Editorial Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Jun 15, 2025; 16(6): 105615
Published online Jun 15, 2025. doi: 10.4239/wjd.v16.i6.105615
Epigenetics and diabetic wound healing: Wilms tumor 1-associated protein as a therapeutic target
Ashraf Al Madhoun, Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15400, Kuwait
ORCID number: Ashraf Al Madhoun (0000-0001-8593-3878).
Author contributions: Al Madhoun A designed the overall concept, reviewed literature, wrote, and edited the manuscript.
Supported by the Kuwait Foundation for the Advancement of Sciences and Dasman Diabetes Institute, No. RACB-2021-007.
Conflict-of-interest statement: The author reports no relevant conflicts of interest for this article.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ashraf Al Madhoun, PhD, Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Jassim AlBahar Street, Dasman 15400, Kuwait. ashraf.madhoun@dasmaninstitute.org
Received: January 30, 2025
Revised: April 3, 2025
Accepted: May 12, 2025
Published online: June 15, 2025
Processing time: 134 Days and 20.3 Hours

Abstract

In this editorial, we highlight the study by Xiao et al. Despite progress in the management of diabetic foot ulcers (DFUs), impaired wound healing remains a significant clinical challenge. Recent studies have highlighted the critical role of epigenetic modifications in diabetic wound healing, with particular emphasis on DNA and RNA methylation pathways. This editorial discusses the findings of Xiao et al, who identified the Wilms tumor 1-associated protein (WTAP) - DNA methyltransferase 1 (DNMT1) axis as a pivotal regulator of endothelial dysfunction in DFUs. WTAP, a regulatory subunit of N6-methyladenosine (m6A) methyltransferase, is upregulated under high-glucose conditions and drives the excessive expression of DNMT1 via m6A modification. This contributes to impaired angiogenesis, reduced cell viability, and delayed wound closure. WTAP knockdown restored endothelial function and significantly improved wound healing in a diabetic mouse model. Furthermore, DNMT1 overexpression abrogated the benefits of WTAP suppression, confirming its downstream effector role. Thus, targeting the WTAP-DNMT1 axis provides a new avenue for DFU management. Moreover, epigenetic interventions that modulate both the m6A and RNA methylation pathways could restore endothelial function and enhance tissue repair in patients with diabetes.

Key Words: Diabetic foot ulcers; Epigenetics; Wilms tumor 1-associated protein; DNA methyltransferase 1; M6A modification; Wound healing

Core Tip: Xiao et al identified the Wilms tumor 1-associated protein (WTAP)-DNA methyltransferase 1 (DNMT1) axis as a pivotal regulator of diabetic foot ulcer healing. Using human umbilical vein endothelial cells and a diabetic mouse model, those researchers demonstrated that WTAP knockdown restored endothelial cell function and enhanced angiogenesis and collagen deposition. Importantly, siRNA-mediated WTAP depletion markedly improved wound closure in the diabetic mice. Furthermore, DNMT1 reversed the beneficial effects of WTAP knockdown, confirming that it is a key downstream effector of WTAP.



INTRODUCTION

Diabetes mellitus (DM), a chronic metabolic disorder, is a leading cause of foot ulcers and lower limb amputations worldwide[1]. Diabetic foot ulcers (DFUs) are of particular concern, affecting approximately 15%-25% of patients with DM throughout their lifetime[2]. DFUs are characterized by ulceration-associated neuropathy and/or peripheral arterial disease resulting from multiple factors, including poor glycemic control, foot injury, infection, foot surgery, and ischemia[3]. Various factors aggravate the development of the condition, increase its recurrence rates, and present multiple challenges to its management. Moreover, the recurrence of DFUs leads to substantial physical, psychological, and financial burdens[4]. Despite advancements in treatment, including debridement, offloading, and infection control, non-healing ulcers remain a major clinical challenge[5].

