Recombinant Xenopus laevis E3 ubiquitin-protein ligase RING2-B (rnf2-b)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on purchasing method and location. Please consult your local distributor for specific delivery timelines.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires advance notification and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which can be used as a reference.
Shelf Life
Shelf life depends on storage conditions, buffer components, temperature, and protein stability. Generally, liquid form is stable for 6 months at -20°C/-80°C, while lyophilized form is stable for 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. Specify your preferred tag type in advance for prioritized development.
Synonyms
rnf2-b; ring1b-b; E3 ubiquitin-protein ligase RING2-B; EC 2.3.2.27; RING finger protein 1B-B; RING1b-B; RING finger protein 2-B; RING-type E3 ubiquitin transferase RING2-B
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-343
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Xenopus laevis (African clawed frog)
Target Names
rnf2-b
Target Protein Sequence
MATPVTAQCS SKTWELSLYE LHRTPQAIMD GTEIAVSPRS LHSELMCPIC LDMLKNTMTT KECLHRFCSD CIVTALRSGN KECPTCRKKL VSKRSLRPDP NFDALISKIY PSRDEYEAHQ DRVLAKLSRL HNQQALSSSI EEGLKMQAMH RAQRVRKHQH ESDNTTFSGG EDNCDSRSHV SNPSVHSNQE AGPSRKRSRA SEDSGAEPDL SHEGGVRSPD PPGGGENGSE IELVFRAHPL LVEKDGYSQT RYVKTTANAT VDHLSKYLAL RIALEEEALR GGAEGVTVGE VSEKQYTIYI CTGAAGGQYT TLNGSLTLEL VNEKYWKISK PLELYYAPTK EQK
Uniprot No.

Target Background

Function

Recombinant Xenopus laevis E3 ubiquitin-protein ligase RING2-B (rnf2-b) is an E3 ubiquitin-protein ligase that mediates monoubiquitination of lysine 119 of histone H2A (H2AK119Ub), playing a crucial role in histone code and gene regulation. H2AK119Ub serves as a marker for epigenetic transcriptional repression. It is an essential component of Polycomb group (PcG) PRC1-like complexes, which are vital for maintaining the transcriptionally repressive state of numerous genes, including Hox genes, throughout development. The PcG PRC1 complex functions through chromatin remodeling and histone modification, resulting in heritable changes in gene expression.

Database Links

UniGene: Xl.76096

Subcellular Location
Nucleus. Cytoplasm. Chromosome.

Q&A

What is the fundamental role of RNF2-B in Xenopus laevis?

RNF2-B in Xenopus laevis functions as an E3 ubiquitin-protein ligase that catalyzes the transfer of ubiquitin to target proteins, marking them for degradation through the proteasome pathway. As a core member of the Polycomb Repressive Complex 1 (PRC1), RNF2-B plays critical roles in gene silencing and chromatin modification processes during development and cellular differentiation. The protein contains a RING finger domain that is essential for its ubiquitin ligase activity, enabling specific substrate recognition and ubiquitination . In the Xenopus model system, RNF2-B contributes to embryonic development through regulation of multiple signaling pathways and transcriptional networks.

What are the recommended protocols for examining RNF2-B ubiquitination activity in vitro?

To effectively examine RNF2-B ubiquitination activity in vitro, researchers should implement a comprehensive experimental design that includes the following methodological approach:

  • Protein preparation: Express and purify recombinant Xenopus laevis RNF2-B and potential substrate proteins (such as p53) using bacterial or insect cell expression systems.

