BRR2C Antibody

Shipped with Ice Packs
In Stock

Description

Molecular Characterization of BRR2C

BRR2C is hypothesized to belong to the Ski2-like family of RNA helicases, characterized by dual helicase cassettes (N- and C-terminal domains). These proteins facilitate RNA structural remodeling, critical for splicing and miRNA biogenesis . Key features include:

  • Domain Architecture: Two ATP-dependent helicase cores (N- and C-helicase cassettes) for RNA unwinding .

  • Conserved Motifs: DEIH (ATPase activity) and SAT (helicase activity) motifs in the N-terminal domain .

  • Post-Translational Modifications: Glycosylation sites in conserved regions, as observed in other immunoglobulin superfamily proteins .

Functional Roles of BRR2C

BRR2C antibodies are tools to study the protein’s involvement in:

  • miRNA Biogenesis: BRR2 homologs (e.g., Brr2a) unwind primary miRNA (pri-miRNA) structures, enabling Microprocessor complex access .

  • Spliceosome Assembly: Helicase activity assists in pre-mRNA splicing by resolving RNA secondary structures .

  • Immune Regulation: Antibody-binding sites on BRR2C may modulate interactions with effector molecules, akin to Fc regions in immunoglobulins .

Antibody Validation and Applications

Validated antibodies against BRR2 homologs demonstrate:

ParameterBrr2a Antibody (Analog)BRD2 Antibody (Control)
Target SpecificityConfirmed via knockout cell lines Validated using BRD2⁻/⁻ HEK293T
Binding AffinityKd=13.3±1.7nMK_d = 13.3 \pm 1.7 \, \text{nM} (N-terminal) Kd=221.2±2.2nMK_d = 221.2 \pm 2.2 \, \text{nM} (C-terminal)
Functional AssaysRIP-qPCR, EMSA, DMS-MaPseq Western blot, ChIP

Key Findings:

  • BRR2C antibodies likely target conformational epitopes in the helicase domains, disrupting ATPase activity .

  • Cross-reactivity with spliceosomal components (e.g., U4/U6 snRNA) has been observed in homologs .

Research Challenges and Gaps

  • Structural Complexity: BRR2C’s dynamic RNA-binding interface complicates antibody design .

  • Validation Standards: Antibody specificity must be confirmed via knockout controls, as false positives persist in commercial reagents .

  • Clinical Relevance: No direct studies link BRR2C antibodies to diseases, though spliceosome dysregulation is implicated in cancers .

Future Directions

  • High-Resolution Epitope Mapping: Cryo-EM or X-ray crystallography to define paratope-CDR interactions .

  • Therapeutic Potential: Engineering BRR2C-blocking antibodies to modulate RNA processing in malignancies .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
BRR2C antibody; At5g61140 antibody; MAF19.14 antibody; DExH-box ATP-dependent RNA helicase DExH14 antibody; EC 3.6.4.13 antibody; BRR2 homolog C antibody; AtBRR2C antibody; Pre-mRNA-splicing helicase BRR2C antibody
Target Names
BRR2C
Uniprot No.

Target Background

Function
BRR2C is an RNA helicase that plays a critical role in pre-mRNA splicing. It functions as a component of the U5 snRNP and U4/U6-U5 tri-snRNP complexes, participating in spliceosome assembly, activation, and disassembly.
Database Links

KEGG: ath:AT5G61140

STRING: 3702.AT5G61140.2

UniGene: At.46196

Protein Families
DExH box helicase family
Subcellular Location
Nucleus.

Q&A

What are the fundamental mechanisms distinguishing bispecific antibodies from conventional monoclonal antibodies?

Bispecific antibodies represent an evolution in antibody engineering, characterized by their ability to simultaneously bind two distinct epitopes, either on the same or different antigens. Unlike conventional monoclonal antibodies that target single epitopes, bispecific antibodies can bridge between different cell types or cellular components, enabling novel therapeutic approaches particularly valuable in cancer immunotherapy and infectious disease treatment . In multiple myeloma treatment, this dual-binding capability allows bispecific antibodies to simultaneously engage cancer cells and immune effector cells, creating a physical link that enhances cytotoxic activity against malignant cells. This mechanism significantly differs from traditional monoclonal antibodies that operate primarily through direct antigen neutralization or complement-dependent cytotoxicity.

What screening and qualification criteria determine patient eligibility for bispecific antibody therapy?

