183]. Further clinical studies are needed to confirm these brilliant results in a larger patients’ sample; nevertheless, future clinical settings must be adjusted to reduce side effects [184].

5.2. Histone Deacetylase Inhibitors and miRNA-Based Therapies

HDAC inhibitors are gaining interest in diabetic nephropathy [50,185]. Non-selective inhibitors such as Vorinostat (SAHA) improved renal function and reduced fibrosis in animal models [186]. Another HDACi Valproic acid (VPA), used for epilepsy and neurologic disorders, was demonstrated to repress the NF-κB pathway, inactivating inflammation and alleviating podocytes and renal cell injury [187]. The same effects on inflammation and fibrosis have been observed with the HDACi trichostatin A (TSA); this compound was able to influence the metabolic memory through the inhibition of TGF-β1, the decrease in extracellular matrix deposition and the escape from the phenomenon of EMT [188]. SIRT1, a class III HDAC, regulates metabolic memory by deacetylating p53 and modulating apoptosis, inflammation, and oxidative stress [113,189]; its activity is negatively regulated by miR-200a-3p, whose inhibition mitigates tubular injury, suggesting its biomarker potential for diabetes- and hypertension-related nephropathy [190].
MiRNAs are both biomarkers and therapeutic targets in DKD. Two strategies have emerged:
  • Inhibition of overexpressed miRNAs (via antisense oligonucleotides, gene knockouts, or “sponges”);
  • Restoration of protective miRNAs (via double-stranded mimics or expression vectors).
Examples include (i) miR-29b mimics, which interfere with Sp1-dependent, TGF-β/Smad3-mediated renal fibrosis, NF-κB–driven renal inflammation, and Th1-associated immune injury [191,192]; and (ii) inhibitors of miR-21 and miR-192, which reduce albuminuria and renal fibrosis, maintaining the structural and functional integrity of the kidney [193,194]. Circulating miRNAs such as miR-1, miR-133a, and miR-126 serve as non-invasive biomarkers for diabetic cardiomyopathy and cardiovascular risk [195,196].
In a study by Al-Kafaji et al., diabetic nephropathy was associated with upregulation of miR-377 and downregulation of miR-192, correlating with albuminuria severity and identifying miR-377 as a positive, and miR-192 as a negative, biomarker of DKD [197]. Recent approaches advocate miRNA panels (e.g., miR-21, miR-34a, miR-133a) to enhance diagnostic and prognostic power, bridging therapy and diagnostics—theranostics—for patient stratification [14,198].

5.3. Integrating Machine Learning and Multi-Omics

Recent advances in Artificial Intelligence (AI) have enabled integration of epigenetic, genomic, transcriptomic, and metabolomic data to predict disease risk and uncover novel therapeutic targets. A recent study from the Middle East that combine EWAS, genome-wide analyses, and metabolomics identifies several previously unrecognized methylated genes, genomic variants, and metabolic pathways implicated in diabetes [199]. These integrative analyses revealed clinically relevant molecular subnetworks, offering new avenues for the development of translational multi-omics applications.
Similarly, another multi-omics investigation integrating metabolomics, transcriptomics, proteomics, and epigenomics identified the transcription factor KLF15 as a critical regulator of diabetic cardiomyopathy, positioning it as a powerful therapeutic target [200].
The application of machine learning algorithms represents a paradigm shift in the management of diabetes and its cardiovascular complications. A compelling example is the Multi-Ethnic Study of Atherosclerosis (MESA) clinical trial (ClinicalTrials.gov ID: NCT00005487), in which machine learning-based modeling of protein–protein interaction networks, when combined with imaging tomography and deep phenotyping, successfully predicted cardiovascular outcomes [201]. Coupling machine learning algorithms with epigenomics and multi-omics datasets can further facilitate the discovery of novel biomarkers for cardiovascular risk stratification in diabetic patients. This integrated approach holds the potential to transform patient care by enabling earlier and more precise diagnoses, individualized risk assessment, and targeted treatment interventions.

6. Conclusions

Further studies are urgently needed to validate candidate biomarkers and identify safer, more effective epigenetic drugs. Integrating multi-omics data will allow stratification of patients predisposed to vascular complications and enhance precision medicine in CKD. Understanding the dynamic role of epigenetic regulation is pivotal for predicting disease trajectory, preventing toxin-induced CKD progression, and overcoming micro- and macrovascular complications.

Author Contributions

Conceptualization, S.C. and I.G.; writing—original draft preparation, S.C., I.G. and A.P.; writing—review and editing, S.C. and I.G.; visualization, M.D., F.S. and A.R.; supervision, A.P., I.G. and M.P.-M.; All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by “HEAL ITALIA—Health Extended Alliance for Innovative Therapies, Advanced Lab-research and Integrated Approaches of Precision Medicine” Cod. PE_00000019 with financial contribution of European Union—NextGenerationUE as part of PNRR MUR—M4C2—Investimento 1.3—Public Call “Partenariati Estesi”—D.D. n. 341/2022.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article. Further inquiries can be directed to the corresponding author.

Acknowledgments

The figures were created with BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
5-aza5-Aza-2′-deoxycytidine
5-mC5-Methylcytosine
ADPAdenine Diphosphate
AGEAdvanced Glycation End Products
AIArtificial Intelligence
AMPKAMP-Activated Protein Kinase
AKIAcute Kidney Injury
AktProtein Kinase B
AR-42Histone Deacetylase Inhibitor AR-42/HDAC-42
AREAntioxidant Response Element
AUHAU RNA Binding Methylglutaconyl-CoA Hydratase
BD1Bromodomain1
BD2Bromodomain2
BETBromodomain and Extra-Terminal Domain
BETiBromodomain and Extra-Terminal Domain Inhibitor
BRDBromodomain Containing Protein
-CH3Methyl group
CaMK2aCalcium/Calmodulin-Dependent Protein Kinase II alpha
CBPCREB-Binding Protein
CBXPolycomb Group Protein Chromobox
CCL-2C-C Motif Chemokine Ligand 2
CDKN1ACyclin Dependent Kinase Inhibitor 1A
CH3CO-Acetyl group
CKDChronic Kidney Disease
COX6A1Cytochrome C Oxidase Subunit 6A1
CpGCytosine-phosphate-Guanine
DAPK3Death-Associated Protein Kinase 3
DCCTDiabetes Control and Complications Trial
DGCR8DiGeorge Syndrome Critical Region Gene 8
DKDDiabetic Kidney Disease
DMNTDNA Methyltransferases
DNMTiDNA Methyltransferase Inhibitor
DRP1Dynamin-Related Protein 1
ECMExtracellular Matrix
EDICEpidemiology of Diabetes Interventions and Complications Study
ELK1ETS Transcription Factor ELK1
EGFEpidermal Growth Factor
eGFREstimated Glomerular Filtration Rate
EMTEpithelial-to-Mesenchymal Transition
EndMTEndothelial-to-Mesenchymal Transition
eNOSEndothelial Nitric Oxide Synthase
ERKExtracellular signal-regulated kinase
ESRDEnd-Stage Renal Disease
EWASEpigenetic-Wide Association Study
EZH2Enhancer of Zeste Homolog 2
FDAFood and Drug Administration
GBD Global Burden Disease, Injuries, and Risk Factors
GFRGlomerular Filtration Rate
GPXGlutathione Peroxidase
GRK5G Protein-coupled Receptor Kinase 5
H1Histone H1
H2AHistone H2A
H2BHistone H2B
H3Histone H3
H4Histone H4
HATsHistone Acetyltransferases
HbA1cGlycated Haemoglobin
HDACHistone Deacetylase
HDACiHistone Deacetylase Inhibitor
HDMHistone Demethylase
HIC-1Hypermethylated in Cancer 1
HMTHistone Methyltransferase
HO-1Heme Oxygenase-1
IRS2Insulin Receptor Substrate 2
JNKJanus Kinase
JMJD3Lysine demethylase 6B
JunDTranscription Factor JunD
JQ-1Bromodomain 4 Inhibitor
KDIGOKidney Disease Improving Global Outcomes Guidelines
KDMLysine Demethylase
KDOQIKidney Disease Outcomes Quality Initiative Guidelines
KMTLysine Methyltransferase
LCATLecithin Cholesterol Acyltransferase
LKB1Liver Kinase B1
MAPKMitogen-Activated Protein Kinase
Mdivi-1Mitochondrial Division Inhibitor-1
MeCP2Methyl-CpG-Binding Protein
MESAMulti-Ethnic Study of Atherosclerosis
MFN2Mitofusin-2
miRNAMicroRNA
mtDNAMitochondrial DNA
mtROSMitochondrial Reactive Oxygen Species
mTORMechanistic Target of Rapamycin
mTORCMechanistic Target of Rapamycin Complex
NAD+Nicotinamide Adenine Dinucleotide
NOXNicotinamide Adenine Dinucleotide Phosphate Oxidase
NF-κBNuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cell
NRF2Nuclear Factor Erythroid 2-Related Factor 2
Pai-1Plasminogen Activator Inhibitor 1
PARPPoly (ADP-ribose) Polymerase
p21Cyclin-Dependent Kinase Inhibitor 1
p53Transformation-Related Protein 53
p65NF-Kappa-B Transcription Factor P65
PINK-1PTEN-Induced Kinase 1
PMPCBPeptidase, Mitochondrial Processing Subunit Beta
PRC2Polycomb Repressive Complex 2
PTENPhosphatase and TENsin Homolog
PTMPost-Translational Modification
RAGE Glycation End Products Receptor
RISCRNA-Induced Silencing Complex
ROSReactive Oxygen Species
RPTORRegulatory Associated Protein of mTOR Complex 1
SAHASuberoylanilide Hydroxamic Acid
SAMS-Adenosyl-l-Methionine
SGI-1027DNA Methyltransferase Inhibitor II
SIRTSilent Mating Type Information Regulation 2 Homolog
SLC1A5Solute Carrier Family 1 (neutral amino acid transporter), Member 5
SLC27A3Solute Carrier Family 27 Member 3
SMADSmall Mother Against Decapentaplegic
SODSuperoxide Dismutase
Sp-1Specificity Protein 1
Suv39H1Suppressor of Variegation 3-9 Homolog 1
TAMM41TAM41 Mitochondrial Translocator Assembly and Maintenance Homolog
TETTen-Eleven Translocation
TGF-αTransforming Growth Factor-alpha
TGF-βTransforming Growth Factor-beta
Th1T helper 1
TSATrichostatin A
TSFMTs Translation Elongation Factor, Mitochondrial
TXNIPThioredoxin-Interacting Protein
UKPDSUnited Kingdom Prospective Diabetes Study
VCAM-1Vascular Cell Adhesion Molecule 1
VPAValproic Acid