  • In vitro ubiquitination assay:

    • Combine purified RNF2-B with E1 (ubiquitin-activating enzyme), E2 (ubiquitin-conjugating enzyme), ATP, ubiquitin, and the substrate protein

    • Incubate at 30°C for 1-2 hours

    • Analyze by SDS-PAGE and western blotting using anti-ubiquitin antibodies

  • Controls and variables:

    ComponentExperimental GroupNegative ControlPositive Control
    E1 enzymePresentPresentPresent
    E2 enzymePresentPresentPresent
    RNF2-BPresentAbsentPresent
    SubstratePresentPresentPresent
    ATPPresentPresentPresent
    Bmi1PresentPresentAbsent
  • Activity verification: Confirm the requirement of Bmi1 for optimal RNF2-B E3 ligase activity through inclusion and exclusion experiments, as research has demonstrated that RNF2 E3 ligase activity requires Bmi1 protein interaction .

  • Mutational analysis: Generate RING domain mutants to validate the critical residues for catalytic activity through site-directed mutagenesis.

This approach allows for quantitative analysis of ubiquitination activity while establishing the specificity of the enzyme-substrate relationship.

How can researchers effectively knockdown RNF2-B expression in Xenopus models for functional studies?

Implementing effective RNF2-B knockdown in Xenopus models requires a multi-faceted approach:

  • Antisense morpholino oligonucleotides (MOs):

    • Design translation-blocking MOs targeting the start codon region

    • Design splice-blocking MOs targeting exon-intron boundaries

    • Inject 5-20 ng of MO into one or both blastomeres at the 1-2 cell stage

    • Include control MOs with 5 nucleotide mismatches for specificity validation

  • CRISPR/Cas9 genome editing:

    • Design sgRNAs targeting conserved regions of RNF2-B

    • Inject Cas9 protein (500 pg) with sgRNA (200-300 pg) into fertilized eggs

    • Confirm editing efficiency through T7 endonuclease assay or direct sequencing

    • Establish F0 or F1 knockout lines for extended studies

  • Validation of knockdown efficiency:

    • Quantitative RT-PCR for transcript levels

    • Western blotting for protein expression

    • Immunohistochemistry for spatial distribution analysis

  • Rescue experiments:

    • Co-inject mRNA encoding RNF2-B (resistant to MO or CRISPR targeting)

    • Assess phenotypic rescue to confirm specificity of knockdown effects

  • Phenotypic analysis:

    • Document developmental abnormalities

    • Analyze affected pathways through target gene expression

    • Evaluate cell cycle progression and apoptosis rates

This methodological framework allows for comprehensive functional analysis while adhering to the experimental design principles of proper controls, variable isolation, and quantifiable outcomes .

What evidence supports RNF2-B's role in p53 regulation?

Substantial evidence supports RNF2-B's role in p53 regulation through direct ubiquitination and proteasomal degradation:

  • Direct binding interaction: Research has demonstrated that RNF2 physically interacts with p53, binding to multiple regions within the DNA binding domain of p53. This interaction occurs with both wild-type and mutated forms of p53 .

  • Ubiquitination activity: RNF2 has been shown to ubiquitinate p53, targeting it for degradation through the proteasome pathway. This activity requires the presence of Bmi1 protein, a component of the polycomb group (PcG) complex .

  • Cell-type specificity: Unlike other E3 ligases such as MDM2 that universally regulate p53, RNF2 exhibits selective activity in specific cell types, particularly in germ-cell tumors and ovarian cancer cells. This specificity suggests a tissue-dependent regulatory mechanism .

  • Functional consequences: Knockdown of RNF2 leads to increased p53 levels, resulting in enhanced p53-mediated cellular responses including cell cycle arrest and apoptosis. These effects can be rescued through simultaneous reduction of p53 expression, confirming the causal relationship .

  • Clinical correlation: Analysis of human ovarian cancer tissues revealed a reverse correlation between RNF2 and p53 expression, with high RNF2 corresponding to low p53 levels in approximately 60% of ovarian serous cystoadenomas and 90% of ovarian mucinous carcinomas .

This multi-layered evidence establishes RNF2-B as a contextually important regulator of p53 stability and function, particularly in reproductive system cancers.

How can researchers distinguish between RNF2-B and other E3 ubiquitin ligases in p53 regulation studies?

