ABSTRACT
O‐GlcNAcylation is a reversible posttranslational modification of proteins that has garnered significant attention in recent years. By regulating the structure and function of proteins, it plays a critical role in various biological processes. Normal O‐GlcNAcylation is essential for maintaining internal homeostasis and is involved in controlling fundamental biological events such as gene expression, the cell cycle regulation, metabolism, and signal transduction. Conversely, aberrant O‐GlcNAcylation is closely linked to the onset and progression of various diseases—including neurodegenerative diseases, cancers, cardiovascular diseases, and immune‐related diseases—where it drives pathological development. Currently, there is a lack of comprehensive reviews systematically addressing the specific mechanisms of O‐GlcNAcylation under both physiological and pathological conditions. Therefore, this article aims to summarize its dual role in maintaining organismal homeostasis and promoting disease pathogenesis, providing an integrated evaluation of the biological significance of this modification in health and diseases. Furthermore, it discusses the potential of O‐GlcNAcylation as a therapeutic target, explores its clinical applications, and analyzes the current challenges and future directions in drug development, thereby offering theoretical insights and research perspectives for related fields.
Keywords: biological processes, diseases, health, mechanisms, O‐GlcNAcylation, therapy
We first introduced the beneficial effects of normal protein O‐GlcNAcylation on health, impacting biological processes such as gene expression, cell cycle progression, metabolism, and signal transduction. Next, we described the role of abnormal protein O‐GlcNAcylation in the development and progression of diseases, including neurodegenerative diseases, cancers, cardiovascular diseases, and immune‐related diseases. Finally, we explored the feasibility of targeting O‐GlcNAcylation for therapeutic purposes and discussed current challenges and future directions, aiming to provide new insights for drug development and the optimization of clinical treatment strategies.

1. Introduction
O‐GlcNAcylation was first discovered in 1980 during research on cell surface glycan structures [1]. As a dynamic and reversible posttranslational modification, it occurs primarily on serine/threonine (Ser/Thr) residues of proteins and is regulated by only two counteracting enzymes: O‐GlcNAc transferase (OGT) and O‐GlcNAc hydrolase (OGA) [2, 3, 4, 5]. As a glycosyltransferase, OGT catalyzes the covalent attachment of the “GlcNAc” moiety from UDP‐GlcNAc to Ser/Thr residues of target proteins, thereby mediating O‐GlcNAcylation. In contrast, OGA hydrolyzes this glycosidic bond to remove the “GlcNAc” modification, facilitating de‐O‐GlcNAcylation [6, 7]. Both OGT and OGA are evolutionarily highly conserved [8, 9, 10, 11], and their genetic deletion leads to developmental defects or even lethality [12, 13], indicating their indispensable roles in maintaining the dynamic balance of protein O‐GlcNAcylation and ensuring structural and functional stability of proteins. The substrate for O‐GlcNAcylation, UDP‐GlcNAc, is mainly produced through the hexosamine biosynthesis pathway (HBP) [14] (Figure 1). Approximately 2–5% of glucose in the body enters this pathway, where it is metabolized along with glutamine, acetyl coenzyme A (acetyl‐CoA) and uridine 5′‐triphosphate (UTP) to generate UDP‐GlcNAc [15]. Consequently, UDP‐GlcNAc is regarded as an intracellular “nutrient sensor” with its levels reflecting the nutritional and metabolic status of the organism [16]. To date, over 5000 proteins, including membrane, nuclear, cytoplasmic, and mitochondrial proteins, have been identified as O‐GlcNAcylation substrates [17, 18, 19], highlighting the broad and crucial role of this modification in regulating protein properties and functions. However, aberrant O‐GlcNAcylation can significantly alter the physicochemical properties and biological functions of proteins, thereby contributing to the development and progression of various diseases [20]. These findings demonstrate that O‐GlcNAcylation plays a critical role in both health and diseases.
FIGURE 1.

Flow diagram of HBP and GlcNAc cycling. Glucose entering the cell is catalyzed by hexokinase to form glucosamine‐6‐phosphate (GlcN‐6P), which is later isomerized to fructose‐6‐phosphate (Fruc‐6P) by the enzyme GPI. GFAT1 is the rate‐limiting enzyme of HBP. Fruc‐6P binds catalytically to the biomolecules glutamine, glucosamine, and acetyl‐CoA via GFAT1 and GNA1 to sequentially produce glucosamine‐6‐phosphate (GlcN‐6P) and N‐acetyl glucosamine‐6‐phosphate (GlcNAc‐6P). Posterior GlcNAc‐6P isomerization to N‐acetylglucosamine 1‐phosphate (GlcNAc‐1P) catalyzed by PGM3. With the participation of UTP, GlcNAc‐1P is catalyzed by UAP1 to produce the end product of HBP, uridine diphosphate‐N‐acetylglucosamine (UDP‐GlcNAc). Finally, UDP‐GlcNAc was used as a substrate to complete the O‐GlcNAcylation and de‐O‐GlcNAcylation modification of the target proteins by the combined action of the enzymes OGT and OGA. Abbreviations: GPI: glucose‐6‐phosphate isomerase; GFAT1: glutamine‐fructose‐6‐phosphate aminotransferase 1; GNK: GlcNAc kinase; GNA1: d‐glucosamine‐6‐phosphate N‐acetyltransferase 1; PGM3: phosphoglucomutase 3; UAP1: UDP‐N‐acetylhexosamine pyrophosphorylase 1; UTP: uridine triphosphate; UDP: uridine diphosphate; OGT: O‐GlcNActransferase; OGA: O‐GlcNAcase.
Research has shown that knockout of the OGT gene has lethal effects on mouse embryos [21, 22] and accelerates cellular senescence and death [23, 24, 25], indicating that normal O‐GlcNAcylation is essential for embryonic development and cellular homeostasis. The health of an organism relies on the orderly execution of numerous biological processes, and O‐GlcNAcylation is extensively involved in these processes: it promotes mRNA transcription and protein translation; regulates transitions between the cell cycle phases (G1, S, G2, and M); influences the metabolism of substances; and participates in the activation and maintenance of various signaling molecules [26, 27]. Thus, O‐GlcNAcylation plays a central role in key biological processes such as gene expression, the cell cycle, metabolism, and signal transduction, serving as a fundamental mechanism for maintaining overall health.
Abnormal O‐GlcNAcylation has been confirmed as a critical factor in the pathogenesis of various diseases. In neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS), dysregulated expression of OGT and OGA in the brain leads to abnormal O‐GlcNAcylation levels, resulting in functional impairment of neuronal proteins and cellular damage [28]. In various malignant tumors, including lung, liver, colorectal, and breast cancers, elevated O‐GlcNAcylation levels significantly promote tumor cell proliferation, invasion, and metastasis [29]. Furthermore, abnormal O‐GlcNAcylation—either increased or decreased—is commonly observed in cardiovascular diseases (CVDs) [30] and immune‐related diseases [31, 32], accelerating disease progression. These findings indicate that dysregulated O‐GlcNAcylation markedly facilitates disease development.
It is evident that the state of normal and abnormal protein O‐GlcNAcylation is closely linked to health and disease. Although research on O‐GlcNAcylation has grown rapidly, comprehensive summaries remain relatively scarce. Therefore, this article first systematically reviews the important role of O‐GlcNAcylation in maintaining health, with an in‐depth discussion of its regulatory mechanisms in gene expression, the cell cycle, metabolism, and signal transduction. Second, it analyzes how aberrant O‐GlcNAcylation promotes the pathogenesis of neurodegenerative diseases, CVDs, cancers, and immune‐related diseases. Finally, the feasibility of targeting O‐GlcNAcylation as a clinical therapeutic strategy is evaluated, current challenges are discussed, and future research directions are proposed, providing a theoretical basis and constructive insights for the clinical management of related diseases.
2. O‐GlcNAcylation is an Important Maintainer of Health
O‐GlcNAcylation serves as an indispensable “vitamin” in the cellular life cycle, playing an essential role in orchestrating various biological processes and contributing significantly to the maintenance of internal homeostasis [33, 34]. This section will focus on how O‐GlcNAcylation promotes key biological events—such as gene expression, the cell cycle, metabolism, and signal transduction—and will elaborate in detail on its substantial contributions to overall organismal health.
2.1. O‐GlcNAcylation Promotes Gene Expression
O‐GlcNAcylation plays a crucial facilitatory role in the process of gene transcription [35]. Transcription—the synthesis of mRNA using a target gene as a template—is the key initial step in gene expression. The functions of numerous protein components involved in this process are precisely regulated by O‐GlcNAcylation. RNA polymerase II (RNAP II) is the central enzyme in transcription. O‐GlcNAcylation modifies Ser/Thr residues within its C‐terminal domain, which promotes the assembly of the preinitiation complex (PIC) and facilitates transcriptional elongation and termination [36, 37]. The formation of PIC is a critical event in transcription initiation. Studies have shown that inhibiting the enzymatic activity of OGT or OGA impedes PIC assembly, indicating that aberrant O‐GlcNAcylation levels significantly disrupt this process [38, 39]. Beyond RNAP II, proteomic analyses have identified 32 additional transcription‐related factors regulated by O‐GlcNAcylation. Dysregulation of this modification can directly lead to functional impairment of these factors, resulting in transcriptional arrest [37]. Histones, as the core components of chromatin structure, promote chromosome decondensation to form linear DNA templates when gene expression is required. O‐GlcNAcylation targets multiple sites on histone tails and acts as a histone modification itself [40, 41, 42, 43, 44, 45, 46, 47, 48, 49], thereby regulating processes like ubiquitination and methylation. This, in turn, promotes chromatin relaxation and facilitates gene transcription [48, 49]. Furthermore, the functions of many transcription‐related components depend on O‐GlcNAcylation, such as TATA‐binding protein (TBP) [50], topoisomerase I (Topo I) [51], and nucleoporins (NUPs) of the nuclear pore complex (NPC) [52]. In addition to directly modulating transcriptional components to promote gene expression, O‐GlcNAcylation also exerts indirect effects by influencing transcription factors. For example, the activity of HOXA1—a transcription factor belonging to the HOX family—is regulated by OGT, and its dysfunction can lead to transcriptional arrest [53, 54, 55, 56, 57, 58, 59]. The activation and stability of other key transcription factors, including Sp1 [60], Hnf‐1 [61], c‐Myc [62, 63], and serum response factor (SRF) [64], also rely on O‐GlcNAcylation [65]. In summary, O‐GlcNAcylation effectively promotes gene transcription by facilitating the assembly of transcriptional machinery and enhancing the activity and stability of transcription factors.
O‐GlcNAcylation plays a key regulatory role in mRNA splicing. As a central step in gene expression regulation, splicing generates multiple mRNA isoforms to enhance transcript diversity and increase the abundance of target mRNA. Studies show that the function of numerous splicing factors depends on O‐GlcNAcylation, which significantly affects accurate gene expression. Treatment of HEK‐293T cells with the OGT inhibitor OSMI‐2 markedly reduces the mRNA abundance of both OGT and OGA. This phenomenon is consistently observed in HCT116 colon cancer cells and mouse embryonic fibroblasts, indicating a conserved role of O‐GlcNAcylation in splicing regulation [66]. Ser/arginine‐rich protein kinase 2 (SRPK2), a key kinase in pre‐mRNA splicing [67, 68], can be O‐GlcNAcylated at Ser490, Thr492, and Thr498, thereby promoting its nuclear translocation and kinase activation [69]. In MCF7 breast cancer cells, reduced O‐GlcNAcylation decreases the mRNA abundance of genes involved in de novo fatty acid synthesis. Further studies confirm that this process is mediated through SRPK2‐regulated pre‐mRNA splicing, demonstrating that O‐GlcNAcylation influences mRNA maturation [69]. A similar mechanism exists in plants. AtACINUS in Arabidopsis, a homolog of mammalian splicing regulators, has been shown to undergo O‐GlcNAcylation [70]. Experiments indicate that ten AtACINUS‐dependent alternative splicing events strongly rely on O‐GlcNAcylation levels, highlighting its important role in plant splicing regulation. Additionally, O‐GlcNAcylation affects transcription and splicing factor activity by modulating AtACINUS, further regulating gene expression [70]. Notably, O‐GlcNAcylation plays a critical role in splicing regulation associated with various diseases. For example, both TDP‐43 [71] (linked to neurodegenerative diseases) and AUF1 [72] (associated with ovarian cancer) are regulated by O‐GlcNAcylation, and aberrant levels lead to splicing dysregulation and disease pathogenesis. In summary, abnormal O‐GlcNAcylation disrupts the function and modification of splicing factors, resulting in pre‐mRNA splicing dysregulation. In contrast, appropriate and precise O‐GlcNAcylation promotes normal pre‐mRNA splicing, ensures accurate and stable gene expression, and plays an essential protective role in maintaining normal cellular physiological functions.
O‐GlcNAcylation plays a key regulatory role in protein translation—the process by which mRNA, transcribed from genes, serves as a template for protein synthesis with the assistance of ribosomes, tRNA, and associated factors. Multiple studies have confirmed that numerous translation factors and the ribosomes themselves are regulated by O‐GlcNAcylation [73]. To date, more than 20 core ribosomal proteins are known to be modulated by this modification, highlighting its close association with the protein translation machinery [74]. Furthermore, stress granules (SGs) and processing bodies (PBs), which are important components of ribonucleoprotein (RNP) granules, are involved in the regulation of mRNA translation and decay. Research indicates that aberrant O‐GlcNAcylation disrupts the proper assembly of SGs and PBs, thereby suppressing protein translation [75, 76, 77, 78, 79]. Additionally, ribosomal protein subunits such as RPL5, RPL10, RPL11, and RPL13a have been identified as targets of O‐GlcNAcylation. Dysregulation of this modification can significantly impair ribosomal function and hinder protein synthesis [80]. Although further investigation is needed to understand how O‐GlcNAcylation regulates other elements within the translation process, its profound impact on ribosomal function is well established. O‐GlcNAcylation is involved in multiple aspects of ribosomal activity, and as the central machinery for protein synthesis, the proper functioning of ribosomes is critically dependent on precise regulation by O‐GlcNAcylation.
Gene expression, one of the most fundamental biological processes, occurs continuously in countless cells throughout an organism. In summary, O‐GlcNAcylation is involved in nearly all stages of gene expression. Key steps—including transcription, mRNA splicing, protein translation, and the functional execution of resultant proteins—are highly dependent on O‐GlcNAcylation regulation (Figure 2 and Table 1). Abnormal O‐GlcNAcylation levels can disrupt these processes, thereby impeding normal gene expression. Therefore, O‐GlcNAcylation plays an indispensable role in ensuring the smooth progression of gene expression.
FIGURE 2.

O‐GlcNAcylation promotes gene expression. In the process of gene expression, O‐GlcNAcylation plays a critical role, running through multiple stages from transcription to translation. First, transcription, as the initial step of gene expression, depends on the regulation of O‐GlcNAcylation to proceed smoothly. Next, the pre‐mRNA generated by transcription undergoes maturation processes such as splicing, which also requires the involvement of O‐GlcNAcylation. Finally, at the stage of protein translation, O‐GlcNAcylation also plays a facilitating role. Thus, OGlcNAcylation is broadly involved in and closely regulates every step of gene expression.
TABLE 1.
Proper protein O‐GlcNAcylation is critical for health.
| O‐GlcNAcylated proteins | O‐GlcNAcylated sites | Biological events regulated by O‐GlcNAcylation | References |
|---|---|---|---|
| O‐GlcNAcylation promotes gene expression | |||
| RNAP II | / | Promotes the assembly of PIC and facilitates transcriptional elongation and termination | [36, 37] |
| PIC | / | Promotes gene transcription | [38, 39] |
| Histone | Multiple sites | Promotes chromatin relaxation and facilitates gene transcription | [40, 41, 42, 43, 44, 45, 46, 47, 48, 49] |
| TBP | / | Promotes gene transcription | [50] |
| Topo I | / | Promotes gene transcription | [51] |
| NPC | / | Promotes gene transcription | [52] |
| HOXA1 | / | Promotes gene transcription | [53, 54, 55, 56, 57, 58, 59] |
| Sp1 | / | Promotes gene transcription | [60] |
| Hnf‐1 | / | Promotes gene transcription | [61] |
| c‐Myc | / | Promotes gene transcription | [62, 63] |
| SRF | / | Promotes gene transcription | [64] |
| OGT | / | Affects mRNA splicing | [66] |
| OGA | / | Affects mRNA splicing | [66] |
| SRPK2 |
Ser490 Thr492 Thr498 |
Affects mRNA splicing | [69] |
| AtACINUS | / | Affects mRNA splicing | [70] |
| TDP‐43 | / | Affects mRNA splicing | [71] |
| AUF1 | / | Affects mRNA splicing | [72] |
| SGs | / | Promotes protein translation | [75, 76, 77, 78, 79] |
| PBs | / | Promotes protein translation | [75, 76, 77, 78, 79] |
| RPL5 | / | Promotes ribosomal function | [80] |
| RPL10 | / | Promotes ribosomal function | [80] |
| RPL11 | / | Promotes ribosomal function | [80] |
| RPL13a | / | Promotes ribosomal function | [80] |
| O‐GlcNAcylation promotes the cell cycle | |||
| CDKs | / | Promotes the G1‐to‐S transition | [82] |
| Cyclin D1 | / | Inhibits ubiquitin–proteasome degradation | [84] |
| β‐Catenin | / | Promotes G0 to enter G1 phase | [85, 86] |
| DDX3X | Ser584 | Inhibits DDX3X ubiquitin–proteasome degradation, promotes translation of cyclin E, and facilitates the G1/S transition | [87, 88] |
| HCF‐1 | Multiple sites | Promotes G1 and M phase progression | [89] |
| Aurora B | / | Promotes spindle function | [91, 96] |
| PLK1 | / | Promotes spindle function | [91, 96] |
| O‐GlcNAcylation promotes metabolism | |||
| HCF‐1 | Ser333 | Promotes hepatic gluconeogenesis | [99] |
| ERRγ |
Ser317 Ser319 |
Enhances the transcriptional activity of ERRγ and promotes the expression of gluconeogenic genes | [101] |
| FOXO1 | / | Promotes hepatic gluconeogenesis | [102, 103, 104, 105, 106] |
| CRTC2 | / | Promotes hepatic gluconeogenesis | [102, 103, 104, 105, 106] |
| PDX1 | / | Promotes its nuclear localization, enhances its DNA‐binding ability, and facilitates the transcription of insulin‐related genes | [107, 108] |
| NeuroD1 | / | Enhances the activity of the insulin gene promoter | [109, 110, 111] |
| O‐GlcNAcylation promotes signal transduction | |||
| β‐catenin |
Ser23 Thr40 Thr41 Thr112 |
Inhibits ubiquitin–proteasome degradation, promotes the activation of the Wnt signaling pathway, and drives tissue homeostasis regeneration | [121, 122, 123] |
| IR‐β | / | Promotes the activation process | [126] |
| IRS1 | / | Promotes the activation process | [126] |
| AKT | / | Promotes the activation process | [126] |
| PDK1 | / | Promotes the activation process | [126] |
| FoxO1 | / | Promotes the activation process | [126] |
| PI3K | / | Promotes the activation process | [126] |
| NF‐κB | / | Enhances its interaction with downstream target genes | [132, 133] |
| TAB1 | Ser395 | Promotes downstream signal transduction | [129, 134] |
| INFs | / | Promotes cytokine signaling | [135] |
| PD‐L1 | / | Promotes cytokine signaling | [136] |
| TGF‐β | / | Promotes cytokine signaling | [137] |
2.2. O‐GlcNAcylation Promotes the Cell Cycle
The cell cycle, encompassing the period from the completion of one division to the next, is a fundamental biological process central to cell proliferation. It consists of interphase (G1, S, G2) and mitotic phase (M). The progression of the cell cycle is tightly regulated by various molecules, among which cyclin‐dependent kinases (CDKs) play a pivotal role. Their function is highly dependent on the maintenance of proper O‐GlcNAcylation levels [81]. Studies indicate that abnormal O‐GlcNAcylation levels significantly impair the expression stability of cyclins and genomic integrity. Elevated O‐GlcNAcylation leads to cyclin dysregulation and genomic instability, whereas inhibition of this modification disrupts the cell cycle progression—for example, by hindering the synthesis of cyclin E in G1 phase, thereby suppressing the G1‐to‐S transition [82]. It has also been reported that inhibiting OGT impedes the G2‐to‐M transition, though the underlying mechanism remains unclear [83]. Cyclin D1, a regulatory partner of CDK4/6, participates in multiple the cell cycle events and is precisely controlled through various modifications such as phosphorylation and ubiquitination. Elevated O‐GlcNAcylation inhibits its ubiquitination‐mediated degradation, enhances its stability, and thereby promotes the cell cycle progression [84]. During G0 phase, cyclin D is abundantly expressed in a β‐catenin‐dependent transcriptional manner, driving cells from quiescence into G1 phase. The stability of β‐catenin in this process also relies on O‐GlcNAcylation [85, 86]. The C‐terminal extension of RNA helicase DDX3X contains two O‐GlcNAcylation sites (Ser584 and Ser588) [87]. Modification at Ser584 inhibits DDX3X degradation via the ubiquitin–proteasome pathway, enhances its stability, promotes translation of cyclin E, and facilitates the G1/S transition [88]. The N‐ and C‐terminal cleavage products of host cell factor 1 (HCF‐1) promote G1 and M phase progression [89]. HCF‐1 itself possesses over 30 O‐GlcNAcylation sites, and its activation and stability are strongly dependent on this modification [90]. These findings collectively demonstrate that homeostasis of O‐GlcNAcylation is essential for the normal function of numerous the cell cycle‐related proteins.
Furthermore, O‐GlcNAcylation is directly involved in spindle assembly and function during mitosis and meiosis. OGT is closely associated with spindle formation, and the proper assembly of centrosomes and spindles depends on stable protein O‐GlcNAcylation [82, 91, 92, 93]. Abnormal levels of this modification disrupt methylation and acetylation patterns of spindle proteins, leading to defective spindle formation [91, 94]. Excessive O‐GlcNAcylation also causes structural abnormalities: during M phase, overexpression promotes the formation of smaller, disorganized compact chromatid structures [95], inhibits CDK1‐mediated transcriptional activation of phosphatase CDC25c, and impairs phosphorylation of CDK1 downstream substrates, thereby disrupting mitotic spindle assembly [91]. Elevated O‐GlcNAcylation can also alter the phosphorylation of spindle proteins through mitotic kinases Aurora B and Polo‐like kinase 1 (PLK1), ultimately impairing spindle function [91, 96]. Under OGA knockdown conditions, sustained high O‐GlcNAcylation suppresses CDK1 phosphorylation, hinders cyclin B/CDK1 complex formation [97], and causes mislocalization of the spindle component Ewing sarcoma breakpoint region 1 protein, resulting in multipolar spindle structures [98]. Both elevated and reduced O‐GlcNAcylation levels significantly disrupt normal spindle structure and function, indicating that dynamic balance of this modification is crucial for maintaining spindle integrity and ensuring orderly the cell cycle progression.
The cell cycle, as the core process of cell proliferation, is closely related to the growth and development of organisms. Studies have shown that O‐GlcNAcylation is extensively involved in regulating the cell cycle, spanning the G0, G1, S, G2, and M phases (Figure 3). A normal and stable level of O‐GlcNAcylation is crucial for maintaining the function of the cell cycle‐related proteins and catalytic enzymes (Table 1). Furthermore, it plays a key supporting role in centrosome duplication and proper spindle assembly. Conversely, abnormal O‐GlcNAcylation can significantly disrupt the cell cycle, leading to dysregulated phase transitions and disturbances in various cycle‐related molecular events. Therefore, maintaining the dynamic balance of O‐GlcNAcylation is essential for ensuring the orderly progression of the cell cycle.
FIGURE 3.

O‐GlcNAcylation promotes the cell cycle. O‐GlcNAcylation is a crucial regulator of the orderly progression of the cell cycle. First, it directly modulates the progression of various phases of the cell cycle, serving as an essential factor for the proper advancement of interphase (G1, S, G2) and mitosis (M). It also facilitates key transitions such as G0‐to‐G1, G1‐to‐S, and G2‐to‐M. Additionally, by maintaining the structural and functional stability of centrosomes and spindles, it ensures the smooth execution of cell division. Thus, O‐GlcNAcylation is an indispensable regulatory factor for the orderly progression of all phases and stages of the cell cycle.
2.3. O‐GlcNAcylation Promotes Metabolism
O‐GlcNAcylation plays a critical regulatory role in the process of gluconeogenesis, a metabolic pathway through which the body converts noncarbohydrate substrates into glucose or glycogen. This process is essential for maintaining blood glucose homeostasis and energy supply during states such as starvation or stress. Studies have shown that under physiological stress, OGT collaborates with HCF‐1 to sense intracellular glucose availability and catalyze the O‐GlcNAcylation of Ser333 on the transcriptional coactivator PGC‐1α. This modification significantly enhances both the activation and protein stability of PGC‐1α, thereby promoting hepatic gluconeogenesis [99]. Estrogen‐related receptor γ (ERRγ), a key positive regulator of liver gluconeogenesis, is also precisely modulated by O‐GlcNAcylation [100]. As an important substrate of OGT, ERRγ can be O‐GlcNAcylated at Ser317 and Ser319. This modification enhances the transcriptional activity of ERRγ, leading to increased expression of downstream gluconeogenic genes and further driving hepatic glucose production [101]. Additionally, O‐GlcNAcylation can upregulate the expression of gluconeogenic genes by modifying transcription factors such as FOXO1 and CRTC2, thereby promoting hepatic gluconeogenesis through multiple pathways [102, 103, 104, 105, 106]. Thus, in response to metabolic demand for gluconeogenesis, O‐GlcNAcylation stabilizes and activates key regulatory proteins through various mechanisms, enhances the transcriptional expression of gluconeogenesis‐related genes, and ultimately facilitates the conversion of noncarbohydrate precursors into glucose, providing essential energy support for the organism.
O‐GlcNAcylation, as a key glycosylation modification, plays a central regulatory role in insulin synthesis and secretion. Its modification level is directly influenced by glucose metabolism, thereby participating in the critical mechanisms regulating insulin secretion. Studies have shown that inhibiting GFAT, the rate‐limiting enzyme of HBP, leads to a significant decrease in O‐GlcNAcylation levels and severely suppresses insulin secretion, fully demonstrating the indispensability of this modification in insulin secretion. At the molecular level, O‐GlcNAcylation functions by modifying multiple key transcription factors. For example, O‐GlcNAcylation of the pancreatic transcription factor PDX1 promotes its nuclear localization, enhances its DNA‐binding ability, and thereby facilitates the transcription of insulin‐related genes [107, 108]. In pancreatic‐derived MIN6 cells, high glucose conditions significantly increase the O‐GlcNAcylation level of PDX1, correspondingly enhancing insulin gene expression and secretion [108]. Similarly, the transcription factor NeuroD1 is also modified by O‐GlcNAcylation. Modified NeuroD1 markedly enhances the activity of the insulin gene promoter [109, 110, 111]. Under high glucose conditions, O‐GlcNAcylated NeuroD1 tends to localize in the nucleus with improved promoter‐binding capacity, effectively promoting insulin gene transcription and protein synthesis [110, 111]. Thus, O‐GlcNAcylation modifies key transcription factors such as PDX1 and NeuroD1, facilitating their nuclear translocation, strengthening their DNA or promoter binding abilities, and synergistically upregulating the transcription of insulin‐related genes, ultimately promoting insulin biosynthesis and secretion.
Cellular metabolism serves as the central hub for energy supply in organisms, directly maintaining energy homeostasis and metabolic stability. Among these processes, O‐GlcNAcylation, as an important posttranslational modification, is closely linked to glucose metabolism. Research has demonstrated that under conditions of hunger or stress, O‐GlcNAcylation enhances insulin secretion and gluconeogenesis, thereby maintaining blood glucose levels and ensuring a continuous energy supply for normal physiological activities. This highlights its crucial role in sustaining health (Figure 4 and Table 1). However, current research on the functions of O‐GlcNAcylation in metabolic pathways beyond glucose metabolism—such as lipid and amino acid metabolism—remains limited. Its regulatory mechanisms and physiological significance require further in‐depth exploration.
FIGURE 4.

O‐GlcNAcylation promotes metabolism. Under extreme hunger or stress, the body's protein O‐GlcNAcylation levels rise significantly, thereby enhancing metabolic processes. First, O‐GlcNAcylation upregulates the expression of gluconeogenesis‐related genes in liver cells, promoting gluconeogenesis and increasing blood glucose levels. Second, OGlcNAcylation stimulates pancreatic β‐cells to synthesize and release insulin, which facilitates glucose uptake and utilization by body cells, thereby lowering blood glucose. Thus, O‐GlcNAcylation serves as a crucial mechanism for energy security under abnormal physiological conditions, maintaining internal homeostasis and overall health.
2.4. O‐GlcNAcylation Promotes Signal Transduction
Cells sense and respond to changing external conditions by detecting physicochemical cues from their environment. This process, known as signal transduction, involves converting extracellular signals into intracellular responses [112, 113]. As a key posttranslational modification, O‐GlcNAcylation regulates the activity and stability of numerous signaling molecules. Through this modification, it facilitates the activation and maintenance of downstream signaling pathways.
O‐GlcNAcylation plays a crucial role in regulating G protein‐coupled receptors (GPCRs) signal transduction. As the largest family of signaling receptors in the human body, GPCRs are widely involved in various biological processes, enabling cells to respond to external stimuli [114, 115]. Among them, the Wnt signaling pathway is a typical GPCR‐mediated pathway that participates in key biological processes such as tissue homeostasis and regeneration, stem cell proliferation, and differentiation [116, 117]. This pathway is precisely regulated by O‐GlcNAcylation [118]. Within the Wnt pathway, β‐catenin serves as a core transcription factor. Under resting cellular conditions, β‐catenin is inactivated through continuous phosphorylation and degraded via ubiquitination, preventing it from performing transcriptional functions [119, 120, 121]. However, during active cell proliferation, OGT catalyzes the O‐GlcNAcylation of multiple amino acid residues on β‐catenin, including Ser23, Thr40, Thr41, and Thr112 [122]. This modification further promotes the de‐phosphorylation and de‐ubiquitination of β‐catenin, significantly enhancing its activation level and protein stability. Activated β‐catenin initiates downstream signaling cascades, promotes the activation of the Wnt signaling pathway, and effectively drives tissue homeostasis regeneration and other related biological processes [121, 122, 123]. Thus, by positively regulating the stability and activity of β‐catenin, O‐GlcNAcylation plays a key role in GPCR/Wnt signal transduction.
