Skip to main content
Kidney Diseases logoLink to Kidney Diseases
. 2025 Jul 1;11(1):518–529. doi: 10.1159/000547202

Lysine Methyltransferases SMYD2 and SMYD3: Emerging Targets in Kidney Diseases

Xinyu Du a, Liyuan Yao a, Shougang Zhuang a,b,
PMCID: PMC12334150  PMID: 40787099

Abstract

Background

The SET and MYND domain-containing (SMYD) protein family is a group of lysine methyltransferases with SET and MYND domains and plays a critical role in regulating gene expression through the methylation of histone and non-histone proteins.

Summary

Studies have linked mutations or overexpression of SMYD2 and SMYD3 to various cancers, including renal carcinoma. Recent research also demonstrates that the expression levels and activity of SMYD2 and SMYD3 are increased in animal models of renal diseases such as autosomal dominant polycystic kidney disease, renal fibrosis, and diabetic nephropathy. Inhibiting either SMYD2 or SMYD3 pharmacologically or genetically can effectively suppress renal tumorigenesis and cystic formation while improving outcomes in renal fibrosis and diabetic nephropathy. Additionally, SMYD2 and SMYD3 are involved in the pathogenesis of acute kidney injury.

Key Messages

This review summarizes the roles of these two lysine methyltransferases in renal diseases, highlights their mechanisms, and emphasizes their potential as therapeutic targets for kidney disorders.

Keywords: Acute kidney injury, Chronic kidney disease, Diabetic nephropathy, Renal cell carcinoma, Epigenetic regulation, Lysine methyltransferase SMYD, Renal fibrosis

Introduction

Aberrant epigenetic modifications are critically involved in the pathogenesis of kidney diseases, with histone methylation being a key research focus. Histone methylation modification enzymes regulate various cellular functions, including heterochromatin formation, X chromosome inactivation, transcriptional regulation, and stem cell maintenance and differentiation [1]. Increasing evidence suggests that the dysregulation or aberrant expression of these enzymes contributes significantly to kidney disease progression by disrupting gene expression programs and altering protein functions, ultimately impairing renal cellular metabolism and homeostasis [2, 3].

The SMYD (SET and MYND domain-containing) family of lysine methyltransferases – comprising SMYD1 through SMYD5 – has attracted growing interest. While initially characterized for their roles in embryogenesis, cardiac and skeletal muscle development, and tumorigenesis [46], members of the SMYD family also play essential roles in renal physiology and pathology. Through histone and non-histone methylation, SMYD proteins modulate key signaling pathways and gene expression networks in renal cells, thereby influencing cellular characteristics and disease-related processes [710]. For example, SMYD2 has been shown to promote the development and progression of both acute kidney injury (AKI) and chronic kidney disease (CKD) by regulating pathological mechanisms such as cell proliferation, apoptosis, inflammation, and fibrosis [1012]. Given these multifaceted roles, the SMYD family is increasingly recognized as a potential therapeutic target in kidney disease research.

Among the SMYD family members, only SMYD2 and SMYD3 exhibit notable expression in renal tissues. This review focuses on the biological functions and regulatory mechanisms of SMYD2 and SMYD3 in kidney cells and across various experimental models of renal diseases. Specifically, we examine their roles in renal tumors, autosomal dominant polycystic kidney disease (ADPKD), AKI, renal fibrosis, and diabetic nephropathy (Table 1). In addition, we discuss the therapeutic potential of targeting SMYD2 and SMYD3 in these pathological conditions.

Table 1.

SMYD2 and SMYD3 inhibition on kidney diseases in various in vitro and in vivo models

Inhibitors or KO mice Models in vitro Models in vivo Effects and mechanisms References
SMYD2 siRNA, AZ505 HK-2 cells, HEK293T cells, OSRC-2 cells, 786-O cells Tumor orthotopic xenograft in nude mice, tumor subcutaneous xenograft in nude mice, lung metastasis mice model Inhibit ccRCC cell growth, migration, and invasion by blocking SMYD2/miR-125b/DKK3 pathway, enhance the synergistic effect with anticancer drugs [13]
SMYD2 siRNA, AZ505 MEK cells, PH2 cells, PN24 cells, mIMCD3 cells Ksp-Cre Pkd1-knockout mice model, tamoxifen-induced conditional Pkd1-knockout mice model Delay renal cyst formation and growth, inhibit STAT3 and NF-κB-p65 activation [10]
SMYD2 siRNA, AZ505 NRK-49F cells, NIH3T3 fibroblasts Ksp-Cre Pkd1-knockout mice model, tamoxifen-induced conditional Pkd1-knockout mice model Ameliorate renal fibrosis, suppress the TGF-β-induced expression of fibrotic genes, activation of renal fibroblasts, and the formation of stress fibers [9]
SMYD2 siRNA, AZ505 NRK-49F cells Mouse model of unilateral ureteral obstruction Attenuate fibrosis, reduce the activation of renal interstitial fibroblasts, inhibit TGF-β1-induced EMT [14]
SMYD2 siRNA, LV-SMYD2, AZ505, LLY507 NRK-52E cells, HK2 cells Cisplatin-induced CKD mouse model Attenuate renal fibrosis, inflammation and apoptosis, inhibit cisplatin-induced EMT [11, 15]
SMYD2 siRNA HK2 cells Glyoxylate-induced CaOx stones mouse model Inhibit CaOx-induced glycolysis, apoptosis, inflammation, and EMT [16]
AZ505 NRK-49F cells Streptozotocin (STZ)-induced DN Attenuate renal fibrosis, inhibit activation of rat renal fibroblasts [17]
AZ505, RTT extract / Streptozotocin (STZ)-induced DN Attenuate renal fibrosis and inflammation [18]
SMYD2 siRNA, AZ505 MTEC cells Cisplatin-induced AKI mouse model Reduce renal dysfunction and renal tubular cell death, prevent renal tubular injury [12]
SMYD3 shRNA TK10 cells, KRC/Y cells, A498 cells, 786-O cells Tumor xenograft in nude mice Inhibit survival, invasion, and growth of RCC cells with VHL mutation [8]
SMYD3 shRNA mIMCD3 cells, HEK293T cells, NIH3T3 fibroblasts Ksp-Cre Pkd1-knockout mice model, tamoxifen-induced conditional Pkd1-knockout mice model Delayed renal cyst growth and improve kidney function, reduced genomic instability, and primary cilia [7]
SMYD3 siRNA, BCI-121 MTEC cells I/R-induced AKI mouse model Aggravate I/R injury and apoptosis, inhibit EGF-induced proliferation and dedifferentiation [19]

SMYD Family

Structure and Function of the SMYD Family

The SMYD (SET and MYND domain-containing) family of proteins is located on chromosome 1q32.3 of the human genome [20] and is characterized by broad yet spatiotemporally regulated expression across mammalian tissues and organs. Despite their functional diversity, SMYD proteins share a structurally conserved architecture consisting of several key domains (Fig. 1). These include the following: (1) SET domain – comprising three subdomains: N-SET (N-terminal), I-SET (insertion), and C-SET (core), this domain is responsible for catalyzing methylation reactions on both histone and non-histone substrates; (2) MYND domain – a zinc finger structure composed of seven cysteine residues and one histidine, primarily involved in mediating protein-protein interactions; (3) CTD – formed by anti-parallel α-helices, the CTD interacts with the I-SET and post-SET domains to create a deep and narrow substrate-binding pocket [6]. The CTD also contributes to enzymatic activation by facilitating the binding of SMYD proteins to substrates or molecular chaperones such as heat shock protein 90 (HSP90) [21].

