International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Sepsis, resulting from a dysregulated host immune response to infection and characterized by the excessive release of inflammatory mediators and abnormal immune cell activation, remains a major challenge in global healthcare (1). In 2017, ~11.0 million sepsis-related mortalities were reported worldwide, representing 19.7% of all global mortalities. Despite a 52.8% decline in the age-standardized sepsis mortality from 1990 to 2017, sepsis remains a leading cause of death, prompting researchers to explore new treatment strategies (2).
The attachment of N-acetylglucosamine (GlcNAc) via an oxygen linkage (O-GlcNAcylation) is a post-translational modification (PTM) of the serine (Ser) and threonine (Thr) residues in proteins. This PTM can regulate the activation, proliferation and apoptosis of immune cells, as well as the production of inflammatory mediators, by influencing various metabolic pathways and cellular processes (3,4). The regulatory role of O-GlcNAcylation is particularly important in the context of sepsis (5), as it not only regulates the immune response to infection but also controls autophagy, the main mechanism by which cells clear pathogens and damaged organelles. Therefore, O-GlcNAcylation may play a critical role in the development of sepsis by affecting immune cell function and the efficiency of autophagy. The present review aims to investigate the underlying mechanisms of O-GlcNAcylation as a bridge between infection immunity and autophagy in sepsis, and to assess its potential as a therapeutic target.
Sepsis is a life-threatening condition characterized by organ dysfunction resulting from dysregulation of the host response to infection (6), and affected ~48.9 million individuals worldwide in 2017, posing a great threat to health (2). The pathogens responsible for sepsis include bacteria, fungi, viruses and parasites.
Immune cells play a key role in the pathogenesis of sepsis. Immunological studies have shown that the host immune response in sepsis involves several sequential or concurrent processes that lead to excessive inflammation and immune suppression. This immune dysfunction results in impaired innate and adaptive immune responses (7).
During the initial phase of sepsis, the body mounts a robust inflammatory response to infection. Immune cells such as macrophages, dendritic cells, neutrophils and lymphocytes are activated following the recognition of pathogen-associated and damage-associated molecular patterns, which triggers the inflammatory response (8). As sepsis progresses, patients often develop immunosuppression, characterized by diminished immune cell function, particularly decreased activity of T cells and B cells, and increased activity of regulatory T (Treg) cells. This immunosuppressive state increases susceptibility to secondary infections and may lead to clinical deterioration and even death (Fig. 1) (8,9).
In sepsis, autophagy plays a vital role in cytoplasmic quality control, cellular metabolism, and innate and adaptive immune responses. It involves a variety of immune cells, including neutrophils, macrophages, T cells and B cells (10,11). In neutrophils, autophagy facilitates the clearance of pathogenic microorganisms, supports cellular function and prevents cell death. Studies have shown that the autophagic activity of neutrophils increases during sepsis, which may help to maintain their survival and function. However, the hyperactivation or inhibition of autophagy can impair the function of neutrophils, thereby affecting the progression of sepsis (12,13). In macrophages, autophagy not only facilitates the clearance of pathogens but also regulates the inflammatory response. During sepsis, the autophagic activity of macrophages may be suppressed, leading to the excessive release of inflammatory mediators and amplification of the inflammatory response. Conversely, the activation of autophagy can attenuate macrophage-mediated inflammation, offering protection against sepsis (14). Autophagy is also essential for the survival and function of T cells. In sepsis, the autophagic activity of T cells is weakened, which results in increased T-cell apoptosis and immunosuppression. In a murine model of sepsis, mice deficient in a T-cell-specific autophagy gene known as autophagy related 7 exhibited a higher mortality rate and greater immunosuppression than was observed in wild-type mice, suggesting a protective role for T-cell autophagy in the regulation of T-cell apoptosis and immunosuppression (10,15). The role of B cells in sepsis has not been fully elucidated; however, autophagy has been indicated to affect the immune response in sepsis by regulating B cell maturation and antibody production, with the activation of autophagy being suggested to contribute to the maintenance of B cell function and prevent immunosuppression (10). In summary, the effect of autophagy on immune cells in sepsis is multifaceted, involving various immune cells, such as neutrophils, macrophages, T cells and B cells. Activation or inhibition of autophagy may have an important effect on the pathogenesis of sepsis, and the regulation of autophagy may be a novel therapeutic strategy for sepsis.
O-GlcNAcylation is a crucial PTM in which GlcNAc molecules attach to Ser or Thr residues in proteins. O-GlcNAc modifications differ from other types of glycosylation in several important aspects (3,16–18): i) They involve the addition of a monosaccharide, unlike other types where complex sugar chains are involved; ii) they primarily occur in the cytoplasm and nucleus, rather than via the Golgi or endoplasmic reticulum pathways; iii) the modification process is dynamic, reversible and regulated by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA); iv) they interact with other PTMs, such as phosphorylation, methylation and ubiquitination, to regulate protein activity and function; and v) they play a role in the regulation of key cellular biological functions. Due to these characteristics, O-GlcNAcylation makes a multifaceted contribution to cell biology and disease development.
O-GlcNAcylation and phosphorylation are two key protein PTMs that regulate a variety of physiological and pathological processes through dynamic competition or synergy. O-GlcNAcylation involves the addition of a single GlcNAc group to Ser or Thr residues, whereas phosphorylation involves the addition of phosphate groups, often at the same or adjacent sites, resulting in a competitive or cooperative regulatory relationship between the two mechanisms (19). For example, in metabolic regulation, O-GlcNAcylation inhibits the Ser9 phosphorylation of glycogen synthase kinase-3β, thereby reducing the hyperphosphorylation of Tau protein, which may protect against neurodegenerative diseases such as Alzheimer's disease (20,21). Similarly, the Ser40 phosphorylation of tyrosine hydroxylase (TH) promotes dopamine synthesis, whereas O-GlcNAcylation reduces TH activity by inhibiting phosphorylation at this site, thereby affecting L-DOPA levels (22). In addition, O-GlcNAcylation can increase the activity of glycogen phosphorylase, suggesting that synergistic effects may exist between these two PTMs in certain contexts (23).
In the pathological process of sepsis, the balance between O-GlcNAcylation and phosphorylation markedly influences the regulation of inflammatory responses and cell death. Recent studies have shown that O-GlcNAcylation can inhibit phosphorylation-driven pro-inflammatory signaling by targeting key inflammation-related proteins. For example, the O-GlcNAcylation of gasdermin D (GSDMD) inhibits its phosphorylation-dependent activation, thereby attenuating lipopolysaccharide (LPS)-induced pyroptosis, a mechanism that may protect against vascular endothelial injury in sepsis (24). Similarly, homeodomain-interacting protein kinase 2 (HIPK2) inhibits hyperinflammation by promoting the acetylation of NF-κB via the phosphorylation of histone deacetylase 3 (HDAC3), while O-GlcNAcylation may affect this pathway indirectly by regulating the activity or stability of HIPK2 (25). The NF-κB acetylation promoted by the HIPK2-mediated phosphorylation of HDAC3 has been shown to ameliorate colitis-associated colorectal cancer and sepsis in mice. In another study, Toll-like receptor 4 (TLR4) was shown to activate ERK1/2 phosphorylation, leading to the downregulation of Kruppel-like factor 4, thereby exacerbating the inflammatory response in sepsis (26).
In sepsis, phosphorylation is often associated with the amplification of pro-inflammatory and cell death signals. For example, peptidylprolyl cis/trans isomerase, NIMA-interacting 1 exacerbates the inflammatory response in septic shock by promoting the phosphorylation of p38 MAPK, which increases the activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome. O-GlcNAcylation can counteract this effect by competitively modifying the same or adjacent sites of p38 MAPK, thereby preventing its phosphorylation and blocking downstream inflammatory signaling (27,28). Notably, O-GlcNAcylation and phosphorylation may act synergistically in certain contexts. For example, O-GlcNAc modification has been shown to increase glycogenolysis by promoting the phosphorylation of glycogen phosphorylase L during energy stress (23). In summary, the dynamic interplay between O-GlcNAcylation and phosphorylation in sepsis reflects a precisely regulated balance that controls inflammatory intensity, cell death patterns and metabolic adaptation. Targeting this interaction may offer novel strategies for the treatment of sepsis.
There is a complex, cross-regulatory relationship between O-GlcNAcylation and ubiquitination, both of which are involved in cellular stress responses, metabolic regulation and disease progression. O-GlcNAc modification affects the stability, activity and interactions of proteins by the dynamic addition or removal of GlcNAc groups, thereby influencing the ubiquitination process (29). For example, O-GlcNAcylation can increase the stability of key proteins by inhibiting their ubiquitination; the O-GlcNAcylation of the circadian regulators brain and muscle ARNT-like 1 and circadian locomotor output cycles kaput prevents their ubiquitin-mediated degradation, thereby maintaining the normal functioning of the biological clock (30). In addition, the synergistic or antagonistic effects of O-GlcNAcylation and ubiquitination play critical roles in processes such as DNA damage repair and cancer metabolism (31,32). The activity of certain E3 ubiquitin ligases, such as members of the cullin-RING ligase (CRL) family, can be regulated by O-GlcNAc modifications, thereby affecting the ubiquitination levels of their substrate proteins (33). The ubiquitin-proteosome system itself is highly complex, with various ubiquitin chain types, including lysine (Lys)48- and Lys63-linked chains, and dynamic modification patterns. These features form a multi-level interaction network with O-GlcNAcylation to coordinate cellular responses to stresses such as oxidative stress and nutrient deprivation (29,34).
