EBNA1BP2 is essential for processing 27S pre-rRNA into mature 18S rRNA, a critical step in 40S ribosomal subunit formation . It associates with nucleolar complexes, including the PeBoW complex, to ensure proper ribosome assembly.
EBNA1BP2 mediates EBV episome attachment to host chromosomes via EBNA1 binding. This interaction promotes viral genome persistence during latency and is linked to transcriptional silencing of host genes near attachment sites (e.g., neuronal and protein kinase A pathways) .
EBNA1BP2 is dysregulated in multiple cancers, with implications for prognosis and therapy:
Cancer Type | EBNA1BP2 Expression | Data Source |
---|---|---|
Bladder (BLCA) | Upregulated | TCGA, TIMER2.0 |
Breast (BRCA) | Upregulated | CPTAC, HPA |
Liver (LIHC) | Upregulated | TCGA, GEPIA2.0 |
Kidney (KICH) | Downregulated | TCGA |
Protein-Level Evidence: Elevated EBNA1BP2 protein levels observed in breast, liver, and lung cancers via immunohistochemistry (HPA) .
Promoter Methylation: Downregulated methylation in most cancers, potentially driving overexpression .
Immune Modulation: EBNA1BP2 expression negatively correlates with hypoxia and positively with immune cell infiltration (B cells, CD8+ T cells) .
EBNA1BP2 expression is influenced by chemical exposures and therapeutics:
Cell Cycle/DNA Repair: EBNA1BP2 is enriched in pathways linked to nucleoplasm organization and RNA binding, with single-cell analyses showing correlation with cell cycle progression .
Transcriptional Repression: EBV episome tethering via EBNA1BP2 recruits H3K9me3 to silence host genes, including tumor suppressors .
EBNA1BP2 is proposed as a pan-cancer prognostic biomarker due to its consistent overexpression in tumors and survival associations . Validated IHC staining in breast, liver, and lung cancers supports its clinical utility .
How do EBNA1BP2 mutations (e.g., G291N) impact ribosome biogenesis in cancer?
Can targeting EBNA1BP2-EBNA1 interactions disrupt EBV latency?
Does EBNA1BP2 modulate immune evasion mechanisms in tumors?
EBNA1BP2 (EBNA1 Binding Protein 2), also known by synonyms NOBP and EBP2, is a protein encoded by the EBNA1BP2 gene (NCBI Gene ID: 10969) in humans . It was initially identified as a protein that interacts with Epstein-Barr virus nuclear antigen 1 (EBNA1) and plays a crucial role in EBV latent infection . This protein is predominantly involved in nucleoplasm functions and RNA binding pathways .
At the molecular level, EBNA1BP2 has 6,025 functional associations with biological entities spanning 8 categories, including molecular profiles, chemicals, functional terms, diseases, phenotypes, structural features, cell types, and genes/proteins, extracted from 98 datasets . These associations highlight its diverse biological roles beyond viral interaction.
To study this protein effectively, researchers typically use antibody-based detection methods, with recombinant EBNA1BP2 protein (such as that derived from E. coli expression systems) serving as a valuable control for validation studies .
In normal cellular physiology, EBNA1BP2 functions primarily in the nucleoplasm compartment where it participates in RNA binding activities . Single-cell analysis demonstrates that EBNA1BP2 expression positively correlates with cell cycle progression and DNA repair processes, suggesting a role in cellular proliferation and genomic stability maintenance .
Mechanistically, EBNA1BP2 interacts with a network of proteins involved in nucleolar functions. Gene ontology (GO) enrichment analysis of EBNA1BP2-correlated genes reveals significant associations with fundamental cellular processes including ribosome biogenesis, RNA processing, and nucleolar organization .
For experimental investigation of these normal functions, researchers should consider using techniques that preserve native protein interactions, such as proximity ligation assays or co-immunoprecipitation combined with mass spectrometry, rather than relying solely on overexpression systems that may disrupt normal stoichiometry of interacting partners.
