RNF122 Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your orders within 1-3 business days of receipt. Delivery times may vary depending on the purchasing method or location. For specific delivery details, please consult your local distributor.
Synonyms
RNF122; RING finger protein 122
Target Names
RNF122
Uniprot No.

Target Background

Function
This antibody may induce necrosis and apoptosis. It may also play a role in cell viability.
Database Links

HGNC: 21147

KEGG: hsa:79845

STRING: 9606.ENSP00000256257

UniGene: Hs.151237

Subcellular Location
Golgi apparatus. Endoplasmic reticulum. Membrane; Single-pass membrane protein.
Tissue Specificity
Widely expressed in several tissues and cell lines.

Q&A

What is RNF122 and why is it significant in research?

RNF122 (Ring Finger Protein 122) is an E3 ubiquitin ligase that plays critical roles in cellular processes including immune regulation, cell viability, and antiviral responses. This protein has gained significant research interest due to its selective negative regulatory role in antiviral innate responses triggered by pattern recognition receptors such as RIG-I. RNF122 is widely expressed in various tissues and cell lines, localizing primarily to the Golgi apparatus, endoplasmic reticulum, and cellular membranes as a single-pass membrane protein . Its importance extends to viral pathogenesis, as it has been shown to promote viral infections such as porcine reproductive and respiratory syndrome virus (PRRSV) through specific ubiquitination mechanisms that stabilize viral proteins and degrade host immune sensors .

What are the key specifications of commercially available RNF122 antibodies with biotin conjugation?

The RNF122 antibody with biotin conjugation is available with specific characteristics that researchers should consider when selecting reagents for their experiments:

ParameterSpecifications
Binding SpecificityAA 71-155 region of RNF122 protein
HostRabbit
ClonalityPolyclonal
ReactivityHuman, Rat (confirmed); Mouse, Dog, Cow, Pig, Horse, Rabbit (predicted)
ApplicationsWestern Blotting, ELISA, Immunohistochemistry (both frozen and paraffin-embedded sections)
Purification MethodProtein A purification
ImmunogenKLH conjugated synthetic peptide derived from human RNF122
IsotypeIgG
Buffer Composition50% Glycerol, 0.01M PBS, pH 7.4 with 0.03% Proclin 300 as preservative
FormLiquid

These specifications provide researchers with critical information for experimental design and antibody selection .

What are the recommended applications for RNF122 biotin-conjugated antibodies?

RNF122 biotin-conjugated antibodies are versatile reagents suitable for multiple research applications. The primary recommended applications include:

  • Western Blotting (WB): For detection of RNF122 protein in tissue or cell lysates with high sensitivity due to the biotin-streptavidin amplification system. The biotin conjugation enables enhanced signal detection through secondary detection with streptavidin-HRP or streptavidin-fluorophore conjugates.

  • Enzyme-Linked Immunosorbent Assay (ELISA): For quantitative detection of RNF122 in solution, utilizing the high-affinity biotin-streptavidin interaction for signal amplification.

  • Immunohistochemistry (IHC): For both frozen (IHC-F) and paraffin-embedded (IHC-P) tissue sections, allowing visualization of RNF122 localization within cells and tissues.

  • Immunofluorescence (IF): For co-localization studies with other cellular markers, particularly useful for confirming the subcellular localization of RNF122 to the Golgi apparatus and endoplasmic reticulum .

The biotin conjugation provides significant advantages for signal amplification and flexibility in detection methods across these applications.

How should RNF122 antibodies be stored and handled for optimal performance?

For optimal performance and longevity of RNF122 biotin-conjugated antibodies, follow these handling and storage protocols:

  • Short-term storage: Keep at 4°C with ice packs during shipping and for up to 2 weeks during active use.

  • Long-term storage: Store at -20°C in a non-frost-free freezer to prevent freeze-thaw cycles.

  • Aliquoting: Upon receipt, divide the antibody into small working aliquots (10-20 μL) to minimize freeze-thaw cycles, as repeated freeze-thaw can degrade the antibody and reduce its effectiveness.

  • Thawing procedure: When needed, thaw aliquots rapidly at room temperature and keep on ice during use.

  • Working dilutions: Prepare fresh working dilutions on the day of the experiment. Do not store diluted antibodies for extended periods.

