UDP-glucose pyrophosphorylase 2 (UGP2) is a critical enzyme encoded by the UGP2 gene (NCBI Gene ID: 7360) that catalyzes the conversion of glucose-1-phosphate and UTP into UDP-glucose, a central metabolite in glycogenesis and glycosylation pathways . This enzyme is ubiquitously expressed in human tissues, with particularly high activity in the liver, pancreas, and skeletal muscle . UGP2’s role spans glycogen synthesis, protein N-glycosylation, and cellular energy metabolism, making it a focal point in cancer biology, genetic disorders, and metabolic regulation .
Glycogen Synthesis: UDP-glucose serves as the direct precursor for glycogen production in liver and muscle tissues .
Protein N-Glycosylation: UDP-glucose is a substrate for glycosyltransferases, modifying proteins like EGFR to regulate their stability and signaling .
Stress Adaptation: UGP2 supports cancer cell survival under nutrient-deprived conditions by maintaining UDP-glucose pools .
Transcriptional Control: UGP2 expression is driven by the YAP-TEAD complex, linking it to oncogenic KRAS signaling in pancreatic ductal adenocarcinoma (PDAC) .
Post-Translational Modifications: Phosphorylation and glycosylation modulate its activity, though specific mechanisms remain under investigation .
Barakat-Perenthaler Syndrome: A homozygous mutation (chr2:64083454A > G) ablates the short UGP2 isoform, causing severe epileptic encephalopathy and neurodevelopmental delays .
Glycosylation Targeting: Inhibiting UGP2 disrupts EGFR maturation, sensitizing PDAC cells to EGFR inhibitors like gefitinib .
Ischemic Angiogenesis: In peripheral artery disease (PAD) models, PFKFB3 silencing upregulates UGP2, enhancing perfusion recovery via UDP-glucose–mediated glycosylation .
Parameter | Specification |
---|---|
Purity | >90% (SDS-PAGE) |
Formulation | 0.25 mg/mL in PBS (pH 7.4) with 30% glycerol and 1 mM DTT |
Storage | -20°C (long-term); 4°C for short-term use |
Applications | Enzyme kinetics, glycosylation assays, drug screening |
Cancer Metabolism: UGP2’s dependency in PDAC and other cancers (DepMap data) highlights its potential as a metabolic vulnerability .
Neurological Disorders: Restoring UGP2 isoform balance could mitigate Barakat-Perenthaler syndrome .
Ischemic Disease: Modulating the PFKFB3-UGP2 axis may improve angiogenesis in PAD .
UGP2 (UDP-glucose pyrophosphorylase 2) is an essential enzyme that catalyzes the formation of UDP-glucose from glucose-1-phosphate and UTP. This reaction is reversible and plays a critical role in carbohydrate metabolism. As the sole enzyme responsible for this reaction in mammalian cells, UGP2 serves as a key intermediary in several critical metabolic pathways. In liver and muscle tissue, UDP-glucose functions as a direct precursor for glycogen synthesis. In lactating mammary glands, UDP-glucose is converted to UDP-galactose, which is subsequently transformed into lactose. The enzyme also produces essential substrates for protein N-glycosylation, a post-translational modification crucial for proper protein folding and function .
UGP2 transfers a glucose moiety from glucose-1-phosphate to MgUTP, forming UDP-glucose and MgPPi. This catalytic function positions UGP2 at the convergence of multiple metabolic pathways that are critical for cellular homeostasis. Interestingly, the eukaryotic UGP2 enzyme has no significant sequence similarity to its prokaryotic counterpart, indicating distinct evolutionary paths . Two transcript variants encoding different isoforms have been identified for the UGP2 gene, suggesting potential differential regulation or function .
UGP2 dysregulation has been linked to several significant diseases, with both neurological conditions and cancers featuring prominently:
Neurological disorders:
Cancers with poor prognosis correlation:
Pancreatic ductal adenocarcinoma (PDAC): High UGP2 expression strongly correlates with worse outcomes, particularly in early-phase and low-grade tumors
Glioblastoma (GBM): UGP2 is aberrantly overexpressed and positively correlated with pathologic grade
Non-small cell lung cancer, stomach adenocarcinoma, and gallbladder cancer: Data from the Cancer Genome Atlas revealed that high UGP2 expression correlates with worse prognosis in these cancer types
While complete deletion of Ugp2 is embryonic lethal in mice, studies in zebrafish have demonstrated that organisms with impaired UGP2 function during development can remain viable, suggesting a potential therapeutic window for targeting UGP2 in disease states . This finding is particularly relevant for developing targeted therapies that modulate rather than completely eliminate UGP2 function.