Fig. 1.

Fig. 1.

Domain architecture of SMYD family proteins. SMYD (SET and MYND domain-containing) family proteins share a conserved structural organization composed of the following key domains arranged sequentially: N-SET domain (N-terminal portion of the SET domain); MYND domain, which contains a zinc finger motif critical for protein-protein interactions; I-SET domain (intervening SET subdomain); C-SET domain (C-terminal portion of the SET domain); post-SET domain, involved in substrate recognition and catalysis; C-terminal domain (CTD), which may contribute to substrate specificity and subcellular localization.

The biochemical activity of SMYD proteins arises from their ability to methylate a broad spectrum of histone and non-histone targets through a substrate-dependent mechanism. Notably, multiple SMYD family members can methylate the same substrate, potentially modifying it simultaneously or in a coordinated fashion. Subcellular localization studies have shown that SMYD family members function in both the nucleus and cytoplasm. Nuclear-localized SMYD proteins influence gene expression and chromatin remodeling, whereas cytoplasmic forms regulate diverse cellular signaling pathways and other biological processes. Collectively, the multifaceted roles of SMYD proteins highlight their importance not only in normal development but also in the pathogenesis of various diseases.

SMYD2

SMYD2 is one of the most extensively studied members of the SMYD family and is distinguished by its unique structural and functional properties. Binding of cofactors induces conformational changes in its CTD, expanding its protein interaction network and enhancing its enzymatic activity [22]. SMYD2 is highly expressed in various tissues, including the heart, brain, liver, kidney, thymus, and ovaries, and is localized to both the nucleus and cytoplasm [20].

Functionally, SMYD2 catalyzes the methylation of histone substrates – specifically H3K4 and H3K36 – as well as a range of non-histone proteins, such as p53 [23], heat shock protein 90 (HSP90) [24], and poly(ADP-ribosyl)transferase 1 [25]. Through these modifications, SMYD2 promotes gene transcription and regulates essential cellular processes including proliferation, differentiation, apoptosis, angiogenesis, and cancer cell invasion and metastasis. It also plays important roles in brain development and the maturation of skeletal muscle and cardiomyocytes, while contributing to the pathogenesis of various cancers, including gastric, bladder, colorectal, and breast cancer [7, 2628]. For example, SMYD2 has been shown to promote breast cancer progression by enhancing the expression of the melanoma cell adhesion molecule through H3K36me2 methylation at its promoter region, thereby facilitating tumor growth and maintaining stem-like properties [29]. In cardiomyocytes, SMYD2 suppresses the intrinsic apoptotic pathway by methylating lysine 370 of the p53 transcription factor, thus inhibiting p53-mediated cell death [30].

SMYD3

The MYND domain of SMYD3 is structurally distinctive, positioned near the substrate-binding pocket and characterized by a positively charged surface that enables direct DNA interaction. This unique feature enhances its histone methyltransferase activity by facilitating substrate recognition and chromatin association [31]. SMYD3 exhibits spatiotemporally regulated expression across various organs, including the heart, skeletal muscle, testis, thymus, brain, kidney, and ovary. Notably, its nuclear accumulation is particularly prominent during the S and G2-M phases of the cell cycle, reflecting its involvement in cell proliferation [32].

SMYD3 methylates a broad range of substrates, including histones – such as H3K4 [33], H4K5 [34], and H4K20 [35] – as well as non-histone proteins like vascular endothelial growth factor receptor 1 [36], mitogen-activated protein kinase kinase kinase 2 [37], AKT1 [38], human epidermal growth factor receptor 2 [39], and enhancer of zeste homolog 2 [40]. Through H3K4 methylation in particular, SMYD3 modulates chromatin structure and transcriptional activity [41]. This epigenetic regulation plays a crucial role in controlling the expression of genes involved in cell cycle progression, differentiation, and signal transduction, including the activation of several oncogenes. Furthermore, SMYD3 is implicated in skeletal muscle development and various malignancies [41, 42]. For example, SMYD3 has been shown to upregulate matrix metalloproteinase-9 expression in diverse cancer cell types – including leukemic, fibrosarcoma, and breast cancer cells – by catalyzing H3K4 methylation at the matrix metalloproteinase-9 promoter region [5].

SMYD2 and SMYD3 in Kidney Diseases

A growing body of evidence indicates that SMYD2 and SMYD3 play pivotal roles in the onset and progression of a wide range of kidney diseases, including renal cell carcinoma (RCC), autosomal dominant polycystic kidney disease (ADPKD), renal fibrosis, diabetic nephropathy, and AKI. Notably, the majority of these findings have been derived from studies using animal models.

Role of SMYD2 in Kidney Diseases

SMYD2 exerts its biological functions primarily through the methylation of both histone and non-histone target proteins. In the cardiovascular system, SMYD2 preserves sarcomere integrity by methylating HSP90 and the HSP90-titin complex, which is essential for maintaining myofibrillar structure and function [24]. In Hirschsprung’s disease, SMYD2 promotes cellular proliferation and migration by modulating METTL3 expression and enhancing m6A RNA methylation [43]. In the context of kidney disease, SMYD2 regulates critical pathological processes – including cell proliferation, apoptosis, inflammation, and fibrosis – through the methylation of histone substrates (e.g., H3K36me3) and non-histone proteins such as NF-κB, STAT3, and phosphatase and tensin homolog (PTEN). Its renal specificity is attributed to two major factors: (1) pathway-specific targeting in renal disease – SMYD2 coordinates the regulation of multiple disease-relevant pathways, including carcinogenesis (via the miR-125b/DKK3 axis), cystogenesis (via the STAT3/NF-κB feedback loop), fibrogenesis (through TGF-β/Smad3 signaling), and AKI (through p53-dependent apoptosis); (2) kidney-specific microenvironmental cues – conditions such as high glucose and hypoxia characteristic of diseased renal tissue aberrantly activate SMYD2. This leads to enhanced methylation of renal substrates such as STAT3 at lysine 685 in ADPKD, and PTEN at lysine 313 in renal fibrosis. These context-dependent modifications underscore SMYD2’s critical involvement in kidney disease pathogenesis and highlight its potential as a therapeutic target.

