UBE2D1 Human, encoded by the UBE2D1 gene, is a ubiquitin-conjugating enzyme (E2) critical for protein ubiquitination—a post-translational modification regulating protein degradation, localization, and activity . As a member of the E2 family, it facilitates the transfer of ubiquitin from E1-activating enzymes to substrate proteins via E3 ligases . Its role spans DNA repair, angiogenesis, and immune responses, making it a pivotal regulator of cellular homeostasis .
Sequence Identity:
Active Site: Contains a conserved catalytic cysteine residue essential for ubiquitin transfer .
Property | Value/Description |
---|---|
Subcellular Localization | Cytosol and plasma membrane |
Key Interactions | BARD1, BRCA1, UBE3A, E6-AP, UHRF1 |
Ubiquitin Linkage | Primarily K48-linked chains |
UBE2D1 modulates vascular endothelial growth factor receptor 2 (VEGFR2) ubiquitination, controlling its plasma membrane levels and downstream signaling . Key findings:
Depletion Effects: Knockdown of UBE2D1 increases VEGFR2 levels, enhancing VEGF-A-stimulated MAPK, PLCγ1, and Akt pathways .
Proteofection Assay: Recombinant UBE2D1 introduction reduces VEGFR2 levels by ~50%, confirming its role in receptor downregulation .
Impact on Tubulogenesis: UBE2D1 deficiency promotes endothelial tube formation, linking it to angiogenic processes .
BRCA1-BARD1 Interaction: UBE2D1 collaborates with BRCA1 and BARD1 in DNA damage response pathways .
UHRF1-Mediated DNA Methylation: Binds the ubiquitin-like domain of UHRF1, enabling H3 ubiquitination and DNMT1 recruitment for DNA methylation maintenance .
p53 and HIF-1α Ubiquitination: Partners with E3 ligases like E6-AP to target tumor suppressors for proteasomal degradation .
Immune Signaling: Required for IRF3 activation in antiviral responses and TNFα-mediated pathways .
Property | UBE2D1 | UBE2D2 | UBE2D3 |
---|---|---|---|
Sequence Identity | – | 89% | 88% |
Key Targets | VEGFR2, p53, HIF-1α | VEGFR2, c-Fos | Rheb, FBW7 |
E3 Ligase Partners | UHRF1, E6-AP | UHRF1, TRAF2 | Cullin3, TRIP12 |
Functional Impact | Angiogenesis, DNA repair | Angiogenesis, immune signaling | mTORC1 activation, proteasomal degradation |
Tumor Suppression: UBE2D1-mediated ubiquitination of p53 balances its degradation and functionality in cancer .
Immune Modulation: Interacts with OTUB1 to regulate K48-linked polyubiquitination in T-cell responses and TNF signaling .
Ubiquitin-conjugating enzyme E2 D1, Stimulator of Fe transport, SFT, UBC4/5 homolog, UbcH5, Ubiquitin carrier protein D1, Ubiquitin-conjugating enzyme E2(17)KB 1, Ubiquitin-conjugating enzyme E2-17 kDa 1, Ubiquitin-protein ligase D1, UBE2D1, SFT, UBC5A, UBCH5A, E2(17)KB1.
MGSSHHHHHH SSGLVPRGSH MGSMALKRIQ KELSDLQRDP PAHCSAGPVG DDLFHWQATI MGPPDSAYQG GVFFLTVHFP TDYPFKPPKI AFTTKIYHPN INSNGSICLD ILRSQWSPAL TVSKVLLSIC SLLCDPNPDD PLVPDIAQIY KSDKEKYNRH AREWTQKYAM.