For detecting EBNA1BP2 expression in research settings, multiple complementary approaches should be employed:
RNA-level detection: Real-time PCR represents a sensitive method for quantifying EBNA1BP2 mRNA expression. The established primer sequences used in published research are:
Protein detection:
Database integration:
For tissues with dense cellular composition, consider using single-cell RNA sequencing to differentiate expression patterns among specific cell populations. When analyzing tumors, microdissection techniques may help isolate cancer cells from stromal components for more precise expression analysis.
EBNA1BP2 exhibits distinct expression patterns across normal and cancerous tissues. Comprehensive analysis using TIMER2.0 and other databases reveals:
Bladder urothelial carcinoma (BLCA)
Breast invasive carcinoma (BRCA)
Cervical squamous cell carcinoma (CESC)
Cholangiocarcinoma (CHOL)
Colon adenocarcinoma (COAD)
Esophageal carcinoma (ESCA)
Head and neck squamous cell carcinoma (HNSC)
Kidney renal clear cell carcinoma (KIRC)
Liver hepatocellular carcinoma (LIHC)
Lung adenocarcinoma (LUAD)
Lung squamous cell carcinoma (LUSC)
Prostate adenocarcinoma (PRAD)
Rectum adenocarcinoma (READ)
Stomach adenocarcinoma (STAD)
Thyroid carcinoma (THCA)
Interestingly, kidney chromophobe (KICH) shows the opposite trend, with lower EBNA1BP2 expression in tumor samples compared to normal tissues . This differential expression suggests tissue-specific regulatory mechanisms that warrant further investigation using tissue-specific cell models and ChIP-seq analysis for identifying regulatory elements.
EBNA1BP2 has emerged as a potential prognostic biomarker across multiple cancer types, with its prognostic significance varying by cancer type. Comprehensive survival analyses using GEPIA2.0 show that low EBNA1BP2 expression correlates with:
Adrenocortical carcinoma (ACC)
Bladder urothelial carcinoma (BLCA)
Brain lower grade glioma (LGG)
Liver hepatocellular carcinoma (LIHC)
Mesothelioma (MESO)
Sarcoma (SARC)
Better disease-free survival (DFS) in:
Adrenocortical carcinoma (ACC)
Head and neck squamous cell carcinoma (HNSC)
Kidney renal papillary cell carcinoma (KIRP)
Brain lower grade glioma (LGG)
Prostate adenocarcinoma (PRAD)
Sarcoma (SARC)
Interestingly, contrary findings were observed in certain cancer types such as kidney renal clear cell carcinoma (KIRC), ovarian cancer (OV), pheochromocytoma and paraganglioma (PCPG), stomach adenocarcinoma (STAD), and thyroid carcinoma (THCA), where higher EBNA1BP2 expression correlated with better outcomes .
To properly assess prognostic value in research settings, investigators should employ multivariate Cox regression models that account for confounding clinical variables and consider cancer molecular subtypes, which may explain these apparently contradictory findings in different tissues.
The mechanisms through which EBNA1BP2 contributes to tumor development appear multifaceted, with several key pathways identified through functional genomics and enrichment analyses:
Cell cycle regulation: At the single-cell level, EBNA1BP2 positively correlates with cell cycle progression and DNA repair processes, potentially promoting cancer cell proliferation .
Immune infiltration: EBNA1BP2 expression associates with immune cell infiltration patterns, including B cells, cancer-associated fibroblasts, and CD8+ T cells, suggesting involvement in tumor microenvironment modulation .
Epigenetic regulation: Promoter methylation analysis reveals downregulated methylation levels of EBNA1BP2 in most cancer types, indicating epigenetic dysregulation as a potential mechanism of overexpression .
Cellular adaptation: EBNA1BP2 expression negatively correlates with hypoxia response, potentially affecting tumor adaptation to microenvironmental stress .
For experimental validation of these mechanisms, CRISPR-Cas9 knockout or knockdown models combined with RNA-seq and ChIP-seq approaches would provide comprehensive insights into transcriptional networks and downstream effectors. Xenograft models with EBNA1BP2 manipulation would help validate its role in tumor growth and metastasis in vivo.