Patient qualification for bispecific antibody therapy typically depends on several clinical parameters. In the context of myeloma treatment, qualification often requires: (1) documentation of previous lines of therapy, as bispecific antibodies are frequently reserved for relapsed/refractory disease; (2) comprehensive screening including cardiac function assessment, neurological evaluation, and infectious disease testing; and (3) evaluation of specific disease characteristics that might influence response or toxicity profiles . Screening typically involves assessments for underlying infections, cytokine release syndrome risk factors, and baseline organ function. Additionally, genetic profiling of the patient's myeloma may be conducted to determine if their specific genetic alterations make them suitable candidates for particular bispecific antibody treatments .

How do researchers distinguish between responders and non-responders to antibody therapy in experimental design?

Researchers employ multiple methodological approaches to differentiate responders from non-responders:

  • Biomarker Profiling: Measuring target antigen expression levels before treatment initiation

  • Immune Cell Enumeration: Quantifying relevant effector cells (e.g., T-cells, NK cells) pre-treatment

  • Cytokine Analysis: Monitoring changes in inflammatory and immune-regulatory cytokines

  • Genetic Analysis: Identifying mutations that may confer resistance

  • Serial Sampling: Collecting specimens at multiple timepoints to track dynamic changes

For bispecific antibodies in myeloma, response assessment typically includes measurement of minimal residual disease, evaluation of circulating plasma cells, and sequential imaging studies to determine depth and durability of response . These multimodal approaches allow for more sophisticated stratification of patient populations beyond simple binary response categories.

What molecular determinants contribute to the neutralization breadth of broadly neutralizing antibodies against viral variants?

The molecular basis for neutralization breadth in broadly neutralizing antibodies (bnAbs) involves several sophisticated structural and biochemical features:

  • Conserved Epitope Targeting: The most effective bnAbs target highly conserved regions that are functionally constrained and less prone to mutation, such as the "silent face" of viral proteins .

  • Structural Flexibility: bnAbs often possess unusually long complementarity-determining regions (CDRs) or unique structural elements that allow them to access recessed, sterically hindered epitopes that are protected from the typical antibody response.

  • Germline Gene Usage: Certain antibody germline genes appear predisposed to developing neutralization breadth. For example, the YYDRxG motif found in certain heavy chain variable regions confers particular advantages in binding conserved viral epitopes .

  • Approach Angle Innovation: As demonstrated in the crystal structures of CC25.54, CC84.24, and CC84.2 antibodies against SARS-CoV-2, certain bnAbs utilize unique approach angles that allow effective neutralization without direct overlap with receptor binding sites .

Research has shown that group 1 and group 2 RBD bnAbs retain neutralizing activity against highly evolved SARS-CoV-2 variants including BA.4/5 and XBB.1.5, with group 2 RBD bnAbs demonstrating particularly robust resistance to escape mutations . This superior breadth derives from their targeting of conserved epitopes that remain relatively unchanged even under significant selective pressure.

How do crystallographic studies inform the rational design of next-generation therapeutic antibodies?

Crystallographic studies provide essential atomic-level insights that drive rational antibody design through several methodological approaches:

  • Epitope Mapping: Crystal structures reveal precise antibody-antigen interfaces, identifying critical contact residues and interaction networks. For example, analysis of CC25.54, CC84.24, and CC84.2 antibodies bound to SARS-CoV-2 RBD at 2.9-3.1Å resolution revealed that these antibodies bind the CR3022 cryptic site using similar approach angles that permit ACE2 competition despite minimal epitope overlap .

  • Paratope Engineering: Structural data enables targeted modifications to antibody binding regions:

AntibodyResolutionBuried Surface AreaLight Chain ContributionHeavy Chain Patterns
CC84.23.0ÅSimilar to othersLarger contributionCDR H3 maintained same contacts
CC25.542.9ÅSimilar to othersVariable contactsCDR H3 maintained same contacts
CC84.243.1ÅSimilar to othersVariable contactsCDR H3 maintained same contacts
  • Germline Impact Analysis: Different germline genes encoding light chains (IGKV3-20 for CC84.2, IGLV3-21 for CC25.54, IGKV1-5 for CC84.24) create distinct interaction patterns while preserving core neutralization capabilities .

  • Conformational Dynamics: Crystal structures capture static snapshots, but analysis of multiple structures helps infer dynamics necessary for binding kinetics optimization.

These methodological approaches demonstrate that structurally-guided antibody design can enhance therapeutic potential by preserving critical binding determinants while modifying framework regions to improve pharmacokinetic properties.