UGP2 expression is regulated through multiple mechanisms that differ between normal physiological conditions and pathological states:
Transcriptional regulation:
The Yes-associated protein 1 (YAP)–TEA domain transcription factor (TEAD) complex directly regulates UGP2 transcription in PDAC cells, identifying UGP2 as a bona fide YAP target gene
YAP, a recognized oncogene, has been identified as an essential mediator of oncogenic KRAS signaling during PDAC progression
YAP can compensate for the inactivation of oncogenic KRAS in several cancer types, suggesting interplay between these major signaling pathways
Pathological upregulation:
In glioblastoma, UGP2 is identified among differentially expressed genes (DEGs), with bioinformatics analysis revealing it as significantly upregulated
Random survival forest modeling identified UGP2 as having a significant effect on GBM prognosis
In PDAC, UGP2 transcription appears to be enhanced as part of the metabolic reprogramming driven by oncogenic signaling
Tissue-specific expression:
While UGP2 is expressed in multiple tissues to support normal metabolism, its expression levels and regulatory mechanisms may vary by tissue type
Two transcript variants encoding different isoforms have been found for UGP2, suggesting potentially different regulatory mechanisms depending on tissue context
Understanding these regulatory mechanisms is crucial for developing therapeutic strategies that target UGP2 in cancer while minimizing adverse effects on normal cellular function.
UGP2 contributes to cancer progression through multiple interconnected molecular mechanisms:
Regulation of glycogen synthesis and energy metabolism:
UGP2 produces UDP-glucose, which serves as a direct precursor for glycogen synthesis
In nutrient-deprived tumor microenvironments characteristic of PDAC, glycogen serves as a critical energy reserve
UGP2 knockdown decreases intracellular glycogen levels in PDAC cells, compromising their ability to survive in low-nutrient conditions
Studies show that cells with reduced UGP2 expression exhibit worse survival in low-glucose environments, with proliferation rescued by UDP-glucose supplementation
Impact on protein N-glycosylation:
UGP2 depletion causes a large-scale decrease in glycan modifications across multiple proteins
Knockdown significantly reduces 141 N-glycosylation modifications across 89 proteins, with most changes not explained by corresponding alterations in total protein levels
This impaired glycosylation affects critical cell surface receptors, including the epidermal growth factor receptor (EGFR)
Proper glycosylation is essential for receptor stability, localization, and function
Alteration of EGFR signaling:
Promotion of cell proliferation, migration, and invasion:
Functional enrichment analysis shows UGP2 involvement in biological processes of cell proliferation, migration, and invasion
Loss-of-function studies demonstrate that UGP2 knockdown decreases glioblastoma cell (U251) growth, migration, and invasion both in vitro and in vivo
In PDAC xenograft models, UGP2 knockdown halts tumor growth and reduces proliferative index as shown by Ki67 staining
The convergence of these mechanisms highlights how UGP2 functions as a central metabolic regulator that supports cancer cell survival, growth, and invasive potential through both energy metabolism and proper protein function.
The relationship between UGP2, YAP-TEAD signaling, and oncogenic KRAS represents a critical regulatory axis in cancer progression:
Transcriptional regulation of UGP2 by YAP-TEAD:
Research has identified UGP2 as a direct transcriptional target of the YAP–TEAD complex in PDAC cells
YAP (Yes-associated protein 1) is a transcriptional co-activator that partners with TEAD (TEA domain) transcription factors to regulate gene expression
This direct regulation establishes UGP2 as a bona fide YAP target gene and integrates it into YAP's broader oncogenic program
YAP as a mediator of KRAS signaling:
Metabolic implications of the KRAS-YAP-UGP2 axis:
Oncogenic KRAS drives metabolic reprogramming in cancer cells to support their increased anabolic demands
YAP contributes to this reprogramming through its target genes, including UGP2
UGP2, in turn, supports crucial metabolic processes including glycogen synthesis and protein N-glycosylation
This cascade provides cancer cells with survival and growth advantages, particularly in nutrient-stressed microenvironments
Therapeutic implications:
The UGP2-YAP-KRAS connection suggests that targeting UGP2 might disrupt KRAS-dependent oncogenic pathways
This is particularly relevant given that KRAS mutations are common in many cancers but have historically been difficult to target directly
Inhibition of UGP2 could potentially provide an alternative approach to counter the effects of oncogenic KRAS signaling
This interconnection underscores UGP2's position as a central node where critical cancer signaling pathways and altered metabolism converge, offering potential insights for therapeutic intervention.
