Open Access

Role of DNA methylation transferase in urinary system diseases: From basic to clinical perspectives (Review)

  • Authors:
    • Yipin Yang
    • Yafen Wang
    • Xiufang Fan
    • Xinwei Xu
    • Huijuan Wang
    • Xinyi Wang
    • Taiyu Shi
    • Jialu Tang
    • Yanmeng Guan
    • Song Li
    • Aimei Wang
  • View Affiliations

  • Published online on: November 19, 2024     https://doi.org/10.3892/ijmm.2024.5460
  • Article Number: 19
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

DNA methylation is one of the earliest discovered and most extensively studied epigenetic regulatory mechanisms. Broadly, DNA methylation refers to the transfer of a methyl group on S‑adenosine‑L‑methionine (SAM) to the C5 site of cytosine, a reaction catalysed by DNA methyltransferase (DNMT). This process can either up‑ or down‑regulate gene expression due to gene promoter methylation, leading to the occurrence of certain diseases. Urinary system diseases, known for their high prevalence and complex pathogenesis, significantly affect the lives and health of patients. Urological tumours, in particular, represent a non‑negligible disease burden worldwide. With the development of epigenetics, an increasing number of studies have demonstrated that DNMT plays an important role in urinary system disease. The present review aims to explore the relationship between DNMT and urinary system diseases and the potential of DNMT in the clinical management of these diseases.

Introduction

The urinary system is narrowly defined to include kidneys, ureters, the bladder and the urethra. In a broader context, it encompasses the cervix, vulva, adnexa, ovaries and fallopian tubes in women, and the prostate, vas deferens, testes and epididymis in men. Diseases can affect each of these organs and impact the entire system. Common urinary system diseases mainly include infections, tumours, stones and inflammation, congenital malformation and trauma. Among these, tumours are the most fatal and have been a major focus of academic research. In 2022, there were 434,419 new cases of kidney cancer and 155,702 kidney cancer-associated mortalities worldwide (1); studies have reflected the rising incidence of urological tumours in recent years (2-4). Non-muscle invasive bladder cancer (NMIBC) has a recurrence rate of ≥70% requiring repeated, costly and invasive testing, making bladder cancer (BCa) the most expensive cancer to treat per capita (5). Urological tumours, therefore, pose a threat to the quality of human life and incur substantial economic costs.

The pathogenesis of urinary system diseases is complex and remains to be actively explored. Epigenetics is an emerging theme in disease pathogenesis, with a major focus on DNA methylation and histone acetylation (6). Previous studies have highlighted that DNA methylation plays an essential role in urinary system diseases recently (5,7,8). DNA methylation is a stable epigenetic marker in humans and has been recognized, along with other regulatory factors, as a major factor influencing gene activity. The DNA methyltransferase (DNMT) family comprises a conserved set of DNA-modifying enzymes central to epigenetic gene regulation (9). A total of three DNMTs, DNMT1, DNMT3A and DNMT3B, catalyze the transfer of a methyl group from S-adenosyl-L-methionine (SAM) to the C5 position of cytosine (10), a process known as methylation. Methylation predominantly occurs at cytosine-phospho-guanine (CpG) islands; which are CpG-rich regions (≥50% of cytosines and guanines) in the genome with a size >200 bp (11,12).

Methylation occurs not only in specific regions of the X chromosome, such as retrotransposon elements and centromere regions, and the result of methylation in these regions usually leads to genes inactivation (13). Studies have shown a significant association between the DNA methylation process in genes and urinary system diseases, with this chemical modification contributing to the development of urinary system disease and holding potential for diagnostic and prognostic applications (14-17). Therefore, the present study systematically explored the pathogenic mechanism and clinical application of DNMTs in urinary system diseases, opening a window of opportunity for improved clinical management.

Overview of urinary system diseases

Urinary system diseases can occur in all organs of the urinary system (kidneys, ureters, bladder and urethra) and spread throughout the system. It manifests itself in a variety of symptoms, including difficulty urinating, abnormal urine (such as changes in colour, clarity or smell), stone formation and a sense of pain. At the same time, it may indirectly cause other health problems, such as high blood pressure, oedema and anemia (18).

Non-oncological diseases of the urinary system

Common non-neoplastic urinary system diseases include autonomic-induced urinary dysfunction, urinary tract infections (UTIs) and urinary stones. Autonomic nerves are essential in urinary tract development, growth factors production and homeostasis control (19-21). The function of the lower urinary tract (storage and periodic elimination of urine) highly depends on the complex neural control system in the brain, spinal cord and peripheral ganglia. Additionally, injury or disease of the corresponding nerve may trigger neurogenic bladder or lower urinary tract dysfunction (22-24).

UTIs are an inflammatory response of the urinary tract epithelium caused by bacterial invasion. It is one of the most common community and hospital-acquired infections (25). Infectious microorganisms, including Gram-negative and Gram-positive bacteria and certain fungi, can cause UTIs via the hematogenous route and ascending routes (26). As one of the most common pathogens, Escherichia coli (E. coli) has multiple virulence factors, including pili, capsule, siderophore receptor, flagella, toxins and lipopolysaccharides, and they work together to cause a UTI (25,27). The conventional UTI treatment is mainly based on the use of antibiotics such as β-lactams, trimethoprim, nitrofurantoin and quinolones; however, antibiotic misuse has led to increased E. coli resistance, driven by genetic variation and horizontal gene transfer (28,29). Consequently, researchers are exploring alternative therapeutic strategies for UTI. The widely conserved respiratory quinoline oxidase cytochrome bd is essential for urothelial intracellular infection; hence, developing drugs targeting this enzyme may be a good research direction (30).

Urolithiasis is a common disease with a growing prevalence and high recurrence rate worldwide (31,32). Urinary stones are classified into two main categories: Metabolic and infected stones. Infectious urinary calculi account for 15% of urinary calculi, often caused by urease positive pathogens (33). The main treatment options include extracorporeal shock wave lithotripsy (ESWL), surgery and drug therapy. ESWL is typically used for renal and upper ureteral stones with diameter ≤2 cm in diameter. In rare diseases such as congenital anatomical anomalies where surgeons have difficulty in accomplishing the treatment of kidney stone disease through ESWL, percutaneous nephrolithotomy, ureteroscopy and minimally invasive surgery. Often they can only accomplish stone removal through open surgery (34).

Urinary system diseases caused by tumours

Epidemiological studies have shown that the incidence of urinary system tumours is increasing globally, significantly impacting the health and quality of life of patients (35,36). According to the global cancer statistics in 2020, there were >2.4 million new cases of urinary tract tumours reported worldwide, accounting for 12.5% of cancer diagnoses and 7.7% of new cancer-associated mortalities (37). Tumours of the urinary system are divided into three categories: i) Urothelial carcinoma of the renal pelvis, ureter and bladder; ii) renal malignant tumours, such as renal cell carcinoma (RCC) and Wilms tumour; and iii) non-urethral epithelial tumours of various organs. The relative rarity of upper tract urothelial carcinoma (UTUC) complicates the establishment of diagnostic and management strategies (38,39). Although advances in optical diagnostic technology should reduce invasive testing for UTUC, urinary cytology remains a primary methods for screening and monitoring urothelial carcinoma (40,41). The mechanisms underlying urinary system tumours remain a significant focus of academic research. Epigenetic alterations, such as histone methylation, acetylation and DNA methylation, are ubiquitous in urological tumours, and have garnered considerable scholarly attention (42).

Overview of DNMTs

The enzymes that build, recognize and eliminate DNA methylation are divided into three categories: i) Writing; ii) erasing; and iii) reading enzymes. DNMTs are known as writing enzymes. The DNMTs are a conserved family of cytosine methylases with a critical role in epigenetic regulation, catalysing DNA methylation by transferring a methyl group from SAM to the fifth carbon of a cytosine residue, forming 5mC (43). DNA methylation occurs mainly at the CpG dinucleotide site, and it occurs less frequently at cytosine-phosphothymine, cytosine-phosphoadenine and cytosine-phosphocytosine sites, which are collectively referred to as non-CpG sites (44).

The human genome encodes five DNMTs: DNMT1, DNMT2, DNMT3A, DNMT3B and DNMT3L. DNMT1, DNMT3A and DNMT3B are canonical cytosine-5 DNMTs that catalyse the addition of methyl groups to genomic DNA. By contrast, DNMT2 and DNMT3L are considered non-canonical family members because they lack catalytic DNMT activity (9). De novo methylation and maintenance methylation are important physiological processes in epigenetics. De novo methylation refers to the methylation of cytosine C5 at a new site on the DNA chain. Maintenance methylation is the process of DNA replication that leaves the methylation of the original site unchanged; that is, the process of methylation modification at the corresponding position of the nascent strand results from the semi-conserved replication of methylated DNA (45). DNMT3A/3B is designated as a de novo DNMT and DNMT1 is designated as a maintenance DNMT. However, DNMT3A/3B participates in DNA maintenance methylation and DNMT1 in de novo methylation (46).

Structure of DNMTs

DNMTs typically comprise an N-terminal regulatory structural domain and a C-terminal catalytic structural domain. DNMT3A has two isoforms while DNMT3B has >30 isoforms. Despite this variation, both share similar domain structures: i) A Pro-T rp-T rp-Pro (pWWP) domain; ii) an ATRX-DNMT3L-DNMT3A (ADD) domain; and iii) a C-terminal catalytic domain. The pWWP domain recognizes H3K36me3 and the ADD domain binds the free N-terminal tail of histone H3 (27121154) (47). Among the three typical cytosine-5 DNMTs, DNMT1 is the most abundant DNMT and is considered to be the critical methyltransferase in mammals (48).

DNMT1 prioritizes the methylation of hemi-methylated DNA, thereby maintaining the epigenetic stability of genetic information. The unique N-terminal regulatory region of DNMT1 contains several structural domains, including the DNA replication foci structural domain (RFD) and the CXXC structural domain, playing the role of the corresponding structural domains. For example, the RFD directs DNMT1 to replicate DNA, while the CXXC domain recognizes and binds DNA containing unmethylated CpG sites.

Activity of DNMTs

The induction of epigenetic alterations, particularly aberrant DNA methylation, is strongly associated with chronic inflammation and various human diseases, including cancer, neurodegenerative diseases and metabolic disorders. Takeshima et al (49) demonstrated that the DNMT activation, triggered by nitrogen oxide (NO) production, is responsible for abnormal methylation in human tissues, indicating that DNMT activity plays a key role in the development and progression of multiple human diseases.

Activity of DNMT1

DNMT1 activity is modulated by a number of molecular interactions. Lymphoid-specific helicase (LSH), a DNA decoupling enzyme belonging to the SNF2 family, has long been recognized to enhance DNMT activity (50,51). Ubiquitin-like, containing PHD and RING finger domains 1 (UHRF1), an E3-ubiquitin protein ligase that interacts with DNMT1, is essential for maintaining DNA methylation when it binds to DNA (9). A previous study has shown that LSH interacts with UHRF1 in an ATPase activity-dependent manner, facilitating DNMT1 recruitment to replication forks and DNA methylation processes. In particular, UHRF1 enhances the binding of LSH to replication forks. This interaction enhances UHRF1 binding to chromatin and promotes UHRF1-catalyzed histone H3 ubiquitination, activating DNMT1 and facilitating its recruitment during the S phase of the cell cycle (52).

DNMT1 activity is affected by its microenvironment. Atrazine, a non-competitive inhibitor of the DNMT reaction, significantly reduces DNMT1 activity. Atrazine exposure at the embryonic stage may, therefore, have toxic effects on various organs by modulating genes (53-56). Duraisamy et al (57) reported that DNMT1 activity in retinal endothelial cells incubated in high glucose initially decreases after 6 h but increases after 24 h.

Studies have shown that DNMT1 fidelity is related to gene complexity and regional differences (58-60). High-complexity repeats and active gene promoter regions exhibit relatively high fidelity, while low-complexity repeats and methylated sequences in intermediate regions show relatively lower fidelity (58-60). However, DNMT1 can correct its inaccuracies through neighbour-guided correction, where changes in local DNA methylation alter the de novo and maintenance activity of DNMT1; at this point, DNMT1 becomes a key 'tuner', effectively improving fidelity (61). Thus, the inaccuracies and neighbour-guided correction of DNMT1 constitute essential mechanisms for maintaining DNA methylation (61).

Activity of DNMT3A and DNMT3B

The activity of DNMT3A and DNMT3B is significant in pathological processes. Studies have shown that eicosatetraenoic acid inhibits histone deacetylase 1 (HDAC1) and DNMT transcription and activity by acting as a natural proliferator-activated receptor γ (PPARγ) ligand and activator (62). Activated peroxisome PPARγ primarily decreases HDAC1 activity, followed by decreases in expression and activity of DNMT 3A and 3B. This process eventually leads to the up-regulation of tumour-suppressor genes (TSGs) (63).

Recently, Zhao et al (64) reported that Oroxylin A can inhibit methionine cycle metabolism in hematopoietic stem cells and reduces the expression of methionine adenosyltransferase 2A, a key enzyme in methionine cycle metabolism, leading to lower levels of SAM, which in turn affects the DNMT activity. The down-regulation of DNMT3A activity inhibits the methylation of cyclic GMP-AMP synthase (cGAS) gene and promotes the activation of the cGAS-STING pathway, thus accelerating the aging of hepatic stellate cells (64). In cases where SAM levels are significantly reduced, DNMT-3A and DNMT-3B may catalyse the demethylation of 5mC through their deamination activity, converting it back to unmodified cytosine. This process may involve the base excision repair pathway, where unmodified cytosine replaces the original methylated cytosine (65).

A recent study reported how genetically encoded photicaged amino acid controls DNMT activity. The technique enables the production of DNMTs, which is otherwise inactive. and can be rapidly activated by light irradiation. The results of this study contribute to a deeper understanding of how cancer-associated DNMT mutations affect their activity in organisms, providing an important foundation for future research and ad.vancements in cancer treatment (66).

Function of DNMTs in T cells

Epigenetic modifications, particularly covalent modifications of DNA itself by DNA methylation, are recognized as critical in determining T cell fate (67,68). Throughout the development of T lymphocytes in the thymus and their subsequent migration to the periphery for differentiation, they are influenced by a series of precisely regulated molecular mechanisms. DNMT3A plays a pivotal role as an epigenetic mechanism component in maintaining the stability of T cell responses (69-71). In mice with DNMT3A knocked out, acute graft-vs.-host disease was observed, accompanied by a significant increase in inflammatory cytokine levels and higher pathological scores in organ tissues. T cells from DNMT3A knockout mice migrated and proliferated earlier in secondary lymphoid organs and showed a tendency to migrate to the small intestine. These findings underscore the vital role for DNMT3A in regulating T cell allogeneic responsiveness (69). Additionally, DNMT1 plays an important regulatory role in Foxp3 expression. In regulatory T (Treg) cells, the Foxp3 promoter region remains hypomethylated (70). Lv et al (71) further explored the important role of DNMT1-promoted Treg differentiation. The results showed that pcDNA3.1(+)-mDNMT-1 blocked polymer protein-promoted IL-10 and Foxp3 expression and Treg differentiation (Fig. 1).

Role of DNMTs in diseases

DNA methylation levels undergo tissue- and organ-specific changes, contributing to various diseases including neurological disorders, cancers, atherosclerosis and osteoporosis. A distinctive feature of cancer is widespread DNA repeat hypomethylation, a phenomenon that affects a large DNA region and is accompanied by frequent local DNA hypermethylation events (72). The degree of hypermethylation has been shown to correlate with cancer aggressiveness (73-75). DNMTs are abnormally expressed in various malignancies, including BCa (76,77), prostate cancer (PCa) (78), colorectal cancer (79) and osteosarcoma (OSA) (80). Genes affected by hypermethylation in cancer cells mainly include TSGs and DNA mismatch repair (MMR) genes. The low expression of TSGs and the silencing of DNA MMR genes are important mechanisms for cancer development (81,82). For instance, DNMT1 mediates hypermethylation of CpG islands in the microRNA (miR)-34a promoter region, leading to miR-34a deficiency and overexpression of Notch proteins, ultimately promoting metastasis and progression of OSA (83). The DNMT-1 inhibitor enhances miR-34a expression in OSA cells, leading to an antitumour effect and may be a promising therapeutic strategy for OSA (83). Studies have shown that hypermethylation of oncogenes and MMR genes due to aberrant DMNT function may activate the MAPK (84), PI3K/ACT (85) and Wnt/b-catenin (75) signalling pathways, which contribute to tumourigenesis (86).