References

  1. Inker, L.A.; Astor, B.C.; Fox, C.H.; Isakova, T.; Lash, J.P.; Peralta, C.A.; Kurella Tamura, M.; Feldman, H.I. KDOQI US commentary on the 2012 KDIGO clinical practice guideline for the evaluation and management of CKD. Am. J. Kidney Dis. 2014, 63, 713–735. [Google Scholar] [CrossRef]
  2. Rysz, J.; Franczyk, B.; Rysz-Gorzynska, M.; Gluba-Brzozka, A. Are Alterations in DNA Methylation Related to CKD Development? Int. J. Mol. Sci. 2022, 23, 7108. [Google Scholar] [CrossRef]
  3. Gilg, J.; Rao, A.; Fogarty, D. UK Renal Registry 16th annual report: Chapter 1 UK renal replacement therapy incidence in 2012: National and centre-specific analyses. Nephron. Clin. Pract. 2014, 125, 1–28. [Google Scholar] [CrossRef]
  4. GBD Chronic Kidney Disease Collaboration, Global, regional, and national burden of chronic kidney disease, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2020, 395, 709–733. [CrossRef]
  5. Ho, H.J.; Shirakawa, H. Oxidative Stress and Mitochondrial Dysfunction in Chronic Kidney Disease. Cells 2022, 12, 88. [Google Scholar] [CrossRef]
  6. Levey, A.S.; Stevens, L.A.; Coresh, J. Conceptual model of CKD: Applications and implications. Am. J. Kidney Dis. 2009, 53, S4–S16. [Google Scholar] [CrossRef] [PubMed]
  7. Rout, P.; Jialal, I. Diabetic Nephropathy; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  8. Tervaert, T.W.; Mooyaart, A.L.; Amann, K.; Cohen, A.H.; Cook, H.T.; Drachenberg, C.B.; Ferrario, F.; Fogo, A.B.; Haas, M.; de Heer, E.; et al. Pathologic classification of diabetic nephropathy. J. Am. Soc. Nephrol. 2010, 21, 556–563. [Google Scholar] [CrossRef]
  9. Liebisch, M.; Wolf, G. Role of Epigenetic Changes in the Pathophysiology of Diabetic Kidney Disease. Glomerular Dis. 2024, 4, 211–226. [Google Scholar] [CrossRef] [PubMed]
  10. Amorim, R.G.; Guedes, G.D.S.; Vasconcelos, S.M.L.; Santos, J.C.F. Kidney Disease in Diabetes Mellitus: Cross-Linking between Hyperglycemia, Redox Imbalance and Inflammation. Arq. Bras. Cardiol. 2019, 112, 577–587, Erratum in Arq. Bras. Cardiol. 2019, 113, 182. [Google Scholar] [CrossRef]
  11. Reidy, K.; Kang, H.M.; Hostetter, T.; Susztak, K. Molecular mechanisms of diabetic kidney disease. J. Clin. Investig. 2014, 124, 2333–2340. [Google Scholar] [CrossRef] [PubMed]
  12. Turkmen, K. Inflammation, oxidative stress, apoptosis, and autophagy in diabetes mellitus and diabetic kidney disease: The Four Horsemen of the Apocalypse. Int. Urol. Nephrol. 2017, 49, 837–844. [Google Scholar] [CrossRef]
  13. 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]
  14. Kuo, F.C.; Chao, C.T.; Lin, S.H. The Dynamics and Plasticity of Epigenetics in Diabetic Kidney Disease: Therapeutic Applications Vis-a-Vis. Int. J. Mol. Sci. 2022, 23, 843. [Google Scholar] [CrossRef] [PubMed]
  15. Giacco, F.; Brownlee, M. Oxidative stress and diabetic complications. Circ. Res. 2010, 107, 1058–1070. [Google Scholar] [CrossRef] [PubMed]
  16. Yapislar, H.; Gurler, E.B. Management of Microcomplications of Diabetes Mellitus: Challenges, Current Trends, and Future Perspectives in Treatment. Biomedicines 2024, 12, 1958. [Google Scholar] [CrossRef]
  17. Thomas, M.C.; Brownlee, M.; Susztak, K.; Sharma, K.; Jandeleit-Dahm, K.A.; Zoungas, S.; Rossing, P.; Groop, P.H.; Cooper, M.E. Diabetic kidney disease. Nat. Rev. Dis. Primers 2015, 1, 15018. [Google Scholar] [CrossRef]
  18. Pal, R.; Bhadada, S.K. AGEs accumulation with vascular complications, glycemic control and metabolic syndrome: A narrative review. Bone 2023, 176, 116884. [Google Scholar] [CrossRef]
  19. Ng, Z.X.; Kuppusamy, U.R.; Iqbal, T.; Chua, K.H. Receptor for advanced glycation end-product (RAGE) gene polymorphism 2245G/A is associated with pro-inflammatory, oxidative-glycation markers and sRAGE in diabetic retinopathy. Gene 2013, 521, 227–233. [Google Scholar] [CrossRef]
  20. de Souza Ferreira, C.; Pennacchi, P.C.; Araujo, T.H.; Taniwaki, N.N.; de Araujo Paula, F.B.; da Silveira Duarte, S.M.; Rodrigues, M.R. Aminoguanidine treatment increased NOX2 response in diabetic rats: Improved phagocytosis and killing of Candida albicans by neutrophils. Eur. J. Pharmacol. 2016, 772, 83–91. [Google Scholar] [CrossRef]
  21. Sugahara, M.; Pak, W.L.W.; Tanaka, T.; Tang, S.C.W.; Nangaku, M. Update on diagnosis, pathophysiology, and management of diabetic kidney disease. Nephrology 2021, 26, 491–500. [Google Scholar] [CrossRef]
  22. Ceriello, A. Hypothesis: The “metabolic memory”, the new challenge of diabetes. Diabetes Res. Clin. Pract. 2009, 86, S2–S6. [Google Scholar] [CrossRef]
  23. El-Osta, A.; Brasacchio, D.; Yao, D.; Pocai, A.; Jones, P.L.; Roeder, R.G.; Cooper, M.E.; Brownlee, M. Transient high glucose causes persistent epigenetic changes and altered gene expression during subsequent normoglycemia. J. Exp. Med. 2008, 205, 2409–2417. [Google Scholar] [CrossRef]
  24. Luna, P.; Guarner, V.; Farias, J.M.; Hernandez-Pacheco, G.; Martinez, M. Importance of Metabolic Memory in the Development of Vascular Complications in Diabetic Patients. J. Cardiothorac. Vasc. Anesth. 2016, 30, 1369–1378. [Google Scholar] [CrossRef] [PubMed]
  25. Testa, R.; Bonfigli, A.R.; Prattichizzo, F.; La Sala, L.; De Nigris, V.; Ceriello, A. The “Metabolic Memory” Theory and the Early Treatment of Hyperglycemia in Prevention of Diabetic Complications. Nutrients 2017, 9, 437. [Google Scholar] [CrossRef] [PubMed]
  26. Dong, H.; Sun, Y.; Nie, L.; Cui, A.; Zhao, P.; Leung, W.K.; Wang, Q. Metabolic memory: Mechanisms and diseases. Signal Transduct. Target. Ther. 2024, 9, 38. [Google Scholar] [CrossRef]
  27. Roy, S.; Sala, R.; Cagliero, E.; Lorenzi, M. Overexpression of fibronectin induced by diabetes or high glucose: Phenomenon with a memory. Proc. Natl. Acad. Sci. USA 1990, 87, 404–408. [Google Scholar] [CrossRef]
  28. Lachin, J.M.; Bebu, I.; Nathan, D.M. DCCT/EDIC Research Group, The Beneficial Effects of Earlier Versus Later Implementation of Intensive Therapy in Type 1 Diabetes. Diabetes Care 2021, 44, 2225–2230. [Google Scholar] [CrossRef]
  29. Wilson-Verdugo, M.; Bustos-Garcia, B.; Adame-Guerrero, O.; Hersch-Gonzalez, J.; Cano-Dominguez, N.; Soto-Nava, M.; Acosta, C.A.; Tusie-Luna, T.; Avila-Rios, S.; Noriega, L.G.; et al. Reversal of high-glucose-induced transcriptional and epigenetic memories through NRF2 pathway activation. Life Sci. Alliance 2024, 7, e202302382. [Google Scholar] [CrossRef] [PubMed]
  30. Nishikawa, T.; Edelstein, D.; Du, X.L.; Yamagishi, S.; Matsumura, T.; Kaneda, Y.; Yorek, M.A.; Beebe, D.; Oates, P.J.; Hammes, H.P.; et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature 2000, 404, 787–790. [Google Scholar] [CrossRef]
  31. Ihnat, M.A.; Thorpe, J.E.; Kamat, C.D.; Szabó, C.; Green, D.E.; Warnke, L.A.; Lacza, Z.; Cselenyák, A.; Ross, K.; Shakir, S.; et al. Reactive oxygen species mediate a cellular ‘memory’ of high glucose stress signalling. Diabetologia 2007, 50, 1523–1531. [Google Scholar] [CrossRef]
  32. Brownlee, M. The pathobiology of diabetic complications: A unifying mechanism. Diabetes 2005, 54, 1615–1625. [Google Scholar] [CrossRef]
  33. Araki, E.; Nishikawa, T. Oxidative stress: A cause and therapeutic target of diabetic complications. J. Diabetes Investig. 2010, 1, 90–96. [Google Scholar] [CrossRef] [PubMed]
  34. Caja, S.; Enríquez, J.A. Mitochondria in endothelial cells: Sensors and integrators of environmental cues. Redox Biol. 2017, 12, 821–827. [Google Scholar] [CrossRef]
  35. Cannito, S.; Giardino, I.; d’Apolito, M.; Pettoello-Mantovani, M.; Scaltrito, F.; Mangieri, D.; Piscazzi, A. The Multifaceted Role of Mitochondria in Angiogenesis. Int. J. Mol. Sci. 2025, 26, 7960. [Google Scholar] [CrossRef]
  36. Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial ROS-induced ROS release: An update and review. Biochim. Biophys. Acta 2006, 1757, 509–517. [Google Scholar] [CrossRef]
  37. Kowluru, R.A.; Alka, K. Mitochondrial Quality Control and Metabolic Memory Phenomenon Associated with Continued Progression of Diabetic Retinopathy. Int. J. Mol. Sci. 2023, 24, 8076. [Google Scholar] [CrossRef]
  38. Hombrebueno, J.R.; Cairns, L.; Dutton, L.R.; Lyons, T.J.; Brazil, D.P.; Moynagh, P.; Curtis, T.M.; Xu, H. Uncoupled turnover disrupts mitochondrial quality control in diabetic retinopathy. JCI Insight 2019, 4, e129760. [Google Scholar] [CrossRef]
  39. Kowluru, R.A.; Mohammad, G.; Kumar, J. Impaired Removal of the Damaged Mitochondria in the Metabolic Memory Phenomenon Associated with Continued Progression of Diabetic Retinopathy. Mol. Neurobiol. 2024, 61, 188–199. [Google Scholar] [CrossRef]
  40. Ho, P.T.B.; Clark, I.M.; Le, L.T.T. MicroRNA-Based Diagnosis and Therapy. Int. J. Mol. Sci. 2022, 23, 7167. [Google Scholar] [CrossRef] [PubMed]
  41. Wenceslau, C.F.; McCarthy, C.G.; Szasz, T.; Spitler, K.; Goulopoulou, S.; Webb, R.C. Working Group on DAMPs in Cardiovascular Disease. Mitochondrial damage-associated molecular patterns and vascular function. Eur. Heart J. 2014, 35, 1172–1177. [Google Scholar] [CrossRef] [PubMed]
  42. Giardino, I.; d’Apolito, M.; Brownlee, M.; Maffione, A.B.; Colia, A.L.; Sacco, M.; Ferrara, P.; Pettoello-Mantovani, M. Vascular toxicity of urea, a new “old player” in the pathogenesis of chronic renal failure induced cardiovascular diseases. Turk. Pediatri Ars. 2017, 52, 187–193. [Google Scholar] [CrossRef]
  43. Vercellino, I.; Sazanov, L.A. The assembly, regulation and function of the mitochondrial respiratory chain. Nat. Rev. Mol. Cell Biol. 2022, 23, 141–161. [Google Scholar] [CrossRef] [PubMed]
  44. Dai, W.; Lu, H.; Chen, Y.; Yang, D.; Sun, L.; He, L. The Loss of Mitochondrial Quality Control in Diabetic Kidney Disease. Front. Cell Dev. Biol. 2021, 9, 706832. [Google Scholar] [CrossRef] [PubMed]
  45. Yang, T.; Qi, F.; Guo, F.; Shao, M.; Song, Y.; Ren, G.; Linlin, Z.; Qin, G.; Zhao, Y. An update on chronic complications of diabetes mellitus: From molecular mechanisms to therapeutic strategies with a focus on metabolic memory. Mol. Med. 2024, 30, 71. [Google Scholar] [CrossRef]