The cross-regulation between O-GlcNAcylation and ubiquitination plays a key role in modulating the intensity of the inflammatory response and maintaining cellular homeostasis. Studies have shown that O-GlcNAc modification can dynamically modulate pro-inflammatory signaling pathways by targeting key components of the ubiquitination system. For example, in macrophages, O-GlcNAc-modified NOD-like receptor X1 (NLRX1) downregulates IL-1β expression by interacting with inhibitor of κB kinase α (IKK-α) and inhibiting its ubiquitination-dependent activation, thereby potentially limiting tissue damage due to excessive inflammatory responses in sepsis (35). In addition, the E3 ubiquitin ligase tripartite motif containing 27 (TRIM27) exacerbates sepsis-related oxidative stress and endothelial dysfunction by promoting the ubiquitin-mediated degradation of peroxisome proliferator-activated receptor γ (PPARγ), and O-GlcNAc modification may counteract the pro-inflammatory effects of TRIM27 by stabilizing PPARγ protein levels (30,36). Notably, the diversity of ubiquitination systems, including different chain linkages such as Lys48- and Lys63-linked chain types, contributes to the dual regulatory features observed in sepsis: Lys48-linked ubiquitination typically targets proteins for proteosomal degradation, helping to clear damaged proteins, whereas Lys63-linked ubiquitination amplifies inflammation by activating pathways such as the NF-κB pathway (29,37). O-GlcNAc modification may balance the inflammatory phenotype of macrophages by selectively regulating the activity of specific E3 ligases, such as E3 ubiquitin-protein ligase COP1, and affecting the ubiquitination status of transcription factors such as CCAAT/enhancer binding protein β (38).
Ubiquitination often serves as a central node for pro-inflammatory and cell death signaling in sepsis, while O-GlcNAc modification may exert a protective effect through multi-level regulatory mechanisms. For example, microbial infection-induced endothelial cell dysfunction is associated with the aberrant ubiquitination of a variety of proteins, such as NADPH oxidase 4 (NOX4), whose stability is regulated by ubiquitination, and its overexpression exacerbates oxidative stress (36,37). O-GlcNAc modification may reduce the production of reactive oxygen species (ROS) by directly modifying NOX4 or regulating the activity of its E3 ligases, such as TRIM27, analogous to its modulatory role in the CRL family ubiquitin ligases involved in DNA damage repair (33). Small-molecule drugs targeting the ubiquitination system, such as deubiquitinase inhibitors, have been shown to modulate the inflammatory cascade in sepsis (39). Similarly, the development of OGT inhibitors or activators may provide new therapeutic strategies by modulating the O-GlcNAc-ubiquitination axis. Notably, there may be synergistic effects between these pathways: For example, O-GlcNAcylation may enhance the stability of NLRX1, promoting its binding to IKK-α and thereby indirectly inhibiting the ubiquitin-dependent activation of the IKK complex; this regulatory homeostasis may exhibit distinct regulatory characteristics at different stages of sepsis (35). In conclusion, the interplay between O-GlcNAcylation and ubiquitination constitutes a complex regulatory network in sepsis. Further elucidation of the underlying mechanism may lay a foundation for the development of targeted interventions to prevent inflammatory imbalance and organ injury.
There is extensive cross-regulation between O-GlcNAc modification and both DNA and histone methylation, which together contribute to the maintenance of epigenetic homeostasis and the regulation of gene expression. Studies have shown that OGT directly modifies DNA methyltransferase 1 through glucose-dependent O-GlcNAcylation, thereby inhibiting its activity, reducing the overall level of genomic methylation and affecting transposon silencing (40,41). At the histone level, OGT is recruited to specific chromatin regions by disruptor of telomeric silencing 1-like, a histone methyltransferase, where it catalyzes the O-GlcNAcylation of histone H2B, which works synergistically with histone H3 Lys79 (H3K79) methylation to regulate gene transcriptional activation (31). In addition, metabolic signals, such as glucose availability, are integrated into the epigenetic regulatory network via O-GlcNAc modification. For example, the c-Myc oncoprotein modulates mitochondrial metabolism and O-GlcNAc cycling, influencing the activity and subcellular localization of DNA and RNA demethylases, thereby linking cellular metabolic state to epigenetic reprogramming (42).
The crosstalk between O-GlcNAcylation and methylation regulates epigenetic reprogramming and metabolic adaptation, which affects the inflammatory response and organ injury. Dysregulation of this epigenetic modification is particularly prominent in monocytes from patients with sepsis and is strongly associated with the aberrant expression of pro-inflammatory cytokines such as TNF-α and IL-6, as well as organ dysfunction (43). In addition, the synergistic interaction of histone methylation with O-GlcNAcylation has a dual role in sepsis: During the endotoxin tolerance stage, G9a-mediated H3K9 methylation and DNA methylation contribute to silencing of the TNF-α gene (44), while O-GlcNAc modification dynamically regulates the expression of pro- and anti-inflammatory genes by regulating DOT1-like histone Lys methyltransferase-dependent H3K79 methylation (31). This multilevel epigenetic-metabolic-ubiquitination regulatory network provides a new paradigm for targeting immunometabolic imbalances in sepsis.
In conclusion, O-GlcNAcylation forms a multi-dimensional regulatory network with other PTMs, including phosphorylation, ubiquitination and methylation, that supports a shift in sepsis treatment from a single anti-inflammatory mode to a systematic regulation based on the dynamic integration of the PTM network. This integrated perspective could provide enable the reversal of immunometabolic imbalances in sepsis.
The PTM of proteins plays a key role in autophagy, either by directly regulating the expression of autophagy-related genes or by modulating key components of the autophagy signaling pathway (45–47). Among these PTMS, O-GlcNAcylation has been shown to be closely associated with the regulation of autophagy; it regulates autophagy by modifying key autophagy-related proteins, thereby affecting their activity, stability and subcellular localization (18). Several proteins with different roles in the autophagy pathway have been identified as potential targets of O-GlcNAcylation (Fig. 2 and Table I) (28,48–60).
Table I.Summary of O-GlcNAc modification sites and their effects on protein function in autophagy regulation. |
The activation of unc-51 like autophagy activating kinase 1 (ULK1) by stressors such as nutrient deprivation (for example, amino acid starvation) and energy stress (for example, ATP depletion/AMP elevation) triggers the autophagic process. ULK1 and ULK2 form protein complexes with various mitosome-associated proteins, including 200 kDa FAK family kinase-interacting protein, autophagy related (ATG)13 and ATG101, which jointly regulate the initiation phase of autophagy (61). These ULK protein complexes play a key regulatory role in the formation of autophagic vesicles and are responsive to signals from multiple signaling pathways. The activity of the ULK1 complex is regulated by both mTOR complex 1 and AMP-activated protein kinase (AMPK) (62,63). Specifically, activation of mTOR via Akt and MAPK signaling inhibits autophagy, while the inhibition of mTOR by AMPK or p53 signaling promotes autophagy. The ULK complex promotes the localized synthesis of phosphatidylinositol-3-phosphate by phosphorylating components of the class III phosphatidylinositol 3-kinase complex, which includes beclin1, vacuolar protein sorting (VPS)15, VPS34 and ATG14 (64). Beclin1 has been identified as a direct target of O-GlcNAcylation, and the O-GlcNAcylation and phosphorylation of beclin1 at the Ser15 site has been shown to regulate autophagy in HTR-8/SVneo cells (48). Bcl-2 inhibits autophagy by interacting with beclin1, and Bcl-2 is also a target of O-GlcNAcylation (49). The O-GlcNAcylation of beclin1 enhances its binding to Bcl-2, thereby suppressing autophagic activity, whereas the O-GlcNAcylation of Bcl-2 regulates autophagy and apoptosis by influencing its expression levels (50,65).
The elongation of phagophores is promoted by two ubiquitin-like conjugation systems: The ATG12-ATG5 system and the microtubule-associated protein 1A/1B-light chain 3 (LC3) system (66). Wang and Hanover (67) demonstrated that the deletion of OGT in Drosophila resulted in a reduction in O-GlcNAc levels and a significant increase in the accumulation of GFP-LGG-1 (an ATG8/LC3 homolog) and its phosphatidylethanolamine-modified form under starvation conditions, indicating enhanced autophagosome formation. Furthermore, OGT knockdown upregulated the mRNA expression of ATG1 and ATG5, whereas OGT overexpression inhibited the transcription of ATG5 and ATG8 (68). These findings suggest that O-GlcNAcylation limits autophagic activity by suppressing the expression of ATG genes. This mechanism may operate synergistically with the stabilization of the beclin1/Bcl-2 complex by O-GlcNAcylation in mammals to maintain the basal autophagy in a low-activity state under normal conditions.
During autophagic fusion, intact autophagosomes fuse with lysosomes to form autolysosomes, which enables the degradation of substances via the action of lysosomal enzymes. O-GlcNAcylation has been shown to regulate the autophagosome-lysosome fusion phase of autophagy in an arsenic-exposed environment by targeting synaptosome-associated protein 29 (SNAP29) (29). In SNAP29-mediated vesicle fusion, autophagosome membranes containing syntaxin 17 (STX17) are linked to lysosomes containing vesicle-associated membrane protein 8 (VAMP8) (69). The O-GlcNAcylation of SNAP29 blocks the assembly of the SNARE complex from STX17, SNAP29 and VAMP8, thereby impairing normal autophagy (70).
O-GlcNAc modification affects cellular metabolism, autotransduction, translation and signal transduction as a nutrient and stress sensor. The maintenance of proper O-GlcNAcylation is crucial for health, as imbalances in O-GlcNAcylation can result in pathological conditions such as tumors, diabetes and neurodegenerative diseases (71–73). A number of studies have focused on the role of O-GlcNAcylation in the regulation of the immune response and maintenance of homeostasis during infection. The present review offers insights into the role of O-GlcNAcylation in the modulation of immune responses against viral, bacterial, fungal and parasitic infections, and explores its potential as a therapeutic target in sepsis (Fig. 3).