In summary, DNMTs plays a crucial role in the pathogenesis of various diseases and can serve as a potential therapeutic targets for the corresponding diseases. With the rapid development of epigenetics, the role and clinical application of DNMT in urinary system diseases have received widespread attention.

Function of DNMTs in various types of urinary system diseases

Growing evidence shows that DNMTs play a significant role in the development of urinary system diseases. Overexpression of DNMTs exacerbates renal fibrosis (87), tubular degeneration (88), polycystic kidney disease (89,90), chronic bladder obstruction (91), as well as the progression of urinary system tumours (5,77,92,93). The aforementioned studies suggest that DNMT is involved in the mechanisms of multiple urinary system diseases development. The present review examines the role and specific mechanisms of DNMTs in seven urological disorders. In conclusion, abnormal expression of DNMTs plays an important role in these diseases (Fig. 2).

Non-oncological diseases of the urinary system Cystitis

Cystitis is a physiological process that characterizes a number of bladder diseases such as UTIs and haemorrhagic cystitis. Cystitis involves aberrant epigenetic alterations through DNA methylation and histone deacetylation. These histone modifications recruit DNMTs, mediate DNA methylation and regulate the expression of genes implicated in pathology (94).

A study used urine specimens from women with interstitial cystitis/painful bladder syndrome (IC/BPS) to assess DNA methylation, and the findings suggest that genes within or downstream of the MAPK pathway exhibit altered methylation in IC/BPS (95). Infection of mammalian tissues with bacteria, viruses and other pathogens results in DNA methylation. In vitro studies have shown that infection of bladder uroepithelial cells with uropathogenic E. coli results in hypermethylation of the TSG CDKN2A (96-98). This hypermethylation may be triggered by lipopolysaccharides (96,99), which activate NF-κB signal transduction via Toll-like receptors, enables mimicking of the inflammation process, and produces certain oxidants such as superoxide and NO (100).

Damaged cytosine products resulting from inflammation, particularly halogenated cytosine residues, are likely to facilitate binding with methyl-binding proteins and enzymatic methylation (101). Uropathogenic E. coli infection modulates the host cell epigenome (96). Systemic chemotherapeutic agents, particularly cyclophosphamide (CPX) and other nitrogen mustard alkylating agents, cause severe clinical manifestations such as cystitis (102). Haldar et al (103) showed that exposure of mouse bladders to CPX results in the DNA methylation of the Ogg1 promoter, which down-regulates gene expression. Furthermore, Choi et al (104) demonstrated that DNA methylation changes are observed in the Calca, tissue inhibitor of metalloproteinase 3 (Timp3), matrix metallopeptidase 2 (Mmp2) and insulin-like growth factor 2 receptor (Igf2r) genes in a mouse model of chronic bladder inflammation induced by CYP (104).

Hypertensive nephropathy

Hypertensive nephropathy is the second leading cause of end-stage renal disease, following diabetes. At present, hypertension has attracted the attention of the medical community due to its high prevalence (105). Data shows that individuals with hypertension account for ~33% of the global population (106,107). Long-term persistent hypertension often leads to chronic renal failure, and in malignant hypertension, renal failure can occur in the short term (108). Studies have shown that the activities of superoxide dismutase (SOD), catalase and glutathione peroxidase are lower in patients with hypertension compared with healthy individuals (109-112). Concurrently, the expression of key enzymes producing reactive oxygen species (ROS), such as NADPH oxidase, increase. Since NADPH oxidase isoforms are highly expressed in the kidneys, oxidative stress is one of the important mechanisms of hypertensive nephropathy (113).

The mechanism of methylation also plays an important role in this process. In an animal study involving Ang-II-induced hypertension, the researchers reported that in addition to the increase in blood pressure, the Ang-II-treated group also had increased expression of DNMT1 and DNMT3A, which act as an antioxidant defence systems to eliminate ROS (113). However, SOD and Sirtuin 1 genes are hypermethylated and inhibited in transcription, which impairs antioxidant function (113,114). Moreover, animal studies have shown that a maternal low-protein diet induces hypermethylation and increased expression of prostaglandin E receptor 1 in the kidneys of young rats, which is associated with subsequent salt-sensitive hypertension in adult rats (115,116). Understanding the methylation mechanism in hypertensive nephropathy will help clarify the specific pathogenesis and potentially lead to novel therapeutic strategies in the future.

Diabetic nephropathy (DN)

DN is a major global public health issue. The number of individuals with diabetes may reach 578 million by 2030 and 700 million by 2045 (117). Approximately 35% of individuals with diabetes develop DN (118), making it the long-term complication of diabetes with the most significant social and family burden (119). Therefore, the study of DN has always been a focal point in the academic community. In recent years, a number of studies have shown that epigenetic changes play a role in the pathogenesis of kidney disease (87,120-122). For example, DNMT1 expression has been observed to be increased in damaged glomerular podocytes of diabetic mice. The actin α4 (ACTN4) gene is hypermethylated in podocytes, but this can be down-regulated using DNA methylation inhibitors such as 5-Aza or epigallocatechin-3-gallate (EGCG). These interventions ensure the integrity of podocytes, effectively reduce the level of urinary protein in diabetic mice, reduce glomerular damage and basement membrane thickening, and protect the kidneys (123,124).

In previous years, studies have reported that the hypermethylation and low expression of the let-7a-3 gene can influence the occurrence and development of DN by targeting UHRF1/DNMT1. Therefore, the let-7a-3 gene is used as the target to inhibit its hypermethylation. Moreover, it may be one of the treatment directions for DN in the future (125,126). Additionally, depletion of the calmodulin regulatory factor 1 (RCAN1) aggravates podocyte injury, impairs the glomerular filtration barrier and increases proteinuria, suggesting that RCAN1 is a protective factor for podocytes (127). However, under high glucose conditions, RCAN1 hypermethylation and decreased expression aggravates kidney injuries, which is one of the mechanisms of DN (128). Hypermethylation of insulin-like growth factor-binding protein-1 (129), methylenetetrahydrofolate reductase (130) and KLOTHO (131) promoters have been associated with DN. EGCG alleviates high glucose-induced kidney injury by reversing KLOTHO promoter methylation (131). Therefore, exploring the relationship between gene methylation and DN helps advance DN pathogenesis research and design new therapeutic strategies.

Chronic kidney disease (CKD)

CKD refers to the structural or functional impairment of the kidneys by various causes (history of renal impairment >3 months) (132). The disease burden of CKD remains substantial and continues to grow globally. Globally, incident cases and DALYs of CKD was 18.99 million and 41.54 million in 2019, respectively (133), imposing a significant societal and individual burden. The clinical manifestations of CKD in different stages are different, but it's typical feature is renal fibrosis. One of the important factors promoting renal fibrosis is epigenetic changes (134-138). Research has confirmed that the methylation of specific genes can promote CKD progression. For example, HOXA5 promoter hypermethylation can enhance the expression of Jagged 1 (JAG1) gene expression, activate the JAG1-Notch signalling pathway and promote renal fibrosis (138). Similarly, the expression of the anti-fibrotic KLOTHO gene in the kidney is decreased due to hypermethylation in the early stage of renal injury. DNMT1/3A inhibition can reduce the loss of the KLOTHO gene, attenuating renal fibrosis and structural damage (137,139). Long-term cadmium exposure induces a compensatory increase in DNMT (140) and promotes hypermethylation of RASAL1, which is associated with a decreased estimated glomerular filtration rate (141). RASAL1 hypermethylation has been confirmed as an important mechanism in CKD in vitro (87,142). The study of epigenetics in CKD offers new diagnostic and therapeutic targets, and provides a new direction for the study of pathogenesis, diagnosis and treatment strategies.

Urinary dysfunction due to neurodegeneration

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the abnormal deposition of α-synuclein (α-Syn), with the main clinical manifestation being movement disorders. With further research, non-motor symptoms of PD are gradually being recognized, with urinary dysfunction being one of the most common autonomic dysfunctions in patients with PD, affecting 27-85% of individuals with the disease (143). Although no studies have directly linked DNMT with urinary symptoms in PD, research has shown that mutations in DNMT1 can lead to defects in DNA methylation activity, resulting in neurodegenerative diseases (144). In addition, the abnormal accumulation of α-Syn in neurons of patients with PD can cause a vicious cycle of α-Syn accumulation by withholding DNMT1, leading to a decrease in the methylation level of the SNCA intron in the α-Syn gene. These findings suggest that DNMT may contribute to urinary dysfunction in PD by interacting with neurodegenerative processes (145).

Urological tumours
RCC

It is estimated that by 2024 kidney cancer will be the sixth most common cancer among men, with an incidence number of 52,380, and the ninth most common cancer among women, with an incidence number of 29,230 (2). The National Cancer Database, which conducted an epidemiological analysis of 262,597 patients diagnosed with renal cell carcinoma (2004-2015) across more than facilities, found that the metastatic rate of renal cell carcinoma was ~11 percent (146), leading to poor prognosis. For example, the 5-year survival rate for the most common clear cell RCC after metastasis is <10% (147). Therefore, early diagnosis and treatment are crucial for improving prognosis. As a malignant tumour, the role of epigenetics in RCC is significant. Overexpression of DNMTs often increases gene promoter hypermethylation, indicating gene inactivation or decreased expression ability (148). TSGs such as von Hippel-Lindau (VHL) (149), p16/CDKN2a, p14ARF, adenomatous polyposis coli (APC), RAS association domain family protein 1A (RASSF1A), B-cell translocation gene 3 (BTG3) and Timp-3 are hypermethylated in patients with RCC (150). The hypermethylation rate of the apoptotic protease-activating factor 1 promoter that induces apoptosis in RCC is ~100%, and the pro-apoptotic gene hypermethylation rate of ASC/TMS1 is ~41.1% (151,152). Promoter methylation of the HIG1 domain family member 1A and death-associated protein kinase 1 (DAPK-1) genes, which inhibit tumour metastasis, are strongly associated with RCC (153).

Additionally, promoter methylation of secreted frizzled-related protein 1 (sFRPs) (154,155), SCUBE3 (156) and GATA-5 (157,158) is associated with poor prognosis in RCC. However, surface hypomethylating agents have shown the potential to effectively reduce kidney damage and improve prognosis (159,160). Screening for specific gene methylation in the blood or urine of patients and applying demethylation drugs to the corresponding targets may become a future research directions for early non-invasive RCC diagnosis.

PCa

PCa is the fifth most common cancer in men, following lung cancer. In 2020, ~1,414,000 new cases were diagnosed, and 375,304 PCa-associated mortalities occurred worldwide (161), placing a significant burden on society. Although the prognosis of PCa is generally favourable, ~15% of the patients with high-risk forms of PCa may die within a few years (162). In the past few years, the rate of early diagnosis of PCa has increased due to the discovery of prostate-specific antigen (PSA) testing, reducing the mortality rate (163). However, due to concerns about the overdiagnosis and treatment of PCa and the complications of screening, the controversy about PSA continues, reducing the willingness to screen in certain countries and slowing the decline of PCa mortality (164,165). Therefore, identifying new targets to replace or supplement PSA screening is one of the hot spots in PCa research (166).

Epigenetic changes are one of the important characteristics of PCa, offering potential new therapeutic directions by using them as a target (167). The androgen receptor (AR) gene is hypermethylated in advanced PCa, and may be related to metastasis (168,169), providing a new prognostic biomarker for PCa. Genes such as ubiquitin carboxyl-terminal hydrolase 1 (170), APC (171), E-Cadherin (172), ASC (173), cyclin d2, glutathione S-transferase pi-1 (GSTP1), retinoic acid receptor β (RARB), CD44 and RASSF1A, which are hypermethylated and suppressed in PCa, have high specificity and sensitivity, and are associated with poor pathological classification of PCa (174,175). Prostate cells are observed in urine, which may serve as important markers for early non-invasive diagnosis and prognosis of PCa in the future (176). The recurrence of PCa after treatment and the time to recurrence are the concerns of patients. Studies have reported that the methylation of CD44 and prostaglandin-endoperoxide synthase 2 (PTGS2) genes are associated with a shorter recurrence time, offering a valuable detection target for predicting the prognosis of PCa after surgery (177). The existing research results of epigenetics are significant for PCa diagnosis and treatment. For example, antitumour drugs such as methylation inhibitors 5-azacytidine and 5-aza-2′-deoxycytidine can specifically inhibit the methylation of certain genes that suppress tumour growth and metastasis and regulate cell cycle, playing a corresponding role in curbing tumour proliferation with fewer side effects; these drugs are likely to be significant in cancer treatment in the future (170,178-181). Therefore, epigenetic modification represents promising directions in PCa research.

BCa

BCa is the most lethal malignant tumour in the urinary system, with 549,393 diagnoses and 199,922 mortalities globally in 2018 (182). The incidence and mortality of BCa continue to rise (183), significantly affecting quality of life. NMIBC has a poor prognosis, with a 5-year recurrence rate of up to 78% (184). Unlike PCa, there is currently no universally accepted screening method for BCa, and patients with NMIBC often endure repeated cystoscopy, imaging and subsequent surgery, which are physically and psychologically stressful (185). Therefore, identifying feasible targets for early non-invasive diagnosis is crucial. The pathogenesis of BCa is complex, and epigenetic changes are one of its important characteristics. The methylation of certain specific genes has been proven to be a good biomarker (186). Promoter methylation of APC is significantly increased in BCa tissue and is associated with disease occurrence and progression, making it a key biomarker for the non-invasive detection of BCa.

Other genes, such as p14ARF (187,188), O6-methylg uanine-DNA-methyltransferase (189), BCL2, CDKN2A, Nidogen (NID) (186), E-cadherin (CDH1) (190), CDH13 (191), RASSF1A (190), DAPK (192,193), fragile histidine triad (FHIT) (194), TIMP3 (195), growth/differentiation factor 15 (GDF15) and Transmembrane protein with an EGF-like and two Follistatin-like domains 2 (TMEFF2) (196) and vimentin (VIM) (196) are hypermethylated in BCa tissues, aiding early diagnosis. A methylation panel comprising of gene such as GDF15, TMEFF2 and VIM (196) has shown improved sensitivity and specificity for detection. DAPK, combined with B-ultrasound examination, has demonstrated consistency with cystoscopy results and has great significance in detecting recurrent BCa (193).

Each cancer has a specific methylation pattern (194), which makes the biomarkers specific and of great application value. However, the degree of gene methylation can vary across different populations due to ethnic differences (197), necessitating the development of other criteria based on ethnic differences. The degree of gene methylation is associated with aging, highlighting aging as a risk factors for BCa (190). These epigenetic biomarkers are important targets for early non-invasive diagnosis and treatment of BCa, and are indicators of treatment efficacy and prognosis. They help to explore the pathogenesis of BCa and identify risk factors related to BCa.

Molecular mechanisms of DNMTs in urinary system diseases

Decline in kidney function

The kidneys are closely related to the urinary system. In clinical practice, if renal function is impaired, it often leads to abnormal urinary drainage function of the urinary system. Studies have shown that epigenetic repair, especially DNA methylation, can severely affect kidney function. Oxidative stress is a major pathological factor contributing to aging-related diseases. Nuclear factor erythroid-derived 2-like 2 (NRF2) and KLOTHO, two major antiaging factors with antioxidant capacities, are suppressed with aging, which is associated with increased incidence of aging-related kidney disorders. A recent study has shown that upregulation of DNMT1/3a/3b expression results in promoter hypermethylation of NRF2 and KLOTHO and decreased expression of NRF2 and KLOTHO, resulting in structural and functional alterations in renal senescence (198). NRF2 inhibition significantly promotes ROS production while suppressing the antioxidant capacity of renal TEC under Ang II conditions (199). Zhao et al (200) showed that ROS production in turn promotes the up-regulation of DNMT1/3a/3b expression, leading to hypermethylation of the NDRG promoter and promoting renal fibrosis. Moreover, ROS activate the NF-κB signalling pathway to induce mitochondrial dysfunction, enhance autophagy and promote apoptosis in renal tubular epithelial cells (201). The Y-box binding protein-1 (YB-1) regulates fibrosis-related genes (for example, Col1a1, Mmp2 and Tgfβ1) and contributes significantly to disease progression. YB-1 binds to specific structural motifs in DNA, thereby recruiting DNMT to the promoter of KLOTHO, resulting in a decrease in KLOTHO expression, which subsequently leads to activation of the Wnt/β-catenin and TGFβ/Smad signalling pathways (202,203). Activation of the Wnt/β-catenin signalling pathway inhibits the mitochondrial biogenesis regulator Pparg coactivator 1α in renal tubular cells and reduces the mitochondrial membrane potential, oxygen consumption and ATP production, which is closely related to renal fibrosis (204). Whereas TGF-β increases DNMT1 and DNMT3a expression by inhibiting miR-152 and miR-30a in fibrotic kidneys thereby inhibiting KLOTHO expression (137). In addition, a recent study by Xiao et al found that the expression of DNMT1, DNMT3a and DNMT3b are significantly increased in CKD, which leads to the hypermethylation of the promoter of HOXA4 and decreases its expression (138). A decrease in the expression of HOXA5 increases the expression of JAG1, which activates the NOTCH signalling pathway, resulting in fibrosis in the kidney (Fig. 3) (138,205).

