BIR2 Antibody refers to an antibody targeting the baculoviral IAP repeat 2 (BIR2) domain of the X-linked inhibitor of apoptosis protein (XIAP). XIAP is a member of the inhibitor of apoptosis (IAP) family, known for regulating caspase activity and apoptosis inhibition . The BIR2 domain (residues 124–240 in humans) is critical for XIAP’s interaction with caspases, E2F1, Sp1, and RIPK2, enabling its roles in apoptosis suppression, immune signaling, and cancer progression .
Apoptosis regulation: BIR2 inhibits caspase-3/7, preventing cell death .
Immune signaling: BIR2-RIPK2 interaction facilitates NOD2-mediated immune responses .
Cancer progression: BIR2 promotes BC invasion via MMP2 activation and Src suppression .
XLP2 syndrome: Mutations in BIR2 (e.g., L207P) impair RIPK2 binding, leading to defective NOD2 signaling and immunodeficiency .
Cancer mechanisms:
| Strategy | Mechanism | Example Compounds/Agents |
|---|---|---|
| SMAC mimetics | Disrupt BIR2-caspase interaction | Compound A (bivalent SMC) |
| PROTACs | Degrade XIAP via BIR2-binding | Experimental PROTACs |
Sample volume: 5 µL
Detection: Competitive FRET assay using GST-tagged BIR2, biotinylated ligand, and anti-GST Terbium cryptate.
Throughput: Compatible with 96-/384-well plates.
Cancer therapy: Targeting BIR2 with SMAC mimetics or PROTACs may overcome chemotherapy resistance .
Immune disorders: Restoring BIR2-RIPK2 interaction could ameliorate NOD2 signaling defects in XLP2 .
Diagnostic potential: BIR2 expression levels may serve as a biomarker for BC aggressiveness .
BIR2/BIRC2 (Baculoviral IAP Repeat Containing 2) is a member of the inhibitor of apoptosis protein (IAP) family that regulates fundamental cell death and survival signaling pathways . It contains multiple BIR (Baculoviral IAP Repeat) domains that mediate protein-protein interactions. BIRC2 plays critical roles in:
Regulation of cell death and apoptosis
Immune signaling pathways, particularly interferon and defense response signaling
Cell-cycle regulation and DNA damage repair processes
Research shows that BIRC2 depletion significantly affects genes involved in interferon response, defense signaling pathways, and cell-cycle regulation as determined by RNA-seq analysis .
When working with BIR2 antibodies, several detection methods have proven effective:
ELISA (Enzyme-Linked Immunosorbent Assay): Sandwich ELISA methods can reliably detect BIRC2 with sensitivity in the pg/mL range (78.125 pg/mL minimum detection limit for some commercial kits)
Western Blotting: Effective for analyzing protein expression levels and post-translational modifications
Immunoprecipitation: Useful for studying protein-protein interactions, particularly between BIR2 and binding partners like RIPK2
Immunohistochemistry: For tissue localization studies
Fluorescence microscopy: For cellular localization studies
The choice of detection method should align with your specific research question. For quantitative detection of BIRC2 in serum, plasma, or tissue homogenates, colorimetric sandwich ELISA provides high sensitivity and excellent specificity .
BIR2 antibodies have been successfully used to analyze multiple sample types:
Serum and plasma samples: Effective for clinical research and biomarker studies
Tissue homogenates: Useful for examining expression in different organs and tissues
Cell lysates: For in vitro studies of protein expression and regulation
Primary cells: Including peripheral blood mononuclear cells (PBMCs) from patients with XIAP mutations
Reconstituted cell lines: Such as XIAP-deficient cell lines used to study BIR2 domain function
Different sample preparation protocols may be required depending on the sample type. For instance, tissue homogenates typically require mechanical disruption and protein extraction buffers, while serum samples may require dilution to minimize matrix effects in assays.
BIR2 antibodies are valuable tools for studying X-linked lymphoproliferative syndrome type-2 (XLP2) caused by mutations in the XIAP BIR2 domain. Methodological approaches include:
Analyzing protein-protein interactions: BIR2 antibodies can help assess how XLP2-associated mutations affect interactions between XIAP and RIPK2. Co-immunoprecipitation experiments have shown that XLP2-BIR2 mutations (including R166I, L207P, and V198M) abolish the XIAP-RIPK2 interaction .
Evaluating ubiquitylation activity: XLP2-BIR2 mutations impair XIAP-mediated ubiquitylation of RIPK2. Using BIR2 antibodies in ubiquitylation assays allows researchers to track how mutations affect this process .