  46. Mohammad, H.P.; Barbash, O.; Creasy, C.L. Targeting epigenetic modifications in cancer therapy: Erasing the roadmap to cancer. Nat. Med. 2019, 25, 403–418. [Google Scholar] [CrossRef] [PubMed]
  47. Schisano, B.; Tripathi, G.; McGee, K.; McTernan, P.G.; Ceriello, A. Glucose oscillations, more than constant high glucose, induce p53 activation and a metabolic memory in human endothelial cells. Diabetologia 2011, 54, 1219–1226. [Google Scholar] [CrossRef]
  48. Brownlee, M.; Hirsch, I.B. Glycemic variability: A hemoglobin A1c-independent risk factor for diabetic complications. JAMA 2006, 295, 1707–1708. [Google Scholar] [CrossRef]
  49. Tonna, S.; El-Osta, A.; Cooper, M.E.; Tikellis, C. Metabolic memory and diabetic nephropathy: Potential role for epigenetic mechanisms. Nat. Rev. Nephrol. 2010, 6, 332–341. [Google Scholar] [CrossRef]
  50. Kushwaha, K.; Garg, S.S.; Gupta, J. Targeting epigenetic regulators for treating diabetic nephropathy. Biochimie 2022, 202, 146–158. [Google Scholar] [CrossRef]
  51. Siebel, A.L.; Fernandez, A.Z.; El-Osta, A. Glycemic memory associated epigenetic changes. Biochem. Pharmacol. 2010, 80, 1853–1859. [Google Scholar] [CrossRef]
  52. Reddy, M.A.; Zhang, E.; Natarajan, R. Epigenetic mechanisms in diabetic complications and metabolic memory. Diabetologia 2015, 58, 443–455. [Google Scholar] [CrossRef] [PubMed]
  53. Tanemoto, F.; Nangaku, M.; Mimura, I. Epigenetic memory contributing to the pathogenesis of AKI-to-CKD transition. Front. Mol. Biosci. 2022, 9, 1003227. [Google Scholar] [CrossRef] [PubMed]
  54. Esteller, M. Epigenetics in cancer. N. Engl. J. Med. 2008, 358, 1148–1159. [Google Scholar] [CrossRef]
  55. Li, E.; Bestor, T.H.; Jaenisch, R. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 1992, 69, 915–926. [Google Scholar] [CrossRef]
  56. Egger, G.; Liang, G.; Aparicio, A.; Jones, P.A. Epigenetics in human disease and prospects for epigenetic therapy. Nature 2004, 429, 457–463. [Google Scholar] [CrossRef] [PubMed]
  57. Robertson, K.D. DNA methylation and chromatin—Unraveling the tangled web. Oncogene 2002, 21, 5361–5379. [Google Scholar] [CrossRef]
  58. Okano, M.; Bell, D.W.; Haber, D.A.; Li, E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 1999, 99, 247–257. [Google Scholar] [CrossRef]
  59. Dabrowski, M.J.; Wojtas, B. Global DNA Methylation Patterns in Human Gliomas and Their Interplay with Other Epigenetic Modifications. Int. J. Mol. Sci. 2019, 20, 3478. [Google Scholar] [CrossRef]
  60. Toth, D.M.; Szeri, F.; Ashaber, M.; Muazu, M.; Szekvolgyi, L.; Aranyi, T. Tissue-specific roles of de novo DNA methyltransferases. Epigenetics Chromatin 2025, 18, 5. [Google Scholar] [CrossRef]
  61. Yang, L.; Yu, S.J.; Hong, Q.; Yang, Y.; Shao, Z.M. Reduced Expression of TET1, TET2, TET3 and TDG mRNAs Are Associated with Poor Prognosis of Patients with Early Breast Cancer. PLoS ONE 2015, 10, e0133896. [Google Scholar] [CrossRef]
  62. Zhang, X.; Zhang, Y.; Wang, C.; Wang, X. TET (Ten-eleven translocation) family proteins: Structure, biological functions and applications. Signal Transduct. Target. Ther. 2023, 8, 297. [Google Scholar] [CrossRef] [PubMed]
  63. Sufiyan, S.; Salam, H.; Ilyas, S.; Amin, W.; Arshad, F.; Fatima, K.; Naeem, S.; Laghari, A.A.; Enam, S.A.; Mughal, N. Prognostic implications of DNA methylation machinery (DNMTs and TETs) expression in gliomas: Correlations with tumor grading and patient survival. J. Neurooncol. 2025, 173, 667–682. [Google Scholar] [CrossRef] [PubMed]
  64. Kouzarides, T. Chromatin modifications and their function. Cell 2007, 128, 693–705. [Google Scholar] [CrossRef] [PubMed]
  65. Hermann, J.; Schurgers, L.; Jankowski, V. Identification and characterization of post-translational modifications: Clinical implications. Mol. Aspects Med. 2022, 86, 101066. [Google Scholar] [CrossRef] [PubMed]
  66. Ji, Y.; Liu, S.; Zhang, Y.; Min, Y.; Wei, L.; Guan, C.; Yu, H.; Zhang, Z. Lysine crotonylation in disease: Mechanisms, biological functions and therapeutic targets. Epigenetics Chromatin 2025, 18, 13. [Google Scholar] [CrossRef]
  67. Raju, C.; Sankaranarayanan, K. Insights on post-translational modifications in fatty liver and fibrosis progression. Biochim. Biophys. Acta Mol. Basis Dis. 2025, 1871, 167659. [Google Scholar] [CrossRef]
  68. Shiio, Y.; Eisenman, R.N. Histone sumoylation is associated with transcriptional repression. Proc. Natl. Acad. Sci. USA 2003, 100, 13225–13230. [Google Scholar] [CrossRef]
  69. Tahiliani, M.; Mei, P.; Fang, R.; Leonor, T.; Rutenberg, M.; Shimizu, F.; Li, J.; Rao, A.; Shi, Y. The histone H3K4 demethylase SMCX links REST target genes to X-linked mental retardation. Nature 2007, 447, 601–605. [Google Scholar] [CrossRef]
  70. Kaniskan, H.U.; Martini, M.L.; Jin, J. Inhibitors of Protein Methyltransferases and Demethylases. Chem. Rev. 2018, 118, 989–1068. [Google Scholar] [CrossRef]
  71. Bu, C.; Xie, Y.; Weng, J.; Sun, Y.; Wu, H.; Chen, Y.; Ye, Y.; Zhou, E.; Yang, Z.; Wang, J. Inhibition of JMJD3 attenuates acute liver injury by suppressing inflammation and oxidative stress in LPS/D-Gal-induced mice. Chem. Biol. Interact. 2025, 418, 111576. [Google Scholar] [CrossRef] [PubMed]
  72. Cao, Y.; Li, Y.; Zhang, N.; Hu, J.; Yin, L.; Pan, Z.; Li, Y.; Du, X.; Zhang, W.; Li, F. Quantitative DNA hypomethylation of ligand Jagged1 and receptor Notch1 signifies occurrence and progression of breast carcinoma. Am. J. Cancer Res. 2015, 5, 1621–1634. [Google Scholar]
  73. Tycko, B. Epigenetic gene silencing in cancer. J. Clin. Investig. 2000, 105, 401–407. [Google Scholar] [CrossRef]
  74. Urli, T.; Greenberg, M.V.C. Epigenetic relay: Polycomb-directed DNA methylation in mammalian development. PLoS Genet. 2025, 21, e1011854. [Google Scholar] [CrossRef] [PubMed]
  75. Wiles, E.T.; Selker, E.U. H3K27 methylation: A promiscuous repressive chromatin mark. Curr. Opin. Genet. Dev. 2017, 43, 31–37. [Google Scholar] [CrossRef]
  76. Sharma, S.; Hampton, J.T.; Kutateladze, T.G.; Liu, W.R. Epigenetic reader chromodomain as a potential therapeutic target. RSC Chem. Biol. 2025, 6, 833–844. [Google Scholar] [CrossRef]
  77. Angrand, P.O. Structure and Function of the Polycomb Repressive Complexes PRC1 and PRC2. Int. J. Mol. Sci. 2022, 23, 5971. [Google Scholar] [CrossRef] [PubMed]
  78. Seto, E.; Yoshida, M. Erasers of histone acetylation: The histone deacetylase enzymes. Cold Spring Harb. Perspect. Biol. 2014, 6, a018713. [Google Scholar] [CrossRef] [PubMed]
  79. Cedar, H.; Bergman, Y. Linking DNA methylation and histone modification: Patterns and paradigms. Nat. Rev. Genet. 2009, 10, 295–304. [Google Scholar] [CrossRef]
  80. Esteve, P.O.; Chin, H.G.; Benner, J.; Feehery, G.R.; Samaranayake, M.; Horwitz, G.A.; Jacobsen, S.E.; Pradhan, S. Regulation of DNMT1 stability through SET7-mediated lysine methylation in mammalian cells. Proc. Natl. Acad. Sci. USA 2009, 106, 5076–5081. [Google Scholar] [CrossRef]
  81. Feng, Q.; Zhang, Y. The MeCP1 complex represses transcription through preferential binding, remodeling, and deacetylating methylated nucleosomes. Genes. Dev. 2001, 15, 827–832. [Google Scholar]
  82. Tan, J.Z.; Yan, Y.; Wang, X.X.; Jiang, Y.; Xu, H.E. EZH2: Biology, disease, and structure-based drug discovery. Acta Pharmacol. Sin. 2014, 35, 161–174. [Google Scholar] [CrossRef]
  83. Wahid, F.; Shehzad, A.; Khan, T.; Kim, Y.Y. MicroRNAs: Synthesis, mechanism, function, and recent clinical trials. Biochim. Biophys. Acta 2010, 1803, 1231–1243. [Google Scholar] [CrossRef]
  84. Nzobokela, J.; Muchaili, L.; Mwambungu, A.; Masenga Sepiso, K.; Kirabo, A. Pathophysiology and emerging biomarkers of cardiovascular-renal-hepato-metabolic syndrome. Front. Cardiovasc. Med. 2025, 12, 1661563. [Google Scholar] [CrossRef]
  85. Zou, C. Advances in the study of miRNAs in chronic kidney disease with cardiovascular complications. Front. Physiol. 2023, 14, 1283597. [Google Scholar] [CrossRef]
  86. Mahjoob, G.; Ahmadi, Y.; Fatima Rajani, H.; Khanbabaei, N.; Abolhasani, S. Circulating microRNAs as predictive biomarkers of coronary artery diseases in type 2 diabetes patients. J. Clin. Lab. Anal. 2022, 36, e24380. [Google Scholar] [CrossRef] [PubMed]
  87. Macgregor-Das, A.M.; Das, S. A microRNA’s journey to the center of the mitochondria. Am. J. Physiol. Heart Circ. Physiol. 2018, 315, H206–H215. [Google Scholar] [CrossRef]
  88. Tang, J.; Yan, H.; Zhuang, S. Histone deacetylases as targets for treatment of multiple diseases. Clin. Sci. 2013, 124, 651–662. [Google Scholar] [CrossRef] [PubMed]
  89. Ibanez-Cabellos, J.S.; Pallardo, F.V.; Garcia-Gimenez, J.L.; Seco-Cervera, M. Oxidative Stress and Epigenetics: miRNA Involvement in Rare Autoimmune Diseases. Antioxidants 2023, 12, 800. [Google Scholar] [CrossRef]
  90. Thomas, M.C. Epigenetic Mechanisms in Diabetic Kidney Disease. Curr. Diab Rep. 2016, 16, 31. [Google Scholar] [CrossRef] [PubMed]
  91. Marumo, T.; Yagi, S.; Kawarazaki, W.; Nishimoto, M.; Ayuzawa, N.; Watanabe, A.; Ueda, K.; Hirahashi, J.; Hishikawa, K.; Sakurai, H.; et al. Diabetes Induces Aberrant DNA Methylation in the Proximal Tubules of the Kidney. J. Am. Soc. Nephrol. 2015, 26, 2388–2397. [Google Scholar] [CrossRef]
  92. Mimura, I.; Chen, Z.; Natarajan, R. Epigenetic alterations and memory: Key players in the development/progression of chronic kidney disease promoted by acute kidney injury and diabetes. Kidney Int. 2025, 107, 434–456. [Google Scholar] [CrossRef]
  93. Dwivedi, R.S.; Herman, J.G.; McCaffrey, T.A.; Raj, D.S. Beyond genetics: Epigenetic code in chronic kidney disease. Kidney Int. 2013, 79, 23–32. [Google Scholar] [CrossRef]
  94. Wing, M.R.; Ramezani, A.; Gill, H.S.; Devaney, J.M.; Raj, D.S. Epigenetics of progression of chronic kidney disease: Fact or fantasy? Semin. Nephrol. 2014, 33, 363–374. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, J.; Shen, F.; Liu, F.; Zhuang, S. Histone Modifications in Acute Kidney Injury. Kidney Dis. 2022, 8, 466–477. [Google Scholar] [CrossRef] [PubMed]