UGP2 plays a crucial role in regulating protein N-glycosylation with significant implications for cancer cell biology:
Mechanism of UGP2's impact on glycosylation:
UGP2 catalyzes the formation of UDP-glucose, which serves as a substrate for numerous glycosylation reactions
UDP-glucose is a critical precursor for N-glycan synthesis, which involves the stepwise addition of sugar moieties to asparagine residues on target proteins
Knockdown of UGP2 in cancer cells results in large-scale decreases in glycan modifications across multiple proteins
Specific glycosylation targets affected:
Proteomic analysis reveals that UGP2 knockdown significantly decreases 141 N-glycosylation modifications spread across 89 different proteins
These modifications are not explained by corresponding changes in total protein levels, indicating a direct effect on the glycosylation process rather than protein expression
The glycosylation changes also do not reflect global alterations in glycan-chain architectures, suggesting specific effects on particular glycosylation sites
Impact on receptor tyrosine kinase function:
A key target affected by UGP2-mediated glycosylation is the epidermal growth factor receptor (EGFR)
UGP2 depletion impairs downstream EGFR signaling, indicating that these glycosylation moieties regulate EGFR function
Proper N-glycosylation is essential for EGFR folding, stability, trafficking to the cell surface, and ligand binding
Biological implications in cancer:
Altered glycosylation affects receptor tyrosine kinase signaling, which drives proliferation, survival, and metastasis
Glycosylation changes can alter cancer cell recognition by the immune system, potentially affecting immune surveillance
Modified glycosylation patterns influence cell adhesion properties, which may impact invasion and metastasis
The specific constellation of glycosylation changes induced by UGP2 dysregulation may create unique vulnerabilities in cancer cells
This regulatory role positions UGP2 as a master controller of protein function through post-translational modification, with wide-ranging effects on signaling networks that drive cancer progression. The specific nature of the glycosylation changes induced by UGP2 alteration suggests potential for targeted therapeutic approaches.
Several complementary experimental approaches have proven effective for studying UGP2 function in cancer:
Bioinformatic analysis:
Mining gene expression datasets (e.g., Gene Expression Omnibus database, The Cancer Genome Atlas) to identify UGP2 expression patterns across cancer types
Differential gene expression analysis using R-based tools and pathway enrichment analysis through Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology
Random survival forest modeling to correlate UGP2 expression with patient outcomes
These approaches identified UGP2 as significantly upregulated in GBM and associated with poor prognosis
Loss-of-function studies:
RNA interference (siRNA or shRNA) for temporary or stable knockdown of UGP2 expression
CRISPR-Cas9 gene editing for complete knockout studies
Functional rescue experiments using UDP-glucose supplementation to confirm specificity of observed phenotypes
These approaches demonstrated that UGP2 knockdown decreases cancer cell growth, migration, and invasion both in vitro and in vivo
In vitro cancer models:
2D cell culture systems using established cancer cell lines (e.g., MiaPaca2 and Suit2 for PDAC, U251 for glioblastoma)
3D culture models to better recapitulate tumor architecture and microenvironment
Metabolic stress experiments (e.g., low glucose conditions) to assess UGP2's role in stress adaptation
Migration and invasion assays to evaluate effects on cancer cell motility
In vivo cancer models:
Proteomic and glycosylation analysis:
Mass spectrometry-based identification of N-glycosylation modifications affected by UGP2 knockdown
Parallel global proteomics to distinguish between changes in glycosylation versus protein expression levels
Glycan chain architecture analysis to characterize specific effects on glycosylation patterns
Signaling pathway analysis:
These complementary approaches provide robust evidence for UGP2's role in cancer cell biology and identify potential vulnerabilities that could be therapeutically exploited.