Development of tumours of the urinary system

Aberrant DNA methylation (genome-wide hypomethylation and site-specific hypermethylation) are observed in numerous types of cancer (206-208). Abnormal DNA methylation has been shown to be strongly associated with numerous types of cancers, particularly with tumour suppressor genes, which play an important role in tumourigenesis and development (209). The active methyl group is transferred to the covalent bond at the 5′ carbon position of cytosine of tumour suppressor genes mediated by the DNMT family, using S-adenosylmethylsulfonine as the methyl carrier. Silencing of gene expression due to hypermethylation of TSGs promoter regions is an important driver of tumourigenesis and progression (210). It remains less well understood how DNMTs are targeted to specific loci in the context of aberrant hypermethylation in cancer cells (211,212). Two possible molecular mechanisms have been proposed (213), one being that during genotoxic stress-induced DNA deletion, the aggregation of DNMT, histone deacetylase complexes and multi-comb complexes at CpG-rich promoters throughout the genome inhibits transcription (214). Alternatively, loss of ten-eleven translocation (TET) activity in cancer cells results in shortening of CpG islands and decreased gene expression through histone H3 lysine 4 mono-methylation (H3K4me1)-induced de novo DNMT invasion of DNA methylation (213). Early findings suggested that DNMT-mediated focal hypermethylation of TSGs directly triggers transcriptional silencing of TSGs and activation of signalling pathways such as Wnt/β-catenin (215), PI3K/AKT (216) and MAPK (217), which promotes cancerous aggravation. These three pathways play important roles in the occurrence and development of cancer.

Multiple studies have shown that Wnt plays a role in maintaining stem cells, assisting their self-renewal and cell generation capacity (218,219), and its abnormal changes are associated with the occurrence and development of a variety of cancers (220,221). For example, APC inactivation leads to the abnormal activation of β-catenin, thereby affecting the Wnt/β-catenin signalling pathway (222), leading to the progression of familial adenomatosis (FAP) to colorectal cancer (223). The phosphatidylinositol 3-kinase (PI3K/Akt) signalling pathway functions in regulating cell cycle, differentiation and metabolism (224), so the abnormality of this signalling pathway is common in cancer. The channel PI3K3A gene mutations or phosphate and tensin homolog (PTEN) gene promoter methylation leading to the decrease of the expression level is often associated with the development of breast cancer (225-227). The serine/threonine protein kinase MAPK, also known as ERK, is a key component of the Raf/MEK/ERK signalling pathway (228). Alterations in this pathway are the most common in human cancers and promote malignant proliferation of cancer cells (229). These three pathways are closely related to DNMT, suggesting that the methylation machinery is involved in the mutation and activation of the pathway (Fig. 4).

Promoter hypermethylation of TSGs is often the main mechanism of their loss of function in prostate cancer. Overall, >30 genes have been reported to be abnormally hypermethylated in prostate cancer, including genes involved in hormonal response, cell cycle regulation, cell invasion and DNA damage repair. Inappropriate silencing of these genes can lead to the occurrence, progression, invasion and metastasis of prostate cancer. The present review found that miR-148a, miR-152 and miR-200b increase DNMT1 expression, which results in the reduction of PTEN expression, which leads to the activation of the PIK3/AKT signalling pathway. In addition, loss of PTEN may lead to decreased expression of NK3 homeobox 1, thereby up-regulating AR-related signalling (230). Dickkopf Wnt signalling pathway inhibitor 3 (DKK3) inhibits the transmission of Wnt/β-catenin signalling pathway, and studies have shown that its expression is decreased in prostate cancer (231-233). Bhattacharyya et al (234) showed that increased DNMT activity and expression lead to methylation of the DKK3 promoter, which leads to the activation of Wnt/β-catenin signalling.

In renal cancers, the p16 and Thrombospondin 1 (THBS-1) genes seem to be hot spots of regional DNA hypermethylation during multistage renal tumourigenesis (235). Inhibition of THBS-1 expression can activate the PI3K/AKT pathway (236). In addition, KLOTHO, a tumour suppressor gene, normally binds to Wnt ligands to block the Wnt pathway (237-240). When DNMT activity is increased, KLOTHO methylation expression is decreased, and Wnt can be over-activated synergistically with TGF-β to induce epithelial-mesenchymal transition (241) and promote the differentiation of cancer stem cells (242), which can promote the occurrence and development of renal cell carcinoma. In conclusion, DNMT may down-regulate THBS-1 through the methylation of THSB-1 promoter and KLOTHO promoter to activate the PI3K/AKT and Wnt signalling pathways to promote cell proliferation in renal cell carcinoma.

In the detection of methylation of bladder cancer protective genes that are also Wnt antagonist genes such as secreted frizzled-related protein 1 (SFRP1), WNT inhibitory factor-1 (WIF1), APC and CDH1, it was found that in all the samples, at least one gene was found to be methylated and then down-regulated, leading to excessive activation of Wnt pathway and uncontrolled proliferation of bladder cells, which promote the occurrence and development of bladder cancer (243,244). In addition to antagonising the Wnt pathway, the SFRP1 gene also inhibits the MAPK pathway (233,245). Therefore, when the activity of DNMT is increased, SFRP1 in the methylated state tends to promote the proliferation of bladder tumour cells. Decreased expression of a tumour suppressor gene may promote cancer cell proliferation through multiple mechanisms, so the importance of methylation is self-evident.

Potential clinical value of DNMT in human urinary system disease

Epigenetic changes are the common mechanism of a variety of diseases in the urinary system. Methylation may lead to specific gene expression decrease or even inactivation, affecting the function of the gene, and eventually leading to lesions (246). Fortunately, there are a variety of studies exploring how to use this feature for clinical use. DNA methylation plays an important role in early diagnosis, accurate treatment and prognosis evaluation. Firstly, some malignant tumours have been found to be associated with the methylation of tumour suppressor genes (247), and the present review found that there are some methylated genes in urinary system diseases such as kidney cancer, bladder cancer, prostate cancer and other malignant tumours that are specific due to the different methylation patterns of different cancers (194). As methylation variation can exist in the whole genome (248), Epigenetic Cancer of the Prostate Test in Urine (epiCaPture), as a detection method for high-risk PCA urine DNA methylation, measures the DNA hypermethylation in the 5′ regulatory region of six genes (GSTP1, SFRP2, IGFBP3, IGFBP7, APC and PTGS2), all of which had previously been reported in PCa combined with PSA, the sensitivity and specificity of PCA assay are >70% (249). DNA methylation is stable and can be detected in almost any body fluid or tissue of the human body (250). Therefore, it is also non-invasive and effective in alleviating the pain of patients. In addition, methylated genes can also be combined with other examination methods such as DAPK combined with B ultrasound examination, which has been confirmed to be as accurate as cystoscopy (193).

At present, a variety of methylated genes have been found to have application value in the diagnosis of diseases. As an epigenetic marker in prostate cancer, GSTP1 has been shown to be hypermethylated in cancer tissues and various body fluid (251). Detection of GSTP1 in urine is even more relevant to the disease status than analysis of Gleason score in biopsy (252). ConfirmMDx, developed by MDxHealth Laboratories, has a negative predictive value of >90% for hypermethylated GSTP1, RASSF1 and APC (253). Therefore, the present study hypothesises that methylated genes can be used as one of the detection markers for early diagnosis in the future. Restoring the function of genes inactivated by methylation is the key to study the treatment strategy. Studies have revealed that reactivating tumour suppressor genes inactivated by methylation through targeted therapy may help treat specific types of cancer (254,255).

In addition to this, the use of specific drugs or techniques to intervene in the DNA methylation process has received widespread attention. For example, the expression of genes silenced by methylation is restored by inhibiting the activity of DNA methyltransferases or facilitating the action of DNA demethylases. At present, a variety of studies have found that DNMT inhibitors or demethylases play an important role in the cure of diseases, including diabetic nephropathy, chronic kidney disease, renal cell carcinoma, prostate cancer and bladder cancer (Table I) (160,198,256-260). For example, RG108, a non-nucleoside inhibitor of DNMT, exerts an antitumour effect by reversing the silencing of antitumour genes such as GSTP1 and APC in prostate cancer (258). In addition to its effect on malignant tumours, DNMT inhibitor SGI-1072 can also reduce renal aging changes by reversing the methylation of anti-aging factor KLOTHO and enhancing its expression (198), and this mechanism is related to the inhibition of oxidative stress, fibrosis and apoptosis (261). OLP, a DNMT inhibitor similar to SGI-1072, has been found to have a demethylation ability in D-galactose-induced senescent renal cells, effectively reducing DNMT1/3a/3b expression, decreasing the loss of antioxidant senescence factors KLOTHO and NRF2, and then ameliorating renal cellular senescence (198). Thus, they may also have a role in chronic kidney disease.

Table I

Abnormally methylated genes and DNMT involved in corresponding processes in urinary system diseases.

Table I

Abnormally methylated genes and DNMT involved in corresponding processes in urinary system diseases.

Urinary system diseaseDNMTHypermethylated genesDNMT inhibitor(Refs.)
Cystitis-CDKN2A, RASSF1A TIMP3, Calca, Timp3 Mmp2, Igf2r, Ogg-(98,103,104)
Hypertensive nephropathyDNMT1
DNMT3a
SOD, Strt1-(113-114)
Diabetic nephropathyDNMT1
DNMT3a
ACTN4, let-7a-3, RCAN1, MTHFR, KLOTHO,5-Aza(123,125-127, 130-131)
Chronic kidney diseaseDNMT1
DNMT3a
HOXA5, KLOTHO, RASAL1SGI-1072, OLP(138,139,141)
Renal cell carcinomaDNMT3b
DNMT3a
DNMT1
VHL, p16/CDKN2a, p14ARF, APC, RASSF1A, BTG3, Timp-3, APAF-1, ASC/TMS1, HIGD1A, DAPK-1, SCUBE3, GATA-5Genistein 5-azacytidine(148-160)
Prostate cancerDNMT3bUCHL1, APC, E-cadherin, ASC, cyclin d2, GSTP1, RARB, CD44, RASSF1A, PTGS2, GSTP, SFRP2, IGFBP3, IGFBP7,Decitabine; RG108(170-177,244, 249,271,272)
Bladder cancerDNMT3b
DNMT3a
APC, MGMT, BCL2, CDKN2A, NID, CDH1, CDH13, p14ARF, RASSF1A, DAPK, FHIT, TIMP3, TMEFF2, GDF15, VIM,Decitabine; 5-azacytidine(186-196)
Neurodegeneration-SNCA-(145)

[i] DNMT, DNA methyltransferase.

In addition, DNMT inhibitors can also be combined with other drugs, such as synergism with the HDAC inhibitor TSC, to effectively inhibit the proliferation of bladder cancer (259). As a pan-DNMT inhibitor, decitabine exerts important antitumour effects in highly DNMT-expressing neuroendocrine prostate cancer, inhibiting tumour progression mainly by suppressing the expression of neuroendocrine profile markers (77). Further study has shown that decitabine can also increase the expression of the drug target B7-h3, and an experiment has demonstrated that decitabine combined with the B7-h3-targeting agent DS-7300a (i-DXd) can effectively enhance the anti-tumour effect (262). As early as 2009, the combination of 5-Azacytidine with cisplatin has been reported to improve antitumour effects and reduce nephrotoxicity (263). 5-Azacytidine inhibits the expression of genes related to oxidative stress and reduces metallothionein gene expression, which reduces nephrotoxicity (263,264). 5-Azacytidine is also known to inhibit the expression of genes related to oxidative stress and reduce metallothionein gene expression, which reduces nephrotoxicity (263,264). The The combination of 5-Azacytidine with cisplatin has been reported to increase sensitivity and resistance (265,266). The reversal of methylation is relatively mild, and the side effects of DNMT inhibitors are relatively small (267), which can also effectively improve the quality of life of patients.

In addition, epigenetic changes, especially gene methylation, can respond to a variety of mechanisms, so it is also beneficial to explore the aetiology and pathogenesis of diseases. For example, if a certain gene methylation degree is found to be higher in hypertensive nephropathy and diabetic nephropathy than in healthy individuals, it can be verified by designed experiments when the causal relationship is not clear, which is helpful to find new treatment ideas. Finally, the prognosis of the disease can also be determined according to the degree of gene methylation. Several studies have confirmed that the hypermethylation of the BNC1 (268), GATA5 (157) and SCUBE3 (156) promoters often predicts poor prognosis of patients with renal cell carcinoma. The methylation levels of LAD1 (154,269) and NEFH (270) can also reflect the outcome of drug treatment of renal cell carcinoma to a certain extent, helping physicians to adjust the treatment plan in time. The promoters of p16INK4a and p14ARF are hypermethylated in bladder cancer, and the degree of methylation is related to the malignant degree of bladder cancer. For example, the methylation of invasive bladder cancer is higher than that of superficial bladder cancer (188). The recurrence of prostate cancer after treatment and the time to recurrence are the concerns of patients. Studies have found that the methylation of CD44 is correlated with progression and metastasis in prostate cancer (271,272) and Yegnasubramanian et al (273) indicated that the hypermethylation of PTGS2 CpG islands predicted an increased risk of prostate cancer recurrence in 2004. Moreover, the methylation of CD44 and PTGS2 genes are associated with a shorter recurrence time, which provides a good detection target for predicting the prognosis of prostate cancer after surgery (177).

Future expectations

Rapid advances in the field of epigenetics not only reveal complex genetic phenomena at the molecular level, but also offer new hope for the treatment of diseases, especially cancer (274). Epigenetic changes have reversible properties, meaning that biological traits can be affected by reversing these changes under certain conditions. DNA methylation, histone modification and regulation of non-coding RNA have become the focus of epigenetic therapy. Currently, several epigenetic-based treatments have been developed, such as DNMTi. However, the main disadvantage of these methods is their lack of specificity, which can lead to off-target effects and other side effects (275-277). The CRISPR/Cas9 technology is a powerful tool that is currently widely used for genome editing. Its modular design is not only suitable for direct modification of DNA, but can also be used to introduce specific epigenetic modifications, such as methylation or demethylation (278). dCas9 fuses with the catalytic domain of DNMT or TET proteins, and the CRISPR/DCas9-DNMT or DCas9-TET system enables precise editing of DNA methylation. Liang et al (279) demonstrated that this fusion protein enables site-specific DNA methylation editing in zebrafish.

Because zebrafish embryos are transparent, grow rapidly and are highly homologous to human genes, they are an ideal model for studying vertebrates. Therefore, DNMTs fusion technology combined with CRISPR/Cas9 system has the potential to be used to specifically edit human DNA methylation in the future.

In the field of urinary tumours, some key epigenetic changes have also been found in prostate cancer. For example, GSTP1 and HOX family member genes are inhibited by hypermethylation in the promoter region. Kardooni et al (232) reported that downregulation of the DKK3 gene is associated with the growth of prostate tumours. The Dkk-3 protein encoded by the DKK3 gene maintains the stable state of normal prostate epithelial cells by regulating the TGF-β/Smad signalling pathway (261). Going forward, using CRISPR/dCas9-DNMT technology, it will be possible to regulate the expression of these genes through methylation or demethylation, thereby controlling tumour growth.

Conclusion

In conclusion, with the deepening understanding of epigenetics, more and more studies have demonstrated that DNMT-mediated aberrant methylation of genes plays an important role in the development of urinary system diseases. The present review systematically reviewed the mechanism and clinical value of DNMT in a variety of urinary system diseases. We found that targeting DNMT to find corresponding therapeutic approaches is of great significance in the treatment of urinary system diseases. It is hypothesised that DNMT will provide novel therapeutic ideas for urinary system diseases.

Availability of data and materials

Not applicable.