  • Buffer conditions: The antibody is supplied in 50% glycerol with 0.01M PBS at pH 7.4 and 0.03% Proclin 300 as a preservative. Maintain these conditions when preparing dilutions .

Proper storage and handling are essential for maintaining antibody integrity and ensuring reproducible experimental results.

How can I optimize western blotting protocols when using biotin-conjugated RNF122 antibodies?

Optimizing western blotting protocols with biotin-conjugated RNF122 antibodies requires specialized approaches to maximize sensitivity while minimizing background:

  • Sample preparation:

    • Lyse cells in RIPA buffer supplemented with protease inhibitors and deubiquitinase inhibitors (particularly N-ethylmaleimide at 10mM) to preserve ubiquitinated forms of RNF122 and its targets.

    • For membrane proteins, include 1% SDS or 8M urea in the lysis buffer to ensure complete solubilization.

  • Blocking optimization:

    • Use 5% BSA rather than milk for blocking to prevent non-specific binding.

    • Add 0.1% Tween-20 to reduce background.

    • Consider adding avidin/biotin blocking steps if endogenous biotin is problematic.

  • Detection system:

    • Utilize streptavidin-HRP at 1:10,000 dilution for optimal signal-to-noise ratio.

    • For enhanced sensitivity, employ commercial signal amplification systems compatible with biotin-streptavidin detection.

  • Controls and validation:

    • Include RNF122-knockdown samples as negative controls.

    • Confirm specificity with competitive blocking using the immunogenic peptide (AA 71-155).

    • When detecting RNF122 (~26 kDa), include appropriate molecular weight markers.

  • Troubleshooting guidance:

    • For high background: Increase washing steps (6×5 minutes each) with TBS-T.

    • For weak signals: Increase primary antibody concentration or incubation time at 4°C overnight.

    • For multiple bands: Validate with RNF122 overexpression to confirm the correct band .

This optimized methodology enables robust and specific detection of RNF122 in western blotting applications.

What experimental approaches can be used to study RNF122's ubiquitination activity in cellular systems?

Investigating RNF122's ubiquitination activity requires carefully designed experimental strategies:

  • Ubiquitination assay design:

    • In vitro assays: Combine purified RNF122, E1 (UBA1), E2 (UBE2D family), ubiquitin, ATP, and potential substrates. Analyze ubiquitination using anti-ubiquitin western blotting.

    • Cell-based assays: Co-transfect cells with tagged RNF122, substrate candidates, and tagged ubiquitin (HA-Ub or Flag-Ub), followed by immunoprecipitation and western blotting.

  • Ubiquitin linkage analysis:

    • Use linkage-specific antibodies (K27, K48, K63) to determine ubiquitin chain types formed by RNF122.

    • Employ ubiquitin mutants (K27R, K48R, K63R) to confirm linkage specificity.

    • Mass spectrometry of immunoprecipitated complexes can identify precise ubiquitination sites and linkage types.

  • Functional consequence assessment:

    • For K63-linked ubiquitination: Analyze protein stability with cycloheximide chase assays.

    • For K48-linked ubiquitination: Use proteasome inhibitors (MG132) to confirm degradation pathways.

    • For K27-linked ubiquitination: Examine effects on protein localization and complex formation.

  • Experimental tools:

    • Mutational analysis: Generate RNF122 RING domain mutants (C92A/C95A) to abolish E3 ligase activity as controls.

    • Domain-specific interactions: Use truncated constructs to map interaction domains with substrates.

    • Proximity labeling: Employ BioID or TurboID fusions with RNF122 to identify proximal proteins as potential substrates .

Research has demonstrated that RNF122 mediates distinct ubiquitination patterns: K63-linked ubiquitination of viral proteins (e.g., PRRSV nsp4) to enhance stability, and K27/K48-linked ubiquitination of host immune sensors (e.g., MDA5) for degradation and immune evasion .

How can I investigate the interaction between RNF122 and pattern recognition receptors in antiviral immunity?

To investigate RNF122's interactions with pattern recognition receptors (PRRs) in antiviral immunity, implement these methodological approaches:

  • Co-immunoprecipitation (Co-IP) strategies:

    • Transfect cells with tagged RNF122 (Myc-RNF122) and tagged PRRs (Flag-MDA5 or Flag-RIG-I).

    • Perform bidirectional Co-IPs (IP with anti-Myc, blot with anti-Flag and vice versa).