Neutrophils are key cells of the innate immune system that rapidly migrate to sites of bacterial infection and remove pathogens via phagocytosis, the release of ROS and necrosis. Neutrophils are the initial responders to injury and infection, and the prognosis of sepsis is poor when neutrophil migration is impaired (74). In both animal models and patients with advanced sepsis, neutrophil function is notably compromised, as evidenced by diminished bacterial clearance, reduced reactivity, decreased ROS production and a significant reduction in neutrophil numbers within infected tissues (75).
O-GlcNAcylation has been found to significantly influence neutrophil chemotaxis and cell migration. For example, treatment with the chemoattractant N-formylmethionyl-leucyl-phenylalanine significantly elevates O-GlcNAcylation levels, thereby promoting neutrophil chemotaxis and migration (76). In addition, glucosamine (GlcN) promotes O-GlcNAcylation by increasing the availability of UDP-GlcNAc, which is the donor substrate for OGT; GlcN enters the hexosamine biosynthetic pathway (HBP) downstream of the rate-limiting enzyme glutamine-fructose-6-phosphate transaminase 1, effectively bypassing it and boosting UDP-GlcNAc production (77). Mechanistically, enhanced O-GlcNAcylation promotes Rac- and phosphoinositide 3-kinase-dependent chemotaxis. Rac is an key small GTPase that regulates neutrophil mobilization by activating downstream MAPK signaling and promoting the phosphorylation of lymphocyte specific protein 1 at Ser243 (78–80). These signaling pathways are critical for neutrophil migration and effector functions. O-GlcNAcylation is inducible in neutrophils and modulates their dynamic responses, potentially providing a novel therapeutic target for the treatment of sepsis.
Macrophages play a key role in resistance to bacterial infections by eliminating pathogens through various mechanisms, including the activation of inducible nitric oxide synthase (iNOS) to produce nitric oxide (NO). NO is a gaseous signaling molecule with antibacterial properties, capable of inhibiting bacterial metabolic enzymes, thereby impairing bacterial viability. In response to bacterial infection or inflammatory stimuli, macrophages increase iNOS expression, thereby increasing NO production (81). NF-κB is a key regulator of the inflammatory response in macrophages and can be modified via O-GlcNAcylation. In RAW264.7 macrophage-like cells, the interaction of OGT with murine Sin3A disrupts NF-κB activation and downregulates iNOS gene expression upon LPS stimulation (82). In microglia, O-GlcNAcylation of the NF-κB subunit cellular-Rel (c-Rel) promotes the formation of the c-Rel-p50 heterodimer and increases iNOS production (83). Notably, the response of the NF-κB subunit p65 to O-GlcNAcylation differs markedly from that of c-Rel. Thiamet-G, a potent and selective inhibitor of OGA, exerts anti-inflammatory effects by promoting the O-GlcNAcylation of p65 in microglia, which subsequently modulates its nuclear localization and reduces the expression of inflammatory genes (84).
Inflammatory diseases such as sepsis are characterized by elevated glucose metabolism in immune cells. Despite macrophages exhibiting increased glycolytic activity and pentose phosphate pathway activation during inflammation, endotoxin exposure inhibits HBP activity, resulting in decreased protein O-GlcNAcylation. Loss of OGT promotes the innate immune response and exacerbates inflammation in sepsis. Specifically, OGT-mediated O-GlcNAcylation of receptor-interacting protein kinase (RIPK)3 at Thr467 disrupts its interaction with RIPK1 and RIPK3 proteins, thereby inhibiting downstream innate immune responses, necroptosis and antibacterial activity (85).
CD8+ T cells are essential for cell-mediated immunity. Upon acute infection, naive CD8+ T cells differentiate into effector cells following antigen stimulation. These effector cells eliminate bacteria by releasing granules containing perforins and granzymes, and secreting cytokines such as interferon (IFN)-γ and TNF-α (86,87). This immune response involves the phosphorylation of various intracellular signaling proteins. Protein O-GlcNAcylation often acts synergistically with phosphorylation (16). Lopez Aguilar et al (88) demonstrated that O-GlcNAcylation was dynamically elevated in murine CD8+ T cells during Listeria infection and in vitro differentiation, with activated effector cells exhibiting increased global levels in vivo, and memory-like cells exhibiting even higher levels (including distinct high-molecular weight modifications) in vitro. Proteomics revealed that O-GlcNAc in effector-like cells primarily targets transcription/translation machinery (such as ribosomal proteins) to enable rapid proliferation, while in memory-like cells it modifies transcriptional regulators, mRNA processing factors and tRNA synthetases, suggesting roles in establishing a poised state. Notably, O-GlcNAc contributes to the ‘histone code’ in both subsets, implicating it in epigenetic regulation of T cell function.
O-GlcNAcylation plays a key regulatory role in the immune response triggered by bacterial infection by dynamically modifying inflammation-related proteins. Pathogen-associated molecular patterns, such as bacterial LPS, can increase O-GlcNAcylation levels in endothelial cells, thereby exacerbating inflammation via the activation of inflammatory signaling pathways (89). At the molecular level, the OGT-mediated glycosylation of NLRX1 facilitates its interaction with IKK-α and promotes the expression of IL-1β in M1 macrophages (35). In parallel, O-GlcNAcylation of the NLRP3 inflammasome not only directly regulates pyroptosis but also promotes the release of inflammatory cytokines by enhancing the activity of the NIMA related kinase 7/NLRP3 axis (90,91). Furthermore, this PTM can create a positive feedback loop, whereby the O-GlcNAcylation of STAT3 further amplifies pro-inflammatory signaling by enhancing JAK2/STAT3 pathway signaling activity (92,93). As an upstream regulator, the phase separation process of TNF receptor-associated factor 6 may also contribute to this regulatory network. Notably, inhibition of O-GlcNAcylation has been shown to effectively block STAT3- and forkhead box protein O1-mediated oxidative stress-induced apoptosis (93–95), suggesting that targeting this modification may be a promising strategy for the alleviation of hyperinflammation during bacterial infections.
These findings suggest that targeting O-GlcNAc modifications may represent a novel therapeutic strategy for the treatment of sepsis by optimizing immune cell function and reducing the tissue damage caused by excessive inflammation.
O-GlcNAcylation has been implicated in the differentiation of T helper (Th)17 cells during fungal infections. Naive CD4+ T cells can differentiate into Th1, Th2, Th17 and Treg cells (96). The downregulation of OGT through microRNA-15b has been shown to inhibit Th17 cell differentiation, thereby contributing to the pathogenesis of multiple sclerosis (97). In addition, elevated O-GlcNAc levels have been shown to promote the secretion of IL-17A by Th17 cells and upregulate the production of lipid ligands for the transcription factor RAR-related orphan receptor γt in diet-induced obese mice (98). Furthermore, in Treg cells, O-GlcNAc-mediated post-translational modifications of the T-cell receptor (TCR) have been demonstrated to increase the stability of forkhead box P3 and activate STAT5, effects that act synergistically to control Treg cell homeostasis and function (96).
Notably, fungal O-GlcNAc metabolism itself directly affects pathogenicity. As a key signaling molecule, GlcNAc regulates morphological transitions, the expression of virulence factors and environmental adaptation in a variety of fungi such as Fusarium. For example, deletion of the OGT homologous gene FpOGT in Fusarium proliferatum inhibits the expression of glucose metabolism-related genes, such as glucokinase, resulting in restricted fungal growth, a downregulated stress response and markedly reduced virulence (99). In addition, GlcNAc has been demonstrated to be a key metabolic target for fungal pathogenesis since it regulates cell wall synthesis, biofilm formation and host tissue invasion pathways (100,101). From a therapeutic perspective, interventions targeting GlcNAc sensing and metabolism, such as GlcNAc analogs or metabolic engineering, have been shown to effectively reduce the virulence of pathogenic fungi (102).
Therefore, in sepsis complicated by fungal infection, the combined modulation of host O-GlcNAc levels to enhance Th17-mediated antifungal immunity while inhibiting fungal GlcNAc-dependent virulence pathways may offer a synergistic therapeutic strategy. Future studies are necessary to further elucidate the molecular mechanisms of the host-fungus O-GlcNAc interaction network and to develop dual regulatory strategies to optimize immunometabolic balance.
Macrophages play a crucial role in the innate immune system, particularly for the early detection of and response to viral infections. Research indicates that O-GlcNAcylation in macrophages is essential for the recognition of RNA viruses. These viruses are detected by cytosolic retinoic acid-inducible gene I-like receptors (RLRs), particularly retinoic acid-inducible gene I and melanoma differentiation-associated gene 5. Upon viral recognition, these receptors engage mitochondrial antiviral signaling protein (MAVS), which activates IFN regulatory factor (IRF)3 and promotes the production of type I IFN (103). In mouse bone marrow-derived macrophages, the absence of OGT disrupts MAVS-dependent RLR signaling and reduces inflammatory cytokine expression (104). Research has further shown that O-GlcNAcylation is crucial for modulating immune responses and increasing infection resistance in macrophages. During influenza A virus infection, O-GlcNAcylation regulates the inflammatory response by modulating the activity of specific transcription factors.
Specifically, O-GlcNAcylation at the Ser430 residue of IRF5 is necessary for its Lys63-linked ubiquitination, which promotes IRF5 activation by facilitating nuclear translocation and interaction with the E3 ubiquitin ligase TRAF6. This modification drives downstream proinflammatory cytokine production during influenza infection (105). Thus, both IRF3 and IRF5 undergo O-GlcNAcylation and ubiquitination, which are crucial for the regulation of macrophage-mediated inflammatory responses following influenza infection. Balancing these modifications is essential to ensure the appropriate intensity of the immune response while preventing excessive inflammation.