Authors' contributions

AW, YY and HW conceived the study. YY, YW, XF and XX wrote and edited the manuscript. XW, TS, YG, SL, AW and JT revised the manuscript. AW put forward constructive opinions on the topic selection of the article. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

This work was supported by the Weifang Municipal Health Commission Project (grant no. WFWSJK-2021-140) and Shandong Province Medicine and Health Science and Technology Development Programme Project (grant no. 2021BJ000026).

References

1 

Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Giaquinto AN and Jemal A: Cancer statistics, 2024. CA Cancer J Clin. 74:12–49. 2024. View Article : Google Scholar : PubMed/NCBI

3 

Zi H, Liu MY, Luo LS, Huang Q, Luo PC, Luan HH, Huang J, Wang DQ, Wang YB, Zhang YY, et al: Global burden of benign prostatic hyperplasia, urinary tract infections, urolithiasis, bladder cancer, kidney cancer, and prostate cancer from 1990 to 2021. Mil Med Res. 11:642024.PubMed/NCBI

4 

Tian YQ, Yang JC, Hu JJ, Ding R, Ye DW and Shang JW: Trends and risk factors of global incidence, mortality, and disability of genitourinary cancers from 1990 to 2019: Systematic analysis for the global burden of disease study 2019. Front Public Health. 11:11193742023. View Article : Google Scholar : PubMed/NCBI

5 

Chen JQ, Salas LA, Wiencke JK, Koestler DC, Molinaro AM, Andrew AS, Seigne JD, Karagas MR, Kelsey KT and Christensen BC: Immune profiles and DNA methylation alterations related with non-muscle-invasive bladder cancer outcomes. Clin Epigenetics. 14:142022. View Article : Google Scholar : PubMed/NCBI

6 

Unnikrishnan A, Freeman WM, Jackson J, Wren JD, Porter H and Richardson A: The role of DNA methylation in epigenetics of aging. Pharmacol Ther. 195:172–185. 2019. View Article : Google Scholar :

7 

Singh NP and Vinod PK: Integrative analysis of DNA methylation and gene expression in papillary renal cell carcinoma. Mol Genet Genomics. 295:807–824. 2020. View Article : Google Scholar : PubMed/NCBI

8 

Deng Q, Du Y, Wang Z, Chen Y, Wang J, Liang H and Zhang D: Identification and validation of a DNA methylation-driven gene-based prognostic model for clear cell renal cell carcinoma. BMC Genomics. 24:3072023. View Article : Google Scholar : PubMed/NCBI

9 

Lyko F: The DNA methyltransferase family: A versatile toolkit for epigenetic regulation. Nat Rev Genet. 19:81–92. 2018. View Article : Google Scholar

10 

Castillo-Aguilera O, Depreux P, Halby L, Arimondo PB and Goossens L: DNA methylation targeting: The DNMT/HMT crosstalk challenge. Biomolecules. 7:32017. View Article : Google Scholar : PubMed/NCBI

11 

Ferreira HJ and Esteller M: CpG islands in cancer: Heads, tails, and sides. Methods Mol Biol. 1766:49–80. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Wu H, Caffo B, Jaffee HA, Irizarry RA and Feinberg AP: Redefining CpG islands using hidden Markov models. Biostatistics. 11:499–514. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Rodriguez-Paredes M and Esteller M: Cancer epigenetics reaches mainstream oncology. Nat Med. 17:330–339. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Li J, Liang Y, Fan J, Xu C, Guan B, Zhang J, Guo B, Shi Y, Wang P, Tan Y, et al: DNA methylation subtypes guiding prognostic assessment and linking to responses the DNA methyltransferase inhibitor SGI-110 in urothelial carcinoma. BMC Med. 20:2222022. View Article : Google Scholar : PubMed/NCBI

15 

Yu JT, Hu XW, Chen HY, Yang Q, Li HD, Dong YH, Zhang Y, Wang JN, Jin J, Wu YG, et al: DNA methylation of FTO promotes renal inflammation by enhancing m6A of PPAR-α in alcohol-induced kidney injury. Pharmacol Res. 163:1052862021. View Article : Google Scholar

16 

Gu Y, Niu S, Wang Y, Duan L, Pan Y, Tong Z, Zhang X, Yang Z, Peng B, Wang X, et al: DMDRMR-mediated regulation of m6A-Modified CDK4 by m6A reader IGF2BP3 drives ccRCC progression. Cancer Res. 81:923–934. 2021. View Article : Google Scholar

17 

Sandholm N, Cole JB, Nair V, Sheng X, Liu H, Ahlqvist E, van Zuydam N, Dahlström EH, Fermin D, Smyth LJ, et al: Genome-wide meta-analysis and omics integration identifies novel genes associated with diabetic kidney disease. Diabetologia. 65:1495–1509. 2022. View Article : Google Scholar : PubMed/NCBI

18 

Johnston WWS: Diseases of the kidneys and urinary system. Med J Aust. 2:57–59. 1947.PubMed/NCBI

19 

Correa-Forero V, Pinilla-Monsalve GD, Valderrama-Chaparro JA and Amaya-Gonzalez P: Cryptococcal meningitis presenting as acute flaccid paralysis: A case report. J Infect Public Health. 13:143–148. 2020. View Article : Google Scholar

20 

Sampaolo S, Esposito T, Gianfrancesco F, Napolitano F, Lombardi L, Lucà R, Roperto F and Di Iorio G: A novel GBE1 mutation and features of polyglucosan bodies autophagy in adult polyglucosan body disease. Neuromuscul Disord. 25:247–252. 2015. View Article : Google Scholar

21 

Roberts NA, Hilton EN, Lopes FM, Singh S, Randles MJ, Gardiner NJ, Chopra K, Coletta R, Bajwa Z, Hall RJ, et al: Lrig2 and Hpse2, mutated in urofacial syndrome, pattern nerves in the urinary bladder. Kidney Int. 95:1138–1152. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Liao L: Evaluation and management of neurogenic bladder: What is new in China? Int J Mol Sci. 16:18580–18600. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Yoshimura N, Ogawa T, Miyazato M, Kitta T, Furuta A, Chancellor MB and Tyagi P: Neural mechanisms underlying lower urinary tract dysfunction. Korean J Urol. 55:81–90. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Archer M, Dogra N, Dovey Z, Ganta T, Jang HS, Khusid JA, Lantz A, Mihalopoulos M, Stockert JA, Zahalka A, et al: Role of α- and β-adrenergic signaling in phenotypic targeting: Significance in benign and malignant urologic disease. Cell Commun Signal. 19:782021. View Article : Google Scholar

25 

Chen N, Li G, Si Y, Ye Y, Zhang T, Chi D, Zhang W, Pan L, Qu G, Lu Y, et al: Development and evaluation of a centrifugal disk system for the rapid detection of multiple pathogens and their antibiotic resistance genes in urinary tract infection. Front Microbiol. 14:11574032023. View Article : Google Scholar : PubMed/NCBI

26 

Simões E Silva AC, Oliveira EA and Mak RH: Urinary tract infection in pediatrics: An overview. J Pediatr (Rio J). 96(Suppl 1): S65–S79. 2020. View Article : Google Scholar

27 

Zulfiqar M, Ubilla CV, Nicola R and Menias CO: Imaging of renal infections and inflammatory disease. Radiol Clin North Am. 58:909–923. 2020. View Article : Google Scholar : PubMed/NCBI

28 

Bachar A, Itzhaki E, Gleizer S, Shamshoom M, Milo R and Antonovsky N: Point mutations in topoisomerase I alter the mutation spectrum in E. coli and impact the emergence of drug resistance genotypes. Nucleic Acids Res. 48:761–769. 2020. View Article : Google Scholar :

29 

Price LB, Johnson JR, Aziz M, Clabots C, Johnston B, Tchesnokova V, Nordstrom L, Billig M, Chattopadhyay S, Stegger M, et al: The epidemic of extended-spectrum-β-lactama se-producing Escherichia coli ST131 is driven by a single highly pathogenic subclone, H30-Rx. mBio. 4:e00377–13. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Beebout CJ, Robertson GL, Reinfeld BI, Blee AM, Morales GH, Brannon JR, Chazin WJ, Rathmell WK, Rathmell JC, Gama V and Hadjifrangiskou M: Uropathogenic Escherichia coli subverts mitochondrial metabolism to enable intracellular bacterial pathogenesis in urinary tract infection. Nat Microbiol. 7:1348–1360. 2022. View Article : Google Scholar : PubMed/NCBI

31 

Spradling K, Ganesan C and Conti S: Medical treatment and prevention of urinary stone disease. Urol Clin North Am. 49:335–344. 2022. View Article : Google Scholar : PubMed/NCBI

32 

Hong SY, Xia QD, Yang YY, Li C, Zhang JQ, Xu JZ, Qin BL, Xun Y and Wang SG: The role of microbiome: A novel insight into urolithiasis. Crit Rev Microbiol. 49:177–196. 2023. View Article : Google Scholar

33 

Hobbs T, Schultz LN, Lauchnor EG, Gerlach R and Lange D: Evaluation of biofilm induced urinary infection stone formation in a novel laboratory model system. J Urol. 199:178–185. 2018. View Article : Google Scholar

34 

Sen H, Seckiner I, Bayrak O, Erturhan S and Demirbağ A: Treatment alternatives for urinary system stone disease in preschool aged children: Results of 616 cases. J Pediatr Urol. 11:34.e1–e5. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Chen F, Zhang Y, Bossé D, Lalani AA, Hakimi AA, Hsieh JJ, Choueiri TK, Gibbons DL, Ittmann M and Creighton CJ: Pan-urologic cancer genomic subtypes that transcend tissue of origin. Nat Commun. 8:1992017. View Article : Google Scholar : PubMed/NCBI

36 

Markowski MC, Boorjian SA, Burton JP, Hahn NM, Ingersoll MA, Maleki Vareki S, Pal SK and Sfanos KS: The microbiome and genitourinary cancer: A collaborative review. Eur Urol. 75:637–646. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

38 

Rouprêt M, Babjuk M, Compérat E, Zigeuner R, Sylvester RJ, Burger M, Cowan NC, Gontero P, Van Rhijn BWG, Mostafid AH, et al: European association of urology guidelines on upper urinary tract urothelial carcinoma: 2017 Update. Eur Urol. 73:111–122. 2018. View Article : Google Scholar

39 

Li Z, Xu H, Gong Y, Chen W, Zhan Y, Yu L, Sun Y, Li A, He S, Guan B, et al: Patient-derived upper tract urothelial carcinoma organoids as a platform for drug screening. Adv Sci (Weinh). 9:e21039992022. View Article : Google Scholar

40 

Zhang ML, Miki Y, Hang JF, Vohra M, Peyton S, McIntire PJ, VandenBussche CJ and Vohra P: A review of upper urinary tract cytology performance before and after the implementation of The Paris system. Cancer Cytopathol. 129:264–274. 2021. View Article : Google Scholar

41 

Bus MTJ, de Bruin DM, Faber DJ, Kamphuis GM, Zondervan PJ, Laguna Pes MP, de Reijke TM, Traxer O, van Leeuwen TG and de la Rosette JJ: Optical diagnostics for upper urinary tract urothelial cancer: Technology, thresholds, and clinical applications. J Endourol. 29:113–123. 2015. View Article : Google Scholar

42 

He Y, Xu W, Xiao YT, Huang H, Gu D and Ren S: Targeting signaling pathways in prostate cancer: Mechanisms and clinical trials. Signal Transduct Target Ther. 7:1982022. View Article : Google Scholar : PubMed/NCBI

43 

Kausar S, Abbas MN and Cui H: A review on the DNA methyltransferase family of insects: Aspect and prospects. Int J Biol Macromol. 186:289–302. 2021. View Article : Google Scholar : PubMed/NCBI

44 

Uysal F and Ozturk S: DNA methyltransferases in mammalian oocytes. Results Probl Cell Differ. 63:211–222. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Okano M, Bell DW, Haber DA and Li E: DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 99:247–257. 1999. View Article : Google Scholar : PubMed/NCBI

46 

Ren W, Gao L and Song J: Structural basis of DNMT1 and DNMT3A-mediated DNA methylation. Genes (Basel). 9:6202018. View Article : Google Scholar : PubMed/NCBI

47 

Chen Z and Zhang Y: Role of mammalian DNA methyltransferases in development. Annu Rev Biochem. 89:135–158. 2020. View Article : Google Scholar

48 

Poh WJ, Wee CP and Gao Z: DNA methyltransferase activity assays: Advances and challenges. Theranostics. 6:369–391. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Takeshima H, Niwa T, Yamashita S, Takamura-Enya T, Iida N, Wakabayashi M, Nanjo S, Abe M, Sugiyama T, Kim YJ and Ushijima T: TET repression and increased DNMT activity synergistically induce aberrant DNA methylation. J Clin Invest. 130:5370–5379. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Huang J, Fan T, Yan Q, Zhu H, Fox S, Issaq HJ, Best L, Gangi L, Munroe D and Muegge K: Lsh, an epigenetic guardian of repetitive elements. Nucleic Acids Res. 32:5019–5028. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Tao Y, Xi S, Shan J, Maunakea A, Che A, Briones V, Lee EY, Geiman T, Huang J, Stephens R, et al: Lsh, chromatin remodeling family member, modulates genome-wide cytosine methylation patterns at nonrepeat sequences. Proc Natl Acad Sci USA. 108:5626–5631. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Han M, Li J, Cao Y, Huang Y, Li W, Zhu H, Zhao Q, Han JJ, Wu Q, Li J, et al: A role for LSH in facilitating DNA methylation by DNMT1 through enhancing UHRF1 chromatin association. Nucleic Acids Res. 48:12116–12134. 2020. View Article : Google Scholar : PubMed/NCBI

53 

Xing H, Wang C, Wu H, Chen D, Li S and Xu S: Effects of atrazine and chlorpyrifos on DNA methylation in the brain and gonad of the common carp. Comp Biochem Physiol C Toxicol Pharmacol. 168:11–19. 2015. View Article : Google Scholar

54 

Wang C, Zhang Z, Yao H, Zhao F, Wang L, Wang X, Xing H and Xu S: Effects of atrazine and chlorpyrifos on DNA methylation in the liver, kidney and gill of the common carp (Cyprinus carpio L.). Ecotoxicol Environ Saf. 108:142–151. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Wirbisky-Hershberger SE, Sanchez OF, Horzmann KA, Thanki D, Yuan C and Freeman JL: Atrazine exposure decreases the activity of DNMTs, global DNA methylation levels, and dnmt expression. Food Chem Toxicol. 109:727–734. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Svedružić ŽM: Dnmt1 structure and function. Prog Mol Biol Transl Sci. 101:221–254. 2011. View Article : Google Scholar

57 

Duraisamy AJ, Mishra M, Kowluru A and Kowluru RA: Epigenetics and Regulation of oxidative stress in diabetic retinopathy. Invest Ophthalmol Vis Sci. 59:4831–4840. 2018. View Article : Google Scholar : PubMed/NCBI

58 

Zhao L, Sun MA, Li Z, Bai X, Yu M, Wang M, Liang L, Shao X, Arnovitz S, Wang Q, et al: The dynamics of DNA methylation fidelity during mouse embryonic stem cell self-renewal and differentiation. Genome Res. 24:1296–1307. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Mohan KN: DNMT1: Catalytic and non-catalytic roles in different biological processes. Epigenomics. 14:629–643. 2022. View Article : Google Scholar : PubMed/NCBI

60 

Arand J, Spieler D, Karius T, Branco MR, Meilinger D, Meissner A, Jenuwein T, Xu G, Leonhardt H, Wolf V and Walter J: In vivo control of CpG and non-CpG DNA methylation by DNA methyltransferases. PLoS Genet. 8:e10027502012. View Article : Google Scholar : PubMed/NCBI

61 

Wang Q, Yu G, Ming X, Xia W, Xu X, Zhang Y, Zhang W, Li Y, Huang C, Xie H, et al: Imprecise DNMT1 activity coupled with neighbor-guided correction enables robust yet flexible epigenetic inheritance. Nat Genet. 52:828–839. 2020. View Article : Google Scholar : PubMed/NCBI

62 

Echeverria F, Ortiz M, Valenzuela R and Videla LA: Long-chain polyunsaturated fatty acids regulation of PPARs, signaling: Relationship to tissue development and aging. Prostaglandins Leukot Essent Fatty Acids. 114:28–34. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Ceccarelli V, Ronchetti S, Marchetti MC, Calvitti M, Riccardi C, Grignani F and Vecchini A: Molecular mechanisms underlying eicosapentaenoic acid inhibition of HDAC1 and DNMT expression and activity in carcinoma cells. Biochim Biophys Acta Gene Regul Mech. 1863:1944812020. View Article : Google Scholar : PubMed/NCBI

64 

Zhao D, Gao Y, Su Y, Zhou Y, Yang T, Li Y, Wang Y, Sun Y, Chen L, Zhang F, et al: Oroxylin A regulates cGAS DNA hypermethylation induced by methionine metabolism to promote HSC senescence. Pharmacol Res. 187:1065902023. View Article : Google Scholar

65 

van der Wijst MG, Venkiteswaran M, Chen H, Xu GL, Plösch T and Rots MG: Local chromatin microenvironment determines DNMT activity: From DNA methyltransferase to DNA demethylase or DNA dehydroxymethylase. Epigenetics. 10:671–676. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Wolffgramm J, Buchmuller B, Palei S, Muñoz-López Á, Kanne J, Janning P, Schweiger MR and Summerer D: Light-activation of DNA-methyltransferases. Angew Chem Int Ed Engl. 60:13507–13512. 2021. View Article : Google Scholar : PubMed/NCBI

67 

Correa LO, Jordan MS and Carty SA: DNA methylation in T-cell development and differentiation. Crit Rev Immunol. 40:135–156. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Lee PP, Fitzpatrick DR, Beard C, Jessup HK, Lehar S, Makar KW, Pérez-Melgosa M, Sweetser MT, Schlissel MS, Nguyen S, et al: A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. Immunity. 15:763–774. 2001. View Article : Google Scholar : PubMed/NCBI

69 

Ktena YP, Koldobskiy MA, Barbato MI, Fu HH, Luznik L, Llosa NJ, Haile A, Klein OR, Liu C, Gamper CJ and Cooke KR: Donor T cell DNMT3a regulates alloreactivity in mouse models of hematopoietic stem cell transplantation. J Clin Invest. 132:e1580472022. View Article : Google Scholar : PubMed/NCBI

70 

Lu CH, Wu CJ, Chan CC, Nguyen DT, Lin KR, Lin SJ, Chen LC, Yen JJ and Kuo ML: DNA methyltransferase inhibitor promotes human CD4+CD25hFOXP3+ regulatory T lymphocyte induction under suboptimal TCR stimulation. Front Immunol. 7:4882016.