    • For endogenous interactions, use biotin-conjugated RNF122 antibodies with streptavidin beads, followed by western blotting for PRRs.

    • Include RNase treatment to determine if interactions are RNA-dependent.

  • Domain mapping:

    • Generate truncation mutants of both RNF122 and PRRs to map interaction interfaces.

    • Focus on the CARD domains of RIG-I and MDA5, which have been implicated in RNF122 interactions.

    • Create point mutations in key residues to pinpoint critical interaction sites.

  • Functional assays:

    • Reporter systems: Utilize IFN-β luciferase reporters to measure the impact of RNF122 on PRR signaling.

    • Viral infection models: Assess viral replication (using plaque assays or qPCR) in cells with RNF122 overexpression or knockdown.

    • Cytokine profiling: Measure IFN-α/β and proinflammatory cytokines by ELISA or qRT-PCR in different RNF122 expression contexts.

  • Microscopy-based approaches:

    • Perform immunofluorescence with biotin-conjugated RNF122 antibodies and fluorescently labeled PRR antibodies to visualize co-localization.

    • Use proximity ligation assays (PLA) to detect protein-protein interactions with high sensitivity.

    • Implement live-cell imaging to track RNF122-PRR interactions during viral infection .

These methodologies can reveal how RNF122 mediates the degradation of MDA5 through K27-linked and K48-linked ubiquitination, thereby inhibiting interferon production and promoting viral replication .

What strategies can be employed to evaluate the role of RNF122 in viral infection models?

To comprehensively evaluate RNF122's role in viral infection models, implement these research strategies:

  • Genetic manipulation approaches:

    • Gene silencing: Design siRNAs or shRNAs targeting porcine or human RNF122, confirming knockdown efficiency by qRT-PCR and western blotting.

    • Overexpression systems: Generate stable cell lines expressing wild-type RNF122 or catalytically inactive mutants (RING domain mutations).

    • CRISPR/Cas9 gene editing: Create RNF122 knockout cell lines for complete loss-of-function studies.

  • Viral infection assessment:

    • Viral replication kinetics: Measure viral titers at multiple time points post-infection using plaque assays, TCID50, or qPCR for viral genomic RNA.

    • Viral protein expression: Quantify viral proteins by western blotting in RNF122-modulated cells.

    • Visualizing infection: Use fluorescently labeled viruses or immunofluorescence to track viral spread in cell culture.

  • Mechanistic investigations:

    • Protein stability assays: Conduct cycloheximide chase experiments to determine if RNF122 affects the half-life of viral proteins (e.g., nsp4 in PRRSV).

    • Ubiquitination profiling: Immunoprecipitate viral proteins and probe for different ubiquitin linkages (K27, K48, K63) in the presence/absence of RNF122.

    • Signaling pathway analysis: Monitor activation of innate immune signaling molecules (IRF3, NF-κB) by phosphorylation status.

  • Transcriptomic and proteomic approaches:

    • Perform RNA-seq to identify genes differentially regulated during viral infection in RNF122-manipulated cells.

    • Use proteomics to catalog ubiquitinated proteins in infected cells with or without RNF122.

  • Validation in physiologically relevant models:

    • Transition from cell lines to primary cells (e.g., PAMs for PRRSV studies).

    • Consider ex vivo tissue explants or organoid cultures when appropriate .

Research has shown that during PRRSV infection, viral proteins (nsp1α, nsp7, nsp9) cooperatively upregulate RNF122 transcription, which in turn enhances virus replication through dual mechanisms: stabilizing viral nsp4 via K63-linked ubiquitination and promoting degradation of host immune sensor MDA5 via K27/K48-linked ubiquitination .

How can I analyze the promoter activity and transcriptional regulation of RNF122?

To investigate promoter activity and transcriptional regulation of RNF122, implement these specialized methodological approaches:

  • Promoter identification and analysis:

    • Bioinformatic prediction: Analyze the 5' upstream region of RNF122 using promoter prediction tools (JASPAR, TRANSFAC).

    • Promoter cloning: Amplify and clone the putative promoter region (~1000 bp upstream of TSS) into luciferase reporter vectors.

    • Deletion series: Create a series of 5' deletion constructs to map the minimal promoter and regulatory elements.

    • Site-directed mutagenesis: Introduce point mutations in predicted transcription factor binding sites to validate their functionality.