SMYD2 and RCC

SMYD2 plays a pivotal role in promoting the development, metastasis, and multidrug resistance of RCC. It is significantly overexpressed in RCC patient tissues and xenograft tumors, and its elevated expression correlates with poor clinical outcomes – including reduced overall and disease-free survival – in patients with clear cell RCC (ccRCC) [13, 44, 45]. The histological similarity between benign eosinophilic tumors and RCC often complicates diagnosis [46]. However, SMYD2 transcript levels have diagnostic utility: they can distinguish papillary RCC (chRCC) from eosinophilic tumors with a sensitivity of 71.0% and specificity of 73.3%, underscoring its potential as both a diagnostic and prognostic biomarker for RCC aggressiveness [44, 45]. Mechanistically, chromatin immunoprecipitation studies have demonstrated that the SMYD2 inhibitor AZ505 blocks SMYD2 binding to the miR-125b promoter, thereby inhibiting the SMYD2/miR-125b/DKK3 oncogenic axis. This suppression leads to reduced proliferation, migration, and invasion of ccRCC cells. In addition, SMYD2 inhibition downregulates the expression of P-glycoprotein (P-gP), a key effector of chemoresistance, thereby enhancing the chemosensitivity of RCC cells [13].

SMYD2 and ADPKD

SMYD2 has emerged as a novel epigenetic regulator of cyst growth in ADPKD [47] (Fig. 2). It is significantly upregulated in renal epithelial cells from PKD1-mutant mice and ADPKD patient samples. Loss of SMYD2 delays cyst formation and progression in PKD1-mutant kidneys. Mechanistically, SMYD2 methylates STAT3 at lysine 685 and NF-κB p65 at lysine 310, thereby modulating their phosphorylation status and promoting proliferation and apoptosis in cystic epithelial cells. Moreover, SMYD2 orchestrates pro-inflammatory cytokine networks through two independent but synergistic positive feedback loops: the SMYD2-STAT3-IL-6-SMYD2 axis and the SMYD2-NF-κB p65-TNFα-SMYD2 axis, which collectively drive cyst growth in ADPKD [10]. In parallel, SMYD2 directly binds to promoters of transforming growth factor-beta (TGF-β) and fibrosis-related genes, regulating their transcription and activating fibroblasts. This establishes a SMYD2-TGF-β-Smad3-SMYD2 feedback loop that exacerbates renal fibrosis associated with ADPKD [9].

Fig. 2.

Fig. 2.

SMYD2-mediated pathways in PKD1-mutant renal epithelial cells and kidneys. In PKD1-mutant renal epithelial cells and kidneys, upregulated SMYD2 contributes to cyst growth and fibrosis through multiple signaling mechanisms. SMYD2 methylates the p65 subunit of NF-κB and STAT3, enhancing their activation. It modulates cytokine networks through two independent positive feedback loops: the SMYD2-STAT3-IL-6-SMYD2 axis and the SMYD2-NF-κB p65-TNFα-SMYD2 axis, thereby amplifying pro-inflammatory and proliferative signaling to promote cystogenesis. Additionally, TGF-β/Smad3 signaling upregulates SMYD2 transcription. In turn, SMYD2 enhances fibrotic gene expression via histone methylation, establishing a SMYD2-TGF-β-Smad3-SMYD2-positive feedback loop that drives renal fibrosis. Key downstream targets include fibronectin (Fn), collagen I (Col1a1), and alpha-smooth muscle actin (α-SMA).

SMYD2 and AKI

SMYD2 plays a significant role in AKI. Both SMYD2 and its histone methylation mark, H3K36me3, are upregulated in murine kidneys following ischemia/reperfusion (I/R) injury, coinciding with increased levels of cleaved caspase-3, p53, and phosphorylated p53 in proximal tubules, as well as activation of JNK-STAT3 signaling pathways [48]. Caspase-3 and p53 mediate apoptotic cell death, while STAT3 contributes to inflammatory responses [49].

Pharmacological inhibition of SMYD2 by AZ505 confers protection against cisplatin-induced AKI. AZ505 treatment reduces histopathological kidney damage, suppresses tubular apoptosis, decreases the release of inflammatory cytokines, and promotes tubular regeneration. Mechanistically, AZ505 inhibits cleaved caspase-3, BAX expression, and p53 phosphorylation, while enhancing the expression of cell cycle proteins and suppressing STAT3 and NF-κB phosphorylation. These effects collectively promote tubular cell proliferation and mitigate inflammation [12, 15].

Although these findings position SMYD2 as a promising therapeutic target in AKI, current evidence largely stems from cisplatin-induced injury models. Further research is required to elucidate SMYD2’s role across diverse AKI etiologies.

SMYD2 and Renal Fibrosis

SMYD2 drives the progression of renal fibrosis (Fig. 3). Both SMYD2 and its associated histone mark H3K36me3 are upregulated in chronically injured kidneys. Inhibition of SMYD2 effectively attenuates fibrosis by suppressing G2/M phase epithelial cell cycle arrest, inhibiting renal interstitial fibroblast activation and proliferation, and modulating inflammatory responses and signaling pathways [14].

Fig. 3.

Fig. 3.

SMYD2-mediated pathways in chronic kidney disease (CKD). SMYD2 drives fibrosis through multiple context-specific mechanisms in CKD. In calcium oxalate (CaOx)-induced nephropathy, SMYD2 methylates PTEN, resulting in the activation of the AKT/mTOR pathway. This promotes glycolytic reprogramming in renal tubular cells, enhancing fibrotic progression. In unilateral ureteral obstruction (UUO) and cisplatin-induced CKD models, upregulated SMYD2 contributes to fibrosis by inducing G2/M cell cycle arrest, renal fibroblast activation, epithelial-to-mesenchymal transition (EMT), and by activating TGF-β/Smad3, ERK, AKT, NF-κB, and STAT3 pathways. In diabetic kidney disease (DKD), SMYD2 is upregulated in response to high glucose (HG) or streptozotocin (STZ) exposure. It promotes NF-κB p65 phosphorylation, nuclear translocation, and transcriptional activation of fibrotic genes via histone methylation, exacerbating renal inflammation and fibrosis.

In human and animal models of calcium oxalate (CaOx) kidney stones, SMYD2 and glycolytic enzymes are elevated. SMYD2 promotes tubular glycolysis and metabolic reprogramming by activating the AKT/mTOR pathway through methylation of PTEN, contributing to tubular injury and fibrosis [16]. In unilateral ureteral obstruction-induced CKD models, treatment with the SMYD2 inhibitor AZ505 suppresses Snail and Twist expression, inhibiting epithelial-to-mesenchymal transition and reducing ERK1/2, STAT3, and NF-κB activation. This leads to decreased extracellular matrix deposition and fibrosis [14]. Additionally, both SMYD2 inhibitors AZ505 and LLY507 ameliorate cisplatin-induced CKD by blocking TGF-β1/Smad3-mediated fibroblast activation, as well as inhibiting NF-κB activation and apoptosis [11, 15].