  96. Villeneuve, L.M.; Reddy, M.A.; Lanting, L.L.; Wang, M.; Meng, L.; Natarajan, R. Epigenetic histone H3 lysine 9 methylation in metabolic memory and inflammatory phenotype of vascular smooth muscle cells in diabetes. Proc. Natl. Acad. Sci. USA 2008, 105, 9047–9052. [Google Scholar] [CrossRef]
  97. De Marinis, Y.; Cai, M.; Bompada, P.; Atac, D.; Kotova, O.; Johansson, M.E.; Garcia-Vaz, E.; Gomez, M.F.; Laakso, M.; Groop, L. Epigenetic regulation of the thioredoxin-interacting protein (TXNIP) gene by hyperglycemia in kidney. Kidney Int. 2016, 89, 342–353. [Google Scholar] [CrossRef]
  98. Yuan, H.; Reddy, M.A.; Sun, G.; Lanting, L.; Wang, M.; Kato, M.; Natarajan, R. Involvement of p300/CBP and epigenetic histone acetylation in TGF-β1-mediated gene transcription in mesangial cells. Am. J. Physiol. Ren. Physiol. 2013, 304, F601–F613. [Google Scholar] [CrossRef] [PubMed]
  99. Mishra, M.; Kowluru, R.A. The Role of DNA Methylation in the Metabolic Memory Phenomenon Associated With the Continued Progression of Diabetic Retinopathy. Investig. Ophthalmol. Vis. Sci. 2016, 57, 5748–5757. [Google Scholar] [CrossRef]
  100. Liao, Y.; Gou, L.; Chen, L.; Zhong, X.; Zhang, D.; Zhu, H.; Lu, X.; Zeng, T.; Deng, X.; Li, Y. NADPH oxidase 4 and endothelial nitric oxide synthase contribute to endothelial dysfunction mediated by histone methylations in metabolic memory. Free Radic. Biol. Med. 2018, 115, 383–394. [Google Scholar] [CrossRef]
  101. Brasacchio, D.; Okabe, J.; Tikellis, C.; Balcerczyk, A.; George, P.; Baker, E.K.; Calkin, A.C.; Brownlee, M.; Cooper, M.E.; El-Osta, A. Hyperglycemia induces a dynamic cooperativity of histone methylase and demethylase enzymes associated with gene-activating epigenetic marks that coexist on the lysine tail. Diabetes 2009, 58, 1229–1236. [Google Scholar] [CrossRef]
  102. Zeng, S.; Wu, X.; Chen, X.; Xu, H.; Zhang, T.; Xu, Y. Hypermethylated in cancer 1 (HIC1) mediates high glucose induced ROS accumulation in renal tubular epithelial cells by epigenetically repressing SIRT1 transcription. Biochim. Biophys. Acta Gene Regul. Mech. 2018, 1861, 917–927. [Google Scholar] [CrossRef]
  103. Rossi, M.N.; Ciolfi, A.; Matteo, V.; Pedace, L.; Nardini, C.; Loricchio, E.; Caiello, I.; Bellomo, F.; Taranta, A.; De Leo, E.; et al. Genome-wide DNA methylation analysis identifies kidney epigenetic dysregulation in a cystinosis mouse model. Front. Cell Dev. Biol. 2025, 13, 1638123. [Google Scholar] [CrossRef]
  104. Stenvinkel, P.; Ekstrom, T.J. Does the uremic milieu affect the epigenotype? J. Ren. Nutr. 2009, 19, 82–85. [Google Scholar] [CrossRef]
  105. Kato, S.; Lindholm, B.; Stenvinkel, P.; Ekstrom, T.J.; Luttropp, K.; Yuzawa, Y.; Yasuda, Y.; Tsuruta, Y.; Maruyama, S. DNA hypermethylation and inflammatory markers in incident Japanese dialysis patients. Nephron Extra 2012, 2, 159–168. [Google Scholar] [CrossRef] [PubMed]
  106. Wielscher, M.; Mandaviya, P.R.; Kuehnel, B.; Joehanes, R.; Mustafa, R.; Robinson, O.; Zhang, Y.; Bodinier, B.; Walton, E.; Mishra, P.P.; et al. DNA methylation signature of chronic low-grade inflammation and its role in cardio-respiratory diseases. Nat. Commun. 2022, 13, 2408. [Google Scholar] [CrossRef]
  107. Vujcic, S.; Kotur-Stevuljevic, J.; Vujcic, Z.; Stojanovic, S.; Beljic Zivkovic, T.; Vuksanovic, M.; Marjanovic Petkovic, M.; Perovic Blagojevic, I.; Koprivica-Uzelac, B.; Ilic-Mijailovic, S.; et al. Global DNA Methylation in Poorly Controlled Type 2 Diabetes Mellitus: Association with Redox and Inflammatory Biomarkers. Int. J. Mol. Sci. 2025, 26, 6716. [Google Scholar] [CrossRef]
  108. Gluck, C.; Qiu, C.; Han, S.Y.; Palmer, M.; Park, J.; Ko, Y.A.; Guan, Y.; Sheng, X.; Hanson, R.L.; Huang, J.; et al. Kidney cytosine methylation changes improve renal function decline estimation in patients with diabetic kidney disease. Nat. Commun. 2019, 10, 2461. [Google Scholar] [CrossRef] [PubMed]
  109. Ingrosso, D.; Perna, A.F. DNA Methylation Dysfunction in Chronic Kidney Disease. Genes 2020, 11, 811. [Google Scholar] [CrossRef] [PubMed]
  110. Wang, Y.Z.; Xu, W.W.; Zhu, D.Y.; Zhang, N.; Wang, Y.L.; Ding, M.; Xie, X.M.; Sun, L.L.; Wang, X.X. Specific expression network analysis of diabetic nephropathy kidney tissue revealed key methylated sites. J. Cell Physiol. 2018, 233, 7139–7147. [Google Scholar] [CrossRef]
  111. Swan, E.J.; Maxwell, A.P.; McKnight, A.J. Distinct methylation patterns in genes that affect mitochondrial function are associated with kidney disease in blood-derived DNA from individuals with Type 1 diabetes. Diabet. Med. 2015, 32, 1110–1115. [Google Scholar] [CrossRef]
  112. Imai, S.; Guarente, L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 2014, 24, 464–471. [Google Scholar] [CrossRef]
  113. Zheng, Z.; Chen, H.; Li, J.; Li, T.; Zheng, B.; Zheng, Y.; Jin, H.; He, Y.; Gu, Q.; Xu, X. Sirtuin 1-mediated cellular metabolic memory of high glucose via the LKB1/AMPK/ROS pathway and therapeutic effects of metformin. Diabetes 2012, 61, 217–228. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, X.; Gao, Y.; Yi, W.; Qiao, Y.; Hu, H.; Wang, Y.; Hu, Y.; Wu, S.; Sun, H.; Zhang, T. Inhibition of miRNA-155 Alleviates High Glucose-Induced Podocyte Inflammation by Targeting SIRT1 in Diabetic Mice. J. Diabetes Res. 2021, 2021, 5597394. [Google Scholar] [CrossRef]
  115. Orimo, M.; Minamino, T.; Miyauchi, H.; Tateno, K.; Okada, S.; Moriya, J.; Komuro, I. Protective role of SIRT1 in diabetic vascular dysfunction. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 889–894. [Google Scholar] [CrossRef] [PubMed]
  116. Li, Q.; Kim, Y.R.; Vikram, A.; Kumar, S.; Kassan, M.; Gabani, M.; Lee, S.K.; Jacobs, J.S.; Irani, K. P66Shc-Induced MicroRNA-34a Causes Diabetic Endothelial Dysfunction by Downregulating Sirtuin1. Arter. Thromb. Vasc. Biol. 2017, 36, 2394–2403. [Google Scholar] [CrossRef]
  117. Huang, K.; Huang, J.; Xie, X.; Wang, S.; Chen, C.; Shen, X.; Liu, P.; Huang, H. Sirt1 resists advanced glycation end products-induced expressions of fibronectin and TGF-beta1 by activating the Nrf2/ARE pathway in glomerular mesangial cells. Free Radic. Biol. Med. 2013, 65, 528–540. [Google Scholar] [CrossRef]
  118. Koyama, T.; Kume, S.; Koya, D.; Araki, S.; Isshiki, K.; Chin-Kanasaki, M.; Sugimoto, T.; Haneda, M.; Sugaya, T.; Kashiwagi, A.; et al. SIRT3 attenuates palmitate-induced ROS production and inflammation in proximal tubular cells. Free Radic. Biol. Med. 2011, 51, 1258–1267. [Google Scholar] [CrossRef]
  119. Juszczak, F.; Arnould, T.; Decleves, A.E. The Role of Mitochondrial Sirtuins (SIRT3, SIRT4 and SIRT5) in Renal Cell Metabolism: Implication for Kidney Diseases. Int. J. Mol. Sci. 2024, 25, 6936. [Google Scholar] [CrossRef]
  120. Dikalova, A.E.; Itani, H.A.; Nazarewicz, R.R.; McMaster, W.G.; Flynn, C.R.; Uzhachenko, R.; Fessel, J.P.; Gamboa, J.L.; Harrison, D.G.; Dikalov, S.I. Sirt3 Impairment and SOD2 Hyperacetylation in Vascular Oxidative Stress and Hypertension. Circ. Res. 2017, 121, 564–574. [Google Scholar] [CrossRef]
  121. Dikalova, S.; Dikalova, A. Mitochondrial deacetylase Sirt3 in vascular dysfunction and hypertension. Curr. Opin. Nephrol. Hypertens. 2024, 31, 151–156. [Google Scholar] [CrossRef] [PubMed]
  122. Pacher, P.; Szabo, C. Role of poly(ADP-ribose) polymerase-1 activation in the pathogenesis of diabetic complications: Endothelial dysfunction, as a common underlying theme. Antioxid. Redox Signal 2008, 7, 1568–1580. [Google Scholar]
  123. Kanasaki, K.; Shi, S.; Kanasaki, M.; He, J.; Nagai, T.; Nakamura, Y.; Ishigaki, Y.; Kitada, M.; Srivastava, S.P.; Koya, D. Linagliptin-mediated DPP-4 inhibition ameliorates kidney fibrosis in streptozotocin-induced diabetic mice by inhibiting endothelial-to-mesenchymal transition in a therapeutic regimen. Diabetes 2014, 63, 2120–2131. [Google Scholar] [CrossRef]
  124. Wang, X.; Lin, B.; Nie, L.; Li, P. microRNA-20b contributes to high glucose-induced podocyte apoptosis by targeting SIRT7. Mol. Med. Rep. 2017, 16, 5667–5674. [Google Scholar] [CrossRef] [PubMed]
  125. Jin, Q.; Ma, F.; Liu, T.; Yang, L.; Mao, H.; Wang, Y.; Peng, L.; Li, P.; Zhan, Y. Sirtuins in kidney diseases: Potential mechanism and therapeutic targets. Cell Commun. Signal 2024, 22, 114. [Google Scholar]
  126. Zhang, Y.; Zhang, C.; Zhang, H.; Zeng, W.; Li, S.; Chen, C.; Song, X.; Sun, J.; Sun, Z.; Cui, C.; et al. ZIPK mediates endothelial cell contraction through myosin light chain phosphorylation and is required for ischemic-reperfusion injury. FASEB J. 2019, 33, 9062–9074. [Google Scholar] [CrossRef]
  127. Li, X.; Liu, J.; Lu, L.; Huang, T.; Hou, W.; Wang, F.; Yu, L.; Wu, F.; Qi, J.; Chen, X.; et al. Sirt7 associates with ELK1 to participate in hyperglycemia memory and diabetic nephropathy via modulation of DAPK3 expression and endothelial inflammation. Transl. Res. 2022, 247, 99–116. [Google Scholar] [CrossRef]
  128. Wegner, M.; Neddermann, D.; Piorunska-Stolzmann, M.; Jagodzinski, P.P. Role of epigenetic mechanisms in the development of chronic complications of diabetes. Diabetes Res. Clin. Pract. 2014, 105, 164–175. [Google Scholar] [CrossRef]
  129. Yang, Y.; Wang, Y.; Fan, X.; Xu, X.; Wang, H.; Wang, X.; Shi, T.; Tang, J.; Guan, Y.; Li, S.; et al. Role of DNA methylation transferase in urinary system diseases: From basic to clinical perspectives (Review). Int. J. Mol. Med. 2025, 55, 19. [Google Scholar] [CrossRef] [PubMed]
  130. Yao, Y.; Song, Q.; Hu, C.; Da, X.; Yu, Y.; He, Z.; Xu, C.; Chen, Q.; Wang, Q.K. Endothelial cell metabolic memory causes cardiovascular dysfunction in diabetes. Cardiovasc. Res. 2022, 118, 196–211. [Google Scholar] [PubMed]
  131. Pedruzzi, L.M.; Cardozo, L.F.; Daleprane, J.B.; Stockler-Pinto, M.B.; Monteiro, E.B.; Leite, M., Jr.; Vaziri, N.D.; Mafra, D. Systemic inflammation and oxidative stress in hemodialysis patients are associated with down-regulation of Nrf2. J. Nephrol. 2015, 28, 495–501. [Google Scholar] [CrossRef]
  132. Bondi, C.D.; Hartman, H.L.; Tan, R.J. NRF2 in kidney physiology and disease. Physiol. Rep. 2024, 12, e15961. [Google Scholar] [CrossRef]
