The RIOX2 Recombinant Monoclonal Antibody (Clone 1E8) is synthesized using a plasmid-based system in mammalian cell lines, such as Expi293F cells. The process involves inserting antibody gene sequences into expression vectors, followed by affinity purification (e.g., affinity chromatography) .
Scalability: Produced in vitro with consistent batch-to-batch performance .
Animal-Free: Ethical alternative to traditional monoclonal antibody production .
Specificity: Targets a synthesized peptide derived from human RIOX2 .
RIOX2 (Ribosomal Oxygenase 2) is a JmjC domain-containing oxygenase that:
Demethylates histone H3K9me3, promoting ribosomal RNA synthesis .
Hydroxylates 60S ribosomal protein L27a (His-39), critical for ribosome biogenesis .
Regulates cell growth and survival, with overexpression linked to cancer progression .
Prostate Cancer: RIOX2 upregulation correlates with poor prognosis and disease-specific survival .
Other Cancers: Elevated expression observed in colon, lung, breast, and liver cancers .
The recombinant antibody is validated for detecting RIOX2 in human samples via ELISA, enabling quantitative analysis of protein levels in cancer research .
Parameter | Detail |
---|---|
Target | Human RIOX2 protein |
Sensitivity | High affinity due to sequence-defined engineering |
Usage | Cancer biomarker studies, epigenetics research |
Prostate Cancer: RIOX2 protein levels are significantly higher in malignant tissues compared to benign counterparts, confirmed via immunohistochemistry .
Genomic Alterations: RIOX2 mRNA upregulation correlates with genetic gain/amplification in prostate cancer .
Prostate Cancer: RIOX2 overexpression is an independent predictor of disease-specific survival .
Mechanism: RIOX2 promotes cell proliferation, anti-apoptosis, and carcinogenesis via c-Myc upregulation .
The recombinant RIOX2 antibody is a monoclonal antibody produced in vitro using the RIOX2 antibody genes, typically expressed from a plasmid within a stable mammalian cell line. These genes ultimately assemble into a fully functional antibody after translation, resulting in the recombinant RIOX2 antibody. This antibody is synthesized against the RIOX2 protein and is purified using affinity chromatography. This recombinant RIOX2 antibody is suitable for use in ELISA to detect the RIOX2 protein from human samples.
RIOX2, a JmjC (Jumonji-C) domain-containing 2-oxoglutarate (2OG)-dependent oxygenase, plays a crucial role in gene transcription within eukaryotic cells. It contributes to cell proliferation, cell cycle transitions, and anti-apoptotic carcinogenic activities. Upregulation of RIOX2 has been observed in various human solid and hematological malignancies, including colon, esophagus, lung, lymphocyte, kidney, nervous system, liver, breast, pancreas, and gastric cancers. Elevated RIOX2 expression has been associated with a poor prognosis. Notably, evidence indicates that downregulation of RIOX2 inhibits cancer cell growth and survival.
RIOX2 (Ribosomal Oxygenase 2) is a protein-coding gene located on chromosome 3q11.2 that is conserved across multiple species including primates, rodents, and other vertebrates . The protein functions as a bifunctional enzyme with dual catalytic activities: it acts as both a histone lysine demethylase that removes methyl groups from trimethylated 'Lys-9' on histone H3 (H3K9me3) and as a ribosomal histidine hydroxylase that catalyzes the hydroxylation of 60S ribosomal protein L27a on 'His-39' .
The demethylation activity of RIOX2 leads to increased ribosomal RNA expression, while its hydroxylation function is believed to play a role in ribosome biogenesis, particularly during the assembly process of pre-ribosomal particles . RIOX2 is also a c-Myc target gene that may significantly influence cell proliferation and growth regulation . It is widely expressed in multiple tissues including thyroid and skin, and has been implicated in diseases such as Squamous Cell Carcinoma and Lung Cancer .
At the subcellular level, RIOX2 is primarily localized to the nucleus and nucleolus, consistent with its roles in transcriptional regulation and ribosome biogenesis . Its involvement in chromatin organization and validated targets of C-MYC transcriptional activation pathways further underscores its importance in fundamental cellular processes .
Recombinant monoclonal antibodies represent a significant advancement over traditional monoclonal antibodies, particularly in terms of production methodology and consistency. While traditional monoclonal antibodies are typically generated through animal immunization followed by hybridoma technology, recombinant monoclonal antibodies are produced using recombinant DNA technology and in vitro cloning methods .