PATHOPHYSIOLOGY OF DFUS

The pathophysiology of DFUs is complex and involves a combination of persistent hyperglycemia, neuropathy, impaired wound healing, and endothelial dysfunction[6]. Prolonged hyperglycemia induces systemic damage, including peripheral nerve dysfunction, which is a key contributor to DFU development[7]. Neuropathy, a major complication of DM, progressively affects sensory, motor, and autonomic nerve functions, further aggravating foot vulnerability[8]. Peripheral sensory neuropathy reduces the patient’s perception of injury, whereas autonomic and motor neuropathy contribute to structural foot deformities, abnormal foot biomechanics, and skin dryness, amplifying vulnerability to callus formation, fissures, and ulceration[9]. Additionally, motor neuropathy alters the foot muscle mechanics, resulting in structural deformities such as hammer toe and Charcot joint[10].

Peripheral arterial disease and impaired healing mechanisms worsen tissue hypoxia, oxidative stress, inflammatory responses, and endothelial tissue dysfunction, mediating insulin resistance, vasoconstriction, and inflammation[11]. These chronic conditions, including DFUs, are associated with progressive tissue loss, complex infections, and cardiovascular disease[12]. Notably, infected ulcers contribute to over 20% of lower limb amputations[13].

DFU treatment is further complicated by nutritional insufficiencies, including deficiencies in zinc and vitamins A, C, and D[14]. Although emerging evidence suggests that vitamin D supplementation may support DFU management, clinical findings remain inconsistent[15]. The socioeconomic burden of DFUs highlights the need for improved diagnostic tools and innovative treatment strategies to effectively address the interconnected pathophysiological mechanisms of the condition.

ROLE OF EPIGENETICS IN DIABETIC WOUND HEALING

Epigenetic mechanisms such as DNA methylation and RNA modifications have recently been revealed as contributors to the development of DM and its complications. Despite the extensive body of research directed toward understanding the epigenetic mechanisms of diabetic complications such as cardiovascular diseases[16], nephropathy[17] and retinopathy[18], little is known about the role of epigenetics in the pathogenesis of DFUs. During wound healing, epigenetic mechanisms, including DNA methylation, histone modifications, and chromatin remodeling, are critical for the regulation of gene expression. Epigenetic alterations affect cellular homeostasis without modulating the underlying DNA sequence, providing control over immune responses, cell proliferation, and tissue repair[19,20]. In diabetic wounds, these processes are dysregulated, causing chronic inflammation, impaired immune cell function, and delayed healing[21]. For example, aberrant DNA methylation in immune cells such as macrophages can induce a proinflammatory environment[22]. Similarly, the disruptions of histone modification and chromatin remodeling alter the expression of genes involved in inflammation and tissue repair. These epigenetic changes are driven by factors such as hyperglycemia, inflammation, and oxidative stress, which are hallmarks of DM[23].

Presently, whether these epigenetic modifications primarily initiate changes in the diabetic wound microenvironment or exacerbate preexisting pathological conditions remains unclear. Despite studies showing that DNA methylation and hydroxymethylation regulate immune cell behavior in DFUs[19,20], direct evidence linking these changes to the onset of DFUs or proving their role in sustaining an already compromised condition remains lacking. Although the chronic nature of DFUs suggests a complex interplay among sustained hyperglycemia, inflammation, oxidative stress, and epigenetic dysregulation, the precise sequence of molecular events leading to impaired wound healing remains unresolved. Further studies are needed to determine the causal relationship between epigenetic dysregulation and DFU progression.

Accordingly, we cannot exclude the possibility that certain epigenetic modulations may serve as biomarkers of exposure to diabetic pathophysiology, possessing potential diagnostic and prognostic insights into DFU risk and progression. At the same time, other epigenetic modulations may provide mechanistic insights into how DM interrupts normal wound healing processes, potentially leading to targeted therapeutic interventions. The concept of epigenetic regulation as both a contributor and a consequence of disease is well documented in other chronic conditions, particularly cancer and cardiovascular diseases, where epigenetic dysregulation has been shown to play a dual role, acting as a key initiator of pathological transformation in some cases while exacerbating disease progression in others[24,25]. Therefore, a deeper exploration of the epigenetic mechanisms in DFUs could yield novel therapeutic strategies aimed at reversing detrimental modifications and restoring cellular homeostasis in chronic wounds.

