UBE2I Human His mediates SUMOylation, a three-step process:
SUMO Activation: E1 enzymes (SAE1/SAE2) activate SUMO via ATP hydrolysis.
SUMO Transfer: UBE2I binds SUMO via a thioester linkage (Cys93).
Substrate Conjugation: UBE2I transfers SUMO to lysine residues in target proteins, often guided by E3 ligases or direct recognition of ΨKxD/E motifs .
HCC: High UBE2I expression correlates with poor prognosis, enhanced migration/invasion, and reduced autophagy markers (LC3A/B, Beclin-1) .
CCA: UBE2I silencing induces p27kip1 nuclear accumulation, arresting cell cycle progression and sensitizing cells to chemotherapy .
Nuclear Speckles: UBE2I localizes to nuclear speckles in mouse oocytes, suggesting roles in RNA processing and transcriptional regulation .
SUMOylation Dynamics: Transcriptional quiescence increases UBE2I-containing bodies, while overexpression disrupts speckle morphology .
UBE2I, also known as UBC9, is a small ubiquitin-like modifier (SUMO) E2 enzyme that plays a critical role in the SUMOylation pathway. Unlike typical E2 enzymes that function in ubiquitination, UBE2I specifically facilitates the conjugation of SUMO to target proteins. UBE2I shares the conserved core ubiquitin conjugating (UBC) domain of approximately 150 amino acid residues found in all E2 proteins, which is critical for its enzymatic function .
Methodologically, UBE2I function can be studied through:
In vitro SUMOylation assays using purified components (similar to ubiquitylation assays described in )
Assessment of thioester bond formation between UBE2I and SUMO
Structure-function analysis comparing UBE2I with other E2 enzymes
E2 Enzyme | Primary Modifier | Key Function | Core Domain Size |
---|---|---|---|
UBE2I | SUMO | SUMOylation | ~150 aa |
UBE2D | Ubiquitin | Ubiquitination | ~150 aa |
UBE2L3 | Ubiquitin | Ubiquitination | ~150 aa |
Production of His-tagged UBE2I for biochemical studies requires a systematic approach:
Cloning strategy:
Expression protocol:
Transform the construct into E. coli BL21(DE3) or Rosetta2(DE3) cells
Grow cultures at 37°C until OD600 reaches 0.6-0.8
Induce protein expression with IPTG (0.5-1mM)
Continue growth at lower temperature (16-20°C) overnight
Purification procedure:
This approach has been successfully used for other E2 enzymes and can be applied to UBE2I .
UBE2I expression shows significant alterations in human cancers, particularly in hepatocellular carcinoma (HCC):
Expression profile:
Methodological approach for expression analysis:
Database mining using GEPIA (http://gepia2.cancer-pku.cn/#index) and Kaplan-Meier plotter (https://kmplot.com/analysis/)[2]
Stratified analysis by gender, race, and clinical parameters
Construction of gene-gene interaction networks using geneMANIA plugin of Cytoscape
Co-expression analysis using Cbioportal (https://www.cbioportal.org/)[2]
Functional significance:
These findings suggest UBE2I could serve as both a prognostic marker and potential therapeutic target in HCC.
Studying UBE2I loading with SUMO requires careful experimental design similar to the E2 loading assays described for ubiquitin E2s:
Basic loading assay protocol:
Prepare reaction mixture containing E1 enzyme (1μg), UBE2I (1μg), and His-tagged SUMO (5μg)
Use buffer containing 10mM HEPES pH7.5, 100mM NaCl, 40μM ATP, and 2mM MgCl2
Incubate for 10 minutes at 30°C
Stop reaction with non-reducing SDS-PAGE sample buffer
Analyze by 4-20% gradient SDS-PAGE and Western blotting using anti-His antibodies
Detection methods:
Western blotting to visualize thioester-linked UBE2I~SUMO intermediate
Mass spectrometry for detailed characterization
Fluorescence-based assays for real-time kinetics
Control experiments:
Catalytic cysteine mutant of UBE2I (negative control)
Omission of ATP (negative control)
Comparison with other E2 enzymes for specificity
This methodology parallels the ubiquitin E2 loading assays described in search result , adapted for SUMO-specific components.