The fundamental difference lies in the production process: traditional monoclonal antibodies are susceptible to spontaneous mutations that can lead to batch-to-batch variations, whereas recombinant monoclonal antibodies are produced from defined genetic sequences under controlled conditions . This controlled production results in several key advantages:
Superior batch-to-batch consistency, which is critical for longitudinal studies where reagent variation could compromise result validity
Enhanced reproducibility and validation capabilities
Ability to be easily engineered or modified to increase binding affinity or specificity
Scalable production that ensures continuous supply for extended research projects
During the conversion from hybridoma to recombinant antibodies, the antibody-encoding genes from hybridoma cell lines are cloned into expression vectors. The resulting recombinant antibody retains the same antigen-binding sequences and specificity as the parental hybridoma, maintaining experimental continuity while improving reliability .
RIOX2 recombinant monoclonal antibodies are versatile research tools applicable across multiple experimental platforms. Based on validated testing data, these antibodies are particularly suitable for:
Western Blot (WB): Effective at dilutions ranging from 1:500-1:2000, allowing for sensitive detection of endogenous RIOX2 protein levels . WB applications are especially valuable for quantifying RIOX2 expression changes following experimental treatments or in different disease states.
Immunocytochemistry (ICC) and Immunofluorescence (IF): Optimal at dilutions of 1:500-1:2500, these applications enable visualization of RIOX2's subcellular localization . Studies have demonstrated clear nuclear and nucleolar staining patterns consistent with RIOX2's biological functions.
Immunoprecipitation (IP): Using 0.2-1μL antibody per mg of lysate allows efficient isolation of RIOX2 and its binding partners . This application is particularly valuable for studying protein-protein interactions and post-translational modifications.
ELISA: High sensitivity at dilutions of 1:5000-20000 makes these antibodies ideal for quantitative detection of RIOX2 in solution .
These applications support diverse research objectives including protein expression analysis, localization studies, protein interaction investigations, and quantitative assessments. For example, immunofluorescence studies have revealed that RIOX2 shows distinct localization patterns that can change in response to cellular stimuli, as demonstrated in experiments with somatostatin receptor systems .
Implementing appropriate controls is critical for ensuring reliable and interpretable results when working with RIOX2 antibodies. A comprehensive control strategy should include:
Positive Controls:
Cell lines with confirmed RIOX2 expression (e.g., A431, Raji, Jurkat, and HeLa cells have been validated to express detectable levels of RIOX2)
Recombinant RIOX2 protein as a reference standard
Tissue sections known to express RIOX2 (thyroid and skin tissues show reliable expression)
Negative Controls:
RIOX2 knockout or knockdown cell lines (using CRISPR-Cas9 or siRNA technology)
Cell lines with naturally low RIOX2 expression
Primary antibody omission control to assess non-specific binding of secondary antibodies
Isotype controls using non-targeting IgG from the same species as the RIOX2 antibody
Specificity Controls:
Competitive blocking experiments using the immunizing peptide
Western blot detection of a single band at the expected molecular weight (53 kDa)
Parallel testing with multiple independent RIOX2 antibodies targeting different epitopes
Treatment Validation Controls:
Cells with manipulated RIOX2 expression through overexpression or downregulation
Treatment conditions known to alter RIOX2 expression (e.g., c-Myc activation/inhibition)
For advanced applications such as ChIP or proximity ligation assays, additional controls specific to these techniques should be incorporated. Remember that proper control selection is application-dependent—for instance, immunofluorescence studies require different controls than immunoprecipitation experiments.
Sample preparation protocols significantly impact RIOX2 antibody performance across different applications. The following methodologies have been optimized for various experimental approaches:
For Western Blot:
Lyse cells in RIPA buffer supplemented with protease inhibitors and phosphatase inhibitors
Homogenize tissues thoroughly using mechanical disruption in cold lysis buffer
Clear lysates by centrifugation at 14,000g for 15 minutes at 4°C
Determine protein concentration using Bradford or BCA assay
Denature samples at 95°C for 5 minutes in reducing sample buffer
Transfer proteins to PVDF membrane (preferred over nitrocellulose for RIOX2 detection)
For Immunofluorescence/Immunocytochemistry:
Fix cells with 4% paraformaldehyde for 15 minutes at room temperature
Block with 10% serum (matching species of secondary antibody) for 1 hour
Incubate with RIOX2 antibody at 1:500-1:2500 dilution overnight at 4°C
Use fluorophore-conjugated secondary antibodies at appropriate dilutions
For Immunoprecipitation:
Prepare cell lysates in non-denaturing buffer containing 1% NP-40 or similar mild detergent
Use 0.2-1 μL antibody per mg of lysate with Protein A/G magnetic beads
Pre-clear lysates with beads alone to reduce non-specific binding
Incubate antibody-bead complex with lysate overnight at 4°C with gentle rotation
General Considerations:
Fresh samples typically yield better results than frozen-thawed material
For small volumes of antibody, briefly centrifuge vials to collect liquid that may become entrapped in the cap during shipping and storage
Store antibodies according to manufacturer recommendations (typically -20°C to -80°C for long-term storage)
These optimized protocols help ensure consistent performance and reliable results when working with RIOX2 antibodies across different experimental platforms.