Natural killer (NK) cells are crucial components of the innate immune system, responsible for targeting and eliminating virus-infected cells. O-GlcNAcylation has been shown to modulate the function of NK cells as a PTM; specifically, the elevated O-GlcNAcylation of NK cells attenuates their cytotoxicity. Notably, certain molecules, such as the glutathione S-transferase-soluble HLA-G1α chain fusion protein, inhibit O-GlcNAcylation in NK cells, thereby enhancing their cytotoxic function (106).
NK group 2D (NKG2D) is an activating receptor on NK cells that facilitates the recognition and elimination of virus-infected cells presenting stress-induced ligands associated with major histocompatibility complex class I molecules (107). The cytotoxic function of NKG2D-expressing NK cells is regulated by the transcription factor enhancer of zeste 2 (EZH2), a core subunit of polycomb repressive complex 2. Inhibition of EZH2 has been shown to accelerate the maturation of NK cells and improve their killing ability. Notably, EZH2 contains five O-GlcNAcylation sites, which are pivotal for its stabilization and function (108–110).
Another transcription factor that induces cytotoxicity by regulating the O-GlcNAcylation of NK cells is c-Myc. It upregulates the expression of granzyme B, thereby increasing the cytotoxic activity of NK cells against virus-infected cells. O-GlcNAcylation helps to maintain the stability and function of c-Myc. The substrate for this modification, UDP-GlcNAc, is generated through glycolysis and the glutamine metabolic pathway. Glutamine depletion reduces UDP-GlcNAc levels in NK cells, subsequently impacting c-Myc function and diminishing cytotoxic activity (111). Thus, O-GlcNAc modification potentially boosts NK cell immune responses, aiding in the elimination of infected cells and protecting against the progression of sepsis.
B cells are activated via the B-cell receptor (BCR) on their surface, a process that is regulated by O-GlcNAcylation. This PTM affects the YN proto-oncogene, an Src family tyrosine kinase, and its interaction with spleen tyrosine kinase to initiate BCR signaling (112). In addition, the O-GlcNAcylation of nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1) and NF-κB further amplifies B cell activation (113). Following BCR crosslinking, the interaction between O-GlcNAc-modified lymphocyte-specific protein 1 (Lsp1) and protein kinase C-β1 is crucial for the regulation of apoptosis in activated B cells. The O-GlcNAcylation of Lsp1 at Ser209 facilitates its phosphorylation at Ser243, thereby modulating Lsp1 function (114). Also, the dysregulation of EZH2 activity or aberrant O-GlcNAcylation may impair antibody production in B cells, indicating the importance of O-GlcNAcylation in the modulation of B cell function and antibody responses (115).
CD8+ cytotoxic T cells identify and destroy infected cells during viral infections. NF-κB and c-Myc are essential transcription factors in the activation of CD8+ T cells, and TCR-mediated stimulation induces their O-GlcNAcylation. The O-GlcNAcylation of c-Rel enhances its binding to the CD28 response element, resulting in the production of proinflammatory cytokines, including IL-2, IFN-γ and granulocyte macrophage-colony-stimulating factor. In addition, site-specific O-GlcNAcylation stabilizes c-Myc, while the inhibition of OGT decreases c-Myc levels (116,117). Furthermore, NFATc1 is mainly O-GlcNAcylated in the cytosol, and translocates to the nucleus after TCR activation. The O-GlcNAc modification of NFAT has been demonstrated to be crucial for its nuclear translocation and function, as the inhibition of OGT decreases TCR-induced IL-2 production and CD69 expression (113). These findings highlight the importance of O-GlcNAcylation in T cells. The inhibition of OGT has also been shown to affect the transcriptional activity of NF-κB in T cells, with the p65 subunit of NF-κB undergoing O-GlcNAcylation (113,118). In addition, the O-GlcNAcylation of DNA fragmentation factor 45 shields proteins from caspase-mediated cleavage during DNA damage-induced T-cell apoptosis, indicating its protective role against apoptotic cell death in T cells (119).
Modulating O-GlcNAc glycosylation levels or its downstream signaling pathways may help to address immune imbalances in patients with sepsis, suppress excessive inflammation, prevent organ dysfunction and ultimately improve clinical outcomes.
The dual role of O-GlcNAcylation in host anti-parasitic immunity and parasite self-regulation has become increasingly evident. This involves an interaction between the regulation of host immune cell function and the metabolic dependence of the parasite on O-GlcNAc pathways. In the anti-parasitic immunity of the host, O-GlcNAc contributes to key pathways that regulate type 2 immune responses. For example, the O-GlcNAcylation of STAT6 in intestinal epithelial cells forms the first line of defense against helminth infection by driving tuft cell proliferation and the release of gasdermin C-dependent alarmins (120). Restoration of STAT6 activation partially rescues the impaired immune responses to helminth infection in OGT-deficient mice, which exhibit defective STAT6 signaling and intestinal villus dysplasia (121). In addition, mast cell function is closely associated with O-GlcNAc modification: In Trichomonas vaginalis infection, leukotriene B4 (LTB4) secreted by the parasite binds to LTB4 receptor 1 on mast cells, elevates O-GlcNAc levels and promotes NADPH oxidase-dependent migration, degranulation and IL-8 secretion (122). However, Tet methylcytosine dioxygenase 2 (TET2)-mediated O-GlcNAcylation affects mast cell differentiation and cytokine production by regulating chromatin accessibility (123). These findings suggest that O-GlcNAc contributes to the anti-parasitic response through the multifaceted regulation of host immune cell functions, including Th2 polarization, epithelial barrier defense and mast cell effector functions.
At the biological level, glycosylation modifications, including O-GlcNAc-related pathways, are essential for the survival, development and pathogenicity of parasites. For example, Plasmodium falciparum relies on the HBP to produce glycosylphosphatidylinositol-anchored proteins, which are critical for the invasion of red blood cells and immune evasion. Therefore, blocking this pathway can lead to the growth arrest of Plasmodium at the schizont stage (124). Although Cryptosporidium lacks certain glycosylases, it compensates by hijacking host glycosylation resources to perform its protein modifications (125). In addition, glycosylation in various parasites, such as Plasmodium and Toxoplasma gondii, affects host immune recognition by altering the function of surface proteins (126,127). Notably, there is an interaction between host and parasite O-GlcNAc metabolism: Parasites may disrupt the glycosylation homeostasis of host immune cells by secreting metabolites such as GlcNAc, while host-targeted drugs against key enzymes involved in parasite glycosylation, such as glutamine:fructose-6-phosphate transaminase (GFAT) in the HBP, can simultaneously inhibit parasite growth and enhance immune clearance (124,127).
In summary, O-GlcNAcylation affects infection outcomes through host-parasite bidirectional regulation: It regulates host type 2 immune responses and effector cell functions through targets such as STAT6 and TET2, while parasites rely on glycosylation pathways to complete their developmental cycles and evade host immunity. Future studies should aim to further elucidate the parasite-specific O-GlcNAc modification network and explore dual intervention strategies, such as the development of GFAT inhibitors combined with immunomodulators, to synergistically enhance anti-parasitic immunity and block pathogen metabolism-dependent virulence mechanisms.
O-GlcNAcylation plays diverse roles in different types of infection by modulating host defense mechanisms and affecting immune cell function. Specifically, macrophage responses to infection reveal a key functional divergence of O-GlcNAcylation: It promotes antiviral defenses but suppresses antibacterial responses. Given the dual role of O-GlcNAcylation in different types of infection, the modulation of O-GlcNAcylation activity may serve as a novel therapeutic strategy. For example, enhancing O-GlcNAcylation may strengthen the host antiviral response, while in bacterial infections, moderate inhibition of O-GlcNAcylation may help to limit inflammatory damage and promote tissue repair. Therefore, targeting O-GlcNAcylation may represent a potential approach for the treatment of infectious diseases such as sepsis.
O-GlcNAc glycosylation is a crucial PTM that influences protein function by interacting with other PTMs, such as phosphorylation and ubiquitination (16). For example, O-GlcNAcylation has been demonstrated to promote protein phosphorylation, including that of Lsp1 in B cells, where it facilitates interaction with F-actin (114). Also, O-GlcNAcylation can modulate the function of ubiquitinated proteins, including macrophage proteins MAVS and IRF5, which depend on downstream ubiquitination signaling for proper immune activation (103–105). Understanding the interplay between O-GlcNAcylation and other PTMs, such as phosphorylation and ubiquitination, is crucial for elucidating the role of O-GlcNAcylation in the regulation of immune cell function in both homeostatic and infectious states.