What statistical methodologies are most appropriate for evaluating comparative efficacy in antibody network meta-analyses?

When conducting network meta-analyses of antibody efficacy, researchers should implement rigorous statistical frameworks:

  • Random-Effects Models: These models account for both within-study and between-study heterogeneity, critical when comparing antibodies with different mechanisms and structural features .

  • Consistency Modeling: Evaluating whether direct and indirect evidence yield similar results using node-split analyses and contribution plots. This is particularly important when comparing diverse antibody types (e.g., nirsevimab, motavizumab, palivizumab, and suptavumab) .

  • Absolute Effect Calculation: Converting odds ratios to absolute effects per 1,000 participants provides more clinically interpretable data:

InterventionRSV Infection ReductionHospitalization ReductionICU Admission Reduction
Nirsevimab-123 (-138 to -100)-54 (-64 to -38)Variable
Palivizumab-108 (-127 to -82)-39 (-48 to -28)Variable
Motavizumab-136 (-146 to -125)-48 (-58 to -33)Superior to palivizumab
  • Heterogeneity Assessment: Evaluating heterogeneity using I² statistics and conducting sensitivity analyses, subgroup analyses, and meta-regression to identify potential effect modifiers .

This methodological approach resulted in moderate- to high-certainty evidence for significant reductions in RSV-related infections and hospitalizations with nirsevimab, palivizumab, and motavizumab compared to placebo in a network meta-analysis of 14 randomized clinical trials involving 18,042 participants .

What experimental controls and validation steps are essential when evaluating neutralization potential of novel antibodies?

Rigorous experimental design for antibody neutralization studies requires multiple methodological controls:

  • Isotype Controls: Inclusion of matched isotype controls to distinguish specific from non-specific effects.

  • Cross-Variant Testing: Systematic evaluation against a panel of variants to assess breadth:

    • Historical isolates (establishing baseline activity)

    • Contemporary circulating variants (establishing current relevance)

    • Engineered escape mutants (probing vulnerability to resistance)

  • Multiple Neutralization Assays: Implementation of complementary assays including:

    • Pseudovirus neutralization

    • Live virus neutralization

    • Cell-cell fusion inhibition

    • Antibody-dependent cellular cytotoxicity (ADCC)

  • Concentration-Response Curves: Full dose-response curves rather than single-point measurements to determine IC₅₀ values with appropriate confidence intervals .

For example, in evaluating SARS-CoV-2 bnAbs, researchers systematically tested neutralization against early variants (Alpha, Beta, Gamma, Delta) and multiple Omicron lineage variants (BA.1, BA.2, BA.2.75, BA.4/5, XBB.1.5), enabling quantification of neutralization changes through geometric mean IC₅₀ comparisons . This comprehensive approach revealed that group 2 RBD bnAbs maintained activity against Omicron variants despite showing intrinsically lower neutralization potency compared to group 1 antibodies.

How should clinical trial designs evolve to better assess bispecific antibody efficacy and safety profiles?

Clinical trial designs for bispecific antibodies require specialized considerations beyond conventional monoclonal antibody studies:

  • Adaptive Designs: Implementation of seamless phase 1/2 designs with real-time dose adjustment based on pharmacokinetic/pharmacodynamic modeling and early efficacy signals.

  • Biomarker-Driven Stratification: Prospective patient stratification based on:

    • Target antigen expression levels

    • Immune effector cell counts and functionality

    • Prior therapy exposure patterns

  • Novel Endpoint Selection: Incorporation of endpoints specifically relevant to bispecific mechanisms:

    • T-cell engagement kinetics

    • Cytokine release dynamics

    • Minimal residual disease assessment

  • Specialized Safety Monitoring: Enhanced monitoring for mechanism-specific toxicities including cytokine release syndrome, neurological events, and infections .

  • Comparative Arms: When ethically appropriate, head-to-head comparison with standard monoclonal antibodies or other bispecifics rather than placebo alone .

Researchers considering bispecific antibody clinical trials should evaluate whether to pursue FDA-approved therapies or investigate experimental bispecifics through clinical trials, weighing factors such as specific disease characteristics, prior treatment history, and genetic profiles .

How can researchers reconcile discrepancies between in vitro neutralization potency and in vivo protection for antibody candidates?

Addressing discordance between in vitro and in vivo antibody performance requires systematic methodological approaches:

  • Fc Function Assessment: Quantification of Fc-mediated effects beyond direct neutralization, including ADCC, antibody-dependent cellular phagocytosis (ADCP), and complement activation.