O‐GlcNAcylation plays a critical role in promoting receptor tyrosine kinases (RTKs) signal transduction. An important class within the RTKs protein family is growth factor receptors, which regulate multiple signaling pathways, such as the protein kinase B (AKT) pathway that controls cell growth and proliferation [124], and the insulin signaling pathway that regulates metabolism and energy storage [125]. Notably, in both the AKT and insulin pathways, several key signaling molecules—including IR‐β, IRS1, AKT, pyruvate dehydrogenase kinase (PDK)1, FoxO1, and PI3K—are direct targets of OGT. Their activation or inactivation processes rely on O‐GlcNAcylation [126]. Furthermore, O‐GlcNAcylation also modulates the MAPK signaling pathway, a classic downstream pathway of RTKs that regulates cell proliferation, differentiation, and apoptosis [127, 128]. Although there are currently few reports of OGT directly modifying MAPK components, existing evidence suggests that O‐GlcNAcylation can enhance interactions between upstream and downstream signaling molecules in the MAPK pathway through crosstalk with phosphorylation modifications, thereby effectively promoting the activation and propagation of this signaling pathway [129].
O‐GlcNAcylation plays a crucial regulatory role in cytokine signal transduction. Various cytokines, such as tumor necrosis factor α (TNFα) and interleukins (ILs), are primarily involved in regulating inflammatory and immune cell processes [130], and O‐GlcNAcylation serves as a key modification in the regulation of these cellular functions [131]. For instance, O‐GlcNAcylation significantly influences biological processes such as cell proliferation and apoptosis by activating nuclear factor κB (NF‐κB) in response to external stimuli [132]. NF‐κB typically exists as a dimer composed of p65 and p50 subunits, with the activity and function of p65 being highly regulated by O‐GlcNAcylation. This modification promotes the nuclear translocation of NF‐κB and enhances its interaction with downstream target genes, thereby effectively initiating related signaling pathways [133]. Additionally, TAB1, an upstream regulatory protein of NF‐κB, is also modulated by O‐GlcNAcylation. Liquid chromatography–tandem mass spectrometry analysis has identified Ser395 of TAB1 as a key site for O‐GlcNAcylation. Mutation at this site significantly inhibits TAB1 activation, and further studies confirm that in ILs signaling pathways, O‐GlcNAcylation actively promotes downstream signal transduction by enhancing TAB1 activity [129, 134]. Beyond TNFα and ILs, the activation processes of other cytokines, such as interferons [135], programmed death‐ligand 1 (PD‐L1) [136], and transforming growth factor β (TGF‐β) [137], also widely depend on O‐GlcNAcylation. These findings collectively demonstrate that O‐GlcNAcylation plays a key positive regulatory role in multiple cytokines signaling pathways, significantly facilitating the smooth progression of inflammatory and immune responses.
In summary, classical signaling pathways such as GPCR, RTK, and cytokine pathways form critical regulatory networks that ensure the proper execution of various biological processes. Dysregulation of these pathways can lead to disruptions in multiple biological functions. Notably, key molecules within these pathways—including receptors, kinases, and transcription factors—are highly dependent on O‐GlcNAcylation to maintain their stability and activation (Table 1). Therefore, during cellular responses to external stimuli, O‐GlcNAcylation not only significantly influences the activation and deactivation of related signaling pathways through precise regulation of signaling molecules but also provides an indispensable molecular foundation for efficient and orderly signal transduction (Figure 5).
FIGURE 5.

O‐GlcNAcylation promotes signal transduction. O‐GlcNAcylation is widely involved in regulating various cellular signal transduction, including GPCRs signaling, RTKs signaling, and cytokine receptor signaling. Specifically, it modifies upstream and downstream signaling molecules and enhances their activation, thereby facilitating the efficient transmission of biological information within the pathways. Thus, this modification plays a crucial role in maintaining the openness and proper functioning of signaling pathways.
3. Abnormal O‐GlcNAcylation Contributes to Disease Pathogenesis
Normal O‐GlcNAcylation plays a crucial role in maintaining overall health, actively regulating various biological processes such as gene expression, the cell cycle, metabolism, and signal transduction. It serves as an essential molecular foundation for sustaining internal homeostasis. However, in the development and progression of numerous diseases, dysregulation of O‐GlcNAcylation often serves as a key driver of pathogenesis. This involves various pathological conditions, including neurodegenerative diseases, CVDs, cancers, immune‐related diseases. Therefore, this section will systematically elaborate on the specific roles of O‐GlcNAcylation abnormalities in these diseases and provide an in‐depth analysis of the associated biological processes and molecular mechanisms driving disease development.
3.1. Abnormal O‐GlcNAcylation Contributes to the Pathogenesis of Neurodegenerative Diseases
Neurodegenerative diseases are a group of progressive neurological disorders characterized primarily by cognitive and physical dysfunction, with the core pathological mechanism lying in the gradual loss of neuronal structure and function. Neurons, as the fundamental units of the nervous system, rely heavily on the precise regulation of various posttranslational modifications to maintain normal function. Among these, O‐GlcNAcylation—a key dynamic modification—is highly abundant in the brain, particularly at synaptic sites. In functional synapses, both OGT and OGA are actively expressed and work together to maintain the dynamic balance of O‐GlcNAcylation at nerve terminals [138]. Numerous proteins closely associated with neuronal structure and function, such as CaMKIV, GluA2, synapsin I, synaptopodin, piccolo, bassoon, cAMP response element‐binding protein, and calcium/calmodulin‐dependent protein kinase II (CaMKII), are precisely regulated by O‐GlcNAcylation [139, 140, 141, 142, 143, 144]. Therefore, O‐GlcNAcylation homeostasis is essential for maintaining normal neuronal physiological function. Disruption of this modification can lead to structural and functional abnormalities in neurons, thereby contributing to the onset and progression of diseases. In recent years, growing evidence has shown that aberrant O‐GlcNAcylation plays a significant role in triggering and promoting various neurodegenerative diseases, including PD, HD, AD, and ALS. The following content will elaborate on the specific mechanisms through which abnormal O‐GlcNAcylation contributes to neurodegenerative diseases.
Abnormal O‐GlcNAcylation levels are closely associated with the onset and progression of AD. As one of the most common neurodegenerative diseases, AD is clinically characterized by memory impairment, mood fluctuations, and cognitive decline [145]. Multiple studies have shown that O‐GlcNAcylation levels in the brain tissues of AD patients are 22–50% lower than those in healthy individuals, suggesting that reduced O‐GlcNAcylation may contribute to AD pathology [146, 147, 148, 149, 150]. At the molecular level, aberrant O‐GlcNAcylation promotes AD progression primarily by affecting the metabolism of Tau protein and β‐amyloid (Aβ). Tau is a microtubule‐associated protein critical for maintaining neuronal cytoskeletal stability. Its activity is regulated by phosphorylation and it contains multiple O‐GlcNAcylation sites, such as Thr123, Ser208, Ser400, Ser409, Ser412, and Ser413 [151]. In AD, Tau undergoes abnormal hyperphosphorylation and aggregates into neurofibrillary tangles (NFTs), which is one of the core pathological features [152, 153]. Cellular and animal studies have demonstrated that elevating O‐GlcNAcylation levels with the OGA inhibitor thiamet‐G significantly suppresses phosphorylation of Tau at sites including Ser396, Thr231, Ser202, and Thr205, thereby inhibiting its hyperphosphorylation and aggregation [154, 155]. This indicates that O‐GlcNAcylation can counteract tauopathy by antagonizing Tau phosphorylation, and its reduction may facilitate AD progression. In addition, Aβ is a peptide fragment produced through sequential cleavage of the conserved type I transmembrane protein APP by α [156], β [157], and γ‐secretase complexes [158, 159], and it accumulates abnormally in the brains of AD patients [160]. Studies have confirmed that O‐GlcNAcylation also regulates Aβ production [161]. In AD model mice overexpressing Aβ, treatment with thiamet‐G significantly reduces Aβ levels [162]. Further mechanistic research reveals that O‐GlcNAcylation modifies the Ser708 residue of the γ‐secretase subunit nicastrin (NCT), thereby inhibiting γ‐secretase activity and reducing Aβ generation [163]. This again suggests that elevated O‐GlcNAcylation suppresses Aβ‐related pathology, while its downregulation promotes amyloid disease. Beyond regulating Tau and Aβ, O‐GlcNAcylation is involved in AD progression through other biological processes such as neuroinflammation and insulin signaling. For example, loss of OGT promotes astrocyte activation and inflammatory responses, leading to cognitive impairment in mice; restoring O‐GlcNAcylation alleviates inflammation and improves cognitive function. Mechanistically, OGT catalyzes O‐GlcNAcylation at Ser384 of the NF‐κB p65 subunit in glial cells, inhibiting NF‐κB signaling and exerting anti‐inflammatory effects. Conversely, its deficiency promotes NF‐κB activation and enhances neuroinflammation [164]. In AD, decreased O‐GlcNAcylation leads to overactivation of the NF‐κB pathway, exacerbating neuroinflammation and accelerating cognitive decline. Additionally, cerebral insulin resistance is another key factor in AD development [165, 166]. Insulin receptor substrate‐1 (IRS‐1) is a critical molecule in the insulin signaling pathway, and phosphorylation at its Ser307 site is closely linked to insulin resistance. Multiple Ser/Thr residues of IRS‐1 can also undergo O‐GlcNAcylation [167, 168]. During AD progression, the decline in O‐GlcNAcylation occurs earlier than IRS‐1 phosphorylation. Since these two modifications may compete for the same or adjacent sites, reduced O‐GlcNAcylation may lift the inhibition on phosphorylation, thereby promoting IRS‐1 phosphorylation, insulin resistance, and further AD development [168]. In summary, abnormally low O‐GlcNAcylation levels significantly promote the initiation and progression of AD by regulating key events including Tau phosphorylation, Aβ production, neuroinflammatory responses, and insulin signaling. Restoring O‐GlcNAcylation levels may represent a potential therapeutic strategy for AD intervention.
PD is the second most common neurodegenerative disease after AD. Its primary clinical features include bradykinesia, rigidity, tremor, and neuropsychiatric symptoms [169]. The characteristic neuropathological hallmarks of PD include the loss of dopaminergic neurons in the substantia nigra and the formation of Lewy bodies composed of abnormally aggregated α‐synuclein [170]. Recent studies have shown that decreased levels of O‐GlcNAcylation are closely associated with these pathological processes [169, 171, 172, 173]. In the development of PD, abnormal phosphorylation of α‐synuclein at Ser87 and Ser129 promotes its aggregation [172] and widespread distribution in presynaptic terminals and synaptic vesicles, interfering with critical functions such as vesicle stability, transport, and neurotransmitter release [174, 175]. α‐synuclein can be modified by O‐GlcNAcylation [172, 173]: cellular experiments have confirmed that O‐GlcNAcylation at Thr72, Thr75, and Thr81 inhibits phosphorylation at Ser87 and Ser129, thereby reducing α‐synuclein aggregation and its neurotoxicity and slowing PD progression [176, 177, 178]. Beyond its regulatory role on α‐synuclein, reduced O‐GlcNAcylation levels also directly impair dopaminergic neuron function [171]. Studies have shown that specific knockout of OGT in dopaminergic neurons leads to neuronal loss, whereas increasing O‐GlcNAcylation supports normal neuronal function and enhances synaptic transmission [179]. In PD mouse models, elevating O‐GlcNAcylation levels by inhibiting OGA significantly alleviates pathological damage in dopaminergic neurons, improves dopamine release deficits and motor dysfunction, and reduces α‐synuclein aggregation [179]. In summary, upregulation of O‐GlcNAcylation is neuroprotective event, while its reduction promotes key pathological processes such as abnormal α‐synuclein aggregation and dopaminergic neuron loss, thereby exacerbating the onset and progression of PD. Therefore, targeting O‐GlcNAcylation may represent a potential therapeutic strategy for PD.
Abnormal O‐GlcNAcylation plays a significant role in the onset and progression of HD. HD is a rare neurodegenerative disease characterized primarily by cognitive and motor dysfunction [180, 181]. Its pathogenesis stems from mutations in the huntingtin gene HTT, leading to misfolding of the encoded huntingtin protein (HTT), which gradually forms oligomers, polymers, and pathogenic mutant HTT fibrils (mHTT fibrils). These abnormal protein aggregates further disrupt key biological processes such as neuronal synaptic transmission, mitochondrial function, and gene transcription, ultimately causing the disease [182, 183]. The structure and function of the HTT protein and its interacting protein HIP1R are regulated by O‐GlcNAcylation [171, 184, 185]. Studies have shown that coexpression of mutant HTT and OGA in mouse neural cells results in reduced O‐GlcNAcylation levels, accompanied by a significant decrease in misfolded HTT proteins and a marked reduction in cell death caused by abnormal mHTT aggregation [185]. This clearly indicates that O‐GlcNAcylation levels are negatively correlated with HD progression, and its elevated expression promotes HD development. Besides, disrupted nucleocytoplasmic transport and distorted nuclear membranes are well‐established pathological features in HD model mice. The NPC, a critical channel for nucleocytoplasmic transport, relies on the proper function of NUPs, whose structural stability and selective permeability are highly dependent on O‐GlcNAcylation [186, 187]. Experiments revealed that treating HD mice with OGA inhibitors increased O‐GlcNAcylation levels and significantly restored nucleocytoplasmic transport function, suggesting that insufficient O‐GlcNAcylation promotes HD pathological progression by impairing NUP function and nucleocytoplasmic transport [186]. In summary, the role of O‐GlcNAcylation in HD is complex and context‐dependent. On one hand, elevated O‐GlcNAcylation promotes the abnormal aggregation of HTT proteins. On the other hand, reduced O‐GlcNAcylation impairs the function of the NPC. Both abnormalities significantly accelerate HD progression, indicating that an imbalance in O‐GlcNAcylation levels—whether increased or decreased—plays a critical role in the development and progression of HD.
Abnormal O‐GlcNAcylation levels play a significant role in the pathological progression of ALS. ALS is a neurodegenerative disease characterized by dysfunction of both upper and lower motor neurons, with clinical manifestations including motor impairment, dysarthria, dysphagia, and behavioral or cognitive abnormalities [188, 189, 190, 191, 192]. One of the hallmark pathological features of ALS is the formation of axonal spheroids composed of neurofilament (NF) proteins [193]. NF proteins are coregulated by O‐GlcNAcylation and phosphorylation, which exhibit crosstalk [194, 195, 196]. In ALS, NF proteins become hyperphosphorylated and form abnormal aggregates [197, 198], while O‐GlcNAcylation levels are significantly reduced in ALS mouse models [199]. This suggests that diminished O‐GlcNAcylation allows phosphorylation to dominate, promoting NF aggregation and accelerating disease progression. Additionally, TDP‐43, a critical RNA/DNA‐binding protein involved in transcriptional regulation and stability maintenance, forms toxic aggregates that contribute to ALS pathogenesis [200, 201, 202]. O‐GlcNAcylation occurs at Thr199 and Thr233 residues of TDP‐43. Studies show that co‐overexpression of OGT and TDP‐43 in SH‐SY5Y neuroblastoma cells effectively suppresses TDP‐43 aggregation and cytotoxicity, indicating a protective role of O‐GlcNAcylation [203]. Reduced O‐GlcNAcylation promotes TDP‐43 aggregation and facilitates ALS development. Moreover, elevated oxidative stress is another key mechanism driving motor neuron degeneration. Nonselenocysteine‐containing phospholipid hydroperoxide glutathione peroxidase (NPGPx), an oxidative stress sensor, is closely associated with ALS. NPGPx knockout mice exhibit ALS‐like phenotypes, including reactive oxygen species (ROS) accumulation and motor neuron death, accompanied by a global reduction in O‐GlcNAcylation. Treatment with an OGA inhibitor in ALS mice significantly alleviates motor neuron death and ROS accumulation, indicating that decreased O‐GlcNAcylation impairs NPGPx function and exacerbates oxidative stress and neuronal death [204]. In summary, normal O‐GlcNAcylation levels may have a protective effect in ALS, whereas its downregulation promotes multiple key pathological processes—including NF assembly, TDP‐43 aggregation, and NPGPx‐mediated oxidative stress—thereby driving ALS progression. Therefore, elevating O‐GlcNAcylation levels may represent a novel therapeutic strategy for ALS.
Abnormal O‐GlcNAcylation levels have been widely recognized as a key molecular hallmark of neurodegenerative diseases. In various types of neurodegenerative diseases, dysregulated O‐GlcNAcylation significantly contributes to disease initiation and progression by impairing the function of critical proteins (Table 2). In AD, abnormally low O‐GlcNAcylation of Tau and Aβ leads to their dysfunction, representing a major pathogenic mechanism. Similarly, in PD, reduced O‐GlcNAcylation not only disrupts normal dopaminergic neuronal function but also exacerbates the abnormal aggregation and pathology of α‐synuclein. In ALS, diminished O‐GlcNAcylation of proteins such as NF, TDP‐43, and NPGPx is closely associated with disease development, resulting in decreased protein stability, increased aggregation propensity, and accelerated neurodegeneration. HD presents a slightly different scenario: hyper‐O‐GlcNAcylation of mHTT protein combined with hypo‐O‐GlcNAcylation of NUP constitutes a critical biological event driving pathological progression. Thus, abnormally low O‐GlcNAcylation plays a central role in the progression of most neurodegenerative diseases, while in certain disorders like HD, abnormally high O‐GlcNAcylation of specific proteins also serves as a non‐negligible pathogenic factor. The balance of this modification is essential for maintaining neuronal homeostasis, and its dysregulation has emerged as a common mechanism underlying neurodegenerative diseases.
TABLE 2.
Dysregulation of protein O‐GlcNAcylation promotes disease progression.
| O‐GlcNAcylated proteins | Changes in O‐GlcNAcylation levels | O‐GlcNAcylated sites | Biological events regulated by O‐GlcNAc | References |
|---|---|---|---|---|
| Abnormal O‐GlcNAcylation contributes to the pathogenesis of neurodegenerative diseases | ||||
| Tau | Downregulation |
Thr123 Ser208 Ser400 Ser409 Ser412 Ser413 |
Promotes abnormal aggregation of Tau protein to form NFTs and facilitates AD progression | [151] |
| Aβ | Downregulation | / | Promotes abnormal aggregation of Aβ and accelerates AD progression | [161] |
| NCT | Downregulation | Ser708 | Promotes abnormal aggregation of Aβ and accelerates AD progression | [163] |
| p65 | Downregulation | Ser384 | Promotes astrocyte activation and inflammatory responses | [164] |
| IRS‐1 | Downregulation | / | Promotes IRS‐1 phosphorylation, insulin resistance, and further AD development | [167, 168] |
| α‐Synuclein | Downregulation |
Thr72 Thr75 Thr81 |
Promotes the aggregation and neurotoxicity of α‐synuclein, thereby contributing to the progression of PD | [176, 177, 178] |
| HTT | Upregulation | / | Promotes the abnormal aggregation of HTT | [171, 184, 185] |
| HIP1R | Upregulation | / | Promotes the abnormal aggregation of HTT | [171, 184, 185] |
| NUPs | Downregulation | / | Impairs NUP function and nucleocytoplasmic transport | [186, 187] |
| NF | Downregulation | / | Promotes the phosphorylation and abnormal aggregation of the NF protein | [199] |
| TDP‐43 | Downregulation | Thr199 Thr233 | Promotes TDP‐43 protein abnormal aggregation and cytotoxicity | [203] |
| NPGPx | Downregulation | / | Impairs NPGPx function and exacerbates oxidative stress and neuronal death | [204] |
| Abnormal O‐GlcNAcylation drives cancer progression | ||||
| RBM14 | Upregulation | Ser521 | Enhances RBM14 transcriptional activity and promotes the proliferation and metastasis of lung cancer cells | [211, 212, 213] |
| NRF2 | Upregulation | Ser103 | Promotes the stabilization and activation of NRF2 | [214] |
| c‐Myc | Upregulation | / | Promotes tumor invasion and metastasis | [217] |
| SAM68 | Upregulation | / | Promotes tumor invasion and metastasis | [218] |
| YEATS2 | Upregulation | / | Promotes tumor invasion and metastasis | [219] |
| PRPS1 | Upregulation | Ser83 Thr166 | Promotes nucleotide synthesis, supporting rapid proliferation, tumor growth, and drug resistance in lung cancer cells | [221] |
| SMAD4 | Upregulation | / | Activates the TGF‐β signaling pathway to induce epithelial–mesenchymal transition | [226] |
| p‐JNK | Upregulation | / | Facilitates JNK/c‐Jun/AP‐1 signal transduction, exacerbate endoplasmic reticulum stress | [231, 232] |
| p‐c‐Jun | Upregulation | / | Facilitates JNK/c‐Jun/AP‐1 signal transduction, exacerbating endoplasmic reticulum stress | [231, 232] |
| AP‐1 | Upregulation | / | Facilitates JNK/c‐Jun/AP‐1 signal transduction, exacerbating endoplasmic reticulum stress | [231, 232] |
| p‐IKKa/IKKb | Upregulation | / | Enhances the DNA‐binding activity of NF‐κB, promoting palmitic acid metabolism | [230] |
| p‐p65 | Upregulation | / | Enhances the DNA‐binding activity of NF‐κB, promoting palmitic acid metabolism | [230] |
| p‐p50 | Upregulation | / | Enhances the DNA‐binding activity of NF‐κB, promoting palmitic acid metabolism | [230] |
| RAF1 | Upregulation | / | Inhibits ubiquitination‐mediated degradation of RAF1. Facilitates liver cancer proliferation and EMT | [234, 235] |
| YAP | Upregulation | Thr241 | Promotes the stabilization and activation of YAP; hepatocarcnogenesis. | [238, 239] |
| TS | Upregulation |
Thr251 Thr306 |
Inhibits TS proteasomal degradation; Inhibits TS proteasomal degradation | [243] |
| YY1 | Upregulation | Thr236 | Activates the expression of oncogenes SLC22A15 and AANAT; facilitates tumorigenesis | [245] |
| YTHDF1 | Upregulation |
Ser196 Ser197 Ser198 |
Promotes c‐Myc oncogene expression and tumor cell proliferation | [246, 247, 248] |
| CSNK2A1 | Upregulation | / | Upregulates the expression of oncogene CSNK2A1 | [249] |
| c‐Myc | Upregulation | Ser415 | Promotes the Warburg effect and tumor cell proliferation | [250, 251] |
| FASN | Upregulation | / | Accelerates tumor growth and metastasis | [254] |
| ITGA5 | Upregulation | / | Promotes tumor cell adhesion, invasion, and EMT | [255, 256, 257, 258, 259] |
| EZH2 | Upregulation | / | Promotes tumor cell metastasis | [260] |
| MORC2 | Upregulation | Thr556 | Promotes the expression of TGF‐β1 downstream target oncogenes CTGF and SNAIL. Promotes the invasion and metastasis of tumor cells | [263] |
| Histone H2A | Upregulation | Ser40 | Inhibits the histone demethylase KDM5B. promotes tumor proliferation and metastasis | [265] |
| FOXM1 | Upregulation | / | Promotes the stabilization and activation of FOXM1; aggravates the malignant behavior of tumor cells | [266, 267] |
| p120 | Upregulation | / | Promotes tumor cell dissociation and distant metastasis | [268] |
| β‐Catenin | Upregulation | / | Promotes tumor cell dissociation and distant metastasis | [268] |
| Cofilin | Upregulation | Ser108 | Promotes tumor cell motility and invasion | [269, 270, 271] |
| MEK2 | Upregulation | Thr13 | Promotes the proliferation and metastasis of breast cancer cells by stabilizing and activating MEK2 | [272] |
| GATAD2B | Upregulation | / | Inhibits ubiquitination and degradation of GATAD2B; enhances CSC self‐renewal and metastatic drug resistance | [273, 274, 275, 276] |
| KLF8 | Upregulation | / | Promotes CSC phenotypes and chemotherapy resistance | [277, 278] |
| Abnormal O‐GlcNAcylation is implicated in the pathogenesis of CVDs | ||||
| c‐Myc | Upregulation | / | Promotes pathological cardiac remodeling | [306, 307, 308, 309] |
| Troponin I | Upregulation | / | Promotes pathological cardiac remodeling | [306, 307, 308, 309] |
| Troponin T | Upregulation | / | Promotes pathological cardiac remodeling | [306, 307, 308, 309] |
| PKAc | Upregulation | / | Promotes myocardial hypertrophy and dysfunction | [313, 314] |
| Abnormal O‐GlcNAcylation promotes the pathogenesis of immune‐related diseases | ||||
| FOXP3 | Downregulation | / | Inhibits Treg cell infiltration and promotes AIH pathology | [319] |
| NF‐κB | Upregulation | / | Promotes the production of Th17 cells and related inflammatory responses, advancing the MS pathological process | [322, 323, 324] |
| p65 | Upregulation | / | Exacerbates RA progression | [327, 328] |
| ACC1 | Upregulation | / | Enhances the activation of transcription factor RORγt; promotes the proinflammatory effects of Th17 cells; promotes the pathological process of RA | [330, 331] |
| NFAT | Upregulation | / | Promotes the activation of T cells and B cells | [338, 339, 342] |
| c‐Myc | Upregulation | / | Promotes T cell activation | [338, 339, 342] |
| NF‐κB | Upregulation | / | Promote the activation of T cells and B cells | [338, 339, 342] |
| RelA | Upregulation | Thr305 | Promotes the release of inflammatory factors | [342, 350] |
| c‐Rel | Upregulation | Ser350 | Promotes the release of inflammatory factors | [342, 350] |
| STAT3 | Upregulation | Thr717 | Inhibits IL‐10 function and promote inflammatory response | [351, 352, 353] |
| EZH2 | Upregulation | Ser75 | Promotes NK cell activation and stabilization | [354, 355, 356] |
| FOXO1 | Upregulation | / | Promotes the cytotoxic effect of NK cells | [357, 358] |
3.2. Abnormal O‐GlcNAcylation Drives Cancer Progression
In recent years, the incidence and mortality rates of cancers have continued to rise, making them a major threat to human health. The development and progression of cancers involve dysregulation of multiple biological processes, including gene mutations, metabolic disorders, and immune evasion, resulting in complex and diverse pathological mechanisms. O‐GlcNAcylation, as an important posttranslational modification of proteins, plays a widespread role in regulating key biological activities such as cellular signal transduction, gene expression, energy metabolism, and immune surveillance. Dysregulated O‐GlcNAcylation is closely associated with the occurrence of various diseases. A growing body of research indicates that O‐GlcNAcylation levels are significantly elevated in various highly prevalent and lethal malignant tumors, including lung cancer, liver cancer, colorectal cancer (CRC), and breast cancer, suggesting its potential critical role in tumorigenesis and progression. However, the molecular mechanisms by which O‐GlcNAcylation promotes the progression of these tumors have not yet been fully elucidated. This section aims to review and discuss recent advances in the regulatory mechanisms of O‐GlcNAcylation in common cancers, and to providing a theoretical basis for related targeted therapeutic strategies.
Lung cancer is one of the most frequently diagnosed malignancies worldwide and a leading cause of cancer‐related mortality [205]. Its development and progression are closely associated with various molecular abnormalities. In recent years, growing evidence has shown that aberrant O‐GlcNAcylation plays a key role in lung cancer progression by influencing various biological processes such as gene expression, signal transduction, and metabolic reprogramming, thereby enhancing tumor proliferation, invasion, metastasis, and therapy resistance [206, 207, 208, 209, 210]. O‐GlcNAcylation significantly enhances oncogenic transcriptional activity by regulating key transcription factors and coactivators. For instance, O‐GlcNAcylation at Ser521 of the nuclear receptor coactivator RBM14 enhances its transcriptional activity, promoting the proliferation and metastasis of lung cancer cells [211, 212, 213]. Similarly, O‐GlcNAcylation at Ser103 of the transcription factor NRF2 by OGT prevents its binding to KEAP1 and subsequent ubiquitin–proteasome degradation, thereby stabilizing and activating NRF2 [214]. This leads to enhanced transcription of downstream antioxidant and prosurvival genes, exacerbating malignant progression and chemotherapy resistance in lung cancer cells [215, 216]. In non‐small cell lung cancer (NSCLC), O‐GlcNAcylation also increases the stability and activity of several transcription‐related proteins, including c‐Myc [217], the nuclear RNA‐binding protein SAM68 [218], and the transcriptional coactivator complex subunit YEATS2 [219], further promoting tumor invasion and metastasis. Beyond transcriptional regulation, O‐GlcNAcylation is directly involved in tumor metabolic reprogramming. Phosphoribosyl pyrophosphate synthetase 1 (PRPS1), a key enzyme in the de novo nucleotide synthesis pathway [220], exhibits enhanced enzymatic activity when O‐GlcNAcylated at Ser83 and Thr166. This promotes nucleotide synthesis, thereby supporting rapid tumor cell proliferation, growth, and drug resistance [221]. Notably, O‐GlcNAcylation also plays a crucial role in maintaining lung cancer stem cell (CSC) properties. The inflammatory cytokine IL‐8, which is highly expressed in lung cancer [222, 223], upregulates glucose transporters GLUT3 and GFAT, facilitating glucose influx into HBP and elevating overall O‐GlcNAcylation levels. This modification helps establish and sustain CSC characteristics, promoting distant metastasis. Inhibition of OGT with OSMI1 significantly reduces CSC populations and suppresses xenograft tumor formation, underscoring the necessity of O‐GlcNAcylation in this process [224]. Furthermore, O‐GlcNAcylation is broadly involved in other malignant behaviors of lung cancer through multiple signaling pathways. For example, the CARM1–USP9X–OGT axis enhances glycolysis and the Warburg effect, promoting tumor growth [225]; O‐GlcNAcylation of SMAD4 activates the TGF‐β signaling pathway to induce epithelial–mesenchymal transition (EMT) [226]; and the IL‐6/STAT3 pathway, regulated by O‐GlcNAcylation, amplifies inflammatory responses, further driving invasion and metastasis [227]. In summary, aberrantly elevated O‐GlcNAcylation drives the initiation and progression of lung cancer through multimolecular and multipathway synergism, making it a promising therapeutic target.
Abnormal O‐GlcNAcylation plays a critical role in the initiation and progression of hepatocellular carcinoma (HCC), one of the most common malignant tumors worldwide [228]. The proportion of HCC cases associated with metabolic dysfunction‐associated steatotic liver disease (MASLD) has been increasing steadily in recent years [229]. A study of 18 MASLD‐HCC patients revealed that 12 exhibited significantly elevated expression of OGT, indicating a strong correlation between aberrant O‐GlcNAcylation and the pathogenesis of MASLD‐HCC [230]. In MASLD‐HCC, highly expressed OGT promotes tumor progression through dual signaling pathways. On one hand, it enhances the expression of p‐JNK and p‐c‐Jun, activates AP‐1, and thereby facilitates JNK/c‐Jun/AP‐1 signal transduction, exacerbating endoplasmic reticulum stress [231, 232]. On the other hand, OGT upregulates the expression of proteins such as p‐IKKa/IKKb, p‐p65, and p‐p50, and enhances the DNA‐binding activity of NF‐κB, promoting palmitic acid metabolism. These changes collectively lead to significantly enhanced growth capacity (p < 0.001), clonogenicity (p < 0.01), and migration and invasion abilities (p < 0.05) in MASLD‐HCC cells [230]. Furthermore, fatty acid synthase (FASN), a key enzyme in fatty acid biosynthesis, also participates in regulating liver cancer progression under elevated O‐GlcNAcylation levels. In HepG2 cells, high O‐GlcNAcylation markedly promotes fatty acid synthesis by activating the PI3K/AKT/mTOR signaling axis, thereby accelerating tumor cell proliferation [233]. Noncoding RNA molecules are also involved in this regulatory network. X‐inactive specific transcript (XIST), a long noncoding RNA highly expressed in liver cancer, promotes O‐GlcNAcylation of the RAF1 oncogene through the XIST/miR‐424‐5p/OGT axis [234, 235]. This modification inhibits ubiquitination‐mediated degradation of RAF1, enhances its stability and activity, and ultimately facilitates liver cancer proliferation and EMT [235]. Additionally, the transcriptional coactivator Yes‐associated protein (YAP), a downstream effector of the Hippo signaling pathway and a known key oncogenic driver, is particularly active under hyperglycemic conditions [236, 237]. Studies confirm that OGT directly modifies the Thr241 residue of the YAP protein, increasing its stability. O‐GlcNAcylated YAP further exacerbates hyperglycemia and hepatic glucose metabolic burden, thereby mediating hyperglycemia‐stimulated hepatocarcinogenesis [238, 239]. In summary, abnormal O‐GlcNAcylation significantly promotes hepatocarcinogenesis, proliferation, invasion, and metastasis by regulating multiple critical biological processes, including gene expression, signal transduction, fatty acid synthesis, and glucose metabolism. It represents a core molecular mechanism in HCC progression.