  133. McClelland, A.D.; Kantharidis, P. microRNA in the development of diabetic complications. Clin. Sci. 2014, 126, 95–110. [Google Scholar] [CrossRef] [PubMed]
  134. Prattichizzo, F.; Giuliani, A.; De Nigris, V.; Pujadas, G.; Ceka, A.; La Sala, L.; Genovese, S.; Testa, R.; Procopio, A.D.; Olivieri, F.; et al. Extracellular microRNAs and endothelial hyperglycaemic memory: A therapeutic opportunity? Diabetes Obes. Metab. 2016, 18, 855–867. [Google Scholar] [CrossRef] [PubMed]
  135. Jin, J.; Wang, X.; Zhi, X.; Meng, D. Epigenetic regulation in diabetic vascular complications. J. Mol. Endocrinol. 2019, 63, R103–R115. [Google Scholar] [CrossRef]
  136. Zhong, X.; Liao, Y.; Chen, L.; Liu, G.; Feng, Y.; Zeng, T.; Zhang, J. The MicroRNAs in the Pathogenesis of Metabolic Memory. Endocrinology 2015, 156, 3157–3168. [Google Scholar] [CrossRef]
  137. Bera, A.; Das, F.; Ghosh-Choudhury, N.; Mariappan, M.M.; Kasinath, B.S.; Ghosh Choudhury, G. Reciprocal regulation of miR-214 and PTEN by high glucose regulates renal glomerular mesangial and proximal tubular epithelial cell hypertrophy and matrix expansion. Am. J. Physiol. Cell Physiol. 2017, 313, C430–C447. [Google Scholar] [CrossRef]
  138. Zhang, Y.; Zhao, S.; Wu, D.; Liu, X.; Shi, M.; Wang, Y.; Zhang, F.; Ding, J.; Xiao, Y.; Guo, B. MicroRNA-22 Promotes Renal Tubulointerstitial Fibrosis by Targeting PTEN and Suppressing Autophagy in Diabetic Nephropathy. J. Diabetes Res. 2018, 2018, 4728645. [Google Scholar] [CrossRef]
  139. Wang, J.Y.; Gao, Y.B.; Zhang, N.; Zou, D.W.; Wang, P.; Zhu, Z.Y.; Li, J.Y.; Zhou, S.N.; Wang, S.C.; Wang, Y.Y.; et al. miR-21 overexpression enhances TGF-β1-induced epithelial-to-mesenchymal transition by target smad7 and aggravates renal damage in diabetic nephropathy. Mol. Cell Endocrinol. 2014, 392, 163–172. [Google Scholar] [CrossRef]
  140. Kato, M.; Zhang, J.; Wang, M.; Lanting, L.; Yuan, H.; Rossi, J.J.; Natarajan, R. MicroRNA-192 in diabetic kidney glomeruli and its function in TGF-beta-induced collagen expression via inhibition of E-box repressors. Proc. Natl. Acad. Sci. USA 2007, 104, 3432–3437. [Google Scholar] [CrossRef] [PubMed]
  141. Barutta, F.; Bellini, S.; Mastrocola, R.; Bruno, G.; Gruden, G. MicroRNA and Microvascular Complications of Diabetes. Int. J. Endocrinol. 2018, 2018, 6890501. [Google Scholar] [CrossRef]
  142. da Cunha, R.S.D.; Azevedo, C.A.B.; Miniskiskosky, G.; Gregório, P.C.; Stinghen, A.E.M. MicroRNAs and vascular damage in chronic kidney disease: Advances and clinical implications. J. Bras. Nefrol. 2025, 47, e20240223. [Google Scholar] [CrossRef] [PubMed]
  143. Satake, E.; Pezzolesi, M.G.; Md Dom, Z.I.; Smiles, A.M.; Niewczas, M.A.; Krolewski, A.S. Circulating miRNA Profiles Associated With Hyperglycemia in Patients With Type 1 Diabetes. Diabetes 2018, 67, 1013–1023. [Google Scholar] [CrossRef] [PubMed]
  144. Ding, M.; Shi, R.; Du, Y.; Chang, P.; Gao, T.; De, D.; Chen, Y.; Li, M.; Li, J.; Li, K.; et al. O-GlcNAcylation-mediated endothelial metabolic memory contributes to cardiac damage via small extracellular vesicles. Cell Metab. 2025, 37, 1344–1363 e6. [Google Scholar] [CrossRef]
  145. Intine, R.V.; Sarras, M.P., Jr. Metabolic memory and chronic diabetes complications: Potential role for epigenetic mechanisms. Curr. Diab Rep. 2013, 12, 551–559. [Google Scholar] [CrossRef] [PubMed]
  146. Yang, A.Y.; Kim, H.; Li, W.; Kong, A.N. Natural compound-derived epigenetic regulators targeting epigenetic readers, writers and erasers. Curr. Top. Med. Chem. 2017, 16, 697–713. [Google Scholar] [CrossRef]
  147. Biswas, S.; Rao, C.M. Epigenetic tools (The Writers, The Readers and The Erasers) and their implications in cancer therapy. Eur. J. Pharmacol. 2018, 837, 8–24. [Google Scholar] [CrossRef]
  148. Contieri, B.; Duarte, B.K.L.; Lazarini, M. Updates on DNA methylation modifiers in acute myeloid leukemia. Ann. Hematol. 2020, 99, 693–701. [Google Scholar] [CrossRef]
  149. Pinto, V.; Bergantim, R.; Caires, H.R.; Seca, H.; Guimaraes, J.E.; Vasconcelos, M.H. Multiple Myeloma: Available Therapies and Causes of Drug Resistance. Cancers 2020, 12, 407. [Google Scholar] [CrossRef]
  150. Damiani, E.; Duran, M.N.; Mohan, N.; Rajendran, P.; Dashwood, R.H. Targeting Epigenetic ‘Readers’ with Natural Compounds for Cancer Interception. J. Cancer Prev. 2020, 25, 189–203. [Google Scholar] [CrossRef]
  151. Arce, C.; Perez-Plasencia, C.; Gonzalez-Fierro, A.; de la Cruz-Hernandez, E.; Revilla-Vazquez, A.; Chavez-Blanco, A.; Trejo-Becerril, C.; Perez-Cardenas, E.; Taja-Chayeb, L.; Bargallo, E.; et al. A proof-of-principle study of epigenetic therapy added to neoadjuvant doxorubicin cyclophosphamide for locally advanced breast cancer. PLoS ONE 2006, 1, e98. [Google Scholar] [CrossRef]
  152. Candelaria, M.; Gallardo-Rincon, D.; Arce, C.; Cetina, L.; Aguilar-Ponce, J.L.; Arrieta, O.; Gonzalez-Fierro, A.; Chavez-Blanco, A.; de la Cruz-Hernandez, E.; Camargo, M.F.; et al. A phase II study of epigenetic therapy with hydralazine and magnesium valproate to overcome chemotherapy resistance in refractory solid tumors. Ann. Oncol. 2007, 18, 1529–1538. [Google Scholar] [CrossRef]
  153. Coronel, J.; Cetina, L.; Pacheco, I.; Trejo-Becerril, C.; Gonzalez-Fierro, A.; de la Cruz-Hernandez, E.; Perez-Cardenas, E.; Taja-Chayeb, L.; Arias-Bofill, D.; Candelaria, M.; et al. A double-blind, placebo-controlled, randomized phase III trial of chemotherapy plus epigenetic therapy with hydralazine valproate for advanced cervical cancer. Preliminary results. Med. Oncol. 2011, 28 (Suppl. 1), S540–S546. [Google Scholar] [CrossRef]
  154. Candelaria, M.; Herrera, A.; Labardini, J.; Gonzalez-Fierro, A.; Trejo-Becerril, C.; Taja-Chayeb, L.; Perez-Cardenas, E.; de la Cruz-Hernandez, E.; Arias-Bofill, D.; Vidal, S.; et al. Hydralazine and magnesium valproate as epigenetic treatment for myelodysplastic syndrome. Preliminary results of a phase-II trial. Ann. Hematol. 2011, 90, 379–387. [Google Scholar] [CrossRef]
  155. Heerboth, S.; Lapinska, K.; Snyder, N.; Leary, M.; Rollinson, S.; Sarkar, S. Use of epigenetic drugs in disease: An overview. Genet. Epigenetics 2014, 6, 9–19. [Google Scholar] [CrossRef]
  156. Farsetti, A.; Illi, B.; Gaetano, C. How epigenetics impacts on human diseases. Eur. J. Intern. Med. 2023, 114, 15–22. [Google Scholar] [CrossRef] [PubMed]
  157. Zhao, H.; Han, Z.; Ji, X.; Luo, Y. Epigenetic Regulation of Oxidative Stress in Ischemic Stroke. Aging Dis. 2016, 7, 295–306. [Google Scholar] [CrossRef] [PubMed]
  158. Guzik, T.J.; Touyz, R.M. Oxidative Stress, Inflammation, and Vascular Aging in Hypertension. Hypertension 2017, 70, 660–667. [Google Scholar] [CrossRef] [PubMed]
  159. Marchant, V.; Trionfetti, F.; Tejedor-Santamaria, L.; Rayego-Mateos, S.; Rotili, D.; Bontempi, G.; Domenici, A.; Mene, P.; Mai, A.; Martin-Cleary, C.; et al. BET Protein Inhibitor JQ1 Ameliorates Experimental Peritoneal Damage by Inhibition of Inflammation and Oxidative Stress. Antioxidants 2023, 12, 2055. [Google Scholar] [CrossRef]
  160. Fraile-Martinez, O.; De Leon-Oliva, D.; Boaru, D.L.; De Castro-Martinez, P.; Garcia-Montero, C.; Barrena-Blazquez, S.; Garcia-Garcia, J.; Garcia-Honduvilla, N.; Alvarez-Mon, M.; Lopez-Gonzalez, L.; et al. Connecting epigenetics and inflammation in vascular senescence: State of the art, biomarkers and senotherapeutics. Front. Genet. 2024, 15, 1345459. [Google Scholar] [CrossRef]
  161. Chen, Z.; Malek, V.; Natarajan, R. Update: The role of epigenetics in the metabolic memory of diabetic complications. Am. J. Physiol. Renal Physiol. 2024, 327, F327–F339. [Google Scholar] [CrossRef]
  162. 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]
  163. 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]
  164. Soriano-Tarraga, C.; Jimenez-Conde, J.; Giralt-Steinhauer, E.; Mola-Caminal, M.; Vivanco-Hidalgo, R.M.; Ois, A.; Rodriguez-Campello, A.; Cuadrado-Godia, E.; Sayols-Baixeras, S.; Elosua, R.; et al. Epigenome-wide association study identifies TXNIP gene associated with type 2 diabetes mellitus and sustained hyperglycemia. Hum. Mol. Genet. 2016, 25, 609–619. [Google Scholar] [CrossRef]
  165. Cooper, M.E.; El-Osta, A.; Allen, T.J.; Watson, A.M.D.; Thomas, M.C.; Jandeleit-Dahm, K.A.M. Metabolic Karma-The Atherogenic Legacy of Diabetes: The 2017 Edwin Bierman Award Lecture. Diabetes 2018, 67, 785–790. [Google Scholar] [CrossRef]
  166. Chen, Z.; Natarajan, R. Epigenetic modifications in metabolic memory: What are the memories, and can we erase them? Am. J. Physiol. Cell Physiol. 2022, 323, C570–C582. [Google Scholar] [CrossRef] [PubMed]
  167. Zhang, X.; Zhao, S.; Yuan, Q.; Zhu, L.; Li, F.; Wang, H.; Kong, D.; Hao, J. TXNIP, a novel key factor to cause Schwann cell dysfunction in diabetic peripheral neuropathy, under the regulation of PI3K/Akt pathway inhibition-induced DNMT1 and DNMT3a overexpression. Cell Death Dis. 2021, 12, 642. [Google Scholar] [CrossRef]
  168. Singh, L.P. Thioredoxin Interacting Protein (TXNIP) and Pathogenesis of Diabetic Retinopathy. J. Clin. Exp. Ophthalmol. 2013, 4, 4172. [Google Scholar] [CrossRef] [PubMed]
  169. Garcia-Calzon, S.; Maguolo, A.; Eichelmann, F.; Edsfeldt, A.; Perfilyev, A.; Maziarz, M.; Lindstrom, A.; Sun, J.; Briviba, M.; Schulze, M.B.; et al. Epigenetic biomarkers predict macrovascular events in individuals with type 2 diabetes. Cell Rep. Med. 2025, 6, 102290. [Google Scholar] [CrossRef] [PubMed]
  170. Park, J.; Guan, Y.; Sheng, X.; Gluck, C.; Seasock, M.J.; Hakimi, A.A.; Qiu, C.; Pullman, J.; Verma, A.; Li, H.; et al. Functional methylome analysis of human diabetic kidney disease. JCI Insight 2019, 4, e128886. [Google Scholar] [CrossRef]
  171. Smyth, L.J.; Patterson, C.C.; Swan, E.J.; Maxwell, A.P.; McKnight, A.J. DNA Methylation Associated with Diabetic Kidney Disease in Blood-Derived DNA. Front. Cell Dev. Biol. 2020, 8, 561907. [Google Scholar] [CrossRef]
  172. Smyth, L.J.; Dahlstrom, E.H.; Syreeni, A.; Kerr, K.; Kilner, J.; Doyle, R.; Brennan, E.; Nair, V.; Fermin, D.; Nelson, R.G.; et al. Epigenome-wide meta-analysis identifies DNA methylation biomarkers associated with diabetic kidney disease. Nat. Commun. 2022, 13, 7891. [Google Scholar] [CrossRef]