When investigating EBNA1BP2 in cancer tissues, several methodological considerations are crucial:
Sample selection and controls:
Include matched tumor and adjacent normal tissues whenever possible
Confirm tissue histology through pathological examination
Consider molecular subtypes of cancers, as EBNA1BP2's role may vary
Expression analysis:
Spatial context:
Consider intratumoral heterogeneity by analyzing multiple regions
Use laser capture microdissection for isolating specific cell populations
Employ spatial transcriptomics for mapping expression patterns within tumor architecture
Data integration:
Single-cell considerations:
These methodological considerations help ensure robust, reproducible results when studying EBNA1BP2 in cancer contexts.
EBNA1BP2 engages in multiple protein-protein interactions that influence its functional role in both normal and pathological contexts. Effective study of these interactions requires:
Interaction identification approaches:
Protein-protein interaction networks can be extracted from BioGRID (Biological General Repository for Interaction Datasets)
Yeast two-hybrid screening for novel interactors
Co-immunoprecipitation followed by mass spectrometry
Proximity-dependent biotin identification (BioID) or APEX labeling for context-specific interactions
Validation methods:
Reciprocal co-immunoprecipitation with specific antibodies
Fluorescence resonance energy transfer (FRET) or bimolecular fluorescence complementation (BiFC) for live-cell interaction studies
Surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) for determining binding kinetics and thermodynamics
Functional characterization:
Mutagenesis of key residues to disrupt specific interactions
Domain mapping to identify interaction interfaces
CRISPR-Cas9 gene editing to create interaction-deficient variants
The amino acid sequence segment ESDESLVTDRELQDAFSRGLLKPGLNVVLEG has been identified as a functional region of EBNA1BP2 , which may be particularly relevant for interaction studies. For studying viral interactions specifically, reconstitution of EBNA1BP2-EBNA1 complexes in vitro using purified components can provide mechanistic insights into structural determinants of these interactions.
The relationship between EBNA1BP2 and immune infiltration has significant implications for research design and interpretation:
Characterizing immune associations:
EBNA1BP2 expression correlates with infiltration of specific immune cell populations, including B cells, cancer-associated fibroblasts, and CD8+ T cells
These associations should be analyzed using multiple computational approaches (TIMER, EPIC, TIDE, QUANTISEQ, CIBERSORT, XCELL, MCPCOUNTER) for robust results
Methodological considerations:
Single-cell RNA sequencing is essential for distinguishing EBNA1BP2 expression in tumor cells versus immune infiltrates
Multiplex immunofluorescence or imaging mass cytometry can provide spatial context for expression relative to immune niches
Flow cytometry sorting of distinct cell populations followed by expression analysis helps determine cell-type-specific effects
Functional validation:
Co-culture experiments with manipulated EBNA1BP2 expression can assess direct effects on immune cell recruitment and function
Conditional knockout models in specific compartments (tumor vs. immune) help delineate cell-autonomous versus non-cell-autonomous effects
Checkpoint blockade response studies in EBNA1BP2-high versus low tumors may reveal therapeutic implications
When designing studies examining this relationship, researchers should carefully account for tumor type and stage, treatment history, and baseline immune characteristics of experimental models, as these factors can significantly influence results and interpretation.
Epigenetic regulation of EBNA1BP2 appears important in cancer contexts, with the promoter methylation level of EBNA1BP2 downregulated in the majority of cancer types . To investigate these mechanisms thoroughly:
Methylation analysis approaches:
Bisulfite sequencing of the EBNA1BP2 promoter region for site-specific methylation patterns
Methylation-specific PCR for targeted assessment of key regulatory regions
Genome-wide methylation arrays (e.g., Illumina MethylationEPIC) for contextualizing EBNA1BP2 methylation within broader epigenetic landscapes
RRBS (Reduced Representation Bisulfite Sequencing) for cost-effective profiling
Chromatin structure analysis:
ChIP-seq for histone modifications (H3K4me3, H3K27ac, H3K27me3) at the EBNA1BP2 locus
ATAC-seq to assess chromatin accessibility of the promoter and enhancer regions
Hi-C or similar techniques to investigate three-dimensional chromatin organization affecting EBNA1BP2 regulation
Transcriptional regulation:
ChIP-seq for transcription factors binding to the EBNA1BP2 promoter
Reporter assays with wild-type and mutated promoter constructs
CRISPR interference or activation (CRISPRi/CRISPRa) targeting regulatory elements
Integrated analysis:
Correlation of methylation patterns with expression levels across cancer types
Multi-omics approaches combining methylation, chromatin, and expression data
Single-cell multi-omics for cellular heterogeneity in epigenetic regulation
When investigating epigenetic mechanisms, researchers should consider developmental stages, environmental exposures, and treatment effects that might influence the epigenetic landscape around EBNA1BP2.