Optimal antibody dilution is critical for achieving the best signal-to-noise ratio while conserving valuable reagents. Based on empirical testing with RIOX2 antibodies, the following application-specific dilution ranges are recommended:
For best results, preliminary titration experiments are recommended when working with new lots of antibody or different sample types. The optimal dilution may vary depending on the expression level of RIOX2 in your specific samples and the detection system being used.
When using enhanced chemiluminescence (ECL) for Western blot detection, secondary antibodies conjugated to HRP typically perform well at 1:10,000 dilution . For fluorescence applications, Alexa Fluor-conjugated secondary antibodies at 1:1000-1:2000 dilutions provide excellent signal with minimal background .
Remember that antibody performance can be affected by storage conditions. Aliquoting antibodies upon receipt and storing at recommended temperatures helps maintain consistency throughout your research project.
Verifying antibody specificity is crucial for reliable experimental outcomes. For RIOX2 antibodies, implement these comprehensive validation approaches:
Genetic Validation:
CRISPR/Cas9 Knockout Verification: Generate RIOX2 knockout cell lines using CRISPR/Cas9 gene editing. The true RIOX2 signal should be absent in knockout lines while present in wild-type cells. Note that CRISPR/Cas9 editing may cause alternative splicing of the targeted mRNA, potentially creating truncated proteins , so multiple knockout strategies targeting different exons are advisable.
siRNA Knockdown Analysis: Reduce RIOX2 expression using siRNA and confirm corresponding reduction in antibody signal by Western blot or immunofluorescence. Quantify the knockdown efficiency by densitometry.
Biochemical Validation:
Molecular Weight Confirmation: Verify detection of a single band at the expected molecular weight of 53 kDa in Western blots .
Peptide Competition Assay: Pre-incubate the antibody with excess immunizing peptide before application to samples. This should block specific binding and eliminate the true RIOX2 signal.
Multiple Antibody Comparison: Test multiple RIOX2 antibodies targeting different epitopes. Consistent detection patterns across antibodies suggest specific recognition.
Functional Validation:
Treatment-Response Correlation: Since RIOX2 is a c-Myc target gene, treatment with c-Myc activators/inhibitors should produce predictable changes in RIOX2 levels that correlate with antibody signal intensity.
Localization Pattern Analysis: Confirm nuclear and nucleolar localization in immunofluorescence studies, consistent with RIOX2's known subcellular distribution .
Post-Translational Modification Detection: Verify antibody detection of RIOX2 under conditions that alter its post-translational modification state, if the antibody is not modification-specific.
Advanced Validation:
Mass Spectrometry Confirmation: Perform immunoprecipitation followed by mass spectrometry identification of the pulled-down protein.
Tissue Cross-Reactivity Panel: Test antibody reactivity across tissues with known RIOX2 expression profiles (e.g., thyroid, skin) .
Inconsistent results when using RIOX2 antibodies can stem from multiple sources. Identifying and addressing these factors is essential for experimental reproducibility:
Antibody-Related Factors:
Batch-to-Batch Variability: Traditional antibodies may show significant variation between lots. Recombinant monoclonal antibodies offer superior consistency , but batch testing is still advisable.
Antibody Degradation: Repeated freeze-thaw cycles, improper storage temperatures, or contamination can degrade antibody quality . Store antibodies at recommended temperatures (-20°C to -80°C) and aliquot upon receipt to minimize freeze-thaw cycles.
Epitope Accessibility Issues: Certain sample preparation methods may mask the epitope recognized by the antibody. If experiencing problems, test alternative fixation and permeabilization protocols.