Intervention strategies targeting O-GlcNAc homeostasis have demonstrated multidimensional potential in the treatment of sepsis. For example, GlcN significantly inhibits LPS-induced activation of the MAPK and NF-κB inflammatory pathways by increasing the O-GlcNAc modification of nucleoplasmic proteins, thereby attenuating lung tissue damage and improving survival in mouse and zebrafish models (128). In addition, OGT-mediated modification of O-GlcNAc at RIPK3 Thr467 has been shown to block the formation of necroptotic complexes and inhibit the inflammatory storm in sepsis (129). Also, an acute increase in overall O-GlcNAc levels, such as that induced by the OGA inhibitor N-butyryl-glucosamine-1,5-lactone O-(phenylcarbamoyl)oxime, significantly reduces mortality in septic shock models by inhibiting the cleavage and activation of GSDMD, a key pyroptosis protein (130,131). At the metabolic level, O-GlcNAcylation regulates metabolic enzymes such as ATP citrate lyase in rat models of juvenile sepsis and helps to modulate systemic inflammatory responses through non-transcriptional pathways, suggesting its multi-target regulatory advantage (132,133). However, the clinical application of OGT/OGA inhibitors faces complex challenges. Specifically, the inhibition of OGT may have off-target effects that interfere with protective mechanisms in sepsis. For example, inhibition of OGT may inadvertently weaken its inhibitory effect on GSDMD-mediated pyroptosis or impair the glutamine-maintained gut mucus barrier by interfering with mucin-type glycosylation, potentially through the cross-inhibition of enzymes such as UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferase (5,24,134). In addition, the compensatory stimulation of OGA or fluctuations in the UDP-GlcNAc metabolic pool triggered by OGT inhibition may disrupt the N-glycosylation of immune receptors such as TLR4, thereby exacerbating inflammatory dysregulation (134–136). However, the spatiotemporal heterogeneity of O-GlcNAcylation, such as the contrasting roles observed between endothelial cells and macrophages, and the lack of cell-specific regulatory tools make it challenging to accurately target the dynamic pathological processes of sepsis (137,138). Future strategies combining nano-targeted delivery and dynamic biomarker monitoring are necessary for the development of spatiotemporally accurate modification approaches that optimize therapeutic benefits while minimizing risks.
In summary, as a key PTM, O-GlcNAcylation acts as a critical link between the immune response to infection and the progression of sepsis. Future studies should aim to fully elucidate the specific role of O-GlcNAcylation in the pathogenesis of sepsis, including the mechanisms by which it activates immune cells, regulates the inflammatory response and controls the autophagic process. In addition, the identification of specific O-GlcNAcylation markers as potential biomarkers may improve the early diagnosis and prognostic assessment of sepsis, providing deeper mechanistic insights. Intervention strategies targeting O-GlcNAcylation may enable the regulation of autophagy and immune responses, thereby providing new therapeutic options for patients with sepsis. Such approaches may include the development of small-molecule drugs to modulate the activity of OGT and OGA, or the design of interventions targeting specific O-GlcNAcylation sites.
Not applicable.
The present study was supported by a grant from the National Natural Science Foundation of China (grant no. 82260135).
Not applicable.
ZH and XL wrote and revised this manuscript. PL designed the subject of review. LZ, YL and XM reviewed and revised the manuscript. Data authentication is not applicable. All authors read and approved the final version of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Nedeva C: Inflammation and cell death of the innate and adaptive immune system during sepsis. Biomolecules. 11:10112021. View Article : Google Scholar : PubMed/NCBI | |
|
Rudd KE, Johnson SC, Agesa KM, Shackelford KA, Tsoi D, Kievlan DR, Colombara DV, Ikuta KS, Kissoon N, Finfer S, et al: Global, regional, and national sepsis incidence and mortality, 1990–2017: Analysis for the global burden of disease study. Lancet. 395:200–211. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Lockridge A and Hanover JA: A nexus of lipid and O-Glcnac metabolism in physiology and disease. Front Endocrinol (Lausanne). 13:9435762022. View Article : Google Scholar : PubMed/NCBI | |
|
Cai H, Xiong W, Zhu H, Wang Q, Liu S and Lu Z: Protein O-GlcNAcylation in multiple immune cells and its therapeutic potential. Front Immunol. 14:12099702023. View Article : Google Scholar : PubMed/NCBI | |
|
Wu D, Su S, Zha X, Wei Y, Yang G, Huang Q, Yang Y, Xia L, Fan S and Peng X: Glutamine promotes O-GlcNAcylation of G6PD and inhibits AGR2 S-glutathionylation to maintain the intestinal mucus barrier in burned septic mice. Redox Biol. 59:1025812023. View Article : Google Scholar : PubMed/NCBI | |
|
Srzić I, Nesek Adam V and Tunjić Pejak D: Sepsis definition: What's new in the treatment guidelines. Acta Clin Croat. 61 (Suppl 1):S67–S72. 2022.PubMed/NCBI | |
|
Wiersinga WJ and van der Poll T: Immunopathophysiology of human sepsis. EBioMedicine. 86:1043632022. View Article : Google Scholar : PubMed/NCBI | |
|
Torres LK, Pickkers P and van der Poll T: Sepsis-induced immunosuppression. Annu Rev Physiol. 84:157–181. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu D, Huang SY, Sun JH, Zhang HC, Cai QL, Gao C, Li L, Cao J, Xu F, Zhou Y, et al: Sepsis-induced immunosuppression: Mechanisms, diagnosis and current treatment options. Mil Med Res. 9:562022.PubMed/NCBI | |
|
Wen X, Xie B, Yuan S and Zhang J: The ‘Self-sacrifice’ of ImmuneCells in sepsis. Front Immunol. 13:8334792022. View Article : Google Scholar : PubMed/NCBI | |
|
Yang L, Zhou L, Li F, Chen X, Li T, Zou Z, Zhi Y and He Z: Diagnostic and prognostic value of Autophagy-related key genes in sepsis and potential correlation with immune cell signatures. Front Cell Dev Biol. 11:12183792023. View Article : Google Scholar : PubMed/NCBI | |
|
Reglero-Real N, Pérez-Gutiérrez L, Yoshimura A, Rolas L, Garrido-Mesa J, Barkaway A, Pickworth C, Saleeb RS, Gonzalez-Nuñez M, Austin-Williams SN, et al: Autophagy modulates endothelial junctions to restrain neutrophil diapedesis during inflammation. Immunity. 54:1989–2004.e9. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu CL, Wang Y, Liu Q, Li HR, Yu CM, Li P, Deng XM and Wang JF: Dysregulation of neutrophil death in sepsis. Front Immunol. 13:9639552022. View Article : Google Scholar : PubMed/NCBI | |
|
Qin Y, Li W, Liu J, Wang F, Zhou W, Xiao L, Zhou P, Wu F, Chen X, Xu S, et al: Andrographolide ameliorates sepsis-induced acute lung injury by promoting autophagy in alveolar macrophages via the RAGE/PI3K/AKT/mTOR pathway. Int Immunopharmacol. 139:1127192024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Bai G, Chen J, Han W, Guo R and Cui N: mTOR deletion ameliorates CD4 + T cell apoptosis during sepsis by improving autophagosome-lysosome fusion. Apoptosis. 27:401–408. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ye L, Ding W, Xiao D, Jia Y, Zhao Z, Ao X and Wang J: O-GlcNAcylation: Cellular physiology and therapeutic target for human diseases. MedComm (2020). 4:e4562023. View Article : Google Scholar : PubMed/NCBI | |
|
Gonzalez-Rellan MJ, Fondevila MF, Dieguez C and Nogueiras R: O-GlcNAcylation: A sweet hub in the regulation of glucose metabolism in health and disease. Front Endocrinol (Lausanne). 13:8735132022. View Article : Google Scholar : PubMed/NCBI | |
|
Chatham JC, Zhang J and Wende AR: Role of O-Linked N-acetylglucosamine protein modification in cellular (patho)physiology. Physiol Rev. 101:427–493. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Q, Zhou S, Lou W, Qian H and Xu Z: Crosstalk between O-GlcNAcylation and phosphorylation in metabolism: Regulation and mechanism. Cell Death Differ. 32:1181–1199. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
El Hajjar L, Page A, Bridot C, Cantrelle FX, Landrieu I and Smet-Nocca C: Regulation of Glycogen Synthase Kinase-3β by Phosphorylation and O-β-Linked N-Acetylglucosaminylation: Implications on tau protein phosphorylation. Biochemistry. 63:1513–1533. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Cardozo CF, Vera A, Quintana-Peña V, Arango-Davila CA and Rengifo J: Regulation of Tau protein phosphorylation by glucosamine-induced O-GlcNAcylation as a neuroprotective mechanism in a brain ischemia-reperfusion model. Int J Neurosci. 133:194–200. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
da Costa Rodrigues B, Dos Santos Lucena MC, Costa A, de Araújo Oliveira I, Thaumaturgo M, Paes-Colli Y, Beckman D, Ferreira ST, de Mello FG, de Melo Reis RA, et al: O-GlcNAcylation regulates tyrosine hydroxylase serine 40 phosphorylation and l-DOPA levels. Am J Physiol Cell Physiol. 328:C825–C835. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Chen YF, Zhu JJ, Li J and Ye XS: O-GlcNAcylation increases PYGL activity by promoting phosphorylation. Glycobiology. 32:101–109. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yu F, Zhang Z, Leng Y and Chen AF: O-GlcNAc modification of GSDMD attenuates LPS-induced endothelial cells pyroptosis. Inflamm Res. 73:5–17. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Correction for Zhang et al. HIPK2 phosphorylates HDAC3 for NF-κB acetylation to ameliorate colitis-associated colorectal carcinoma and sepsis. Proc Natl Acad Sci USA. 122:e25060471222025. View Article : Google Scholar : PubMed/NCBI | |