Pathway analysis of EBNA1BP2-correlated genes provides valuable insights for therapeutic targeting strategies:
Identification of relevant pathways:
Network-based approaches:
Generate protein-protein interaction networks using BioGRID and similar databases
Identify hub proteins or signaling nodes that connect EBNA1BP2 to critical cellular functions
Perform gene set enrichment analysis (GSEA) to identify enriched pathways in EBNA1BP2-high versus low tumors
Therapeutic implications:
Target synthetic lethal interactions by identifying genes whose inhibition is selectively toxic in EBNA1BP2-high cells
Explore vulnerability to RNA metabolism inhibitors, given EBNA1BP2's association with RNA binding pathways
Investigate combinatorial approaches targeting both EBNA1BP2-dependent pathways and immune checkpoints, based on immune infiltration correlations
Experimental validation:
High-throughput drug screening in isogenic cell lines with EBNA1BP2 knockdown/overexpression
CRISPR-Cas9 screening to identify synthetic lethal interactions
Patient-derived xenograft models stratified by EBNA1BP2 expression for preclinical therapeutic evaluation
The correlation between EBNA1BP2 and cell cycle/DNA repair processes suggests potential synergy with existing therapies targeting these pathways, such as PARP inhibitors or cell cycle checkpoint inhibitors, which should be systematically evaluated.
Several important contradictions and knowledge gaps exist in current EBNA1BP2 research:
Cancer-type specific effects:
While low EBNA1BP2 expression correlates with better prognosis in most cancers (ACC, BLCA, LGG, LIHC, MESO, SARC, UCS), the opposite trend is observed in others (KIRC, OV, PCPG, STAD, THCA)
The molecular basis for these differential effects remains unexplained and requires tissue-specific mechanistic studies
Causality versus correlation:
Current evidence primarily establishes correlations between EBNA1BP2 expression and cancer outcomes
Causal relationships and mechanisms by which EBNA1BP2 influences tumor biology need further investigation through interventional studies
EBV-dependent versus independent functions:
While EBNA1BP2 was initially identified through its interaction with the Epstein-Barr virus, its broader roles independent of viral infection remain poorly characterized
The extent to which viral interaction influences EBNA1BP2's cellular functions requires clarification
Regulatory mechanisms:
While promoter methylation changes have been observed, the comprehensive regulatory network controlling EBNA1BP2 expression, including transcription factors and non-coding RNAs, remains incomplete
The stimuli and signaling pathways that modulate EBNA1BP2 expression under physiological and pathological conditions are not well defined
Protein structure-function relationships:
Detailed structural information about EBNA1BP2 and how it relates to functional interactions is limited
The specific domains mediating protein-protein interactions and their conformational dynamics need characterization
Addressing these gaps requires integrative approaches combining structural biology, systems biology, and functional genomics in relevant model systems.