Designing effective UBE2I knockdown studies requires systematic optimization:
siRNA approach (as used in HCC studies) :
Design multiple siRNA sequences targeting different UBE2I regions
Include scrambled siRNA controls
Optimize transfection conditions for each cell line
Confirm knockdown efficiency by Western blot and qRT-PCR
Experimental design considerations:
Determine optimal timepoint for analysis (24-72h post-transfection)
Include appropriate functional assays:
Migration assays (wound healing, transwell)
Invasion assays (Matrigel-coated transwell)
Proliferation assays (MTT, colony formation)
Analyze pathway effects through Western blot of key markers
Results analysis:
This approach has successfully demonstrated that UBE2I silencing leads to decreased cell migration, invasion and proliferation in HCC cells .
Understanding UBE2I's structural determinants of specificity requires comprehensive structure-function analysis:
Key structural features to analyze:
Methodological approaches:
Comparative structural analysis:
Superimpose UBE2I structure with other E2 structures
Analyze structural differences around the active site
Compare electrostatic surface potentials
Identify regions that correlate with differential activity
This approach parallels the systematic structure-function analysis described for other E2 enzymes , adapted specifically for UBE2I's role in SUMOylation.
The mechanisms by which UBE2I promotes cancer cell migration and invasion can be elucidated through comprehensive pathway analysis:
RNA-sequencing approach:
Pathway analysis:
Validation experiments:
Confirm expression changes of key genes by qRT-PCR
Validate protein-level changes by Western blot
Perform rescue experiments to establish causality
This systematic approach has revealed potential pathways through which UBE2I influences cancer progression in HCC models .
Mass spectrometry (MS) provides powerful tools for characterizing UBE2I-mediated SUMOylation products:
Sample preparation strategy:
MS analysis approach:
Digest samples with trypsin or other suitable proteases
For SUMO remnant identification, use specific antibodies to enrich modified peptides
Perform LC-MS/MS analysis using high-resolution instruments
Apply database searching to identify modified proteins and sites
Data interpretation:
Identify SUMOylation sites based on characteristic mass shifts
Quantify relative SUMOylation levels
Compare modification patterns between experimental conditions
Integrate with protein-protein interaction data
This methodology adapts approaches used for characterizing ubiquitylation products of E2-HECT E3 pairs to the analysis of UBE2I-mediated SUMOylation.
UBE2I's relationship with autophagy pathways in cancer can be systematically investigated:
Experimental approach:
Mechanistic investigation:
Examine key autophagy regulators (Beclin-1, ATG proteins) after UBE2I manipulation
Investigate mTOR pathway components and their activation status
Analyze RNA-seq data for autophagy-related gene expression changes
Use reporter assays to monitor autophagy activity
Functional relevance:
Determine whether autophagy modulation mediates UBE2I's effects on:
Cell migration and invasion
Proliferation and survival
Response to therapeutic agents
Perform rescue experiments by co-modulating UBE2I and autophagy
Western blot analysis in HCC studies has established a connection between downregulated UBE2I expression and autophagy pathways, suggesting this may be a mechanism by which UBE2I influences cancer progression .
Post-translational modifications (PTMs) of UBE2I constitute an important regulatory layer:
Key PTMs affecting UBE2I:
Phosphorylation at specific residues
Acetylation
Self-SUMOylation
Ubiquitination affecting stability
Methodological approach to study PTMs:
Mass spectrometry to identify modification sites
Site-directed mutagenesis to create non-modifiable mutants
Phospho-specific or acetylation-specific antibodies
In vitro enzymatic assays comparing modified vs unmodified UBE2I
Functional assessment:
Compare activity of wild-type vs. mutant UBE2I in SUMOylation assays
Analyze subcellular localization using confocal microscopy
Examine interactions with E3 ligases and substrates
Assess stability and turnover rates
This approach parallels methods used to study regulatory mechanisms of other E2 enzymes in the ubiquitin system .