Abnormal O‐GlcNAcylation plays a critical role in the initiation and progression of CRC, the fourth deadliest cancer worldwide, causing approximately 90,000 deaths annually [240]. The malignant progression of CRC is closely associated with dysregulation of various molecular mechanisms, among which aberrant elevation of O‐GlcNAcylation has become a major research focus. Studies have shown that in CRC cell lines HT29 and HCT116, the overall O‐GlcNAcylation level, OGT, and GFAT—the rate‐limiting enzyme of HBP—are significantly higher than in normal colon cells [241]. Furthermore, dozens of proteins in CRC tissues exhibit abnormally high O‐GlcNAcylation, which suggests a strong correlation between upregulated O‐GlcNAcylation and CRC pathogenesis [242]. In nucleic acid metabolism, thymidylate synthase (TS), a key enzyme in de novo dTMP synthesis essential for DNA synthesis and damage repair, is O‐GlcNAcylated at Thr251 and Thr306 by highly expressed OGT in CRC. This modification inhibits TS proteasomal degradation, enhances its stability, and ultimately promotes tumor cell proliferation [243]. At the gene expression level, the transcription factor YY1, overexpressed in various cancers including CRC [244], is stabilized through O‐GlcNAcylation at Thr236. Modified YY1 activates the expression of oncogenes SLC22A15 and AANAT, facilitating tumorigenesis [245]. Additionally, OGT modifies the Ser196‐198 sites of the m6A reader protein YTHDF1, strengthening its interaction with exportin protein Crm1, promoting its cytoplasmic localization and translation of the downstream oncoprotein c‐Myc, thereby driving cell proliferation [246, 247, 248]. O‐GlcNAcylation also directly upregulates the expression of oncogenes such as casein kinase 2 alpha 1 (CSNK2A1) [249]. These findings demonstrate that O‐GlcNAcylation promotes CRC progression by regulating transcription, translation, and oncoprotein stability. Metabolic reprogramming is a hallmark of cancer. The Warburg effect—enhanced aerobic glycolysis—commonly observed in CRC is also regulated by O‐GlcNAcylation [250, 251]: OGT mediates glycosylation of c‐Myc at Ser415, enhancing its stability and activity, which upregulates PDK2, suppresses mitochondrial pyruvate metabolism and ROS generation, ultimately reinforcing glycolysis, inhibiting the tricarboxylic acid cycle, and promoting tumor cell proliferation [252]. In lipid metabolism, FASN is highly expressed in CRC [253] and positively regulates the expression of OGT and GFPT1, further elevating O‐GlcNAcylation levels and forming a positive feedback loop that accelerates tumor growth and metastasis [254]. Cell–cell and cell–matrix interactions are also modulated by O‐GlcNAcylation. Integrin α5 (ITGA5) shows enhanced stability after O‐GlcNAcylation, promoting cell adhesion, invasion, and EMT [255, 256, 257, 258, 259]. Moreover, the activity and stability of EZH2 are increased due to elevated O‐GlcNAcylation, leading to altered expression of E‐cadherin and β‐catenin and further facilitating tumor metastasis [260]. In the tumor immune microenvironment (TME), the phenotypic polarization of tumor‐associated macrophages (TAMs) is influenced by O‐GlcNAcylation. In colorectal adenocarcinoma (COAD), OGT collaborates with the deubiquitinating enzyme USP18 to inhibit STAT2 degradation, promoting M2 polarization of TAMs and supporting tumor immune evasion and growth [261]. Finally, studies have revealed that SET domain‐containing protein 5 in CRC upregulates the expression of CSC markers such as ESRRB, CD133, and KLF4 by facilitating OGT‐mediated O‐GlcNAcylation of RNAP II, endowing tumor cells with stem‐like properties—a process dependent on O‐GlcNAcylation [262]. In summary, abnormal O‐GlcNAcylation comprehensively drives CRC progression by regulating gene expression, nucleic acid and metabolic reprogramming, fatty acid synthesis, cellular interactions, immune microenvironment remodeling, and cancer stemness, underscoring its pivotal role as a core molecular mechanism in CRC.
Breast cancer, a global health concern, ranks among the top malignancies in women in terms of both incidence and mortality, posing a serious threat to female lives. Recent studies have revealed that aberrant O‐GlcNAcylation plays a key role in promoting the development and progression of breast cancer. In breast cancer, highly expressed TGF‐β1 enhances the stability of GFAT (the rate‐limiting enzyme of the HBP) and activates the mTOR/MYC signaling cascade. This cascade ultimately upregulates OGT expression and elevates global O‐GlcNAcylation levels [263, 264]. Substantial evidence indicates that O‐GlcNAcylation is abnormally elevated in breast cancer and significantly promotes disease progression. On one hand, O‐GlcNAcylation influences gene expression by regulating key transcription factors and chromatin‐remodeling proteins. For example, the Thr556 residue of the chromatin remodeler MORC2 can be O‐GlcNAcylated, and modified MORC2 promotes the expression of TGF‐β1 downstream target genes CTGF and SNAIL, both of which are critical for breast cancer invasion and metastasis [263]. Inhibiting O‐GlcNAcylation markedly reduces the invasive ability of breast cancer cells, indicating that this modification drives tumor progression through MORC2‐mediated transcriptional mechanisms [263]. In triple‐negative breast cancer, O‐GlcNAcylation at Ser40 of histone H2A inhibits the histone demethylase KDM5B, thereby promoting tumor proliferation and metastasis [265]. Additionally, the stability of the oncogenic transcription factor FOXM1 is regulated by O‐GlcNAcylation: under normal conditions, FOXM1 undergoes SIRT1‐mediated ubiquitination and degradation, whereas high levels of O‐GlcNAcylation in breast cancer inhibit the AMPK–SIRT1 axis, enhancing FOXM1 stability and exacerbating malignant behavior [266, 267]. On the other hand, O‐GlcNAcylation promotes tumor progression by affecting cell adhesion, motility, and signal transduction. For instance, OGT overexpression modifies p120 and β‐catenin, leading to reduced E‐cadherin on the cell surface, weakened intercellular adhesion, and facilitated tumor cell detachment and distant dissemination [268]. Meanwhile, O‐GlcNAcylation at Ser108 of the motility‐related protein cofilin enhances its stability, promoting cell movement and invasion [269, 270, 271]. MEK2, an upstream kinase in the ERK signaling pathway, undergoes O‐GlcNAcylation at Thr13, which promotes Thr394 autophosphorylation, enhancing MEK2 stability and activation, thereby driving breast cancer cell proliferation. Mutation at this site significantly reduces tumor proliferation and metastasis [272]. Furthermore, O‐GlcNAcylation promotes drug resistance and metastasis by maintaining CSC properties. GATAD2B, a component of the NuRD complex and a substrate of OGT, is stabilized through O‐GlcNAcylation, which inhibits its ubiquitination and degradation, thereby enhancing CSC self‐renewal and metastatic drug resistance [273, 274, 275, 276]. Similarly, KLF8, a key regulator of mammosphere formation in breast cancer, is also modified by O‐GlcNAcylation, further promoting CSC phenotypes and chemotherapy resistance [277, 278]. In summary, aberrantly elevated O‐GlcNAcylation in breast cancer drives disease progression through multiple mechanisms, including enhancing oncogene expression, disrupting cell adhesion, activating motility‐related proteins, and maintaining stem cell properties, collectively promoting tumor proliferation, invasion, metastasis, and drug resistance. Notably, upregulated O‐GlcNAcylation significantly facilitates EMT and distant metastasis in breast cancer.
In summary, aberrant O‐GlcNAcylation not only significantly promotes the progression of highly prevalent and lethal malignancies such as lung cancer, liver cancer, CRC, and breast cancer, but also plays a critical role in the development and progression of various other malignant solid tumors, including thyroid [279, 280, 281], gastric [282, 283, 284], esophageal [285], cervical [286, 287, 288, 289, 290], ovarian [291, 292, 293, 294, 295], and renal [296, 297] cancers. Thus, abnormal O‐GlcNAcylation can disrupt protein functions, thereby affecting core biological processes such as gene expression, signal transduction, metabolic regulation, and immune modulation, broadly driving the proliferation, invasion, and metastasis of epithelial‐derived malignancies (Table 2).
3.3. Abnormal O‐GlcNAcylation is Implicated in the Pathogenesis of CVDs
CVDs are the leading cause of death and disability worldwide [298, 299, 300]. In recent years, growing evidence has shown that the occurrence and progression of CVDs are closely associated with abnormal levels of O‐GlcNAcylation [301, 302, 303, 304].
Elevated O‐GlcNAcylation plays a key role in the development and progression of cardiac hypertrophy and heart failure (HF) [305]. Cardiac hypertrophy is a typical pathological response of the heart to chronic overload, commonly triggered by conditions such as aortic stenosis (AS), aortic coarctation, and hypertension. These conditions can ultimately lead to severe clinical outcomes including HF and myocardial infarction (MI). Multiple studies have confirmed that O‐GlcNAcylation levels are significantly increased across various heart disease models. In patients with AS, both OGT protein and mRNA expression levels were markedly elevated, with an overall increase of 64.7 ± 7.6% in protein O‐GlcNAcylation compared with the control group. In rat models of aortic coarctation‐induced cardiac hypertrophy and HF, O‐GlcNAcylation levels increased by 80.6 ± 13.1% and 58.2 ±10.0%, respectively. Additionally, hypertensive rat models showed a 47.2 ± 6.5% rise in O‐GlcNAcylation [305]. These data consistently indicate a significant positive correlation between elevated O‐GlcNAcylation and pathological conditions such as AS, aortic coarctation, and hypertension. Functional studies further reveal the pathogenic role of O‐GlcNAcylation in cardiac pathology. Even in the absence of pathological stress, transgenic overexpression of OGT in mouse hearts was sufficient to induce ventricular arrhythmias and premature death. Conversely, reducing O‐GlcNAcylation by overexpressing OGA alleviated cardiac injury and maintained heart function, demonstrating that O‐GlcNAcylation itself has direct disease‐promoting effects [303].
At the molecular level, elevated O‐GlcNAcylation promotes pathological cardiac remodeling through multiple pathways. For instance, excessive O‐GlcNAc modification of key proteins such as c‐Myc, troponin I, and troponin T exacerbates the process of cardiac remodeling [306, 307, 308, 309]. Furthermore, in HF and cardiomyopathy, widespread protein modifications mediated by OGT suppress the activity of genes related to mitochondrial metabolism, suggesting a potential role in promoting cardiac decompensation by impairing mitochondrial function [303]. Additionally, increased O‐GlcNAcylation activates the mTOR signaling pathway, which is known to be closely associated with cardiomyocyte growth and hypertrophy [310, 311], thereby further aggravating pathological cardiac hypertrophy and HF [312]. Similarly, this modification can induce a hypertrophic phenotype by activating the protein kinase A (PKA) pathway. A study by Chen et al. demonstrated that OGT directly modifies the PKA catalytic subunit PKAc via O‐GlcNAcylation. The modified PKAc promotes the synthesis of atrial natriuretic peptide and β‐myosin heavy chain, thereby driving cardiac hypertrophy and dysfunction [313, 314]. Abnormally elevated O‐GlcNAc levels have also been detected in animal models of MI. More importantly, in a postinfarction HF model, further increasing O‐GlcNAcylation levels with the OGA inhibitor PUGNAc led to a significant decline in cardiac contractile function, reaffirming the causal relationship between elevated modification levels and worsening cardiac function [305]. Thus, abnormally elevated O‐GlcNAcylation significantly promotes the development of cardiac hypertrophy, HF, and other cardiovascular pathologies by affecting multiple critical signaling pathways and cellular processes, making it an important pathogenic factor.
The aforementioned study indicates that abnormal O‐GlcNAcylation levels are closely associated with the development and progression of CVDs, but its role is bidirectional. On one hand, substantial evidence suggests that high levels of O‐GlcNAcylation significantly promote CVDs; on the other hand, multiple studies have also pointed out that its low expression similarly accelerates CVDs progression. Specifically, an abnormal decrease in O‐GlcNAcylation is associated with an increased risk of major adverse cardiovascular events (MACEs; including cardiovascular death, nonfatal acute MI, and nonfatal ischemic stroke). A 2025 retrospective study divided 1947 CVD patients into a high‐expression group (974 cases) and a low‐expression group (973 cases) based on their average serum OGT levels. After long‐term follow‐up, the MACEs incidence in the high‐expression group was 7.6% (74 cases), significantly lower than the 10.4% (100 cases) in the low‐expression group (p = 0.032), suggesting that low O‐GlcNAcylation expression may increase the risk of MACEs [315]. Moreover, consistent trends were observed in animal models. A study using a tamoxifen‐induced MI model compared the outcomes between OGT‐knockout mice and wild‐type C57BL/6 mice. The results showed that 4 weeks after MI, the survival rate in the OGT‐knockout group was 64%, significantly lower than the 80% in the wild‐type group, further indicating that the loss of O‐GlcNAcylation may exacerbate the progression of HF [316]. These findings demonstrate that both excessive elevation and abnormal reduction of O‐GlcNAcylation may promote the development of CVDs through different mechanisms.
In summary, experimental observations and molecular mechanism studies indicate that the level of O‐GlcNAcylation significantly influences the occurrence and progression of CVDs. High expression of O‐GlcNAcylation markedly promotes pathological processes such as AS, arterial constriction, and hypertension, thereby exacerbating HF, myocardial hypertrophy, and ischemic myocardial injury (Table 2). Conversely, its low expression also contributes to the occurrence of MACEs and accelerates the deterioration of HF. These findings suggest that maintaining the homeostasis of O‐GlcNAcylation is crucial for the structural and functional integrity of the cardiovascular system. Abnormal expression levels—whether elevated or reduced—may significantly drive the development and progression of CVDs.
3.4. Abnormal O‐GlcNAcylation Promotes the Pathogenesis of Immune‐Related Diseases
O‐GlcNAcylation is a crucial posttranslational modification essential for normal immune system function. It contributes to immune homeostasis by precisely regulating the functions of various immune cells, the activity of immune molecules, and related biological processes. However, aberrant O‐GlcNAcylation can disrupt immune homeostasis and trigger the onset or exacerbation of various immune‐related diseases. For instance, in autoimmune diseases and organ transplant rejection, aberrant O‐GlcNAcylation promotes pathogenesis by influencing immune cell activation and infiltration and modulating inflammatory factor secretion. This section will systematically elaborate on the specific mechanisms by which abnormal O‐GlcNAcylation contributes to the pathogenesis and progression of these diseases.
Autoimmune hepatitis (AIH) is an immune‐mediated inflammatory disease of the liver parenchyma, characterized pathologically by CD4⁺ and CD8⁺ T lymphocyte infiltration leading to hepatocyte injury [317, 318]. As a key organ in maintaining immune homeostasis, the liver promotes immune tolerance to hepatocyte autoantigens by facilitating the infiltration of regulatory T cells (Tregs), thereby suppressing autoimmune responses. In AIH patients, the number of Tregs is significantly reduced, and their immunoregulatory function is critically dependent on the expression of the transcription factor FOXP3. The expression of FOXP3 is regulated by multiple mechanisms, among which O‐GlcNAcylation serves as an important regulatory pathway. Comparative studies using Eogt−⁄− and Eogt⁺⁄⁺ mouse models revealed that Eogt−⁄− mice lacking O‐GlcNAcylation modification exhibited more severe AIH pathological phenotypes, suggesting a protective role of this modification in suppressing AIH pathogenesis. Further molecular mechanism studies indicated that O‐GlcNAcylation promotes FOXP3 expression by activating the Notch signaling pathway, thereby enhancing Tregs differentiation and liver infiltration. However, under AIH conditions, abnormally reduced O‐GlcNAcylation levels lead to decreased Notch signaling activity, restricted FOXP3 expression, impaired Treg function, and insufficient infiltration, ultimately exacerbating autoimmune responses and promoting AIH progression [319]. Thus, abnormally reduced O‐GlcNAcylation contributes to the development and progression of AIH by impairing Treg function and infiltration.
Multiple Sclerosis (MS) is an inflammatory autoimmune disorder characterized by chronic demyelination and neurodegenerative changes in the central nervous system. Its core pathological mechanism involves the differentiation of CD4⁺ T cells into the Th17 subset and the excessive production of IL‐17, which mediates autoimmune inflammatory responses [320, 321]. In this process, microRNA‐15b, a member of the microRNA family, plays a critical regulatory role. This molecule is aberrantly expressed in various autoimmune diseases, neurodegenerative diseases, and cancers. Studies have shown that miR‐15b expression is significantly downregulated in CD4⁺ T cells of MS patients, and its level is negatively correlated with Th17 cell differentiation. Mechanistic studies further demonstrate that reduced miR‐15b expression enhances Th17 cell differentiation and infiltration, thereby driving MS progression. At the molecular level, this regulation depends on O‐GlcNAcylation modification of the nuclear transcription factor NF‐κB by OGT. When miR‐15b expression decreases, this modification process is enhanced, boosting NF‐κB transcriptional activity and upregulating its downstream target gene retinoic acid‐related orphan receptor γt (RORγt). As a key transcription factor for Th17 cell differentiation, increased RORγt expression further promotes Th17 cell generation and related inflammatory responses, ultimately exacerbating MS pathogenesis [322, 323, 324]. Therefore, the aberrantly elevated O‐GlcNAcylation of NF‐κB significantly promotes the pathogenesis of MS by modulating Th17 cell function.
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovitis and bone damage [325]. Its pathological mechanism involves abnormal activation of inflammatory factors and immune cell infiltration, and while the exact process remains incompletely understood, studies indicate a close association with O‐GlcNAcylation [326]. During the onset of RA, TNFα, a key regulator of inflammation, promotes O‐GlcNAcylation events during osteoclastogenesis. Research by Kim et al. demonstrated that TNFα enhances O‐GlcNAcylation of the transcription factor p65. This modification increases p65's DNA‐binding capacity and transcriptional activity, thereby exacerbating RA progression [327, 328]. Additionally, IL‐1β plays a significant role in sustaining chronic inflammation in RA. It induces O‐GlcNAcylation of downstream signaling molecules, further driving disease development [329]. Similar to MS, RA joint tissues exhibit extensive infiltration of Th17 cells differentiated from CD4+ T cells. Studies show that O‐GlcNAcylation of acetyl‐CoA carboxylase 1 (ACC1) in RA enhances the activation of transcription factor RORγt. This, in turn, promotes the production of the highly proinflammatory cytokine IL‐17A by Th17 cells, ultimately intensifying the inflammatory response [330, 331]. Thus, O‐GlcNAcylation significantly contributes to RA progression by participating in inflammatory factor regulatory networks and promoting immune cell infiltration and cytokine release, making it a potential target for therapeutic intervention.
Organ transplantation is a critical treatment for end‐stage organ failure, yet it is often complicated by various postoperative issues, with immune rejection representing the most significant and potentially fatal complication. Similar to autoimmune diseases, this process is primarily driven by immune cell‐mediated inflammatory infiltration and tissue damage. Notably, the mTOR signaling pathway is frequently hyperactivated in transplant recipients and regulates multiple cellular processes, including metabolism and O‐GlcNAcylation [332]. As an important posttranslational modification, O‐GlcNAcylation is widely involved in the differentiation, maturation, and functional regulation of immune cells such as T cells, B cells, macrophages, and NK cells, playing a significant role in transplant immune rejection. First, O‐GlcNAcylation exacerbates cellular immune responses by promoting T cell maturation and activation while inhibiting apoptosis. Studies show that during T cell receptor (TCR) β‐chain rearrangement, knockdown of OGT leads to a significant reduction in CD4⁺ CD8⁺ T cells, while OGT overexpression promotes their generation, indicating that O‐GlcNAcylation levels directly affect TCR rearrangement efficiency and T cell development [333, 334, 335]. During activation, OGT expression markedly increases in T cells [336, 337]; siRNA‐mediated OGT inhibition significantly reduces the synthesis and release of the key activator IL‐2 [338, 339, 340, 341]. O‐GlcNAcylation also modulates the activity of critical transcription factors involved in T cell activation, such as NFAT, c‐Myc, and NF‐κB, whose activation is essential for IL‐2 transcription [338, 339, 342]. Furthermore, reducing O‐GlcNAcylation levels upregulates PD‐1 expression and promotes CD8⁺ T cell apoptosis, whereas overexpression exerts an antiapoptotic effect [343, 344]. These findings collectively demonstrate that O‐GlcNAcylation enhances T cell‐mediated immune responses in multiple ways. Second, O‐GlcNAcylation significantly promotes B cell survival, activation, and antibody production. B cell survival depends on the BAFF/BAFFR signaling pathway. OGT deficiency reduces the expression of key molecules—p50, p65, RelB, and p52—leading to B cell apoptosis. [345, 346, 347]. Under low OGT conditions, the proportions of splenic follicular B cells and mature B cells in mice decrease, and the activity of germinal center B cells, memory B cells, and plasma cells is suppressed, indicating that O‐GlcNAcylation is essential for maintaining B cell homeostasis and antibody responses [348]. Conversely, OGT overexpression enhances O‐GlcNAcylation of NF‐κB p65 and NFATc1, further elevating B cell activation [349]. Thus, O‐GlcNAcylation significantly promotes antibody‐mediated rejection by strengthening humoral immune responses. Finally, O‐GlcNAcylation broadly regulates the functions of innate immune cells such as macrophages and NK cells, exacerbating inflammatory responses. In macrophages, OGT‐mediated O‐GlcNAcylation of NF‐κB subunits RelA and c‐Rel at Thr305 and Ser350 enhance their transcriptional activity and stability, promoting the release of inflammatory factors such as NO and ILs [342, 350]. Meanwhile, O‐GlcNAcylation of STAT3 at Thr717 can suppress the production or signaling of the anti‐inflammatory cytokine IL‐10, further amplifying proinflammatory responses [351, 352, 353]. In NK cells, O‐GlcNAcylation at the Ser75 site of histone methyltransferase EZH2 promotes cell activation and homeostasis [354, 355, 356]. Additionally, O‐GlcNAcylation inactivates the transcription factor FOXO1 by promoting its de‐phosphorylation, thereby relieving its suppression of NK cell function and enhancing cytotoxicity [357, 358]. Clearly, O‐GlcNAcylation plays a central role in immune rejection after organ transplantation. By promoting the activation, survival, and function of T and B cells, as well as enhancing the inflammatory effects of macrophages and NK cells, it drives cellular, humoral, and innate immune responses in multiple dimensions, ultimately leading to rejection. Targeting the O‐GlcNAcylation signaling pathway may offer new strategies for antirejection therapy.
In summary, the stability of O‐GlcNAcylation levels is crucial for maintaining immune system homeostasis. Under physiological conditions, moderate O‐GlcNAcylation supports the normal functioning of the immune regulatory network. However, abnormal O‐GlcNAcylation can significantly disrupt immune balance, leading to functional disorders and abnormal infiltration of various immune cells—such as T cells, B cells, NK cells, and macrophages—thereby promoting the development and progression of multiple immune‐related diseases (Table 2). These include AIH, MS, RA, and organ transplant rejection.
4. Prospects of Targeted O‐GlcNAcylation Therapy
O‐GlcNAcylation plays a critical role in the development and progression of various diseases, including neurodegenerative diseases, cancers, CVDs, and immune‐related disorders. Given its significant contribution to pathological processes, targeting O‐GlcNAcylation demonstrates broad clinical application prospects. However, systematic exploration and comprehensive summaries focusing on this target remain relatively limited. This section aims to discuss the feasibility of O‐GlcNAcylation as a therapeutic target, along with current challenges and future research directions. The goal is to provide new perspectives for optimizing clinical treatment strategies and advancing drug development for related diseases.
4.1. Feasibility of O‐GlcNAcylation as a Therapeutic Target
Abnormal O‐GlcNAcylation is widely involved in the occurrence and progression of various diseases, playing a significant role in promoting pathological processes. Its expression and functional impact vary considerably across different diseases: in cancers and CVDs, aberrant elevation of O‐GlcNAcylation markedly accelerates disease progression, whereas in neurodegenerative and immune‐related disorders, either increases or decreases in its levels can exacerbate pathogenesis. Overall, O‐GlcNAcylation serves as a key regulatory mechanism in multiple diseases, and its dysregulation is an important driver of disease development. Therefore, targeting O‐GlcNAcylation offers a promising strategy to effectively halt disease progression and has important implications for optimizing clinical treatment approaches. In eukaryotes, O‐GlcNAcylation is catalyzed by two highly conserved enzymes: OGT and OGA. The absence of isoenzymes means that interventions targeting OGT and OGA avoid issues such as functional redundancy, off‐target effects, or reduced efficacy, thereby ensuring high specificity. Moreover, the sole substrate for this modification, UDP‐GlcNAc, is exclusively supplied by HBP. Targeting HBP does not trigger metabolic compensation or alternative pathways. Thus, for diseases highly dependent on O‐GlcNAcylation, interventions aimed at OGT, OGA, or HBP not only represent promising therapeutic strategies but also demonstrate broad prospects for clinical translation.
OGT is a glycosyltransferase that covalently modifies target proteins by catalyzing O‐GlcNAcylation, playing a role in regulating various cellular processes. To date, several classes of OGT inhibitors have been developed, including UDP‐GlcNAc analogs (such as Alloxan [359, 360, 361, 362, 363, 364], UDP‐5S‐GlcNAc [365, 366, 367], BADGP [368, 369, 370, 371, 372], and Ac‐5SGlcNAc [373, 374, 375]), high‐throughput screening‐derived inhibitors (such as ST045849 [376, 377, 378, 379, 380], BZX [381, 382], and the OSMI series [383, 384, 385, 386, 387, 388, 389]), bisubstrate inhibitors (such as goblin1‐2 [390, 391]), and benzoxazolinones [392] (Table 3). However, these inhibitors generally suffer from limitations such as low specificity, high cytotoxicity, poor cell permeability, or significant off‐target effects. Among them, the small molecule OSMI is widely used in disease mechanism research and preclinical models due to its relatively stable physicochemical properties, low side effects, and good cellular permeability. Particularly in malignant tumor research, OSMI demonstrates multifaceted inhibitory effects. For example, in prostate cancer, OSMI effectively inhibits OGT‐mediated super‐enhancer‐dependent gene expression and the function of KIF1A protein, which drives neuroendocrine differentiation, thereby significantly suppressing tumor cell proliferation [393, 394]. In pancreatic ductal adenocarcinoma, OGT promotes tumor recurrence by modifying and stabilizing the transcription factor SOX2, while OSMI treatment markedly reduces the O‐GlcNAcylation level and stability of SOX2, thus decreasing recurrence [395]. In NSCLC, OSMI enhances the sensitivity of STK11/KRAS/LKB1‐mutant tumors to cisplatin by interfering with OGT's regulation of the HBP [396]. Furthermore, multiple studies have confirmed that OSMI, either alone or in combination with chemotherapeutic agents, significantly induces apoptosis in prostate cancer cell line PC‐3 [397], CRC cell lines HCT116 and LS174T [398, 399], liver cancer cell lines HCC and HepG2 [400, 401, 402, 403], and bladder cancer cell line YTS‐1 [404]—an effect also validated in vitro. Beyond malignant tumors, OSMI shows potential therapeutic value in CVDs. High OGT activity catalyzes O‐GlcNAcylation at Ser280 of CaMKII, triggering massive mitochondrial ROS release and leading to ventricular cardiomyocyte injury [405]. OSMI intervention effectively inhibits this modification process and alleviates ROS‐mediated myocardial damage. Thus, OGT demonstrates significant potential as a therapeutic target in both cancers and CVDs, and its inhibitor OSMI exhibits broad biological activity and promising application prospects in basic and preclinical research. Although more data are needed to confirm the effectiveness of OGT as a therapeutic target in other diseases with abnormally high O‐GlcNAcylation, existing evidence provides a strong basis for its further development.
TABLE 3.