UBE2D1 is a ubiquitin-conjugating enzyme (E2) encoded by the UBE2D1 gene in humans. It functions as a critical component in the protein ubiquitination process, which is an important cellular mechanism for targeting abnormal or short-lived proteins for degradation. The ubiquitination process involves three classes of enzymes working in cascade:
Ubiquitin-activating enzymes (E1s) - activate ubiquitin in an ATP-dependent manner
Ubiquitin-conjugating enzymes (E2s, including UBE2D1) - transfer activated ubiquitin
Ubiquitin-protein ligases (E3s) - recognize specific protein substrates
UBE2D1 functions by interacting with both E1 ubiquitin-activating enzymes and E3 ubiquitin-protein ligases to facilitate the transfer of ubiquitin to target proteins . This enzyme has been shown to participate in the ubiquitination of several important regulatory proteins, including the tumor suppressor p53 and the hypoxia-inducible transcription factor HIF1alpha .
Based on subcellular fractionation experiments, UBE2D1 is predominantly localized in the cytoplasm of human cells. This has been confirmed through blotting analysis of proteins from nuclear and cytoplasmic fractions . This cytoplasmic localization is consistent with its role in the ubiquitin-proteasome system, where many ubiquitination reactions and subsequent protein degradation events occur in the cytosolic compartment.
Interestingly, transcript analysis revealed that UBE2D1 upregulation is specific to cancer cells and not observed in hepatitis cells without HCC, suggesting that UBE2D1 could potentially serve as an early biomarker for HCC development . The dysregulation of UBE2D1 in cancer can occur through several mechanisms:
Genomic copy number gain - a frequent alteration in HCC
Inflammation-mediated regulation - high serum IL-6 levels are associated with UBE2D1 upregulation
DNA damage response pathways - continuous IL-6 exposure can lead to genomic instability and alterations in UBE2D1 expression
Multiple lines of evidence demonstrate a strong association between UBE2D1 and hepatocellular carcinoma (HCC) progression:
UBE2D1 plays a critical role in regulating p53 protein levels through ubiquitin-dependent degradation, which directly impacts several p53-dependent cellular processes:
Cell proliferation: UBE2D1 overexpression promotes cell proliferation in p53 wild-type cell lines by reducing p53 levels. This effect was not observed in p53 mutant or p53-deficient cell lines, confirming the p53-dependent mechanism .
Apoptosis regulation: UBE2D1 represses cell apoptosis in p53 wild-type cells but not in p53 mutated or deficient cells. This suggests that UBE2D1 affects apoptotic pathways primarily through its regulation of p53 .
Tumor growth: In vivo studies showed that UBE2D1 overexpression accelerates tumor growth, while UBE2D1 knockdown inhibits tumor growth in xenograft models, likely through modulation of p53-mediated tumor suppression .
The specificity of UBE2D1's effect on p53 wild-type cells but not p53 mutant or deficient cells provides strong evidence for a direct mechanistic link between UBE2D1-mediated ubiquitination and p53-dependent cellular processes .
The relationship between IL-6, UBE2D1, and cancer development represents a novel mechanistic pathway in carcinogenesis, particularly in hepatocellular carcinoma:
Clinical correlation: High serum IL-6 levels in HCC patients are associated with genomic gain of UBE2D1 .
Genomic instability: Continuous IL-6 exposure activates DNA damage response and promotes genomic instability by repressing the DNA damage checkpoint protein RAD51B .
Mechanistic pathway: This forms an IL-6/RAD51B/UBE2D1 axis where continuous IL-6 exposure leads to genomic alterations, including copy number gain of UBE2D1 .
Functional consequence: The resultant UBE2D1 overexpression enhances degradation of p53, promoting cancer cell proliferation and survival .
Synergistic effect: Continuous IL-6 can significantly facilitate HCC growth, especially in cells with genomic gain of UBE2D1, suggesting a cooperative interaction between inflammatory signaling and genetic alterations .
This relationship highlights how chronic inflammation (represented by continuous IL-6 exposure) can drive genetic alterations that promote cancer development, offering insights into how inflammation-associated cancers may evolve.
Several complementary techniques have proven effective for detecting UBE2D1 expression in tissue samples:
Immunohistochemistry (IHC):
Protocol: 5 μm paraffin sections are prepared from tissue samples. UBE2D1 antibody (Abcam) is applied at 1:100 dilution. Vectastain Elite ABC kits and ImmPACT DAB Substrate are used for detection .