71 

Lv Q, Shi C, Qiao S, Cao N, Guan C, Dai Y and Wei Z: Alpinetin exerts anti-colitis efficacy by activating AhR, regulating miR-302/DNMT-1/CREB signals, and therefore promoting Treg differentiation. Cell Death Dis. 9:8902018. View Article : Google Scholar : PubMed/NCBI

72 

Ehrlich M: DNA hypermethylation in disease: Mechanisms and clinical relevance. Epigenetics. 14:1141–1163. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Ellinger J, El Kassem N, Heukamp LC, Matthews S, Cubukluoz F, Kahl P, Perabo FG, Müller SC, von Ruecker A and Bastian PJ: Hypermethylation of cell-free serum DNA indicates worse outcome in patients with bladder cancer. J Urol. 179:346–352. 2008. View Article : Google Scholar

74 

Marsit CJ, Karagas MR, Danaee H, Liu M, Andrew A, Schned A, Nelson HH and Kelsey KT: Carcinogen exposure and gene promoter hypermethylation in bladder cancer. Carcinogenesis. 27:112–116. 2006. View Article : Google Scholar

75 

Yates DR, Rehman I, Abbod MF, Meuth M, Cross SS, Linkens DA, Hamdy FC and Catto JW: Promoter hypermethylation identifies progression risk in bladder cancer. Clin Cancer Res. 13:2046–2053. 2007. View Article : Google Scholar : PubMed/NCBI

76 

Thompson D, Lawrentschuk N and Bolton D: New approaches to targeting epigenetic regulation in bladder cancer. Cancers (Basel). 15:18562023. View Article : Google Scholar : PubMed/NCBI

77 

Nunes SP, Henrique R, Jerónimo C and Paramio JM: DNA methylation as a therapeutic target for bladder cancer. Cells. 9:18502020. View Article : Google Scholar : PubMed/NCBI

78 

Wang R and Liu X: Epigenetic regulation of prostate cancer. Genes Dis. 7:606–613. 2020. View Article : Google Scholar : PubMed/NCBI

79 

Yang Z, Chu B, Tu Y, Li L, Chen D, Huang S, Huang W, Fan W, Li Q, Zhang C, et al: Dual inhibitors of DNMT and HDAC remodels the immune microenvironment of colorectal cancer and enhances the efficacy of anti-PD-L1 therapy. Pharmacol Res. 206:1072712024. View Article : Google Scholar : PubMed/NCBI

80 

Wang JH, Zeng Z, Sun J, Chen Y and Gao X: A novel small-molecule antagonist enhances the sensitivity of osteosarcoma to cabozantinib in vitro and in vivo by targeting DNMT-1 correlated with disease severity in human patients. Pharmacol Res. 173:1058692021. View Article : Google Scholar : PubMed/NCBI

81 

Li KK, Li F, Li QS, Yang K and Jin B: DNA methylation as a target of epigenetic therapeutics in cancer. Anticancer Agents Med Chem. 13:242–247. 2013. View Article : Google Scholar

82 

Nagaraju GP, Wu C, Merchant N, Chen Z, Lesinski GB and El-Rayes BF: Epigenetic effects of inhibition of heat shock protein 90 (HSP90) in human pancreatic and colon cancer. Cancer Lett. 402:110–116. 2017. View Article : Google Scholar : PubMed/NCBI

83 

Manara MC, Valente S, Cristalli C, Nicoletti G, Landuzzi L, Zwergel C, Mazzone R, Stazi G, Arimondo PB, Pasello M, et al: A quinoline-based DNA methyltransferase inhibitor as a possible adjuvant in osteosarcoma therapy. Mol Cancer Ther. 17:1881–1892. 2018. View Article : Google Scholar : PubMed/NCBI

84 

Jin T, Hao J and Fan D: Nicotine induces aberrant hypermethylation of tumor suppressor genes in pancreatic epithelial ductal cells. Biochem Biophys Res Commun. 499:934–940. 2018. View Article : Google Scholar : PubMed/NCBI

85 

Bazzichetto C, Conciatori F, Pallocca M, Falcone I, Fanciulli M, Cognetti F, Milella M and Ciuffreda L: PTEN as a prognostic/predictive biomarker in cancer: An unfulfilled promise? Cancers (Basel). 11:4352019. View Article : Google Scholar : PubMed/NCBI

86 

Carbajo-Garcia MC, Corachán A, Segura-Benitez M, Monleón J, Escrig J, Faus A, Pellicer A, Cervelló I and Ferrero H: 5-aza-2′-deoxycitidine inhibits cell proliferation, extracellular matrix formation and Wnt/β-catenin pathway in human uterine leiomyomas. Reprod Biol Endocrinol. 19:1062021. View Article : Google Scholar

87 

Bechtel W, McGoohan S, Zeisberg EM, Müller GA, Kalbacher H, Salant DJ, Müller CA, Kalluri R and Zeisberg M: Methylation determines fibroblast activation and fibrogenesis in the kidney. Nat Med. 16:544–550. 2010. View Article : Google Scholar : PubMed/NCBI

88 

Kato M and Natarajan R: Epigenetics and epigenomics in diabetic kidney disease and metabolic memory. Nat Rev Nephrol. 15:327–345. 2019. View Article : Google Scholar : PubMed/NCBI

89 

Bowden SA, Rodger EJ, Chatterjee A, Eccles MR and Stayner C: Recent discoveries in epigenetic modifications of polycystic kidney disease. Int J Mol Sci. 22:133272021. View Article : Google Scholar : PubMed/NCBI

90 

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. 26:2388–2397. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Sidler M, Aitken KJ, Jiang JX, Yadav P, Lloyd E, Ibrahim M, Choufani S, Weksberg R and Bägli D: Inhibition of DNA methylation during chronic obstructive bladder disease (COBD) improves function, pathology and expression. Sci Rep. 11:173072021. View Article : Google Scholar : PubMed/NCBI

92 

Li Y, Luo Q, Li Z, Wang Y, Zhu C, Li T and Li X: Long non-coding RNA IRAIN inhibits VEGFA expression via enhancing its DNA methylation leading to tumor suppression in renal carcinoma. Front Oncol. 10:10822020. View Article : Google Scholar : PubMed/NCBI

93 

Huang YQ, Guan H, Liu CH, Liu DC, Xu B, Jiang L, Lin ZX and Chen M: Association between RASSF1A promoter methylation and renal cell cancer susceptibility: A meta-analysis. Genet Mol Res. 15:gmr.150269942016.

94 

Bayarsaihan D: Epigenetic mechanisms in inflammation. J Dent Res. 90:9–17. 2011. View Article : Google Scholar :

95 

Bradley MS, Burke EE, Grenier C, Amundsen CL, Murphy SK and Siddiqui NY: A genome-scale DNA methylation study in women with interstitial cystitis/bladder pain syndrome. Neurourol Urodyn. 37:1485–1493. 2018. View Article : Google Scholar : PubMed/NCBI

96 

Tolg C and Bägli DJ: Uropathogenic Escherichia coli infection: Potential importance of epigenetics. Epigenomics. 4:229–235. 2012. View Article : Google Scholar : PubMed/NCBI

97 

Emam M, Cánovas A, Islas-Trejo AD, Fonseca PAS, Medrano JF and Mallard B: Transcriptomic profiles of monocyte-derived macrophages in response to Escherichia coli is associated with the host genetics. Sci Rep. 10:2712020. View Article : Google Scholar : PubMed/NCBI

98 

Tolg C, Sabha N, Cortese R, Panchal T, Ahsan A, Soliman A, Aitken KJ, Petronis A and Bägli DJ: Uropathogenic E. coli infection provokes epigenetic downregulation of CDKN2A (p16INK4A) in uroepithelial cells. Lab Invest. 91:825–836. 2011. View Article : Google Scholar : PubMed/NCBI

99 

Tatemichi M, Hata H, Tazawa H and Nakadate T: Lipopolysaccharide induces aberrant hypermethylation of Hic-1 in mouse embryonic fibroblasts lacking p53 gene. Anticancer Res. 28:2101–2108. 2008.PubMed/NCBI

100 

Yang RB, Mark MR, Gray A, Huang A, Xie MH, Zhang M, Goddard A, Wood WI, Gurney AL and Godowski PJ: Toll-like receptor-2 mediates lipopolysaccharide-induced cellular signalling. Nature. 395:284–288. 1998. View Article : Google Scholar : PubMed/NCBI

101 

Valinluck V and Sowers LC: Inflammation-mediated cytosine damage: A mechanistic link between inflammation and the epigenetic alterations in human cancers. Cancer Res. 67:5583–5586. 2007. View Article : Google Scholar : PubMed/NCBI

102 

Ding H, Chen J, Su M, Lin Z, Zhan H, Yang F, Li W, Xie J, Huang Y, Liu X, et al: BDNF promotes activation of astrocytes and microglia contributing to neuroinflammation and mechanical allodynia in cyclophosphamide-induced cystitis. J Neuroinflammation. 17:192020. View Article : Google Scholar : PubMed/NCBI

103 

Haldar S, Dru C, Mishra R, Tripathi M, Duong F, Angara B, Fernandez A, Arditi M and Bhowmick NA: Histone deacetylase inhibitors mediate DNA damage repair in ameliorating hemorrhagic cystitis. Sci Rep. 6:392572016. View Article : Google Scholar : PubMed/NCBI

104 

Choi IS, Yu K, Kim J, De Guzman E, Weisenberger DJ, Oghamian S, Kim HJ, Lee KH, Carroll C, Trinh BN, et al: Alterations in deoxyribonucleic acid (DNA) methylation patterns of Calca, Timp3, Mmp2, and Igf2r are associated with chronic cystitis in a cyclophosphamide-induced mouse model. Urology. 82:253.e9–e15. 2013. View Article : Google Scholar : PubMed/NCBI

105 

Mennuni S, Rubattu S, Pierelli G, Tocci G, Fofi C and Volpe M: Hypertension and kidneys: Unraveling complex molecular mechanisms underlying hypertensive renal damage. J Hum Hypertens. 28:74–79. 2014. View Article : Google Scholar

106 

Tomaszewski M and Itoh H: ISH2022KYOTO hypertension zero declaration. Cardiovasc Res. 119:e1362023. View Article : Google Scholar

107 

NCD Risk Factor Collaboration (NCD-RisC): Worldwide trends in hypertension prevalence and progress in treatment and control from 1990 to 2019: A pooled analysis of 1201 population-representative studies with 104 million participants. Lancet. 398:957–980. 2021. View Article : Google Scholar : PubMed/NCBI

108 

Pinheiro LC and Oliveira-Paula GH: Sources and effects of oxidative stress in hypertension. Curr Hypertens Rev. 16:166–180. 2020. View Article : Google Scholar

109 

Rodriguez-Iturbe B, Sepassi L, Quiroz Y, Ni Z, Wallace DC and Vaziri ND: Association of mitochondrial SOD deficiency with salt-sensitive hypertension and accelerated renal senescence. J Appl Physiol (1985). 102:255–260. 2007. View Article : Google Scholar

110 

Godin N, Liu F, Lau GJ, Brezniceanu ML, Chénier I, Filep JG, Ingelfinger JR, Zhang SL and Chan JS: Catalase overexpression prevents hypertension and tubular apoptosis in angiotensinogen transgenic mice. Kidney Int. 77:1086–1097. 2010. View Article : Google Scholar : PubMed/NCBI

111 

Chen Y, Li X, Wang S, Miao R and Zhong J: Targeting iron metabolism and ferroptosis as novel therapeutic approaches in cardiovascular diseases. Nutrients. 15:5912023. View Article : Google Scholar : PubMed/NCBI

112 

Vaziri ND, Lin CY, Farmand F and Sindhu RK: Superoxide dismutase, catalase, glutathione peroxidase and NADPH oxidase in lead-induced hypertension. Kidney Int. 63:186–194. 2003. View Article : Google Scholar

113 

Pushpakumar S, Ren L, Juin SK, Majumder S, Kulkarni R and Sen U: Methylation-dependent antioxidant-redox imbalance regulates hypertensive kidney injury in aging. Redox Biol. 37:1017542020. View Article : Google Scholar : PubMed/NCBI

114 

Cencioni C, Spallotta F, Martelli F, Valente S, Mai A, Zeiher AM and Gaetano C: Oxidative stress and epigenetic regulation in ageing and age-related diseases. Int J Mol Sci. 14:17643–17663. 2013. View Article : Google Scholar : PubMed/NCBI

115 

Miyoshi M, Sato M, Saito K, Otani L, Shirahige K, Miura F, Ito T, Jia H and Kato H: Maternal protein restriction alters the renal ptger1 DNA methylation state in SHRSP offspring. Nutrients. 10:14362018. View Article : Google Scholar : PubMed/NCBI

116 

Jia H, Miyoshi M, Li X, Furukawa K, Otani L, Shirahige K, Miura F, Ito T and Kato H: The epigenetic legacy of maternal protein restriction: Renal ptger1 DNA methylation changes in hypertensive rat offspring. Nutrients. 15:39572023. View Article : Google Scholar : PubMed/NCBI

117 

Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N, Colagiuri S, Guariguata L, Motala AA, Ogurtsova K, et al: Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International diabetes federation diabetes atlas, 9th edition. Diabetes Res Clin Pract. 157:1078432019. View Article : Google Scholar

118 

Lin E and Erickson KF: Payer mix among patients receiving dialysis. JAMA. 324:900–901. 2020. View Article : Google Scholar : PubMed/NCBI

119 

Thomas MC, Brownlee M, Susztak K, Sharma K, Jandeleit-Dahm KA, Zoungas S, Rossing P, Groop PH and Cooper ME: Diabetic kidney disease. Nat Rev Dis Primers. 1:150182015. View Article : Google Scholar : PubMed/NCBI

120 

Sun G, Reddy MA, Yuan H, Lanting L, Kato M and Natarajan R: Epigenetic histone methylation modulates fibrotic gene expression. J Am Soc Nephrol. 21:2069–2080. 2010. View Article : Google Scholar : PubMed/NCBI

121 

Schlosser P, Tin A, Matias-Garcia PR, Thio CHL, Joehanes R, Liu H, Weihs A, Yu Z, Hoppmann A, Grundner-Culemann F, et al: Meta-analyses identify DNA methylation associated with kidney function and damage. Nat Commun. 12:71742021. View Article : Google Scholar : PubMed/NCBI