SMYD2 and Diabetic Kidney Disease

Diabetic kidney disease (DKD), a major complication of diabetes, is characterized by glomerular hypertrophy, proteinuria, reduced glomerular filtration rate, and progressive fibrosis, ultimately leading to renal failure. Pathological features include mesangial hyperplasia, thickening of the glomerular basement membrane, capillary occlusion, podocyte injury, and fibroblast activation [50]. Hyperglycemia promotes fibroblast transdifferentiation into myofibroblasts, which secretes excessive extracellular matrix, driving extensive fibrosis [51, 52]. Nuclear factor kappa B (NF-κB) plays a key role in regulating this phenotypic transition of fibroblasts [53].

SMYD2 contributes to DKD pathogenesis by activating renal fibroblasts under hyperglycemic conditions. SMYD2 expression is upregulated in the kidneys of streptozotocin-induced diabetic mice [54]. Both Ranunculus ternatus extracts and the SMYD2 inhibitor AZ505 reduce SMYD2 levels, H3K36me3 histone methylation, and NF-κB p65 phosphorylation, exerting anti-inflammatory and anti-fibrotic effects in diabetic nephropathy models [17, 18]. In NRK49F fibroblast cells, high glucose induces nuclear co-localization of phosphorylated NF-κB p65 (p-p65) and SMYD2. Treatment with AZ505 blocks p-p65 nuclear translocation and expression, thereby inhibiting hyperglycemia-induced fibroblast activation [17].

Role of SMYD3 in Kidney Disease

Current research highlights that SMYD3 regulates cell fate decisions through epigenetic reprogramming of target gene transcription, a fundamental mechanism conserved across various organ diseases. In cancer, SMYD3 promotes tumor progression by activating kinases and facilitating degradation of tumor suppressors [41]. During skeletal muscle development, SMYD3 modulates differentiation by targeting the myogenic regulator myogenin [55]. In vascular remodeling, SMYD3 enhances vascular smooth muscle cell proliferation and migration by increasing H3K4me3 marks at specific gene promoters [56]. In kidney disease (Fig. 4), SMYD3 epigenetically contributes to the progression of RCC and ADPKD, while also facilitating repair processes in AKI. It displays three kidney-specific features: (1) an oncogenic mechanism dependent on a von Hippel-Lindau (VHL)-deficient microenvironment; (2) regulation of ciliary function via α-tubulin methylation; and (3) a context-dependent protective role during AKI regeneration.

Fig. 4.

Fig. 4.

SMYD3-associated pathways across kidney disease models. SMYD3 exerts distinct roles across different renal pathologies via epigenetic regulation: (1) ischemia/reperfusion (I/R)-induced acute kidney injury (AKI): SMYD3 enhances H3K4me3 enrichment at the EGFR promoter, promoting its transcription. This upregulates EGFR/AKT signaling, supporting tubular cell survival, dedifferentiation, and proliferation, thereby facilitating post-injury renal repair. (2) Renal cell carcinoma (RCC): SMYD3 is recruited by lncRNA CDKN2B-AS1 and CBP to the NUF2 promoter, increasing H3K4me3 and H3K27ac to drive NUF2 expression and tumor progression. In parallel, the VHL/HIF-2α/SMYD3 cascade directs SMYD3 to the EGFR promoter, enhancing H3K4me3 deposition and EGFR transcription, further promoting RCC malignancy. (3) Autosomal dominant polycystic kidney disease (ADPKD): SMYD3 modulates key PKD-associated signaling pathways (e.g., NF-κB p65, STAT3, ERK1/2, and cyclin D1) that regulate cell proliferation, apoptosis, and the cell cycle. Additionally, SMYD3 methylates α-tubulin-K40, disrupting mitotic integrity and ciliary function, contributing to genomic instability and cystogenesis.

SMYD3 and RCC

SMYD3 expression is markedly elevated in RCC tumors and is associated with advanced disease stage, aggressive histological subtypes, high nuclear grade, and poorer patient survival outcomes [8, 57]. Mechanistically, the long noncoding RNA CDKN2B-AS1 recruits SMYD3 along with the histone acetyltransferase CREB-binding protein to the promoter region of NUF2, promoting enrichment of H3K4me3 and H3K27ac marks, thereby enhancing NUF2 transcription and facilitating ccRCC progression [58]. Additionally, SMYD3 knockdown significantly impairs RCC cell proliferation, reduces colony formation, diminishes tumorigenic potential, and induces apoptosis [8]. Through the VHL/HIF-2α/SMYD3 signaling axis, SMYD3 catalyzes H3K4me2 and H3K4me3 modifications at the epidermal growth factor receptor (EGFR) promoter, increasing SP1 transcription factor binding and upregulating EGFR expression, thereby driving RCC progression [8].

SMYD3 and ADPKD

In ADPKD, loss of polycystin-1 (encoded by PKD1) and disruption of primary cilia are critical drivers of cyst growth [59]. Chromosomal instability contributes to clonal heterogeneity, further accelerating cyst progression, a hallmark of ADPKD [60]. SMYD3 is upregulated in PKD1-mutant mice and human ADPKD kidneys, where it enhances transcriptional activity through increased H3K4me3 deposition. Notably, SMYD3 knockout delays cyst development and improves renal function in PKD1-mutant mice [7]. Mechanistically, SMYD3 influences key PKD-related signaling pathways – such as NF-κB p65, STAT3, ERK1/2, CDK4, CDK6, and cyclin D1 – regulating cell proliferation, apoptosis, and cell cycle progression. Moreover, SMYD3 modulates primary cilia assembly by localizing to centrosomes and promotes genomic instability by methylating α-tubulin at lysine 40 (K40), thereby exacerbating cyst growth [7].

SMYD3 and AKI

Our recent studies demonstrated that SMYD3 is upregulated in murine kidneys following I/R injury. Pharmacological inhibition of SMYD3 using the selective inhibitor BCI-121 worsened tubular injury, increased apoptosis, and impaired renal function in this model [19]. Further investigations revealed that BCI-121 also suppressed renal epithelial cell differentiation and proliferation. Mechanistically, SMYD3 promotes EGFR expression and activates EGFR/AKT signaling by mediating H3K4me3 enrichment at the EGFR promoter. Considering the pivotal role of EGFR in tubular cell survival and regeneration after AKI [61], SMYD3-mediated activation of the EGFR pathway likely plays a critical protective role in renal recovery.

SMYD3 and Renal Fibrosis

Although SMYD3’s role in renal fibrosis remains to be fully elucidated, it has been shown to regulate renal fibroblast proliferation. Recent studies using iTRAQ technology identified elevated SMYD3 and H3K4me3 levels in renal tissues from rats with fetal growth restriction [62]. Moreover, SMYD3 expression strongly correlates with proliferation and reprogramming efficiency in porcine fibroblasts. SMYD3 upregulation enhances fibroblast proliferation and improves developmental potential in nuclear transfer embryos [63]. Similarly, overexpression of SMYD3 promotes proliferation and increases the efficiency of induced pluripotent stem cell generation in bovine embryonic fibroblasts [64]. Given that renal fibroblast activation and proliferation are key drivers of CKD progression, it is plausible that SMYD3 plays an important role in renal fibrosis and CKD pathogenesis. This hypothesis warrants further investigation.