|
Li C, Yu L, Mai C, Mu T and Zeng Y: KLF4 down-regulation resulting from TLR4 promotion of ERK1/2 phosphorylation underpins inflammatory response in sepsis. J Cell Mol Med. 25:2013–2024. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Dong R, Xue Z, Fan G, Zhang N, Wang C, Li G and Da Y: Pin1 Promotes NLRP3 inflammasome activation by phosphorylation of p38 MAPK Pathway in Septic Shock. Front Immunol. 12:6202382021. View Article : Google Scholar : PubMed/NCBI | |
|
Jin L, Yuan F, Dai G, Yao Q, Xiang H, Wang L, Xue B, Shan Y and Liu X: Blockage of O-linked GlcNAcylation induces AMPK-dependent autophagy in bladder cancer cells. Cell Mol Biol Lett. 25:172020. View Article : Google Scholar : PubMed/NCBI | |
|
Sheng X, Xia Z, Yang H and Hu R: The ubiquitin codes in cellular stress responses. Protein Cell. 15:157–190. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li MD, Ruan HB, Hughes ME, Lee JS, Singh JP, Jones SP, Nitabach MN and Yang X: O-GlcNAc signaling entrains the circadian clock by inhibiting BMAL1/CLOCK ubiquitination. Cell Metab. 17:303–310. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Xu B, Zhang C, Jiang A, Zhang X, Liang F, Wang X, Li D, Liu C, Liu X, Xia J, et al: Histone methyltransferase Dot1L recruits O-GlcNAc transferase to target chromatin sites to regulate histone O-GlcNAcylation. J Biol Chem. 298:1021152022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Wei D, Li C, Yang X, Su K, Wang T, Zou R, Wang L, Cun D, Tang B, et al: O-GlcNAcylation with ubiquitination stabilizes METTL3 to promoting HMGB1 degradation to inhibit ferroptosis and enhance gemcitabine resistance in pancreatic cancer. Mol Med. 31:2282025. View Article : Google Scholar : PubMed/NCBI | |
|
Koo SY, Park EJ, Noh HJ, Jo SM, Ko BK, Shin HJ and Lee CW: Ubiquitination links DNA damage and repair signaling to cancer metabolism. Int J Mol Sci. 24:84412023. View Article : Google Scholar : PubMed/NCBI | |
|
Dikic I and Schulman BA: An expanded lexicon for the ubiquitin code. Nat Rev Mol Cell Biol. 24:273–287. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Chen L, Li Y, Zeng S, Duan S, Huang Z and Liang Y: The interaction of O-GlcNAc-modified NLRX1 and IKK-α modulates IL-1β expression in M1 macrophages. In Vitro Cell Dev Biol Anim. 58:408–418. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ning M, Liu Y, Wang D, Wei J, Hu G and Xing P: Knockdown of TRIM27 alleviated sepsis-induced inflammation, apoptosis, and oxidative stress via suppressing ubiquitination of PPARγ and reducing NOX4 expression. Inflamm Res. 71:1315–1325. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang J, He Y and Zhou D: The role of ubiquitination in microbial infection induced endothelial dysfunction: Potential therapeutic targets for sepsis. Expert Opin Ther Targets. 27:827–839. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yu Y, Fu Q and Li J, Zen X and Li J: E3 ubiquitin ligase COP1-mediated CEBPB ubiquitination regulates the inflammatory response of macrophages in sepsis-induced myocardial injury. Mamm Genome. 35:56–67. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li ZQ, Chen X and Wang Y: Small molecules targeting ubiquitination to control inflammatory diseases. Drug Discov Today. 26:2414–2422. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
O-GlcNAc transferase is a key regulator of DNA methylation and transposon silencing. Nat Struct Mol Biol. 32:1137–1138. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Shin H, Leung A, Costello KR, Senapati P, Kato H, Moore RE, Lee M, Lin D, Tang X, Pirrotte P, et al: Inhibition of DNMT1 methyltransferase activity via glucose-regulated O-GlcNAcylation alters the epigenome. Elife. 12:e855952023. View Article : Google Scholar : PubMed/NCBI | |
|
Lin AP, Qiu Z, Ethiraj P, Sasi B, Jaafar C, Rakheja D and Aguiar R: MYC, mitochondrial metabolism and O-GlcNAcylation converge to modulate the activity and subcellular localization of DNA and RNA demethylases. Leukemia. 36:1150–1159. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Lorente-Sorolla C, Garcia-Gomez A, Català-Moll F, Toledano V, Ciudad L, Avendaño-Ortiz J, Maroun-Eid C, Martín-Quirós A, Martínez-Gallo M, Ruiz-Sanmartín A, et al: Inflammatory cytokines and organ dysfunction associate with the aberrant DNA methylome of monocytes in sepsis. Genome Med. 11:662019. View Article : Google Scholar : PubMed/NCBI | |
|
El Gazzar M, Yoza BK, Chen X, Hu J, Hawkins GA and McCall CE: G9a and HP1 couple histone and DNA methylation to TNFalpha transcription silencing during endotoxin tolerance. J Biol Chem. 283:32198–32208. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu Z, Ren W, Li S, Gao L and Zhi K: Functional significance of O-linked N-acetylglucosamine protein modification in regulating autophagy. Pharmacol Res. 202:1071202024. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Ren W, Zheng J, Li S, Zhi K and Gao L: Role of O-linked N-acetylglucosamine protein modification in oxidative stress-induced autophagy: A novel target for bone remodeling. Cell Commun Signal. 22:3582024. View Article : Google Scholar : PubMed/NCBI | |
|
Jeon M, Park J, Yang E, Baek HJ and Kim H: Regulation of autophagy by protein methylation and acetylation in cancer. J Cell Physiol. 237:13–28. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Q, Na Q and Song W: Moderate mammalian target of rapamycin inhibition induces autophagy in HTR8/SVneo cells via O-linked β-N-acetylglucosamine signaling. J Obstet Gynaecol Res. 43:1585–1596. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Qiu Z, Cui J, Huang Q, Qi B and Xia Z: Roles of O-GlcNAcylation in mitochondrial homeostasis and cardiovascular diseases. Antioxidants (Basel). 13:5712024. View Article : Google Scholar : PubMed/NCBI | |
|
Marsh SA, Powell PC, Dell'italia LJ and Chatham JC: Cardiac O-GlcNAcylation blunts autophagic signaling in the diabetic heart. Life Sci. 92:648–656. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Rahman MA, Cho Y, Hwang H and Rhim H: Pharmacological Inhibition of O-GlcNAc transferase promotes mTOR-Dependent autophagy in rat cortical neurons. Brain Sci. 10:9582020. View Article : Google Scholar : PubMed/NCBI | |
|
Pyo KE, Kim CR, Lee M, Kim JS, Kim KI and Baek SH: ULK1 O-GlcNAcylation is crucial for activating VPS34 via ATG14L during Autophagy initiation. Cell Rep. 25:2878–2890.e4. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Shi Y, Yan S, Shao GC, Wang J, Jian YP, Liu B, Yuan Y, Qin K, Nai S, Huang X, et al: O-GlcNAcylation stabilizes the autophagy-initiating kinase ULK1 by inhibiting chaperone-mediated autophagy upon HPV infection. J Biol Chem. 298:1023412022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Li C, Shuai W, Chen T, Gong Y, Hu H, Wei Y, Kong B and Huang H: Maresin2 fine-tunes ULK1 O-GlcNAcylation to improve post myocardial infarction remodeling. Eur J Pharmacol. 962:1762232024. View Article : Google Scholar : PubMed/NCBI | |
|
Alghusen IM, Carman MS, Wilkins HM, Strope TA, Gimore C, Fedosyuk H, Shawa J, Ephrame SJ, Denson AR, Wang X, et al: O-GlcNAc impacts mitophagy via the PINK1-dependent pathway. Front Aging Neurosci. 16:13879312024. View Article : Google Scholar : PubMed/NCBI | |
|
Guo B, Liang Q, Li L, Hu Z, Wu F, Zhang P, Ma Y, Zhao B, Kovács AL, Zhang Z, et al: O-GlcNAc-modification of SNAP-29 regulates autophagosome maturation. Nat Cell Biol. 16:1215–1226. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Dodson M, Liu P, Jiang T, Ambrose AJ, Luo G, Rojo de la Vega M, Cholanians AB, Wong PK, Chapman E and Zhang DD: Increased O-GlcNAcylation of SNAP29 drives Arsenic-induced autophagic dysfunction. Mol Cell Biol. 38:e00595–e00517. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Pellegrini FR, De Martino S, Fianco G, Ventura I, Valente D, Fiore M, Trisciuoglio D and Degrassi F: Blockage of autophagosome-lysosome fusion through SNAP29 O-GlcNAcylation promotes apoptosis via ROS production. Autophagy. 19:2078–2093. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Huang L, Yuan P, Yu P, Kong Q, Xu Z, Yan X, Shen Y, Yang J, Wan R, Hong K, et al: O-GlcNAc-modified SNAP29 inhibits autophagy-mediated degradation via the disturbed SNAP29-STX17-VAMP8 complex and exacerbates myocardial injury in type I diabetic rats. Int J Mol Med. 42:3278–3290. 2018.PubMed/NCBI | |
|
Zhou F, Yang X, Zhao H, Liu Y, Feng Y, An R, Lv X, Li J and Chen B: Down-regulation of OGT promotes cisplatin resistance by inducing autophagy in ovarian cancer. Theranostics. 8:5200–5212. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Patel A and Faesen AC: Metamorphosis by ATG13 and ATG101 in human autophagy initiation. Autophagy. 20:968–969. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X and Jia J: Magnolol improves Alzheimer's disease-like pathologies and cognitive decline by promoting autophagy through activation of the AMPK/mTOR/ULK1 pathway. Biomed Pharmacother. 161:1144732023. View Article : Google Scholar : PubMed/NCBI | |