DNA methylation and the hydroxymethylation of cytosine residues are key epigenetic mechanisms that silence gene expression[26]. Abnormal DNA methylation patterns have been observed in tissue-resident immune cells of diabetic wounds. For example, increased DNA methyltransferase 1 (DNMT1) activity in diabetic macrophages promotes a proinflammatory state and suppresses angiogenesis and extracellular matrix assembly[27,28]. Conversely, the hydroxymethylation of the regulatory region of matrix metalloproteinase 9 by TET enzymes elevates the expression of this matrix-degrading enzyme and impairs keratinocyte function and wound closure[29].

Histone modifications such as methylation and acetylation are often disrupted in diabetic wounds, causing improper gene activation or silencing[30]. For instance, Jumonji domain containing-3 (a histone demethylase) and mixed-lineage leukemia protein 1 (a histone methyltransferase) are often overexpressed in diabetic wounds, promoting macrophage-mediated inflammation[31-33]. Similarly, histone acetylation, which is associated with active gene transcription, is reduced in diabetic wounds, resulting in genes involved in tissue repair being silenced. For example, the decreased acetylation of histone H3 at the extracellular superoxide dismutase [Cu-Zn] (SOD3) gene promoter impairs antioxidative defenses, thereby exacerbating oxidative stress in diabetic wounds[34]. These observations highlight how histone modifications may serve as both markers and mediators of chronic DFUs.

Chromatin remodeling and nucleosome repositioning represent other critical epigenetic mechanisms in wound healing. In diabetic wounds, chromatin remodeling is often impaired, leading to improper gene expression and dysfunctional immune responses[35]. For example, impaired keratinocyte differentiation is associated with a low activity level of BRG1 ATPase, a key component of the SWI/SNF chromatin remodeling complex[36].

In summary, the epigenetic modulation of DNA introduces a complex layer of gene expression regulation, which is essential for effective wound healing. Dysregulation of these processes, especially in DM, can significantly impair tissue repair and regeneration. Therefore, understanding these disruptions provides opportunities for the development of therapeutic strategies to correct epigenetic errors and improve clinical outcomes in patients with chronic wounds.

KEY FINDINGS: THE WTAP-DNMT1 AXIS - A NOVEL EPIGENETIC MECHANISM

Xiao et al[37] introduced the novel concept of a Wilms tumor 1-associated protein (WTAP)-DNMT1 axis as a key epigenetic mechanism in diabetic wound healing. As a crucial component of the N6-methyladenosine (m6A) methyltransferase complex, WTAP was found to be significantly upregulated in human umbilical vein endothelial cells exposed to high glucose conditions. This increase in WTAP expression was associated with the impairment of cellular processes required for effective wound healing, including endothelial cell viability and migration as well as angiogenesis. These findings suggest that WTAP plays a detrimental role in the wound-healing process under diabetic conditions.

This study further revealed that WTAP exerts its effects through an epigenetic mechanism by modulating the m6A modification of DNMT1 mRNA, thereby enhancing its expression and causing endothelial dysfunction[37]. Furthermore, WTAP knockdown resulted in a significant reduction in the m6A modification of the DNMT1 transcripts and suppressed their expression. Notably, the reversal of DNMT1 expression restored the functions of the endothelial cells, improving their viability and migration as well as angiogenesis. These findings suggest that the WTAP-mediated m6A methylation of DNMT1 mRNA plays a critical role in impairing diabetic wound healing.

Next, the WTAP-DNMT1 axis was validated in a mouse model. WTAP knockdown in diabetic mice significantly accelerated wound healing, as evidenced by progressive wound closure, enhanced re-epithelialization, and increased collagen deposition. These observations suggest that the downregulation of WTAP promotes an effective and rapid wound-healing response under diabetic conditions. This study also demonstrated that the overexpression of DNMT1 reversed the beneficial effects of WTAP knockdown, confirming DNMT1 as a key downstream effector of WTAP. These mechanistic insights emphasize the importance of the WTAP-DNMT1 axis as a major player in diabetes-related deficits in wound healing and endothelial dysfunction[37].