  • Transcription factor binding studies:

    • Chromatin Immunoprecipitation (ChIP): Use antibodies against predicted transcription factors (e.g., HLTF, E2F complex) to immunoprecipitate bound DNA, followed by qPCR for RNF122 promoter regions.

    • Electrophoretic Mobility Shift Assay (EMSA): Detect direct binding of nuclear proteins to labeled RNF122 promoter fragments.

    • DNA-affinity precipitation: Use biotinylated promoter fragments with streptavidin beads to pull down binding proteins, followed by mass spectrometry identification.

  • Functional validation experiments:

    • Luciferase reporter assays: Measure promoter activity under different stimuli (e.g., viral infection, cytokine treatment).

    • Transcription factor overexpression/knockdown: Evaluate how modulating specific transcription factors affects RNF122 expression.

    • Viral protein effects: Assess how viral proteins (e.g., nsp1α, nsp7, nsp9) impact RNF122 promoter activity through reporter assays.

  • Epigenetic regulation assessment:

    • Bisulfite sequencing: Analyze CpG methylation patterns in the RNF122 promoter under different conditions.

    • Chromatin accessibility: Perform ATAC-seq to determine if chromatin structure around the RNF122 promoter changes during infection.

    • Histone modification ChIP: Examine active (H3K4me3, H3K27ac) and repressive (H3K27me3) histone marks at the RNF122 locus .

Research has identified the core promoter of RNF122 between -550 to -470 bp upstream of the transcription start site, which displays significant differential transcriptional activities. Furthermore, transcription factors HLTF (inhibited by viral nsp1α and nsp7) and E2F complex (regulated by viral nsp9) have been shown to modulate RNF122 transcription during viral infection .

How can I address non-specific binding when using biotin-conjugated RNF122 antibodies?

Non-specific binding is a common challenge when working with biotin-conjugated antibodies. Implement these solutions to improve specificity:

  • Blocking optimization strategies:

    • Endogenous biotin blocking: Pre-treat samples with avidin followed by biotin to mask endogenous biotin. Commercial kits (e.g., Vector Laboratories Avidin/Biotin Blocking Kit) are effective for this purpose.

    • Buffer adjustments: Add 0.1-0.3% Triton X-100 or Tween-20 to reduce hydrophobic interactions.

    • Blocking agents: Use 5% BSA rather than milk, as milk contains biotin that can interfere with detection.

    • Carrier proteins: Add 1-5% normal serum from the same species as the secondary reagent to reduce non-specific interactions.

  • Antibody dilution optimization:

    • Perform a dilution series (1:100, 1:250, 1:500, 1:1000) to identify the optimal concentration that provides specific signal with minimal background.

    • Consider a two-step detection protocol: primary antibody followed by streptavidin-conjugated reporter for increased specificity.

  • Validation techniques:

    • Include RNF122 knockdown or knockout samples as negative controls.

    • Perform peptide competition assays using the immunogenic peptide (AA 71-155) to confirm signal specificity.

    • Include isotype controls at the same concentration as the primary antibody.

  • Sample preparation considerations:

    • For tissues with high endogenous biotin (liver, kidney, brain), additional blocking steps are essential.

    • For fixation-sensitive epitopes, optimize fixation conditions and consider antigen retrieval methods that preserve the target epitope .

These strategies can significantly reduce non-specific binding while maintaining detection sensitivity in various applications using biotin-conjugated RNF122 antibodies.

What are the best approaches for detecting low-abundance RNF122 in complex biological samples?

Detecting low-abundance RNF122 in complex samples requires specialized sensitivity-enhancing strategies:

  • Sample enrichment techniques:

    • Subcellular fractionation: Isolate Golgi and ER fractions where RNF122 is concentrated to enhance signal-to-noise ratio.

    • Immunoprecipitation: Perform IP with RNF122 antibodies prior to analysis to concentrate the target protein.

    • Protein concentration methods: Use TCA precipitation or methanol/chloroform extraction to concentrate proteins from dilute samples.

  • Signal amplification methods:

    • Tyramide signal amplification (TSA): This enzyme-mediated reporter deposition technique can increase sensitivity 10-100 fold for immunohistochemistry and immunofluorescence.

    • Rolling circle amplification (RCA): For oligonucleotide-linked detection systems, providing exponential signal enhancement.

    • Multi-layer detection: Employ biotin-conjugated primary followed by streptavidin-HRP, then biotinylated tyramide, and finally streptavidin-fluorophore.