Scoring: Expression is evaluated on a four-level scale: 0 (-), 1(+), 2(++), 3(+++) .
Quantification: Quantitative analyses of immunohistochemistry images can be performed using Image Software Pro Plus 6.0 .
Western Blotting:
Real-time PCR:
The combined use of these techniques provides comprehensive information about UBE2D1 expression at protein and transcript levels, as well as its cellular distribution.
Researchers can employ several approaches to modulate UBE2D1 expression in experimental models:
Lentiviral-based overexpression:
siRNA-mediated knockdown:
CRISPR/Cas9 gene editing:
Can be used for creating complete knockout models
Allows for precise genomic modifications
Useful for studying long-term effects of UBE2D1 loss
Inhibitor-based approaches:
These approaches can be combined with appropriate functional assays (cell proliferation, apoptosis, colony formation, in vivo tumor growth) to elucidate the biological roles of UBE2D1 in various contexts.
Several biochemical and cellular assays can be used to measure UBE2D1 enzyme activity:
Ubiquitination assays:
E3 ligase autoubiquitination assays:
Substrate-specific ubiquitination assays:
Ubiquitin discharge assays:
Binding assays:
The UBE2D family includes several members (UBE2D1, UBE2D2, UBE2D3, and UBE2D4) that share high sequence similarity but may have distinct functional roles:
Substrate specificity: Despite structural similarities, UBE2D family members may interact with different E3 ligases or recognize different substrates with varying affinities. Research approaches to investigate this include:
Comparative binding assays with different E3 ligases
Substrate profiling using proteomics approaches
Selective knockdown of individual UBE2D family members followed by ubiquitome analysis
Tissue-specific expression: UBE2D family members may show different expression patterns across tissues, suggesting specialized roles. This can be explored through:
Analysis of public RNA-seq databases for tissue-specific expression
Immunohistochemistry studies comparing expression patterns
Cell type-specific functional studies
Regulatory mechanisms: Different UBE2D family members may be regulated through distinct mechanisms. Investigation approaches include:
Promoter analysis for transcription factor binding sites
Post-translational modification profiling
Protein stability and turnover studies
Response to cellular stress: UBE2D family members may respond differently to various cellular stresses such as DNA damage, oxidative stress, or hypoxia. This can be studied through:
Stress-induction experiments followed by expression analysis
Functional studies in stress response pathways
Analysis of stress-induced post-translational modifications
Understanding these functional divergences is crucial for developing specific targeting strategies and interpreting the biological significance of alterations in individual UBE2D family members.
Developing selective inhibitors of UBE2D1 represents an important research direction, particularly given its role in cancer. Several strategies can be employed:
Structure-based design: Using crystal structures of UBE2D1 to identify unique binding pockets or interaction surfaces that differ from other UBE2D family members. The approach includes:
In silico screening of compound libraries against specific UBE2D1 pockets
Structure-activity relationship studies to optimize lead compounds
Fragment-based drug discovery approaches
Linked-domain inhibitor approach: This strategy has shown promise in research settings:
Multi-mechanistic inhibition: Targeting UBE2D1 through multiple mechanisms simultaneously:
Exploiting binding stoichiometry: Understanding the binding stoichiometry between UBE2D1 and inhibitors is crucial:
Allosteric inhibition: Identifying allosteric sites that are unique to UBE2D1 and developing compounds that induce conformational changes to inhibit its activity.
The development of UBE2D1-specific inhibitors could provide valuable tools for basic research and potentially therapeutic applications in diseases where UBE2D1 is dysregulated.