As suggested by the authors, targeting the WTAP-DNMT1 axis may be a potential therapeutic strategy for diabetic wound healing. Suppressing WTAP or DNMT1 might, in principle, reverse endothelial cell dysfunction, enhance angiogenesis, and ultimately improve wound repair in patients with DM. Thus, this study opens new avenues for therapeutic interventions aimed at treating chronic complications arising from impaired wound healing in DM and offers prospects for better clinical management of the disease[37].

BROADER SIGNIFICANCE OF RNA METHYLATION IN DIABETES

The discovery of the role of WTAP in diabetic wound healing underscores the broader significance of RNA methylation in metabolic diseases. M6A modifications have been implicated in various cellular processes, including glucose metabolism, inflammation, and vascular function[38]. The dysregulation of m6A-modifying enzymes such as methyltransferase-like 3 (METTL3) and fat mass and obesity-associated (FTO) has been linked to DM-associated complications. For instance, METTL3 regulates apoptosis and inflammation in endothelial cells under high-glucose conditions[39,40]. Similarly, FTO-mediated m6A demethylation influences inflammation and vascular dysfunction[41]. Taken together, these findings highlight the potential of targeting m6A-related pathways as a therapeutic strategy for DM and its complications.

THERAPEUTIC IMPLICATIONS AND FUTURE DIRECTIONS

Targeting WTAP is a promising strategy for treating endothelial dysfunction, thereby improving DFU treatment in patients with DM. Although small-molecule inhibitors or RNA-based therapies that modulate WTAP activity have emerged as a novel class of treatments, achieving their high specificity is crucial for preventing off-target effects and preserving normal cellular functions. Additionally, combination therapies with existing treatments (e.g., growth factors or angiogenesis-promoting agents) for targeting WTAP may act synergistically to further enhance DFU treatment. These approaches illustrate the potential of multifaceted interventions in dealing with the complex pathophysiology of diabetic wounds.

In this regard, RNA-based therapeutic strategies also hold great promise[42]. The precise targeting of WTAP with small interfering RNAs (siRNAs), antisense oligonucleotides (ASOs), or mRNA-based therapies can help restore endothelial function. At the post-transcriptional level, both siRNAs and ASOs can silence WTAP, thereby reducing its expression and mitigating its pathological effects on diabetic wounds. Furthermore, the introduction of modified WTAP variants or regulatory elements through mRNA can counteract the deleterious functions of WTAP while preserving its physiological roles. Moreover, recent advances in lipid nanoparticle delivery systems have enhanced the feasibility of RNA-based therapies by enabling targeted delivery to cells in the wound microenvironment.

Beyond wound healing, the interaction between WTAP and DNMT1 in other diabetic complications such as nephropathy and retinopathy require further investigation. Studying the WTAP-DNMT1 axis in this context may reveal additional therapeutic avenues and broaden the clinical relevance of these findings. However, before these discoveries can be translated into clinical applications, further validations of the safety, efficacy, and pharmacokinetics of such treatments in larger animal models and human trials are required. Additionally, high levels of WTAP can serve as a biomarker of poor wound healing, facilitating early diagnosis and personalized treatment approaches. With the increasing recognition of RNA-based therapeutics as powerful tools in precision medicine, continued research on their application in DFUs will unlock new treatment paradigms, offering more targeted and effective solutions for managing chronic wounds and other metabolic dysfunctions.

CONCLUSION

The study by Xiao et al[37] represents a significant advancement in our understanding of diabetic wound healing. The identification of WTAP as a key epigenetic regulator as well as its interaction with DNMT1 has revealed a novel pathway that mediates endothelial dysfunction under hyperglycemic conditions. These findings not only provide new insights into the molecular mechanisms of diabetic wound pathology but also offer innovative therapeutic strategies. The increasing global burden of DM has also increased the demand for effective treatment interventions for its complications. Indeed, epigenetic therapies, including WTAP inhibitors, have great potential for treating diabetic wounds and managing DM-associated complications. Further studies are necessary to validate these findings, advance the development of targeted therapies, and explore the broader implications of RNA methylation in disease progression. Bridging the gap between molecular biology and clinical innovation is essential to improve the outcomes for millions of people living with DM.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Endocrinology and metabolism

Country of origin: Kuwait

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Chen HH S-Editor: Li L L-Editor: A P-Editor: Xu ZH