Sample-Related Factors:
Protein Degradation: Inadequate protease inhibition can lead to RIOX2 degradation. Always use fresh protease inhibitor cocktails in sample preparation.
Post-Translational Modifications: Changes in phosphorylation, methylation, or other modifications may affect antibody recognition depending on the epitope location.
Sample Heterogeneity: Variation in RIOX2 expression between cell populations or tissue regions can cause inconsistent results. Consider single-cell techniques for heterogeneous samples.
Technical Factors:
Inconsistent Sample Loading: Variation in protein loading can be misinterpreted as biological differences. Always normalize to housekeeping proteins or total protein staining.
Detection System Variability: ECL reagent degradation or inconsistent imaging exposure times can cause apparent signal variations. Standardize detection methods and include calibration controls.
Buffer Composition Differences: Small changes in buffer pH, salt concentration, or detergent content can significantly impact antibody performance. Maintain consistent buffer preparations.
Biological Factors:
Cell Cycle Dependence: RIOX2 expression and localization may vary throughout the cell cycle. Synchronize cells when possible or account for this variation in experimental design.
Stress Response Effects: Cellular stress during experimental manipulation can alter RIOX2 levels. Minimize handling stress and include appropriate controls.
Cell Confluence Effects: RIOX2 expression may change with cell density. Standardize cell confluence levels between experiments.
Troubleshooting Strategy:
Systematically test each variable while keeping others constant
Include positive and negative controls in each experiment
Document all experimental conditions meticulously
Consider using alternative detection methods to confirm results
By addressing these potential sources of variability, researchers can achieve more consistent and reliable results when working with RIOX2 antibodies.
Interpreting changes in RIOX2 expression requires careful consideration of its biological context and methodological factors. Follow these guidelines for robust data interpretation:
Quantitative Assessment Framework:
Baseline Expression Establishment: Before interpreting treatment effects, thoroughly characterize baseline RIOX2 expression in your experimental system. This includes protein levels, subcellular localization, and potential isoform expression.
Multi-Method Confirmation: Validate expression changes using complementary techniques. For example, if Western blot shows increased RIOX2 levels, confirm with qRT-PCR for mRNA expression and immunofluorescence for protein localization changes.
Temporal Dynamics Analysis: RIOX2 responses may be time-dependent. Perform time-course experiments to distinguish transient from sustained effects and identify optimal analysis timepoints.
Functional Context Interpretation:
c-Myc Pathway Coordination: Since RIOX2 is a c-Myc target gene , correlate RIOX2 expression changes with c-Myc activity and other c-Myc-regulated genes to establish pathway context.
Dual Enzymatic Function Consideration: Remember that RIOX2 has both histone demethylase and ribosomal hydroxylase activities . Changes in RIOX2 expression may affect either or both functions, potentially impacting different downstream processes.
Subcellular Localization Shifts: Pay attention to changes in RIOX2 localization patterns. Redistribution between nucleoplasm and nucleolus may indicate functional shifts between chromatin regulation and ribosome biogenesis roles .
Experimental Design Considerations:
Dose-Response Relationships: Establish dose-dependent effects of treatments on RIOX2 expression to identify threshold concentrations and saturation points.
Cell Type Specificity: RIOX2 responses may vary between cell types. What increases expression in one cell type might decrease it in another due to different regulatory networks.
Reversibility Testing: Determine whether RIOX2 expression changes are reversible after treatment withdrawal, which helps distinguish adaptive from permanent alterations.
Biological Significance Assessment:
Correlation with Cellular Phenotypes: Link RIOX2 expression changes to functional outcomes like proliferation rates, differentiation status, or ribosome biogenesis markers.
Disease Context Relevance: For cancer-related studies, interpret RIOX2 changes in light of its associations with squamous cell carcinoma and lung cancer .
Systems Biology Perspective: Position RIOX2 alterations within broader pathway contexts using bioinformatics approaches like gene set enrichment analysis.
The example in the search results showed that when HEK-293 cells stably transfected with SSTR2 were treated with somatostatin-14, RIOX2 antibody immunofluorescence revealed internalization of receptors from the plasma membrane to perinuclear vesicle clusters . This demonstrates how RIOX2 antibodies can be used to track dynamic protein localization changes in response to stimuli.