Single-cell technologies provide transformative insights into EBNA1BP2 function within heterogeneous tumor environments:
Cellular heterogeneity mapping:
Single-cell RNA sequencing reveals cell-type-specific expression patterns of EBNA1BP2 across tumor ecosystems
t-SNE visualization of EBNA1BP2 expression across tumor samples helps identify distinct cellular clusters with differential expression
CancerSEA analysis at the single-cell level reveals correlations between EBNA1BP2 expression and functional states such as cell cycle progression, DNA repair, and hypoxia response
Methodological approaches:
scRNA-seq coupled with computational lineage inference to track EBNA1BP2 expression changes during tumor evolution
CITE-seq or REAP-seq for simultaneous profiling of EBNA1BP2 mRNA and surface proteins on immune cells
Spatial transcriptomics to map EBNA1BP2 expression within architectural features of tumors
Single-cell ATAC-seq to correlate chromatin accessibility with EBNA1BP2 expression
Functional relationships:
Integration of single-cell data with spatial information to understand EBNA1BP2's role in tumor-stroma-immune interactions
Correlation of EBNA1BP2 expression with markers of stemness, differentiation, and drug resistance at single-cell resolution
Trajectory analysis to identify cellular states where EBNA1BP2 expression changes dynamically
Therapeutic implications:
Identification of rare cell populations with extreme EBNA1BP2 expression that may drive therapeutic resistance
Determination of optimal cellular targets based on co-expression patterns with druggable pathways
Prediction of treatment response heterogeneity based on EBNA1BP2-associated cellular states
For effective single-cell studies of EBNA1BP2, researchers should consider technical factors such as dissociation protocols that preserve cellular states, depth of sequencing adequate for detecting moderately expressed genes, and computational approaches that account for technical artifacts while preserving biological variation.
Based on current evidence, several high-priority research directions for EBNA1BP2 in cancer biology include:
Mechanistic dissection of tissue-specific effects: Investigating why EBNA1BP2 has opposing prognostic significance in different cancer types through tissue-specific knockout models and transcriptomic profiling.
Therapeutic targeting: Developing approaches to modulate EBNA1BP2 function or expression, potentially through:
Small molecule inhibitors targeting protein-protein interactions
Degrader technologies (PROTACs) targeting EBNA1BP2 for ubiquitin-mediated degradation
RNA-based therapeutics to modulate expression levels
Biomarker development: Validating EBNA1BP2 as a prognostic or predictive biomarker in prospective clinical trials, particularly in cancer types where it shows strong survival correlations.
Immune modulation: Exploring how EBNA1BP2 influences the tumor immune microenvironment and response to immunotherapies, given its correlations with immune cell infiltration.
Structural biology: Determining the three-dimensional structure of EBNA1BP2 and its complexes to enable structure-based drug design and mechanistic insights.
For effective advancement of these research directions, collaborative approaches combining clinical samples, diverse model systems, and integrative multi-omics analyses will be essential to develop a comprehensive understanding of EBNA1BP2's role in cancer biology and its potential as a therapeutic target.
To address contradictory findings in EBNA1BP2 research, investigators should implement the following experimental design principles:
Standardized methodology:
Context-specific investigation:
Study EBNA1BP2 in multiple cell types and cancer models simultaneously
Analyze effects within specific molecular subtypes of cancers
Consider microenvironmental factors that may influence EBNA1BP2 function
Multi-level analysis:
Integrate genomic, transcriptomic, proteomic, and functional data
Correlate EBNA1BP2 genetic alterations with expression and function
Employ both in vitro and in vivo models for validation
Mechanistic focus:
Use CRISPR-Cas9 for precise genetic manipulation rather than relying solely on overexpression or siRNA
Employ domain-specific mutations to dissect functional regions
Utilize inducible systems to study temporal aspects of EBNA1BP2 function
Transparent reporting:
Clearly document experimental conditions and cellular context
Report all outcomes, including negative results
Share raw data through appropriate repositories
The EBNA1BP2 gene is located on chromosome 1 and encodes a protein that is approximately 37.2 kDa in size . The protein consists of several domains that facilitate its interaction with other cellular components. Notably, EBNA1BP2 contains a conserved 200-300 amino acid block at its C-terminus, which is essential for its function .
One of the most intriguing aspects of EBNA1BP2 is its interaction with the Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA1). EBNA1 is a multifunctional DNA-binding protein that plays a key role in the replication and maintenance of the EBV episome within infected cells . The interaction between EBNA1 and EBNA1BP2 is crucial for the stable segregation of EBV episomes during cell division, ensuring the persistence of the virus in the host cell .
Due to its role in rRNA processing and its interaction with EBV, EBNA1BP2 is a subject of interest in various research fields, including virology, molecular biology, and cancer research. Studies on EBNA1BP2 can provide insights into the mechanisms of ribosome biogenesis and the persistence of viral infections in host cells .