UPP1 is a cytosolic, dimeric enzyme encoded by the UPP1 gene on human chromosome 7. Key features include:
Molecular Weight: 29.3 kDa (recombinant form expressed in E. coli) .
Structure: Comprises 310 amino acids (Ala2-Ala310) with an N-terminal 6-His tag in recombinant forms .
Catalytic Mechanism: Binds substrates such as uridine and 5-fluorouracil (5-FU) through a flexible active site, enabling hinge-like domain motions for ligand exchange .
Specific Activity Calculation
The enzymatic activity of recombinant UPP1 is quantified using the formula:
Parameter | Formula |
---|---|
Specific Activity | |
Example: Under standard assay conditions (1 mM uridine, 0.1 µg enzyme), UPP1 exhibits robust activity .
UPP1 sustains nucleotide pools and energy metabolism via:
Pyrimidine Salvage: Converts uridine to uracil, recycling pyrimidine bases for RNA/DNA synthesis .
Metabolic Flexibility: In nutrient-limited conditions, UPP1-derived ribose-1-phosphate fuels glycolysis and the pentose phosphate pathway, supporting tumor survival .
Uridine Homeostasis: Regulates plasma uridine levels, influencing cellular responses to stress and chemotherapy .
UPP1 is overexpressed in numerous malignancies, correlating with aggressive phenotypes:
Chemotherapy Synergy: UPP1 activates prodrugs like 5-FU and capecitabine, improving their cytotoxicity in cancers .
Inhibitors: Targeting UPP1 reduces uridine catabolism, elevating endogenous uridine levels to protect healthy tissues during chemotherapy .
Glycolysis Targeting: UPP1-high tumors exhibit resistance to glycolysis inhibitors (e.g., 2-DG), suggesting combinational therapies .
In vitro: UPP1 knockdown reduces ATP production, lactate levels, and cell proliferation in lung adenocarcinoma .
In vivo: UPP1 overexpression accelerates tumor growth and metastasis in xenograft models .
Prognostic Marker: High UPP1 expression predicts poor outcomes in oral squamous cell carcinoma and glioblastoma .
Drug Resistance: UPP1-mediated AKT activation blunts gemcitabine efficacy in bladder cancer, reversible via MK2206 (AKT inhibitor) .
Epigenetic Regulation: Histone acetylation (H3K27ac, H3K9ac) upregulates UPP1, highlighting epigenetic therapies like CBP/p300 inhibitors .
UPP1 (Uridine Phosphorylase 1) is an enzyme that catalyzes the reversible phosphorolysis of uridine (or 2'-deoxyuridine) to uracil and ribose-1-phosphate (or deoxyribose-1-phosphate) . This reaction plays a critical role in the pyrimidine salvage pathway, which enables cells to recycle nucleosides for nucleotide synthesis.
The functional analysis of UPP1 requires multiple methodological approaches:
Enzyme activity assays using LC-MS with isotope-labeled substrates (e.g., 13C9,15N2-uridine)
Expression analysis via RT-qPCR, western blotting, or immunohistochemistry
Genetic manipulation through siRNA knockdown or CRISPR-Cas9 gene editing
In normal physiology, UPP1 helps maintain nucleoside homeostasis, but its dysregulation in cancer contributes to various pathological processes including altered immune responses and tumor progression .
UPP1 exhibits a distinct expression pattern across human tissues and cell populations:
Immune cells: Highest expression is observed in neutrophils, particularly in mature (Ly6G-high) neutrophils. Expression increases during neutrophil maturation and activation, especially after inflammatory triggers .
Myeloid lineage: Significant expression across myeloid cells, as demonstrated by CD11b-DTR transgenic mouse models where depletion of CD11b-expressing cells resulted in decreased serum uracil levels .
T-cells: Minimal to undetectable expression under normal conditions .
Brain tissue: Lower expression in normal brain compared to glioma tissue, with particular upregulation in the mesenchymal subtype of glioma .