  • Detection system optimization:

    • Enhanced chemiluminescence (ECL) substrates: Use super-signal or femto-signal ECL reagents for western blotting.

    • Fluorescent detection: Consider Quantum dots or near-infrared fluorophores for improved signal-to-background ratio.

    • Extended exposure times: For western blots, use incremental exposure times to capture weak signals without saturating strong signals.

  • Technical considerations:

    • Reduce background: Implement additional washing steps with increased stringency (higher salt concentration or detergent).

    • Block interfering substances: For tissue samples, use commercial biotin-blocking reagents before applying biotin-conjugated antibodies.

    • Minimize protein loss: Reduce sample handling steps and use low-protein-binding tubes .

These approaches can dramatically improve detection of low-abundance RNF122 in complex biological samples while maintaining specificity.

How should I design experiments to study the differential ubiquitination patterns mediated by RNF122?

Designing experiments to study differential ubiquitination patterns mediated by RNF122 requires sophisticated approaches to distinguish between K27, K48, and K63 linkages:

  • Ubiquitin mutant panel design:

    • Lysine-to-arginine mutations: Express ubiquitin constructs with specific lysine mutations (K27R, K48R, K63R) to prevent formation of specific linkages.

    • Single-lysine ubiquitin: Use ubiquitin variants where all lysines except one (K27-only, K48-only, K63-only) are mutated to determine predominant linkage types.

    • Linkage-specific tandem ubiquitin: Express engineered tandem ubiquitin constructs that mimic specific linkages to serve as size markers.

  • Detection strategies for specific linkages:

    • Linkage-specific antibodies: Employ commercially available antibodies specific for K27-, K48- or K63-linked polyubiquitin chains.

    • Mass spectrometry approaches: Use targeted MS/MS to identify and quantify specific ubiquitin branch points and linkages, with heavy isotope-labeled ubiquitin as internal standard.

    • UbiCREST assay: Apply linkage-specific deubiquitinases (DUBs) to cleave specific chain types and analyze the resulting patterns.

  • Substrate-specific analysis protocols:

    • For viral nsp4 K63-linkage study: Co-express Flag-nsp4, HA-ubiquitin, and Myc-RNF122 in cells, immunoprecipitate with anti-Flag, then probe with anti-K63 ubiquitin antibodies.

    • For MDA5 K27/K48-linkage analysis: Similarly co-express components, immunoprecipitate the substrate, and probe with linkage-specific antibodies.

    • Stability correlation: Perform cycloheximide chase experiments in parallel to correlate ubiquitin linkage patterns with protein half-life.

  • Controls and validations:

    • E3 ligase-dead control: Include RNF122 RING domain mutants (C92A/C95A) that lack E3 ligase activity.

    • Proteasome inhibition: Use MG132 (for K48 linkages) or specific inhibitors of non-proteasomal degradation pathways (for K27 linkages).

    • E2 enzyme specificity: Test different E2 conjugating enzymes to determine which partners with RNF122 to form specific linkages .

Research has demonstrated that RNF122 can mediate distinct ubiquitination patterns with different functional outcomes: K63-linked ubiquitination enhances stability (as seen with viral nsp4), while K27/K48-linked ubiquitination typically leads to degradation (as observed with MDA5) .

What controls should be included when studying RNF122's role in immune signaling pathways?

When investigating RNF122's role in immune signaling pathways, incorporate these essential controls to ensure experimental validity:

  • Genetic controls for RNF122 manipulation:

    • Expression controls: Include empty vector controls for overexpression experiments and non-targeting siRNA/shRNA for knockdown studies.

    • Functional mutants: Compare wild-type RNF122 with catalytically inactive mutants (RING domain mutations C92A/C95A) to distinguish between scaffold and enzymatic functions.

    • Rescue experiments: Perform reconstitution with siRNA-resistant RNF122 constructs after knockdown to confirm specificity.

  • Pathway activation controls:

    • Positive controls: Include canonical pathway activators (e.g., poly(I:C) for MDA5, 5'ppp-RNA for RIG-I, LPS for TLR4) at standardized doses.

    • Pathway inhibitors: Use established inhibitors (e.g., BX-795 for TBK1, BAY11-7082 for NF-κB) as negative controls.

    • Temporal controls: Collect samples at multiple time points to capture the dynamic nature of immune signaling.