Post-translational modifications (PTMs) likely play important roles in regulating UBE2D1 function, though this area requires further investigation. Research approaches and considerations include:
Identification of PTMs:
Mass spectrometry-based proteomics to identify phosphorylation, ubiquitination, SUMOylation, or other modifications
Site-specific antibodies to detect known modifications
Bioinformatic prediction of potential modification sites
Functional consequences of modifications:
Site-directed mutagenesis of modified residues to mimic or prevent modification
In vitro activity assays comparing modified and unmodified forms
Cellular studies examining how modifications affect substrate targeting
Modification dynamics in response to stimuli:
Investigation of how cellular stresses (oxidative stress, DNA damage, hypoxia) affect UBE2D1 modifications
Temporal analysis of modification patterns during cell cycle progression
Changes in modifications in response to growth factors or cytokines
Cross-talk between different modifications:
How one modification might influence the occurrence or effects of others
Hierarchical relationships between modifications
Competitive modifications at the same or adjacent sites
Potential therapeutic implications:
Targeting enzymes responsible for UBE2D1 modifications
Developing compounds that stabilize or disrupt modification-dependent interactions
Using modification patterns as biomarkers for disease states
Understanding the complex regulation of UBE2D1 through post-translational modifications could reveal new opportunities for therapeutic intervention and explain context-dependent functions of this enzyme.
UBE2D1 shows potential as a biomarker in cancer diagnostics, particularly for hepatocellular carcinoma (HCC). Implementation strategies include:
Standardization of detection methods and large-scale validation studies would be necessary before clinical implementation of UBE2D1 as a cancer biomarker.
Several therapeutic approaches could potentially target the UBE2D1 pathway in cancer:
Direct UBE2D1 inhibition:
Linked-domain inhibitors like UBOX-UBL show promise in research settings with high affinity (400nM ± 170nM)
Advanced inhibitors such as UBOX-UbvD1short demonstrate robust inhibition at concentrations as low as 20nM
Further optimization of these research tools could lead to clinically viable inhibitors
Disruption of UBE2D1-E3 ligase interactions:
Small molecules designed to interfere with specific UBE2D1-E3 interactions
Peptide-based inhibitors mimicking key interaction interfaces
Allosteric modulators affecting conformation of interaction surfaces
Targeting upstream regulators:
p53 stabilization strategies:
Since a major oncogenic effect of UBE2D1 is p53 degradation, approaches to stabilize p53 could counteract UBE2D1 effects
Combination with existing MDM2 inhibitors, which also target p53 degradation
Development of dual-specificity compounds affecting multiple p53 degradation pathways
Exploiting synthetic lethality:
Identifying cellular contexts where UBE2D1 inhibition would be selectively lethal to cancer cells
Combination strategies with DNA damaging agents or other stress inducers
Targeting parallel survival pathways in UBE2D1-overexpressing tumors
The development of these approaches would require further understanding of UBE2D1's role in different cancer types and careful assessment of potential toxicities, as ubiquitination pathways are fundamental to cellular homeostasis.
While this area needs further investigation, several mechanisms may link UBE2D1 dysregulation to therapeutic resistance in cancer:
p53-mediated resistance:
Stress response modulation:
UBE2D1 may regulate other stress response proteins beyond p53
Altered stress response pathways could enable cancer cells to survive therapeutic stress
Understanding these broader targets requires comprehensive substrate identification studies
Genomic instability contribution:
Inflammatory microenvironment effects:
Potential impact on drug metabolism:
Ubiquitination pathways can affect the stability and function of drug transporters and metabolizing enzymes
Altered UBE2D1 activity might influence intracellular drug concentrations
This could affect a broad spectrum of therapeutics independent of direct resistance mechanisms
Research investigating correlations between UBE2D1 expression and treatment outcomes in cancer patients would help clarify its role in therapeutic resistance and guide the development of strategies to overcome such resistance.