  • Tissue Distribution Analysis: Evaluation of antibody biodistribution to relevant anatomical compartments, particularly for respiratory or neurotropic pathogens.

  • Immune Environment Reconstitution: In vitro systems incorporating relevant immune components:

    • Co-culture with immune effector cells

    • Addition of complement components

    • Physiologically relevant matrices

  • Pharmacokinetic/Pharmacodynamic Modeling: Integration of exposure data with temporal neutralization requirements.

This methodological framework helps explain observations such as those seen with group 2 RBD bnAbs against SARS-CoV-2, which despite showing lower neutralization potency in vitro (IC₅₀ values in the μg/ml range), demonstrated superior resistance to antigenic drift in Omicron variants compared to more potent group 1 antibodies . Such findings highlight the importance of comprehensive assessment beyond simple neutralization potency.

What analytical approaches help distinguish between antibody resistance due to epitope mutations versus allosteric conformational changes?

Distinguishing between direct epitope mutations and allosteric mechanisms of antibody escape requires integrated analytical methodologies:

  • Deep Mutational Scanning: Systematic evaluation of single amino acid substitutions across the entire target protein to identify direct and indirect effects on antibody binding.

  • Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS): Measurement of conformational dynamics and protein breathing that may reveal allosteric mechanisms not apparent in static structures.

  • Cryo-EM Analysis of Alternative Conformations: Visualization of conformational ensembles rather than single states to identify population shifts induced by distant mutations.

  • Computational Molecular Dynamics: Simulation of protein motion and conformational changes using physics-based modeling to predict how distal mutations influence epitope presentation.

  • Correlative Mutation Analysis: Statistical approaches examining co-varying residues that may reveal networks of functionally linked positions.

These methodologies help explain observations such as the differential impact of Omicron mutations on group 1 versus group 2 RBD bnAbs, where group 1 antibodies showed substantial neutralization loss (geometric mean IC₅₀ drop = 14-105-fold) while group 2 antibodies remained relatively resistant (geometric mean IC₅₀ drop = 2-17-fold) .

What emerging technological platforms show promise for developing antibodies with enhanced breadth and potency?

Several advanced technological platforms are revolutionizing antibody discovery and optimization:

  • AI-Driven Antibody Design: Machine learning algorithms trained on antibody-epitope interaction databases to predict mutations that enhance affinity while preserving breadth.

  • Yeast Display Evolution with Deep Sequencing: High-throughput directed evolution coupled with next-generation sequencing to identify rare variants with exceptional properties.

  • Structurally-Guided Germline Targeting: Rational design approaches focusing on antibody germline genes predisposed to breadth, such as those containing the YYDRxG motif in heavy chain CDR3 regions .

  • Multivalent Antibody Engineering: Development of tri-specific or higher-order multivalent antibodies to simultaneously target multiple conserved epitopes, reducing escape potential.

  • Germline-Targeted Immunogen Design: Reverse engineering of immunogens that specifically activate B-cell receptors with genetic features associated with breadth development.

These technological advances offer promising avenues for developing next-generation antibodies against rapidly evolving pathogens and treatment-resistant cancers, potentially yielding therapeutics with substantially improved clinical outcomes compared to current options.

How might sequence-structure-function relationships inform optimization of antibody binding properties?

Leveraging sequence-structure-function relationships for antibody optimization involves several methodological approaches:

  • CDR Grafting and Framework Optimization: Transplanting binding determinants while enhancing stability:

    • Identification of critical binding residues through alanine scanning

    • Incorporation into optimized framework regions with improved stability

    • Retention of key structural elements that maintain appropriate binding geometry

  • Paratope Refinement Based on Structural Insights: Crystal structures of antibodies like CC25.54, CC84.24, and CC84.2 reveal how different germline-encoded light chains (IGKV3-20, IGLV3-21, IGKV1-5) create distinct interaction patterns with antigens while preserving core neutralization capabilities .

  • Somatic Hypermutation Analysis: Studying natural antibody maturation pathways to identify beneficial mutation patterns that could be recapitulated or enhanced through rational design.

  • Framework Compatibility Assessment: Evaluating how different framework regions influence the presentation and flexibility of CDR loops, potentially enhancing breadth through subtle conformational effects.

These approaches demonstrate that understanding the relationship between antibody sequence, three-dimensional structure, and functional properties enables rational optimization strategies that preserve essential binding characteristics while enhancing pharmacokinetics, stability, and manufacturability.

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.