|
Ge Y, Zhou M, Chen C, Wu X and Wang X: Role of AMPK mediated pathways in autophagy and aging. Biochimie. 195:100–113. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Pizzimenti C, Fiorentino V, Ruggeri C, Franchina M, Ercoli A, Tuccari G and Ieni A: Autophagy involvement in Non-neoplastic and neoplastic endometrial pathology: The state of the art with a focus on carcinoma. Int J Mol Sci. 25:121182024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang TF, Feng ZQ, Sun YW, Zhao SJ, Zou HY, Hao HS, Du WH, Zhao XM, Zhu HB and Pang YW: Disruption of O-GlcNAcylation homeostasis induced ovarian granulosa cell injury in bovine. Int J Mol Sci. 23:78152022. View Article : Google Scholar : PubMed/NCBI | |
|
Xu J, Gu J, Pei W, Zhang Y, Wang L and Gao J: The role of lysosomal membrane proteins in autophagy and related diseases. FEBS J. 291:3762–3785. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Wang P and Hanover JA: Nutrient-driven O-GlcNAc cycling influences autophagic flux and neurodegenerative proteotoxicity. Autophagy. 9:604–606. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Park S, Lee Y, Pak JW, Kim H, Choi H, Kim JW, Roth J and Cho JW: O-GlcNAc modification is essential for the regulation of autophagy in Drosophila melanogaster. Cell Mol Life Sci. 72:3173–3183. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Zheng D, Tong M, Zhang S, Pan Y, Zhao Y, Zhong Q and Liu X: Human YKT6 forms priming complex with STX17 and SNAP29 to facilitate autophagosome-lysosome fusion. Cell Rep. 43:1137602024. View Article : Google Scholar : PubMed/NCBI | |
|
Ben Ahmed A, Lemaire Q, Scache J, Mariller C, Lefebvre T and Vercoutter-Edouart AS: O-GlcNAc dynamics: The sweet side of protein trafficking regulation in mammalian cells. Cells. 12:13962023. View Article : Google Scholar : PubMed/NCBI | |
|
Du P, Zhang X, Lian X, Hölscher C and Xue G: O-GlcNAcylation and its roles in neurodegenerative diseases. J Alzheimers Dis. 97:1051–1068. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
He XF, Hu X, Wen GJ, Wang Z and Lin WJ: O-GlcNAcylation in cancer development and immunotherapy. Cancer Lett. 566:2162582023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu C, Dong W, Li J, Kong Y and Ren X: O-GlcNAc modification and its role in diabetic retinopathy. Metabolites. 12:7252022. View Article : Google Scholar : PubMed/NCBI | |
|
Bruserud Ø, Mosevoll KA, Bruserud Ø, Reikvam H and Wendelbo Ø: The regulation of neutrophil migration in patients with sepsis: The complexity of the molecular mechanisms and their modulation in sepsis and the heterogeneity of sepsis patients. Cells. 12:10032023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou YY and Sun BW: Recent advances in neutrophil chemotaxis abnormalities during sepsis. Chin J Traumatol. 25:317–324. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Kneass ZT and Marchase RB: Neutrophils exhibit rapid Agonist-induced increases in protein-associated O-GlcNAc. J Biol Chem. 279:45759–45765. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Cong R, Sun L, Yang J, Cui H, Ji X, Zhu J, Gu JH and He B: Protein O-GlcNAcylation alleviates small intestinal injury induced by ischemia-reperfusion and oxygen-glucose deprivation. Biomed Pharmacother. 138:1114772021. View Article : Google Scholar : PubMed/NCBI | |
|
Hossain M, Qadri SM, Xu N, Su Y, Cayabyab FS, Heit B and Liu L: Endothelial LSP1 modulates extravascular neutrophil chemotaxis by regulating nonhematopoietic vascular PECAM-1 Expression. J Immunol. 195:2408–2416. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Machin PA, Johnsson AE, Massey EJ, Pantarelli C, Chetwynd SA, Chu JY, Okkenhaug H, Segonds-Pichon A, Walker S, Malliri A, et al: Dock2 generates characteristic spatiotemporal patterns of Rac activity to regulate neutrophil polarisation, migration and phagocytosis. Front Immunol. 14:11808862023. View Article : Google Scholar : PubMed/NCBI | |
|
Kneass ZT and Marchase RB: Protein O-GlcNAc modulates motility-associated signaling intermediates in neutrophils. J Biol Chem. 280:14579–14585. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Wu KK, Xu X, Wu M, Li X, Hoque M, Li G, Lian Q, Long K, Zhou T, Piao H, et al: MDM2 induces pro-inflammatory and glycolytic responses in M1 macrophages by integrating iNOS-nitric oxide and HIF-1α pathways in mice. Nat Commun. 15:86242024. View Article : Google Scholar : PubMed/NCBI | |
|
Hwang SY, Hwang JS, Kim SY and Han IO: O-GlcNAc transferase inhibits LPS-mediated expression of inducible nitric oxide synthase through an increased interaction with mSin3A in RAW264.7 cells. Am J Physiol Cell Physiol. 305:C601–C608. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Hwang SY, Hwang JS, Kim SY and Han IO: O-GlcNAcylation and p50/p105 binding of c-Rel are dynamically regulated by LPS and glucosamine in BV2 microglia cells. Br J Pharmacol. 169:1551–1560. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
He Y, Ma X, Li D and Hao J: Thiamet G mediates neuroprotection in experimental stroke by modulating microglia/macrophage polarization and inhibiting NF-κB p65 signaling. J Cereb Blood Flow Metab. 37:2938–2951. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Seo J, Kim Y, Ji S, Kim HB, Jung H, Yi EC, Lee YH, Shin I, Yang WH and Cho JW: O-GlcNAcylation of RIPK1 rescues red blood cells from necroptosis. Front Immunol. 14:11604902023. View Article : Google Scholar : PubMed/NCBI | |
|
Janas ML, Groves P, Kienzle N and Kelso A: IL-2 regulates perforin and granzyme gene expression in CD8+ T cells independently of its effects on survival and proliferation. J Immunol. 175:8003–8010. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Reina-Campos M, Scharping NE and Goldrath AW: CD8+ T cell metabolism in infection and cancer. Nat Rev Immunol. 21:718–738. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lopez Aguilar A, Gao Y, Hou X, Lauvau G, Yates JR and Wu P: Profiling of protein O-GlcNAcylation in murine CD8+ effector- and Memory-like T cells. ACS Chem Biol. 12:3031–3038. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Chen H, Shi Y, Ying J, Dong Z, Wang Y, Zheng Y and Ruan P: O-linked N-acetylglucosamine modification induced by lipopolysaccharide is involved in inflammatory signaling pathway in endothelial cells. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 35:164–169. 2023.(In Chinese). PubMed/NCBI | |
|
Zeng Z, Liao X and Zhao X: O-GlcNAc transferase mediates O-GlcNAcylation of NLRP3 regulates pyroptosis in spinal cord injury. Brain Res Bull. 222:1112332025. View Article : Google Scholar : PubMed/NCBI | |
|
He C, Wu Q, Zeng Z, Yang Y, He H, Hu M and Liu S: OGT-induced O-GlcNAcylation of NEK7 protein aggravates osteoarthritis progression by enhancing NEK7/NLRP3 axis. Autoimmunity. 57:23192022024. View Article : Google Scholar : PubMed/NCBI | |
|
Yaqin Z, Kehan W, Yi Z, Naijian W, Wei Q and Fei M: Resveratrol alleviates inflammatory bowel disease by inhibiting JAK2/STAT3 pathway activity via the reduction of O-GlcNAcylation of STAT3 in intestinal epithelial cells. Toxicol Appl Pharmacol. 484:1168822024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang CC, Li Y, Jiang CY, Le QM, Liu X, Ma L and Wang FF: O-GlcNAcylation mediates H2O2-induced apoptosis through regulation of STAT3 and FOXO1. Acta Pharmacol Sin. 45:714–727. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Peng J, Xia Y, Pan C, Li Y, Gu W, Wang J, Wang C, Wang Y, Song J, et al: Sufu limits sepsis-induced lung inflammation via regulating phase separation of TRAF6. Theranostics. 13:3761–3780. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Matsui S, Ri C, Bolanos LC, Choi K, Shibamiya A, Ishii A, Takaishi K, Oshima-Hasegawa N, Tsukamoto S, Takeda Y, et al: Metabolic reprogramming regulated by TRAF6 contributes to the leukemia progression. Leukemia. 38:1032–1045. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Qiang A, Slawson C and Fields PE: The Role of O-GlcNAcylation in immune cell activation. Front Endocrinol (Lausanne). 12:5966172021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu R, Ma X, Chen L, Yang Y, Zeng Y, Gao J, Jiang W, Zhang F, Li D, Han B, et al: MicroRNA-15b Suppresses Th17 differentiation and is associated with pathogenesis of multiple sclerosis by targeting O-GlcNAc transferase. J Immunol. 198:2626–2639. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Machacek M, Saunders H, Zhang Z, Tan EP, Li J, Li T, Villar MT, Artigues A, Lydic T, Cork G, et al: Elevated O-GlcNAcylation enhances pro-inflammatory Th17 function by altering the intracellular lipid microenvironment. J Biol Chem. 294:8973–8990. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Thomaidis T, Maderer A, Formentini A, Bauer S, Trautmann M, Schwarz M, Neumann W, Kittner JM, Schad A, Link KH, et al: Proteins of the VEGFR and EGFR pathway as predictive markers for adjuvant treatment in patients with stage II/III colorectal cancer: Results of the FOGT-4 trial. J Exp Clin Cancer Res. 33:832014. View Article : Google Scholar : PubMed/NCBI | |
|