  • Readout validation controls:

    • Reporter assays: Include pathway-specific positive controls (constitutively active IRF3-5D for IFN-β reporters) and negative controls (dominant-negative constructs).

    • qRT-PCR normalization: Use multiple reference genes (GAPDH, β-actin, 18S rRNA) for accurate normalization of immune gene expression.

    • Protein phosphorylation: Compare total and phospho-specific antibodies for signaling molecules (IRF3, TBK1, IκB) in parallel.

  • Viral infection controls:

    • UV-inactivated virus: To distinguish between effects requiring viral replication versus initial recognition.

    • Timing controls: Evaluate early (0-6h) versus late (24-48h) infection time points to separate direct sensing from secondary effects.

    • MOI titration: Use multiple MOIs to ensure observations are robust across infection conditions.

  • Cell-type controls:

    • Test findings in multiple relevant cell types (e.g., epithelial cells, macrophages, dendritic cells).

    • Include primary cells where possible to validate findings from cell lines .

These comprehensive controls ensure that observed effects are specifically attributable to RNF122's function in immune signaling pathways, minimizing the risk of experimental artifacts or misinterpretation.

How can RNF122 antibodies be used to investigate novel therapeutic targets for viral infections?

RNF122 antibodies can be leveraged in multiple research approaches to identify and validate novel therapeutic targets for viral infections:

  • Target identification strategies:

    • Proximity-based interactome mapping: Use biotin-conjugated RNF122 antibodies for immunoprecipitation followed by mass spectrometry to identify novel interaction partners during viral infection.

    • Co-localization screening: Apply RNF122 antibodies in high-content imaging to identify proteins that co-localize with RNF122 during specific phases of infection.

    • Genetic interaction studies: Combine RNF122 knockdown with genome-wide CRISPR screens to identify synthetic lethal interactions that could represent therapeutic targets.

  • Target validation methodologies:

    • Viral replication dependency: Use RNF122 antibodies to track how modulation of candidate targets affects RNF122 localization, expression, and activity during infection.

    • Ubiquitination profiling: Employ immunoprecipitation with RNF122 antibodies followed by ubiquitin linkage-specific antibodies to determine how candidate targets influence RNF122's ubiquitination patterns.

    • Structure-function analysis: Use epitope mapping with various RNF122 antibodies to identify critical functional domains that interact with potential therapeutic targets.

  • Drug discovery applications:

    • Small molecule screening: Develop high-throughput assays using RNF122 antibodies to identify compounds that disrupt specific RNF122 interactions or functions.

    • Peptide inhibitor design: Use structural information from antibody epitope mapping to design peptides that target critical RNF122 interaction interfaces.

    • Antibody-drug conjugates: Exploit biotin-conjugated RNF122 antibodies for targeted delivery of antiviral compounds to sites of viral replication.

  • Translational research approaches:

    • Biomarker development: Evaluate RNF122 levels or localization patterns as potential biomarkers for viral infection progression or treatment response.

    • Ex vivo validation: Apply RNF122 antibodies in patient-derived samples or organoid cultures to validate targets in physiologically relevant systems.

    • Combination therapy assessment: Use RNF122 antibodies to evaluate how modulation of ubiquitination pathways might synergize with existing antiviral therapies .

Research has established that RNF122 plays critical roles in viral immune evasion through both enhancement of viral protein stability and degradation of host immune sensors. These functions position RNF122 and its interaction network as promising therapeutic targets for disrupting viral replication strategies .

What emerging technologies could enhance the study of RNF122's function in cellular pathways?

Emerging technologies offer unprecedented opportunities to advance our understanding of RNF122's functions in cellular pathways:

  • Advanced imaging technologies:

    • Super-resolution microscopy: Techniques like STORM, PALM, or STED can resolve RNF122 localization with nanometer precision, revealing previously undetectable spatial relationships with interaction partners.

    • Live-cell ubiquitination sensors: FRET-based reporters that detect specific ubiquitin linkage formation in real-time within living cells.

    • Correlative light-electron microscopy (CLEM): Combine fluorescence localization of RNF122 with ultrastructural context from electron microscopy.

  • Genome and proteome editing approaches:

    • Base editors and prime editors: Make precise modifications to RNF122 or substrates without introducing double-strand breaks.

    • Ubiquitin-specific proteomics: Combine CRISPR screens with ubiquitin remnant profiling to systematically identify substrates and regulatory networks.

