ELOB Human, encoded by the ELOB gene (NCBI Gene ID: 6923), is a 118-amino acid ubiquitin-like protein critical for transcriptional elongation and protein degradation. As a core subunit of the Elongin complex (SIII), it regulates RNA Polymerase II activity and serves as a regulatory component in Cullin-RING E3 ubiquitin ligase complexes (CRLs). ELOB’s dual roles in transcription and ubiquitination make it a pivotal target in molecular biology and disease research .
ELOB/ELOC binds to elongin A (ELOA), forming the Elongin complex, which:
Suppresses RNA Polymerase II Pausing: Stabilizes the polymerase at template-encoded arrest sites, particularly under low nucleotide conditions .
Structural Interaction: Cryo-EM data show ELOB/ELOC anchoring ELOA to RNA Polymerase II’s RPB2 domain, inducing conformational changes near the active site .
The ELOB/ELOC heterodimer interacts with BC-box motifs in substrate recognition proteins (e.g., VHL, SOCS-box proteins), linking them to Cullin-RING ligases:
Overexpression in Tumors: ELOB is upregulated in breast cancer, correlating with poor prognosis. It promotes proliferation via ubiquitination of p14/ARF, an oncoprotein .
Therapeutic Target: Knockdown of ELOB suppresses tumor growth by stabilizing p14/ARF, suggesting ELOB inhibitors may restore tumor suppressor activity .
ELOB/ELOC interacts with EPOP to recruit Polycomb repressors to PcG target genes, generating bivalent chromatin domains critical for stem cell pluripotency .
ELOB is utilized in:
Store at 4°C if entire vial will be used within 2-4 weeks. Store, frozen at -20°C for longer periods of time.
For long term storage it is recommended to add a carrier protein (0.1% HSA or BSA).
Avoid multiple freeze-thaw cycles.
ELOB is a 118 amino acid protein that functions as a regulatory subunit initially identified in RNA polymerase II elongation processes. It forms part of the Elongin (SIII) complex alongside ELOA (transcriptionally active component) and ELOC (another regulatory subunit) . This complex plays a critical role in regulating the elongation process of RNA polymerase II by preventing transient pausing of the polymerase at various sites within the transcriptional unit. Beyond transcriptional regulation, ELOB serves as a pivotal element in the ELOB/c-Cullin2/5-SOCS-box E3 ubiquitin-protein ligase complex, which catalyzes the ubiquitination and subsequent degradation of various target proteins .
ELOB functions as an integral component of several ubiquitin-proteasome systems (UPSs), most notably in Cullin-RING ligases (CRLs) such as CRL2 and CRL5. In the CRL2 complex specifically, ELOB interacts with RBX1, Cullin-2, and connexin ELOC, forming a structural scaffold that facilitates the binding of substrate recognition proteins . One well-studied example involves the Von Hippel-Lindau (VHL) protein, which acts as a recognition protein interacting with the CRL2 complex. To study these interactions experimentally, coimmunoprecipitation assays can be performed using ELOB antibodies (such as ab151743 from Abcam) followed by immunoblotting for suspected interaction partners .
ELOB demonstrates widespread expression across human tissues, suggesting its fundamental importance in cellular processes. According to data from the Human Protein Atlas, ELOB is expressed in neural tissues (including hippocampal formation, amygdala, basal ganglia, midbrain, spinal cord, cerebral cortex, cerebellum, hypothalamus), endocrine organs (thyroid, parathyroid, adrenal, and pituitary glands), digestive system (esophagus, stomach, intestines, liver, gallbladder, pancreas), reproductive organs, cardiovascular tissues, and the immune system . For researchers examining tissue-specific ELOB expression, immunohistochemistry staining protocols using anti-ELOB antibodies (such as sc-133090 from Santa Cruz) are recommended, with subsequent evaluation using the H-score semi-quantitative method (ranging from 0-300) .
Researchers can employ multiple complementary techniques to quantify ELOB expression:
RNA-seq analysis: Utilizing transcriptomic data from databases such as TCGA (The Cancer Genome Atlas) or GEO (Gene Expression Omnibus) for large-scale comparative studies. Differential expression analysis can be performed using the limma package in R, with filtering criteria of P < 0.05 and |log2 fold change| > 0.58 .
Western blot quantification: For protein-level validation, researchers should use validated antibodies against ELOB and appropriate loading controls. Cell lysates should be generated and analyzed via standardized immunoblotting protocols .
Immunohistochemistry: For tissue-specific expression patterns, IHC staining using tissue arrays with 5-micron thickness sections. After dehydration and peroxidase blocking, apply ELOB antibody and incubate for 30 minutes, followed by detection using appropriate systems such as DakoCytomation EnVision + System-HRP .