To distinguish between RNF2-B and other E3 ubiquitin ligases (such as MDM2) in p53 regulation studies, researchers should implement the following methodological approach:

  • Cell-type specific analysis:

    • Conduct comparative studies across diverse cell types as RNF2 selectively degrades p53 in specific cell lines (like germ-cell tumors), while MDM2 exhibits more universal activity

    • Create a systematic panel of cell lines to identify RNF2-B-dependent versus MDM2-dependent regulation

  • Co-factor dependency assessment:

    • Evaluate Bmi1 requirement through knockdown or inhibition experiments

    • RNF2 requires Bmi1 interaction for optimal E3 ligase activity, providing a distinguishing characteristic

  • Subcellular localization studies:

    • Perform fractionation experiments and immunofluorescence to determine where p53 degradation occurs

    • RNF2 primarily targets nuclear p53 for degradation in specific cell types

  • Domain-specific interactions:

    • Map the binding regions through deletion constructs and co-immunoprecipitation

    • Identify unique interaction sites that differentiate between E3 ligases

  • Response to cellular stress:

    • Compare p53 stability in response to different cellular stresses

    • Analyze whether RNF2-B and other E3 ligases respond differently to specific stimuli

  • Quantitative contribution analysis:

    E3 LigaseCell Typep53 Half-life InfluenceCo-factor RequirementsSubcellular Activity
    RNF2-BGerm-cell tumorsMajor decreaseBmi1-dependentNuclear
    RNF2-BOvarian tumorsMajor decreaseBmi1-dependentNuclear
    RNF2-BNon-reproductive tissueMinimal effectBmi1-dependentNuclear
    MDM2UniversalMajor decreasep300-enhancedCytoplasmic and nuclear

This differentiated approach allows researchers to delineate the unique contributions of RNF2-B to p53 regulation, particularly in reproductive system cancers where it appears to play a specialized role compared to more ubiquitous E3 ligases.

What is the mechanism through which RNF2-B influences Wnt/β-catenin signaling?

RNF2-B influences Wnt/β-catenin signaling through a specific mechanism involving TCF7L1 (also known as TCF3) regulation:

  • TCF7L1 destabilization: Upon activation of Wnt signaling, RNF2 promotes the degradation of TCF7L1, a transcriptional repressor, through ubiquitination. This occurs through direct interaction between RNF2 and TCF7L1, marking the latter for proteasomal degradation .

  • Threshold regulation: RNF2 affects the threshold for Wnt activation by modulating TCF7L1 levels. When RNF2 is knocked down, there is stabilization of TCF7L1, which raises the threshold required for effective Wnt signaling activation .

  • Nuclear localization: The regulatory interaction occurs primarily in the nucleus, where RNF2 targets TCF7L1. Immunocytochemistry assays have demonstrated that nuclear TCF7L1 is stabilized following RNF2 knockdown .

  • Downstream target gene regulation: RNF2 knockdown leads to maintained high levels of TCF7L1, resulting in decreased transcription of Wnt target genes including c-myc, Axin2, TCF1, and LEF1. This has been validated through both luciferase reporter assays and RT-PCR analysis of target gene expression .

  • β-catenin interaction: The expression of active β-catenin (ABC) is decreased in stable RNF2 knockdown cells compared to control cells, suggesting that RNF2 modulates not only TCF7L1 levels but also potentially impacts active β-catenin pools .

This mechanism establishes RNF2-B as an important regulator of the Wnt signaling pathway, functioning through control of the TCF7L1 repressor, which has significant implications for understanding developmental processes and cancer progression where Wnt signaling plays crucial roles.

How can researchers experimentally validate the interaction between RNF2-B and TCF7L1 in Xenopus models?