Distinct Roles of SMYD2 and SMYD3 in Kidney Diseases

While SMYD2 and SMYD3 share pathogenic roles in promoting renal tumors, ADPKD, fibrosis, and diabetic nephropathy, their functional roles and underlying mechanisms diverge in AKI. SMYD2 exacerbates tubular damage by activating p53-dependent apoptosis, cleaved caspase 3, and JNK-STAT3 inflammatory pathways in a murine model of cisplatin-induced AKI [12]. In contrast, SMYD3 exerts a protective effect on the renal epithelium by sustaining EGFR/AKT-mediated cell survival and regeneration in a murine model of I/R-induced AKI [19]. The reasons behind this dichotomy remain unclear but may relate to the specific AKI model employed. Further studies are warranted to comprehensively assess the distinct functional roles of SMYD2 and SMYD3 across diverse AKI models.

SMYD2 and SMYD3 as Potential Therapeutic Targets for Kidney Diseases

SMYD2

SMYD2 is upregulated in renal tumors, AKI, and CKD, with its expression closely correlating with disease onset and progression. Thus, SMYD2 represents a promising therapeutic target for treating renal tumors as well as acute and CKDs.

SMYD2 inhibitors are categorized into five structural classes: benzoxazinones (e.g., AZ505), diphenylpiperazines (e.g., LLY-507), aminopyrazolone (e.g., BAY-598), noncompetitive inhibitors (EPZ series), and SAM-competitive inhibitors (e.g., PFI-5) [65]. Among these, BAY-598 and EPZ compounds demonstrate favorable oral bioavailability but exhibit limited cellular activity [66, 67]. AZ505 is the most extensively studied in kidney disease models, showing therapeutic efficacy against fibrosis, ADPKD, and AKI; however, long-term safety and human data remain lacking. Notably, 6-month administration of AZ505 (10 mg/kg, i.p.) significantly delayed cyst formation in mice with good tolerability and no observed weight loss [14]. LLY-507 offers superior pharmacokinetics, high selectivity, and low cytotoxicity [68] and effectively attenuated fibrosis in cisplatin-induced CKD models with efficacy comparable to AZ505 and without weight loss [11].

Significant progress has been made in elucidating SMYD2’s role in cancer progression. SMYD2 is frequently upregulated across multiple cancers, where its expression correlates with enhanced proliferation, metastasis, immune evasion, and drug resistance. The SMYD2 inhibitor AZ505 suppresses triple-negative breast cancer growth by inhibiting methylation mediated by STAT3, NF-κB p65, and protein tyrosine phosphatase non-receptor type 13 (PTPN13) [27]. Additionally, AZ505 reduces proliferation in human renal cancer primary cells and xenograft models, while also enhancing chemosensitivity in RCC [13]. Given cisplatin’s widespread use as a chemotherapeutic agent and AZ505’s protective effects against cisplatin-induced kidney injury, dual targeting of SMYD2 could both potentiate cisplatin’s antitumor efficacy and confer renal protection. Therefore, AZ505 holds promise as a chemotherapeutic adjuvant in RCC and as a novel therapeutic agent for CKDs such as ADPKD, potentially improving overall patient outcomes.

SMYD3

SMYD3 is overexpressed in RCC, correlating with tumor progression and poor patient prognosis. Silencing SMYD3 markedly inhibits RCC growth and invasiveness, making it a promising therapeutic target. Several high-selectivity SMYD3 inhibitors have been developed, including BCI-121, nano-EM127, BAY-6035, EPZ031686, EPZ030456, and GSK2807 [69]. Among these, EM127 and BAY-6035 show significantly higher binding affinity compared to the micromolar-potency inhibitor BCI-121 [7072]. EM127 inhibits SMYD3 through covalent binding to Cys186, while BAY-6035 is highly selective and inactive against 34 other methyltransferases. In contrast, BCI-121’s lower potency raises concerns about off-target effects. However, EM127, BAY-6035, and related compounds exhibit poor membrane permeability – likely due to nitrogenous heterocycles – and pose risks of CYP450-mediated drug-drug interactions, necessitating careful metabolite toxicity evaluation [7072]. EPZ031686 improves lipophilicity by blocking hydrogen-bond donors, showing strong cellular activity, excellent oral bioavailability, and metabolic stability [73], making it a promising candidate for targeted kidney disease therapy. Both BCI-121 (administered at 50 mg/kg thrice weekly for 6 weeks) and nano-EM127 effectively inhibit SMYD3 in vivo [74, 75]. Key challenges remain: EM127’s low permeability and potential toxicity linked to irreversible inhibition, and BCI-121’s off-target risks. Notably, EM127’s sustained inhibition may benefit chronic diseases like ADPKD and cancer.

Despite promising preclinical results with SMYD2/3 inhibitors such as AZ505 in kidney disease models, clinical translation faces several hurdles: (1) tissue-specific paradoxes – SMYD2 is cardioprotective [30] but exacerbates apoptosis in AKI [12], so systemic administration could cause unpredictable off-target effects. SMYD3 is oncogenic in RCC but promotes renal tubular regeneration in AKI, meaning its inhibition might impair tissue repair. (2) Lack of human pharmacokinetic/pharmacodynamic data – no clinical trials have been reported for these inhibitors. Although AZ505 and LLY-507 appear safe in animal studies, their long-term human safety remains uncharacterized. (3) Model limitations – most SMYD2 research in AKI uses cisplatin-induced models; studies in ischemia/reperfusion or sepsis models are needed. Similarly, the role of SMYD3 in renal fibrosis is mostly inferred from fibroblast data, requiring direct evidence from CKD models.

Conclusions and Perspectives

The SMYD family regulates transcription and diverse cellular processes, including proliferation, differentiation, and apoptosis [69, 76]. Among its five members, SMYD2 and SMYD3 have emerged as key epigenetic regulators in kidney diseases, modulating cystogenesis, fibrosis, diabetic nephropathy, and AKI through methylation of both histone and non-histone substrates [7, 9, 10, 12, 14, 18, 77]. While SMYD2 has been extensively studied in renal tumors [13, 45], ADPKD [9, 10], fibrosis [11, 14, 16], and AKI [12], research on SMYD3 remains nascent, primarily focusing on RCC and ADPKD [7, 8]. Despite accumulating evidence implicating both enzymes in diverse renal pathologies, the precise molecular mechanisms remain incompletely understood. Both SMYD2 and SMYD3 engage multiple signaling pathways, such as TGF-β and STAT3, and methylate various substrates, complicating mechanistic delineation [9, 14, 69, 76]. Notably, SMYD2 displays context-dependent functions – promoting fibrosis in ADPKD while exacerbating inflammation during AKI [10, 12]. Thus, systemic inhibition risks unintended off-target effects. Addressing inhibitor specificity and pathway complexity is essential for therapeutic translation. Future research should prioritize comprehensive mapping of non-histone substrates and context-dependent interactions. Moreover, it is critical to evaluate the efficacy and mechanisms of various SMYD2/3 inhibitors across diverse animal models of kidney disease and to employ combinatorial approaches for deeper mechanistic insights – foundational steps toward realizing their full therapeutic potential.