Single-cell RNA sequencing: For cellular heterogeneity analysis, scRNA-seq data can be analyzed using the "Seurat" package and t-distributed Stochastic Neighbor Embedding (tSNE) method for non-linear dimensionality reduction and visualization .
ELOB functions as a critical component of E3 ubiquitin ligase complexes, particularly within the CRL2 (Cullin2-RBX1-ELOB E3 ligase) complex. This complex facilitates the ubiquitination and subsequent proteasomal degradation of target proteins. The mechanistic process involves:
ELOB forms a complex with ELOC and Cullin-2/RBX1
This complex recruits substrate recognition proteins (like VHL)
Together, they coordinate the transfer of ubiquitin to target substrates
Polyubiquitinated substrates are then recognized by the 26S proteasome and degraded
To experimentally investigate this process, researchers should employ in vivo ubiquitination assays. For example, to assess endogenous p14/ARF ubiquitination (a known ELOB target), transfect cells with siRNA targeting ELOB, treat with proteasome inhibitor MG-132 for 2 hours, lyse cells in 1% SDS-containing buffer, boil lysates for 10 minutes, dilute in SDS-free buffer, perform immunoprecipitation with anti-p14/ARF antibody, and analyze by immunoblotting with anti-ubiquitin antibody .
ELOB was initially characterized for its role in transcriptional regulation as part of the Elongin (SIII) complex. This complex enhances RNA polymerase II activity by suppressing transient pausing during transcriptional elongation. The precise molecular mechanisms involve:
Association of the Elongin complex (ELOA, ELOB, ELOC) with RNA polymerase II
ELOA directly interacts with polymerase to stimulate elongation rates
ELOB and ELOC serve as regulatory subunits stabilizing the complex and potentially mediating additional protein interactions
This dual functionality - in both transcriptional regulation and protein degradation pathways - positions ELOB as a multifunctional adaptor protein at the intersection of gene expression and protein turnover. Researchers studying this function should use chromatin immunoprecipitation (ChIP) assays to identify genomic regions where ELOB-containing complexes associate with transcriptional machinery .
Multiple lines of evidence connect ELOB expression to breast cancer:
Expression analysis: Bioinformatic analysis of TCGA and FUSCC (Fudan University Shanghai Cancer Center) databases demonstrates significant ELOB overexpression in breast cancer tissues compared to normal tissues .
Prognostic correlation: Higher ELOB expression correlates with unfavorable prognosis in breast cancer patients, as validated through Kaplan-Meier survival analysis .
Pathway enrichment: GSEA and KEGG pathway analyses reveal that elevated ELOB expression associates with multiple cancer-promoting pathways, including cell cycle regulation, DNA replication, proteasome function, and PI3K-Akt signaling .
Functional validation: Both in vivo and in vitro experiments confirm that downregulation of ELOB significantly suppresses breast cancer cell proliferation .
Tissue microarray validation: Analysis of breast cancer tissue microarrays and western blot analysis of patient samples demonstrates elevated ELOB expression in tumor tissues compared to adjacent normal tissues .
For researchers investigating ELOB in cancer contexts, a combination of public database mining (TCGA, cBioPortal), tissue microarray analysis, and functional validation through genetic manipulation (siRNA knockdown, overexpression constructs) is recommended .