Lung tissue: Expression levels increase in pre-metastatic lung environment in the presence of primary mammary tumors .
Methodology for studying tissue distribution includes single-cell RNA sequencing, flow cytometry sorting of specific cell populations, and immunohistochemical analysis of tissue samples.
For robust assessment of UPP1 activity in biological samples, researchers should employ complementary techniques:
For enzymatic activity:
LC-MS measurement: Using isotope-labeled substrates (13C9,15N2-uridine) and quantifying the labeled product (13C4,15N2-uracil) provides the most direct measure of UPP1 catalytic activity .
Serum uracil quantification: Serves as an indirect measure of UPP1 activity in vivo .
For expression analysis:
RT-qPCR: Quantifies UPP1 mRNA levels with high sensitivity .
Single-cell RNA sequencing: Allows identification of UPP1-expressing cell populations within heterogeneous samples .
Proteomics: Western blotting and mass spectrometry for protein quantification.
For functional assessment:
Genetic manipulation: siRNA knockdown followed by cellular function assays (proliferation, migration, invasion) .
Pharmacological inhibition: Using UPP1-specific inhibitors with subsequent phenotypic analysis.
Co-culture experiments: Particularly useful for studying UPP1's effects on immune cell interactions .
These methodological approaches should be selected based on the specific research question and available samples.
UPP1 exerts dual effects on cancer progression and immune function through several mechanisms:
Cancer progression effects:
Promotes tumor cell proliferation and invasion, as demonstrated in glioma cell lines (U251 and LN229) through EdU and Transwell assays .
Contributes to metastasis formation, particularly in mammary cancer spreading to the lungs .
Elevates expression of PD-L1 through the PI3K/AKT/mTOR pathway in lung adenocarcinoma .
Increases release of immunosuppressive cytokines, with TGF-β1 being particularly prominent .
Immune system interactions:
Alters neutrophil behavior by modifying adhesion molecule expression, leading to decreased neutrophil motility in the pre-metastatic lung .
UPP1-expressing neutrophils suppress T-cell proliferation, creating an immunosuppressive environment .
Affects macrophage recruitment and function in the tumor microenvironment .
Associates with immune modulators CD274, CD276, CD28, and ICOSLG in gliomas .
Influences immune cell composition, including dendritic cells, B cells, T cells, MDSCs, Tregs, and macrophages .
The relationship between UPP1 and immune function appears bidirectional, as inflammatory signals can increase UPP1 expression in neutrophils, which then contributes to immunosuppression in cancer contexts .
UPP1 expression is regulated through multiple layers of control that differ between normal and pathological states:
Transcriptional regulation:
Inflammatory signals: LPS challenge potently increases Upp1 expression in neutrophils isolated from bone marrow, blood, and spleen .
Maturation factors: Upp1 expression increases during neutrophil maturation, with higher levels in Ly6G-high (mature) compared to Ly6G-intermediate (immature) neutrophils .
Malignancy signals: UPP1 expression correlates positively with WHO grade in gliomas, suggesting regulation by malignancy-associated factors .
Molecular subtypes: In gliomas, UPP1 is significantly upregulated in the mesenchymal subtype compared to other molecular subtypes .
Post-translational regulation:
Enzymatic activity can be modulated independently of expression levels.
UPP1 may be involved in protein turnover regulation via the ubiquitin-proteasome pathway .
Methodological approaches to study regulation:
Promoter analysis and chromatin immunoprecipitation to identify transcription factors.
Reporter gene assays to assess promoter activity under different conditions.
Phosphoproteomics to identify post-translational modifications affecting enzyme activity.
Metabolic profiling to understand how substrate availability influences UPP1 function.
Understanding these regulatory mechanisms could reveal potential intervention points for therapeutic targeting.
UPP1-related biomarkers hold significant promise for cancer diagnostics and prognostication:
Expression-based biomarkers:
UPP1 mRNA levels: Higher expression correlates with worse survival in glioma patients .