Conflict of Interest Statement

Shougang Zhuang was a member of the journal’s Editorial Board at the time of submission. The other authors declared no competing interest in this work.

Funding Sources

This study was supported by grants from the National Natural Science Foundation of China (82370698 and 82070700 to S.Z.) and US National Institutes of Health (1R56DK135540-01 to S.Z.).

Author Contributions

Xinyu Du and Liyuan Yao are responsible for collecting literature and drafting the review. Shougang Zhuang revised the manuscript. All authors read and approved the final manuscript.

Funding Statement

This study was supported by grants from the National Natural Science Foundation of China (82370698 and 82070700 to S.Z.) and US National Institutes of Health (1R56DK135540-01 to S.Z.).

References

  • 1. Martin C, Zhang Y. The diverse functions of histone lysine methylation. Nat Rev Mol Cell Biol. 2005;6(11):838–49. [DOI] [PubMed] [Google Scholar]
  • 2. Li T, Yu C, Zhuang S. Histone methyltransferase EZH2: a potential therapeutic target for kidney diseases. Front Physiol. 2021;12:640700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Guo C, Dong G, Liang X, Dong Z. Epigenetic regulation in AKI and kidney repair: mechanisms and therapeutic implications. Nat Rev Nephrol. 2019;15(4):220–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Du SJ, Tan X, Zhang J. SMYD proteins: key regulators in skeletal and cardiac muscle development and function. Anat Rec. 2014;297(9):1650–62. [DOI] [PubMed] [Google Scholar]
  • 5. Han TS, Kim DS, Son MY, Cho HS. SMYD family in cancer: epigenetic regulation and molecular mechanisms of cancer proliferation, metastasis, and drug resistance. Exp Mol Med. 2024;56(11):2325–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Spellmon N, Holcomb J, Trescott L, Sirinupong N, Yang Z. Structure and function of SET and MYND domain-containing proteins. Int J Mol Sci. 2015;16(1):1406–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Agborbesong E, Zhou JX, Zhang H, Li LX, Harris PC, Calvet JP, et al. Overexpression of SMYD3 promotes autosomal dominant polycystic kidney disease by mediating cell proliferation and genome instability. Biomedicines. 2024;12(3):603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Liu C, Liu L, Wang K, Li XF, Ge LY, Ma RZ, et al. VHL-HIF-2α axis-induced SMYD3 upregulation drives renal cell carcinoma progression via direct trans-activation of EGFR. Oncogene. 2020;39(21):4286–98. [DOI] [PubMed] [Google Scholar]
  • 9. Li LX, Zhang L, Agborbesong E, Zhang X, Zhou JX, Li X. Cross talk between lysine methyltransferase Smyd2 and TGF-β-Smad3 signaling promotes renal fibrosis in autosomal dominant polycystic kidney disease. Am J Physiol Ren Physiol. 2022;323(2):F227–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Li LX, Fan LX, Zhou JX, Grantham JJ, Calvet JP, Sage J, et al. Lysine methyltransferase SMYD2 promotes cyst growth in autosomal dominant polycystic kidney disease. J Clin Investig. 2017;127(7):2751–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Chen M, Zuo S, Chen S, Li X, Zhang T, Yang D, et al. Pharmacological inhibition of SMYD2 protects against cisplatin-induced renal fibrosis and inflammation. J Pharmacol Sci. 2023;153(1):38–45. [DOI] [PubMed] [Google Scholar]
  • 12. Cui B, Hou X, Liu M, Li Q, Yu C, Zhang S, et al. Pharmacological inhibition of SMYD2 protects against cisplatin-induced acute kidney injury in mice. Front Pharmacol. 2022;13:829630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Yan L, Ding B, Liu H, Zhang Y, Zeng J, Hu J, et al. Inhibition of SMYD2 suppresses tumor progression by down-regulating microRNA-125b and attenuates multi-drug resistance in renal cell carcinoma. Theranostics. 2019;9(26):8377–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Liu L, Liu F, Guan Y, Zou J, Zhang C, Xiong C, et al. Critical roles of SMYD2 lysine methyltransferase in mediating renal fibroblast activation and kidney fibrosis. FASEB J. 2021;35(7):e21715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Zuo S, Yuan H, Li X, Chen M, Peng R, Chen S, et al. SMYD2 promotes renal tubular cell apoptosis and chronic kidney disease following cisplatin nephrotoxicity. FASEB J. 2025;39(10):e70651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Pan S, Yuan T, Xia Y, Yu W, Li H, Rao T, et al. SMYD2 promotes calcium oxalate-induced glycolysis in renal tubular epithelial cells via PTEN methylation. Biomedicines. 2024;12(10):2279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Zuo S, Li X, Chen S, Chen M, Peng R, Zou X, et al. Mechanism underlying the role of SMYD2 in mediating high glucose-induced activation of rat renal fibroblasts. China J Mod Med. 2024;34(4):29–38. [Google Scholar]
  • 18. Xu W, Peng R, Chen S, Wu C, Wang X, Yu T, et al. Ranunculus ternatus Thunb extract attenuates renal fibrosis of diabetic nephropathy via inhibiting SMYD2. Pharm Biol. 2022;60(1):300–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Du X, Shen F, Yu C, Wang Y, Yu J, Yao L, et al. SMYD3 as an epigenetic regulator of renal tubular cell survival and regeneration following acute kidney injury in mice. FASEB J. 2025;39(9):e70533. [DOI] [PubMed] [Google Scholar]
  • 20. Brown MA, Sims RJ, Gottlieb PD, Tucker PW. Identification and characterization of Smyd2: a split SET/MYND domain-containing histone H3 lysine 36-specific methyltransferase that interacts with the Sin3 histone deacetylase complex. Mol Cancer. 2006;5:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Brown MA, Foreman K, Harriss J, Das C, Zhu L, Edwards M, et al. C-terminal domain of SMYD3 serves as a unique HSP90-regulated motif in oncogenesis. Oncotarget. 2015;6(6):4005–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Jiang Y, Sirinupong N, Brunzelle J, Yang Z. Crystal structures of histone and p53 methyltransferase SmyD2 reveal a conformational flexibility of the autoinhibitory C-terminal domain. PLoS One. 2011;6(6):e21640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Wang Y, Jin G, Guo Y, Cao Y, Niu S, Fan X, et al. SMYD2 suppresses p53 activity to promote glucose metabolism in cervical cancer. Exp Cell Res. 2021;404(2):112649. [DOI] [PubMed] [Google Scholar]
  • 24. Voelkel T, Andresen C, Unger A, Just S, Rottbauer W, Linke WA. Lysine methyltransferase Smyd2 regulates Hsp90-mediated protection of the sarcomeric titin springs and cardiac function. Biochim Biophys Acta. 2013;1833(4):812–22. [DOI] [PubMed] [Google Scholar]