Researchers should employ a multi-tiered approach:
Genetic manipulation:
Protein-protein interaction studies:
Protein stability assessment:
Localization studies:
In vivo validation:
ELOB regulates p14/ARF through a precise molecular mechanism:
As a core component of the Cullin2-RBX1-ELOB E3 ligase (CRL2) complex, ELOB facilitates the recognition and ubiquitination of p14/ARF
This ubiquitination marks p14/ARF for proteasomal degradation
When ELOB is downregulated, p14/ARF degradation is impaired, leading to p14/ARF accumulation
The anticancer effects of ELOB depletion can be rescued by simultaneous knockdown of p14/ARF, confirming the mechanistic relationship
To experimentally validate this relationship, researchers should:
Perform coimmunoprecipitation to demonstrate physical interaction between components of the CRL2 complex and p14/ARF
Conduct in vivo ubiquitination assays after MG-132 treatment
Assess p14/ARF protein levels following ELOB manipulation using western blot
Perform rescue experiments with simultaneous manipulation of both ELOB and p14/ARF
ELOB-containing E3 ligase complexes intersect with multiple cancer-related signaling pathways:
Cell cycle regulation: GSEA analysis indicates that high ELOB expression correlates with cell cycle pathway activation
DNA replication: ELOB expression levels associate with DNA replication pathways, suggesting a role in regulating proteins involved in genome replication
PI3K-Akt signaling: Pathway enrichment analysis demonstrates a connection between ELOB expression and PI3K-Akt pathway activity, a key cancer-promoting signaling axis
Proteasome pathway: Naturally, ELOB's role in ubiquitination connects to the proteasomal degradation system
To comprehensively map these pathway interactions, researchers should:
Use RNA-seq following ELOB manipulation to identify global transcriptional changes
Conduct phospho-proteomic analysis to detect altered signaling node activity
Perform pathway analysis using tools like GSEA, GO enrichment, and KEGG pathway analysis with the R package "clusterProfiler"
Validate key interactions through targeted inhibition of identified pathway components
When creating stable ELOB knockdown models, researchers should consider:
Essential gene concerns: ELOB's fundamental role in cellular processes may make complete knockout lethal or severely growth-inhibiting, requiring inducible systems or partial knockdown approaches
Off-target effects: Carefully design siRNA sequences to avoid off-target effects; validate with multiple independent sequences and rescue experiments
Functional redundancy: Consider potential compensation by related proteins (e.g., other elongin family members)
Experimental validation: Confirm knockdown efficiency at both RNA and protein levels using qRT-PCR and western blotting
Phenotypic assessment: Thoroughly characterize models using proliferation assays, cell cycle analysis, and apoptosis measurements to distinguish between specific and non-specific effects
For optimal results, researchers should employ lentiviral delivery of shRNA or CRISPR-Cas9 systems for stable integration, with tetracycline-inducible promoters to control the degree and timing of ELOB depletion .
To study ELOB interactions in complex cellular contexts:
Proximity labeling approaches:
BioID or TurboID fusion proteins to identify proteins in close proximity to ELOB
APEX2-based proximity labeling for temporal resolution of interaction networks
Mass spectrometry-based interactomics:
Immunoprecipitation of tagged ELOB followed by mass spectrometry
SILAC or TMT labeling for quantitative comparison across conditions
Fluorescence-based interaction studies:
FRET (Förster Resonance Energy Transfer) or BiFC (Bimolecular Fluorescence Complementation) to visualize interactions in living cells
High-content imaging for spatiotemporal dynamics
Genetic interaction mapping:
CRISPR screening approaches to identify synthetic lethal interactions
Combinatorial genetic manipulation to map functional interactions
Single-cell analysis:
Based on ELOB's role in cancer progression, several therapeutic approaches warrant investigation:
Direct ELOB inhibition:
Small molecule inhibitors disrupting ELOB interactions with other complex components
Peptide-based inhibitors mimicking key interaction interfaces
CRL2 complex disruption:
Substrate-specific approaches:
Blocking recognition of specific substrates like p14/ARF
Developing proteolysis-targeting chimeras (PROTACs) to redirect ELOB-containing complexes
Combinatorial approaches:
Pairing ELOB targeting with inhibitors of intersecting pathways like PI3K-Akt
Synthetic lethality-based combinations
Gene therapy approaches:
siRNA delivery systems specifically targeting ELOB
CRISPR-based approaches for precise genetic manipulation
To identify potential therapeutic compounds, researchers can utilize the Connectivity Map (CMap) analysis platform (https://clue.io), which can discover small molecules and inform clinical trials by identifying compounds with expression signatures inverse to ELOB-driven signatures .
Understanding cross-species conservation is crucial for translational research:
Evolutionary conservation: The mouse Elob gene (MGI:1914923) shares significant homology with human ELOB, suggesting conserved fundamental functions .
Expression patterns: Both human and mouse ELOB show widespread tissue distribution, though some tissue-specific differences may exist .
Functional conservation: The core role in ubiquitin ligase complexes appears conserved, though substrate specificity may vary between species.
Model considerations: When using mouse models:
Validate key findings in human cell lines or tissues
Consider potential differences in protein interaction networks
Assess pathway conservation through comparative genomics
Evaluate phenotypic differences following genetic manipulation
Translational limitations: Researchers should be aware that certain cancer-promoting mechanisms, particularly those involving p14/ARF, may have species-specific aspects requiring careful validation across models .