UPP1high tumor cell clusters: Identified through single-cell RNA sequencing in lung adenocarcinoma, associated with invasive front location and poorer outcomes .
Mesenchymal subtype marker: UPP1 could serve as a potential biomarker for identifying the aggressive mesenchymal subtype of glioma .
Functional biomarkers:
Serum uracil levels: As a product of UPP1 activity, elevated uracil might serve as a minimally invasive biomarker .
Immunosuppressive cytokine profile: TGF-β1 and other immunosuppressive cytokines associated with UPP1 expression .
Immune checkpoint molecules: UPP1 expression correlates with immune modulators CD274, CD276, CD28, and ICOSLG .
Predictive biomarkers:
Treatment response prediction: UPP1 effectively predicts immunotherapy responses in multiple cancer cohorts .
Drug sensitivity indicator: UPP1high tumors showed relatively increased sensitivity to Bosutinib and Dasatinib in patient-derived organoids .
Methodological approaches for biomarker development include multivariate analysis of large patient datasets, machine learning algorithms for pattern recognition, and validation across independent cohorts with diverse treatment histories.
UPP1 plays a multifaceted role in establishing the pre-metastatic niche, particularly in the context of mammary cancer metastasis to the lung:
Neutrophil-mediated mechanisms:
UPP1 alters the expression of adhesion molecules on neutrophils, resulting in decreased neutrophil motility specifically in the pre-metastatic lung .
This modified neutrophil behavior creates a permissive environment for circulating tumor cells to establish secondary tumors .
Extracellular matrix remodeling:
UPP1 generates uracil, which increases fibronectin deposition in the extracellular microenvironment .
Enhanced fibronectin content creates a favorable scaffold for metastatic seeding and growth .
Immunosuppressive microenvironment:
UPP1-expressing neutrophils suppress T-cell proliferation, inhibiting anti-tumor immune responses in the pre-metastatic site .
This immune evasion mechanism protects arriving metastatic cells from elimination .
Experimental evidence and methodology:
Knockout or inhibition of UPP1 in mice with mammary tumors produces three significant effects:
These findings demonstrate that UPP1 expression, particularly in neutrophils, prepares the "soil" of the pre-metastatic niche, making it more receptive for circulating tumor cells to establish metastases. Methodologically, this research employed genetic knockout models, pharmacological inhibition approaches, and comprehensive analysis of the lung microenvironment.
UPP1 orchestrates immune evasion across multiple cancer types through distinct but overlapping molecular mechanisms:
In gliomas:
Immune checkpoint regulation: UPP1 is significantly associated with immune checkpoint molecules CD274 (PD-L1), CD276, CD28, and ICOSLG .
Immune cell infiltration: High UPP1 expression correlates with altered immune cell composition, including changes in dendritic cells, B cells, T cells, MDSCs, Tregs, and macrophages .
Macrophage function: UPP1 silencing enhances recruitment and activation of macrophages, suggesting its normal expression suppresses macrophage-mediated anti-tumor immunity .
In lung adenocarcinoma:
Cytokine secretion: UPP1 upregulation increases release of immunosuppressive cytokines, with TGF-β1 being particularly prominent .
PD-L1 expression: UPP1 elevates PD-L1 expression through the PI3K/AKT/mTOR pathway, contributing to CD8+ T cell suppression .
Spatial location: UPP1high tumor cells are primarily located at the tumor invasive front, strategically positioned to interact with infiltrating immune cells .
In mammary cancer:
T-cell suppression: UPP1-expressing neutrophils directly suppress T-cell proliferation .
Neutrophil recruitment: UPP1 affects neutrophil behavior and recruitment to pre-metastatic sites .