  173. Marchiori, M.; Maguolo, A.; Perfilyev, A.; Maziarz, M.; Martinell, M.; Gomez, M.F.; Ahlqvist, E.; Garcia-Calzon, S.; Ling, C. Blood-Based Epigenetic Biomarkers Associated with Incident Chronic Kidney Disease in Individuals with Type 2 Diabetes. Diabetes 2025, 74, 439–450. [Google Scholar] [CrossRef]
  174. Akhouri, V.; Majumder, S.; Gaikwad, A.B. Targeting DNA methylation in diabetic kidney disease: A new perspective. Life Sci. 2023, 335, 122256. [Google Scholar] [CrossRef] [PubMed]
  175. Chen, G.; Chen, H.; Ren, S.; Xia, M.; Zhu, J.; Liu, Y.; Zhang, L.; Tang, L.; Sun, L.; Liu, H.; et al. Aberrant DNA methylation of mTOR pathway genes promotes inflammatory activation of immune cells in diabetic kidney disease. Kidney Int. 2019, 96, 409–420. [Google Scholar] [CrossRef] [PubMed]
  176. Al-Dabet, M.M.; Shahzad, K.; Elwakiel, A.; Sulaj, A.; Kopf, S.; Bock, F.; Gadi, I.; Zimmermann, S.; Rana, R.; Krishnan, S.; et al. Reversal of the renal hyperglycemic memory in diabetic kidney disease by targeting sustained tubular p21 expression. Nat. Commun. 2020, 13, 5062. [Google Scholar] [CrossRef]
  177. Feng, S.; Peden, E.K.; Guo, Q.; Lee, T.H.; Li, Q.; Yuan, Y.; Chen, C.; Huang, F.; Cheng, J. Downregulation of the endothelial histone demethylase JMJD3 is associated with neointimal hyperplasia of arteriovenous fistulas in kidney failure. J. Biol. Chem. 2022, 298, 101816. [Google Scholar] [CrossRef]
  178. Sanchez-Ceinos, J.; Hussain, S.; Khan, A.W.; Zhang, L.; Almahmeed, W.; Pernow, J.; Cosentino, F. Repressive H3K27me3 drives hyperglycemia-induced oxidative and inflammatory transcriptional programs in human endothelium. Cardiovasc. Diabetol. 2024, 23, 122. [Google Scholar] [CrossRef] [PubMed]
  179. Li, T.; Yu, C.; Zhuang, S. Histone Methyltransferase EZH2: A Potential Therapeutic Target for Kidney Diseases. Front. Physiol. 2021, 12, 640700. [Google Scholar] [CrossRef]
  180. Siddiqi, F.S.; Majumder, S.; Thai, K.; Abdalla, M.; Hu, P.; Advani, S.L.; White, K.E.; Bowskill, B.B.; Guarna, G.; Dos Santos, C.C.; et al. The Histone Methyltransferase Enzyme Enhancer of Zeste Homolog 2 Protects against Podocyte Oxidative Stress and Renal Injury in Diabetes. J. Am. Soc. Nephrol. 2015, 27, 2021–2034. [Google Scholar] [CrossRef]
  181. Sekine, S.; Youle, R.J. PINK1 import regulation; a fine system to convey mitochondrial stress to the cytosol. BMC Biol. 2018, 16, 2. [Google Scholar] [CrossRef]
  182. Mu, J.; Zhang, D.; Tian, Y.; Xie, Z.; Zou, M.H. BRD4 inhibition by JQ1 prevents high-fat diet-induced diabetic cardiomyopathy by activating PINK1/Parkin-mediated mitophagy in vivo. J. Mol. Cell Cardiol. 2021, 149, 1–14. [Google Scholar] [CrossRef] [PubMed]
  183. Ray, K.K.; Nicholls, S.J.; Buhr, K.A.; Ginsberg, H.N.; Johansson, J.O.; Kalantar-Zadeh, K.; Kulikowski, E.; Toth, P.P.; Wong, N.; Sweeney, M.; et al. Effect of Apabetalone Added to Standard Therapy on Major Adverse Cardiovascular Events in Patients With Recent Acute Coronary Syndrome and Type 2 Diabetes: A Randomized Clinical Trial. JAMA 2020, 323, 1565–1573. [Google Scholar] [CrossRef]
  184. Brandts, J.; Ray, K.K. Apabetalone—BET protein inhibition in cardiovascular disease and Type 2 diabetes. Future Cardiol. 2020, 16, 385–395. [Google Scholar] [CrossRef]
  185. Sasaki, K.; Masaki, T. Epigenetic histone modifications in kidney disease and epigenetic memory. Clin. Exp. Nephrol. 2025, 29, 1129–1138. [Google Scholar] [CrossRef]
  186. Liu, M.; Qiao, Z.; Zhang, Y.; Zhan, P.; Yi, F. Histone Deacetylases Take Center Stage on Regulation of Podocyte Function. Kidney Dis. 2020, 6, 236–246, Erratum in Kidney Dis. 2020, 6, 382–383. [Google Scholar] [CrossRef]
  187. Khan, S.; Jena, G.; Tikoo, K.; Kumar, V. Valproate attenuates the proteinuria, podocyte and renal injury by facilitating autophagy and inactivation of NF-kappaB/iNOS signaling in diabetic rat. Biochimie 2015, 110, 1–16. [Google Scholar] [CrossRef]
  188. Yoshikawa, M.; Hishikawa, K.; Marumo, T.; Fujita, T. Inhibition of histone deacetylase activity szouuppresses epithelial-to-mesenchymal transition induced by TGF-beta1 in human renal epithelial cells. J. Am. Soc. Nephrol. 2007, 18, 58–65. [Google Scholar] [CrossRef]
  189. Huang, K.; Gao, X.; Wei, W. The crosstalk between Sirt1 and Keap1/Nrf2/ARE anti-oxidative pathway forms a positive feedback loop to inhibit FN and TGF-beta1 expressions in rat glomerular mesangial cells. Exp. Cell Res. 2017, 361, 63–72. [Google Scholar] [CrossRef]
  190. Martinez-Arroyo, O.; Flores-Chova, A.; Mendez-Debaets, M.; Garcia-Ferran, L.; Escriva, L.; Forner, M.J.; Redon, J.; Cortes, R.; Ortega, A. Inhibiting miR-200a-3p Increases Sirtuin 1 and Mitigates Kidney Injury in a Tubular Cell Model of Diabetes and Hypertension-Related Renal Damage. Biomolecules 2025, 15, 995. [Google Scholar] [CrossRef] [PubMed]
  191. Chen, H.Y.; Zhong, X.; Huang, X.R.; Meng, X.M.; You, Y.; Chung, A.C.; Lan, H.Y. MicroRNA-29b inhibits diabetic nephropathy in db/db mice. Mol. Ther. 2014, 22, 842–853. [Google Scholar] [CrossRef] [PubMed]
  192. Gondaliya, P.; Dasare, A.; Srivastava, A.; Kalia, K. miR29b regulates aberrant methylation in In-Vitro diabetic nephropathy model of renal proximal tubular cells. PLoS ONE 2018, 13, e0208044, Erratum in PLoS ONE, 2019, 14, e0211591. [Google Scholar] [CrossRef]
  193. Putta, S.; Lanting, L.; Sun, G.; Lawson, G.; Kato, M.; Natarajan, R. Inhibiting microRNA-192 ameliorates renal fibrosis in diabetic nephropathy. J. Am. Soc. Nephrol. 2012, 23, 458–469. [Google Scholar] [CrossRef]
  194. Dhas, Y.; Arshad, N.; Biswas, N.; Jones, L.D.; Ashili, S. MicroRNA-21 Silencing in Diabetic Nephropathy: Insights on Therapeutic Strategies. Biomedicines 2023, 11, 2583. [Google Scholar] [CrossRef]
  195. de Gonzalo-Calvo, D.; van der Meer, R.W.; Rijzewijk, L.J.; Smit, J.W.; Revuelta-Lopez, E.; Nasarre, L.; Escola-Gil, J.C.; Lamb, H.J.; Llorente-Cortes, V. Serum microRNA-1 and microRNA-133a levels reflect myocardial steatosis in uncomplicated type 2 diabetes. Sci. Rep. 2017, 7, 47. [Google Scholar] [CrossRef]
  196. Napoli, C.; Benincasa, G.; Schiano, C.; Salvatore, M. Differential epigenetic factors in the prediction of cardiovascular risk in diabetic patients. Eur. Heart J. Cardiovasc. Pharmacother. 2020, 6, 239–247. [Google Scholar] [CrossRef]
  197. Al-Kafaji, G.; Al-Muhtaresh, H.A. Expression of microRNA377 and microRNA192 and their potential as blood-based biomarkers for early detection of type 2 diabetic nephropathy. Mol. Med. Rep. 2018, 18, 1171–1180. [Google Scholar]
  198. De, A.; Sarkar, A.; Banerjee, T.; Bhowmik, R.; Sar, S.; Shaharyar, M.A.; Karmakar, S.; Ghosh, N. MicroRNA: Unveiling novel mechanistic and theranostic pathways in diabetic cardiomyopathy. Front. Pharmacol. 2025, 16, 1613844. [Google Scholar] [CrossRef] [PubMed]
  199. Yousri, N.A.; Albagha, O.M.E.; Hunt, S.C. Integrated epigenome, whole genome sequence and metabolome analyses identify novel multi-omics pathways in type 2 diabetes: A Middle Eastern study. BMC Med. 2023, 21, 347. [Google Scholar] [CrossRef] [PubMed]
  200. Zhou, L.; Mei, S.; Ma, X.; Wuyun, Q.; Cai, Z.; Chen, C.; Ding, H.; Yan, J. Multi-omics insights into the pathogenesis of diabetic cardiomyopathy: Epigenetic and metabolic profiles. Epigenomics 2025, 17, 33–48. [Google Scholar] [CrossRef] [PubMed]
  201. Zhang, M.; Su, Q.; Lu, Y.; Zhao, M.; Niu, B. Application of machine learning approaches for protein-protein interactions prediction. Med. Chem. 2017, 13, 506–514. [Google Scholar] [CrossRef]
Figure 1. Molecular pathophysiology of hyperglycemic memory in the development of vascular complications. The process is principally driven by four arms. (A) Initiation by formation of Advanced Glycation End Products: hyperglycemia promotes the non-enzymatic formation of advanced end products, followed by the binding to their specific receptor, subsequently activating a cascade of pro-inflammatory and pro-oxidant signals; (B) ROS overproduction and mitochondrial damage: mitochondrial dysfunctions, characterized by increased fission, decreased fusion, impaired quality control via mitophagy, and compromised respiratory efficiency are induced and result in the overproduction of reactive oxygen species; (C) Epigenetic pathological modification: the cumulative cellular stress from AGE overproduction and excessive reactive oxygen species induce an epigenetic reprogramming, including alteration of DNA methylation, aberrant post-translational histone modifications, and dysregulation of non-coding RNAs; (D) Persistent altered gene expression: transient or sustained hyperglycemia induces durable cellular and epigenetic reprogramming characterized by endoplasmic reticulum stress and the chronic activation of transcription factors, which drives the expression of pro-inflammatory mediators, promotes extracellular matrix remodeling, and increases endothelium stiffness. AGE: Advanced Glycation End Products; RAGE: Receptor for Advanced Glycation End Products; mtDNA: mitochondrial DNA; ROS: reactive oxygen species; DNMT: DNA methyltransferases; ncRNA: non-coding RNA; ER: endoplasmic reticulum; ECM: extracellular matrix; ↓: increase; ↑: reduction. Created with Biorender.com.