A comprehensive bioinformatics approach to UBE2I research utilizes multiple resources:
Expression and clinical correlation databases:
HCCDB: Database specific for hepatocellular carcinoma data
TIMER: Tool for immune infiltrate analysis across cancers
Kaplan-Meier plotter (https://kmplot.com/analysis/): Survival analysis tool
GEPIA (http://gepia2.cancer-pku.cn/#index): Gene expression profiling and analysis
Cbioportal (https://www.cbioportal.org/): For identifying co-expressed genes
Network analysis tools:
Pathway and enrichment analysis:
Structural analysis tools:
This integrated approach has been successfully applied in UBE2I research in HCC and structure-function studies of E2 enzymes .
Identifying novel UBE2I substrates requires a multi-faceted experimental approach:
Proteomics-based identification:
SUMO-remnant immunoaffinity profiling
Stable isotope labeling (SILAC) comparing control vs. UBE2I-manipulated cells
Purification of SUMOylated proteins using His-tagged SUMO pulldown
Mass spectrometry analysis to identify modified proteins
Bioinformatics prediction:
Use SUMOylation site prediction algorithms
Analyze gene expression data for co-regulation with UBE2I
Integrate with protein-protein interaction networks
Validation strategy:
In vitro SUMOylation assays with candidate substrates
Site-directed mutagenesis of predicted SUMOylation sites
Cell-based validation using co-immunoprecipitation
Functional studies to determine consequences of substrate SUMOylation
Cancer-specific considerations:
Compare substrate profiles across different cancer types
Correlate substrate SUMOylation with cancer progression markers
Analyze clinical relevance of identified substrates
This methodology builds on approaches used for E2-substrate identification in the ubiquitin system , adapted for UBE2I-specific SUMOylation research.
Proper interpretation of RNA-sequencing data after UBE2I manipulation requires careful analysis:
Experimental design considerations:
Data analysis pipeline:
Pathway analysis approach:
Validation and integration:
Confirm key findings by qRT-PCR
Validate protein-level changes by Western blot
Integrate with functional data
Connect transcriptional changes to phenotypic effects
This approach has been successfully applied in UBE2I research in HCC, revealing complex transcriptional networks influenced by UBE2I expression .
Ubiquitin-Conjugating Enzyme E2I, also known as UBC9, is a crucial enzyme in the ubiquitin-proteasome system, which is responsible for protein degradation and regulation within cells. The human recombinant version of this enzyme, tagged with a His (histidine) tag, is widely used in research to study its function and interactions.
The UBE2I gene encodes the UBC9 protein, which belongs to the ubiquitin-conjugating enzyme family. The human recombinant UBC9 is produced in E. coli and typically includes a His tag for purification purposes. This recombinant protein has a molecular weight of approximately 19.5 kDa and consists of 171 amino acids .
UBC9 plays a pivotal role in the second step of the ubiquitination process. In this process, ubiquitin, a small regulatory protein, is first activated by an E1 enzyme. The activated ubiquitin is then transferred to the active site cysteine residue of the E2 enzyme, UBC9. UBC9 subsequently interacts with an E3 ligase, which facilitates the transfer of ubiquitin to target proteins .
Apart from its role in ubiquitination, UBC9 is also involved in SUMOylation, a process similar to ubiquitination but involving Small Ubiquitin-like Modifier (SUMO) proteins. UBC9 accepts SUMO proteins from the E1 complex and catalyzes their attachment to target proteins. This modification can alter the function, localization, or stability of the target proteins .
The human recombinant UBC9 with a His tag is extensively used in biochemical and structural studies. The His tag allows for easy purification of the protein using affinity chromatography. Researchers use this recombinant protein to investigate the mechanisms of ubiquitination and SUMOylation, as well as to identify potential therapeutic targets for diseases related to protein misregulation .