122 

Smyth LJ, Dahlström EH, Syreeni A, Kerr K, Kilner J, Doyle R, Brennan E, Nair V, Fermin D, Nelson RG, et al: Epigenome-wide meta-analysis identifies DNA methylation biomarkers associated with diabetic kidney disease. Nat Commun. 13:78912022. View Article : Google Scholar : PubMed/NCBI

123 

He C, Wang D, Wang R, Huang Y, Huang X, Shen S, Lv J and Wu M: Epigallocatechin gallate induces the demethylation of actinin alpha 4 to inhibit diabetic nephropathy renal fibrosis via the NF-KB signaling pathway in vitro. Dose Response. 20:155932582211057042022. View Article : Google Scholar : PubMed/NCBI

124 

Zhang L, Zhang Q, Liu S, Chen Y, Li R, Lin T, Yu C, Zhang H, Huang Z, Zhao X, et al: DNA methyltransferase 1 may be a therapy target for attenuating diabetic nephropathy and podocyte injury. Kidney Int. 92:140–153. 2017. View Article : Google Scholar : PubMed/NCBI

125 

Peng R, Liu H, Peng H, Zhou J, Zha H, Chen X, Zhang L, Sun Y, Yin P, Wen L, et al: Promoter hypermethylation of let-7a-3 is relevant to its down-expression in diabetic nephropathy by targeting UHRF1. Gene. 570:57–63. 2015. View Article : Google Scholar : PubMed/NCBI

126 

Zhou J, Peng R, Li T, Luo X, Peng H, Zha H, Yin P, Wen L and Zhang Z: A potentially functional polymorphism in the regulatory region of let-7a-2 is associated with an increased risk for diabetic nephropathy. Gene. 527:456–461. 2013. View Article : Google Scholar : PubMed/NCBI

127 

Lane BM, Murray S, Benson K, Bierzynska A, Chryst-Stangl M, Wang L, Wu G, Cavalleri G, Doyle B, Fennelly N, et al: A rare autosomal dominant variant in regulator of calcineurin type 1 (RCAN1) gene confers enhanced calcineurin activity and may cause FSGS. J Am Soc Nephrol. 32:1682–1695. 2021. View Article : Google Scholar : PubMed/NCBI

128 

Li H, Zhang W, Zhong F, Das GC, Xie Y, Li Z, Cai W, Jiang G, Choi J, Sidani M, et al: Epigenetic regulation of RCAN1 expression in kidney disease and its role in podocyte injury. Kidney Int. 94:1160–1176. 2018. View Article : Google Scholar : PubMed/NCBI

129 

Hasan NS, Gamal El Dine H, Kamel SA, Hamed M, Youssef RN, Mahmoud Hassan E, Abdelrahman AH, Musa NI, Ali A and Awadallah E: Association of genetic and epigenetic changes of insulin like growth factor binding protein-1 in Egyptian patients with type 2 diabetes mellitus. Diabetes Res Clin Pract. 200:1106772023. View Article : Google Scholar : PubMed/NCBI

130 

Yang XH, Cao RF, Yu Y, Sui M, Zhang T, Xu JY and Wang XM: A study on the correlation between MTHFR promoter methylation and diabetic nephropathy. Am J Transl Res. 8:4960–4967. 2016.PubMed/NCBI

131 

Yang XH, Zhang BL, Zhang XM, Tong JD, Gu YH, Guo LL and Jin HM: EGCG attenuates renal damage via reversing Klotho hypermethylation in diabetic db/db mice and HK-2 cells. Oxid Med Cell Longev. 2020:60927152020. View Article : Google Scholar : PubMed/NCBI

132 

Tomson CRV, Cheung AK, Mann JFE, Chang TI, Cushman WC, Furth SL, Hou FF, Knoll GA, Muntner P, Pecoits-Filho R, et al: Management of blood pressure in patients with chronic kidney disease not receiving dialysis: Synopsis of the 2021 KDIGO clinical practice guideline. Ann Intern Med. 174:1270–1281. 2021. View Article : Google Scholar : PubMed/NCBI

133 

Ying M, Shao X, Qin H, Yin P, Lin Y, Wu J, Ren J and Zheng Y: Disease burden and epidemiological trends of chronic kidney disease at the global, regional, national levels from 1990 to 2019. Nephron. 148:113–123. 2024. View Article : Google Scholar

134 

Ko YA, Mohtat D, Suzuki M, Park AS, Izquierdo MC, Han SY, Kang HM, Si H, Hostetter T, Pullman JM, et al: Cytosine methylation changes in enhancer regions of core pro-fibrotic genes characterize kidney fibrosis development. Genome Biol. 14:R1082013. View Article : Google Scholar : PubMed/NCBI

135 

Morgado-Pascual JL, Marchant V, Rodrigues-Diez R, Dolade N, Suarez-Alvarez B, Kerr B, Valdivielso JM, Ruiz-Ortega M and Rayego-Mateos S: Epigenetic modification mechanisms involved in inflammation and fibrosis in renal pathology. Mediators Inflamm. 2018:29310492018. View Article : Google Scholar

136 

Panizo S, Martinez-Arias L, Alonso-Montes C, Cannata P, Martín-Carro B, Fernández-Martín JL, Naves-Díaz M, Carrillo-López N and Cannata-Andía JB: Fibrosis in chronic kidney disease: Pathogenesis and consequences. Int J Mol Sci. 22:4082021. View Article : Google Scholar : PubMed/NCBI

137 

Yin S, Zhang Q, Yang J, Lin W, Li Y, Chen F and Cao W: TGFβ-incurred epigenetic aberrations of miRNA and DNA methyltransferase suppress Klotho and potentiate renal fibrosis. Biochim Biophys Acta Mol Cell Res. 1864:1207–1216. 2017. View Article : Google Scholar : PubMed/NCBI

138 

Xiao X, Wang W, Guo C, Wu J, Zhang S, Shi H, Kwon S, Chen J and Dong Z: Hypermethylation leads to the loss of HOXA5, resulting in JAG1 expression and NOTCH signaling contributing to kidney fibrosis. Kidney Int. 106:98–114. 2024. View Article : Google Scholar : PubMed/NCBI

139 

Li Y, Chen F, Wei A, Bi F, Zhu X, Yin S, Lin W and Cao W: Klotho recovery by genistein via promoter histone acetylation and DNA demethylation mitigates renal fibrosis in mice. J Mol Med (Berl). 97:541–552. 2019. View Article : Google Scholar : PubMed/NCBI

140 

Takiguchi M, Achanzar WE, Qu W, Li G and Waalkes MP: Effects of cadmium on DNA-(Cytosine-5) methyltransferase activity and DNA methylation status during cadmium-induced cellular transformation. Exp Cell Res. 286:355–365. 2003. View Article : Google Scholar : PubMed/NCBI

141 

Zhang C, Liang Y, Lei L, Zhu G, Chen X, Jin T and Wu Q: Hypermethylations of RASAL1 and KLOTHO is associated with renal dysfunction in a Chinese population environmentally exposed to cadmium. Toxicol Appl Pharmacol. 271:78–85. 2013. View Article : Google Scholar : PubMed/NCBI

142 

Azuma M, Koyama D, Kikuchi J, Yoshizawa H, Thasinas D, Shiizaki K, Kuro-o M, Furukawa Y and Kusano E: Promoter methylation confers kidney-specific expression of the Klotho gene. FASEB J. 26:4264–4274. 2012. View Article : Google Scholar : PubMed/NCBI

143 

Chen Z, Li G and Liu J: Autonomic dysfunction in Parkinson's disease: Implications for pathophysiology, diagnosis, and treatment. Neurobiol Dis. 134:1047002020. View Article : Google Scholar

144 

Wang W, Zhao X, Shao Y, Duan X, Wang Y, Li J, Li J, Li D, Li X and Wong J: Mutation-induced DNMT1 cleavage drives neurodegenerative disease. Sci Adv. 7:eabe85112021. View Article : Google Scholar : PubMed/NCBI

145 

Ye S, Zhong J, Huang J, Chen L, Yi L, Li X, Lv J, Miao J, Li H, Chen D and Li C: Protective effect of plastrum testudinis extract on dopaminergic neurons in a Parkinson's disease model through DNMT1 nuclear translocation and SNCA's methylation. Biomed Pharmacother. 141:1118322021. View Article : Google Scholar : PubMed/NCBI

146 

Patel HD, Gupta M, Joice GA, Srivastava A, Alam R, Allaf ME and Pierorazio PM: Clinical stage migration and survival for renal cell carcinoma in the United States. Eur Urol Oncol. 2:343–348. 2019. View Article : Google Scholar : PubMed/NCBI

147 

Turajlic S, Swanton C and Boshoff C: Kidney cancer: The next decade. J Exp Med. 215:2477–2479. 2018. View Article : Google Scholar : PubMed/NCBI

148 

Lin RK and Wang YC: Dysregulated transcriptional and post-translational control of DNA methyltransferases in cancer. Cell Biosci. 4:462014. View Article : Google Scholar : PubMed/NCBI

149 

Arai E and Kanai Y: Genetic and epigenetic alterations during renal carcinogenesis. Int J Clin Exp Pathol. 4:58–73. 2010.

150 

Bahadoram S, Davoodi M, Hassanzadeh S, Bahadoram M, Barahman M and Mafakher L: Renal cell carcinoma: An overview of the epidemiology, diagnosis, and treatment. G Ital Nefrol. 39:2022–vol3. 2022.PubMed/NCBI

151 

Christoph F, Kempkensteffen C, Weikert S, Köllermann J, Krause H, Miller K, Schostak M and Schrader M: Methylation of tumour suppressor genes APAF-1 and DAPK-1 and in vitro effects of demethylating agents in bladder and kidney cancer. Br J Cancer. 95:1701–1707. 2006. View Article : Google Scholar : PubMed/NCBI

152 

Christoph F, Weikert S, Kempkensteffen C, Krause H, Schostak M, Köllermann J, Miller K and Schrader M: Promoter hypermethylation profile of kidney cancer with new proapoptotic p53 target genes and clinical implications. Clin Cancer Res. 12:5040–5046. 2006. View Article : Google Scholar : PubMed/NCBI

153 

Liu Q, Jin J, Ying J, Cui Y, Sun M, Zhang L, Fan Y, Xu B and Zhang Q: Epigenetic inactivation of the candidate tumor suppressor gene ASC/TMS1 in human renal cell carcinoma and its role as a potential therapeutic target. Oncotarget. 6:22706–22723. 2015. View Article : Google Scholar : PubMed/NCBI

154 

Joosten SC, Deckers IA, Aarts MJ, Hoeben A, van Roermund JG, Smits KM, Melotte V, van Engeland M and Tjan-Heijnen VC: Prognostic DNA methylation markers for renal cell carcinoma: A systematic review. Epigenomics. 9:1243–1257. 2017. View Article : Google Scholar : PubMed/NCBI

155 

Mo S, Su Z, Heng B, Chen W, Shi L, Du X and Lai C: SFRP1 promoter methylation and renal carcinoma risk: A systematic review and meta-analysis. J Nippon Med Sch. 85:78–86. 2018. View Article : Google Scholar : PubMed/NCBI

156 

Morris MR, Ricketts CJ, Gentle D, McRonald F, Carli N, Khalili H, Brown M, Kishida T, Yao M, Banks RE, et al: Genome-wide methylation analysis identifies epigenetically inactivated candidate tumour suppressor genes in renal cell carcinoma. Oncogene. 30:1390–1401. 2011. View Article : Google Scholar

157 

Peters I, Eggers H, Atschekzei F, Hennenlotter J, Waalkes S, Tränkenschuh W, Grosshennig A, Merseburger AS, Stenzl A, Kuczyk MA and Serth J: GATA5 CpG island methylation in renal cell cancer: A potential biomarker for metastasis and disease progression. BJU Int. 110:E144–E152. 2012. View Article : Google Scholar : PubMed/NCBI

158 

Peters I, Gebauer K, Dubrowinskaja N, Atschekzei F, Kramer MW, Hennenlotter J, Tezval H, Abbas M, Scherer R, Merseburger AS, et al: GATA5 CpG island hypermethylation is an independent predictor for poor clinical outcome in renal cell carcinoma. Oncol Rep. 31:1523–1530. 2014. View Article : Google Scholar : PubMed/NCBI

159 

Rohilla S, Singh M, Priya S, Almalki WH, Haniffa SM, Subramaniyan V, Fuloria S, Fuloria NK, Sekar M, Singh SK, et al: Exploring the mechanical perspective of a new anti-tumor agent: Melatonin. J Environ Pathol Toxicol Oncol. 42:1–16. 2023. View Article : Google Scholar : PubMed/NCBI

160 

Majid S, Dar AA, Ahmad AE, Hirata H, Kawakami K, Shahryari V, Saini S, Tanaka Y, Dahiya AV, Khatri G and Dahiya R: BTG3 tumor suppressor gene promoter demethylation, histone modification and cell cycle arrest by genistein in renal cancer. Carcinogenesis. 30:662–670. 2009. View Article : Google Scholar : PubMed/NCBI

161 

Wang L, Lu B, He M, Wang Y, Wang Z and Du L: Prostate cancer incidence and mortality: Global status and temporal trends in 89 countries from 2000 to 2019. Front Public Health. 10:8110442022. View Article : Google Scholar : PubMed/NCBI

162 

Chang AJ, Autio KA, Roach M III and Scher HI: High-risk prostate cancer-classification and therapy. Nat Rev Clin Oncol. 11:308–323. 2014. View Article : Google Scholar : PubMed/NCBI

163 

Jemal A, Culp MB, Ma J, Islami F and Fedewa SA: Prostate cancer incidence 5 years after US preventive services task force recommendations against screening. J Natl Cancer Inst. 113:64–71. 2021. View Article : Google Scholar :

164 

Zargar H, van den Bergh R, Moon D, Lawrentschuk N, Costello A and Murphy D: The impact of the United States preventive services task force (USPTSTF) recommendations against prostate-specific antigen (PSA) testing on PSA testing in Australia. BJU Int. 119:110–115. 2017. View Article : Google Scholar

165 

Curry SJ, Krist AH and Owens DK: Annual report to the nation on the status of cancer, part II: Recent changes in prostate cancer trends and disease characteristics. Cancer. 125:317–318. 2019. View Article : Google Scholar

166 

Hatakeyama S, Yoneyama T, Tobisawa Y and Ohyama C: Recent progress and perspectives on prostate cancer biomarkers. Int J Clin Oncol. 22:214–221. 2017. View Article : Google Scholar :

167 

Jerónimo C, Bastian PJ, Bjartell A, Carbone GM, Catto JW, Clark SJ, Henrique R, Nelson WG and Shariat SF: Epigenetics in prostate cancer: Biologic and clinical relevance. Eur Urol. 60:753–766. 2011. View Article : Google Scholar : PubMed/NCBI

168 

Zhao SG, Chen WS, Li H, Foye A, Zhang M, Sjöström M, Aggarwal R, Playdle D, Liao A, Alumkal JJ, et al: The DNA methylation landscape of advanced prostate cancer. Nat Genet. 52:778–789. 2020. View Article : Google Scholar : PubMed/NCBI

169 

Zhao Y, Hu X, Yu H, Liu X, Sun H and Shao C: Alternations of gene expression in PI3K and AR pathways and DNA methylation features contribute to metastasis of prostate cancer. Cell Mol Life Sci. 79:4362022. View Article : Google Scholar : PubMed/NCBI

170 

Ummanni R, Jost E, Braig M, Lohmann F, Mundt F, Barett C, Schlomm T, Sauter G, Senff T, Bokemeyer C, et al: Ubiquitin carboxyl-terminal hydrolase 1 (UCHL1) is a potential tumour suppressor in prostate cancer and is frequently silenced by promoter methylation. Mol Cancer. 10:1292011. View Article : Google Scholar : PubMed/NCBI

171 

Chen Y, Li J, Yu X, Li S, Zhang X, Mo Z and Hu Y: APC gene hypermethylation and prostate cancer: A systematic review and meta-analysis. Eur J Hum Genet. 21:929–935. 2013. View Article : Google Scholar : PubMed/NCBI

172 

Zhang SQ, Zhang GQ and Zhang L: Correlation between methylation of the E-cadherin gene and malignancy of prostate cancer. Genet Mol Res. 15:gmr.150280462016.