Identifying the substrate spectrum of UBE2D1 is crucial for understanding its cellular functions. Several bioinformatic approaches can facilitate this:
Proteomic data analysis:
Analysis of global ubiquitinome data from mass spectrometry studies
Comparative ubiquitinome analysis between UBE2D1-depleted and control cells
Identification of ubiquitination sites enriched in UBE2D1-expressing cells
Protein-protein interaction networks:
Integration of experimental protein-protein interaction data
Network analysis to identify proteins that interact with both UBE2D1 and known E3 partners
Pathway enrichment analysis of potential substrate networks
Structural modeling and docking:
In silico docking of potential substrates to UBE2D1-E3 complexes
Molecular dynamics simulations to assess binding stability
Identification of common structural motifs in known substrates
Machine learning approaches:
Development of predictive models based on features of known UBE2D1 substrates
Training algorithms to recognize ubiquitination sites preferred by UBE2D1
Integration of multiple data types (structure, sequence, expression) for improved prediction
Evolutionary conservation analysis:
Identification of conserved ubiquitination sites across species
Correlation with UBE2D1 conservation patterns
Enrichment analysis for conserved sites in specific protein families
These computational approaches should be integrated with experimental validation to confirm predicted substrates and understand their biological significance in the context of UBE2D1 function.
When facing contradictory data regarding UBE2D1 function, researchers should employ a systematic approach to resolution:
Context-dependent analysis:
Carefully evaluate experimental conditions across contradictory studies
Consider cell type-specific factors that might influence UBE2D1 function
Analyze expression levels of UBE2D1 partners (E3 ligases, substrates) across different experimental systems
Technical validation:
Reproduce key experiments using multiple methodologies
Validate antibody specificity and tool compounds thoroughly
Employ genetic approaches (CRISPR/Cas9) alongside RNAi or inhibitor-based methods
Substrate-specific evaluation:
Determine whether contradictions are general or substrate-specific
Perform detailed kinetic analyses of UBE2D1 activity toward different substrates
Consider the influence of post-translational modifications on substrate recognition
Meta-analysis approaches:
Systematic review of available literature with quality assessment
Quantitative synthesis of results across multiple studies
Identification of moderator variables that might explain discrepancies
Collaborative resolution:
Direct collaboration between labs reporting contradictory results
Exchange of materials and protocols to identify sources of variation
Standardization of key assays to improve reproducibility
By employing these systematic approaches, researchers can work toward resolving contradictions and developing a more comprehensive understanding of UBE2D1 function in different biological contexts.
Analyzing UBE2D1 expression across cancer datasets requires robust statistical approaches:
Differential expression analysis:
Parametric methods: t-tests for pairwise comparisons, ANOVA for multi-group comparisons
Non-parametric alternatives: Wilcoxon rank-sum test, Kruskal-Wallis test
Adjustment for multiple testing using methods like Benjamini-Hochberg procedure to control false discovery rate
Survival analysis:
Correlation analyses:
Pearson correlation for linear relationships between UBE2D1 and other continuous variables
Spearman correlation for non-parametric assessment of associations
Partial correlation to account for confounding variables
Integration of multi-omics data:
Principal component analysis (PCA) or t-SNE for dimensionality reduction
Unsupervised clustering methods to identify patient subgroups
Regularized regression methods (LASSO, Ridge) for building predictive models
Batch effect correction:
ComBat or similar methods to adjust for batch effects when integrating data from different sources
Quantile normalization to make expression distributions comparable across datasets
Proper data scaling and transformation to meet statistical assumptions
Validation approaches:
Cross-validation for assessing model performance
Independent validation cohorts to confirm findings
Bootstrapping to estimate confidence intervals
Several emerging technologies hold promise for advancing UBE2D1 research:
CRISPR-based technologies:
Base editing and prime editing for introducing precise mutations in UBE2D1
CRISPRi/CRISPRa for reversible modulation of UBE2D1 expression
CRISPR screens to identify synthetic lethal interactions with UBE2D1
Advanced proteomics approaches:
Proximity labeling methods (BioID, APEX) to identify local UBE2D1 interactors
Degradomics to comprehensively identify substrates affected by UBE2D1
Targeted mass spectrometry for quantifying low-abundance ubiquitinated species
Single-cell technologies:
Single-cell RNA-seq to evaluate UBE2D1 expression heterogeneity
Single-cell proteomics to analyze protein-level variations
Spatial transcriptomics to map UBE2D1 expression in complex tissues
Live-cell imaging techniques:
FRET-based sensors for monitoring UBE2D1 activity in real-time
Optogenetic approaches to spatiotemporally control UBE2D1 function
Super-resolution microscopy to visualize UBE2D1-mediated ubiquitination events
Structural biology advances:
Cryo-EM for capturing dynamic UBE2D1-E3-substrate complexes
Hydrogen-deuterium exchange mass spectrometry for probing conformational changes
Integrative structural biology combining multiple data types
Organoid and patient-derived models:
Patient-derived organoids to study UBE2D1 in disease-relevant contexts
Humanized mouse models for evaluating UBE2D1 inhibitors
Organ-on-chip systems for studying UBE2D1 in complex tissue environments
Integration of these technologies could provide unprecedented insights into UBE2D1 function and facilitate the development of targeted therapeutic approaches.