References
1.  Rathnayake A, Saboo A, Malabu UH, Falhammar H. Lower extremity amputations and long-term outcomes in diabetic foot ulcers: A systematic review. World J Diabetes. 2020;11:391-399.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 23]  [Cited by in RCA: 45]  [Article Influence: 9.0]  [Reference Citation Analysis (4)]
2.  McDermott K, Fang M, Boulton AJM, Selvin E, Hicks CW. Etiology, Epidemiology, and Disparities in the Burden of Diabetic Foot Ulcers. Diabetes Care. 2023;46:209-221.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 212]  [Cited by in RCA: 350]  [Article Influence: 175.0]  [Reference Citation Analysis (1)]
3.  Edmonds M, Manu C, Vas P. The current burden of diabetic foot disease. J Clin Orthop Trauma. 2021;17:88-93.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 39]  [Cited by in RCA: 186]  [Article Influence: 46.5]  [Reference Citation Analysis (0)]
4.  Qian L, Yan S, Ting ST, Han ZM, Qi T. Complications and psychological impact of pressure ulcers on patients and caregivers. Int Wound J. 2024;21:e14836.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 11]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
5.  Rasouli M, Goli R, Nokashti HH. Surgical debridement and maggot debridement therapy can survive patient with diabetic foot ulcer after foot trauma: A case report. Int J Surg Case Rep. 2024;121:109990.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
6.  Kim J. The pathophysiology of diabetic foot: a narrative review. J Yeungnam Med Sci. 2023;40:328-334.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 14]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
7.  Parveen K, Hussain MA, Anwar S, Elagib HM, Kausar MA. Comprehensive review on diabetic foot ulcers and neuropathy: Treatment, prevention and management. World J Diabetes. 2025;16:100329.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
8.  Volmer-Thole M, Lobmann R. Neuropathy and Diabetic Foot Syndrome. Int J Mol Sci. 2016;17:917.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 130]  [Cited by in RCA: 212]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
9.  Wang X, Yuan CX, Xu B, Yu Z. Diabetic foot ulcers: Classification, risk factors and management. World J Diabetes. 2022;13:1049-1065.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in CrossRef: 2]  [Cited by in RCA: 70]  [Article Influence: 23.3]  [Reference Citation Analysis (3)]
10.  Bus SA, Maas M, Michels RP, Levi M. Role of intrinsic muscle atrophy in the etiology of claw toe deformity in diabetic neuropathy may not be as straightforward as widely believed. Diabetes Care. 2009;32:1063-1067.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 60]  [Cited by in RCA: 58]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
11.  Lee SH, Kim SH, Kim KB, Kim HS, Lee YK. Factors Influencing Wound Healing in Diabetic Foot Patients. Medicina (Kaunas). 2024;60:723.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
12.  Gallagher KA, Mills JL, Armstrong DG, Conte MS, Kirsner RS, Minc SD, Plutzky J, Southerland KW, Tomic-Canic M; American Heart Association Council on Peripheral Vascular Disease;  Council on Cardiovascular and Stroke Nursing;  Council on Clinical Cardiology;  and Council on Lifestyle and Cardiometabolic Health. Current Status and Principles for the Treatment and Prevention of Diabetic Foot Ulcers in the Cardiovascular Patient Population: A Scientific Statement From the American Heart Association. Circulation. 2024;149:e232-e253.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 18]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
13.  Armstrong DG, Tan TW, Boulton AJM, Bus SA. Diabetic Foot Ulcers: A Review. JAMA. 2023;330:62-75.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 216]  [Cited by in RCA: 436]  [Article Influence: 218.0]  [Reference Citation Analysis (0)]
14.  Bechara N, Gunton JE, Flood V, Hng TM, McGloin C. Associations between Nutrients and Foot Ulceration in Diabetes: A Systematic Review. Nutrients. 2021;13:2576.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
15.  Dai J, Jiang C, Chen H, Chai Y. Vitamin D and diabetic foot ulcer: a systematic review and meta-analysis. Nutr Diabetes. 2019;9:8.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 19]  [Cited by in RCA: 37]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