173 

Miyauchi T, Takahashi M, Mitsuzuka K, Saiki Y, Okubo T, Vertino PM, Goto A, Arai Y, Horii A and Fukushige S: Aberrant hypermethylation-mediated suppression of PYCARD is extremely frequent in prostate cancer with gleason score ≥7. Dis Markers. 2021:88589052021. View Article : Google Scholar

174 

Singal R, Ferdinand L, Reis IM and Schlesselman JJ: Methylation of multiple genes in prostate cancer and the relationship with clinicopathological features of disease. Oncol Rep. 12:631–637. 2004.PubMed/NCBI

175 

Zhang W, Zeng S, Gong L, Zhang D and Hu X: Gene methylation status in focus of advanced prostate cancer diagnostics and improved individual outcomes. Transl Androl Urol. 12:1813–1826. 2023. View Article : Google Scholar

176 

van der Pol Y, Moldovan N, Ramaker J, Bootsma S, Lenos KJ, Vermeulen L, Sandhu S, Bahce I, Pegtel DM, Wong SQ, et al: The landscape of cell-free mitochondrial DNA in liquid biopsy for cancer detection. Genome Biol. 24:2292023. View Article : Google Scholar : PubMed/NCBI

177 

Woodson K, O'Reilly KJ, Ward DE, Walter J, Hanson J, Walk EL and Tangrea JA: CD44 and PTGS2 methylation are independent prognostic markers for biochemical recurrence among prostate cancer patients with clinically localized disease. Epigenetics. 1:183–186. 2006. View Article : Google Scholar

178 

Boiron M: Genetics and cellular cellular biology of cancer. Pathol Biol (Paris). 38:765–767. 1990.In French. PubMed/NCBI

179 

Zorn CS, Wojno KJ, McCabe MT, Kuefer R, Gschwend JE and Day ML: 5-aza-2′-deoxycytidine delays androgen-independent disease and improves survival in the transgenic adenocarcinoma of the mouse prostate mouse model of prostate cancer. Clin Cancer Res. 13:2136–2143. 2007. View Article : Google Scholar : PubMed/NCBI

180 

Shang D, Liu Y, Liu Q, Zhang F, Feng L, Lv W and Tian Y: Synergy of 5-aza-2′-deoxycytidine (DAC) and paclitaxel in both androgen-dependent and -independent prostate cancer cell lines. Cancer Lett. 278:82–87. 2009. View Article : Google Scholar : PubMed/NCBI

181 

Thibault A, Figg WD, Bergan RC, Lush RM, Myers CE, Tompkins A, Reed E and Samid D: A phase II study of 5-aza-2′deoxycytidine (decitabine) in hormone independent metastatic (D2) prostate cancer. Tumori. 84:87–89. 1998. View Article : Google Scholar : PubMed/NCBI

182 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

183 

Teoh JYC, Huang J, Ko WYK, Lok V, Choi P, Ng CF, Sengupta S, Mostafid H, Kamat AM, Black PC, et al: Global trends of bladder cancer incidence and mortality, and their associations with tobacco use and gross domestic product per capita. Eur Urol. 78:893–906. 2020. View Article : Google Scholar : PubMed/NCBI

184 

Sylvester RJ, van der Meijden AP, Oosterlinck W, Witjes JA, Bouffioux C, Denis L, Newling DW and Kurth K: Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: A combined analysis of 2596 patients from seven EORTC trials. Eur Urol. 49:466–477. 2006. View Article : Google Scholar : PubMed/NCBI

185 

Dobruch J and Oszczudlowski M: Bladder cancer: Current challenges and future directions. Medicina (Kaunas). 57:7492021. View Article : Google Scholar : PubMed/NCBI

186 

Scher MB, Elbaum MB, Mogilevkin Y, Hilbert DW, Mydlo JH, Sidi AA, Adelson ME, Mordechai E and Trama JP: Detecting DNA methylation of the BCL2, CDKN2A and NID2 genes in urine using a nested methylation specific polymerase chain reaction assay to predict bladder cancer. J Urol. 188:2101–2107. 2012. View Article : Google Scholar : PubMed/NCBI

187 

Bai ZJ, Liu Q, Wang XS and Liu WY: APC promoter methylation is correlated with development and progression of bladder cancer, but not linked to overall survival: A meta-analysis. Neoplasma. 66:470–480. 2019. View Article : Google Scholar : PubMed/NCBI

188 

Kawamoto K, Enokida H, Gotanda T, Kubo H, Nishiyama K, Kawahara M and Nakagawa M: p16INK4a and p14ARF methylation as a potential biomarker for human bladder cancer. Biochem Biophys Res Commun. 339:790–796. 2006. View Article : Google Scholar

189 

Jahed M, Ebadi N, Mivehchi M, Majidizadeh T, Shahshanipour M, Asgari M, Ghadakzadeh S and Hosseini SA: MGMT hypermethylation and BCL-2 overexpression associated with superficial bladder cancer and recurrence. Cancer Biomark. 16:627–632. 2016. View Article : Google Scholar : PubMed/NCBI

190 

Bayramov B, Gunes S, Buyukalpelli R, Aydin O and Henkel R: Promoter methylation analysis of CDH1 and p14ARF genes in patients with urothelial bladder cancer. Onco Targets Ther. 11:4189–4196. 2018. View Article : Google Scholar : PubMed/NCBI

191 

Chen F, Huang T, Ren Y, Wei J, Lou Z, Wang X, Fan X, Chen Y, Weng G and Yao X: Clinical significance of CDH13 promoter methylation as a biomarker for bladder cancer: A meta-analysis. BMC Urol. 16:522016. View Article : Google Scholar : PubMed/NCBI

192 

Dai L, Ma C, Zhang Z, Zeng S, Liu A, Tang S, Ren Q, Sun Y and Xu C: DAPK promoter methylation and bladder cancer risk: A systematic review and meta-analysis. PLoS One. 11:e01672282016. View Article : Google Scholar : PubMed/NCBI

193 

Wang D, Qiu Z and Wu C: Diagnostic value of the combination of DAPK methylation in urinary sediment and B ultrasound for recurrent urinary bladder cancer. World J Surg Oncol. 21:2672023. View Article : Google Scholar : PubMed/NCBI

194 

Han SY, Iliopoulos D, Druck T, Guler G, Grubbs CJ, Pereira M, Zhang Z, You M, Lubet RA, Fong LY and Huebner K: CpG methylation in the Fhit regulatory region: Relation to Fhit expression in murine tumors. Oncogene. 23:3990–3998. 2004. View Article : Google Scholar : PubMed/NCBI

195 

Kandimalla R, van Tilborg AA and Zwarthoff EC: DNA methylation-based biomarkers in bladder cancer. Nat Rev Urol. 10:327–335. 2013. View Article : Google Scholar : PubMed/NCBI

196 

Costa VL, Henrique R, Danielsen SA, Duarte-Pereira S, Eknaes M, Skotheim RI, Rodrigues A, Magalhães JS, Oliveira J, Lothe RA, et al: Three epigenetic biomarkers, GDF15, TMEFF2, and VIM, accurately predict bladder cancer from DNA-based analyses of urine samples. Clin Cancer Res. 16:5842–5851. 2010. View Article : Google Scholar : PubMed/NCBI

197 

Sun J, Chen Z, Zhu T, Yu J, Ma K, Zhang H, He Y, Luo X and Zhu J: Hypermethylated SFRP1, but none of other nine genes 'informative' for western countries, is valuable for bladder cancer detection in Mainland China. J Cancer Res Clin Oncol. 135:1717–1727. 2009. View Article : Google Scholar : PubMed/NCBI

198 

Gao Q, Chen F, Zhang L, Wei A, Wang Y, Wu Z and Cao W: Inhibition of DNA methyltransferase aberrations reinstates antioxidant aging suppressors and ameliorates renal aging. Aging Cell. 21:e135262022. View Article : Google Scholar :

199 

Li XT, Song JW, Zhang ZZ, Zhang MW, Liang LR, Miao R, Liu Y, Chen YH, Liu XY and Zhong JC: Sirtuin 7 mitigates renal ferroptosis, fibrosis and injury in hypertensive mice by facilitating the KLF15/Nrf2 signaling. Free Radic Biol Med. 193:459–473. 2022. View Article : Google Scholar : PubMed/NCBI

200 

Zhao Y, Fan X, Wang Q, Zhen J, Li X, Zhou P, Lang Y, Sheng Q, Zhang T, Huang T, et al: ROS promote hyper-methylation of NDRG2 promoters in a DNMTS-dependent manner: Contributes to the progression of renal fibrosis. Redox Biol. 62:1026742023. View Article : Google Scholar : PubMed/NCBI

201 

Ming S, Tian J, Ma K, Pei C, Li L, Wang Z, Fang Z, Liu M, Dong H, Li W, et al: Oxalate-induced apoptosis through ERS-ROS-NF-κB signalling pathway in renal tubular epithelial cell. Mol Med. 28:882022. View Article : Google Scholar

202 

Doi S, Zou Y, Togao O, Pastor JV, John GB, Wang L, Shiizaki K, Gotschall R, Schiavi S, Yorioka N, et al: Klotho inhibits transforming growth factor-beta1 (TGF-beta1) signaling and suppresses renal fibrosis and cancer metastasis in mice. J Biol Chem. 286:8655–8665. 2011. View Article : Google Scholar : PubMed/NCBI

203 

Brandt S, Bernhardt A, Häberer S, Wolters K, Gehringer F, Reichardt C, Krause A, Geffers R, Kahlfuß S, Jeron A, et al: Comparative analysis of acute kidney injury models and related fibrogenic responses: Convergence on methylation patterns regulated by cold shock protein. Cells. 13:3672024. View Article : Google Scholar : PubMed/NCBI

204 

Miao J, Liu J, Niu J, Zhang Y, Shen W, Luo C, Liu Y, Li C, Li H, Yang P, et al: Wnt/β-catenin/RAS signaling mediates age-related renal fibrosis and is associated with mitochondrial dysfunction. Aging Cell. 18:e130042019. View Article : Google Scholar

205 

Edeling M, Ragi G, Huang S, Pavenstadt H and Susztak K: Developmental signalling pathways in renal fibrosis: The roles of Notch, Wnt and Hedgehog. Nat Rev Nephrol. 12:426–439. 2016. View Article : Google Scholar : PubMed/NCBI

206 

Wu J, Wei Y, Li T, Lin L, Yang Z and Ye L: DNA methylation-mediated lowly expressed AOX1 promotes cell migration and invasion of prostate cancer. Urol Int. 107:517–525. 2023. View Article : Google Scholar

207 

Ding M, Wang Q, Zhu W, Chang J, Liao H and Xiao G: DNA methylation-mediated low expression of ZNF582 promotes the proliferation, migration, and invasion of clear cell renal cell carcinoma. Clin Exp Nephrol. 27:24–31. 2023. View Article : Google Scholar

208 

Heichman KA and Warren JD: DNA methylation biomarkers and their utility for solid cancer diagnostics. Clin Chem Lab Med. 50:1707–1721. 2012. View Article : Google Scholar : PubMed/NCBI

209 

Du Y, Xu Y, Guo X, Tan C, Zhu X, Liu G, Lyu X and Bei C: Methylation-regulated tumor suppressor gene PDE7B promotes HCC invasion and metastasis through the PI3K/AKT signaling pathway. BMC Cancer. 24:6242024. View Article : Google Scholar : PubMed/NCBI

210 

Lopez-Serra P and Esteller M: DNA methylation-associated silencing of tumor-suppressor microRNAs in cancer. Oncogene. 31:1609–1622. 2012. View Article : Google Scholar :

211 

Zhu L, Tang N, Hang H, Zhou Y, Dong J, Yang Y, Mao L, Qiu Y, Fu X and Cao W: Loss of Claudin-1 incurred by DNMT aberration promotes pancreatic cancer progression. Cancer Lett. 586:2166112024. View Article : Google Scholar : PubMed/NCBI

212 

Lam TW, Tong JH, To KF, Chan A, Liew CT, Lai PB and Wong N: Correlative analysis of DNA methyltransferase expression and promoter hypermethylation of tumor suppressor genes in hepatocellular carcinoma. Cancer Genomics Proteomics. 3:271–277. 2006.PubMed/NCBI

213 

Nishiyama A and Nakanishi M: Navigating the DNA methylation landscape of cancer. Trends Genet. 37:1012–1027. 2021. View Article : Google Scholar : PubMed/NCBI

214 

O'Hagan HM, Wang W, Sen S, Destefano Shields C, Lee SS, Zhang YW, Clements EG, Cai Y, Van Neste L, Easwaran H, et al: Oxidative damage targets complexes containing DNA methyltransferases, SIRT1, and polycomb members to promoter CpG Islands. Cancer Cell. 20:606–619. 2011. View Article : Google Scholar : PubMed/NCBI

215 

Xiao X, Chen M, Sang Y, Xue J, Jiang K, Chen Y, Zhang L, Yu S, Lv W, Li Y, et al: Methylation-mediated silencing of ATF3 promotes thyroid cancer progression by regulating prognostic genes in the MAPK and PI3K/AKT pathways. Thyroid. 33:1441–1454. 2023. View Article : Google Scholar : PubMed/NCBI

216 

Zhu S, Xu H, Chen R, Shen Q, Yang D, Peng H, Tong J and Fu Q: DNA methylation and miR-92a-3p-mediated repression of HIP1R promotes pancreatic cancer progression by activating the PI3K/AKT pathway. J Cell Mol Med. 27:788–802. 2023. View Article : Google Scholar : PubMed/NCBI

217 

Jin Y, Cui D, Ren J, Wang K, Zeng T and Gao L: CACNA2D3 is downregulated in gliomas and functions as a tumor suppressor. Mol Carcinog. 56:945–959. 2017. View Article : Google Scholar

218 

Clevers H and Nusse R: Wnt/β-catenin signaling and disease. Cell. 149:1192–1205. 2012. View Article : Google Scholar : PubMed/NCBI

219 

van Es JH, Haegebarth A, Kujala P, Itzkovitz S, Koo BK, Boj SF, Korving J, van den Born M, van Oudenaarden A, Robine S and Clevers H: A critical role for the Wnt effector Tcf4 in adult intestinal homeostatic self-renewal. Mol Cell Biol. 32:1918–1927. 2012. View Article : Google Scholar : PubMed/NCBI

220 

Yao H, Ashihara E and Maekawa T: Targeting the Wnt/β-catenin signaling pathway in human cancers. Expert Opin Ther Targets. 15:873–887. 2011. View Article : Google Scholar : PubMed/NCBI

221 

Yang XZ, Cheng TT, He QJ, Lei ZY, Chi J, Tang Z, Liao QX, Zhang H, Zeng LS and Cui SZ: LINC01133 as ceRNA inhibits gastric cancer progression by sponging miR-106a-3p to regulate APC expression and the Wnt/β-catenin pathway. Mol Cancer. 17:1262018. View Article : Google Scholar

222 

Korinek V, Barker N, Morin PJ, van Wichen D, de Weger R, Kinzler KW, Vogelstein B and Clevers H: Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC-/- colon carcinoma. Science. 275:1784–1787. 1997. View Article : Google Scholar : PubMed/NCBI

223 

Nishisho I, Nakamura Y, Miyoshi Y, Miki Y, Ando H, Horii A, Koyama K, Utsunomiya J, Baba S and Hedge P: Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science. 253:665–669. 1991. View Article : Google Scholar : PubMed/NCBI

224 

Yuan TL and Cantley LC: PI3K pathway alterations in cancer: Variations on a theme. Oncogene. 27:5497–5510. 2008. View Article : Google Scholar : PubMed/NCBI

225 

Stemke-Hale K, Gonzalez-Angulo AM, Lluch A, Neve RM, Kuo WL, Davies M, Carey M, Hu Z, Guan Y, Sahin A, et al: An integrative genomic and proteomic analysis of PIK3CA, PTEN, and AKT mutations in breast cancer. Cancer Res. 68:6084–6091. 2008. View Article : Google Scholar : PubMed/NCBI

226 

Jouali F, Marchoudi N, Talbi S, Bilal B, El Khasmi M, Rhaissi H and Fekkak J: Detection of PIK3/AKT pathway in Moroccan population with triple negative breast cancer. BMC Cancer. 18:9002018. View Article : Google Scholar : PubMed/NCBI

227 

Hennessy BT, Smith DL, Ram PT, Lu Y and Mills GB: Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov. 4:988–1004. 2005. View Article : Google Scholar : PubMed/NCBI

228 

Park JI: MAPK-ERK pathway. Int J Mol Sci. 24:96662023. View Article : Google Scholar : PubMed/NCBI