RIOX2's dual functions as both a histone lysine demethylase and a ribosomal histidine hydroxylase present unique research opportunities. Here are sophisticated approaches using RIOX2 antibodies to dissect these distinct enzymatic activities:
Chromatin Immunoprecipitation (ChIP) Analysis:
Use RIOX2 antibodies for ChIP followed by sequencing (ChIP-seq) to map genome-wide binding sites, identifying regions where RIOX2 may act as a histone demethylase.
Perform sequential ChIP (ChIP-reChIP) with RIOX2 antibodies followed by H3K9me3-specific antibodies to identify genomic regions where both proteins co-localize.
Compare ChIP-seq data before and after experimental treatments that differentially affect RIOX2's demethylase function to identify condition-specific binding patterns.
Proximity Ligation Assays (PLA):
Combine RIOX2 antibodies with antibodies against histone H3K9me3 in PLA to visualize and quantify their physical proximity in situ.
Similarly, use RIOX2 and ribosomal protein L27a antibodies in PLA to investigate the ribosomal hydroxylase function in different cellular compartments.
Compare PLA signals between different cell types or treatment conditions to identify factors that shift RIOX2's functional balance between its dual roles.
Co-Immunoprecipitation (Co-IP) Studies:
Use RIOX2 antibodies for Co-IP followed by mass spectrometry to identify the complete interactome of RIOX2 in different cellular compartments.
Perform directed Co-IP experiments to detect interactions with chromatin-modifying complexes versus ribosomal assembly factors.
Analyze how these interaction profiles change under conditions that affect cell growth, differentiation, or stress responses.
Functional Enzymatic Assays:
Immunoprecipitate RIOX2 using specific antibodies and perform in vitro enzymatic assays to measure both demethylase and hydroxylase activities from the same sample.
Create domain-specific RIOX2 antibodies that preferentially recognize conformations associated with either enzymatic function.
Develop antibodies that specifically recognize the active sites of each enzymatic function to directly inhibit one activity while leaving the other intact.
Fluorescence Resonance Energy Transfer (FRET):
Combine fluorescently-labeled RIOX2 antibodies with labeled H3K9me3 or L27a antibodies to detect proximity in living cells through FRET.
Monitor FRET signals in real-time during cell cycle progression or differentiation to track dynamic shifts in RIOX2's functional associations.
Super-Resolution Microscopy:
Use RIOX2 antibodies in combination with super-resolution techniques (STORM, PALM, SIM) to precisely localize RIOX2 within nuclear subdomains.
Perform multi-color super-resolution imaging to map RIOX2's spatial relationships with chromatin marks versus ribosomal components at nanometer resolution.
These advanced approaches enable researchers to dissect the complex dual functionality of RIOX2, providing insights into how this bifunctional enzyme coordinates chromatin regulation and ribosome biogenesis in different cellular contexts.
RIOX2's associations with squamous cell carcinoma and lung cancer highlight its potential significance in oncogenesis. Here are sophisticated research strategies using RIOX2 antibodies to investigate its role in cancer progression:
Tissue Microarray (TMA) Analysis:
Use RIOX2 antibodies for immunohistochemical staining of cancer TMAs containing samples from multiple patients at different disease stages.
Correlate RIOX2 expression patterns with clinicopathological parameters, patient outcomes, and other molecular markers.
Perform multiplexed immunofluorescence with RIOX2 and other cancer-associated proteins to create comprehensive protein expression profiles.
Patient-Derived Xenograft (PDX) Models:
Establish PDX models from primary tumors and track RIOX2 expression during passage and metastasis using antibody-based methods.
Test how therapeutic interventions affect RIOX2 expression and localization in PDX models.
Compare RIOX2 expression in primary tumors versus their corresponding PDX models to identify potential selective pressures.
Cancer Cell Line Encyclopedia (CCLE) Validation:
Use RIOX2 antibodies to validate mRNA expression data from the CCLE across diverse cancer cell lines.
Identify cell line models with distinctive RIOX2 expression patterns for mechanistic studies.
Correlate RIOX2 protein levels with drug sensitivity profiles to identify potential therapeutic vulnerabilities.
Circulating Tumor Cell (CTC) Analysis:
Develop protocols using RIOX2 antibodies to detect and characterize CTCs from cancer patients.
Compare RIOX2 expression in primary tumors, metastatic lesions, and CTCs from the same patient.
Investigate whether RIOX2 expression in CTCs correlates with metastatic potential or treatment resistance.
Functional Studies in 3D Culture Models:
Manipulate RIOX2 expression in cancer cells and assess effects on spheroid formation, invasion, and resistance to apoptosis.