16.  Pirola L, Balcerczyk A, Tothill RW, Haviv I, Kaspi A, Lunke S, Ziemann M, Karagiannis T, Tonna S, Kowalczyk A, Beresford-Smith B, Macintyre G, Kelong M, Hongyu Z, Zhu J, El-Osta A. Genome-wide analysis distinguishes hyperglycemia regulated epigenetic signatures of primary vascular cells. Genome Res. 2011;21:1601-1615.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 173]  [Cited by in RCA: 164]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
17.  Advani A, Huang Q, Thai K, Advani SL, White KE, Kelly DJ, Yuen DA, Connelly KA, Marsden PA, Gilbert RE. Long-term administration of the histone deacetylase inhibitor vorinostat attenuates renal injury in experimental diabetes through an endothelial nitric oxide synthase-dependent mechanism. Am J Pathol. 2011;178:2205-2214.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 121]  [Cited by in RCA: 128]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
18.  Zhong Q, Kowluru RA. Epigenetic changes in mitochondrial superoxide dismutase in the retina and the development of diabetic retinopathy. Diabetes. 2011;60:1304-1313.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 152]  [Cited by in RCA: 169]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
19.  Barrero MJ, Boué S, Izpisúa Belmonte JC. Epigenetic mechanisms that regulate cell identity. Cell Stem Cell. 2010;7:565-570.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 82]  [Cited by in RCA: 90]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
20.  Portela A, Esteller M. Epigenetic modifications and human disease. Nat Biotechnol. 2010;28:1057-1068.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1865]  [Cited by in RCA: 1964]  [Article Influence: 140.3]  [Reference Citation Analysis (0)]
21.  Mannar V, Boro H, Patel D, Agstam S, Dalvi M, Bundela V. Epigenetics of the Pathogenesis and Complications of Type 2 Diabetes Mellitus. touchREV Endocrinol. 2023;19:46-53.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
22.  Singh R, Chandel S, Dey D, Ghosh A, Roy S, Ravichandiran V, Ghosh D. Epigenetic modification and therapeutic targets of diabetes mellitus. Biosci Rep. 2020;40:BSR20202160.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 44]  [Cited by in RCA: 44]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
23.  Keating ST, Plutzky J, El-Osta A. Epigenetic Changes in Diabetes and Cardiovascular Risk. Circ Res. 2016;118:1706-1722.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 85]  [Cited by in RCA: 108]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
24.  Nise MS, Falaturi P, Erren TC. Epigenetics: origins and implications for cancer epidemiology. Med Hypotheses. 2010;74:377-382.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 9]  [Cited by in RCA: 10]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
25.  Lorenzen JM, Martino F, Thum T. Epigenetic modifications in cardiovascular disease. Basic Res Cardiol. 2012;107:245.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 96]  [Cited by in RCA: 105]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
26.  Dhar GA, Saha S, Mitra P, Nag Chaudhuri R. DNA methylation and regulation of gene expression: Guardian of our health. Nucleus (Calcutta). 2021;64:259-270.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 14]  [Cited by in RCA: 113]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
27.  Wang X, Cao Q, Yu L, Shi H, Xue B, Shi H. Epigenetic regulation of macrophage polarization and inflammation by DNA methylation in obesity. JCI Insight. 2016;1:e87748.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 90]  [Cited by in RCA: 150]  [Article Influence: 16.7]  [Reference Citation Analysis (0)]
28.  Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99:247-257.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4452]  [Cited by in RCA: 4272]  [Article Influence: 164.3]  [Reference Citation Analysis (0)]
29.  Zhang J, Yang C, Wang C, Liu D, Lao G, Liang Y, Sun K, Luo H, Tan Q, Ren M, Yan L. AGE-induced keratinocyte MMP-9 expression is linked to TET2-mediated CpG demethylation. Wound Repair Regen. 2016;24:489-500.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 22]  [Cited by in RCA: 38]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