Optimal analytical methods for measuring UGP2 enzymatic activity and its metabolic products include:
Direct enzymatic activity assays:
Spectrophotometric coupled enzyme assays that measure the rate of UDP-glucose formation
Radiometric assays using 14C-labeled glucose-1-phosphate to trace product formation
Colorimetric phosphate release assays that quantify the pyrophosphate generated during the reaction
These methods provide quantitative measurement of UGP2 catalytic function in purified systems or cell lysates
UDP-glucose quantification:
Liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) for sensitive and specific quantification of UDP-glucose levels in cell or tissue extracts
High-performance liquid chromatography (HPLC) with UV detection
Enzymatic cycling assays that amplify the detection signal for increased sensitivity
These techniques allow researchers to directly measure the primary product of UGP2 activity
Glycogen content analysis:
Periodic acid-Schiff (PAS) staining for histological visualization of glycogen in tissue samples
Enzymatic methods using amyloglucosidase to break down glycogen followed by glucose measurement
Glycogen extraction and quantification using anthrone reagent colorimetric assay
These approaches showed that UGP2 knockdown decreases intracellular glycogen levels in cancer cells
Isotope tracing studies:
13C-labeled glucose tracing combined with mass spectrometry to track carbon flow through UGP2 into glycogen and other metabolic products
Nuclear magnetic resonance (NMR) spectroscopy for structural analysis of labeled metabolic products
These methods provide dynamic information about UGP2's role in cellular metabolism
N-glycosylation profiling:
Lectin microarrays for broad profiling of glycosylation patterns
MALDI-TOF mass spectrometry for glycan structure analysis
Glycoproteomics using mass spectrometry to identify specific N-glycosylation sites affected by UGP2 activity
These techniques revealed that UGP2 knockdown significantly decreases 141 N-glycosylation modifications across 89 proteins
Metabolic flux analysis:
Stable isotope-resolved metabolomics (SIRM) to quantify metabolic flux through UGP2-dependent pathways
Computational modeling of metabolic networks incorporating UGP2 activity
These approaches provide systems-level insights into how UGP2 influences cellular metabolism
In situ methods:
Fluorescence resonance energy transfer (FRET)-based sensors for real-time monitoring of UDP-glucose in living cells
Activity-based protein profiling (ABPP) to assess UGP2 activity in complex biological samples
These techniques allow for spatial and temporal resolution of UGP2 activity
These analytical methods, especially when used in combination, provide comprehensive insights into UGP2's enzymatic function and metabolic impact in both normal and pathological contexts.
Designing effective UGP2 inhibitors for cancer therapy requires careful consideration of several key factors:
Structural and biochemical considerations:
UGP2 is the sole enzyme responsible for UDP-glucose synthesis in mammalian cells, suggesting potential for high specificity
Understanding active site structure and catalytic mechanism to design competitive inhibitors
Exploring allosteric sites that might offer alternative means of inhibition
Considering both the catalytic domain and potential regulatory domains as targets
Selectivity profiling:
Therapeutic window assessment:
While complete deletion of UGP2 is embryonic lethal in mice, partial inhibition may be tolerated in adult tissues
Determining the degree of inhibition needed for anti-cancer effects versus toxicity to normal tissues
Zebrafish studies suggest that organisms with impaired UGP2 function during development can remain viable, indicating potential for a therapeutic window
Cancer-specific considerations:
Metabolic bypass mechanisms:
Anticipating potential metabolic adaptations or bypass pathways that might confer resistance
Considering combination approaches that target both UGP2 and potential compensatory pathways
Designing inhibitors that might synergize with glycolysis inhibitors or other metabolic therapies
Delivery and pharmacokinetic considerations:
For brain tumors like glioblastoma, inhibitors must cross the blood-brain barrier
For pancreatic cancer, considering the dense stromal environment that often limits drug delivery
Optimizing drug-like properties including stability, half-life, and tumor penetration
Biomarker development:
Identifying markers that predict sensitivity to UGP2 inhibition, such as YAP activity or glycogen dependence
Developing methods to monitor UGP2 inhibition in vivo, such as glycosylation profiling
Creating companion diagnostics to guide patient selection
Alternative targeting strategies:
Exploring targeted protein degradation approaches (e.g., PROTACs) as alternatives to catalytic inhibition
Considering disruption of protein-protein interactions, such as those involving the YAP-TEAD complex that regulates UGP2 expression
Investigating opportunities to target UGP2 indirectly through upstream regulatory mechanisms
These considerations highlight both the challenges and opportunities in developing UGP2 inhibitors as cancer therapeutics. The unique position of UGP2 at the intersection of multiple metabolic pathways and cancer signaling networks makes it a promising but complex target.