To experimentally validate the interaction between RNF2-B and TCF7L1 in Xenopus models, researchers should implement the following comprehensive methodological workflow:

  • Co-immunoprecipitation (Co-IP) studies:

    • Generate antibodies specific to Xenopus RNF2-B and TCF7L1 or use epitope-tagged constructs

    • Perform reciprocal Co-IPs from Xenopus embryo lysates at different developmental stages

    • Include appropriate negative controls (IgG, unrelated proteins) and positive controls

    • Analyze by western blotting to detect protein-protein interactions

  • Proximity ligation assay (PLA):

    • Perform PLA in Xenopus tissue sections to visualize endogenous protein interactions

    • Quantify interaction signals across different developmental stages and tissue types

    • Compare wild-type and Wnt-activated conditions to assess signal modulation

  • Bimolecular Fluorescence Complementation (BiFC):

    • Generate fusion constructs of RNF2-B and TCF7L1 with split fluorescent protein fragments

    • Inject mRNA encoding these constructs into Xenopus embryos

    • Visualize interaction through fluorescence microscopy in living embryos

    • Map interaction domains through deletion construct analysis

  • In vitro ubiquitination assays:

    • Express and purify recombinant Xenopus RNF2-B and TCF7L1

    • Conduct in vitro ubiquitination assays to demonstrate direct modification

    • Perform mass spectrometry to identify specific ubiquitination sites on TCF7L1

  • Functional validation through rescue experiments:

    Experimental ConditionRNF2-B StatusTCF7L1 ConstructExpected Outcome
    ControlEndogenousEndogenousNormal Wnt response
    RNF2-B knockdownDepletedEndogenousElevated TCF7L1, reduced Wnt response
    RNF2-B knockdown + rescueRestoredEndogenousNormalized TCF7L1, restored Wnt response
    RNF2-B knockdownDepletedUbiquitination-resistant mutantPersistent high TCF7L1, blocked Wnt response
  • Developmental context analysis:

    • Analyze interaction dynamics during key developmental transitions

    • Correlate with Wnt pathway activation patterns

    • Map spatial distribution through whole-mount immunostaining

This multi-faceted approach provides robust validation of the RNF2-B/TCF7L1 interaction through complementary techniques, establishing both the physical association and functional significance of this regulatory relationship in Xenopus development.

How does RNF2-B overexpression contribute to cancer progression?

RNF2-B overexpression contributes to cancer progression through multiple interconnected mechanisms:

  • p53 degradation and tumor suppressor inactivation: RNF2 directly targets p53 for ubiquitination and proteasomal degradation in specific cancer types. This negative regulation of p53 inhibits its tumor suppressor functions, including cell cycle arrest and apoptosis induction. Studies have demonstrated that RNF2 knockdown restores p53 levels, leading to increased apoptosis and reduced tumor growth .

  • Tissue-specific oncogenic activity: RNF2 shows increased expression in various tumor types compared to normal tissue counterparts. Research has identified particularly high RNF2 expression in reproductive system cancers, with upregulation in 88% of ovarian serous cystoadenomas, 90% of ovarian mucinous carcinomas, and 60% of clear cell carcinomas . This tissue-specific pattern suggests contextual oncogenic functions.

  • Cell cycle dysregulation: RNF2 overexpression contributes to bypassing cell cycle checkpoints. When RNF2 is depleted in cancer cells, G1 arrest increases significantly (28% G0/G1 in RNF2 shRNA-transfected cells compared with 17% G0/G1 in control cells), indicating its role in promoting cell cycle progression .

  • Anti-apoptotic effects: Cancer cells with high RNF2 levels show resistance to apoptosis. Terminal transferase dUTP nick-end labeling (TUNEL) and Annexin V staining have demonstrated that RNF2 depletion significantly increases apoptosis in cancer cells, with this effect being p53-dependent .

  • Wnt signaling modulation: RNF2 promotes Wnt pathway activation through degradation of the TCF7L1 repressor. This contributes to increased expression of Wnt target genes involved in proliferation and stemness, including c-myc, Axin2, TCF1, and LEF1 .