For comprehensive ELOB expression analysis across cancer datasets:
Data acquisition:
Extract RNA-seq data with clinical information from TCGA (https://tcga-data.nci.nih.gov/tcga/)
Access additional datasets through cBioPortal (https://www.cbioportal.org/)
Differential expression analysis:
Survival analysis:
Employ Kaplan-Meier method with log-rank test
Utilize online tools like Kaplan-Meier plotter (https://kmplot.com/analysis/)
Methylation analysis:
Use the Methsurv database (https://biit.cs.ut.ee/methsurv/) to analyze DNA methylation patterns
Correlate methylation status with expression and clinical outcomes
Pathway and network analysis:
For integrative multi-omics analysis of ELOB:
Data layer integration:
Combine genomic (mutations, CNVs), transcriptomic (expression), proteomic, and epigenomic (methylation) data
Use tools like MultiOmics Factor Analysis (MOFA) for dimensionality reduction across data types
Single-cell multi-omics:
Network construction and analysis:
Build integrative networks incorporating expression, protein-protein interactions, and pathway information
Identify key nodes and regulatory relationships
Use weighted gene co-expression network analysis (WGCNA) to identify modules associated with ELOB expression
Causal inference approaches:
Apply computational causal inference to identify potential regulatory relationships
Use methods like Mendelian randomization to establish causal relationships where genetic variants are available
Visualization and interpretation:
This comprehensive approach enables researchers to place ELOB in broader cellular contexts and identify novel functional relationships that may not be apparent from single-omics analyses.
Several critical questions remain unexplored:
Developmental roles: How does ELOB function during embryonic development and tissue differentiation? Mouse models suggest broad expression across tissue types, but detailed developmental functions remain unclear .
Tissue-specific functions: Does ELOB have tissue-specific roles beyond its core function in ubiquitination? The widespread expression pattern suggests potential specialized functions in different cellular contexts .
Non-canonical functions: Beyond its established roles in transcriptional regulation and protein degradation, does ELOB participate in other cellular processes?
Regulatory mechanisms: How is ELOB itself regulated at transcriptional, post-transcriptional, and post-translational levels?
Cancer type specificity: Why does ELOB overexpression appear particularly impactful in breast cancer, and does this extend to other cancer types?
Therapeutic resistance: Does ELOB contribute to therapeutic resistance mechanisms in cancer treatment?
Non-oncogenic roles in disease: Does ELOB dysregulation contribute to non-cancer pathologies?
To address these questions, researchers should employ conditional knockout models, tissue-specific approaches, and comprehensive multi-omics analyses across development and disease contexts.
Cutting-edge technologies poised to enhance ELOB research include:
Spatial transcriptomics and proteomics:
Mapping ELOB expression and its targets with spatial resolution in tissues
Correlating with pathological features in disease states
CRISPR-based functional genomics:
Base editing for precise genetic manipulation
CRISPRi/CRISPRa for reversible modulation of ELOB expression
CRISPR screens to identify synthetic lethal interactions
Structural biology approaches:
Cryo-EM to resolve structures of ELOB-containing complexes
Hydrogen-deuterium exchange mass spectrometry for dynamic structural information
AlphaFold and other AI-based structure prediction for interaction modeling
Single-molecule techniques:
Live-cell imaging of ELOB dynamics
Single-molecule pulldown to analyze complex stoichiometry
Super-resolution microscopy for subcellular localization
Organoid and patient-derived models:
3D organoid systems to study ELOB in more physiologically relevant contexts
Patient-derived xenografts to validate findings in heterogeneous tumor environments
These technologies will provide unprecedented insights into ELOB biology and potentially reveal novel therapeutic opportunities across multiple disease contexts.
Elongin-B is a small protein with a molecular mass of approximately 13.1 kDa and consists of 118 amino acids . It is a non-glycosylated polypeptide chain produced in Escherichia coli (E. coli) and purified using standard chromatography techniques . The protein is typically stored in a sterile, filtered, colorless solution containing Tris-HCl, DTT, PMSF, and glycerol to maintain its stability .
The primary function of Elongin-B is to initiate the elongation phase of transcription by RNA polymerase II. It achieves this by suppressing the transient pausing of the polymerase at various sites within transcription units . This suppression is essential for the efficient transcription of genes and the proper regulation of gene expression.
Elongin-B forms a heterotrimeric complex with elongins A and C. This complex is known as the SIII complex and is involved in the regulation of transcription elongation . The von Hippel-Lindau (VHL) tumor suppressor protein also interacts with elongins B and C, inhibiting transcription elongation . This interaction is crucial for the regulation of gene expression and the prevention of uncontrolled cell growth, which can lead to tumor formation.
Recombinant human Elongin-B is widely used in research to study the mechanisms of transcription elongation and the regulation of gene expression. It is also used to investigate the interactions between elongins and other proteins, such as the VHL tumor suppressor protein . The recombinant protein is suitable for various applications, including SDS-PAGE and functional studies .