Methodological approaches to study these mechanisms:
Single-cell RNA sequencing to identify cell-specific expression patterns
Cytometry by time-of-flight (CyTOF) for detailed immune cell profiling
Co-culture experiments to assess cellular interactions
Cytokine arrays to measure secreted immunomodulatory factors
In vivo models with genetic or pharmacological manipulation of UPP1
These findings collectively demonstrate that UPP1 functions as a master regulator of the immunosuppressive tumor microenvironment across different cancer types, though with tissue-specific mechanisms that require tailored therapeutic approaches.
Machine learning offers powerful tools for uncovering UPP1-associated gene signatures and functional pathways. A comprehensive methodological approach includes:
Data integration and preprocessing:
Combine multiple data types: single-cell RNA sequencing, bulk RNA sequencing, proteomics, and clinical data .
Normalize and batch-correct data from different sources.
Perform feature selection to identify the most informative variables.
Supervised learning methods:
LASSO (Least Absolute Shrinkage and Selection Operator) regression: Used to identify prognostic genes associated with UPP1 .
Random Forest models: For classification of UPP1-high versus UPP1-low tumors and prediction of associated features.
Support Vector Machines: To identify robust biomarkers across cancer types.
Unsupervised learning approaches:
Clustering algorithms: To identify patient subgroups based on UPP1 and associated gene expression patterns.
Dimensionality reduction techniques: Such as t-SNE or UMAP to visualize relationships between UPP1 and other genes.
Network analysis: To construct protein-protein interaction networks centered on UPP1.
Pathway analysis integration:
Gene Set Enrichment Analysis (GSEA): To identify pathways enriched in UPP1-high versus UPP1-low tumors .
Gene Ontology analysis: Revealed UPP1 association with immune and inflammatory response pathways .
Gene Sets Variation Analysis: Showed correlation between UPP1 and MHC-II and LCK pathways .
Validation and application:
Cross-validation techniques to ensure model robustness.
Testing on independent cohorts to confirm predictive power.
Application to drug response prediction: The R package oncoPredict was used to predict drug responses related to UPP1 .
As demonstrated in glioma research, this integrative machine learning approach successfully identified UPP1 as a novel driver of tumorigenesis and immune evasion, with significant implications for patient prognosis and treatment response .
Targeting UPP1 in cancer treatment involves multiple complementary approaches:
Direct enzyme inhibition:
Development of specific UPP1 catalytic site inhibitors.
Structure-based drug design leveraging knowledge of UPP1's enzymatic mechanism.
Small molecule screening to identify novel inhibitory compounds.
Expression modulation:
siRNA or antisense oligonucleotides for transient UPP1 knockdown.
CRISPR-based approaches for permanent genetic modification.
Promoter-targeting strategies to reduce transcription.
Pathway-based approaches:
Combination with immunotherapy: Since UPP1 is associated with immune checkpoint molecules, combining UPP1 inhibition with immune checkpoint blockade might enhance efficacy .
PI3K/AKT/mTOR pathway inhibitors: As UPP1 elevates PD-L1 through this pathway in lung adenocarcinoma, combining UPP1 targeting with pathway inhibitors could be synergistic .
Anti-TGF-β1 therapies: Since UPP1 increases TGF-β1 production, combining UPP1 inhibition with TGF-β blockade might be effective .
Precision medicine strategies:
Biomarker-guided therapy: High UPP1 expression could identify patients likely to benefit from specific kinase inhibitors (Bosutinib and Dasatinib) in lung adenocarcinoma .
Cancer-type specific approaches: Since UPP1 has varying mechanisms across cancer types, therapeutic strategies should be tailored accordingly.
Methodological considerations:
In vivo models to assess therapeutic efficacy and immune effects.
Sequential or concurrent drug administration strategies.
Development of delivery systems to target specific cell populations (e.g., neutrophils vs. tumor cells).
Experimental evidence suggests that targeting UPP1 could simultaneously inhibit tumor cell proliferation and invasion while enhancing anti-tumor immune responses, making it a promising multi-modal therapeutic target .