Use RIOX2 antibodies to monitor protein expression and localization in 3D culture systems that better recapitulate tumor architecture.
Combine with live-cell imaging to track dynamic changes in RIOX2 expression during specific phases of invasion or colony formation.
Experimental Metastasis Models:
Compare RIOX2 expression between primary tumors and experimental metastases using antibody-based detection methods.
Analyze whether RIOX2 knockdown affects metastatic potential in animal models.
Investigate RIOX2's role in specific steps of the metastatic cascade (invasion, circulation, extravasation, colonization).
Therapeutic Targeting Assessment:
Develop function-blocking antibodies against RIOX2 to directly inhibit its activities in cancer cells.
Test combinations of RIOX2 inhibition with standard chemotherapeutics or targeted agents.
Use RIOX2 antibodies to monitor treatment responses and resistance mechanisms.
The study by Zhou et al. (2019) demonstrated that Mina53 (RIOX2) regulates the differentiation and proliferation of leukemia cells , providing a foundation for further investigation of its roles in hematological malignancies. Similarly, research by Xuan et al. (2018) revealed that Mina53 deficiency leads to glioblastoma cell apoptosis by inducing DNA replication stress and diminishing DNA damage response , highlighting its potential as a therapeutic target in brain tumors.
RIOX2's role as a ribosomal histidine hydroxylase that modifies 60S ribosomal protein L27a on 'His-39' positions it as a significant player in ribosome biogenesis. Here are sophisticated research approaches using RIOX2 antibodies to investigate this critical cellular process:
Nucleolar Proteomics:
Use RIOX2 antibodies for immunoprecipitation from nucleolar extracts followed by mass spectrometry to identify its ribosome-associated interaction partners.
Compare RIOX2 interactomes under normal conditions versus ribosomal stress induced by actinomycin D or other nucleolar disruptors.
Perform temporal proteomics during synchronous ribosome biogenesis to track dynamic associations of RIOX2 with pre-ribosomal particles.
Ribosome Assembly Monitoring:
Combine RIOX2 antibodies with antibodies against various pre-rRNA processing factors in co-immunoprecipitation experiments to map RIOX2's involvement in specific assembly steps.
Use antibodies in sucrose gradient fractionation experiments to track RIOX2 association with different pre-ribosomal particles.
Develop proximity labeling techniques using RIOX2 antibodies to identify proteins in close proximity during active ribosome assembly.
Hydroxylation Activity Assessment:
Develop antibodies specifically recognizing hydroxylated His-39 on L27a to directly measure RIOX2's enzymatic output.
Compare hydroxylation levels between normal and disease states using these modification-specific antibodies.
Investigate how oxygen tension affects RIOX2-mediated hydroxylation using hypoxia models and correlate with ribosome function.
Nucleolar Stress Response:
Track RIOX2 localization during nucleolar stress using immunofluorescence to determine if it redistributes from the nucleolus.
Analyze how RIOX2 expression and post-translational modifications change during ribosomal stress.
Investigate whether RIOX2 participates in p53-dependent or p53-independent nucleolar stress response pathways.
Translation Efficiency Studies:
Correlate RIOX2 levels and L27a hydroxylation status with global and transcript-specific translation efficiency.
Perform polysome profiling coupled with RIOX2 immunoprecipitation to identify mRNAs preferentially translated by RIOX2-modified ribosomes.
Investigate whether RIOX2-modified ribosomes have altered affinity for specific translation factors or regulatory RNAs.
Structural Biology Approaches:
Use RIOX2 antibodies to purify native ribosomal complexes for cryo-electron microscopy studies.
Develop conformation-specific antibodies that recognize RIOX2 in its catalytically active state when bound to ribosomal substrates.
Employ antibody-based proximity labeling to map the structural environment of RIOX2 binding sites on pre-ribosomes.
Developmental and Tissue-Specific Analysis:
Track RIOX2 expression and L27a hydroxylation across different tissues and developmental stages to identify contexts with distinctive ribosome modification patterns.
Investigate tissue-specific consequences of RIOX2 depletion on ribosome biogenesis and function.
Analyze whether RIOX2-modified ribosomes have specialized functions in particular cell types or physiological contexts.
These approaches leverage RIOX2 antibodies to uncover the mechanistic details of how this enzyme contributes to ribosome biogenesis, potentially revealing new regulatory principles in this fundamental cellular process.