  • Clinical correlation with poor outcomes: The reverse correlation between high RNF2 expression and low p53 levels in human ovarian cancer tissues suggests a mechanistic link to cancer progression and potentially treatment resistance .

This multifaceted contribution to cancer biology positions RNF2-B as both a potential biomarker and therapeutic target, particularly in reproductive system malignancies where its overexpression appears most prevalent and functionally significant.

What experimental approaches can evaluate RNF2-B as a therapeutic target in cancer models?

To evaluate RNF2-B as a therapeutic target in cancer models, researchers should implement a comprehensive experimental strategy that addresses efficacy, specificity, and translational potential:

  • Target validation in vitro:

    • Generate stable RNF2-B knockdown cancer cell lines using shRNA or CRISPR/Cas9

    • Compare cell proliferation, colony formation, cell cycle progression, and apoptosis rates between knockout and control cells

    • Perform rescue experiments with wild-type or catalytically inactive RNF2-B to confirm specificity

    • Evaluate p53 and TCF7L1 levels as markers of target engagement

  • Small molecule inhibitor development and testing:

    • Design or screen for compounds targeting the RING domain or Bmi1 interaction interface

    • Assess biochemical inhibition of E3 ligase activity using in vitro ubiquitination assays

    • Evaluate cellular efficacy through monitoring of p53 and TCF7L1 stabilization

    • Determine selectivity profiles against other RING-type E3 ligases

  • Combination therapy approaches:

    Therapeutic ApproachMechanismExpected OutcomePotential Biomarkers
    RNF2-B inhibitor + DNA damaging agentsEnhance p53 responseIncreased apoptosisp53 levels, PUMA, p21
    RNF2-B inhibitor + Wnt inhibitorsDual pathway blockadeReduced stemnessAxin2, LEF1, TCF1
    RNF2-B inhibitor + Immune checkpoint inhibitorsEnhance immune responseIncreased T-cell infiltrationPD-L1, CD8+ T-cells
  • In vivo efficacy studies:

    • Generate xenograft models using RNF2-B overexpressing cancer cells

    • Implement genetic approaches (inducible shRNA) or pharmacological inhibition

    • Monitor tumor growth using volumetric measurements (V = L × W² /2)

    • Assess mechanism of action through immunohistochemistry and molecular analyses of tumor samples

  • Patient-derived xenograft (PDX) models:

    • Establish PDX models from cancer types with high RNF2-B expression

    • Correlate RNF2-B expression levels with treatment response

    • Identify potential predictive biomarkers for patient stratification

  • Translational biomarker development:

    • Develop IHC protocols for RNF2-B, p53, and TCF7L1 detection in patient samples

    • Correlate expression patterns with clinical outcomes

    • Establish cutoff values for potential patient selection in clinical trials

This integrated approach provides a robust framework for evaluating RNF2-B as a therapeutic target, from mechanistic validation to translational applications, with particular emphasis on reproductive system cancers where RNF2-B overexpression has been most clearly documented.

How can Xenopus laevis RNF2-B studies inform human cancer research?

Xenopus laevis RNF2-B studies provide several unique advantages that can significantly inform human cancer research:

  • Evolutionary conservation analysis: Comparing the structure and function of Xenopus RNF2-B with human RNF2 reveals evolutionarily conserved domains and mechanisms. With the Xenopus laevis genome containing 44,456 genes including 34,476 protein-coding genes , comparative genomics approaches can identify core functional elements preserved across species that are likely essential for cancer-related processes.

  • Developmental context elucidation: Xenopus embryos offer an accessible system to study RNF2-B function during normal development, providing insights into how its dysregulation contributes to oncogenesis. The transparent nature of embryos allows visualization of signaling pathway dynamics in real-time, particularly for Wnt signaling which is frequently dysregulated in human cancers.

