UBE2S (Ubiquitin-conjugating enzyme E2 S), also known as E2-EPF, is a 26 kDa protein belonging to the ubiquitin-conjugating enzyme (E2) family. It facilitates the transfer of ubiquitin from E1-activating enzymes to target proteins, forming Lys11-linked polyubiquitin chains critical for proteasomal degradation .
UBE2S collaborates with the anaphase-promoting complex/cyclosome (APC/C) to degrade mitotic regulators (e.g., securin, cyclin B). It elongates Lys11-linked chains initiated by UBE2C/UBCH10, ensuring proper mitotic exit .
Priming Phase: UBE2C/UBCH10 adds initial ubiquitin to APC/C substrates.
Elongation Phase: UBE2S extends chains, enhancing proteasomal recognition .
UBE2S is overexpressed in multiple cancers, driving tumor progression through:
Hypoxia Signaling: Degradation of VHL stabilizes HIF-1α, promoting angiogenesis .
DNA Damage Response: Modifies chromatin to regulate transcription silencing .
Chemoresistance: Activates PTEN-AKT pathways, reducing sensitivity to 5-FU/oxaliplatin in HCC .
Prognostic Value: High UBE2S expression correlates with poor survival in glioma (HR = 1.8, P < 0.01) and HCC (P < 0.001) .
Diagnostic Utility: Elevated levels in cancer tissues vs. nonmalignant controls .
UBE2S promotes epithelial-mesenchymal transition (EMT) in cervical cancer by degrading epithelial markers, enhancing metastasis .
UBE2S is an E2 ubiquitin-conjugating enzyme that catalyzes the formation of K11-linked polyubiquitin chains. Its primary function involves protein ubiquitination, a post-translational modification process that regulates protein degradation and cellular signaling pathways. UBE2S specifically promotes K11-linked polyubiquitination, which differs from the classical K48-linked chains that typically mark proteins for proteasomal degradation .
To study UBE2S function, researchers typically employ techniques such as RNA interference (shRNA or siRNA) to knock down UBE2S expression or overexpression systems using lentiviral vectors. For instance, the cDNA sequence of UBE2S can be inserted into pCDH lentivirus vectors and transfected into cells to study gain-of-function effects . Expression changes are then validated using quantitative real-time PCR and Western blot analysis.
UBE2S expression is significantly upregulated in multiple cancer types compared to normal tissues. In hepatocellular carcinoma (HCC), both mRNA and protein levels of UBE2S are higher in tumor tissues than in adjacent normal tissues . Similarly, in lung cancer, immunohistochemical assays and TCGA database analyses have demonstrated elevated UBE2S expression in cancer tissues compared to normal tissues .
To investigate UBE2S expression levels, researchers commonly employ:
Immunohistochemistry (IHC) on tissue microarrays
Western blot analysis of tissue or cell lysates
Quantitative real-time PCR for mRNA expression
Analysis of public databases such as TCGA
For instance, a study of lung adenocarcinoma used microarrays containing 30 tumor samples across different stages and 30 adjacent normal samples, which were subjected to immunohistochemistry staining of UBE2S .
UBE2S interacts with multiple signaling pathways that are critical for cancer development and progression:
Cell cycle regulation: UBE2S interacts with TRIM28 in the nucleus to enhance ubiquitination of p27, facilitating its degradation and promoting cell cycle progression .
Wnt/β-Catenin pathway: UBE2S modifies β-Catenin at K19 via K11-linked polyubiquitin chain, enhancing β-Catenin stability and promoting its cellular accumulation .
VHL/HIF-1α and VHL/JAK2/STAT3 pathways: UBE2S interacts directly with VHL to upregulate these pathways, enhancing malignant properties of HCC cells .
P53, Integrin, and ILK signaling: Microarray analysis of UBE2S knockdown cells revealed regulation of genes involved in these pathways .
To study these interactions, researchers use co-immunoprecipitation assays, pathway-specific reporter assays, and gene expression profiling following UBE2S manipulation.
UBE2S has been discovered to exist in a dimeric state that confers auto-inhibition by blocking a catalytically critical ubiquitin binding site. This dimerization is stimulated by the lysine-rich C-terminal extension of UBE2S, which is also required for the recruitment of this E2 to the APC/C (Anaphase-Promoting Complex/Cyclosome) and is auto-ubiquitinated as substrate abundance becomes limiting .