Reagents for regulating protein O‐GlcNAcylation levels.
| Reagent | Type | Advantage | Disadvantage | References |
|---|---|---|---|---|
| OGT inhibitors (downregulation of O‐GlcNAcylation) | ||||
| Alloxan (2,4,5,6‐tetraoxypyrimidine) | Substrate analogues | Early tools for exploring the biological functions of O‐GlcNAc | Lacks of specificity; off‐target effects, promotes ROS generation; cytotoxicity; leading to apoptosis and necrosis of pancreatic β cells | [359, 360, 361, 362, 363, 364] |
| UDP‐5S‐GlcNAc | Substrate analogues | Exhibits extremely high specificity | Utilized by OGT at a much lower rate compared with UDP‐GlcNAc; reduces the intracellular pool of UDP‐GlcNAc by hijacking HBP; affects other glycosyltransferases by either direct or indirect inhibition | [365, 366, 367] |
| BADGP | Substrate analogues | Highly cell permeable | Inhibitory efficacy is low | [368, 369, 370, 371, 372] |
| Ac‐5SGlcNAc | Substrate analogues | High specificity; highly cell permeable | There has been no evaluation of its efficiency and safety in vivo. | [373, 374, 375] |
| ST045849 (compound 4) | HTS‐derived inhibitors | Can penetrate cells and possess cellular vitality | Relatively low inhibitory potency and impact on protein stability | [376, 377, 378, 379, 380] |
| BZX (compound 5) | HTS‐derived inhibitors | Inhibitory efficacy is good and cell viability | Cross‐links the active site of OGT (Lys842 and Cys917) through a double‐displacement mechanism, with potential toxic and off‐target effects | [381, 382] |
| OSMI series | HTS‐derived inhibitors | High specificity; highly cell permeable | Inhibitory efficacy is low; off‐target effect; incomplete specificity; leads to unreliable reversible inactivation of OGT; affects cell viability | [383, 384, 385, 386, 387, 388, 389] |
| Goblin1‐2 | Bisubstrate inhibitors | Highly specific and glucose concentration‐dependent inhibition | Lacks of cell permeability | [ 390 , 391 ] |
| OGA inhibitors (upregulation of O‐GlcNAcylation) | ||||
| PUGNAc | OGA inhibitors | Highly cell permeable | Lacks of selectivity and specificity; relatively low inhibitory potency; cytotoxic | [406, 407] |
| Thiamet‐G | OGA inhibitors | High selectivity and efficiency; highly cell permeable | Nonspecific global impact; cytotoxic; inducible insulin resistance | [408, 409] |
| NButGT | OGA inhibitors | Highly efficient and little impact on cell viability | Off‐target effects; low blood–brain barrier penetration efficiency; synaptic toxicity | [410, 411, 412] |
| NAG‐thiazoline | OGA inhibitors | Highly cell permeable; high efficiency and specificity | Short half‐life, unstable structure, easily metabolized and eliminated; lack of selectivity in vivo; easy to produce off‐target effects; cytotoxic | [413] |
| GlcNAcstatin | OGA inhibitors | Highly efficient | Lacks of cell permeability; metabolized or eliminated more rapidly in the body; off‐target effects | [414, 415] |
| HBP inhibitors (downregulation of O‐GlcNAcylation) | ||||
| Azaserine (O‐diazoacetyl‐l‐serine) | GFAT inhibitors | Highly efficient | Severe systemic toxicity, carcinogenicity, and low selectivity to GFAT | [420] |
| DON (6‐diazo‐5‐oxo‐l‐norleucine) | GFAT inhibitors | Highly efficient; antitumor proliferation | Has low selectivity for GFAT; causes systemic toxicity | [421, 422, 423, 424, 425] |
OGA is a hydrolase that specifically cleaves the covalent bond between O‐GlcNAc and proteins, thereby removing O‐GlcNAc modifications from target proteins, a process known as de‐O‐GlcNAcylation. Reduced levels of O‐GlcNAcylation have been observed in various diseases, including neurodegenerative diseases such as AD, PD, HD, and ALS, as well as certain immune‐related diseases. Therefore, inhibiting OGA to restore normal protein glycosylation levels is considered a potential therapeutic strategy. Several OGA inhibitors have been developed, including PUGNAc [406, 407], Thiamet‐G [408, 409], NButGT [410, 411, 412], NAG‐thiazoline [413], and GlcNAcstatin [414, 415] (Table 3). As the earliest and most widely used inhibitor, PUGNAc has demonstrated protective effects in multiple neurodegenerative disease models: in AD models, it increases O‐GlcNAcylation of Tau protein and reduces its hyperphosphorylation, thereby slowing the decline in learning and memory functions [416]; in PD and ALS models, PUGNAc enhances neuronal activity and delays disease progression [417]. Although PUGNAc shows certain therapeutic effects, its poor substrate selectivity and structural limitations hinder its clinical application [418]. To overcome these drawbacks, researchers developed GlcNAcstatin based on the structure of PUGNAc, which exhibits high selectivity for human OGA (hOGA) [419]. Currently, research on GlcNAcstatin, NButGT, Thiamet‐G, and other inhibitors in diseases associated with O‐GlcNAcylation deficiency remains relatively limited. Nevertheless, the neuroprotective effects demonstrated by PUGNAc fully indicate that targeting OGA for drug intervention is both feasible and worthy of further exploration.
The HBP is the sole metabolic pathway in cells for UDP‐GlcNAc, the substrate required for O‐GlcNAcylation. Inhibiting HBP effectively reduces UDP‐GlcNAc production, thereby decreasing overall protein O‐GlcNAcylation levels. Thus, targeting HBP, particularly its key rate‐limiting enzyme GFAT, represents a promising strategy for correcting abnormal O‐GlcNAcylation. Currently known GFAT inhibitors primarily include glutamine analogs, such as O‐diazoacetyl‐l‐serine (azaserine) [420] and 6‐diazo‐5‐oxo‐l‐norleucine (DON) [421, 422, 423, 424, 425] (Table 3). These compounds not only significantly inhibit tumor cell proliferation and other malignant phenotypes but also demonstrate therapeutic potential in CVDs [426]. Studies have shown that inhibiting GFAT activity with DON or azaserine effectively alleviates CVDs and improves pathological conditions [426]. Notably, a synergistic effect exists between inhibiting HBP and directly inhibiting OGT. Simultaneously targeting both pathways can further enhance the regulation of O‐GlcNAcylation. Therefore, targeting HBP not only helps improve the efficacy of OGT inhibitors but also practically validates the feasibility of targeting HBP in the treatment of diseases associated with elevated O‐GlcNAcylation levels.
In summary, aberrant O‐GlcNAcylation is a significant driver of disease progression. This modification is uniquely dependent on the HBP for substrate supply, with its addition and removal catalyzed exclusively by OGT and OGA, respectively. Due to this well‐defined and streamlined mechanism, O‐GlcNAcylation represents a promising therapeutic target. Indeed, interventions targeting this process in various disease models have successfully attenuated pathological progression and shown promising therapeutic efficacy. Therefore, developing treatment strategies focused on modulating O‐GlcNAcylation is highly feasible.
4.2. Current Challenges and Future Directions
O‐GlcNAcylation, as a crucial posttranslational modification of proteins, is extensively involved in various vital biological processes such as cellular signal transduction, transcriptional regulation, and metabolic homeostasis. Its dysregulation is closely associated with the development and progression of multiple diseases. Therefore, targeting the O‐GlcNAc cycling pathway—including inhibiting OGT, OGA, or HBP—has emerged as an important therapeutic strategy. However, this approach still faces several key challenges in translational applications. The primary issue lies in the complex relationship between the biological characteristics of O‐GlcNAcylation and disease pathology. This modification is widespread throughout the body and plays a critical role in maintaining homeostasis, whereas related diseases such as malignant tumors, neurodegenerative diseases, and CVDs often exhibit tissue‐specific manifestations. Thus, an ideal therapeutic strategy must precisely regulate abnormal O‐GlcNAcylation levels at the lesion site while preserving systemic physiological functions, thereby avoiding side effects caused by broad inhibition. Achieving such spatial selectivity remains a major focus and challenge in current research. Second, existing drugs targeting O‐GlcNAcylation still exhibit significant limitations in potency and specificity. Numerous inhibitors developed to date—such as those targeting OGT, OGA, or GFAT—commonly suffer from issues like low target selectivity, poor cell permeability, high cytotoxicity, and off‐target effects. For instance, the classic OGT inhibitor Alloxan not only has limited efficacy but also induces ROS generation leading to cell death and may simultaneously affect OGA activity. Benzoxazolinone‐based molecules can cause irreversible OGT inactivation, which lacks controllability, while BADGP may compromise antipathogen defense by interfering with host immune responses [427, 428]. These limitations underscore the urgent need to develop novel inhibitors with high selectivity, stability, and low toxicity. Although targeting O‐GlcNAcylation still faces numerous obstacles, addressing two key issues—tissue‐specific regulation and drug efficacy/safety—could significantly advance its clinical translation. Future research should focus on designing precise intervention strategies for localized pathological sites and optimizing compound structures to enhance selectivity and biocompatibility, thereby offering new avenues for treating related diseases.
Despite its promise as a therapeutic strategy for various diseases, targeting O‐GlcNAcylation faces two major challenges: the inadequate efficacy of current inhibitors and their lack of spatial specificity. Overcoming these limitations will be a central focus of future research.
To tackle the issue of insufficient inhibitors performance, there are currently two main optimization pathways. The first involves improving performance through drug iteration. This entails systematically optimizing the structure, efficacy, and side effects of previous‐generation drugs to develop next‐generation drugs with stronger inhibitory effects, higher selectivity, and lower toxicity. For example, the OGA inhibitor GlcNAcstatin was optimized based on the structure of PUGNAc, demonstrating superior inhibitory efficiency and specificity for OGA [429, 430]. Similarly, starting from OSMI1, researchers have successively developed more specific follow‐up versions such as OSMI2, OSMI3, and OSMI4. The second pathway involves exploring new types of chemical drugs or biological agents. Currently, interventions for diseases related to abnormal O‐GlcNAcylation levels primarily rely on inhibitors: diseases with high O‐GlcNAcylation often target OGT or GFAT (a rate‐limiting enzyme in the HBP pathway), while those with low levels often inhibit OGA. However, research on corresponding agonists has lagged. In fact, developing agonists for OGT, OGA, or GFAT holds significant potential. For instance, OGA agonists could work synergistically with OGT or GFAT inhibitors to reduce O‐GlcNAcylation levels, potentially benefiting cancers and CVDs. Conversely, OGT or GFAT agonists could be combined with OGA inhibitors to increase O‐GlcNAcylation levels, offering new approaches for neurodegenerative diseases and some autoimmune disorders. Additionally, antagonistic antibody drugs targeting OGT, OGA, or GFAT are highly promising, as they can block their targets with high specificity and sensitivity, complementing existing inhibitors. Novel drugs regulating the expression of these genes (e.g., siRNA, antisense oligonucleotides) are also worth exploring, particularly for O‐GlcNAcylation‐related diseases caused by abnormal expression.
In addressing the challenge of insufficient spatial specificity, targeted delivery technologies represented by nanomaterials and engineered exosomes offer promising solutions. Various nanomaterials can serve as drug carriers due to their excellent biocompatibility and potential for targeted release. For instance, liposomes—micron‐scale phospholipid vesicles with a bilayer membrane structure—can encapsulate diverse drug molecules and release them in specific organs, making them widely used in interventions for tumors and CVDs [431]. A diverse range of other materials, including nanoparticles, hydrogels, micelles, dendrimers, mesoporous materials, adenoviruses, lysozymes, elastin‐like polypeptides, and chitosan, also demonstrate excellent delivery capabilities [432, 433, 434, 435, 436, 437, 438, 439, 440, 441, 442, 443, 444, 445, 446, 447, 448, 449, 450, 451, 452, 453, 454, 455]. By surface‐modifying these materials and loading them with OGT, OGA, or GFAT inhibitors, high‐precision delivery to specific tissues or lesion sites can be achieved. Exosomes, natural vesicles secreted by cells, play a key role in intercellular communication and material transport. Their low immunogenicity, high biocompatibility, and excellent stability make them ideal drug carriers. Notably, exosomes possess a natural “homing” ability to target diseased areas, and diseased cells (such as tumor cells) produce exosomes in large quantities. Through engineering approaches—loading exosomes with specific inhibitors and modifying their surface molecules—their targeted delivery capacity can be further enhanced, enabling more precise drug delivery to lesion sites. This improves therapeutic efficacy while reducing systemic side effects.
In summary, the main challenges of targeting O‐GlcNAcylation therapeutically are the low efficacy and significant side effects of existing inhibitors, coupled with a lack of tissue or cellular specificity. To overcome these hurdles, future research should focus on three core strategies: iterating and improving upon current drugs; developing novel therapeutics; and utilizing advanced targeted delivery systems for precise spatial drug delivery. Together, these approaches form a critical pathway for the field's advancement.
5. Summary
O‐GlcNAcylation is an emerging posttranslational modification that primarily targets Ser/Thr residues, extensively modulating protein structure and function. Since its discovery, it has been consistently shown to play an indispensable role in sustaining normal biological activities and overall health—spanning gene expression, signal transduction, metabolism, and the cell cycle. As a fundamental biological process, gene expression is profoundly shaped by O‐GlcNAcylation. This modification contributes to the assembly of transcription initiation complexes, guides mRNA splicing, and fine‐tunes the initiation, elongation, termination stages of protein translation—thereby exerting precise control over the entire flow of genetic information. In signal transduction, O‐GlcNAcylation exerts significant influence over the activation of diverse signaling cascades and the stability of signaling molecules. For instance, by modifying β‐catenin in the Wnt/β‐catenin pathway, it enhances the protein's stability and thereby supports tissue homeostasis and regeneration. Concurrently, O‐GlcNAcylation regulates RTK pathways such as AKT and MAPK, which are central to cell proliferation, differentiation, apoptosis, and energy storage. Moreover, it can activate cytokine‐related pathways such as TNFα and ILs, enabling cells to adapt to external stimuli and underscoring its critical role in health maintenance. As the core process governing energy supply, metabolism is dynamically regulated by O‐GlcNAcylation. Under energy‐deficient conditions such as fasting, this modification promotes the expression of key gluconeogenic genes—including FOXO1, CRTC2, PGC‐1α, and ERRγ. Conversely, in hyperglycemic environments, it boosts insulin secretion by modulating transcription factors such as PDX1 and NeuroD1. These mechanisms illustrate the central role of O‐GlcNAcylation in maintaining energy balance and metabolic homeostasis. The cell cycle, which underlies growth and proliferation, is strongly supported by O‐GlcNAcylation. It facilitates the orderly progression through G1, S, G2, and M phases and ensures the accurate execution of phase‐specific events. In contrast, deficiency or dysregulation of O‐GlcNAcylation markedly impedes the cell cycle progression, further emphasizing its necessity for cellular proliferation. O‐GlcNAcylation participates broadly and deeply in multiple core biological processes essential for organismal homeostasis. Its multifaceted functions across gene expression, signal transduction, metabolism, and the cell cycle collectively affirm its critical contribution to health and physiological stability.
As research advances, a growing body of data indicates that abnormal elevation or reduction in O‐GlcNAcylation levels is present in various diseases and plays a critical role in their onset and progression. Aberrant O‐GlcNAcylation is widely involved in multiple pathological processes, including CVDs, cancers, immune‐related diseases, neurodegenerative diseases, contributing to disease progression through direct or indirect mechanisms. In CVDs and cancers, O‐GlcNAcylation is often abnormally elevated. In the cardiovascular system, excessively high O‐GlcNAcylation can promote HF by activating the mTOR pathway or induce ventricular cardiomyocyte hypertrophy through PKA pathway activation, significantly exacerbating cardiovascular pathology. In cancers, elevated O‐GlcNAcylation facilitates key processes such as de novo nucleic acid synthesis, oncogene expression, transcription factor activation, CSC formation, immune evasion, and metabolic reprogramming of glucose and lipids, thereby driving the proliferation, invasion, metastasis, and drug resistance of malignancies like lung, liver, colorectal, and breast cancers. Abnormally high O‐GlcNAcylation also plays an important role in immune‐related diseases. Elevated modification levels can lead to dysfunction of T and B cells, enhance their tissue infiltration capacity, and trigger sustained inflammatory responses, thereby promoting conditions such as AIH, MS, RA, and organ transplant rejection. In most neurodegenerative diseases, O‐GlcNAcylation levels are abnormally decreased and similarly contribute to disease progression. Low O‐GlcNAcylation can cause abnormal Tau aggregation in AD, α‐synuclein accumulation and dopamine neuron loss in PD, dysfunction of NPCs in HD, and impaired functions of NF, TDP‐43, and NPGPx in ALS. Notably, in HD, abnormal aggregation of HTT under high O‐GlcNAcylation conditions is also a key pathogenic mechanism. These facts demonstrate that dysregulated O‐GlcNAcylation is a key driver of disease pathogenesis.
O‐GlcNAcylation is significantly dysregulated in a range of diseases. Whether abnormally elevated or reduced, this modification drives disease initiation and progression, establishing it as a crucial pathological regulator. Consequently, targeting O‐GlcNAcylation has emerged as a promising therapeutic strategy. Current approaches to modulate aberrant O‐GlcNAcylation primarily fall into two categories: using OGT or GFAT inhibitors to curb abnormal elevations, or employing OGA inhibitors to augment deficient levels. However, existing inhibitors generally suffer from poor selectivity, off‐target effects, and significant side effects, which limit their clinical application. Additionally, current strategies lack spatial specificity, leading to unintended drug effects in nontarget tissues and compromising both efficacy and safety. To enhance the feasibility and effectiveness of targeting O‐GlcNAcylation, future research should focus on three key strategies: optimizing existing drugs, developing new inhibitors, and utilizing targeted delivery systems. This multipronged approach aims to improve drug performance and local concentration while reducing systemic exposure and related side effects. By integrating drug optimization with innovative delivery systems, O‐GlcNAcylation holds promise as a highly efficient and precise therapeutic target, ultimately accelerating its clinical translation.
Finally, this paper systematically summarizes the beneficial roles of O‐GlcNAcylation in various biological processes and thoroughly analyzes how its dysregulation contributes to disease pathogenesis, thereby elucidating its critical functions in both health and disease. Building on this foundation, we evaluate the feasibility, challenges, and future research directions of targeting O‐GlcNAcylation therapeutically, with the aim of accelerating its clinical translation.
Author Contributions
Z.R., C.C. and C.Y.: Writing—original draft and visualization. X.L. and C.Y.: Conceptualization, supervision, and review. J.O. and G.W.: Literature collection. All authors have read and agreed to the published version of the manuscript.
Conflicts of Interest
The authors declare no conflicts of interests.
Ethics Statement
The authors have nothing to report.
Funding
This work was supported by the Basic Research Program of Guangzhou Municipal Science and Technology Bureau (Grant number: SL2022A03J00629); the Natural Science Foundation of China (NSFC) (Grant number: 82000714); the Guangdong Medical Science and Technology Research Fund (Grant number: A2024367); and the Plan on Enhancing Scientific Research in GMU (Grant number: GMUCR2024‐01008).
Acknowledgments
On behalf of all authors, we would like to express our sincere gratitude to the First Affiliated Hospital of Guangzhou Medical University for their invaluable support. We also wish to extend our appreciation to “Adobe Systems Incorporated” for providing “Adobe Illustrator,” the vector graphics software used to create all the figures in this article.
Ran Z., Chen C., Ou J., Wu G., Yang C., and Liu X., “Protein O‐GlcNAcylation in Health and Diseases.” MedComm 6, no. 12 (2025): e70536. 10.1002/mco2.70536
Zhihong Ran and Chuanbao Chen contributed equally to this work.
Contributor Information
Chao Yang, Email: cyang18@gzhmu.edu.cn.
Xiaoyou Liu, Email: liuxy@gzhmu.edu.cn.
Data Availability Statement
The authors have nothing to report.
References
- 1. Torres C. R. and Hart G. W., “Topography and Polypeptide Distribution of Terminal N‐Acetylglucosamine Residues on the Surfaces of Intact Lymphocytes. Evidence for O‐Linked Glcnac,” Journal of Biological Chemistry 259, no. 5 (1984): 3308–3317, 10.1016/S0021-9258(17)43295-9. [DOI] [PubMed] [Google Scholar]
- 2. Hart G. W., Housley M. P., and Slawson C., “Cycling of O‐Linked Beta‐N‐Acetylglucosamine on Nucleocytoplasmic Proteins,” Nature 446, no. 7139 (2007): 1017–1022, 10.1038/nature05815. [DOI] [PubMed] [Google Scholar]
- 3. Joiner C. M., Li H., Jiang J., et al., “Structural Characterization of the O‐Glcnac Cycling Enzymes: Insights into Substrate Recognition and Catalytic Mechanisms,” Current Opinion in Structural Biology 56 (2019): 97–106, 10.1016/j.sbi.2018.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Kreppel L. K., Blomberg M. A., and Hart G. W., “Dynamic Glycosylation of Nuclear and Cytosolic Proteins. Cloning and Characterization of a Unique O‐Glcnac Transferase With Multiple Tetratricopeptide Repeats,” Journal of Biological Chemistry 272, no. 14 (1997): 9308–9315, 10.1074/jbc.272.14.9308. [DOI] [PubMed] [Google Scholar]
- 5. Lubas W. A., Frank D. W., Krause M., et al., “O‐Linked Glcnac Transferase Is a Conserved Nucleocytoplasmic Protein Containing Tetratricopeptide Repeats,” Journal of Biological Chemistry 272, no. 14 (1997): 9316–9324, 10.1074/jbc.272.14.9316. [DOI] [PubMed] [Google Scholar]
- 6. Hart G. W., Slawson C., Ramirez‐Correa G., et al., “Cross Talk between O‐Glcnacylation and Phosphorylation: Roles in Signaling, Transcription, and Chronic Disease,” Annual Review of Biochemistry 80 (2011): 825–858, 10.1146/annurev-biochem-060608-102511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Hart G. W., Housley M. P., and Slawson C., “Cycling of O‐Linked Beta‐N‐Acetylglucosamine on Nucleocytoplasmic Proteins,” Nature 446, no. 7139 (2007): 1017–1022, 10.1038/nature05815. [DOI] [PubMed] [Google Scholar]
- 8. Joud M., Moller M., and Olsson M. L., “Identification of Human Glycosyltransferase Genes Expressed in Erythroid Cells Predicts Potential Carbohydrate Blood Group Loci,” Scientific Reports 8, no. 1 (2018): 6040, 10.1038/s41598-018-24445-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Karczewski K. J., Francioli L. C., Tiao G., et al., “The Mutational Constraint Spectrum Quantified from Variation in 141,456 Humans,” Nature 581, no. 7809 (2020): 434–443, 10.1038/s41586-020-2308-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Resto M., Kim B., Fernandez A. G., et al., “O‐Glcnacase Is an Rna Polymerase II Elongation Factor Coupled to Pausing Factors Spt5 and Tif1Beta,” Journal of Biological Chemistry 291, no. 43 (2016): 22703–22713, 10.1074/jbc.M116.751420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Love D. C., Ghosh S., Mondoux M. A., et al., “Dynamic O‐Glcnac Cycling at Promoters of Caenorhabditis Elegans Genes Regulating Longevity, Stress, and Immunity,” Proceedings of the National Academy of Sciences of the United States of America 107, no. 16 (2010): 7413–7418, 10.1073/pnas.0911857107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Love D. C. and Hanover J. A., “The Hexosamine Signaling Pathway: Deciphering the “O‐Glcnac Code”,” Science Stke no. 312 (2005): re13. [DOI] [PubMed] [Google Scholar]
- 13. Wells L., Gao Y., Mahoney J. A., et al., “Dynamic O‐Glycosylation of Nuclear and Cytosolic Proteins: Further Characterization of the Nucleocytoplasmic Beta‐N‐Acetylglucosaminidase, O‐Glcnacase,” Journal of Biological Chemistry 277, no. 3 (2002): 1755–1761, 10.1074/jbc.M109656200. [DOI] [PubMed] [Google Scholar]
- 14. Li X., Gong W., Wang H., et al., “O‐Glcnac Transferase Suppresses Inflammation and Necroptosis by Targeting Receptor‐Interacting Serine/Threonine‐Protein Kinase 3,” Immunity 50, no. 3 (2019): 576–590, 10.1016/j.immuni.2019.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Hardiville S. and Hart G. W., “Nutrient Regulation of Signaling, Transcription, and Cell Physiology by O‐Glcnacylation,” Cell Metabolism 20, no. 2 (2014): 208–213, 10.1016/j.cmet.2014.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Wei S., Zhao Q., Zheng K., et al., “Gfat1‐Linked Tab1 Glutamylation Sustains P38 Mapk Activation and Promotes Lung Cancer Cell Survival under Glucose Starvation,” Cell Discovery 8, no. 1 (2022): 77, 10.1038/s41421-022-00423-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Hart G. W., Slawson C., Ramirez‐Correa G., et al., “Cross Talk between O‐Glcnacylation and Phosphorylation: Roles in Signaling, Transcription, and Chronic Disease,” Annual Review of Biochemistry 80 (2011): 825–858, 10.1146/annurev-biochem-060608-102511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Hart G. W., Housley M. P., and Slawson C., “Cycling of O‐Linked Beta‐N‐Acetylglucosamine on Nucleocytoplasmic Proteins,” Nature 446, no. 7139 (2007): 1017–1022, 10.1038/nature05815. [DOI] [PubMed] [Google Scholar]
- 19. Groves J. A., Lee A., Yildirir G., et al., “Dynamic O‐Glcnacylation and Its Roles in the Cellular Stress Response and Homeostasis,” Cell Stress & Chaperones 18, no. 5 (2013): 535–558, 10.1007/s12192-013-0426-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Chang Y. H., Weng C. L., and Lin K. I., “O‐Glcnacylation and Its Role in the Immune System,” Journal of Biomedical Science 27, no. 1 (2020): 57, 10.1186/s12929-020-00648-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Yang Y. R., Song M., Lee H., et al., “O‐Glcnacase Is Essential for Embryonic Development and Maintenance of Genomic Stability,” Aging Cell 11, no. 3 (2012): 439–448, 10.1111/j.1474-9726.2012.00801.x. [DOI] [PubMed] [Google Scholar]
- 22. O'Donnell N., Zachara N. E., Hart G. W., et al., “Ogt‐Dependent X‐Chromosome‐Linked Protein Glycosylation Is a Requisite Modification in Somatic Cell Function and Embryo Viability,” Molecular and Cellular Biology 24, no. 4 (2004): 1680–1690, 10.1128/MCB.24.4.1680-1690.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Shafi R., Iyer S. P., Ellies L. G., et al., “The O‐Glcnac Transferase Gene Resides on the X Chromosome and Is Essential for Embryonic Stem Cell Viability and Mouse Ontogeny,” Proceedings of the National Academy of Sciences of the United States of America 97, no. 11 (2000): 5735–5739, 10.1073/pnas.100471497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Yang Y. R., Song M., Lee H., et al., “O‐Glcnacase Is Essential for Embryonic Development and Maintenance of Genomic Stability,” Aging Cell 11, no. 3 (2012): 439–448, 10.1111/j.1474-9726.2012.00801.x. [DOI] [PubMed] [Google Scholar]