UGP2 expression shows significant correlations with clinical outcomes across multiple cancer types, though with some variation:
Pancreatic ductal adenocarcinoma (PDAC):
Glioblastoma and other brain tumors:
UGP2 expression is aberrantly overexpressed in human glioma and positively correlated with pathologic grade
UGP2 has been identified as having a significant effect on GBM prognosis based on random survival forest modeling
Expression is associated with biological processes of cell proliferation, migration, and invasion
Other solid tumors:
Cancer types without significant correlation:
Multi-cancer analysis:
The pattern of correlation between UGP2 expression and outcomes across multiple cancer types suggests its role may be most critical in highly glycolytic cancers or those existing in nutrient-deprived microenvironments
The strongest associations appear in cancers known for metabolic reprogramming and aggressive growth patterns
These clinical correlations provide important validation of the biological findings regarding UGP2's role in cancer progression and suggest its potential utility as both a prognostic biomarker and therapeutic target, particularly in PDAC and glioblastoma.
Several potential biomarkers could predict sensitivity to UGP2-targeted therapies:
UGP2 expression levels:
High baseline expression of UGP2 may indicate greater dependency on its function
Immunohistochemistry or RNA-based quantification of UGP2 in tumor samples could serve as a primary screening biomarker
Cancer types shown to have poor prognosis with high UGP2 expression (PDAC, glioblastoma, non-small cell lung cancer) may be particularly sensitive
YAP/TEAD activity markers:
KRAS mutation status:
Glycogen content:
N-glycosylation profiles:
Metabolic stress markers:
Since UGP2 appears particularly important under nutrient-deprived conditions, markers of metabolic stress might predict sensitivity
Hypoxia markers (HIF-1α, CA9)
Nutrient stress response proteins (GRP78, ATF4)
These might identify tumors existing in microenvironments where UGP2 function is most critical
Combination biomarker panels:
Integrating multiple markers into a predictive signature may provide greater accuracy
Machine learning approaches applied to multi-omic data could identify complex patterns associated with UGP2 dependency
Combining genomic, transcriptomic, and metabolomic markers may capture the multifaceted role of UGP2
Functional assays:
Ex vivo drug sensitivity testing using patient-derived organoids or explants
Metabolic flux analysis to assess real-time dependency on UGP2-related pathways
These functional approaches could provide direct evidence of sensitivity beyond static biomarkers
Developing and validating these potential biomarkers would be an essential component of any clinical development program for UGP2-targeted therapies, enabling appropriate patient selection and potentially improving therapeutic outcomes.
The most promising future directions in UGP2 research for cancer therapy span several interconnected areas:
Therapeutic development:
Design and optimization of small molecule UGP2 inhibitors based on structural insights and enzymatic mechanisms
Development of alternative approaches such as targeted protein degradation (PROTACs) directed at UGP2
Exploration of combinatorial approaches targeting UGP2 alongside other metabolic vulnerabilities or standard chemotherapies
These development efforts could provide first-in-class therapies for aggressive cancers with limited treatment options
Expanded cancer type investigation:
While PDAC and glioblastoma show strong evidence for UGP2 dependency, expanding research to other cancer types with poor prognosis correlations is warranted
Systematic screening across diverse cancer types to identify those most dependent on UGP2 function
Investigation of rare cancers that may share metabolic features with PDAC or glioblastoma
Deeper mechanistic understanding:
Biomarker development and validation:
Prospective validation of UGP2 expression as a prognostic marker across cancer types
Development of companion diagnostics to identify patients most likely to benefit from UGP2-targeted therapy
Multi-omic approaches to create integrated predictive signatures
Resistance mechanisms:
Identification of potential metabolic bypass pathways that may confer resistance to UGP2 inhibition
Understanding adaptive responses to UGP2 targeting in different genetic and microenvironmental contexts
Development of strategies to prevent or overcome resistance
Expanded therapeutic applications:
Investigation of UGP2's role in metastasis and exploration of its potential as an anti-metastatic target
Evaluation of UGP2 inhibition in combination with immunotherapy
Assessment of UGP2's role in cancer stem cell maintenance and therapeutic resistance
Translational research platforms:
Development of patient-derived organoid models to test UGP2 targeting in personalized medicine approaches
Creation of genetically engineered mouse models with inducible UGP2 knockdown to evaluate systemic effects of inhibition
Implementation of glycomic and metabolomic platforms in clinical trials to monitor UGP2 inhibition
These research directions collectively address the most critical questions surrounding UGP2 as a therapeutic target and could significantly advance our ability to exploit this metabolic vulnerability in cancer treatment. The convergent evidence from multiple studies highlights UGP2's central role in cancer metabolism and its potential as a therapeutic target worthy of continued investigation .