Researchers investigating UBE2S dimerization have employed:
Nuclear magnetic resonance (NMR) spectroscopy to detect concentration-dependent changes in molecular tumbling
Pull-down experiments using differentially tagged UBE2S (His6- and HA-tagged)
Crystallography to determine protein structure (PDB: 6S98 for wild-type UBC domain and PDB: 6S96 for C118A mutant)
The structural data revealed significant differences in crystal packing between wild-type and mutant UBE2S:
UBE2S UBC WT (PDB: 6S98) | UBE2S UBC C118A (PDB: 6S96) |
---|---|
Space group: P 1 2 1 1 | Space group: P 6 1 |
Unit cell parameters (Å): 44.8 49.05 71.93 | Unit cell parameters (Å): 120.9 120.9 45.3 |
Unit cell angles (°): 90 106.03 90 | Unit cell angles (°): 90 90 120 |
Dimerization-deficient UBE2S mutants showed more rapid turnover in cells and did not promote mitotic slippage during prolonged drug-induced mitotic arrest, suggesting that dimerization attenuates the auto-ubiquitination-induced turnover of UBE2S when the APC/C is not fully active .
Whole-exome sequencing of HCC tissues has identified novel somatic mutations in UBE2S, including p.Gly57Ala and p.Lys63Asn. Functional predictions indicated that these amino acid substitutions are potentially deleterious . While wild-type UBE2S promotes cancer progression, these mutations may further enhance its oncogenic properties.
To study these mutations, researchers can employ:
Site-directed mutagenesis to generate mutant UBE2S constructs
Functional assays comparing wild-type and mutant UBE2S effects on:
Cell proliferation (MTT, BrdU incorporation)
Invasion and migration (Transwell, wound healing)
Cell cycle progression (flow cytometry)
In vivo tumor growth (xenograft models)
Structural analyses to determine how mutations affect protein conformation and interaction capabilities
UBE2S plays a critical role in cell cycle progression, particularly at the G1/S phase transition. Mechanistically, UBE2S interacts with TRIM28 in the nucleus, and together they enhance the ubiquitination of p27 to facilitate its degradation and promote cell cycle progression .
Research methodologies to investigate this include:
Flow cytometry analysis of cell cycle distribution following UBE2S knockdown or overexpression
BrdU incorporation assays to measure DNA synthesis and cell proliferation
Western blot analysis of cell cycle regulators (p27, cyclins, CDKs)
Co-immunoprecipitation to detect UBE2S interactions with TRIM28 and other cell cycle proteins
Ubiquitination assays to measure p27 ubiquitination levels
Studies have demonstrated that UBE2S knockdown in HCC and lung cancer cells leads to G1 phase arrest, decreased BrdU incorporation, and increased p27 levels . Conversely, UBE2S overexpression promotes G1/S transition and enhances cell proliferation. These findings establish UBE2S as a key regulator of cell cycle progression in cancer cells.
Several complementary techniques can be employed to study UBE2S protein-protein interactions:
Co-immunoprecipitation (Co-IP): This is the primary method used to detect interactions between UBE2S and partners like TRIM28, VHL, and β-Catenin. Researchers typically use antibodies against UBE2S or its tagged version (HA, Flag, His) to pull down protein complexes, followed by immunoblotting for potential interacting partners .
Proximity ligation assay (PLA): This technique provides visualization of protein interactions in situ with high specificity and sensitivity.
Fluorescence resonance energy transfer (FRET) or bimolecular fluorescence complementation (BiFC): These techniques allow for real-time visualization of protein interactions in living cells.
Yeast two-hybrid screening: This can be used for unbiased identification of novel UBE2S-interacting proteins.
Mass spectrometry-based approaches: After immunoprecipitation, mass spectrometry can identify novel interacting partners in an unbiased manner.
For studying UBE2S dimerization specifically, researchers have used pull-down experiments with His6- and HA-tagged UBE2S, along with NMR analyses under reducing conditions to prevent disulfide-induced aggregation .
To modulate UBE2S expression for functional studies, researchers have employed several approaches:
RNA interference:
Overexpression systems:
CRISPR-Cas9 gene editing:
For complete knockout or introducing specific mutations.
Small molecule inhibitors:
Validation of expression modulation should be performed using:
Quantitative real-time PCR for mRNA levels
Western blot for protein levels
Functional assays to confirm phenotypic changes
Researchers have employed various experimental models to study UBE2S in cancer:
Cell line models:
HCC cell lines (HepG2, Hep3B, Huh7, SMMC-7721)
Lung cancer cell lines (A549, H1299)
Colorectal cancer cell lines
Patient-derived samples:
Tissue microarrays containing multiple patient samples
Fresh frozen tissues for protein and RNA extraction
Animal models:
3D culture systems:
Spheroid cultures and organoids can bridge the gap between 2D cell cultures and in vivo models.