- 25. Wang T., Birsoy K., Hughes N. W., et al., “Identification and Characterization of Essential Genes in the Human Genome,” Science 350, no. 6264 (2015): 1096–1101, 10.1126/science.aac7041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Hardiville S. and Hart G. W., “Nutrient Regulation of Gene Expression by O‐Glcnacylation of Chromatin,” Current Opinion in Chemical Biology 33 (2016): 88–94, 10.1016/j.cbpa.2016.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. He X. F., Hu X., Wen G. J., et al., “O‐Glcnacylation in Cancer Development and Immunotherapy,” Cancer Letters 566 (2023): 216258, 10.1016/j.canlet.2023.216258. [DOI] [PubMed] [Google Scholar]
- 28. Lee B. E., Suh P. G., and Kim J. I., “O‐Glcnacylation in Health and Neurodegenerative Diseases,” Experimental and Molecular Medicine 53, no. 11 (2021): 1674–1682, 10.1038/s12276-021-00709-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. He X. F., Hu X., Wen G. J., et al., “O‐Glcnacylation in Cancer Development and Immunotherapy,” Cancer Letters 566 (2023): 216258, 10.1016/j.canlet.2023.216258. [DOI] [PubMed] [Google Scholar]
- 30. Chatham J. C., Zhang J., and Wende A. R., “Role of O‐Linked N‐Acetylglucosamine Protein Modification in Cellular (Patho)Physiology,” Physiological Reviews 101, no. 2 (2021): 427–493, 10.1152/physrev.00043.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Zhu Q., Zhou L., Yang Z., et al., “O‐Glcnacylation Plays a Role in Tumor Recurrence of Hepatocellular Carcinoma Following Liver Transplantation,” Medical Oncology 29, no. 2 (2012): 985–993, 10.1007/s12032-011-9912-1. [DOI] [PubMed] [Google Scholar]
- 32. Mukherjee M. M., Biesbrock D., and Hanover J. A., “Galectin‐3: Integrator of Signaling via Hexosamine Flux,” Biomolecules 15, no. 7 (2025), 10.3390/biom15071028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Levine Z. G. and Walker S., “The Biochemistry of O‐Glcnac Transferase: Which Functions Make It Essential in Mammalian Cells?,” Annual Review of Biochemistry 85 (2016): 631–657, 10.1146/annurev-biochem-060713-035344. [DOI] [PubMed] [Google Scholar]
- 34. Shafi R., Iyer S. P., Ellies L. G., et al., “The O‐Glcnac Transferase Gene Resides on the X Chromosome and Is Essential for Embryonic Stem Cell Viability and Mouse Ontogeny,” Proceedings of the National Academy of Sciences of the United States of America 97, no. 11 (2000): 5735–5739, 10.1073/pnas.100471497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Hardiville S. and Hart G. W., “Nutrient Regulation of Signaling, Transcription, and Cell Physiology by O‐Glcnacylation,” Cell Metabolism 20, no. 2 (2014): 208–213, 10.1016/j.cmet.2014.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Lewis B. A. and Hanover J. A., “O‐Glcnac and the Epigenetic Regulation of Gene Expression,” Journal of Biological Chemistry 289, no. 50 (2014): 34440–34448, 10.1074/jbc.R114.595439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Parker M. P., Peterson K. R., and Slawson C., “O‐Glcnacylation and O‐Glcnac Cycling Regulate Gene Transcription: Emerging Roles in Cancer,” Cancers 13, no. 7 (2021), 10.3390/cancers13071666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Ranuncolo S. M., Ghosh S., Hanover J. A., et al., “Evidence of the Involvement of O‐Glcnac‐Modified Human Rna Polymerase II Ctd in Transcription in Vitro and in Vivo,” Journal of Biological Chemistry 287, no. 28 (2012): 23549–23561, 10.1074/jbc.M111.330910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Lewis B. A., Burlingame A. L., and Myers S. A., “Human Rna Polymerase II Promoter Recruitment in Vitro Is Regulated by O‐Linked N‐Acetylglucosaminyltransferase (Ogt),” Journal of Biological Chemistry 291, no. 27 (2016): 14056–14061, 10.1074/jbc.M115.684365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Sakabe K., Wang Z., and Hart G. W., “Beta‐N‐Acetylglucosamine (O‐Glcnac) Is Part of the Histone Code,” Proceedings of the National Academy of Sciences of the United States of America 107, no. 46 (2010): 19915–19920, 10.1073/pnas.1009023107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Zhang S., Roche K., Nasheuer H., et al., “Modification of Histones by Sugar Beta‐N‐Acetylglucosamine (Glcnac) Occurs on Multiple Residues, Including Histone H3 Serine 10, and Is Cell Cycle‐Regulated,” Journal of Biological Chemistry 286, no. 43 (2011): 37483–37495, 10.1074/jbc.M111.284885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Fong J. J., Nguyen B. L., Bridger R., et al., “Beta‐N‐Acetylglucosamine (O‐Glcnac) Is a Novel Regulator of Mitosis‐Specific Phosphorylations on Histone H3,” Journal of Biological Chemistry 287, no. 15 (2012): 12195–12203, 10.1074/jbc.M111.315804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Chen Q., Chen Y., Bian C., et al., “Tet2 Promotes Histone O‐Glcnacylation during Gene Transcription,” Nature 493, no. 7433 (2013): 561–564, 10.1038/nature11742. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Xu Q., Yang C., Du Y., et al., “Ampk Regulates Histone H2B O‐Glcnacylation,” Nucleic Acids Research 42, no. 9 (2014): 5594–5604, 10.1093/nar/gku236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Delporte A., De Zaeytijd J., De Storme N., et al., “Cell Cycle‐Dependent O‐Glcnac Modification of Tobacco Histones and Their Interaction With the Tobacco Lectin,” Plant Physiology and Biochemistry 83 (2014): 151–158, 10.1016/j.plaphy.2014.07.021. [DOI] [PubMed] [Google Scholar]
- 46. Dehennaut V., Leprince D., and Lefebvre T., “O‐Glcnacylation, an Epigenetic Mark. Focus on the Histone Code, Tet Family Proteins, and Polycomb Group Proteins,” Frontiers in Endocrinology 5 (2014): 155, 10.3389/fendo.2014.00155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Wang P., Peng C., Liu X., et al., “Ogt Mediated Histone H2B S112 Glcnacylation Regulates Dna Damage Response,” Journal of Genetics and Genomics 42, no. 9 (2015): 467–475, 10.1016/j.jgg.2015.07.002. [DOI] [PubMed] [Google Scholar]
- 48. Leturcq M., Lefebvre T., and Vercoutter‐Edouart A., “O‐Glcnacylation and Chromatin Remodeling in Mammals: An up‐to‐Date Overview,” Biochemical Society Transactions 45, no. 2 (2017): 323–338, 10.1042/BST20160388. [DOI] [PubMed] [Google Scholar]
- 49. Fujiki R., Hashiba W., Sekine H., et al., “Glcnacylation of Histone H2B Facilitates Its Monoubiquitination,” Nature 480, no. 7378 (2011): 557–560, 10.1038/nature10656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Hardiville S., Banerjee P. S., Selen Alpergin E. S., et al., “Tata‐Box Binding Protein O‐Glcnacylation at T114 Regulates Formation of the B‐Tfiid Complex and Is Critical for Metabolic Gene Regulation,” Molecular Cell 77, no. 5 (2020): 1143–1152, 10.1016/j.molcel.2019.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Noach N., Segev Y., Levi I., et al., “Modification of Topoisomerase I Activity by Glucose and by O‐Glcnacylation of the Enzyme Protein,” Glycobiology 17, no. 12 (2007): 1357–1364, 10.1093/glycob/cwm105. [DOI] [PubMed] [Google Scholar]
- 52. Eustice M., Bond M. R., and Hanover J. A., “O‐Glcnac Cycling and the Regulation of Nucleocytoplasmic Dynamics,” Biochemical Society Transactions 45, no. 2 (2017): 427–436, 10.1042/BST20160171. [DOI] [PubMed] [Google Scholar]
- 53. Butkinaree C., Park K., and Hart G. W., “Extensive Crosstalk With Phosphorylation to Regulate Signaling and Transcription in Response to Nutrients and Stress,” Biochimica Et Biophysica Acta 1800, no. 2 (2010): 96–106, 10.1016/j.bbagen.2009.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Ozcan S., Andrali S. S., and Cantrell J. E. L., “Modulation of Transcription Factor Function by O‐Glcnac Modification,” Biochimica Et Biophysica Acta 1799, no. 5–6 (2010): 353–364, 10.1016/j.bbagrm.2010.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Housley M. P., Rodgers J. T., Udeshi N. D., et al., “O‐Glcnac Regulates Foxo Activation in Response to Glucose,” Journal of Biological Chemistry 283, no. 24 (2008): 16283–16292, 10.1074/jbc.M802240200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Kuo M., Zilberfarb V., Gangneux N., et al., “O‐Glycosylation of Foxo1 Increases Its Transcriptional Activity towards the Glucose 6‐Phosphatase Gene,” Febs Letters 582, no. 5 (2008): 829–834, 10.1016/j.febslet.2008.02.010. [DOI] [PubMed] [Google Scholar]
- 57. Fardini Y., Masson E., Boudah O., et al., “O‐Glcnacylation of Foxo1 in Pancreatic Beta Cells Promotes Akt Inhibition Through an Igfbp1‐Mediated Autocrine Mechanism,” Faseb Journal 28, no. 2 (2014): 1010–1021, 10.1096/fj.13-238378. [DOI] [PubMed] [Google Scholar]
- 58. Choe S., Ladam F., and Sagerstrom C. G., “Tale Factors Poise Promoters for Activation by Hox Proteins,” Developmental Cell 28, no. 2 (2014): 203–211, 10.1016/j.devcel.2013.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Draime A., Bridoux L., Belpaire M., et al., “The O‐Glcnac Transferase Ogt Interacts With and Post‐Translationally Modifies the Transcription Factor Hoxa1,” Febs Letters 592, no. 7 (2018): 1185–1201, 10.1002/1873-3468.13015. [DOI] [PubMed] [Google Scholar]
- 60. Jackson S. P. and Tjian R., “O‐Glycosylation of Eukaryotic Transcription Factors: Implications for Mechanisms of Transcriptional Regulation,” Cell 55, no. 1 (1988): 125–133, 10.1016/0092-8674(88)90015-3. [DOI] [PubMed] [Google Scholar]
- 61. Lichtsteiner S. and Schibler U., “A Glycosylated Liver‐Specific Transcription Factor Stimulates Transcription of the Albumin Gene,” Cell 57, no. 7 (1989): 1179–1187, 10.1016/0092-8674(89)90055-X. [DOI] [PubMed] [Google Scholar]
- 62. Chou T. Y., Hart G. W., and Dang C. V., “C‐Myc Is Glycosylated at Threonine 58, a Known Phosphorylation Site and a Mutational Hot Spot in Lymphomas,” Journal of Biological Chemistry 270, no. 32 (1995): 18961–18965, 10.1074/jbc.270.32.18961. [DOI] [PubMed] [Google Scholar]
- 63. Chou T. Y., Dang C. V., and Hart G. W., “Glycosylation of the C‐Myc Transactivation Domain,” Proceedings of the National Academy of Sciences of the United States of America 92, no. 10 (1995): 4417–4421, 10.1073/pnas.92.10.4417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Reason A. J., Morris H. R., Panico M., et al., “Localization of O‐Glcnac Modification on the Serum Response Transcription Factor,” Journal of Biological Chemistry 267, no. 24 (1992): 16911–16921, 10.1016/S0021-9258(18)41871-6. [DOI] [PubMed] [Google Scholar]
- 65. Comer F. I. and Hart G. W., “O‐Glcnac and the Control of Gene Expression,” Biochimica Et Biophysica Acta 1473, no. 1 (1999): 161–171, 10.1016/S0304-4165(99)00176-2. [DOI] [PubMed] [Google Scholar]
- 66. Tan Z., Fei G., Paulo J. A., et al., “O‐Glcnac Regulates Gene Expression by Controlling Detained Intron Splicing,” Nucleic Acids Research 48, no. 10 (2020): 5656–5669, 10.1093/nar/gkaa263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Mathew R., Hartmuth K., Mohlmann S., et al., “Phosphorylation of Human Prp28 by Srpk2 Is Required for Integration of the U4/U6‐U5 Tri‐Snrnp into the Spliceosome,” Nature Structural & Molecular Biology 15, no. 5 (2008): 435–443, 10.1038/nsmb.1415. [DOI] [PubMed] [Google Scholar]
- 68. Wang H. Y., Lin W., Dyck J. A., et al., “Srpk2: A Differentially Expressed Sr Protein‐Specific Kinase Involved in Mediating the Interaction and Localization of Pre‐Mrna Splicing Factors in Mammalian Cells,” Journal of Cell Biology 140, no. 4 (1998): 737–750, 10.1083/jcb.140.4.737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Tan W., Jiang P., Zhang W., et al., “Posttranscriptional Regulation of De Novo Lipogenesis by Glucose‐Induced O‐Glcnacylation,” Molecular Cell 81, no. 9 (2021): 1890–1904, 10.1016/j.molcel.2021.02.009. [DOI] [PubMed] [Google Scholar]
- 70. Bi Y., Deng Z., Ni W., et al., “Arabidopsis Acinus Is O‐Glycosylated and Regulates Transcription and Alternative Splicing of Regulators of Reproductive Transitions,” Nature Communications 12, no. 1 (2021): 945, 10.1038/s41467-021-20929-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Zhao M., Yao X., Wei P., et al., “O‐Glcnacylation of Tdp‐43 Suppresses Proteinopathies and Promotes Tdp‐43'S Mrna Splicing Activity,” Embo Reports 22, no. 6 (2021): e51649, 10.15252/embr.202051649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Wang J., Liu N., Wei X., et al., “Regulation of Auf1 Alternative Splicing by Hnrnpa1 and Srsf2 Modulate the Sensitivity of Ovarian Cancer Cells to Cisplatin,” Cellular Oncology 47, no. 6 (2024): 2349–2366, 10.1007/s13402-024-01023-8. [DOI] [PubMed] [Google Scholar]
- 73. Zeidan Q., Wang Z., De Maio A., et al., “O‐Glcnac Cycling Enzymes Associate With the Translational Machinery and Modify Core Ribosomal Proteins,” Molecular Biology of the Cell 21, no. 12 (2010): 1922–1936, 10.1091/mbc.e09-11-0941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Hart G. W., “Nutrient Regulation of Signaling and Transcription,” Journal of Biological Chemistry 294, no. 7 (2019): 2211–2231, 10.1074/jbc.AW119.003226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Anderson P. and Kedersha N., “Rna Granules,” Journal of Cell Biology 172, no. 6 (2006): 803–808, 10.1083/jcb.200512082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Anderson P. and Kedersha N., “Stress Granules: The Tao of Rna Triage,” Trends in Biochemical Sciences 33, no. 3 (2008): 141–150, 10.1016/j.tibs.2007.12.003. [DOI] [PubMed] [Google Scholar]
- 77. Kedersha N. and Anderson P., “Mammalian Stress Granules and Processing Bodies,” Methods in Enzymology 431 (2007): 61–81. [DOI] [PubMed] [Google Scholar]
- 78. Parker R. and Sheth U., “P Bodies and the Control of Mrna Translation and Degradation,” Molecular Cell 25, no. 5 (2007): 635–646, 10.1016/j.molcel.2007.02.011. [DOI] [PubMed] [Google Scholar]
- 79. Eulalio A., Behm‐Ansmant I., and Izaurralde E., “P Bodies: At the Crossroads of Post‐Transcriptional Pathways,” Nature Reviews Molecular Cell Biology 8, no. 1 (2007): 9–22, 10.1038/nrm2080. [DOI] [PubMed] [Google Scholar]
- 80. Ohn T., Kedersha N., Hickman T., et al., “A Functional Rnai Screen Links O‐Glcnac Modification of Ribosomal Proteins to Stress Granule and Processing Body Assembly,” Nature Cell Biology 10, no. 10 (2008): 1224–1231, 10.1038/ncb1783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Malumbres M. and Barbacid M., “Cell Cycle, Cdks and Cancer: A Changing Paradigm,” Nature Reviews Cancer 9, no. 3 (2009): 153–166, 10.1038/nrc2602. [DOI] [PubMed] [Google Scholar]
- 82. Slawson C., Zachara N. E., Vosseller K., et al., “Perturbations in O‐Linked Beta‐N‐Acetylglucosamine Protein Modification Cause Severe Defects in Mitotic Progression and Cytokinesis,” Journal of Biological Chemistry 280, no. 38 (2005): 32944–32956, 10.1074/jbc.M503396200. [DOI] [PubMed] [Google Scholar]
- 83. Dehennaut V., Lefebvre T., Sellier C., et al., “O‐Linked N‐Acetylglucosaminyltransferase Inhibition Prevents G2/M Transition in Xenopus Laevis Oocytes,” Journal of Biological Chemistry 282, no. 17 (2007): 12527–12536, 10.1074/jbc.M700444200. [DOI] [PubMed] [Google Scholar]
- 84. Masclef L., Dehennaut V., Mortuaire M., et al., “Cyclin D1 Stability Is Partly Controlled by O‐Glcnacylation,” Frontiers in Endocrinology 10 (2019): 106, 10.3389/fendo.2019.00106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Olivier‐Van Stichelen S., Dehennaut V., Buzy A., et al., “O‐Glcnacylation Stabilizes Beta‐Catenin Through Direct Competition With Phosphorylation at Threonine 41,” Faseb Journal 28, no. 8 (2014): 3325–3338, 10.1096/fj.13-243535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Olivier‐Van Stichelen S., Drougat L., Dehennaut V., et al., “Serum‐Stimulated Cell Cycle Entry Promotes Ncogt Synthesis Required for Cyclin D Expression,” Oncogenesis 1, no. 12 (2012): e36, 10.1038/oncsis.2012.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Mitchell C. W., Czajewski I., and van Aalten D. M. F., “Bioinformatic Prediction of Putative Conveyers of O‐Glcnac Transferase Intellectual Disability,” Journal of Biological Chemistry 298, no. 9 (2022): 102276, 10.1016/j.jbc.2022.102276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Mitchell C. W., Yuan H., Sonderstrup‐Jensen M., et al., “O‐Glcnacylation of the Intellectual Disability Protein Ddx3X Exerts Proteostatic Cell Cycle Control,” Open Biology 15, no. 7 (2025): 250064, 10.1098/rsob.250064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Julien E. and Herr W., “Proteolytic Processing Is Necessary to Separate and Ensure Proper Cell Growth and Cytokinesis Functions of Hcf‐1,” Embo Journal 22, no. 10 (2003): 2360–2369, 10.1093/emboj/cdg242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Woo C. M., Lund P. J., Huang A. C., et al., “Mapping and Quantification of over 2000 O‐Linked Glycopeptides in Activated Human T Cells With Isotope‐Targeted Glycoproteomics (Isotag),” Molecular & Cellular Proteomics 17, no. 4 (2018): 764–775, 10.1074/mcp.RA117.000261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Wang Z., Udeshi N. D., Slawson C., et al., “Extensive Crosstalk between O‐Glcnacylation and Phosphorylation Regulates Cytokinesis,” Science Signaling 3, no. 104 (2010): ra2, 10.1126/scisignal.2000526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Dehennaut V., Slomianny M., Page A., et al., “Identification of Structural and Functional O‐Linked N‐Acetylglucosamine‐Bearing Proteins in Xenopus Laevis Oocyte,” Molecular & Cellular Proteomics 7, no. 11 (2008): 2229–2245, 10.1074/mcp.M700494-MCP200. [DOI] [PubMed] [Google Scholar]
- 93. Slawson C. and Duncan F. E., “Sweet Action: The Dynamics of O‐Glcnacylation during Meiosis in Mouse Oocytes,” Molecular Reproduction and Development 82, no. 12 (2015): 915, 10.1002/mrd.22577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Sakabe K. and Hart G. W., “O‐Glcnac Transferase Regulates Mitotic Chromatin Dynamics,” Journal of Biological Chemistry 285, no. 45 (2010): 34460–34468, 10.1074/jbc.M110.158170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Tan E. P., Caro S., Potnis A., et al., “O‐Linked N‐Acetylglucosamine Cycling Regulates Mitotic Spindle Organization,” Journal of Biological Chemistry 288, no. 38 (2013): 27085–27099, 10.1074/jbc.M113.470187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Slawson C., Lakshmanan T., Knapp S., et al., “A Mitotic Glcnacylation/Phosphorylation Signaling Complex Alters the Posttranslational State of the Cytoskeletal Protein Vimentin,” Molecular Biology of the Cell 19, no. 10 (2008): 4130–4140, 10.1091/mbc.e07-11-1146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Fisher D., Krasinska L., Coudreuse D., et al., “Phosphorylation Network Dynamics in the Control of Cell Cycle Transitions,” Journal of Cell Science 125, no. Pt 20 (2012): 4703–4711, 10.1242/jcs.106351. [DOI] [PubMed] [Google Scholar]
- 98. Lanza C., Tan E. P., Zhang Z., et al., “Reduced O‐Glcnacase Expression Promotes Mitotic Errors and Spindle Defects,” Cell Cycle 15, no. 10 (2016): 1363–1375, 10.1080/15384101.2016.1167297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Ruan H., Han X., Li M., et al., “O‐Glcnac Transferase/Host Cell Factor C1 Complex Regulates Gluconeogenesis by Modulating Pgc‐1Alpha Stability,” Cell Metabolism 16, no. 2 (2012): 226–237, 10.1016/j.cmet.2012.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Kim D., Ryu D., Koh M., et al., “Orphan Nuclear Receptor Estrogen‐Related Receptor Gamma (Errgamma) Is Key Regulator of Hepatic Gluconeogenesis,” Journal of Biological Chemistry 287, no. 26 (2012): 21628–21639, 10.1074/jbc.M111.315168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Misra J., Kim D., Jung Y. S., et al., “O‐Glcnacylation of Orphan Nuclear Receptor Estrogen‐Related Receptor Gamma Promotes Hepatic Gluconeogenesis,” Diabetes 65, no. 10 (2016): 2835–2848, 10.2337/db15-1523. [DOI] [PubMed] [Google Scholar]
- 102. Urasaki Y., Pizzorno G., and Le T. T., “Chronic Uridine Administration Induces Fatty Liver and Pre‐Diabetic Conditions in Mice,” PLoS ONE 11, no. 1 (2016): e0146994, 10.1371/journal.pone.0146994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Dentin R., Hedrick S., Xie J., et al., “Hepatic Glucose Sensing via the Creb Coactivator Crtc2,” Science 319, no. 5868 (2008): 1402–1405, 10.1126/science.1151363. [DOI] [PubMed] [Google Scholar]
- 104. Housley M. P., Rodgers J. T., Udeshi N. D., et al., “O‐Glcnac Regulates Foxo Activation in Response to Glucose,” Journal of Biological Chemistry 283, no. 24 (2008): 16283–16292, 10.1074/jbc.M802240200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Housley M. P., Udeshi N. D., Rodgers J. T., et al., “A Pgc‐1Alpha‐O‐Glcnac Transferase Complex Regulates Foxo Transcription Factor Activity in Response to Glucose,” Journal of Biological Chemistry 284, no. 8 (2009): 5148–5157, 10.1074/jbc.M808890200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Kuo M., Zilberfarb V., Gangneux N., et al., “O‐Glcnac Modification of Foxo1 Increases Its Transcriptional Activity: A Role in the Glucotoxicity Phenomenon?,” Biochimie 90, no. 5 (2008): 679–685, 10.1016/j.biochi.2008.03.005. [DOI] [PubMed] [Google Scholar]
- 107. Babu D. A., Deering T. G., and Mirmira R. G., “A Feat of Metabolic Proportions: Pdx1 Orchestrates Islet Development and Function in the Maintenance of Glucose Homeostasis,” Molecular Genetics and Metabolism 92, no. 1–2 (2007): 43–55, 10.1016/j.ymgme.2007.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Gao Y., Miyazaki J., and Hart G. W., “The Transcription Factor Pdx‐1 Is Post‐Translationally Modified by O‐Linked N‐Acetylglucosamine and this Modification Is Correlated With Its Dna Binding Activity and Insulin Secretion in Min6 Beta‐Cells,” Archives of Biochemistry and Biophysics 415, no. 2 (2003): 155–163, 10.1016/S0003-9861(03)00234-0. [DOI] [PubMed] [Google Scholar]
- 109. Chae J. H., Stein G. H., and Lee J. E., “Neurod: The Predicted and the Surprising,” Molecules and Cells 18, no. 3 (2004): 271–288, 10.1016/S1016-8478(23)13113-X. [DOI] [PubMed] [Google Scholar]
- 110. Petersen H. V., Jensen J. N., Stein R., et al., “Glucose Induced Mapk Signalling Influences Neurod1‐Mediated Activation and Nuclear Localization,” Febs Letters 528, no. 1–3 (2002): 241–245, 10.1016/S0014-5793(02)03318-5. [DOI] [PubMed] [Google Scholar]
- 111. Andrali S. S., Qian Q., and Ozcan S., “Glucose Mediates the Translocation of Neurod1 by O‐Linked Glycosylation,” Journal of Biological Chemistry 282, no. 21 (2007): 15589–15596, 10.1074/jbc.M701762200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Cheng X. and Case L. B., “Phase Separation in Chemical and Mechanical Signal Transduction,” Current Opinion in Cell Biology 85 (2023): 102243, 10.1016/j.ceb.2023.102243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Ullo M. F. and Case L. B., “How Cells Sense and Integrate Information from Different Sources,” Wires Mechanisms of Disease 15, no. 4 (2023): e1604, 10.1002/wsbm.1604. [DOI] [PubMed] [Google Scholar]
- 114. Wootten D., Christopoulos A., Marti‐Solano M., et al., “Mechanisms of Signalling and Biased Agonism in G Protein‐Coupled Receptors,” Nature Reviews Molecular Cell Biology 19, no. 10 (2018): 638–653, 10.1038/s41580-018-0049-3. [DOI] [PubMed] [Google Scholar]
- 115. Hauser A. S., Attwood M. M., Rask‐Andersen M., et al., “Trends in Gpcr Drug Discovery: New Agents, Targets and Indications,” Nature Reviews Drug Discovery 16, no. 12 (2017): 829–842, 10.1038/nrd.2017.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Logan C. Y. and Nusse R., “The Wnt Signaling Pathway in Development and Disease,” Annual Review of Cell and Developmental Biology 20 (2004): 781–810, 10.1146/annurev.cellbio.20.010403.113126. [DOI] [PubMed] [Google Scholar]
- 117. Steinhart Z. and Angers S., “Wnt Signaling in Development and Tissue Homeostasis,” Development (Cambridge, England) 145, no. 11 (2018), 10.1242/dev.146589. [DOI] [PubMed] [Google Scholar]
- 118. Shafi R., Iyer S. P., Ellies L. G., et al., “The O‐Glcnac Transferase Gene Resides on the X Chromosome and Is Essential for Embryonic Stem Cell Viability and Mouse Ontogeny,” Proceedings of the National Academy of Sciences of the United States of America 97, no. 11 (2000): 5735–5739, 10.1073/pnas.100471497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Hart M., Concordet J. P., Lassot I., et al., “The F‐Box Protein Beta‐Trcp Associates With Phosphorylated Beta‐Catenin and Regulates Its Activity in the Cell,” Current Biology 9, no. 4 (1999): 207–210, 10.1016/S0960-9822(99)80091-8. [DOI] [PubMed] [Google Scholar]
- 120. Liu C., Li Y., Semenov M., et al., “Control of Beta‐Catenin Phosphorylation/Degradation by a Dual‐Kinase Mechanism,” Cell 108, no. 6 (2002): 837–847, 10.1016/S0092-8674(02)00685-2. [DOI] [PubMed] [Google Scholar]
- 121. Zhu Y. and Hart G. W., “Dual‐Specificity Rna Aptamers Enable Manipulation of Target‐Specific O‐Glcnacylation and Unveil Functions of O‐Glcnac on Beta‐Catenin,” Cell 186, no. 2 (2023): 428–445, 10.1016/j.cell.2022.12.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Olivier‐Van S. S., Dehennaut V., Buzy A., et al., “O‐Glcnacylation Stabilizes Beta‐Catenin Through Direct Competition With Phosphorylation at Threonine 41,” Faseb Journal 28, no. 8 (2014): 3325–3338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Li V. S., Ng S. S., Boersema P. J., et al., “Wnt Signaling Through Inhibition of Beta‐Catenin Degradation in an Intact Axin1 Complex,” Cell 149, no. 6 (2012): 1245–1256, 10.1016/j.cell.2012.05.002. [DOI] [PubMed] [Google Scholar]
- 124. Du Z. and Lovly C. M., “Mechanisms of Receptor Tyrosine Kinase Activation in Cancer,” Molecular Cancer 17, no. 1 (2018): 58, 10.1186/s12943-018-0782-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Hua Q., “Insulin: A Small Protein With a Long Journey,” Protein & Cell 1, no. 6 (2010): 537–551, 10.1007/s13238-010-0069-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Yang X., Ongusaha P. P., Miles P. D., et al., “Phosphoinositide Signalling Links O‐Glcnac Transferase to Insulin Resistance,” Nature 451, no. 7181 (2008): 964–969, 10.1038/nature06668. [DOI] [PubMed] [Google Scholar]
- 127. Sabio G. and Davis R. J., “Tnf and Map Kinase Signalling Pathways,” Seminars in Immunology 26, no. 3 (2014): 237–245, 10.1016/j.smim.2014.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Keshet Y. and Seger R., “The Map Kinase Signaling Cascades: A System of Hundreds of Components Regulates a Diverse Array of Physiological Functions,” Methods in Molecular Biology 661 (2010): 3–38. [DOI] [PubMed] [Google Scholar]
- 129. Nelson Z. M., Leonard G. D., and Fehl C., “Tools for Investigating O‐Glcnac in Signaling and Other Fundamental Biological Pathways,” Journal of Biological Chemistry 300, no. 2 (2024): 105615, 10.1016/j.jbc.2023.105615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Leonard W. J. and Lin J. X., “Cytokine Receptor Signaling Pathways,” Journal of Allergy and Clinical Immunology 105, no. 5 (2000): 877–888, 10.1067/mai.2000.106899. [DOI] [PubMed] [Google Scholar]
- 131. Qiang A., Slawson C., and Fields P. E., “The Role of O‐Glcnacylation in Immune Cell Activation,” Frontiers in Endocrinology 12 (2021): 596617, 10.3389/fendo.2021.596617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Zinatizadeh M. R., Schock B., Chalbatani G. M., et al., “The Nuclear Factor Kappa B (Nf‐Kb) Signaling in Cancer Development and Immune Diseases,” Genes & Diseases 8, no. 3 (2021): 287–297, 10.1016/j.gendis.2020.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Yang W. H., Park S. Y., Nam H. W., et al., “Nfkappab Activation Is Associated With Its O‐Glcnacylation State under Hyperglycemic Conditions,” Proceedings of the National Academy of Sciences of the United States of America 105, no. 45 (2008): 17345–17350, 10.1073/pnas.0806198105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Pathak S., Borodkin V. S., Albarbarawi O., et al., “O‐Glcnacylation of Tab1 Modulates Tak1‐Mediated Cytokine Release,” Embo Journal 31, no. 6 (2012): 1394–1404, 10.1038/emboj.2012.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Wang Q., Fang P., He R., et al., “O‐Glcnac Transferase Promotes Influenza a Virus‐Induced Cytokine Storm by Targeting Interferon Regulatory Factor‐5,” Science Advances 6, no. 16 (2020): eaaz7086, 10.1126/sciadv.aaz7086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Zhu Q., Wang H., Chai S., et al., “O‐Glcnacylation Promotes Tumor Immune Evasion by Inhibiting Pd‐L1 Lysosomal Degradation,” Proceedings of the National Academy of Sciences of the United States of America 120, no. 13 (2023): e2078171176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Zhang J., Ten D. P., Wuhrer M., et al., “Role of Glycosylation in Tgf‐Beta Signaling and Epithelial‐to‐Mesenchymal Transition in Cancer,” Protein & Cell 12, no. 2 (2021): 89–106, 10.1007/s13238-020-00741-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Cole R. N. and Hart G. W., “Cytosolic O‐Glycosylation Is Abundant in Nerve Terminals,” Journal of Neurochemistry 79, no. 5 (2001): 1080–1089, 10.1046/j.1471-4159.2001.00655.x. [DOI] [PubMed] [Google Scholar]