UPP1 expression demonstrates consistent but context-dependent correlations with clinical outcomes:
Gliomas:
Prognostic value: Higher UPP1 expression correlates with worse patient survival .
Grade association: UPP1 expression level is positively correlated with WHO grade of glioma .
Molecular subtype: Significantly upregulated in the aggressive mesenchymal subtype of glioma .
IDH status correlation: Likely associated with IDH-wildtype status (implied by its connection to mesenchymal subtype and poorer prognosis) .
Lung adenocarcinoma:
Tumor heterogeneity: UPP1high tumor cell clusters, identified through single-cell RNA sequencing, are associated with the invasive front of tumors .
Immunosuppressive microenvironment: UPP1high tumor cells display stronger association with immunosuppressive components in the TME .
Drug sensitivity: UPP1high tumors exhibit relatively increased sensitivity to specific kinase inhibitors (Bosutinib and Dasatinib) .
Mammary cancer:
Metastatic potential: UPP1 expression is associated with increased lung metastasis formation .
Neutrophil association: UPP1 expression in neutrophils correlates with their immunosuppressive behavior .
Immunotherapy context:
Predictive biomarker: UPP1 effectively predicts immunotherapy responses across multiple cancer cohorts .
Paradoxical effect: Interestingly, high UPP1 expression is associated with better survival in immunotherapy cohorts, despite its generally poor prognostic value in untreated cancers .
Methodological approaches for outcome correlation:
Kaplan-Meier survival analysis with log-rank tests.
Cox proportional hazards regression for multivariate analysis.
ROC curve analysis for assessing predictive power for treatment response.
Meta-analysis across multiple independent cohorts to establish consistency.
These findings suggest that UPP1 expression is a clinically relevant biomarker with potential applications in prognostication, treatment selection, and monitoring therapeutic response across multiple cancer types.
Uridine Phosphorylase 1 (UPP1) is a critical enzyme involved in the pyrimidine salvage pathway, which is essential for maintaining uridine homeostasis in the body. This enzyme catalyzes the reversible phosphorolysis of uridine to uracil and ribose-1-phosphate . The human recombinant form of UPP1 has been extensively studied due to its significant role in various biochemical processes and its potential therapeutic applications.
UPP1 belongs to the family of pentosyltransferases and is expressed in the cytosol of most tissues . It is particularly abundant in certain tumors, where it plays a crucial role in the activation of pyrimidine nucleoside analogues used in chemotherapy, such as 5-fluorouracil (5-FU) and its prodrug, capecitabine . The enzyme’s ability to catalyze the conversion of uridine to uracil and ribose-1-phosphate is vital for the salvage of pyrimidine nucleotides, which are necessary for DNA and RNA synthesis .
Recombinant human UPP1 is typically produced using Escherichia coli (E. coli) as the expression system. The gene encoding UPP1 is cloned into an appropriate expression vector, which is then introduced into E. coli cells. The recombinant protein is expressed with an N-terminal His-tag to facilitate purification. After expression, the cells are lysed, and the protein is purified using affinity chromatography techniques . The purified protein is then subjected to various assays to confirm its activity and purity.
The primary reaction catalyzed by UPP1 is the reversible phosphorolysis of uridine to uracil and ribose-1-phosphate . This reaction is crucial for the pyrimidine salvage pathway, allowing cells to recycle uridine and maintain nucleotide pools. Additionally, UPP1 plays a significant role in the activation of chemotherapeutic agents. For example, it converts 5-FU to 5-fluorouridine, which is further phosphorylated to its active form by uridine kinase .
The elevated expression of UPP1 in certain tumors has made it a target for cancer therapy. Inhibitors of UPP1, such as 5-benzylacyclouridine (BAU), have been developed to modulate the cytotoxic side effects of 5-FU and its derivatives . By inhibiting UPP1, these compounds can increase the therapeutic index of chemotherapeutic agents, selectively protecting normal tissues with lower UPP1 activity .