For analyzing clinical relevance, researchers commonly use:
Immunohistochemistry on tissue microarrays
Analysis of public databases (TCGA, GEO)
Correlation of UBE2S expression with clinicopathological features and patient survival
High UBE2S expression has been consistently associated with poor clinical outcomes across multiple cancer types:
To establish these correlations, researchers typically:
Perform IHC on tissue microarrays containing samples from patients with comprehensive clinical data
Use Kaplan-Meier survival analysis to correlate expression with patient outcomes
Conduct multivariate Cox regression analysis to determine if UBE2S is an independent prognostic factor
Analyze public databases (TCGA) to validate findings in larger cohorts
These clinical correlations reinforce the biological significance of UBE2S in cancer progression and highlight its potential as a prognostic biomarker.
Several therapeutic approaches targeting UBE2S are being explored:
Small molecule inhibitors:
Combination therapies:
Gene therapy approaches:
Delivery of UBE2S-targeting shRNA or siRNA could potentially suppress tumor growth.
Targeting UBE2S-regulated pathways:
Inhibitors of downstream pathways activated by UBE2S, such as Wnt/β-Catenin, VHL/HIF-1α, or JAK2/STAT3 signaling, may be effective in UBE2S-overexpressing tumors.
For preclinical evaluation of these approaches, researchers should:
Assess efficacy in multiple cell line and animal models
Evaluate specificity using rescue experiments
Determine potential toxicities in normal cells
Investigate mechanisms of resistance
UBE2S has significant potential as a biomarker for cancer:
To develop UBE2S as a clinical biomarker, researchers should:
Validate expression patterns in large, multi-center cohorts
Develop standardized assays for UBE2S detection
Determine optimal cutoff values for high versus low expression
Evaluate UBE2S in combination with other biomarkers for improved accuracy
Assess UBE2S in liquid biopsies (circulating tumor cells, cell-free DNA) for non-invasive testing
UBE2S has been implicated in cellular differentiation processes, particularly in embryonic stem (ES) cell differentiation into mesoendoderm lineages. Research has shown that Ube2s-promoted β-Catenin accumulation partially released the dependence on exogenous molecules for the process of ES cell differentiation into mesoendoderm lineages .
Research approaches to investigate this role include:
In vitro differentiation assays with ES cells
Gene expression profiling during differentiation
Lineage tracing in development models
Knockout or knockdown studies in developmental systems
This emerging area of research suggests that UBE2S may have important functions beyond cancer, potentially playing roles in embryonic development and tissue homeostasis.
UBE2S specifically catalyzes the formation of K11-linked polyubiquitin chains, which differs from the classical K48-linked chains typically associated with protein degradation. UBE2S modifies β-Catenin at K19 via K11-linked polyubiquitin chain, resulting in enhanced stability rather than degradation .
To investigate ubiquitin chain topology and its effects:
Use ubiquitin mutants (with specific lysine residues mutated to arginine)
Employ mass spectrometry to identify ubiquitination sites and chain types
Perform in vitro ubiquitination assays with purified components
Use linkage-specific antibodies to detect specific ubiquitin chain types
Understanding how different ubiquitin chain topologies affect substrate fate could reveal new mechanisms of protein regulation and potentially identify novel therapeutic approaches targeting specific ubiquitination events.
Ubiquitin Conjugating Enzyme E2S (UBE2S) is a member of the ubiquitin-conjugating enzyme family, also known as E2 enzymes. These enzymes play a crucial role in the ubiquitination process, which is essential for protein degradation and regulation within the cell. UBE2S is particularly significant due to its involvement in various cellular processes, including cell cycle regulation, DNA repair, and signal transduction.
UBE2S is characterized by its ability to receive ubiquitin from a ubiquitin-activating enzyme (E1) and subsequently interact with a ubiquitin ligase (E3) to conjugate ubiquitin to substrate proteins . This process is vital for targeting proteins for degradation via the proteasome, thus maintaining protein homeostasis within the cell .
The ubiquitin-proteasome system (UPS) orchestrates the specific poly-ubiquitination of protein substrates in humans, which can lead to their degradation . UBE2S, as part of this system, ensures the proper turnover of proteins, preventing the accumulation of damaged or misfolded proteins that could potentially lead to cellular dysfunction and disease.
Recombinant human UBE2S is produced using recombinant DNA technology, which involves inserting the gene encoding UBE2S into a suitable expression system, such as bacteria or yeast. This allows for the large-scale production of UBE2S, which can be used for various research and therapeutic purposes. Recombinant UBE2S retains the functional properties of the native enzyme, making it a valuable tool for studying the ubiquitination process and its implications in health and disease .
UBE2S is widely used in research to understand its role in the ubiquitination process and its impact on cellular functions. Studies have shown that UBE2S is involved in maintaining proteostasis during aging, ensuring protein quality control, and preventing the accumulation of poly-ubiquitinated proteins . These findings highlight the importance of UBE2S in cellular homeostasis and its potential as a therapeutic target for age-related diseases and other conditions associated with protein misfolding and aggregation.