Figure 1. Molecular pathophysiology of hyperglycemic memory in the development of vascular complications. The process is principally driven by four arms. (A) Initiation by formation of Advanced Glycation End Products: hyperglycemia promotes the non-enzymatic formation of advanced end products, followed by the binding to their specific receptor, subsequently activating a cascade of pro-inflammatory and pro-oxidant signals; (B) ROS overproduction and mitochondrial damage: mitochondrial dysfunctions, characterized by increased fission, decreased fusion, impaired quality control via mitophagy, and compromised respiratory efficiency are induced and result in the overproduction of reactive oxygen species; (C) Epigenetic pathological modification: the cumulative cellular stress from AGE overproduction and excessive reactive oxygen species induce an epigenetic reprogramming, including alteration of DNA methylation, aberrant post-translational histone modifications, and dysregulation of non-coding RNAs; (D) Persistent altered gene expression: transient or sustained hyperglycemia induces durable cellular and epigenetic reprogramming characterized by endoplasmic reticulum stress and the chronic activation of transcription factors, which drives the expression of pro-inflammatory mediators, promotes extracellular matrix remodeling, and increases endothelium stiffness. AGE: Advanced Glycation End Products; RAGE: Receptor for Advanced Glycation End Products; mtDNA: mitochondrial DNA; ROS: reactive oxygen species; DNMT: DNA methyltransferases; ncRNA: non-coding RNA; ER: endoplasmic reticulum; ECM: extracellular matrix; ↓: increase; ↑: reduction. Created with Biorender.com.
Genes 16 01442 g001
Figure 2. Schematic representation of the organization and packaging of elements of the nucleosome. (A) Nucleosome as the fundamental unit of the epigenome: the structure of the nucleosome consists of an octamer of core histone proteins (two copies each of H2A, H2B, H3, and H4) and the H1 linker histone, around which DNA is wrapped. Histone tails undergo post-translational modifications at different amino acid residues and biochemical groups added at the N-terminal region are represented (e.g., acetyl, methyl, and phosphoryl group); (B) Dynamics of DNA methylation: enzymatic processes governing DNA methylation and demethylation. DNA methyltransferases (DNMT1, DNMT3A, DNMT3B) catalyze the addition of a methyl group to the 5′ position of cytosine, forming 5-methylcytosine (5-mC); conversely, the ten-eleven translocation family of dioxygenases initiates active demethylation by oxidizing 5-mC to 5-hydroxymethylcytosine; (C) Chromatin architecture and function as a consequence of epigenetic marks deposition: covalent modifications of histone tails can shape chromatin conformation, impacting on gene expression. The acetylation of lysine residues break down the affinity between positive charge of lysines and negative charge of DNA, inducing relaxation of the chromatin structure and facilitating gene transcription; conversely, DNA methylation enhances affinity between DNA and histones, resulting in condensed chromatin (in form of heterochromatin) and inactivation of the transcription. SAM: S-adenosyl-methionine; TET: ten-eleven translocation. Created with Biorender.com.
Figure 2. Schematic representation of the organization and packaging of elements of the nucleosome. (A) Nucleosome as the fundamental unit of the epigenome: the structure of the nucleosome consists of an octamer of core histone proteins (two copies each of H2A, H2B, H3, and H4) and the H1 linker histone, around which DNA is wrapped. Histone tails undergo post-translational modifications at different amino acid residues and biochemical groups added at the N-terminal region are represented (e.g., acetyl, methyl, and phosphoryl group); (B) Dynamics of DNA methylation: enzymatic processes governing DNA methylation and demethylation. DNA methyltransferases (DNMT1, DNMT3A, DNMT3B) catalyze the addition of a methyl group to the 5′ position of cytosine, forming 5-methylcytosine (5-mC); conversely, the ten-eleven translocation family of dioxygenases initiates active demethylation by oxidizing 5-mC to 5-hydroxymethylcytosine; (C) Chromatin architecture and function as a consequence of epigenetic marks deposition: covalent modifications of histone tails can shape chromatin conformation, impacting on gene expression. The acetylation of lysine residues break down the affinity between positive charge of lysines and negative charge of DNA, inducing relaxation of the chromatin structure and facilitating gene transcription; conversely, DNA methylation enhances affinity between DNA and histones, resulting in condensed chromatin (in form of heterochromatin) and inactivation of the transcription. SAM: S-adenosyl-methionine; TET: ten-eleven translocation. Created with Biorender.com.
Genes 16 01442 g002
Figure 3. The central role of hyperglycemia-induced changes in driving the progression of chronic kidney disease. Transient or persistent hyperglycemia acts as the primum movens to establish the memory phenomenon and to initiate the cascade of downstream pathological events, centered around mitochondrial, endothelial, and vascular dysfunctions. At the cellular level, high glucose impacts on mitochondrial functions, inducing overproduction of reactive oxygen species through oxidative phosphorylation, resulting in fission/fusion imbalance and damage to the mitochondrial DNA. These processes are regulated by an epigenetic memory, able to create changes in DNA methylation, post-translational modification (e.g., Sirtuin activation/repression), and miRNA deregulations. Hyperglycemia also promotes endothelial dysfunctions, with activation of inflammatory pathways with involvement of molecular and epigenetic targets, resulting in imbalance in vascular homeostasis. The initial stimulus, coupled with endothelial and mitochondrial dysfunction, triggers a vicious cycle within the kidney, producing oxidative stress, accumulation of nephrotoxins, and chronic inflammation. These factors could contribute to the progression of chronic kidney disease, with fibrosis, senescence, apoptosis, ultimately predisposing to the onset of end-stage renal disease. Moreover, the systemic nature of endothelial dysfunction gives rise to widespread vascular complications, classified in microvascular and macrovascular complications, such as retinopathy and artery disease. The dysfunction of cardiomyocytes and of vascular smooth muscle cells can be involved, finally, in structural damages and adverse cardiac outcomes, such as atherosclerotic cardiovascular disease, left ventricular hypertrophy, and heart failure. CKD, chronic kidney disease; ESRD, end-stage renal disease; DNMT, DNA methyltransferase; mtDNA, mitochondrial DNA; ROS, reactive oxygen species; TCA, tricarboxylic acid; ASCVD, atherosclerotic cardiovascular disease; LVH, left ventricular hypertrophy; HF, heart failure; ↓: increase; ↑: reduction. Created with Biorender.com.
Figure 3. The central role of hyperglycemia-induced changes in driving the progression of chronic kidney disease. Transient or persistent hyperglycemia acts as the primum movens to establish the memory phenomenon and to initiate the cascade of downstream pathological events, centered around mitochondrial, endothelial, and vascular dysfunctions. At the cellular level, high glucose impacts on mitochondrial functions, inducing overproduction of reactive oxygen species through oxidative phosphorylation, resulting in fission/fusion imbalance and damage to the mitochondrial DNA. These processes are regulated by an epigenetic memory, able to create changes in DNA methylation, post-translational modification (e.g., Sirtuin activation/repression), and miRNA deregulations. Hyperglycemia also promotes endothelial dysfunctions, with activation of inflammatory pathways with involvement of molecular and epigenetic targets, resulting in imbalance in vascular homeostasis. The initial stimulus, coupled with endothelial and mitochondrial dysfunction, triggers a vicious cycle within the kidney, producing oxidative stress, accumulation of nephrotoxins, and chronic inflammation. These factors could contribute to the progression of chronic kidney disease, with fibrosis, senescence, apoptosis, ultimately predisposing to the onset of end-stage renal disease. Moreover, the systemic nature of endothelial dysfunction gives rise to widespread vascular complications, classified in microvascular and macrovascular complications, such as retinopathy and artery disease. The dysfunction of cardiomyocytes and of vascular smooth muscle cells can be involved, finally, in structural damages and adverse cardiac outcomes, such as atherosclerotic cardiovascular disease, left ventricular hypertrophy, and heart failure. CKD, chronic kidney disease; ESRD, end-stage renal disease; DNMT, DNA methyltransferase; mtDNA, mitochondrial DNA; ROS, reactive oxygen species; TCA, tricarboxylic acid; ASCVD, atherosclerotic cardiovascular disease; LVH, left ventricular hypertrophy; HF, heart failure; ↓: increase; ↑: reduction. Created with Biorender.com.
Genes 16 01442 g003
Table 1. HDAC enzymes classes.
Table 1. HDAC enzymes classes.
ClassEnzymesCommon Name/CharacteristicsMechanism
IHDAC1, HDAC2, HDAC3, HDAC8“Classical” HDACsTraditional deacetylation mechanism
IIHDAC4, HDAC5, HDAC6, HDAC7, HDAC9, HDAC10“Classical” HDACsTraditional deacetylation mechanism
IIISIRT 1–7SirtuinsNAD+-dependent mechanism
IVHDAC11“Classical” HDACsShares only weak homology with Class I and II
HDAC, histone deacetylases; SIRT, silent information regulator Sirtuin; NAD+, nicotinamide adenine dinucleotide.
Table 2. List of FDA-approved epigenetic drugs.
Table 2. List of FDA-approved epigenetic drugs.
Drug NameTargetIndicationYear
Azacitidine (Vidaza®)DNMTiMyelodysplastic syndromes,
Chronic myelomonocytic leukemia,
Acute myeloid leukemia
2004
2004
2007
Decitabine (Dacogen®)
Decitabine-Cedazuridine (Inqovi®)
DNMTiMyelodysplastic syndromes,
Intermediate/high-risk myelodysplastic syndromes
2006
2020
Valproic Acid (Depakin®)Class I/II HDACEpilepsy, bipolar disorder, migraine2010
Vorinostat (Zolinza®)Class I/II HDACCutaneous T-cell lymphoma2006
Romidepsin (Istodax®)HDAC6Cutaneous T-cell lymphoma,
Peripheral T-cell lymphoma (withdrawn 2021)
2009
2011
Belinostat (Beleodaq®)Non-selective HDACiRelapsed or refractory peripheral T-cell lymphoma2014
Panobinostat (Farydak®)Non-selective HDACiRelapsed multiple myeloma (discontinued 2021)2015
Tazemetostat (Tazverik®)EZH2iMetastatic or locally advanced epithelioid sarcoma,
Relapsed or refractory follicular lymphoma
2020
2020
DNMT, DNA methyltransferase; HDAC, histone deacetylases; EZH2, enhancer of zeste homolog 2.
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