- 139. Erickson J. R., Pereira L., Wang L., et al., “Diabetic Hyperglycaemia Activates Camkii and Arrhythmias by O‐Linked Glycosylation,” Nature 502, no. 7471 (2013): 372–376, 10.1038/nature12537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Vosseller K., Trinidad J. C., Chalkley R. J., et al., “O‐Linked N‐Acetylglucosamine Proteomics of Postsynaptic Density Preparations Using Lectin Weak Affinity Chromatography and Mass Spectrometry,” Molecular & Cellular Proteomics 5, no. 5 (2006): 923–934, 10.1074/mcp.T500040-MCP200. [DOI] [PubMed] [Google Scholar]
- 141. Khidekel N., Ficarro S. B., Peters E. C., et al., “Exploring the O‐Glcnac Proteome: Direct Identification of O‐Glcnac‐Modified Proteins from the Brain,” Proceedings of the National Academy of Sciences of the United States of America 101, no. 36 (2004): 13132–13137, 10.1073/pnas.0403471101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Taylor E. W., Wang K., Nelson A. R., et al., “O‐Glcnacylation of Ampa Receptor Glua2 Is Associated With a Novel Form of Long‐Term Depression at Hippocampal Synapses,” Journal of Neuroscience 34, no. 1 (2014): 10–21, 10.1523/JNEUROSCI.4761-12.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Dias W. B., Cheung W. D., Wang Z., et al., “Regulation of Calcium/Calmodulin‐Dependent Kinase IV by O‐Glcnac Modification,” Journal of Biological Chemistry 284, no. 32 (2009): 21327–21337, 10.1074/jbc.M109.007310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Rexach J. E., Clark P. M., Mason D. E., et al., “Dynamic O‐Glcnac Modification Regulates Creb‐Mediated Gene Expression and Memory Formation,” Nature Chemical Biology 8, no. 3 (2012): 253–261, 10.1038/nchembio.770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Breijyeh Z. and Karaman R., “Comprehensive Review on Alzheimer's Disease: Causes and Treatment,” Molecules (Basel, Switzerland) 25, no. 24 (2020), 10.3390/molecules25245789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Liu F., Shi J., Tanimukai H., et al., “Reduced O‐Glcnacylation Links Lower Brain Glucose Metabolism and Tau Pathology in Alzheimer's Disease,” Brain 132, no. Pt 7 (2009): 1820–1832, 10.1093/brain/awp099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Balana A. T., Levine P. M., Craven T. W., et al., “O‐Glcnac Modification of Small Heat Shock Proteins Enhances Their Anti‐Amyloid Chaperone Activity,” Nature Chemistry 13, no. 5 (2021): 441–450, 10.1038/s41557-021-00648-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Park J., Ha H. J., Chung E. S., et al., “O‐Glcnacylation Ameliorates the Pathological Manifestations of Alzheimer's Disease by Inhibiting Necroptosis,” Science Advances 7, no. 3 (2021), 10.1126/sciadv.abd3207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Liu F., Iqbal K., Grundke‐Iqbal I., et al., “O‐Glcnacylation Regulates Phosphorylation of Tau: A Mechanism Involved in Alzheimer's Disease,” Proceedings of the National Academy of Sciences of the United States of America 101, no. 29 (2004): 10804–10809, 10.1073/pnas.0400348101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Arnold C. S., Johnson G. V., Cole R. N., et al., “The Microtubule‐Associated Protein Tau Is Extensively Modified With O‐Linked N‐Acetylglucosamine,” Journal of Biological Chemistry 271, no. 46 (1996): 28741–28744, 10.1074/jbc.271.46.28741. [DOI] [PubMed] [Google Scholar]
- 151. Yuzwa S. A., Yadav A. K., Skorobogatko Y., et al., “Mapping O‐Glcnac Modification Sites on Tau and Generation of a Site‐Specific O‐Glcnac Tau Antibody,” Amino Acids 40, no. 3 (2011): 857–868, 10.1007/s00726-010-0705-1. [DOI] [PubMed] [Google Scholar]
- 152. Iqbal K., Alonso A. C., Chen S., et al., “Tau Pathology in Alzheimer Disease and Other Tauopathies,” Biochimica Et Biophysica Acta 1739, no. 2–3 (2005): 198–210, 10.1016/j.bbadis.2004.09.008. [DOI] [PubMed] [Google Scholar]
- 153. Kopeikina K. J., Hyman B. T., and Spires‐Jones T. L., “Soluble Forms of Tau Are Toxic in Alzheimer's Disease,” Translational Neuroscience 3, no. 3 (2012): 223–233, 10.2478/s13380-012-0032-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Yuzwa S. A., Macauley M. S., Heinonen J. E., et al., “A Potent Mechanism‐Inspired O‐Glcnacase Inhibitor That Blocks Phosphorylation of Tau in Vivo,” Nature Chemical Biology 4, no. 8 (2008): 483–490, 10.1038/nchembio.96. [DOI] [PubMed] [Google Scholar]
- 155. Yuzwa S. A., Shan X., Macauley M. S., et al., “Increasing O‐Glcnac Slows Neurodegeneration and Stabilizes Tau against Aggregation,” Nature Chemical Biology 8, no. 4 (2012): 393–399, 10.1038/nchembio.797. [DOI] [PubMed] [Google Scholar]
- 156. Lammich S., Kojro E., Postina R., et al., “Constitutive and Regulated Alpha‐Secretase Cleavage of Alzheimer's Amyloid Precursor Protein by a Disintegrin Metalloprotease,” Proceedings of the National Academy of Sciences of the United States of America 96, no. 7 (1999): 3922–3927, 10.1073/pnas.96.7.3922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Vassar R., Bennett B. D., Babu‐Khan S., et al., “Beta‐Secretase Cleavage of Alzheimer's Amyloid Precursor Protein by the Transmembrane Aspartic Protease Bace,” Science 286, no. 5440 (1999): 735–741, 10.1126/science.286.5440.735. [DOI] [PubMed] [Google Scholar]
- 158. Evin G., Cappai R., Li Q. X., et al., “Candidate Gamma‐Secretases in the Generation of the Carboxyl Terminus of the Alzheimer's Disease Beta a4 Amyloid: Possible Involvement of Cathepsin D,” Biochemistry 34, no. 43 (1995): 14185–14192, 10.1021/bi00043a024. [DOI] [PubMed] [Google Scholar]
- 159. De Strooper B., Saftig P., Craessaerts K., et al., “Deficiency of Presenilin‐1 Inhibits the Normal Cleavage of Amyloid Precursor Protein,” Nature 391, no. 6665 (1998): 387–390, 10.1038/34910. [DOI] [PubMed] [Google Scholar]
- 160. Busche M. A. and Hyman B. T., “Synergy between Amyloid‐Beta and Tau in Alzheimer's Disease,” Nature Neuroscience 23, no. 10 (2020): 1183–1193, 10.1038/s41593-020-0687-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Griffith L. S., Mathes M., and Schmitz B., “Beta‐Amyloid Precursor Protein Is Modified With O‐Linked N‐Acetylglucosamine,” Journal of Neuroscience Research 41, no. 2 (1995): 270–278, 10.1002/jnr.490410214. [DOI] [PubMed] [Google Scholar]
- 162. Kim C., Nam D. W., Park S. Y., et al., “O‐Linked Beta‐N‐Acetylglucosaminidase Inhibitor Attenuates Beta‐Amyloid Plaque and Rescues Memory Impairment,” Neurobiology of Aging 34, no. 1 (2013): 275–285, 10.1016/j.neurobiolaging.2012.03.001. [DOI] [PubMed] [Google Scholar]
- 163. Shah S., Lee S. F., Tabuchi K., et al., “Nicastrin Functions as a Gamma‐Secretase‐Substrate Receptor,” Cell 122, no. 3 (2005): 435–447, 10.1016/j.cell.2005.05.022. [DOI] [PubMed] [Google Scholar]
- 164. Dong X., Shu L., Zhang J., et al., “Ogt‐Mediated O‐Glcnacylation Inhibits Astrocytes Activation Through Modulating Nf‐Kappab Signaling Pathway,” Journal of Neuroinflammation 20, no. 1 (2023): 146, 10.1186/s12974-023-02824-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Bedse G., Di Domenico F., Serviddio G., et al., “Aberrant Insulin Signaling in Alzheimer's Disease: Current Knowledge,” Frontiers in Neuroscience 9 (2015): 204, 10.3389/fnins.2015.00204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Salkovic‐Petrisic M. and Hoyer S., “Central Insulin Resistance as a Trigger for Sporadic Alzheimer‐Like Pathology: An Experimental Approach,” Journal of Neural Transmission Supplementum no. 72 (2007): 217–233. [DOI] [PubMed] [Google Scholar]
- 167. Klein A. L., Berkaw M. N., Buse M. G., et al., “O‐Linked N‐Acetylglucosamine Modification of Insulin Receptor Substrate‐1 Occurs in Close Proximity to Multiple Sh2 Domain Binding Motifs,” Molecular & Cellular Proteomics 8, no. 12 (2009): 2733–2745, 10.1074/mcp.M900207-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Dos S. J., Vizuete A., Hansen F., et al., “Early and Persistent O‐Glcnac Protein Modification in the Streptozotocin Model of Alzheimer's Disease,” Journal of Alzheimers Disease 61, no. 1 (2018): 237–249. [DOI] [PubMed] [Google Scholar]
- 169. Tysnes O. B. and Storstein A., “Epidemiology of Parkinson's Disease,” Journal of Neural Transmission 124, no. 8 (2017): 901–905, 10.1007/s00702-017-1686-y. [DOI] [PubMed] [Google Scholar]
- 170. Braak H., Del T. K., Rub U., et al., “Staging of Brain Pathology Related to Sporadic Parkinson's Disease,” Neurobiology of Aging 24, no. 2 (2003): 197–211, 10.1016/S0197-4580(02)00065-9. [DOI] [PubMed] [Google Scholar]
- 171. Lee B. E., Kim H. Y., Kim H. J., et al., “O‐Glcnacylation Regulates Dopamine Neuron Function, Survival and Degeneration in Parkinson Disease,” Brain 143, no. 12 (2020): 3699–3716, 10.1093/brain/awaa320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Zhang J., Li X., and Li J. D., “The Roles of Post‐Translational Modifications on Alpha‐Synuclein in the Pathogenesis of Parkinson's Diseases,” Frontiers in Neuroscience 13 (2019): 381, 10.3389/fnins.2019.00381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Wang Z., Udeshi N. D., O'Malley M., et al., “Enrichment and Site Mapping of O‐Linked N‐Acetylglucosamine by a Combination of Chemical/Enzymatic Tagging, Photochemical Cleavage, and Electron Transfer Dissociation Mass Spectrometry,” Molecular & Cellular Proteomics 9, no. 1 (2010): 153–160, 10.1074/mcp.M900268-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Kim W. S., Kagedal K., and Halliday G. M., “Alpha‐Synuclein Biology in Lewy Body Diseases,” Alzheimers Research & Therapy 6, no. 5 (2014): 73, 10.1186/s13195-014-0073-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Bras I. C. and Outeiro T. F., “Alpha‐Synuclein: Mechanisms of Release and Pathology Progression in Synucleinopathies,” Cells 10, no. 2 (2021), 10.3390/cells10020375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Marotta N. P., Lin Y. H., Lewis Y. E., et al., “O‐Glcnac Modification Blocks the Aggregation and Toxicity of the Protein Alpha‐Synuclein Associated With Parkinson's Disease,” Nature Chemistry 7, no. 11 (2015): 913–920, 10.1038/nchem.2361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Levine P. M., Galesic A., Balana A. T., et al., “Alpha‐Synuclein O‐Glcnacylation Alters Aggregation and Toxicity, Revealing Certain Residues as Potential Inhibitors of Parkinson's Disease,” Proceedings of the National Academy of Sciences of the United States of America 116, no. 5 (2019): 1511–1519, 10.1073/pnas.1808845116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Galesic A., Rakshit A., Cutolo G., et al., “Comparison of N‐Acetyl‐Glucosamine to Other Monosaccharides Reveals Structural Differences for the Inhibition of Alpha‐Synuclein Aggregation,” Acs Chemical Biology 16, no. 1 (2021): 14–19, 10.1021/acschembio.0c00716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Lee B. E., Suh P. G., and Kim J. I., “O‐Glcnacylation in Health and Neurodegenerative Diseases,” Experimental and Molecular Medicine 53, no. 11 (2021): 1674–1682, 10.1038/s12276-021-00709-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. McColgan P. and Tabrizi S. J., “Huntington's Disease: A Clinical Review,” European Journal of Neurology 25, no. 1 (2018): 24–34, 10.1111/ene.13413. [DOI] [PubMed] [Google Scholar]
- 181. Walker F. O., “Huntington's Disease,” Lancet 369, no. 9557 (2007): 218–228, 10.1016/S0140-6736(07)60111-1. [DOI] [PubMed] [Google Scholar]
- 182. Tabrizi S. J., Flower M. D., Ross C. A., et al., “Huntington Disease: New Insights into Molecular Pathogenesis and Therapeutic Opportunities,” Nature Reviews Neurology 16, no. 10 (2020): 529–546, 10.1038/s41582-020-0389-4. [DOI] [PubMed] [Google Scholar]
- 183. Ho L. W., Brown R., Maxwell M., et al., “Wild Type Huntingtin Reduces the Cellular Toxicity of Mutant Huntingtin in Mammalian Cell Models of Huntington's Disease,” Journal of Medical Genetics 38, no. 7 (2001): 450–452, 10.1136/jmg.38.7.450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Wulff‐Fuentes E., Berendt R. R., Massman L., et al., “The Human O‐Glcnacome Database and Meta‐Analysis,” Scientific Data 8, no. 1 (2021): 25, 10.1038/s41597-021-00810-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Kumar A., Singh P. K., Parihar R., et al., “Decreased O‐Linked Glcnacylation Protects from Cytotoxicity Mediated by Huntingtin Exon1 Protein Fragment,” Journal of Biological Chemistry 289, no. 19 (2014): 13543–13553, 10.1074/jbc.M114.553321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Grima J. C., Daigle J. G., Arbez N., et al., “Mutant Huntingtin Disrupts the Nuclear Pore Complex,” Neuron 94, no. 1 (2017): 93–107, 10.1016/j.neuron.2017.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Zhu Y., Liu T. W., Madden Z., et al., “Post‐Translational O‐Glcnacylation Is Essential for Nuclear Pore Integrity and Maintenance of the Pore Selectivity Filter,” Journal of Molecular Cell Biology 8, no. 1 (2016): 2–16, 10.1093/jmcb/mjv033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Goutman S. A., Hardiman O., Al‐Chalabi A., et al., “Recent Advances in the Diagnosis and Prognosis of Amyotrophic Lateral Sclerosis,” Lancet Neurology 21, no. 5 (2022): 480–493, 10.1016/S1474-4422(21)00465-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Beeldman E., Govaarts R., de Visser M., et al., “Progression of Cognitive and Behavioural Impairment in Early Amyotrophic Lateral Sclerosis,” Journal of Neurology Neurosurgery and Psychiatry 91, no. 7 (2020): 779–780, 10.1136/jnnp-2020-322992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Pender N., Pinto‐Grau M., and Hardiman O., “Cognitive and Behavioural Impairment in Amyotrophic Lateral Sclerosis,” Current Opinion in Neurology 33, no. 5 (2020): 649–654, 10.1097/WCO.0000000000000862. [DOI] [PubMed] [Google Scholar]
- 191. Crockford C., Newton J., Lonergan K., et al., “Als‐Specific Cognitive and Behavior Changes Associated With Advancing Disease Stage in Als,” Neurology 91, no. 15 (2018): e1370–e1380, 10.1212/WNL.0000000000006317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Bersano E., Sarnelli M. F., Solara V., et al., “Decline of Cognitive and Behavioral Functions in Amyotrophic Lateral Sclerosis: A Longitudinal Study,” Amyotrophic Lateral Sclerosis and Frontotemporal Degeneration 21, no. 5–6 (2020): 373–379, 10.1080/21678421.2020.1771732. [DOI] [PubMed] [Google Scholar]
- 193. Corbo M. and Hays A. P., “Peripherin and Neurofilament Protein Coexist in Spinal Spheroids of Motor Neuron Disease,” Journal of Neuropathology and Experimental Neurology 51, no. 5 (1992): 531–537, 10.1097/00005072-199209000-00008. [DOI] [PubMed] [Google Scholar]
- 194. Dong D. L., Xu Z. S., Chevrier M. R., et al., “Glycosylation of Mammalian Neurofilaments. Localization of Multiple O‐Linked N‐Acetylglucosamine Moieties on Neurofilament Polypeptides L and M,” Journal of Biological Chemistry 268, no. 22 (1993): 16679–16687, 10.1016/S0021-9258(19)85471-6. [DOI] [PubMed] [Google Scholar]
- 195. Deng Y., Li B., Liu F., et al., “Regulation between O‐Glcnacylation and Phosphorylation of Neurofilament‐M and Their Dysregulation in Alzheimer Disease,” Faseb Journal 22, no. 1 (2008): 138–145, 10.1096/fj.07-8309com. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Dong D. L., Xu Z. S., Hart G. W., et al., “Cytoplasmic O‐Glcnac Modification of the Head Domain and the Ksp Repeat Motif of the Neurofilament Protein Neurofilament‐H,” Journal of Biological Chemistry 271, no. 34 (1996): 20845–20852, 10.1074/jbc.271.34.20845. [DOI] [PubMed] [Google Scholar]
- 197. Xiao S., McLean J., and Robertson J., “Neuronal Intermediate Filaments and Als: A New Look at an Old Question,” Biochimica Et Biophysica Acta 1762, no. 11–12 (2006): 1001–1012, 10.1016/j.bbadis.2006.09.003. [DOI] [PubMed] [Google Scholar]
- 198. Lobsiger C. S., Garcia M. L., Ward C. M., et al., “Altered Axonal Architecture by Removal of the Heavily Phosphorylated Neurofilament Tail Domains Strongly Slows Superoxide Dismutase 1 Mutant‐Mediated Als,” Proceedings of the National Academy of Sciences of the United States of America 102, no. 29 (2005): 10351–10356, 10.1073/pnas.0503862102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Ludemann N., Clement A., Hans V. H., et al., “O‐Glycosylation of the Tail Domain of Neurofilament Protein M in Human Neurons and in Spinal Cord Tissue of a Rat Model of Amyotrophic Lateral Sclerosis (Als),” Journal of Biological Chemistry 280, no. 36 (2005): 31648–31658, 10.1074/jbc.M504395200. [DOI] [PubMed] [Google Scholar]
- 200. Cohen T. J., Lee V. M., and Trojanowski J. Q., “Tdp‐43 Functions and Pathogenic Mechanisms Implicated in Tdp‐43 Proteinopathies,” Trends in Molecular Medicine 17, no. 11 (2011): 659–667, 10.1016/j.molmed.2011.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Morgan S. and Orrell R. W., “Pathogenesis of Amyotrophic Lateral Sclerosis,” British Medical Bulletin 119, no. 1 (2016): 87–98, 10.1093/bmb/ldw026. [DOI] [PubMed] [Google Scholar]
- 202. Saberi S., Stauffer J. E., Schulte D. J., et al., “Neuropathology of Amyotrophic Lateral Sclerosis and Its Variants,” Neurologic Clinics 33, no. 4 (2015): 855–876, 10.1016/j.ncl.2015.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Zhao M. J., Yao X., Wei P., et al., “O‐Glcnacylation of Tdp‐43 Suppresses Proteinopathies and Promotes Tdp‐43'S Mrna Splicing Activity,” Embo Reports 22, no. 6 (2021): e51649, 10.15252/embr.202051649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Hsieh Y. L., Su F. Y., Tsai L. K., et al., “Npgpx‐Mediated Adaptation to Oxidative Stress Protects Motor Neurons from Degeneration in Aging by Directly Modulating O‐Glcnacase,” Cell Reports 29, no. 8 (2019): 2134–2143, 10.1016/j.celrep.2019.10.053. [DOI] [PubMed] [Google Scholar]
- 205. Thai A. A., Solomon B. J., Sequist L. V., et al., “Lung Cancer,” Lancet 398, no. 10299 (2021): 535–554, 10.1016/S0140-6736(21)00312-3. [DOI] [PubMed] [Google Scholar]
- 206. Liu Y. Y., Liu H. Y., Yu T. J., et al., “O‐Glcnacylation of Morc2 at Threonine 556 by Ogt Couples Tgf‐Beta Signaling to Breast Cancer Progression,” Cell Death and Differentiation 29, no. 4 (2022): 861–873, 10.1038/s41418-021-00901-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Lin L., Yuan Q., Gu J., et al., “Carm1‐Mediated Ogt Arginine Methylation Promotes Non‐Small Cell Lung Cancer Glycolysis by Stabilizing Ogt,” Cell Death & Disease 15, no. 12 (2024): 927, 10.1038/s41419-024-07313-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Luanpitpong S., Rodboon N., Samart P., et al., “A Novel Trpm7/O‐Glcnac Axis Mediates Tumour Cell Motility and Metastasis by Stabilising C‐Myc and Caveolin‐1 in Lung Carcinoma,” British Journal of Cancer 123, no. 8 (2020): 1289–1301, 10.1038/s41416-020-0991-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Lin C. H., Liao C. C., Wang S. Y., et al., “Comparative O‐Glcnac Proteomic Analysis Reveals a Role of O‐Glcnacylated Sam68 in Lung Cancer Aggressiveness,” Cancers 14, no. 1 (2022), 10.3390/cancers14010243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Ge X., Peng X., Li M., et al., “Ogt Regulated O‐Glcnacylation Promotes Migration and Invasion by Activating Il‐6/Stat3 Signaling in Nsclc Cells,” Pathology Research and Practice 225 (2021): 153580, 10.1016/j.prp.2021.153580. [DOI] [PubMed] [Google Scholar]
- 211. Iwasaki T., Chin W. W., and Ko L., “Identification and Characterization of Rrm‐Containing Coactivator Activator (Coaa) as Trbp‐Interacting Protein, and Its Splice Variant as a Coactivator Modulator (Coam),” Journal of Biological Chemistry 276, no. 36 (2001): 33375–33383, 10.1074/jbc.M101517200. [DOI] [PubMed] [Google Scholar]
- 212. Auboeuf D., Dowhan D. H., Li X., et al., “Coaa, a Nuclear Receptor Coactivator Protein at the Interface of Transcriptional Coactivation and Rna Splicing,” Molecular and Cellular Biology 24, no. 1 (2004): 442–453, 10.1128/MCB.24.1.442-453.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Kweon T. H., Jung H., Ko J. Y., et al., “O‐Glcnacylation of Rbm14 Contributes to Elevated Cellular O‐Glcnac Through Regulation of Oga Protein Stability,” Cell Reports 43, no. 5 (2024): 114163, 10.1016/j.celrep.2024.114163. [DOI] [PubMed] [Google Scholar]
- 214. Itoh K., Wakabayashi N., Katoh Y., et al., “Keap1 Represses Nuclear Activation of Antioxidant Responsive Elements by Nrf2 Through Binding to the Amino‐Terminal Neh2 Domain,” Genes & Development 13, no. 1 (1999): 76–86, 10.1101/gad.13.1.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Zhang Y., Sun C., Ma L., et al., “O‐Glcnacylation Promotes Malignancy and Cisplatin Resistance of Lung Cancer by Stabilising Nrf2,” Clinical and Translational Medicine 14, no. 10 (2024): e70037, 10.1002/ctm2.70037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Vomund S., Schafer A., Parnham M. J., et al., “Nrf2, the Master Regulator of Anti‐Oxidative Responses,” International Journal of Molecular Sciences 18, no. 12 (2017), 10.3390/ijms18122772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Luanpitpong S., Rodboon N., Samart P., et al., “A Novel Trpm7/O‐Glcnac Axis Mediates Tumour Cell Motility and Metastasis by Stabilising C‐Myc and Caveolin‐1 in Lung Carcinoma,” British Journal of Cancer 123, no. 8 (2020): 1289–1301, 10.1038/s41416-020-0991-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Lin C. H., Liao C. C., Wang S. Y., et al., “Comparative O‐Glcnac Proteomic Analysis Reveals a Role of O‐Glcnacylated Sam68 in Lung Cancer Aggressiveness,” Cancers 14, no. 1 (2022), 10.3390/cancers14010243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Zhao J., Zhao Z., Zhou W., et al., “Yeats2 O‐Glcnacylation Promotes Chromatin Association of the Atac Complex and Lung Cancer Tumorigenesis,” Journal of Biological Chemistry 301, no. 7 (2025): 110388, 10.1016/j.jbc.2025.110388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Khorana H. G., Fernandes J. F., and Kornberg A., “Pyrophosphorylation of Ribose 5‐Phosphate in the Enzymatic Synthesis of 5‐Phosphorylribose 1‐Pyrophosphate,” Journal of Biological Chemistry 230, no. 2 (1958): 941–948. [PubMed] [Google Scholar]
- 221. Chen L., Zhou Q., Zhang P., et al., “Direct Stimulation of De Novo Nucleotide Synthesis by O‐Glcnacylation,” Nature Chemical Biology 20, no. 1 (2024): 19–29, 10.1038/s41589-023-01354-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Alfaro C., Sanmamed M. F., Rodriguez‐Ruiz M. E., et al., “Interleukin‐8 in Cancer Pathogenesis, Treatment and Follow‐Up,” Cancer Treatment Reviews 60 (2017): 24–31, 10.1016/j.ctrv.2017.08.004. [DOI] [PubMed] [Google Scholar]
- 223. David J. M., Dominguez C., Hamilton D. H., et al., “The Il‐8/Il‐8R Axis: A Double Agent in Tumor Immune Resistance,” Vaccines 4, no. 3 (2016), 10.3390/vaccines4030022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Shimizu M. and Tanaka N., “Il‐8‐Induced O‐Glcnac Modification via Glut3 and Gfat Regulates Cancer Stem Cell‐Like Properties in Colon and Lung Cancer Cells,” Oncogene 38, no. 9 (2019): 1520–1533, 10.1038/s41388-018-0533-4. [DOI] [PubMed] [Google Scholar]
- 225. Lin L., Yuan Q., Gu J., et al., “Carm1‐Mediated Ogt Arginine Methylation Promotes Non‐Small Cell Lung Cancer Glycolysis by Stabilizing Ogt,” Cell Death & Disease 15, no. 12 (2024): 927, 10.1038/s41419-024-07313-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Kim Y. J., Kang M. J., Kim E., et al., “O‐Glcnac Stabilizes Smad4 by Inhibiting Gsk‐3Beta‐Mediated Proteasomal Degradation,” Scientific Reports 10, no. 1 (2020): 19908, 10.1038/s41598-020-76862-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227. Ge X., Peng X., Li M., et al., “Ogt Regulated O‐Glcnacylation Promotes Migration and Invasion by Activating Il‐6/Stat3 Signaling in Nsclc Cells,” Pathology Research and Practice 225 (2021): 153580, 10.1016/j.prp.2021.153580. [DOI] [PubMed] [Google Scholar]
- 228. Torre L. A., Bray F., Siegel R. L., et al., “Global Cancer Statistics,” Ca‐a Cancer Journal for Clinicians 65, no. 2 (2012): 87–108. [DOI] [PubMed] [Google Scholar]
- 229. El‐Serag H. B., “Hepatocellular Carcinoma,” New England Journal of Medicine 365, no. 12 (2011): 1118–1127, 10.1056/NEJMra1001683. [DOI] [PubMed] [Google Scholar]
- 230. Xu W., Zhang X., Wu J. L., et al., “O‐Glcnac Transferase Promotes Fatty Liver‐Associated Liver Cancer Through Inducing Palmitic Acid and Activating Endoplasmic Reticulum Stress,” Journal of Hepatology 67, no. 2 (2017): 310–320, 10.1016/j.jhep.2017.03.017. [DOI] [PubMed] [Google Scholar]
- 231. Nakagawa H., Umemura A., Taniguchi K., et al., “Er Stress Cooperates With Hypernutrition to Trigger Tnf‐Dependent Spontaneous Hcc Development,” Cancer Cell 26, no. 3 (2014): 331–343, 10.1016/j.ccr.2014.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Garcia‐Ruiz C., Mato J. M., Vance D., et al., “Acid Sphingomyelinase‐Ceramide System in Steatohepatitis: A Novel Target Regulating Multiple Pathways,” Journal of Hepatology 62, no. 1 (2015): 219–233, 10.1016/j.jhep.2014.09.023. [DOI] [PubMed] [Google Scholar]
- 233. Raab S., Gadault A., Very N., et al., “Dual Regulation of Fatty Acid Synthase (Fasn) Expression by O‐Glcnac Transferase (Ogt) and Mtor Pathway in Proliferating Liver Cancer Cells,” Cellular and Molecular Life Sciences 78, no. 13 (2021): 5397–5413, 10.1007/s00018-021-03857-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Matallanas D., Birtwistle M., Romano D., et al., “Raf Family Kinases: Old Dogs Have Learned New Tricks,” Genes & Cancer 2, no. 3 (2011): 232–260, 10.1177/1947601911407323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Ning D., Chen J., Du P., et al., “The Crosstalk Network of Xist/Mir‐424‐5P/Ogt Mediates Raf1 Glycosylation and Participates in the Progression of Liver Cancer,” Liver International 41, no. 8 (2021): 1933–1944, 10.1111/liv.14904. [DOI] [PubMed] [Google Scholar]
- 236. Wang J., Ma L., Weng W., et al., “Mutual Interaction between Yap and Creb Promotes Tumorigenesis in Liver Cancer,” Hepatology 58, no. 3 (2013): 1011–1020, 10.1002/hep.26420. [DOI] [PubMed] [Google Scholar]
- 237. Perra A., Kowalik M. A., Ghiso E., et al., “Yap Activation Is an Early Event and a Potential Therapeutic Target in Liver Cancer Development,” Journal of Hepatology 61, no. 5 (2014): 1088–1096, 10.1016/j.jhep.2014.06.033. [DOI] [PubMed] [Google Scholar]
- 238. Ryu T. Y., Park J., and Scherer P. E., “Hyperglycemia as a Risk Factor for Cancer Progression,” Diabetes & Metabolism Journal 38, no. 5 (2014): 330–336, 10.4093/dmj.2014.38.5.330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Zhang X., Qiao Y., Wu Q., et al., “The Essential Role of Yap O‐Glcnacylation in High‐Glucose‐Stimulated Liver Tumorigenesis,” Nature Communications 8 (2017): 15280, 10.1038/ncomms15280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Dekker E., Tanis P. J., Vleugels J., et al., “Colorectal Cancer,” Lancet 394, no. 10207 (2019): 1467–1480, 10.1016/S0140-6736(19)32319-0. [DOI] [PubMed] [Google Scholar]
- 241. Steenackers A., Olivier‐Van S. S., Baldini S. F., et al., “Silencing the Nucleocytoplasmic O‐Glcnac Transferase Reduces Proliferation, Adhesion, and Migration of Cancer and Fetal Human Colon Cell Lines,” Frontiers in Endocrinology 7 (2016): 46, 10.3389/fendo.2016.00046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Phueaouan T., Chaiyawat P., Netsirisawan P., et al., “Aberrant O‐Glcnac‐Modified Proteins Expressed in Primary Colorectal Cancer,” Oncology Reports 30, no. 6 (2013): 2929–2936, 10.3892/or.2013.2794. [DOI] [PubMed] [Google Scholar]
- 243. Kristensen M. H., Weidinger M., Bzorek M., et al., “Correlation between Thymidylate Synthase Gene Variants, Rna and Protein Levels in Primary Colorectal Adenocarcinomas,” Journal of International Medical Research 38, no. 2 (2010): 484–497, 10.1177/147323001003800212. [DOI] [PubMed] [Google Scholar]
- 244. Chen Z., Han S., Huang W., et al., “Microrna‐215 Suppresses Cell Proliferation, Migration and Invasion of Colon Cancer by Repressing Yin‐Yang 1,” Biochemical and Biophysical Research Communications 479, no. 3 (2016): 482–488, 10.1016/j.bbrc.2016.09.089. [DOI] [PubMed] [Google Scholar]
- 245. Zhu G., Qian M., Lu L., et al., “O‐Glcnacylation of Yy1 Stimulates Tumorigenesis in Colorectal Cancer Cells by Targeting Slc22a15 and Aanat,” Carcinogenesis 40, no. 9 (2019): 1121–1131. [DOI] [PubMed] [Google Scholar]