Gao YZ, Wang YT, He S, Li H, Wang Y and Wu Z: FpOGT is required for fungal growth, stress response, and virulence of Fusarium proliferatum by affecting the expression of glucokinase and other glucose metabolism-related genes. Phytopathol Res. 6:22024. View Article : Google Scholar | |
|
Naseem S and Konopka JB: N-acetylglucosamine regulates virulence properties in microbial pathogens. PLoS Pathog. 11:e10049472015. View Article : Google Scholar : PubMed/NCBI | |
|
Ansari S, Kumar V, Bhatt DN, Irfan M and Datta A: N-acetylglucosamine sensing and metabolic engineering for attenuating human and plant pathogens. Bioengineering (Basel). 9:642022. View Article : Google Scholar : PubMed/NCBI | |
|
Seo J, Park YS, Kweon TH, Kang J, Son S, Kim HB, Seo YR, Kang MJ, Yi EC, Lee YH, et al: O-Linked N-acetylglucosamine modification of mitochondrial antiviral signaling protein regulates antiviral signaling by modulating its activity. Front Immunol. 11:5892592020. View Article : Google Scholar : PubMed/NCBI | |
|
Song N, Qi Q, Cao R, Qin B, Wang B, Wang Y, Zhao L, Li W, Du X, Liu F, et al: MAVS O-GlcNAcylation is essential for host antiviral immunity against lethal RNA viruses. Cell Rep. 28:2386–2396.e5. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Fang P, He R, Li M, Yu H, Zhou L, Yi Y, Wang F, Rong Y, Zhang Y, et al: O-GlcNAc transferase promotes influenza A virus-induced cytokine storm by targeting interferon regulatory factor-5. Sci Adv. 6:eaaz70862020. View Article : Google Scholar : PubMed/NCBI | |
|
Yao AY, Tang HY, Wang Y, Feng MF and Zhou RL: Inhibition of the activating signals in NK92 cells by recombinant GST-sHLA-G1a chain. Cell Res. 14:155–160. 2004. View Article : Google Scholar : PubMed/NCBI | |
|
Mariuzza RA, Singh P, Karade SS, Shahid S and Sharma VK: Recognition of self and viral ligands by NK cell receptors. Immunol Rev. 329:e134352025. View Article : Google Scholar : PubMed/NCBI | |
|
Yu M, Su Z, Huang X, Zhou Y, Zhang X, Wang B, Wang Z, Liu Y, Xing N, Xia M, et al: Histone methyltransferase Ezh2 negatively regulates NK cell terminal maturation and function. J Leukoc Biol. 110:1033–1045. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lo PW, Shie JJ, Chen CH, Wu CY, Hsu TL and Wong CH: O-GlcNAcylation regulates the stability and enzymatic activity of the histone methyltransferase EZH2. Proc Natl Acad Sci USA. 115:7302–7307. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Feinberg D, Ramakrishnan P, Wong DP, Asthana A and Parameswaran R: Inhibition of O-GlcNAcylation decreases the cytotoxic function of natural killer cells. Front Immunol. 13:8412992022. View Article : Google Scholar : PubMed/NCBI | |
|
Loftus RM, Assmann N, Kedia-Mehta N, O'Brien KL, Garcia A, Gillespie C, Hukelmann JL, Oefner PJ, Lamond AI, Gardiner CM, et al: Amino acid-dependent cMyc expression is essential for NK cell metabolic and functional responses in mice. Nat Commun. 9:23412018. View Article : Google Scholar : PubMed/NCBI | |
|
Ramakrishnan P: O-GlcNAcylation and immune cell signaling: A review of known and a preview of unknown. J Biol Chem. 300:1073492024. View Article : Google Scholar : PubMed/NCBI | |
|
Chang YH, Weng CL and Lin KI: O-GlcNAcylation and its role in the immune system. J Biomed Sci. 27:572020. View Article : Google Scholar : PubMed/NCBI | |
|
Wu JL, Wu HY, Tsai DY, Chiang MF, Chen YJ, Gao S, Lin CC, Lin CH, Khoo KH, Chen YJ and Lin KI: Temporal regulation of Lsp1 O-GlcNAcylation and phosphorylation during apoptosis of activated B cells. Nat Commun. 7:125262016. View Article : Google Scholar : PubMed/NCBI | |
|
Guo M, Price MJ, Patterson DG, Barwick BG, Haines RR, Kania AK, Bradley JE, Randall TD, Boss JM and Scharer CD: EZH2 represses the B cell transcriptional program and regulates Antibody-secreting cell metabolism and antibody production. J Immunol. 200:1039–1052. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Ramakrishnan P, Clark PM, Mason DE, Peters EC, Hsieh-Wilson LC and Baltimore D: Activation of the transcriptional function of the NF-κB protein c-Rel by O-GlcNAc glycosylation. Sci Signal. 6:ra752013. View Article : Google Scholar : PubMed/NCBI | |
|
Swamy M, Pathak S, Grzes KM, Damerow S, Sinclair LV, van Aalten DM and Cantrell DA: Glucose and glutamine fuel protein O-GlcNAcylation to control T cell self-renewal and malignancy. Nat Immunol. 17:712–720. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Liu AR and Ramakrishnan P: Regulation of nuclear Factor-kappaB Function by O-GlcNAcylation in inflammation and cancer. Front Cell Dev Biol. 9:7517612021. View Article : Google Scholar : PubMed/NCBI | |
|
Saha A and Fernández-Tejada A: Chemical biology tools to interrogate the roles of O-GlcNAc in immunity. Front Immunol. 13:10898242022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao M, Ren K, Xiong X, Xin Y, Zou Y, Maynard JC, Kim A, Battist AP, Koneripalli N, Wang Y, et al: Epithelial STAT6 O-GlcNAcylation drives a concerted anti-helminth alarmin response dependent on tuft cell hyperplasia and Gasdermin C. Immunity. 55:13272022. View Article : Google Scholar : PubMed/NCBI | |
|
Xiong X, Huang R, Li Z, Yang C, Wang Q, Ruan HB and Xu L: Intestinal epithelial STAT6 activation rescues the defective Anti-helminth responses caused by ogt deletion. Int J Mol Sci. 23:111372022. View Article : Google Scholar : PubMed/NCBI | |
|
Min A, Lee YA, Kim KA and Shin MH: BLT1-mediated O-GlcNAcylation is required for NOX2-dependent migration, exocytotic degranulation and IL-8 release of human mast cell induced by Trichomonas vaginalis-secreted LTB4. Microbes Infect. 20:376–384. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Montagner S, Leoni C, Emming S, Della Chiara G, Balestrieri C, Barozzi I, Piccolo V, Togher S, Ko M, Rao A, et al: TET2 regulates mast cell differentiation and proliferation through catalytic and Non-catalytic activities. Cell Rep. 15:1566–1579. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Alberione MP, Avalos-Padilla Y, Rangel GW, Ramírez M, Romero-Uruñuela T, Fenollar A, Crispim M, Smith TK, Llinás M and Izquierdo L: Hexosamine biosynthesis disruption impairs GPI production and arrests Plasmodium falciparum growth at schizont stages. PLoS Pathog. 21:e10128322025. View Article : Google Scholar : PubMed/NCBI | |
|
Wang D, Wang C and Zhu G: Genomic reconstruction and features of glycosylation pathways in the apicomplexan Cryptosporidium parasites. Front Mol Biosci. 9:10510722022. View Article : Google Scholar : PubMed/NCBI | |
|
Gowda DC and Miller LH: Glycosylation in malaria parasites: What do we know. Trends Parasitol. 40:131–146. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang N, Jiang N and Chen Q: Key regulators of parasite biology viewed through a Post-translational modification repertoire. Proteomics. December 17–2024.(Epub ahead of print). View Article : Google Scholar | |
|
Hwang JS, Kim KH, Park J, Kim SM, Cho H, Lee Y and Han IO: Glucosamine improves survival in a mouse model of sepsis and attenuates sepsis-induced lung injury and inflammation. J Biol Chem. 294:608–622. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Gong W, Wang H, Li T, Attri KS, Lewis RE, Kalil AC, Bhinderwala F, Powers R, Yin G, et al: O-GlcNAc transferase suppresses inflammation and necroptosis by targeting Receptor-Interacting Serine/Threonine-protein kinase 3. Immunity. 50:576–590.e6. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Lang Y, Li J and Zhang L: O-GlcNAcylation dictates pyroptosis. Front Immunol. 15:15135422024. View Article : Google Scholar : PubMed/NCBI | |
|
Ferron M, Cadiet J, Persello A, Prat V, Denis M, Erraud A, Aillerie V, Mevel M, Bigot E, Chatham JC, et al: O-GlcNAc stimulation: A new metabolic approach to treat septic shock. Sci Rep. 9:187512019. View Article : Google Scholar : PubMed/NCBI | |
|
Denis M, Dupas T, Persello A, Dontaine J, Bultot L, Betus C, Pelé T, Dhot J, Erraud A, Maillard A, et al: An O-GlcNAcylomic approach reveals ACLY as a potential target in sepsis in the young rat. Int J Mol Sci. 22:92362021. View Article : Google Scholar : PubMed/NCBI | |
|
Dupas T, Persello A, Blangy-Letheule A, Denis M, Erraud A, Aillerie V, Leroux AA, Rivière M, Lebreton J, Tessier A, et al: Beneficial effects of O-GlcNAc stimulation in a young rat model of sepsis: Beyond modulation of gene expression. Int J Mol Sci. 23:64302022. View Article : Google Scholar : PubMed/NCBI | |
|
Trapannone R, Rafie K and van Aalten DM: O-GlcNAc transferase inhibitors: Current tools and future challenges. Biochem Soc Trans. 44:88–93. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Elbatrawy AA, Kim EJ and Nam G: O-GlcNAcase: Emerging mechanism, substrate recognition and Small-Molecule inhibitors. ChemMedChem. 15:1244–1257. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Ortiz-Meoz RF, Jiang J, Lazarus MB, Orman M, Janetzko J, Fan C, Duveau DY, Tan ZW, Thomas CJ and Walker S: A small molecule that inhibits OGT activity in cells. ACS Chem Biol. 10:1392–1397. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Fehl C and Hanover JA: Tools, tactics and objectives to interrogate cellular roles of O-GlcNAc in disease. Nat Chem Biol. 18:8–17. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng SS, Mody AC and Woo CM: Opportunities for therapeutic modulation of O-GlcNAc. Chem Rev. 124:12918–13019. 2024. View Article : Google Scholar : PubMed/NCBI |