While UBE2D1 has been extensively studied in cancer, several other potential roles deserve investigation:
Neurodegenerative disorders:
Given the importance of ubiquitination in protein quality control, UBE2D1 may influence the accumulation of misfolded proteins in neurodegenerative diseases
Research could explore UBE2D1's role in degrading disease-associated proteins like tau, alpha-synuclein, or huntingtin
Mouse models with conditional UBE2D1 deletion in neurons could reveal neurological phenotypes
Inflammatory diseases:
Iron metabolism disorders:
Developmental processes:
Analysis of UBE2D1 expression patterns during embryonic development
Investigation of its role in stem cell maintenance and differentiation
Potential involvement in developmental signaling pathways like Wnt or Notch
Cellular stress responses:
Exploration of UBE2D1's role in oxidative stress, hypoxia, and DNA damage responses beyond p53 and HIF1alpha
Investigation of UBE2D1 in cellular adaptation to proteotoxic stress
Potential involvement in autophagy regulation
These unexplored areas could reveal new biological functions of UBE2D1 and potentially identify additional therapeutic opportunities beyond cancer.
Understanding UBE2D1 regulation could significantly contribute to precision medicine approaches:
Biomarker-guided therapies:
UBE2D1 expression or genomic alterations could serve as biomarkers for patient stratification
High UBE2D1 expression correlates with poorer survival in HCC, suggesting potential as a prognostic indicator
Analysis of UBE2D1 status could guide treatment selection, particularly for therapies targeting the p53 pathway
Personalized inhibitor strategies:
Development of UBE2D1 inhibitors with varying mechanisms of action to address different clinical contexts
Dose adjustment based on individual UBE2D1 expression levels
Combination strategies tailored to specific UBE2D1-dependent alterations
Genetic context considerations:
Monitoring disease progression:
Tracking changes in UBE2D1 expression or activity during treatment
Development of non-invasive methods to assess UBE2D1-dependent processes
Use as a marker for early detection of recurrence
Target population identification:
Integration of UBE2D1 information into comprehensive molecular profiling could enhance treatment planning and monitoring, ultimately improving patient outcomes through more precise therapeutic strategies.
Ubiquitin Conjugating Enzyme E2D1, also known as UBE2D1, is a crucial component of the ubiquitin-proteasome system (UPS), which is responsible for the targeted degradation of proteins within eukaryotic cells. This enzyme plays a pivotal role in the ubiquitination process, acting as an intermediary that transfers ubiquitin from the E1 activating enzyme to the E3 ligase, which then attaches ubiquitin to the substrate protein.
Ubiquitination is a post-translational modification that involves the attachment of ubiquitin, a small regulatory protein, to lysine residues on target proteins. This process typically involves three main steps:
UBE2D1 is involved in various cellular processes, including:
Recombinant UBE2D1 is produced using recombinant DNA technology, which involves cloning the UBE2D1 gene into an expression vector, introducing the vector into a host cell (such as E. coli), and inducing the expression of the UBE2D1 protein. The recombinant protein is then purified for use in research and therapeutic applications.
Recombinant UBE2D1 is widely used in biochemical and structural studies to understand the mechanisms of ubiquitination and its role in various cellular processes. It is also used in drug discovery and development, particularly in the design of inhibitors that target the ubiquitin-proteasome system for the treatment of diseases such as cancer and neurodegenerative disorders .