- 246. Li J., Ahmad M., Sang L., et al., “O‐Glcnacylation Promotes the Cytosolic Localization of the M(6)a Reader Ythdf1 and Colorectal Cancer Tumorigenesis,” Journal of Biological Chemistry 299, no. 6 (2023): 104738, 10.1016/j.jbc.2023.104738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Shi H., Wei J., and He C., “Where, When, and How: Context‐Dependent Functions of Rna Methylation Writers, Readers, and Erasers,” Molecular Cell 74, no. 4 (2019): 640–650, 10.1016/j.molcel.2019.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Wang X., Zhao B. S., Roundtree I. A., et al., “N(6)‐Methyladenosine Modulates Messenger Rna Translation Efficiency,” Cell 161, no. 6 (2015): 1388–1399, 10.1016/j.cell.2015.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Yu Z., He H., Jiang B., et al., “O‐Glcnacylation of Csnk2a1 by Ogt Is Involved in the Progression of Colorectal Cancer,” Molecular Biotechnology 67, no. 1 (2025): 272–283, 10.1007/s12033-024-01049-1. [DOI] [PubMed] [Google Scholar]
- 250. Zhong X., He X., Wang Y., et al., “Warburg Effect in Colorectal Cancer: The Emerging Roles in Tumor Microenvironment and Therapeutic Implications,” Journal of Hematology & Oncology 15, no. 1 (2022): 160, 10.1186/s13045-022-01358-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Pavlova N. N. and Thompson C. B., “The Emerging Hallmarks of Cancer Metabolism,” Cell Metabolism 23, no. 1 (2016): 27–47, 10.1016/j.cmet.2015.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Wang H., Sun J., Sun H., et al., “The Ogt‐C‐Myc‐Pdk2 Axis Rewires the Tca Cycle and Promotes Colorectal Tumor Growth,” Cell Death and Differentiation 31, no. 9 (2024): 1157–1169, 10.1038/s41418-024-01315-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Luo X., Cheng C., Tan Z., et al., “Emerging Roles of Lipid Metabolism in Cancer Metastasis,” Molecular Cancer 16, no. 1 (2017): 76, 10.1186/s12943-017-0646-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Drury J., Geisen M. E., Tessmann J. W., et al., “Overexpression of Fatty Acid Synthase Upregulates Glutamine‐Fructose‐6‐Phosphate Transaminase 1 and O‐Linked N‐Acetylglucosamine Transferase to Increase O‐Glcnac Protein Glycosylation and Promote Colorectal Cancer Growth,” International Journal of Molecular Sciences 25, no. 9 (2024), 10.3390/ijms25094883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Murillo C. A., Rychahou P. G., and Evers B. M., “Inhibition of Alpha5 Integrin Decreases Pi3K Activation and Cell Adhesion of Human Colon Cancers,” Surgery 136, no. 2 (2004): 143–149, 10.1016/j.surg.2004.04.006. [DOI] [PubMed] [Google Scholar]
- 256. Pelillo C., Bergamo A., Mollica H., et al., “Colorectal Cancer Metastases Settle in the Hepatic Microenvironment Through Alpha5Beta1 Integrin,” Journal of Cellular Biochemistry 116, no. 10 (2015): 2385–2396, 10.1002/jcb.25189. [DOI] [PubMed] [Google Scholar]
- 257. Nam E. H., Lee Y., Zhao X. F., et al., “Zeb2‐Sp1 Cooperation Induces Invasion by Upregulating Cadherin‐11 and Integrin Alpha5 Expression,” Carcinogenesis 35, no. 2 (2014): 302–314, 10.1093/carcin/bgt340. [DOI] [PubMed] [Google Scholar]
- 258. Nam E. H., Lee Y., Moon B., et al., “Twist1 and Ap‐1 Cooperatively Upregulate Integrin Alpha5 Expression to Induce Invasion and the Epithelial‐Mesenchymal Transition,” Carcinogenesis 36, no. 3 (2015): 327–337, 10.1093/carcin/bgv005. [DOI] [PubMed] [Google Scholar]
- 259. Yu M., Chu S., Fei B., et al., “O‐Glcnacylation of Itga5 Facilitates the Occurrence and Development of Colorectal Cancer,” Experimental Cell Research 382, no. 2 (2019): 111464, 10.1016/j.yexcr.2019.06.009. [DOI] [PubMed] [Google Scholar]
- 260. Jiang M., Xu B., Li X., et al., “O‐Glcnacylation Promotes Colorectal Cancer Metastasis via the Mir‐101‐O‐Glcnac/Ezh2 Regulatory Feedback Circuit,” Oncogene 38, no. 3 (2019): 301–316, 10.1038/s41388-018-0435-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Wang M., Qiao L., Jin L., et al., “Ogt‐Regulated O‐Glcnacylation Promotes the Malignancy of Colorectal Cancer by Activating Stat2 to Induce Macrophage M2: Ogt Protein Macromolecule Action,” International Journal of Biological Macromolecules 311, no. Pt 3 (2025): 144057, 10.1016/j.ijbiomac.2025.144057. [DOI] [PubMed] [Google Scholar]
- 262. Cho H. I., Jo S., Kim M. S., et al., “Setd5 Regulates the Ogt‐Catalyzed O‐Glcnacylation of Rna Polymerase II, Which Is Involved in the Stemness of Colorectal Cancer Cells,” Scientific Reports 13, no. 1 (2023): 19885, 10.1038/s41598-023-46923-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263. Liu Y. Y., Liu H. Y., Yu T. J., et al., “O‐Glcnacylation of Morc2 at Threonine 556 by Ogt Couples Tgf‐Beta Signaling to Breast Cancer Progression,” Cell Death and Differentiation 29, no. 4 (2022): 861–873, 10.1038/s41418-021-00901-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Sodi V. L., Khaku S., Krutilina R., et al., “Mtor/Myc Axis Regulates O‐Glcnac Transferase Expression and O‐Glcnacylation in Breast Cancer,” Molecular Cancer Research 13, no. 5 (2015): 923–933, 10.1158/1541-7786.MCR-14-0536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265. Uno Y. and Hayakawa K., “O‐Glcnacylation on Serine 40 of Histone H2a Promotes Proliferation and Invasion in Triple‐Negative Breast Cancer,” Scientific Reports 15, no. 1 (2025): 10170, 10.1038/s41598-025-95394-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Ferrer C. M., Lu T. Y., Bacigalupa Z. A., et al., “O‐Glcnacylation Regulates Breast Cancer Metastasis via Sirt1 Modulation of Foxm1 Pathway,” Oncogene 36, no. 4 (2017): 559–569, 10.1038/onc.2016.228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267. Koo C. Y., Muir K. W., and Lam E. W., “Foxm1: From Cancer Initiation to Progression and Treatment,” Biochimica Et Biophysica Acta 1819, no. 1 (2012): 28–37, 10.1016/j.bbagrm.2011.09.004. [DOI] [PubMed] [Google Scholar]
- 268. Gu Y., Mi W., Ge Y., et al., “Glcnacylation Plays an Essential Role in Breast Cancer Metastasis,” Cancer Research 70, no. 15 (2010): 6344–6351, 10.1158/0008-5472.CAN-09-1887. [DOI] [PubMed] [Google Scholar]
- 269. Ghosh M., Song X., Mouneimne G., et al., “Cofilin Promotes Actin Polymerization and Defines the Direction of Cell Motility,” Science 304, no. 5671 (2004): 743–746, 10.1126/science.1094561. [DOI] [PubMed] [Google Scholar]
- 270. Sidani M., Wessels D., Mouneimne G., et al., “Cofilin Determines the Migration Behavior and Turning Frequency of Metastatic Cancer Cells,” Journal of Cell Biology 179, no. 4 (2007): 777–791, 10.1083/jcb.200707009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Huang X., Pan Q., Sun D., et al., “O‐Glcnacylation of Cofilin Promotes Breast Cancer Cell Invasion,” Journal of Biological Chemistry 288, no. 51 (2013): 36418–36425, 10.1074/jbc.M113.495713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Xu Y., Sheng X., Zhao T., et al., “O‐Glcnacylation of Mek2 Promotes the Proliferation and Migration of Breast Cancer Cells,” Glycobiology 31, no. 5 (2021): 571–581, 10.1093/glycob/cwaa103. [DOI] [PubMed] [Google Scholar]
- 273. Schmolka N., Karemaker I. D., Cardoso D. S. R., et al., “Dissecting the Roles of Mbd2 Isoforms and Domains in Regulating Nurd Complex Function during Cellular Differentiation,” Nature Communications 14, no. 1 (2023): 3848, 10.1038/s41467-023-39551-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274. Wang B., Li C., Ming J., et al., “The Nurd Complex Cooperates With Sall4 to Orchestrate Reprogramming,” Nature Communications 14, no. 1 (2023): 2846, 10.1038/s41467-023-38543-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275. Grzeskowiak C. L., Kundu S. T., Mo X., et al., “In Vivo Screening Identifies Gatad2B as a Metastasis Driver in Kras‐Driven Lung Cancer,” Nature Communications 9, no. 1 (2018): 2732, 10.1038/s41467-018-04572-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Le Minh G., Merzy J., Esquea E. M., et al., “Gatad2B O‐Glcnacylation Regulates Breast Cancer Stem‐Like Potential and Drug Resistance,” Cells 14, no. 6 (2025), 10.3390/cells14060398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277. Akella N. M., Le Minh G., Ciraku L., et al., “O‐Glcnac Transferase Regulates Cancer Stem‐Like Potential of Breast Cancer Cells,” Molecular Cancer Research 18, no. 4 (2020): 585–598, 10.1158/1541-7786.MCR-19-0732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278. Le Minh G., Esquea E. M., Dhameliya T. T., et al., “Kruppel‐Like Factor 8 Regulates Triple Negative Breast Cancer Stem Cell‐Like Activity,” Frontiers in Oncology 13 (2023): 1141834, 10.3389/fonc.2023.1141834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279. Li X., Wu Z., He J., et al., “Ogt Regulated O‐Glcnacylation Promotes Papillary Thyroid Cancer Malignancy via Activating Yap,” Oncogene 40, no. 30 (2021): 4859–4871, 10.1038/s41388-021-01901-7. [DOI] [PubMed] [Google Scholar]
- 280. Zhang P., Wang C., Ma T., et al., “O‐Glcnacylation Enhances the Invasion of Thyroid Anaplastic Cancer Cells Partially by Pi3K/Akt1 Pathway,” Oncotargets and Therapy 8 (2015): 3305–3313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Cheng Y. U., Li H., Li J., et al., “O‐Glcnacylation Enhances Anaplastic Thyroid Carcinoma Malignancy,” Oncology Letters 12, no. 1 (2016): 572–578, 10.3892/ol.2016.4647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282. Zhang P., Luo Z., Xu Y., et al., “Hypoxia‐Induced Circprelid2 Promotes Gastric Cancer Metastasis by Facilitating Zeb2 Translation via Pcbp1 O‐Glcnacylation,” Advanced Science (2025): e05396, 10.1002/advs.202505396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283. Jiang M., Wu N., Xu B., et al., “Fatty Acid‐Induced Cd36 Expression via O‐Glcnacylation Drives Gastric Cancer Metastasis,” Theranostics 9, no. 18 (2019): 5359–5373, 10.7150/thno.34024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284. Wang G., Xu Z., Sun J., et al., “O‐Glcnacylation Enhances Reticulon 2 Protein Stability and Its Promotive Effects on Gastric Cancer Progression,” Cellular Signalling 108 (2023): 110718, 10.1016/j.cellsig.2023.110718. [DOI] [PubMed] [Google Scholar]
- 285. Qiao Z., Dang C., Zhou B., et al., “Downregulation of O‐Linked N‐Acetylglucosamine Transferase by Rna Interference Decreases Mmp9 Expression in Human Esophageal Cancer Cells,” Oncology Letters 11, no. 5 (2016): 3317–3323, 10.3892/ol.2016.4428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Wu H., Song S., Yan A., et al., “Rack1 Promotes the Invasive Activities and Lymph Node Metastasis of Cervical Cancer via Galectin‐1,” Cancer Letters 469 (2020): 287–300, 10.1016/j.canlet.2019.11.002. [DOI] [PubMed] [Google Scholar]
- 287. Zeng Q., Zhao R. X., Chen J., et al., “O‐Linked Glcnacylation Elevated by Hpv E6 Mediates Viral Oncogenesis,” Proceedings of the National Academy of Sciences of the United States of America 113, no. 33 (2016): 9333–9338, 10.1073/pnas.1606801113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Zhang C., Jiang H., Yuan L., et al., “Circvprbp Inhibits Nodal Metastasis of Cervical Cancer by Impeding Rack1 O‐Glcnacylation and Stability,” Oncogene 42, no. 11 (2023): 793–807, 10.1038/s41388-023-02595-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289. Zhang C., Jiang H., Yuan L., et al., “Correction: Circvprbp Inhibits Nodal Metastasis of Cervical Cancer by Impeding Rack1 O‐Glcnacylation and Stability,” Oncogene 44, no. 23 (2025): 1887–1890, 10.1038/s41388-025-03440-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290. Ali A., Kim S. H., Kim M. J., et al., “O‐Glcnacylation of Nf‐Kappab Promotes Lung Metastasis of Cervical Cancer Cells via Upregulation of Cxcr4 Expression,” Molecules and Cells 40, no. 7 (2017): 476–484, 10.14348/molcells.2017.2309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291. Yan T., Ma X., Zhou K., et al., “A Novel Csn5/Crt O‐Glcnac/Er Stress Regulatory Axis in Platinum Resistance of Epithelial Ovarian Cancer,” International Journal of Biological Sciences 20, no. 4 (2024): 1279–1296, 10.7150/ijbs.89700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292. Qian L., Yang X., Li S., et al., “Reduced O‐Glcnacylation of Snap‐23 Promotes Cisplatin Resistance by Inducing Exosome Secretion in Ovarian Cancer,” Cell Death Discovery 7, no. 1 (2021): 112, 10.1038/s41420-021-00489-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293. Rogalska A., Forma E., Brys M., et al., “Hyperglycemia‐Associated Dysregulation of O‐Glcnacylation and Hif1a Reduces Anticancer Action of Metformin in Ovarian Cancer Cells (Skov‐3),” International Journal of Molecular Sciences 19, no. 9 (2018), 10.3390/ijms19092750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Peng J., Liu J., Ning M., et al., “Fluzoparib Disrupts Golgi Apparatus to Inhibit O‐Glcnacylation and Nuclear Translocation of Beta‐Catenin to Attenuate Ovarian Cancer Invasion and Metastasis,” International Immunopharmacology 162 (2025): 115158, 10.1016/j.intimp.2025.115158. [DOI] [PubMed] [Google Scholar]
- 295. Xia L., Mei J., Huang M., et al., “O‐Glcnacylation in Ovarian Tumorigenesis and Its Therapeutic Implications,” Translational Oncology 51 (2025): 102220, 10.1016/j.tranon.2024.102220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296. Yan Z., Li Y., Wang M., et al., “O‐Glcnacylation of Dj‐1 Suppresses Ferroptosis in Renal Cell Carcinoma by Affecting the Transsulfuration Pathway,” International Immunopharmacology 148 (2025): 114098, 10.1016/j.intimp.2025.114098. [DOI] [PubMed] [Google Scholar]
- 297. Wang L., Chen S., Zhang J., et al., “Suppressed Ogt Expression Inhibits Cell Proliferation and Modulates Egfr Expression in Renal Cell Carcinoma,” Cancer Management and Research 11 (2019): 2215–2223, 10.2147/CMAR.S190642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298. Olvera L. E., Ballard B. D., and Jan A., “Cardiovascular Disease,” (2025). [PubMed]
- 299. Flora G. D. and Nayak M. K., “A Brief Review of Cardiovascular Diseases, Associated Risk Factors and Current Treatment Regimes,” Current Pharmaceutical Design 25, no. 38 (2019): 4063–4084, 10.2174/1381612825666190925163827. [DOI] [PubMed] [Google Scholar]
- 300. Townsend N., Kazakiewicz D., Lucy W. F., et al., “Epidemiology of Cardiovascular Disease in Europe,” Nature Reviews Cardiology 19, no. 2 (2022): 133–143, 10.1038/s41569-021-00607-3. [DOI] [PubMed] [Google Scholar]
- 301. Wang H. F., Wang Y. X., Zhou Y. P., et al., “Protein O‐Glcnacylation in Cardiovascular Diseases,” Acta Pharmacologica Sinica 44, no. 1 (2023): 8–18, 10.1038/s41401-022-00934-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302. Wright J. N., Collins H. E., Wende A. R., et al., “O‐Glcnacylation and Cardiovascular Disease,” Biochemical Society Transactions 45, no. 2 (2017): 545–553, 10.1042/BST20160164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303. Umapathi P., Mesubi O. O., Banerjee P. S., et al., “Excessive O‐Glcnacylation Causes Heart Failure and Sudden Death,” Circulation 143, no. 17 (2021): 1687–1703, 10.1161/CIRCULATIONAHA.120.051911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304. Ferron M., Denis M., Persello A., et al., “Protein O‐Glcnacylation in Cardiac Pathologies: Past, Present, Future,” Frontiers in Endocrinology 9 (2018): 819, 10.3389/fendo.2018.00819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305. Lunde I. G., Aronsen J. M., Kvaloy H., et al., “Cardiac O‐Glcnac Signaling Is Increased in Hypertrophy and Heart Failure,” Physiological Genomics 44, no. 2 (2012): 162–172, 10.1152/physiolgenomics.00016.2011. [DOI] [PubMed] [Google Scholar]
- 306. Chou T. Y., Hart G. W., and Dang C. V., “C‐Myc Is Glycosylated at Threonine 58, a Known Phosphorylation Site and a Mutational Hot Spot in Lymphomas,” Journal of Biological Chemistry 270, no. 32 (1995): 18961–18965, 10.1074/jbc.270.32.18961. [DOI] [PubMed] [Google Scholar]
- 307. Zhong W., Mao S., Tobis S., et al., “Hypertrophic Growth in Cardiac Myocytes Is Mediated by Myc Through a Cyclin D2‐Dependent Pathway,” Embo Journal 25, no. 16 (2006): 3869–3879, 10.1038/sj.emboj.7601252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308. Ramirez‐Correa G. A., Jin W., Wang Z., et al., “O‐Linked Glcnac Modification of Cardiac Myofilament Proteins: A Novel Regulator of Myocardial Contractile Function,” Circulation Research 103, no. 12 (2008): 1354–1358, 10.1161/CIRCRESAHA.108.184978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309. Dubois‐Deruy E., Belliard A., Mulder P., et al., “Interplay between Troponin T Phosphorylation and O‐N‐Acetylglucosaminylation in Ischaemic Heart Failure,” Cardiovascular Research 107, no. 1 (2015): 56–65, 10.1093/cvr/cvv136. [DOI] [PubMed] [Google Scholar]
- 310. Jewell J. L. and Guan K. L., “Nutrient Signaling to Mtor and Cell Growth,” Trends in Biochemical Sciences 38, no. 5 (2013): 233–242, 10.1016/j.tibs.2013.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311. Sciarretta S., Volpe M., and Sadoshima J., “Mammalian Target of Rapamycin Signaling in Cardiac Physiology and Disease,” Circulation Research 114, no. 3 (2014): 549–564, 10.1161/CIRCRESAHA.114.302022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312. Tran D. H., May H. I., Li Q., et al., “Chronic Activation of Hexosamine Biosynthesis in the Heart Triggers Pathological Cardiac Remodeling,” Nature Communications 11, no. 1 (2020): 1771, 10.1038/s41467-020-15640-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 313. Xie S., Jin N., Gu J., et al., “O‐Glcnacylation of Protein Kinase a Catalytic Subunits Enhances Its Activity: A Mechanism Linked to Learning and Memory Deficits in Alzheimer's Disease,” Aging Cell 15, no. 3 (2016): 455–464, 10.1111/acel.12449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314. Chen X., Zhang L., He H., et al., “Increased O‐Glcnacylation Induces Myocardial Hypertrophy,” In Vitro Cellular & Developmental Biology‐Animal 56, no. 9 (2020): 735–743, 10.1007/s11626-020-00503-z. [DOI] [PubMed] [Google Scholar]
- 315. Yu W., Hu S., Xu X., et al., “Association between O‐Glcnac Transferase Activity and Major Adverse Cardiovascular Events: Findings from the China Peace‐Mpp Cohort,” Bmc Cardiovascular Disorders 25, no. 1 (2025): 281, 10.1186/s12872-025-04732-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316. Watson L. J., Facundo H. T., Ngoh G. A., et al., “O‐Linked Beta‐N‐Acetylglucosamine Transferase Is Indispensable in the Failing Heart,” Proceedings of the National Academy of Sciences of the United States of America 107, no. 41 (2010): 17797–17802, 10.1073/pnas.1001907107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317. Krawitt E. L., “Autoimmune Hepatitis,” New England Journal of Medicine 354, no. 1 (2006): 54–66, 10.1056/NEJMra050408. [DOI] [PubMed] [Google Scholar]
- 318. Taubert R., Hardtke‐Wolenski M., Noyan F., et al., “Intrahepatic Regulatory T Cells in Autoimmune Hepatitis Are Associated With Treatment Response and Depleted With Current Therapies,” Journal of Hepatology 61, no. 5 (2014): 1106–1114, 10.1016/j.jhep.2014.05.034. [DOI] [PubMed] [Google Scholar]
- 319. Hao X., Li Y., Wang J., et al., “Deficient O‐Glcnac Glycosylation Impairs Regulatory T Cell Differentiation and Notch Signaling in Autoimmune Hepatitis,” Frontiers in Immunology 9 (2018): 2089, 10.3389/fimmu.2018.02089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320. Korn T., Bettelli E., Oukka M., et al., “Il‐17 and Th17 Cells,” Annual Review of Immunology 27 (2009): 485–517, 10.1146/annurev.immunol.021908.132710. [DOI] [PubMed] [Google Scholar]
- 321. Dong C., “Th17 Cells in Development: An Updated View of Their Molecular Identity and Genetic Programming,” Nature Reviews Immunology 8, no. 5 (2008): 337–348, 10.1038/nri2295. [DOI] [PubMed] [Google Scholar]
- 322. Ivanov I. I., McKenzie B. S., Zhou L., et al., “The Orphan Nuclear Receptor Rorgammat Directs the Differentiation Program of Proinflammatory Il‐17+ T Helper Cells,” Cell 126, no. 6 (2006): 1121–1133, 10.1016/j.cell.2006.07.035. [DOI] [PubMed] [Google Scholar]
- 323. Yang X. O., Pappu B. P., Nurieva R., et al., “T Helper 17 Lineage Differentiation Is Programmed by Orphan Nuclear Receptors Ror Alpha and Ror Gamma,” Immunity 28, no. 1 (2008): 29–39, 10.1016/j.immuni.2007.11.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324. Liu R., Ma X., Chen L., et al., “Microrna‐15B Suppresses Th17 Differentiation and Is Associated With Pathogenesis of Multiple Sclerosis by Targeting O‐Glcnac Transferase,” Journal of Immunology 198, no. 7 (2017): 2626–2639, 10.4049/jimmunol.1601727. [DOI] [PubMed] [Google Scholar]
- 325. Smolen J. S., Aletaha D., and McInnes I. B., “Rheumatoid Arthritis,” Lancet 388, no. 10055 (2016): 2023–2038, 10.1016/S0140-6736(16)30173-8. [DOI] [PubMed] [Google Scholar]
- 326. Yan X., Zheng J., Ren W., et al., “O‐Glcnacylation: Roles and Potential Therapeutic Target for Bone Pathophysiology,” Cell Communication and Signaling 22, no. 1 (2024): 279, 10.1186/s12964-024-01659-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 327. Li Y. N., Chen C. W., Trinh‐Minh T., et al., “Dynamic Changes in O‐Glcnacylation Regulate Osteoclast Differentiation and Bone Loss via Nucleoporin 153,” Bone Research 10, no. 1 (2022): 51, 10.1038/s41413-022-00218-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 328. Kim H. B., Lee S. W., Mun C. H., et al., “O‐Linked N‐Acetylglucosamine Glycosylation of P65 Aggravated the Inflammation in both Fibroblast‐Like Synoviocytes Stimulated by Tumor Necrosis Factor‐Alpha and Mice With Collagen Induced Arthritis,” Arthritis Research & Therapy 17, no. 1 (2015): 248, 10.1186/s13075-015-0762-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 329. Jacques C., Floris I., and Lejeune B., “Ultra‐Low Dose Cytokines in Rheumatoid Arthritis, Three Birds With One Stone as the Rationale of the 2Larth((R)) Micro‐Immunotherapy Treatment,” International Journal of Molecular Sciences 22, no. 13 (2021), 10.3390/ijms22136717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330. Huang N., Dong H., Luo Y., et al., “Th17 Cells in Periodontitis and Its Regulation by a20,” Frontiers in Immunology 12 (2021): 742925, 10.3389/fimmu.2021.742925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331. Machacek M., Saunders H., Zhang Z., et al., “Elevated O‐Glcnacylation Enhances Pro‐Inflammatory Th17 Function by Altering the Intracellular Lipid Microenvironment,” Journal of Biological Chemistry 294, no. 22 (2019): 8973–8990, 10.1074/jbc.RA119.008373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 332. Sodi V. L., Khaku S., Krutilina R., et al., “Mtor/Myc Axis Regulates O‐Glcnac Transferase Expression and O‐Glcnacylation in Breast Cancer,” Molecular Cancer Research 13, no. 5 (2015): 923–933, 10.1158/1541-7786.MCR-14-0536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 333. Shah D. K. and Zuniga‐Pflucker J. C., “An Overview of the Intrathymic Intricacies of T Cell Development,” Journal of Immunology 192, no. 9 (2014): 4017–4023, 10.4049/jimmunol.1302259. [DOI] [PubMed] [Google Scholar]
- 334. Swamy M., Pathak S., Grzes K. M., et al., “Glucose and Glutamine Fuel Protein O‐Glcnacylation to Control T Cell Self‐Renewal and Malignancy,” Nature Immunology 17, no. 6 (2016): 712–720, 10.1038/ni.3439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335. O'Donnell N., Zachara N. E., Hart G. W., et al., “Ogt‐Dependent X‐Chromosome‐Linked Protein Glycosylation Is a Requisite Modification in Somatic Cell Function and Embryo Viability,” Molecular and Cellular Biology 24, no. 4 (2004): 1680–1690, 10.1128/MCB.24.4.1680-1690.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336. Abramowitz L. K. and Hanover J. A., “T Cell Development and the Physiological Role of O‐Glcnac,” Febs Letters 592, no. 23 (2018): 3943–3949, 10.1002/1873-3468.13159. [DOI] [PubMed] [Google Scholar]
- 337. Buck M. D., O'Sullivan D., and Pearce E. L., “T Cell Metabolism Drives Immunity,” Journal of Experimental Medicine 212, no. 9 (2015): 1345–1360, 10.1084/jem.20151159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 338. Swamy M., Pathak S., Grzes K. M., et al., “Glucose and Glutamine Fuel Protein O‐Glcnacylation to Control T Cell Self‐Renewal and Malignancy,” Nature Immunology 17, no. 6 (2016): 712–720, 10.1038/ni.3439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339. Golks A., Tran T. T., Goetschy J. F., et al., “Requirement for O‐Linked N‐Acetylglucosaminyltransferase in Lymphocytes Activation,” Embo Journal 26, no. 20 (2007): 4368–4379, 10.1038/sj.emboj.7601845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 340. Lund P. J., Elias J. E., and Davis M. M., “Global Analysis of O‐Glcnac Glycoproteins in Activated Human T Cells,” Journal of Immunology 197, no. 8 (2016): 3086–3098, 10.4049/jimmunol.1502031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 341. Abramowitz L. K., Olivier‐Van S. S., and Hanover J. A., “Chromosome Imbalance as a Driver of Sex Disparity in Disease,” Journal of Genomics 2 (2014): 77–88, 10.7150/jgen.8123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 342. Ramakrishnan P., Clark P. M., Mason D. E., et al., “Activation of the Transcriptional Function of the Nf‐Kappab Protein C‐Rel by O‐Glcnac Glycosylation,” Science Signaling 6, no. 290 (2013): ra75, 10.1126/scisignal.2004097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 343. Zhu Q., Wang H., Chai S., et al., “O‐Glcnacylation Promotes Tumor Immune Evasion by Inhibiting Pd‐L1 Lysosomal Degradation,” Proceedings of the National Academy of Sciences of the United States of America 120, no. 13 (2023): e2078171176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 344. Yuan Y., Wang L., Ge D., et al., “Exosomal O‐Glcnac Transferase from Esophageal Carcinoma Stem Cell Promotes Cancer Immunosuppression Through Up‐Regulation of Pd‐1 in Cd8(+) T Cells,” Cancer Letters 500 (2021): 98–106, 10.1016/j.canlet.2020.12.012. [DOI] [PubMed] [Google Scholar]
- 345. Wu J. L., Chiang M. F., Hsu P. H., et al., “O‐Glcnacylation Is Required for B Cell Homeostasis and Antibody Responses,” Nature Communications 8, no. 1 (2017): 1854, 10.1038/s41467-017-01677-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 346. Schiemann B., Gommerman J. L., Vora K., et al., “An Essential Role for Baff in the Normal Development of B Cells Through a Bcma‐Independent Pathway,” Science 293, no. 5537 (2001): 2111–2114, 10.1126/science.1061964. [DOI] [PubMed] [Google Scholar]
- 347. Sasaki Y., Casola S., Kutok J. L., et al., “Tnf Family Member B Cell‐Activating Factor (Baff) Receptor‐Dependent and ‐Independent Roles for Baff in B Cell Physiology,” Journal of Immunology 173, no. 4 (2004): 2245–2252, 10.4049/jimmunol.173.4.2245. [DOI] [PubMed] [Google Scholar]
- 348. Wu J. L., Chiang M. F., Hsu P. H., et al., “O‐Glcnacylation Is Required for B Cell Homeostasis and Antibody Responses,” Nature Communications 8, no. 1 (2017): 1854, 10.1038/s41467-017-01677-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 349. Mackay F., Figgett W. A., Saulep D., et al., “B‐Cell Stage and Context‐Dependent Requirements for Survival Signals from Baff and the B‐Cell Receptor,” Immunological Reviews 237, no. 1 (2010): 205–225, 10.1111/j.1600-065X.2010.00944.x. [DOI] [PubMed] [Google Scholar]
- 350. Allison D. F., Wamsley J. J., Kumar M., et al., “Modification of Rela by O‐Linked N‐Acetylglucosamine Links Glucose Metabolism to Nf‐Kappab Acetylation and Transcription,” Proceedings of the National Academy of Sciences of the United States of America 109, no. 42 (2012): 16888–16893, 10.1073/pnas.1208468109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 351. Li X., Zhang Z., Li L., et al., “Myeloid‐Derived Cullin 3 Promotes Stat3 Phosphorylation by Inhibiting Ogt Expression and Protects against Intestinal Inflammation,” Journal of Experimental Medicine 214, no. 4 (2017): 1093–1109, 10.1084/jem.20161105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 352. Staples K. J., Smallie T., Williams L. M., et al., “Il‐10 Induces Il‐10 in Primary Human Monocyte‐Derived Macrophages via the Transcription Factor Stat3,” Journal of Immunology 178, no. 8 (2007): 4779–4785, 10.4049/jimmunol.178.8.4779. [DOI] [PubMed] [Google Scholar]
- 353. Takeda K., Clausen B. E., Kaisho T., et al., “Enhanced Th1 Activity and Development of Chronic Enterocolitis in Mice Devoid of Stat3 in Macrophages and Neutrophils,” Immunity 10, no. 1 (1999): 39–49, 10.1016/S1074-7613(00)80005-9. [DOI] [PubMed] [Google Scholar]