  • 25. Piao L, Kang D, Suzuki T, Masuda A, Dohmae N, Nakamura Y, et al. The histone methyltransferase SMYD2 methylates PARP1 and promotes poly(ADP-ribosyl)ation activity in cancer cells. Neoplasia. 2014;16(3):257–64.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Komatsu S, Ichikawa D, Hirajima S, Nagata H, Nishimura Y, Kawaguchi T, et al. Overexpression of SMYD2 contributes to malignant outcome in gastric cancer. Br J Cancer. 2015;112(2):357–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Li LX, Zhou JX, Calvet JP, Godwin AK, Jensen RA, Li X. Lysine methyltransferase SMYD2 promotes triple negative breast cancer progression. Cell Death Dis. 2018;9(3):326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Yue M, Liu T, Yan G, Luo X, Wang L. LINC01605, regulated by the EP300-SMYD2 complex, potentiates the binding between METTL3 and SPTBN2 in colorectal cancer. Cancer Cell Int. 2021;21(1):504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Li X, Wang Y, Zhang Y, Liu B. Overexpression of MCAM induced by SMYD2-H3K36me2 in breast cancer stem cell properties. Breast Cancer. 2022;29(5):854–68. [DOI] [PubMed] [Google Scholar]
  • 30. Sajjad A, Novoyatleva T, Vergarajauregui S, Troidl C, Schermuly RT, Tucker HO, et al. Lysine methyltransferase Smyd2 suppresses p53-dependent cardiomyocyte apoptosis. Biochim Biophys Acta. 2014;1843(11):2556–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Foreman KW, Brown M, Park F, Emtage S, Harriss J, Das C, et al. Structural and functional profiling of the human histone methyltransferase SMYD3. PLoS One. 2011;6(7):e22290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Hamamoto R, Furukawa Y, Morita M, Iimura Y, Silva FP, Li M, et al. SMYD3 encodes a histone methyltransferase involved in the proliferation of cancer cells. Nat Cell Biol. 2004;6(8):731–40. [DOI] [PubMed] [Google Scholar]
  • 33. Zhu W, Wang C, Xue L, Liu L, Yang X, Liu Z, et al. The SMYD3-MTHFD1L-formate metabolic regulatory axis mediates mitophagy to inhibit M1 polarization in macrophages. Int Immunopharmacol. 2022;113(Pt A):109352. [DOI] [PubMed] [Google Scholar]
  • 34. Van Aller GS, Reynoird N, Barbash O, Huddleston M, Liu S, Zmoos AF, et al. Smyd3 regulates cancer cell phenotypes and catalyzes histone H4 lysine 5 methylation. Epigenetics. 2012;7(4):340–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Nigam N, Bernard B, Sevilla S, Kim S, Dar MS, Tsai D, et al. SMYD3 represses tumor-intrinsic interferon response in HPV-negative squamous cell carcinoma of the head and neck. Cell Rep. 2023;42(7):112823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Kunizaki M, Hamamoto R, Silva FP, Yamaguchi K, Nagayasu T, Shibuya M, et al. The lysine 831 of vascular endothelial growth factor receptor 1 is a novel target of methylation by SMYD3. Cancer Res. 2007;67(22):10759–65. [DOI] [PubMed] [Google Scholar]
  • 37. Ikram S, Rege A, Negesse MY, Casanova AG, Reynoird N, Green EM. The SMYD3-MAP3K2 signaling axis promotes tumor aggressiveness and metastasis in prostate cancer. Sci Adv. 2023;9(46):eadi5921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Yoshioka Y, Suzuki T, Matsuo Y, Nakakido M, Tsurita G, Simone C, et al. SMYD3-mediated lysine methylation in the PH domain is critical for activation of AKT1. Oncotarget. 2016;7(46):75023–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Yoshioka Y, Suzuki T, Matsuo Y, Tsurita G, Watanabe T, Dohmae N, et al. Protein lysine methyltransferase SMYD3 is involved in tumorigenesis through regulation of HER2 homodimerization. Cancer Med. 2017;6(7):1665–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Wang P, Zhao L, Rui Y, Ding Y. SMYD3 regulates gastric cancer progression and macrophage polarization through EZH2 methylation. Cancer Gene Ther. 2023;30(4):575–81. [DOI] [PubMed] [Google Scholar]
  • 41. Bernard BJ, Nigam N, Burkitt K, Saloura V. SMYD3: a regulator of epigenetic and signaling pathways in cancer. Clin Epigenet. 2021;13(1):45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Fujii T, Tsunesumi S, Yamaguchi K, Watanabe S, Furukawa Y. Smyd3 is required for the development of cardiac and skeletal muscle in zebrafish. PLoS One. 2011;6(8):e23491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Hou X, Yang Y, Wang C, Huang Z, Zhang M, Yang J, et al. H3K36 methyltransferase SMYD2 affects cell proliferation and migration in Hirschsprung’s disease by regulating METTL3. J Cell Physiol. 2024;239(9):e31402. [DOI] [PubMed] [Google Scholar]
  • 44. Pires-Luís AS, Vieira-Coimbra M, Vieira FQ, Costa-Pinheiro P, Silva-Santos R, Dias PC, et al. Expression of histone methyltransferases as novel biomarkers for renal cell tumor diagnosis and prognostication. Epigenetics. 2015;10(11):1033–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Ferreira MJ, Pires-Luís AS, Vieira-Coimbra M, Costa-Pinheiro P, Antunes L, Dias PC, et al. SETDB2 and RIOX2 are differentially expressed among renal cell tumor subtypes, associating with prognosis and metastization. Epigenetics. 2017;12(12):1057–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Brimo F, Epstein JI. Selected common diagnostic problems in urologic pathology: perspectives from a large consult service in genitourinary pathology. Arch Pathol Lab Med. 2012;136(4):360–71. [DOI] [PubMed] [Google Scholar]
  • 47. Aguilar A. Polycystic kidney disease: SMYD2 is a novel epigenetic regulator of cyst growth. Nat Rev Nephrol. 2017;13(9):513. [DOI] [PubMed] [Google Scholar]
  • 48. Xu W, Wu C, Wang X, Jian J, Yang Y, Zhang N, et al. Relationship between histone H3K36 trimethylation and acute renal injury induced by ischemia/reperfusion. Chin J Pathophysiology. 2020;36(3):525–31. [Google Scholar]
  • 49. Grynberg K, Ma FY, Nikolic-Paterson DJ. The JNK signaling pathway in renal fibrosis. Front Physiol. 2017;8:829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Stanigut AM, Tuta L, Pana C, Alexandrescu L, Suceveanu A, Blebea NM, et al. Autophagy and mitophagy in diabetic kidney disease-A literature review. Int J Mol Sci. 2025;26(2):806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Nangaku M, Hirakawa Y, Mimura I, Inagi R, Tanaka T. Epigenetic changes in the acute kidney injury-to-chronic kidney disease transition. Nephron. 2017;137(4):256–9. [DOI] [PubMed] [Google Scholar]