229 

Ullah R, Yin Q, Snell AH and Wan L: RAF-MEK-ERK pathway in cancer evolution and treatment. Semin Cancer Biol. 85:123–154. 2022. View Article : Google Scholar

230 

Gurbuz V, Sozen S, Bilen CY and Konac E: miR-148a, miR-152 and miR-200b promote prostate cancer metastasis by targeting DNMT1 and PTEN expression. Oncol Lett. 22:8052021. View Article : Google Scholar : PubMed/NCBI

231 

Bhattacharyya S, Feferman L and Tobacman JK: Dihydrotestosterone inhibits arylsulfatase B and Dickkopf Wnt signaling pathway inhibitor (DKK)-3 leading to enhanced Wnt signaling in prostate epithelium in response to stromal Wnt3A. Prostate. 79:689–700. 2019. View Article : Google Scholar : PubMed/NCBI

232 

Kardooni H, Gonzalez-Gualda E, Stylianakis E, Saffaran S, Waxman J and Kypta RM: CRISPR-mediated reactivation of DKK3 expression attenuates TGF-β signaling in prostate cancer. Cancers (Basel). 10:1652018. View Article : Google Scholar

233 

Lodygin D, Epanchintsev A, Menssen A, Diebold J and Hermeking H: Functional epigenomics identifies genes frequently silenced in prostate cancer. Cancer Res. 65:4218–4227. 2005. View Article : Google Scholar : PubMed/NCBI

234 

Bhattacharyya S, Feferman L and Tobacman JK: Chondroitin sulfatases differentially regulate Wnt signaling in prostate stem cells through effects on SHP2, phospho-ERK1/2, and Dickkopf Wnt signaling pathway inhibitor (DKK3). Oncotarget. 8:100242–100260. 2017. View Article : Google Scholar : PubMed/NCBI

235 

Arai E, Kanai Y, Ushijima S, Fujimoto H, Mukai K and Hirohashi S: Regional DNA hypermethylation and DNA methyltransferase (DNMT) 1 protein overexpression in both renal tumors and corresponding nontumorous renal tissues. Int J Cancer. 119:288–296. 2006. View Article : Google Scholar : PubMed/NCBI

236 

Liao F, Liao Z, Zhang T, Jiang W, Zhu P, Zhao Z, Shi H, Zhao D, Zhou N and Huang X: ECFC-derived exosomal THBS1 mediates angiogenesis and osteogenesis in distraction osteogenesis via the PI3K/AKT/ERK pathway. J Orthop Translat. 37:12–22. 2022. View Article : Google Scholar : PubMed/NCBI

237 

Rubinek T and Wolf I: The role of alpha-Klotho as a universal tumor suppressor. Vitam Horm. 101:197–214. 2016. View Article : Google Scholar : PubMed/NCBI

238 

Wang Y and Sun Z: Current understanding of Klotho. Ageing Res Rev. 8:43–51. 2009. View Article : Google Scholar :

239 

Chen B, Ma X, Liu S, Zhao W and Wu J: Inhibition of lung cancer cells growth, motility and induction of apoptosis by Klotho, a novel secreted Wnt antagonist, in a dose-dependent manner. Cancer Biol Ther. 13:1221–1228. 2012. View Article : Google Scholar : PubMed/NCBI

240 

Camilli TC, Xu M, O'Connell MP, Chien B, Frank BP, Subaran S, Indig FE, Morin PJ, Hewitt SM and Weeraratna AT: Loss of Klotho during melanoma progression leads to increased filamin cleavage, increased Wnt5A expression, and enhanced melanoma cell motility. Pigment Cell Melanoma Res. 24:175–186. 2011. View Article : Google Scholar

241 

Li X, Lu P, Shao XF, Jiang T, Liu F and Li G: Klotho regulates epithelial-to-mesenchymal transition in vitro via Wnt/β-catenin pathway and attenuates chronic allograft dysfunction in a rat renal transplant model. Ann Transplant. 26:e9300662021. View Article : Google Scholar

242 

Prud'homme GJ: Cancer stem cells and novel targets for antitumor strategies. Curr Pharm Des. 18:2838–2849. 2012. View Article : Google Scholar : PubMed/NCBI

243 

Costa VL, Henrique R, Ribeiro FR, Carvalho JR, Oliveira J, Lobo F, Teixeira MR and Jerónimo C: Epigenetic regulation of Wnt signaling pathway in urological cancer. Epigenetics. 5:343–351. 2010. View Article : Google Scholar : PubMed/NCBI

244 

Henrique R, Ribeiro FR, Fonseca D, Hoque MO, Carvalho AL, Costa VL, Pinto M, Oliveira J, Teixeira MR, Sidransky D and Jerónimo C: High promoter methylation levels of APC predict poor prognosis in sextant biopsies from prostate cancer patients. Clin Cancer Res. 13:6122–6129. 2007. View Article : Google Scholar : PubMed/NCBI

245 

Rogler A, Kendziorra E, Giedl J, Stoehr C, Taubert H, Goebell PJ, Wullich B, Stöckle M, Lehmann J, Petsch S, et al: Functional analyses and prognostic significance of SFRP1 expression in bladder cancer. J Cancer Res Clin Oncol. 141:1779–1790. 2015. View Article : Google Scholar : PubMed/NCBI

246 

Robertson KD and Jones PA: DNA methylation: Past, present and future directions. Carcinogenesis. 21:461–467. 2000. View Article : Google Scholar : PubMed/NCBI

247 

Myöhänen SK, Baylin SB and Herman JG: Hypermethylation can selectively silence individual p16ink4A alleles in neoplasia. Cancer Res. 58:591–593. 1998.PubMed/NCBI

248 

Kucuk O: Cancer biomarkers. Mol Aspects Med. 45:1–2. 2015. View Article : Google Scholar : PubMed/NCBI

249 

O'Reilly E, Tuzova AV, Walsh AL, Russell NM, O'Brien O, Kelly S, Dhomhnallain ON, DeBarra L, Dale CM, Brugman R, et al: epiCaPture: A urine DNA methylation test for early detection of aggressive prostate cancer. JCO Precis Oncol. 2019:PO.18.001342019.PubMed/NCBI

250 

Mikeska T and Craig JM: DNA methylation biomarkers: Cancer and beyond. Genes (Basel). 5:821–864. 2014. View Article : Google Scholar : PubMed/NCBI

251 

Mahon KL, Qu W, Devaney J, Paul C, Castillo L, Wykes RJ, Chatfield MD, Boyer MJ, Stockler MR, Marx G, et al: Methylated glutathione S-transferase 1 (mGSTP1) is a potential plasma free DNA epigenetic marker of prognosis and response to chemotherapy in castrate-resistant prostate cancer. Br J Cancer. 111:1802–1809. 2014. View Article : Google Scholar : PubMed/NCBI

252 

Ibrahim J, Peeters M, Van Camp G and Opde Beeck K: Methylation biomarkers for early cancer detection and diagnosis: Current and future perspectives. Eur J Cancer. 178:91–113. 2023. View Article : Google Scholar

253 

Waterhouse RL Jr, Van Neste L, Moses KA, Barnswell C, Silberstein JL, Jalkut M, Tutrone R, Sylora J, Anglade R, Murdock M, et al: Evaluation of an epigenetic assay for predicting repeat prostate biopsy outcome in African American men. Urology. 128:62–65. 2019. View Article : Google Scholar

254 

Yamagishi M, Kuze Y, Kobayashi S, Nakashima M, Morishima S, Kawamata T, Makiyama J, Suzuki K, Seki M, Abe K, et al: Mechanisms of action and resistance in histone methylation-targeted therapy. Nature. 627:221–228. 2024. View Article : Google Scholar : PubMed/NCBI

255 

Sgro A, Cursons J, Waryah C, Woodward EA, Foroutan M, Lyu R, Yeoh GCT, Leedman PJ and Blancafort P: Epigenetic reactivation of tumor suppressor genes with CRISPRa technologies as precision therapy for hepatocellular carcinoma. Clin Epigenetics. 15:732023. View Article : Google Scholar : PubMed/NCBI

256 

Patra A, Deb M, Dahiya R and Patra SK: 5-Aza-2′-deoxycytidine stress response and apoptosis in prostate cancer. Clin Epigenetics. 2:339–348. 2011. View Article : Google Scholar

257 

Issa JP: Decitabine. Curr Opin Oncol. 15:446–451. 2003. View Article : Google Scholar : PubMed/NCBI

258 

Graça I, Sousa EJ, Baptista T, Almeida M, Ramalho-Carvalho J, Palmeira C, Henrique R and Jerónimo C: Anti-tumoral effect of the non-nucleoside DNMT inhibitor RG108 in human prostate cancer cells. Curr Pharm Des. 20:1803–1811. 2014. View Article : Google Scholar

259 

Wang C, Lei L, Xu Y, Li Y, Zhang J, Xu Y and Si S: Trichostatin C synergistically interacts with DNMT inhibitor to induce antineoplastic effect via inhibition of axl in bladder and lung cancer cells. Pharmaceuticals (Basel). 17:4252024. View Article : Google Scholar : PubMed/NCBI

260 

Laranjeira ABA, Hollingshead MG, Nguyen D, Kinders RJ, Doroshow JH and Yang SX: DNA damage, demethylation and anticancer activity of DNA methyltransferase (DNMT) inhibitors. Sci Rep. 13:59642023. View Article : Google Scholar : PubMed/NCBI

261 

Kim SJ, Cheresh P, Eren M, Jablonski RP, Yeldandi A, Ridge KM, Budinger GRS, Kim DH, Wolf M, Vaughan DE and Kamp DW: Klotho, an antiaging molecule, attenuates oxidant-induced alveolar epithelial cell mtDNA damage and apoptosis. Am J Physiol Lung Cell Mol Physiol. 313:L16–L26. 2017. View Article : Google Scholar : PubMed/NCBI

262 

Yamada Y, Venkadakrishnan VB, Mizuno K, Bakht M, Ku SY, Garcia MM and Beltran H: Targeting DNA methylation and B7-H3 in RB1-deficient and neuroendocrine prostate cancer. Sci Transl Med. 15:eadf67322023. View Article : Google Scholar : PubMed/NCBI

263 

Tikoo K, Ali IY, Gupta J and Gupta C: 5-Azacytidine prevents cisplatin induced nephrotoxicity and potentiates anticancer activity of cisplatin by involving inhibition of metallothionein, pAKT and DNMT1 expression in chemical induced cancer rats. Toxicol Lett. 191:158–166. 2009. View Article : Google Scholar : PubMed/NCBI

264 

Hosokawa O, Okabe M, Saito S, Saito T and Kurasaki M: Protective role of metallothionein on DNA damage in rat kidney caused by cis-diamminedichloroplatinum. Pharmacol Toxicol. 86:276–282. 2000. View Article : Google Scholar : PubMed/NCBI

265 

Francia G, Green SK, Bocci G, Man S, Emmenegger U, Ebos JM, Weinerman A, Shaked Y and Kerbel RS: Down-regulation of DNA mismatch repair proteins in human and murine tumor spheroids: Implications for multicellular resistance to alkylating agents. Mol Cancer Ther. 4:1484–1494. 2005. View Article : Google Scholar : PubMed/NCBI

266 

Plumb JA, Strathdee G, Sludden J, Kaye SB and Brown R: Reversal of drug resistance in human tumor xenografts by 2′-deoxy-5-azacytidine-induced demethylation of the hMLH1 gene promoter. Cancer Res. 60:6039–6044. 2000.PubMed/NCBI

267 

Dai X, Ren T, Zhang Y and Nan N: Methylation multiplicity and its clinical values in cancer. Expert Rev Mol Med. 23:e22021. View Article : Google Scholar : PubMed/NCBI

268 

Ricketts CJ, Hill VK and Linehan WM: Tumor-specific hypermethylation of epigenetic biomarkers, including SFRP1, predicts for poorer survival in patients from the TCGA kidney renal clear cell carcinoma (KIRC) project. PLoS One. 9:e856212014. View Article : Google Scholar : PubMed/NCBI

269 

Peters I, Dubrowinskaja N, Abbas M, Seidel C, Kogosov M, Scherer R, Gebauer K, Merseburger AS, Kuczyk MA, Grünwald V and Serth J: DNA methylation biomarkers predict progression-free and overall survival of metastatic renal cell cancer (mRCC) treated with antiangiogenic therapies. PLoS One. 9:e914402014. View Article : Google Scholar : PubMed/NCBI

270 

Dubrowinskaja N, Gebauer K, Peters I, Hennenlotter J, Abbas M, Scherer R, Tezval H, Merseburger AS, Stenzl A, Grünwald V, et al: Neurofilament heavy polypeptide CpG island methylation associates with prognosis of renal cell carcinoma and prediction of antivascular endothelial growth factor therapy response. Cancer Med. 3:300–309. 2014. View Article : Google Scholar : PubMed/NCBI

271 

Lou W, Krill D, Dhir R, Becich MJ, Dong JT, Frierson HF Jr, Isaacs WB, Isaacs JT and Gao AC: Methylation of the CD44 metastasis suppressor gene in human prostate cancer. Cancer Res. 59:2329–2331. 1999.PubMed/NCBI

272 

Kito H, Suzuki H, Ichikawa T, Sekita N, Kamiya N, Akakura K, Igarashi T, Nakayama T, Watanabe M, Harigaya K and Ito H: Hypermethylation of the CD44 gene is associated with progression and metastasis of human prostate cancer. Prostate. 49:110–115. 2001. View Article : Google Scholar : PubMed/NCBI

273 

Yegnasubramanian S, Kowalski J, Gonzalgo ML, Zahurak M, Piantadosi S, Walsh PC, Bova GS, De Marzo AM, Isaacs WB and Nelson WG: Hypermethylation of CpG islands in primary and metastatic human prostate cancer. Cancer Res. 64:1975–1986. 2004. View Article : Google Scholar : PubMed/NCBI

274 

Pan Y, Liu G, Zhou F, Su B and Li Y: DNA methylation profiles in cancer diagnosis and therapeutics. Clin Exp Med. 18:1–14. 2018. View Article : Google Scholar

275 

Altucci L and Rots MG: Epigenetic drugs: From chemistry via biology to medicine and back. Clin Epigenetics. 8:562016. View Article : Google Scholar : PubMed/NCBI

276 

Egger G, Liang G, Aparicio A and Jones PA: Epigenetics in human disease and prospects for epigenetic therapy. Nature. 429:457–463. 2004. View Article : Google Scholar : PubMed/NCBI

277 

Cossio FP, Esteller M and Berdasco M: Towards a more precise therapy in cancer: Exploring epigenetic complexity. Curr Opin Chem Biol. 57:41–49. 2020. View Article : Google Scholar : PubMed/NCBI

278 

Kondrashov A and Karpova E: Notes on functional modules in the assembly of CRISPR/Cas9-mediated epigenetic modifiers. Methods Mol Biol. 2198:401–428. 2021. View Article : Google Scholar

279 

Liang F, Dong Z, Ye J, Hu W, Bhandari RK, Mai K and Wang X: In vivo DNA methylation editing in zebrafish. Epigenetics. 18:21923262023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2025
Volume 55 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang Y, Wang Y, Fan X, Xu X, Wang H, Wang X, Shi T, Tang J, Guan Y, Li S, Li S, et al: Role of DNA methylation transferase in urinary system diseases: From basic to clinical perspectives (Review). Int J Mol Med 55: 19, 2025.
APA
Yang, Y., Wang, Y., Fan, X., Xu, X., Wang, H., Wang, X. ... Wang, A. (2025). Role of DNA methylation transferase in urinary system diseases: From basic to clinical perspectives (Review). International Journal of Molecular Medicine, 55, 19. https://doi.org/10.3892/ijmm.2024.5460
MLA
Yang, Y., Wang, Y., Fan, X., Xu, X., Wang, H., Wang, X., Shi, T., Tang, J., Guan, Y., Li, S., Wang, A."Role of DNA methylation transferase in urinary system diseases: From basic to clinical perspectives (Review)". International Journal of Molecular Medicine 55.2 (2025): 19.
Chicago
Yang, Y., Wang, Y., Fan, X., Xu, X., Wang, H., Wang, X., Shi, T., Tang, J., Guan, Y., Li, S., Wang, A."Role of DNA methylation transferase in urinary system diseases: From basic to clinical perspectives (Review)". International Journal of Molecular Medicine 55, no. 2 (2025): 19. https://doi.org/10.3892/ijmm.2024.5460