Assessing NOD2 signaling: BIR2 antibodies can be used to investigate how XLP2-BIR2 mutations impair NOD1/2-dependent immune signaling in:
Patient sample analysis: BIR2 antibodies enable comparative studies between healthy donors and XLP2 patients to identify functional defects in NOD2-dependent immune responses .
When investigating BIR2-RIPK2 interactions, consider these methodological approaches:
Co-immunoprecipitation studies: Use BIR2 antibodies to pull down protein complexes and analyze RIPK2 association. Research has shown that the XIAP BIR2 domain is crucial for RIPK2 binding, and XLP2-BIR2 mutations abolish this interaction .
Domain mapping experiments: Utilize truncated constructs to identify specific interaction regions. Studies confirm that the interaction between XIAP and RIPK2 requires the XIAP BIR2 domain and does not involve other domains of XIAP .
Mutational analysis: Introduce specific mutations in the BIR2 domain to identify critical residues for RIPK2 binding. Six XLP2-BIR2 mutations have been shown to abrogate co-purification of endogenous RIPK2 with XIAP .
Functional assays: Measure downstream signaling (e.g., NF-κB activation) to assess the functional consequences of disrupted BIR2-RIPK2 interactions. XIAP XLP2-BIR2 variants are unable to restore NOD2-induced NF-κB activation in XIAP-deficient cells .
Inhibitor studies: Use Smac mimetic compounds (SMCs) to competitively disrupt BIR2-RIPK2 interactions and analyze the functional outcomes .
BIR2 antibodies are valuable tools in cancer immunology research, particularly given BIRC2's role in immune evasion mechanisms. Here's how to approach this research:
Analysis of tumor immune microenvironment:
Knockdown/knockout validation studies:
Combination therapy assessment:
Immune signaling pathway analysis:
To investigate BIR2's involvement in DNA damage repair pathways, consider these methodological approaches:
DNA damage induction and repair kinetics:
Use BIR2 antibodies to track BIRC2 expression and localization during DNA damage response
Induce DNA damage with agents like etoposide and monitor repair over time
Research shows that overexpression of BIRC2 delays DNA damage repair, as evidenced by persistent levels of γH2AX even 6 hours after drug removal
Cell cycle analysis in relation to BIRC2 expression:
Chromatin immunoprecipitation (ChIP):
Co-immunoprecipitation with DNA repair proteins:
Identify BIRC2 interactions with DNA repair machinery components
Map these interactions to specific phases of the DNA damage response
When using BIR2 antibodies in ELISA, follow these optimized protocols for reliable results:
Sample preparation:
For serum/plasma: Dilute samples appropriately in the provided sample dilution buffer
For tissue homogenates: Process tissues in homogenization buffer, centrifuge to remove debris, and quantify protein concentration
Perform all sample preparation steps on ice to prevent protein degradation
Procedure for sandwich ELISA (based on commercial kit protocols):
Quality control measures:
Assay performance characteristics:
For successful immunoprecipitation experiments with BIR2 antibodies, follow these optimization strategies:
Cell lysis conditions:
Use gentle lysis buffers that preserve protein-protein interactions (e.g., RIPA buffer with protease inhibitors)
For nuclear interactions (such as BIRC2-histone H3), include specific nuclear extraction protocols
Optimize detergent concentration to maintain protein interactions while solubilizing membrane proteins
Antibody selection and validation:
Validate antibody specificity using western blotting before immunoprecipitation
For studying specific domains (like BIR2 or BIR3), use domain-specific antibodies
Consider using epitope-tagged constructs when studying mutated versions of BIRC2/XIAP
Control experiments:
Detection of interacting partners:
For known interactions (like BIRC2-RIPK2), use specific antibodies against the partner protein
For novel interactions, consider mass spectrometry analysis of immunoprecipitates
When studying ubiquitylation events, include deubiquitinase inhibitors in lysis buffers
Research has shown that all six XLP2-BIR2 mutations abrogate the co-purification of endogenous RIPK2 with XIAP, while binding to other partners like TAB1 remains unaffected .
BIR2 antibodies are essential tools for evaluating how small molecule inhibitors affect BIRC2 function:
Monitoring target engagement:
Functional outcome assessment:
Cellular localization studies:
Protein degradation analysis:
Combination therapy studies:
When conducting cancer research with BIR2 antibodies, include these essential controls:
Expression level controls:
Functional validation controls:
Antibody validation controls:
Peptide blocking experiments to confirm antibody specificity
Secondary antibody-only controls to identify non-specific binding
Multiple antibodies targeting different epitopes of BIRC2 to confirm results
Treatment response controls:
Patient sample controls:
Age and gender-matched normal samples
Samples spanning different cancer stages and grades
Treatment-naive versus post-treatment samples
Research has shown that knockdown of BIRC2 in mouse melanoma or breast cancer cells increases intratumoral activated CD8+ T cells, providing a methodological framework for studying BIRC2's role in cancer immune evasion .