  • 52. Sun YBY, Qu X, Caruana G, Li J. The origin of renal fibroblasts/myofibroblasts and the signals that trigger fibrosis. Differentiation. 2016;92(3):102–7. [DOI] [PubMed] [Google Scholar]
  • 53. Sato Y, Yanagita M. Resident fibroblasts in the kidney: a major driver of fibrosis and inflammation. Inflamm Regen. 2017;37:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Jian J, Zhang N, Yu T, Wang X, Wu C, Xu W, et al. Expression and significance of histone methyltransferase SMYD2 in renal tissue of diabetic nephropathy mice and its expression in renal tissue of diabetic nephropathy mice. J Pract Med. 2020;36(23):3194–8. [Google Scholar]
  • 55. Codato R, Perichon M, Divol A, Fung E, Sotiropoulos A, Bigot A, et al. The SMYD3 methyltransferase promotes myogenesis by activating the myogenin regulatory network. Sci Rep. 2019;9(1):17298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Yang D, Su Z, Wei G, Long F, Zhu YC, Ni T, et al. H3K4 methyltransferase Smyd3 mediates vascular smooth muscle cell proliferation, migration, and neointima formation. Arterioscler Thromb Vasc Biol. 2021;41(6):1901–14. [DOI] [PubMed] [Google Scholar]
  • 57. Chen H, Si T, Jiang Y, Wang D, Zhou Y, Lv J. Relationships between progrosis,immune infiltration and expression of SMYD3 in pan-cancer. Basic Clin Med. 2023;43(1):68–75. [Google Scholar]
  • 58. Xie X, Lin J, Fan X, Zhong Y, Chen Y, Liu K, et al. LncRNA CDKN2B-AS1 stabilized by IGF2BP3 drives the malignancy of renal clear cell carcinoma through epigenetically activating NUF2 transcription. Cell Death Dis. 2021;12(2):201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Ma M, Tian X, Igarashi P, Pazour GJ, Somlo S. Loss of cilia suppresses cyst growth in genetic models of autosomal dominant polycystic kidney disease. Nat Genet. 2013;45(9):1004–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Burtey S, Riera M, Ribe E, Pennenkamp P, Rance R, Luciani J, et al. Centrosome overduplication and mitotic instability in PKD2 transgenic lines. Cell Biol Int. 2008;32(10):1193–8. [DOI] [PubMed] [Google Scholar]
  • 61. Tang J, Liu N, Zhuang S. Role of epidermal growth factor receptor in acute and chronic kidney injury. Kidney Int. 2013;83(5):804–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Liu X, Wang J, Gao L, Liu H, Liu C. iTRAQ-Based proteomic analysis of neonatal kidney from offspring of protein restricted rats reveals abnormalities in intraflagellar transport proteins. Cell Physiol Biochem. 2017;44(1):185–99. [DOI] [PubMed] [Google Scholar]
  • 63. Zhang Y. Preliminary study of the effect of SMYD3 gene in pig fibroblast proliferation and reprogramming. Guangxi University; 2018. [Google Scholar]
  • 64. Bai H, Dong Y, Yue Y, Li Y, Yu H. Effect of overexpression of SET and MYND domain containing protein 3 gene (SMYD3) in bovine (Bos taurus) embryonic fibroblast cells (bEFs) on the cells proliferation and the efficiency for induced pluripotent stem cells (iPSCs). J Agric Biotechnol. 2015;23(10):1282–92. [Google Scholar]
  • 65. Zheng Q, Zhang W, Rao GW. Protein lysine methyltransferase SMYD2: a promising small molecule target for cancer therapy. J Med Chem. 2022;65(15):10119–32. [DOI] [PubMed] [Google Scholar]
  • 66. Eggert E, Hillig RC, Koehr S, Stöckigt D, Weiske J, Barak N, et al. Discovery and characterization of a highly potent and selective aminopyrazoline-based in vivo probe (BAY-598) for the protein lysine methyltransferase SMYD2. J Med Chem. 2016;59(10):4578–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Thomenius MJ, Totman J, Harvey D, Mitchell LH, Riera TV, Cosmopoulos K, et al. Small molecule inhibitors and CRISPR/Cas9 mutagenesis demonstrate that SMYD2 and SMYD3 activity are dispensable for autonomous cancer cell proliferation. PLoS One. 2018;13(6):e0197372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Nguyen H, Allali-Hassani A, Antonysamy S, Chang S, Chen LH, Curtis C, et al. LLY-507, a cell-active, potent, and selective inhibitor of protein-lysine methyltransferase SMYD2. J Biol Chem. 2015;290(22):13641–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Rubio-Tomás T. Novel insights into SMYD2 and SMYD3 inhibitors: from potential anti-tumoural therapy to a variety of new applications. Mol Biol Rep. 2021;48(11):7499–508. [DOI] [PubMed] [Google Scholar]
  • 70. Gradl S, Steuber H, Weiske J, Szewczyk MM, Schmees N, Siegel S, et al. Discovery of the SMYD3 inhibitor BAY-6035 using thermal shift assay (TSA)-Based high-throughput screening. SLAS Discov. 2021;26(8):947–60. [DOI] [PubMed] [Google Scholar]
  • 71. Parenti MD, Naldi M, Manoni E, Fabini E, Cederfelt D, Talibov VO, et al. Discovery of the 4-aminopiperidine-based compound EM127 for the site-specific covalent inhibition of SMYD3. Eur J Med Chem. 2022;243:114683. [DOI] [PubMed] [Google Scholar]
  • 72. Peserico A, Germani A, Sanese P, Barbosa AJ, Di Virgilio V, Fittipaldi R, et al. A SMYD3 small-molecule inhibitor impairing cancer cell growth. J Cell Physiol. 2015;230(10):2447–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Mitchell LH, Boriack-Sjodin PA, Smith S, Thomenius M, Rioux N, Munchhof M, et al. Novel oxindole sulfonamides and sulfamides: EPZ031686, the first orally bioavailable small molecule SMYD3 inhibitor. ACS Med Chem Lett. 2016;7(2):134–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Sanese P, De Marco K, Lepore Signorile M, La Rocca F, Forte G, Latrofa M, et al. The novel SMYD3 inhibitor EM127 impairs DNA repair response to chemotherapy-induced DNA damage and reverses cancer chemoresistance. J Exp Clin Cancer Res. 2024;43(1):151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Huang Y, Tang M, Hu Z, Cai B, Chen G, Jiang L, et al. SMYD3 promotes endometrial cancer through epigenetic regulation of LIG4/XRCC4/XLF complex in non-homologous end joining repair. Oncogenesis. 2024;13(1):3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Padilla A, Manganaro JF, Huesgen L, Roess DA, Brown MA, Crans DC. Targeting epigenetic changes mediated by members of the SMYD family of lysine methyltransferases. Molecules. 2023;28(4):2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Li LX, Zhou JX, Wang X, Zhang H, Harris PC, Calvet JP, et al. Cross-talk between CDK4/6 and SMYD2 regulates gene transcription, tubulin methylation, and ciliogenesis. Sci Adv. 2020;6(44):eabb3154. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Kidney Diseases are provided here courtesy of Karger Publishers

RESOURCES