    • Optogenetic control: Engineer light-responsive RNF122 variants to achieve spatiotemporal control of its E3 ligase activity.

  • Single-cell technologies:

    • Single-cell proteomics: Analyze RNF122 expression and modification states at single-cell resolution to capture heterogeneity in responses.

    • Spatial transcriptomics: Map RNF122 mRNA expression in tissue contexts with spatial information preserved.

    • Multi-modal single-cell analysis: Simultaneously profile transcriptome, proteome, and ubiquitinome in individual cells to construct comprehensive pathway models.

  • Computational and systems biology approaches:

    • Deep learning for interaction prediction: Apply neural networks to predict novel RNF122 substrates or regulators based on sequence and structural features.

    • Network analysis algorithms: Identify key nodes and vulnerabilities in RNF122-centered interaction networks.

    • Molecular dynamics simulations: Model the structural dynamics of RNF122 interactions with E2 enzymes and substrates to guide inhibitor design.

  • Organoid and microphysiological systems:

    • Organ-on-chip models: Study RNF122 function in microfluidic devices that recapitulate tissue architecture and physiology.

    • Patient-derived organoids: Examine RNF122 function in disease-relevant organoid models with genetic diversity .

These emerging technologies promise to reveal new dimensions of RNF122 biology, particularly its context-dependent functions across different cell types, tissues, and disease states.

How can comparative studies between different species advance our understanding of RNF122 function?

Comparative studies across species can provide valuable insights into conserved and divergent aspects of RNF122 function:

  • Evolutionary analysis approaches:

    • Phylogenetic profiling: Construct comprehensive evolutionary trees of RNF122 across species to identify conserved domains and lineage-specific adaptations.

    • Selection pressure analysis: Calculate Ka/Ks ratios to identify regions under positive selection, potentially revealing host-pathogen conflict zones.

    • Ancestral sequence reconstruction: Infer and synthesize ancestral RNF122 proteins to test functional evolution hypotheses.

  • Cross-species experimental strategies:

    • Heterologous expression: Test functionality of RNF122 orthologs from different species in standardized cellular backgrounds to identify species-specific activities.

    • Domain swapping experiments: Create chimeric proteins with domains from different species to map functional conservation and divergence.

    • Cross-reactivity studies: Test existing RNF122 antibodies against multiple species to develop tools for comparative studies.

  • Multi-species infection models:

    • Parallel viral challenge experiments: Compare RNF122 responses to viral infection across human, porcine, and murine systems.

    • Cross-species substrate identification: Determine whether RNF122 targets the same substrates in different species during pathogen challenge.

    • Transgenic humanization approaches: Express human RNF122 in model organisms to test species-specific immune evasion mechanisms.

  • Structural comparative biology:

    • Comparative structural modeling: Generate and compare structural models of RNF122 from different species to identify conserved binding pockets.

    • Binding interface conservation analysis: Map conservation of amino acids at interaction surfaces to predict functional conservation.

    • Co-evolution analysis: Identify co-evolving residues between RNF122 and its binding partners across species.

  • Methodological considerations:

    • Use biotin-conjugated antibodies with predicted cross-reactivity to facilitate comparative studies.

    • Design degenerate primers for conserved regions to clone RNF122 from diverse species.

    • Develop species-specific cell systems to test RNF122 function in native contexts .

Research has shown that porcine RNF122 plays a critical role in PRRSV infection, yet the conservation of this mechanism across species remains to be fully elucidated. The combination of evolutionary analysis with functional studies can reveal how RNF122's role in immune regulation has evolved in response to species-specific pathogen pressures .

What considerations are important when designing RNF122 knockdown or knockout experiments?

Designing effective RNF122 knockdown or knockout experiments requires careful consideration of multiple factors:

  • Target selection and validation strategies:

    • Isoform specificity: Design siRNAs/shRNAs or CRISPR guide RNAs that target all known RNF122 splice variants or specifically target individual isoforms as needed.

    • Off-target prediction: Use bioinformatic tools to predict and minimize off-target effects, particularly for other RING finger domain proteins.

    • Knockdown validation: Confirm RNF122 reduction at both mRNA (qRT-PCR) and protein (western blotting with biotin-conjugated antibodies) levels.