30.  Strahl BD, Allis CD. The language of covalent histone modifications. Nature. 2000;403:41-45.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 6156]  [Cited by in RCA: 6125]  [Article Influence: 245.0]  [Reference Citation Analysis (0)]
31.  De Santa F, Totaro MG, Prosperini E, Notarbartolo S, Testa G, Natoli G. The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing. Cell. 2007;130:1083-1094.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 696]  [Cited by in RCA: 753]  [Article Influence: 41.8]  [Reference Citation Analysis (1)]
32.  Robert I, Aussems M, Keutgens A, Zhang X, Hennuy B, Viatour P, Vanstraelen G, Merville MP, Chapelle JP, de Leval L, Lambert F, Dejardin E, Gothot A, Chariot A. Matrix Metalloproteinase-9 gene induction by a truncated oncogenic NF-kappaB2 protein involves the recruitment of MLL1 and MLL2 H3K4 histone methyltransferase complexes. Oncogene. 2009;28:1626-1638.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 30]  [Cited by in RCA: 34]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
33.  Kimball AS, Joshi A, Carson WF 4th, Boniakowski AE, Schaller M, Allen R, Bermick J, Davis FM, Henke PK, Burant CF, Kunkel SL, Gallagher KA. The Histone Methyltransferase MLL1 Directs Macrophage-Mediated Inflammation in Wound Healing and Is Altered in a Murine Model of Obesity and Type 2 Diabetes. Diabetes. 2017;66:2459-2471.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 55]  [Cited by in RCA: 75]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
34.  Yasuda H, Ohashi A, Nishida S, Kamiya T, Suwa T, Hara H, Takeda J, Itoh Y, Adachi T. Exendin-4 induces extracellular-superoxide dismutase through histone H3 acetylation in human retinal endothelial cells. J Clin Biochem Nutr. 2016;59:174-181.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 17]  [Cited by in RCA: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
35.  den Dekker A, Davis FM, Kunkel SL, Gallagher KA. Targeting epigenetic mechanisms in diabetic wound healing. Transl Res. 2019;204:39-50.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 92]  [Cited by in RCA: 156]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
36.  Indra AK, Dupé V, Bornert JM, Messaddeq N, Yaniv M, Mark M, Chambon P, Metzger D. Temporally controlled targeted somatic mutagenesis in embryonic surface ectoderm and fetal epidermal keratinocytes unveils two distinct developmental functions of BRG1 in limb morphogenesis and skin barrier formation. Development. 2005;132:4533-4544.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 94]  [Cited by in RCA: 97]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
37.  Xiao RJ, Wang TJ, Wu DY, Yang SF, Gao H, Gan PD, Yi YY, Zhang YL. N6-methyladenosine methyltransferase Wilms tumor 1-associated protein impedes diabetic wound healing through epigenetically activating DNA methyltransferase 1. World J Diabetes. 2025;16:102126.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
38.  Ren Y, Li Z, Li J, Liang R, Wang Z, Bai Y, Yang Y, Tang Q, Fu Y, Zhang X, Zhang Y, Yu Y, Xiong Y. m(6) A mRNA methylation: Biological features, mechanisms, and therapeutic potentials in type 2 diabetes mellitus. Obes Rev. 2023;24:e13639.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
39.  Song B, Zeng Y, Cao Y, Zhang J, Xu C, Pan Y, Zhao X, Liu J. Emerging role of METTL3 in inflammatory diseases: mechanisms and therapeutic applications. Front Immunol. 2023;14:1221609.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 24]  [Reference Citation Analysis (0)]
40.  Li N, Wei X, Dai J, Yang J, Xiong S. METTL3: a multifunctional regulator in diseases. Mol Cell Biochem. 2025;.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
41.  Zhou C, She X, Gu C, Hu Y, Ma M, Qiu Q, Sun T, Xu X, Chen H, Zheng Z. FTO fuels diabetes-induced vascular endothelial dysfunction associated with inflammation by erasing m6A methylation of TNIP1. J Clin Invest. 2023;133:e160517.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 31]  [Reference Citation Analysis (0)]
42.  Liu WW, Zheng SQ, Li T, Fei YF, Wang C, Zhang S, Wang F, Jiang GM, Wang H. RNA modifications in cellular metabolism: implications for metabolism-targeted therapy and immunotherapy. Signal Transduct Target Ther. 2024;9:70.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 30]  [Reference Citation Analysis (0)]