Cannito, S.; Giardino, I.; D’Apolito, M.; Ranaldi, A.; Scaltrito, F.; Pettoello-Mantovani, M.; Piscazzi, A. From Metabolic to Epigenetic Memory: The Impact of Hyperglycemia-Induced Epigenetic Signature on Kidney Disease Progression and Complications. Genes 2025, 16, 1442. https://doi.org/10.3390/genes16121442

AMA Style

Cannito S, Giardino I, D’Apolito M, Ranaldi A, Scaltrito F, Pettoello-Mantovani M, Piscazzi A. From Metabolic to Epigenetic Memory: The Impact of Hyperglycemia-Induced Epigenetic Signature on Kidney Disease Progression and Complications. Genes. 2025; 16(12):1442. https://doi.org/10.3390/genes16121442

Chicago/Turabian Style

Cannito, Sara, Ida Giardino, Maria D’Apolito, Alessandra Ranaldi, Francesca Scaltrito, Massimo Pettoello-Mantovani, and Annamaria Piscazzi. 2025. "From Metabolic to Epigenetic Memory: The Impact of Hyperglycemia-Induced Epigenetic Signature on Kidney Disease Progression and Complications" Genes 16, no. 12: 1442. https://doi.org/10.3390/genes16121442

APA Style

Cannito, S., Giardino, I., D’Apolito, M., Ranaldi, A., Scaltrito, F., Pettoello-Mantovani, M., & Piscazzi, A. (2025). From Metabolic to Epigenetic Memory: The Impact of Hyperglycemia-Induced Epigenetic Signature on Kidney Disease Progression and Complications. Genes, 16(12), 1442. https://doi.org/10.3390/genes16121442

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