  • Experimental design considerations:

    • Timing optimization: For transient knockdown, determine the optimal timeframe where RNF122 depletion is maximal (typically 24-72h post-transfection).

    • Delivery method selection: Choose appropriate transfection methods based on cell type (lipid-based reagents, electroporation, or viral vectors).

    • Concentration titration: Test multiple siRNA/shRNA concentrations to identify the minimum effective dose that minimizes off-target effects.

  • Control implementation:

    • Rescue experiments: Co-express siRNA-resistant RNF122 to confirm phenotype specificity.

    • Multiple targeting sequences: Use at least 3 independent siRNAs/shRNAs targeting different regions of RNF122 to confirm consistent phenotypes.

    • Scrambled and non-targeting controls: Include proper negative controls with similar GC content and no known targets.

  • For CRISPR/Cas9 knockout approaches:

    • Guide RNA design: Create multiple guide RNAs targeting early exons or critical functional domains (RING finger domain).

    • Clonal isolation vs. pool screening: Consider whether clonal isolation or population analysis is more appropriate based on experimental goals.

    • Verification methods: Confirm gene disruption by sequencing, western blotting, and functional assays.

    • Conditional systems: Consider inducible CRISPR systems (Tet-on/off) for temporal control of knockout.

  • Phenotypic analysis framework:

    • Temporal considerations: Examine both immediate and long-term consequences of RNF122 depletion.

    • Pathway-specific readouts: Measure changes in interferon signaling, viral replication, and protein stability.

    • Compensatory mechanism assessment: Monitor potential upregulation of related E3 ligases that might compensate for RNF122 loss .

Research has shown that RNF122 knockdown can significantly impair viral replication for viruses like PRRSV, while enhancing antiviral immune responses through the preservation of pattern recognition receptors like MDA5. Carefully designed RNF122 depletion experiments are essential for validating its potential as a therapeutic target .

How can RNF122 antibodies contribute to the development of novel diagnostic tools for viral infections?

RNF122 antibodies, particularly biotin-conjugated variants, offer unique opportunities for developing innovative diagnostic approaches for viral infections:

  • Biomarker development strategies:

    • Expression level diagnostics: Develop quantitative assays (ELISA, electrochemiluminescence) using RNF122 antibodies to measure changes in RNF122 levels during viral infection.

    • Modification-specific detection: Generate antibodies that specifically recognize ubiquitinated forms of RNF122 or its substrates as infection-specific biomarkers.

    • Localization pattern analysis: Create imaging-based diagnostics that detect infection-induced changes in RNF122 subcellular distribution.

  • Multiplex diagnostic platforms:

    • Antibody arrays: Incorporate biotin-conjugated RNF122 antibodies in protein microarrays alongside other innate immunity markers.

    • Bead-based multiplexing: Conjugate RNF122 antibodies to coded microbeads for flow cytometry-based multiplex detection.

    • Biosensor integration: Immobilize RNF122 antibodies on electrochemical or optical biosensors for rapid detection systems.

  • Point-of-care testing approaches:

    • Lateral flow assays: Develop rapid tests utilizing gold nanoparticle-conjugated RNF122 antibodies for field-deployable diagnostics.

    • Microfluidic devices: Integrate RNF122 antibodies in microfluidic channels for automated sample processing and detection.

    • Smartphone-based readouts: Couple antibody-based detection with smartphone camera analysis for resource-limited settings.

  • Advanced diagnostic methodologies:

    • Single-cell diagnostics: Use RNF122 antibodies in mass cytometry or imaging mass cytometry to identify infected cells within heterogeneous populations.

    • Exosome-based detection: Capture exosomes from infected cells using RNF122 antibodies to detect infection-related changes in exosomal cargo.

    • Digital PCR coupling: Combine antibody-based cell sorting of RNF122-expressing cells with digital PCR for ultrasensitive viral detection.

  • Clinical implementation considerations:

    • Reference range establishment: Determine normal variation in RNF122 levels across different tissues and demographic groups.

    • Standardization protocols: Develop calibrators and controls for consistent quantification across different testing platforms.

    • Longitudinal monitoring: Evaluate the utility of RNF122 as a prognostic marker by correlating levels with disease progression .

Research has demonstrated that RNF122 expression is significantly modulated during viral infections, particularly by PRRSV non-structural proteins. This virus-induced alteration makes RNF122 a promising biomarker candidate for detecting active infection and monitoring treatment response .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.