Combination strategies could significantly enhance the efficacy of UGP2-targeted approaches through several mechanisms:
Co-targeting metabolic dependencies:
Combining UGP2 inhibition with glycolysis inhibitors (e.g., 2-deoxyglucose) to comprehensively disrupt glucose metabolism
Pairing with glutaminase inhibitors to block alternative energy sources, as cancer cells might attempt to compensate for UGP2 inhibition through glutamine metabolism
Targeting both glycogen synthesis (via UGP2) and glycogen breakdown (via glycogen phosphorylase inhibitors) to completely block glycogen utilization
These approaches would create synthetic lethality by blocking multiple metabolic escape routes
Enhancing oncogenic pathway inhibition:
Combining UGP2 inhibition with YAP-TEAD inhibitors, given their regulatory relationship
Pairing with KRAS pathway inhibitors in KRAS-mutant cancers, as UGP2 appears connected to KRAS signaling through YAP
Adding EGFR inhibitors to capitalize on the impaired EGFR signaling observed with UGP2 depletion
These combinations could disrupt both metabolic and signaling dependencies of cancer cells
Overcoming stromal barriers:
In PDAC, combining UGP2 inhibition with stroma-targeting agents to improve drug delivery
Pairing with hyaluronidase to decrease interstitial pressure and enhance penetration of UGP2 inhibitors
These approaches would address the physical barriers that often limit therapeutic efficacy in PDAC
Enhancing immunotherapy:
Investigating whether UGP2 inhibition affects tumor cell recognition by immune cells through altered glycosylation patterns
Combining with immune checkpoint inhibitors if UGP2 inhibition creates a more immunogenic tumor phenotype
These combinations could convert "cold" tumors to "hot" immunologically responsive tumors
Targeting cancer stem cells:
Exploring UGP2 inhibition in combination with cancer stem cell-targeting agents
Investigating whether glycogen metabolism and protein glycosylation are particularly important for cancer stem cell maintenance
These approaches could address the cellular heterogeneity that often leads to therapeutic resistance
Enhancing standard chemotherapy:
Combining UGP2 inhibition with gemcitabine or FOLFIRINOX in PDAC
Pairing with temozolomide in glioblastoma
Testing whether metabolic stress induced by UGP2 inhibition sensitizes cancer cells to DNA-damaging agents
These combinations could improve outcomes with established treatment regimens
Synthetic lethal approaches:
Screening for genetic contexts that create enhanced sensitivity to UGP2 inhibition
Identifying complementary targets that, when inhibited alongside UGP2, create catastrophic metabolic failure
These approaches could identify patient subgroups likely to show dramatic responses
Each of these combination strategies addresses a different aspect of cancer biology and could be tailored to specific cancer types based on their molecular and metabolic profiles. The unique position of UGP2 at the intersection of glycogen metabolism and protein glycosylation makes it particularly attractive for combination approaches that exploit multiple cancer vulnerabilities simultaneously.
UGP2 catalyzes the conversion of glucose-1-phosphate and uridine triphosphate (UTP) to UDP-glucose and pyrophosphate (PPi). This reaction is crucial for the formation of UDP-glucose, which serves as a glycosyl donor in the biosynthesis of glycogen and glycoproteins . The enzyme’s activity is dependent on the presence of magnesium ions (Mg2+), which act as cofactors in the reaction .
UDP-glucose is a central metabolite in the biosynthesis of glycogen, a storage form of glucose, and in the glycosylation of proteins and lipids. Glycosylation is a critical post-translational modification that affects protein folding, stability, and function. UGP2’s role in these processes underscores its importance in maintaining cellular homeostasis and energy balance .
Recent studies have highlighted the significance of UGP2 in cancer metabolism, particularly in pancreatic ductal adenocarcinoma (PDAC). UGP2 is upregulated in some cancers, and its expression is regulated by the Yes-associated protein 1 (YAP)-TEA domain transcription factor (TEAD) complex . Loss of UGP2 leads to decreased intracellular glycogen levels and defects in N-glycosylation targets, including the epidermal growth factor receptor (EGFR), which are crucial for cell growth and survival . These findings suggest that UGP2 could be a potential therapeutic target for cancer treatment.
Human recombinant UGP2 is produced using recombinant DNA technology, which involves inserting the human UGP2 gene into a suitable expression system, such as bacteria or yeast. This allows for the large-scale production of the enzyme for research and therapeutic purposes. Recombinant UGP2 retains the same biochemical properties and functions as the native enzyme, making it a valuable tool for studying carbohydrate metabolism and developing potential treatments for metabolic disorders and cancers .