- 354. Bugide S., Green M. R., and Wajapeyee N., “Inhibition of Enhancer of Zeste Homolog 2 (Ezh2) Induces Natural Killer Cell‐Mediated Eradication of Hepatocellular Carcinoma Cells,” Proceedings of the National Academy of Sciences of the United States of America 115, no. 15 (2018): E3509–E3518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 355. Yin J., Leavenworth J. W., Li Y., et al., “Ezh2 Regulates Differentiation and Function of Natural Killer Cells Through Histone Methyltransferase Activity,” Proceedings of the National Academy of Sciences of the United States of America 112, no. 52 (2015): 15988–15993, 10.1073/pnas.1521740112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 356. Yin J., Leavenworth J. W., Li Y., et al., “Ezh2 Regulates Differentiation and Function of Natural Killer Cells Through Histone Methyltransferase Activity,” Proceedings of the National Academy of Sciences of the United States of America 112, no. 52 (2015): 15988–15993, 10.1073/pnas.1521740112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 357. Chang Y. H., Weng C. L., and Lin K. I., “O‐Glcnacylation and Its Role in the Immune System,” Journal of Biomedical Science 27, no. 1 (2020): 57, 10.1186/s12929-020-00648-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 358. Deng Y., Kerdiles Y., Chu J., et al., “Transcription Factor Foxo1 Is a Negative Regulator of Natural Killer Cell Maturation and Function,” Immunity 42, no. 3 (2015): 457–470, 10.1016/j.immuni.2015.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 359. Lenzen S. and Panten U., “Alloxan: History and Mechanism of Action,” Diabetologia 31, no. 6 (1988): 337–342, 10.1007/BF02341500. [DOI] [PubMed] [Google Scholar]
- 360. Lenzen S. and Panten U., “Alloxan: History and Mechanism of Action,” Diabetologia 31, no. 6 (1988): 337–342, 10.1007/BF02341500. [DOI] [PubMed] [Google Scholar]
- 361. Macauley M. S., Whitworth G. E., Debowski A. W., et al., “O‐Glcnacase Uses Substrate‐Assisted Catalysis: Kinetic Analysis and Development of Highly Selective Mechanism‐Inspired Inhibitors,” Journal of Biological Chemistry 280, no. 27 (2005): 25313–25322, 10.1074/jbc.M413819200. [DOI] [PubMed] [Google Scholar]
- 362. Konrad R. J., Zhang F., Hale J. E., et al., “Alloxan Is an Inhibitor of the Enzyme O‐Linked N‐Acetylglucosamine Transferase,” Biochemical and Biophysical Research Communications 293, no. 1 (2002): 207–212, 10.1016/S0006-291X(02)00200-0. [DOI] [PubMed] [Google Scholar]
- 363. Liu J., Yu Y., Fan Y. Z., et al., “Cardiovascular Effects of Endomorphins in Alloxan‐Induced Diabetic Rats,” Peptides 26, no. 4 (2005): 607–614, 10.1016/j.peptides.2004.11.017. [DOI] [PubMed] [Google Scholar]
- 364. Zhang H., Gao G., and Brunk U. T., “Extracellular Reduction of Alloxan Results in Oxygen Radical‐Mediated Attack on Plasma and Lysosomal Membranes,” Apmis 100, no. 4 (1992): 317–325, 10.1111/j.1699-0463.1992.tb00878.x. [DOI] [PubMed] [Google Scholar]
- 365. Kapuria V., Rohrig U. F., Waridel P., et al., “The Conserved Threonine‐Rich Region of the Hcf‐1(Pro) Repeat Activates Promiscuous Ogt:Udp‐Glcnac Glycosylation and Proteolysis Activities,” Journal of Biological Chemistry 293, no. 46 (2018): 17754–17768, 10.1074/jbc.RA118.004185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 366. Liu T. W., Zandberg W. F., Gloster T. M., et al., “Metabolic Inhibitors of O‐Glcnac Transferase That Act in Vivo Implicate Decreased O‐Glcnac Levels in Leptin‐Mediated Nutrient Sensing,” Angewandte Chemie‐International Edition 57, no. 26 (2018): 7644–7648, 10.1002/anie.201803254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 367. Gloster T. M., Zandberg W. F., Heinonen J. E., et al., “Hijacking a Biosynthetic Pathway Yields a Glycosyltransferase Inhibitor Within Cells,” Nature Chemical Biology 7, no. 3 (2011): 174–181, 10.1038/nchembio.520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 368. Wang T. F., Feng Z. Q., Sun Y. W., et al., “Disruption of O‐Glcnacylation Homeostasis Induced Ovarian Granulosa Cell Injury in Bovine,” International Journal of Molecular Sciences 23, no. 14 (2022), 10.3390/ijms23147815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 369. Haltiwanger R. S., Grove K., and Philipsberg G. A., “Modulation of O‐Linked N‐Acetylglucosamine Levels on Nuclear and Cytoplasmic Proteins in Vivo Using the Peptide O‐Glcnac‐Beta‐N‐Acetylglucosaminidase Inhibitor O‐(2‐Acetamido‐2‐Deoxy‐D‐Glucopyranosylidene)Amino‐N‐Phenylcarbamate,” Journal of Biological Chemistry 273, no. 6 (1998): 3611–3617, 10.1074/jbc.273.6.3611. [DOI] [PubMed] [Google Scholar]
- 370. Yuzwa S. A., Macauley M. S., Heinonen J. E., et al., “A Potent Mechanism‐Inspired O‐Glcnacase Inhibitor That Blocks Phosphorylation of Tau in Vivo,” Nature Chemical Biology 4, no. 8 (2008): 483–490, 10.1038/nchembio.96. [DOI] [PubMed] [Google Scholar]
- 371. James L. R., Tang D., Ingram A., et al., “Flux Through the Hexosamine Pathway Is a Determinant of Nuclear Factor Kappab‐ Dependent Promoter Activation,” Diabetes 51, no. 4 (2002): 1146–1156, 10.2337/diabetes.51.4.1146. [DOI] [PubMed] [Google Scholar]
- 372. Hennebicq‐Reig S., Lesuffleur T., Capon C., et al., “Permanent Exposure of Mucin‐Secreting Ht‐29 Cells to Benzyl‐N‐Acetyl‐Alpha‐D‐Galactosaminide Induces Abnormal O‐Glycosylation of Mucins and Inhibits Constitutive and Stimulated Muc5Ac Secretion,” Biochemical Journal 334, no. Pt 1 (1998): 283–295, 10.1042/bj3340283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 373. Olivier‐Van S. S., Drougat L., Dehennaut V., et al., “Serum‐Stimulated Cell Cycle Entry Promotes Ncogt Synthesis Required for Cyclin D Expression,” Oncogenesis 1, no. 12 (2012): e36, 10.1038/oncsis.2012.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 374. Gloster T. M., Zandberg W. F., Heinonen J. E., et al., “Hijacking a Biosynthetic Pathway Yields a Glycosyltransferase Inhibitor Within Cells,” Nature Chemical Biology 7, no. 3 (2011): 174–181, 10.1038/nchembio.520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 375. Liu T. W., Zandberg W. F., Gloster T. M., et al., “Metabolic Inhibitors of O‐Glcnac Transferase That Act in Vivo Implicate Decreased O‐Glcnac Levels in Leptin‐Mediated Nutrient Sensing,” Angewandte Chemie‐International Edition 57, no. 26 (2018): 7644–7648, 10.1002/anie.201803254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 376. Lima V. V., Giachini F. R., Carneiro F. S., et al., “O‐Glcnacylation Contributes to the Vascular Effects of Et‐1 via Activation of the Rhoa/Rho‐Kinase Pathway,” Cardiovascular Research 89, no. 3 (2011): 614–622, 10.1093/cvr/cvq338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 377. Trapannone R., Rafie K., and van Aalten D. M., “O‐Glcnac Transferase Inhibitors: Current Tools and Future Challenges,” Biochemical Society Transactions 44, no. 1 (2016): 88–93, 10.1042/BST20150189. [DOI] [PubMed] [Google Scholar]
- 378. Rafie K., Gorelik A., Trapannone R., et al., “Thio‐Linked Udp‐Peptide Conjugates as O‐Glcnac Transferase Inhibitors,” Bioconjugate Chemistry 29, no. 6 (2018): 1834–1840, 10.1021/acs.bioconjchem.8b00194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 379. Gross B. J., Kraybill B. C., and Walker S., “Discovery of O‐Glcnac Transferase Inhibitors,” Journal of the American Chemical Society 127, no. 42 (2005): 14588–14589, 10.1021/ja0555217. [DOI] [PubMed] [Google Scholar]
- 380. Trapannone R., Rafie K., and van Aalten D. M., “O‐Glcnac Transferase Inhibitors: Current Tools and Future Challenges,” Biochemical Society Transactions 44, no. 1 (2016): 88–93, 10.1042/BST20150189. [DOI] [PubMed] [Google Scholar]
- 381. Rehman S., Obaid A., Naz A., et al., “Model‐Based in Silico Analysis of the Pi3K/Akt Pathway: The Elucidation of Cross‐Talk between Diabetes and Breast Cancer,” Peerj 6 (2018): e5917, 10.7717/peerj.5917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 382. Jiang J., Lazarus M. B., Pasquina L., et al., “A Neutral Diphosphate Mimic Crosslinks the Active Site of Human O‐Glcnac Transferase,” Nature Chemical Biology 8, no. 1 (2011): 72–77, 10.1038/nchembio.711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 383. Yang Y., Yan Y., Yin J., et al., “O‐Glcnacylation of Ythdf2 Promotes Hbv‐Related Hepatocellular Carcinoma Progression in an N(6)‐Methyladenosine‐Dependent Manner,” Signal Transduction and Targeted Therapy 8, no. 1 (2023): 63, 10.1038/s41392-023-01316-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 384. Wu Y. K., Liu M., Zhou H. L., et al., “O‐Linked Beta‐N‐Acetylglucosamine Transferase Regulates Macrophage Polarization in Diabetic Periodontitis: In Vivo and in Vitro Study,” World Journal of Diabetes 16, no. 3 (2025): 95092, 10.4239/wjd.v16.i3.95092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 385. Itkonen H. M., Urbanucci A., Martin S. E., et al., “High Ogt Activity Is Essential for Myc‐Driven Proliferation of Prostate Cancer Cells,” Theranostics 9, no. 8 (2019): 2183–2197, 10.7150/thno.30834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 386. Yuzwa S. A., Macauley M. S., Heinonen J. E., et al., “A Potent Mechanism‐Inspired O‐Glcnacase Inhibitor That Blocks Phosphorylation of Tau in Vivo,” Nature Chemical Biology 4, no. 8 (2008): 483–490, 10.1038/nchembio.96. [DOI] [PubMed] [Google Scholar]
- 387. Trapannone R., Rafie K., and van Aalten D. M., “O‐Glcnac Transferase Inhibitors: Current Tools and Future Challenges,” Biochemical Society Transactions 44, no. 1 (2016): 88–93, 10.1042/BST20150189. [DOI] [PubMed] [Google Scholar]
- 388. Rafie K., Gorelik A., Trapannone R., et al., “Thio‐Linked Udp‐Peptide Conjugates as O‐Glcnac Transferase Inhibitors,” Bioconjugate Chemistry 29, no. 6 (2018): 1834–1840, 10.1021/acs.bioconjchem.8b00194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 389. Ortiz‐Meoz R. F., Jiang J., Lazarus M. B., et al., “A Small Molecule That Inhibits Ogt Activity in Cells,” Acs Chemical Biology 10, no. 6 (2015): 1392–1397, 10.1021/acschembio.5b00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 390. Borodkin V. S., Schimpl M., Gundogdu M., et al., “Bisubstrate Udp‐Peptide Conjugates as Human O‐Glcnac Transferase Inhibitors,” Biochemical Journal 457, no. 3 (2014): 497–502, 10.1042/BJ20131272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 391. Song Y., “Catch in Nucleus,” Nature Chemical Biology 19, no. 11 (2023): 1291, 10.1038/s41589-023-01470-8. [DOI] [PubMed] [Google Scholar]
- 392. Villagrasa M., Guillamon M., Navarro A., et al., “Development of a Pressurized Liquid Extraction‐Solid‐Phase Extraction Followed by Liquid Chromatography‐Electrospray Ionization Tandem Mass Spectrometry Method for the Quantitative Determination of Benzoxazolinones and Their Degradation Products in Agricultural Soil,” Journal of Chromatography A 1179, no. 2 (2008): 190–197. [DOI] [PubMed] [Google Scholar]
- 393. Zhou Q., Yang M., Fu J., et al., “Kif1a Promotes Neuroendocrine Differentiation in Prostate Cancer by Regulating the Ogt‐Mediated O‐Glcnacylation,” Cell Death & Disease 15, no. 11 (2024): 796, 10.1038/s41419-024-07142-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 394. Itkonen H. M., Urbanucci A., Martin S. E., et al., “High Ogt Activity Is Essential for Myc‐Driven Proliferation of Prostate Cancer Cells,” Theranostics 9, no. 8 (2019): 2183–2197, 10.7150/thno.30834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 395. Sharma N. S., Gupta V. K., Dauer P., et al., “O‐Glcnac Modification of Sox2 Regulates Self‐Renewal in Pancreatic Cancer by Promoting Its Stability,” Theranostics 9, no. 12 (2019): 3410–3424, 10.7150/thno.32615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 396. Zhang C., Yin X., Jiang J., et al., “Downregulation of Gfpt2 Enhances Cisplatin Chemotherapy Sensitivity in Stk11/Kras Mutant Non‐Small Cell Lung Cancer by Regulating the Hexosamine Biosynthesis Pathway, Resisting Tumor Growth,” Cytokine 191 (2025): 156943, 10.1016/j.cyto.2025.156943. [DOI] [PubMed] [Google Scholar]
- 397. Makwana V., Dukie A. S., and Rudrawar S., “Investigating the Impact of Ogt Inhibition on Doxorubicin‐ And Docetaxel‐Induced Cytotoxicity in Pc‐3 and Wpmy‐1 Cells,” International Journal of Toxicology 39, no. 6 (2020): 586–593, 10.1177/1091581820948433. [DOI] [PubMed] [Google Scholar]
- 398. Lee D. E., Lee G. Y., Lee H. M., et al., “Synergistic Apoptosis by Combination of Metformin and an O‐Glcnacylation Inhibitor in Colon Cancer Cells,” Cancer Cell International 23, no. 1 (2023): 108, 10.1186/s12935-023-02954-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 399. Loison I., Pioger A., Paget S., et al., “O‐Glcnacylation Inhibition Redirects the Response of Colon Cancer Cells to Chemotherapy from Senescence to Apoptosis,” Cell Death & Disease 15, no. 10 (2024): 762, 10.1038/s41419-024-07131-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 400. Lee S. J., Lee D. E., Choi S. Y., et al., “Osmi‐1 Enhances Trail‐Induced Apoptosis Through Er Stress and Nf‐Kappab Signaling in Colon Cancer Cells,” International Journal of Molecular Sciences 22, no. 20 (2021), 10.3390/ijms222011073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 401. Lee S. J. and Kwon O. S., “O‐Glcnac Transferase Inhibitor Synergistically Enhances Doxorubicin‐Induced Apoptosis in Hepg2 Cells,” Cancers 12, no. 11 (2020), 10.3390/cancers12113154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 402. Lin B., Chai S., Zhang Q., et al., “Sialic Acid‐Modified O‐Glcnac Transferase Inhibitor Liposome Presents Antitumor Effect in Hepatocellular Carcinoma,” Molecular Pharmaceutics 21, no. 1 (2024): 102–112, 10.1021/acs.molpharmaceut.3c00451. [DOI] [PubMed] [Google Scholar]
- 403. Lee J., Koo G. B., Park J., et al., “Downregulation of O‐Glcnacylation Enhances Etoposide‐Induced P53‐Mediated Apoptosis in Hepg2 Human Liver Cancer Cells,” Febs Open Bio 15, no. 7 (2025): 1176–1188, 10.1002/2211-5463.70028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 404. Wu J., Tan Z., Li H., et al., “Melatonin Reduces Proliferation and Promotes Apoptosis of Bladder Cancer Cells by Suppressing O‐Glcnacylation of Cyclin‐Dependent‐Like Kinase 5,” Journal of Pineal Research 71, no. 3 (2021): e12765, 10.1111/jpi.12765. [DOI] [PubMed] [Google Scholar]
- 405. Lu S., Liao Z., Lu X., et al., “Hyperglycemia Acutely Increases Cytosolic Reactive Oxygen Species via O‐Linked Glcnacylation and Camkii Activation in Mouse Ventricular Myocytes,” Circulation Research 126, no. 10 (2020): e80–e96, 10.1161/CIRCRESAHA.119.316288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 406. Whitworth G. E., Macauley M. S., Stubbs K. A., et al., “Analysis of Pugnac and Nag‐Thiazoline as Transition State Analogues for Human O‐Glcnacase: Mechanistic and Structural Insights into Inhibitor Selectivity and Transition State Poise,” Journal of the American Chemical Society 129, no. 3 (2007): 635–644, 10.1021/ja065697o. [DOI] [PubMed] [Google Scholar]
- 407. Park S. Y., Ryu J., and Lee W., “O‐Glcnac Modification on Irs‐1 and Akt2 by Pugnac Inhibits Their Phosphorylation and Induces Insulin Resistance in Rat Primary Adipocytes,” Experimental and Molecular Medicine 37, no. 3 (2005): 220–229, 10.1038/emm.2005.30. [DOI] [PubMed] [Google Scholar]
- 408. Zhou W., Tang Q., Wang S., et al., “Local Thiamet‐G Delivery by a Thermosensitive Hydrogel Confers Ischemic Cardiac Repair via Myeloid M2‐Like Activation in a Stat6 O‐Glcnacylation‐Dependent Manner,” International Immunopharmacology 131 (2024): 111883, 10.1016/j.intimp.2024.111883. [DOI] [PubMed] [Google Scholar]
- 409. Yuzwa S. A., Macauley M. S., Heinonen J. E., et al., “A Potent Mechanism‐Inspired O‐Glcnacase Inhibitor That Blocks Phosphorylation of Tau in Vivo,” Nature Chemical Biology 4, no. 8 (2008): 483–490, 10.1038/nchembio.96. [DOI] [PubMed] [Google Scholar]
- 410. Denis M., Dupas T., Persello A., et al., “An O‐Glcnacylomic Approach Reveals Acly as a Potential Target in Sepsis in the Young Rat,” International Journal of Molecular Sciences 22, no. 17 (2021), 10.3390/ijms22179236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 411. Adibekian A., Martin B. R., Wang C., et al., “Click‐Generated Triazole Ureas as Ultrapotent in Vivo‐Active Serine Hydrolase Inhibitors,” Nature Chemical Biology 7, no. 7 (2011): 469–478, 10.1038/nchembio.579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 412. Wieboldt R. and Laubli H., “Glycosaminoglycans in Cancer Therapy,” American Journal of Physiology‐Cell Physiology 322, no. 6 (2022): C1187–C1200, 10.1152/ajpcell.00063.2022. [DOI] [PubMed] [Google Scholar]
- 413. Dorfmueller H. C., Borodkin V. S., Schimpl M., et al., “Glcnacstatins Are Nanomolar Inhibitors of Human O‐Glcnacase Inducing Cellular Hyper‐O‐Glcnacylation,” Biochemical Journal 420, no. 2 (2009): 221–227, 10.1042/BJ20090110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 414. Dorfmueller H. C., Borodkin V. S., Schimpl M., et al., “Glcnacstatin: A Picomolar, Selective O‐Glcnacase Inhibitor That Modulates Intracellular O‐Glcnacylation Levels,” Journal of the American Chemical Society 128, no. 51 (2006): 16484–16485, 10.1021/ja066743n. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 415. Adibekian A., Martin B. R., Wang C., et al., “Click‐Generated Triazole Ureas as Ultrapotent in Vivo‐Active Serine Hydrolase Inhibitors,” Nature Chemical Biology 7, no. 7 (2011): 469–478, 10.1038/nchembio.579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 416. Xie S., Jin N., Gu J., et al., “O‐Glcnacylation of Protein Kinase a Catalytic Subunits Enhances Its Activity: A Mechanism Linked to Learning and Memory Deficits in Alzheimer's Disease,” Aging Cell 15, no. 3 (2016): 455–464, 10.1111/acel.12449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 417. Herzog R., Bender T. O., Vychytil A., et al., “Dynamic O‐Linked N‐Acetylglucosamine Modification of Proteins Affects Stress Responses and Survival of Mesothelial Cells Exposed to Peritoneal Dialysis Fluids,” Journal of the American Society of Nephrology 25, no. 12 (2014): 2778–2788, 10.1681/ASN.2013101128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 418. Whitworth G. E., Macauley M. S., Stubbs K. A., et al., “Analysis of Pugnac and Nag‐Thiazoline as Transition State Analogues for Human O‐Glcnacase: Mechanistic and Structural Insights into Inhibitor Selectivity and Transition State Poise,” Journal of the American Chemical Society 129, no. 3 (2007): 635–644, 10.1021/ja065697o. [DOI] [PubMed] [Google Scholar]
- 419. Dorfmueller H. C., Borodkin V. S., Schimpl M., et al., “Cell‐Penetrant, Nanomolar O‐Glcnacase Inhibitors Selective against Lysosomal Hexosaminidases,” Chemistry & Biology 17, no. 11 (2010): 1250–1255, 10.1016/j.chembiol.2010.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 420. Ivanov V. I. and Karpeisky M. Y., “Dynamic Three‐Dimensional Model for Enzymic Transamination,” Advances in Enzymology and Related Areas of Molecular Biology 32 (1969): 21–53. [DOI] [PubMed] [Google Scholar]
- 421. Coffey G. L., Ehrlich J., Fisher M. W., et al., “6‐Diazo‐5‐Oxo‐L‐Norleucine, a New Tumor‐Inhibitory Substance. I. Biologic Studies,” Antibiotics and Chemotherapy (Northfield) 6, no. 8 (1956): 487–497. [PubMed] [Google Scholar]
- 422. Stock C. C., Reilly H. C., Buckley S. M., et al., “Azaserine, a New Tumour‐Inhibitory Substance; Studies With Crocker Mouse Sarcoma 180,” Nature 173, no. 4393 (1954): 71–72. [DOI] [PubMed] [Google Scholar]
- 423. Wellen K. E., Lu C., Mancuso A., et al., “The Hexosamine Biosynthetic Pathway Couples Growth Factor‐Induced Glutamine Uptake to Glucose Metabolism,” Genes & Development 24, no. 24 (2010): 2784–2799, 10.1101/gad.1985910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 424. Rais R., Jancarik A., Tenora L., et al., “Discovery of 6‐Diazo‐5‐Oxo‐L‐Norleucine (Don) Prodrugs With Enhanced Csf Delivery in Monkeys: A Potential Treatment for Glioblastoma,” Journal of Medicinal Chemistry 59, no. 18 (2016): 8621–8633, 10.1021/acs.jmedchem.6b01069. [DOI] [PubMed] [Google Scholar]
- 425. Le A., Lane A. N., Hamaker M., et al., “Glucose‐Independent Glutamine Metabolism via Tca Cycling for Proliferation and Survival in B Cells,” Cell Metabolism 15, no. 1 (2012): 110–121, 10.1016/j.cmet.2011.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 426. Bolanle I. O., Riches‐Suman K., Williamson R., et al., “Emerging Roles of Protein O‐Glcnacylation in Cardiovascular Diseases: Insights and Novel Therapeutic Targets,” Pharmacological Research 165 (2021): 105467, 10.1016/j.phrs.2021.105467. [DOI] [PubMed] [Google Scholar]
- 427. Olvera A., Martinez J. P., Casadella M., et al., “Benzyl‐2‐Acetamido‐2‐Deoxy‐Alpha‐D‐Galactopyranoside Increases Human Immunodeficiency Virus Replication and Viral Outgrowth Efficacy in Vitro,” Frontiers in Immunology 8 (2017): 2010, 10.3389/fimmu.2017.02010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 428. Hennebicq‐Reig S., Lesuffleur T., Capon C., et al., “Permanent Exposure of Mucin‐Secreting Ht‐29 Cells to Benzyl‐N‐Acetyl‐Alpha‐D‐Galactosaminide Induces Abnormal O‐Glycosylation of Mucins and Inhibits Constitutive and Stimulated Muc5Ac Secretion,” Biochemical Journal 334, no. Pt 1 (1998): 283–295, 10.1042/bj3340283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 429. Dorfmueller H. C., Borodkin V. S., Schimpl M., et al., “Glcnacstatins Are Nanomolar Inhibitors of Human O‐Glcnacase Inducing Cellular Hyper‐O‐Glcnacylation,” Biochemical Journal 420, no. 2 (2009): 221–227, 10.1042/BJ20090110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 430. Dorfmueller H. C., Borodkin V. S., Schimpl M., et al., “Glcnacstatin: A Picomolar, Selective O‐Glcnacase Inhibitor That Modulates Intracellular O‐Glcnacylation Levels,” Journal of the American Chemical Society 128, no. 51 (2006): 16484–16485, 10.1021/ja066743n. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 431. Xun Z., Li T., and Xue X., “The Application Strategy of Liposomes in Organ Targeting Therapy,” Wiley Interdisciplinary Reviews‐Nanomedicine and Nanobiotechnology 16, no. 2 (2024): e1955, 10.1002/wnan.1955. [DOI] [PubMed] [Google Scholar]
- 432. Yu H., Jin F., Liu D., et al., “Ros‐Responsive Nano‐Drug Delivery System Combining Mitochondria‐Targeting Ceria Nanoparticles With Atorvastatin for Acute Kidney Injury,” Theranostics 10, no. 5 (2020): 2342–2357, 10.7150/thno.40395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 433. Bruni R., Possenti P., Bordignon C., et al., “Ultrasmall Polymeric Nanocarriers for Drug Delivery to Podocytes in Kidney Glomerulus,” Journal of Controlled Release 255 (2017): 94–107, 10.1016/j.jconrel.2017.04.005. [DOI] [PubMed] [Google Scholar]
- 434. Lai X., Geng X., Tan L., et al., “A Ph‐Responsive System Based on Fluorescence Enhanced Gold Nanoparticles for Renal Targeting Drug Delivery and Fibrosis Therapy,” International Journal of Nanomedicine 15 (2020): 5613–5627, 10.2147/IJN.S260069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 435. Qiao H., Sun M., Su Z., et al., “Kidney‐Specific Drug Delivery System for Renal Fibrosis Based on Coordination‐Driven Assembly of Catechol‐Derived Chitosan,” Biomaterials 35, no. 25 (2014): 7157–7171, 10.1016/j.biomaterials.2014.04.106. [DOI] [PubMed] [Google Scholar]
- 436. Guo L., Luo S., Du Z., et al., “Targeted Delivery of Celastrol to Mesangial Cells Is Effective against Mesangioproliferative Glomerulonephritis,” Nature Communications 8, no. 1 (2017): 878, 10.1038/s41467-017-00834-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 437. Chen D., Han S., Zhu Y., et al., “Kidney‐Targeted Drug Delivery via Rhein‐Loaded Polyethyleneglycol‐Co‐Polycaprolactone‐Co‐Polyethylenimine Nanoparticles for Diabetic Nephropathy Therapy,” International Journal of Nanomedicine 13 (2018): 3507–3527, 10.2147/IJN.S166445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 438. Williams R. M., Shah J., Ng B. D., et al., “Mesoscale Nanoparticles Selectively Target the Renal Proximal Tubule Epithelium,” Nano Letters 15, no. 4 (2015): 2358–2364, 10.1021/nl504610d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 439. Han S. J., Williams R. M., D'Agati V., et al., “Selective Nanoparticle‐Mediated Targeting of Renal Tubular Toll‐Like Receptor 9 Attenuates Ischemic Acute Kidney Injury,” Kidney International 98, no. 1 (2020): 76–87, 10.1016/j.kint.2020.01.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 440. Liu S., Zhao M., Zhou Y., et al., “A Self‐Assembling Peptide Hydrogel‐Based Drug Co‐Delivery Platform to Improve Tissue Repair after Ischemia‐Reperfusion Injury,” Acta Biomaterialia 103 (2020): 102–114, 10.1016/j.actbio.2019.12.011. [DOI] [PubMed] [Google Scholar]
- 441. Gu M., Lu L., Wei Q., et al., “Improved Oral Bioavailability and Anti‐Chronic Renal Failure Activity of Chrysophanol via Mixed Polymeric Micelles,” Journal of Microencapsulation 38, no. 1 (2021): 47–60, 10.1080/02652048.2020.1849440. [DOI] [PubMed] [Google Scholar]
- 442. Binkhathlan Z., Ali R., Qamar W., et al., “Pharmacokinetic and Tissue Distribution of Orally Administered Cyclosporine a‐Loaded Poly(Ethylene Oxide)‐Block‐Poly(Epsilon‐Caprolactone) Micelles versus Sandimmune((R)) in Rats,” Pharmaceutical Research 38, no. 1 (2021): 51–65, 10.1007/s11095-021-02990-5. [DOI] [PubMed] [Google Scholar]
- 443. Qin X., Xu Y., Zhou X., et al., “An Injectable Micelle‐Hydrogel Hybrid for Localized and Prolonged Drug Delivery in the Management of Renal Fibrosis,” Acta Pharmaceutica Sinica B 11, no. 3 (2021): 835–847, 10.1016/j.apsb.2020.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 444. Katsumi H., Takashima R., Suzuki H., et al., “S‐Nitrosylated L‐Serine‐Modified Dendrimer as a Kidney‐Targeting Nitric Oxide Donor for Prevention of Renal Ischaemia/Reperfusion Injury,” Free Radical Research 54, no. 11–12 (2020): 841–847, 10.1080/10715762.2019.1697437. [DOI] [PubMed] [Google Scholar]
- 445. Kobayashi H., Jo S. K., Kawamoto S., et al., “Polyamine Dendrimer‐Based Mri Contrast Agents for Functional Kidney Imaging to Diagnose Acute Renal Failure,” Journal of Magnetic Resonance Imaging 20, no. 3 (2004): 512–518, 10.1002/jmri.20147. [DOI] [PubMed] [Google Scholar]
- 446. Ding W. Y., Kuzmuk V., Hunter S., et al., “Adeno‐Associated Virus Gene Therapy Prevents Progression of Kidney Disease in Genetic Models of Nephrotic Syndrome,” Science Translational Medicine 15, no. 708 (2023): eabc8226, 10.1126/scitranslmed.abc8226. [DOI] [PubMed] [Google Scholar]
- 447. Haas M., Kluppel A. C., Wartna E. S., et al., “Drug‐Targeting to the Kidney: Renal Delivery and Degradation of a Naproxen‐Lysozyme Conjugate in Vivo,” Kidney International 52, no. 6 (1997): 1693–1699, 10.1038/ki.1997.504. [DOI] [PubMed] [Google Scholar]
- 448. Pan X., Xie F., Xiao D., et al., “Design, Synthesis, and Renal Targeting of Methylprednisolone‐Lysozyme,” International Journal of Molecular Sciences 21, no. 6 (2020), 10.3390/ijms21061922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 449. Dolman M. E., Fretz M. M., Segers G. J., et al., “Renal Targeting of Kinase Inhibitors,” International Journal of Pharmaceutics 364, no. 2 (2008): 249–257, 10.1016/j.ijpharm.2008.04.040. [DOI] [PubMed] [Google Scholar]
- 450. Zheng X. P., Nie Q., Feng J., et al., “Kidney‐Targeted Baicalin‐Lysozyme Conjugate Ameliorates Renal Fibrosis in Rats With Diabetic Nephropathy Induced by Streptozotocin,” Bmc Nephrology 21, no. 1 (2020): 174, 10.1186/s12882-020-01833-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 451. Bidwell G. R., Fokt I., Priebe W., et al., “Development of Elastin‐Like Polypeptide for Thermally Targeted Delivery of Doxorubicin,” Biochemical Pharmacology 73, no. 5 (2007): 620–631, 10.1016/j.bcp.2006.10.028. [DOI] [PubMed] [Google Scholar]
- 452. Kuna M., Mahdi F., Chade A. R., et al., “Molecular Size Modulates Pharmacokinetics, Biodistribution, and Renal Deposition of the Drug Delivery Biopolymer Elastin‐Like Polypeptide,” Scientific Reports 8, no. 1 (2018): 7923, 10.1038/s41598-018-24897-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 453. Wang X., Lin Y., Zeng Y., et al., “Effects of Mycophenolic Acid‐Glucosamine Conjugates on the Base of Kidney Targeted Drug Delivery,” International Journal of Pharmaceutics 456, no. 1 (2013): 223–234, 10.1016/j.ijpharm.2013.07.064. [DOI] [PubMed] [Google Scholar]
- 454. Yuan Z. X., Sun X., Gong T., et al., “Randomly 50% N‐Acetylated Low Molecular Weight Chitosan as a Novel Renal Targeting Carrier,” Journal of Drug Targeting 15, no. 4 (2007): 269–278, 10.1080/10611860701289875. [DOI] [PubMed] [Google Scholar]
- 455. Shang S., Li X., Wang H., et al., “Targeted Therapy of Kidney Disease With Nanoparticle Drug Delivery Materials,” Bioactive Materials 37 (2024): 206–221, 10.1016/j.bioactmat.2024.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The authors have nothing to report.