  • Rapid functional validation: Xenopus embryos enable efficient functional studies through microinjection of morpholinos or CRISPR components, allowing rapid assessment of gene function modifications. This system can validate potential cancer-related interactions, such as the RNF2-B regulation of TCF7L1 in Wnt signaling , before pursuing more resource-intensive mammalian models.

  • Pathway interaction mapping: The Xenopus system facilitates comprehensive analysis of RNF2-B interactions with multiple pathways simultaneously. Studies have demonstrated that RNF2 affects both p53-mediated apoptosis and Wnt/β-catenin signaling , providing a platform to understand how these pathways intersect in cancer progression.

  • Drug screening applications: Xenopus embryos can be utilized for initial screening of compounds targeting RNF2-B activity, offering a whole-organism context for assessing both efficacy and toxicity before advancing to mammalian models.

  • Tissue-specific regulation insights: The tissue-specific activity of RNF2 in degrading p53, particularly in reproductive system cancers , can be examined developmentally in Xenopus to understand the underlying regulatory mechanisms of this selectivity.

By leveraging these unique advantages of the Xenopus model system, researchers can gain fundamental insights into RNF2-B biology that directly inform and accelerate human cancer research, particularly for reproductive system malignancies where RNF2 overexpression has been most clearly documented.

What are the current challenges and future directions in RNF2-B research?

Current challenges and future directions in RNF2-B research span multiple dimensions from basic molecular mechanisms to translational applications:

  • Substrate specificity determination:

    • Current challenge: Beyond p53 and TCF7L1 , the complete substrate repertoire of RNF2-B remains poorly characterized, limiting our understanding of its broader cellular functions.

    • Future direction: Implementation of proteome-wide approaches such as ubiquitin remnant profiling (K-ε-GG) in RNF2-B overexpression and knockout models to systematically identify the substrate landscape.

  • Tissue-specific regulatory mechanisms:

    • Current challenge: The basis for RNF2-B's selective activity in specific tissues, particularly reproductive system cancers, remains incompletely understood.

    • Future direction: Single-cell analysis of RNF2-B activity across diverse tissues to map regulatory networks and cofactor availability that dictate context-dependent functions.

  • Functional redundancy with other E3 ligases:

    • Current challenge: Overlapping functions between RNF2-B and other E3 ligases (like MDM2 for p53 regulation) complicate targeted therapeutic development.

    • Future direction: Comprehensive comparative studies using CRISPR screens to identify synthetic lethal interactions that could be exploited for cancer-specific targeting.

  • Therapeutic targeting approaches:

    • Current challenge: Developing selective inhibitors of RNF2-B that don't affect other RING-domain E3 ligases remains difficult due to structural similarities.

    • Future direction: Focus on disrupting specific protein-protein interactions, particularly the RNF2-Bmi1 interface that is required for optimal E3 ligase activity .

  • Integrated pathway modeling:

    Research AreaCurrent UnderstandingKnowledge GapFuture Approach
    p53 regulationDirect ubiquitinationContext dependencySystems biology modeling
    Wnt signalingTCF7L1 degradationIntegration with other pathwaysMulti-omics integration
    Cancer progressionCorrelation with outcomesCausal mechanismsLongitudinal patient studies
    Drug resistanceLimited dataPotential contributionResistant model development
  • Clinical biomarker development:

    • Current challenge: While RNF2-B overexpression correlates with cancer progression, standardized assessment methods for patient stratification are lacking.

    • Future direction: Development and validation of clinical assays for RNF2-B expression and activity that can guide therapeutic decisions.

  • Model system limitations:

    • Current challenge: Translating findings between Xenopus laevis and human systems requires careful validation due to species differences.

    • Future direction: Development of humanized Xenopus models through targeted replacement of key regulatory elements to enhance translational relevance.

These multifaceted challenges and directions highlight the complexity of RNF2-B biology while pointing toward integrated approaches that combine basic mechanistic studies with translational applications to fully leverage this protein's potential as both a cancer biomarker and therapeutic target.

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 2024 Thebiotek. All Rights Reserved.