Non-specific binding is a common challenge when working with antibodies. Here are methodological solutions specifically for BIR2 antibody research:
Optimize blocking conditions:
Test different blocking agents (BSA, non-fat milk, normal serum)
Increase blocking time (1-2 hours at room temperature or overnight at 4°C)
Consider commercial blocking buffers formulated to reduce background
Antibody dilution optimization:
Sample preparation refinement:
Washing protocol optimization:
Cross-reactivity assessment:
When investigating BIR2 domain mutations, particularly in the context of XLP2, consider these methodological approaches:
Mutation selection and validation:
Functional readouts:
Interaction studies:
Design co-immunoprecipitation experiments to assess how mutations affect binding to known partners like RIPK2
Include TAB1 interaction analysis as a control (binds to BIR1 domain, should be unaffected by BIR2 mutations)
Consider proximity ligation assays for in situ detection of protein interactions
Comparative analysis framework:
Research has demonstrated that XLP2-BIR2 mutations severely impair NOD1/2-dependent immune signaling in primary cells from XLP2 patients and in reconstituted XIAP-deficient cell lines .
When faced with contradictory results across different detection methods, use this systematic approach:
Method-specific factors assessment:
ELISA: Consider detection limits (typically 78.125 pg/mL for BIRC2), sample matrix effects, and potential cross-reactivity
Western blotting: Evaluate protein denaturation effects, epitope accessibility, and transfer efficiency
Immunoprecipitation: Assess buffer conditions that might disrupt protein-protein interactions
Antibody characteristics comparison:
Epitope location: Different antibodies may target distinct regions of BIRC2
Clonality: Monoclonal vs. polyclonal antibodies may yield different results
Affinity and specificity: Higher affinity antibodies may detect lower expression levels
Biological context analysis:
Cell/tissue type differences: BIRC2 expression and function varies across tissues
Experimental conditions: Stress, cytokine stimulation, or drug treatments may affect BIRC2 detection
Post-translational modifications: May alter epitope recognition
Validation strategies:
Use multiple antibodies targeting different epitopes
Employ genetic approaches (siRNA, CRISPR) to confirm specificity
Include recombinant BIRC2 protein as a positive control
Data integration approach:
Consider each method's strengths and limitations
Weigh results based on technical quality and reproducibility
Develop a consensus interpretation that accounts for methodological differences
BIR2 antibodies offer powerful tools for investigating BIRC2's role in cancer immunotherapy resistance:
Tumor microenvironment profiling:
Mechanistic studies:
Biomarker development:
Use BIR2 antibodies to assess BIRC2 expression in patient tumor samples before and during immunotherapy
Correlate expression levels with treatment response to determine predictive value
Combination therapy design:
Monitor BIRC2 expression during treatment with BIRC2 inhibitors combined with immune checkpoint inhibitors
Use BIR2 antibodies to confirm target engagement in preclinical studies
Resistance mechanism identification:
Compare BIRC2 expression and localization in immunotherapy-responsive versus resistant tumors
Investigate downstream signaling pathways affected by BIRC2 that contribute to resistance
Research demonstrating that knockdown of BIRC2 expression in mouse melanoma or breast cancer cells impairs tumor growth by increasing intratumoral activated CD8+ T cells provides a foundation for these studies .
Recent advances have enabled more detailed study of BIRC2 BIR3-histone H3 interactions:
Structural analysis techniques:
ChIP-sequencing applications:
Use BIR2 antibodies in ChIP-seq to map genome-wide binding sites of BIRC2
Integrate with histone modification maps to understand the functional context of binding
Correlate with gene expression data to identify regulated genes
CRISPR-based screening:
Deploy CRISPR screens to identify genes that modulate BIRC2-H3 interactions
Use BIR2 antibodies to validate hit genes that affect nuclear localization or function
Live-cell imaging approaches:
Utilize fluorescently tagged BIRC2 constructs combined with H3 markers to visualize interactions in real-time
Apply FRET or BRET techniques to measure protein-protein proximity in living cells
Small molecule intervention:
| Method | Application | Advantages | Limitations |
|---|---|---|---|
| X-ray/Cryo-EM | Structural determination | High resolution | Requires purified proteins |
| NMR spectroscopy | Binding kinetics | Dynamic information | Size limitations |
| ChIP-seq | Genome-wide binding | In vivo relevance | Antibody specificity dependent |
| CRISPR screening | Functional genomics | Unbiased approach | Complex data analysis |
| Live-cell imaging | Real-time dynamics | Physiological context | Potential tag interference |