The term "BRG3" can refer to different entities depending on the context. In some studies, BRG3 is associated with a cell line used in cancer research, while in others, it refers to an E3 ligase involved in plant biology.
The BRG3 cell line is mentioned in research related to glioma and antiangiogenic therapy. Studies have shown that targeting β1 integrin in BRG3 cells can significantly impact their adhesion, migration, and proliferation capabilities, which are crucial for tumor growth and resistance to certain therapies .
Effect | Description |
---|---|
Adhesion | Reduced adhesion to extracellular matrix (ECM) |
Migration | Decreased mesenchymal-type migration |
Proliferation | Attenuated proliferation in vitro |
Tumor Growth | Impaired growth in vivo |
In plant biology, BRG3 refers to an E3 ligase involved in the regulation of tomato ripening. Persulfidation of BRG3 affects its ubiquitination activity, impacting the transcription of genes related to ripening .
Mechanism | Effect on Ripening |
---|---|
Persulfidation of BRG3 | Reduces ubiquitination activity |
Interaction with WRKY71 | Enhances WRKY71 binding to CAS1 promoter |
Outcome | Delays tomato ripening |
While there is no specific information on a "BRG3 Antibody," antibodies are widely used in research for targeting specific proteins or cell lines. For example, antibodies against β1 integrin have been used to study its role in tumor growth and resistance . Similarly, humanized antibodies are developed for therapeutic purposes, such as targeting BAG3 in pancreatic cancer .
Target | Disease | Antibody Type |
---|---|---|
β1 Integrin | Glioma | Monoclonal |
BAG3 | Pancreatic Cancer | Humanized |
This antibody targets a probable E3 ubiquitin-protein ligase. It does not affect the stability of DELLA proteins.
Gene Function References:
BTG3 (B-cell translocation gene 3) is a member of the BTG/TOB family of antiproliferative proteins. Its primary function involves regulating cell cycle progression, specifically by impeding the transition from G0/G1 to S phase when overexpressed . The protein is ubiquitously expressed in human tissues, with notably higher expression observed in the ventricular zone of the developing central nervous system, as well as in reproductive and lymphoid tissues including ovary, testis, prostate, thymus, and lung . BTG3 functions as a negative regulator of cell proliferation, potentially playing important roles in development, differentiation, and tumor suppression. The protein is known by several alternative names including "abundant in neuroepithelium area protein" (ANA) and protein Tob5, reflecting its varied tissue distribution and biological roles .
Despite similar abbreviations that sometimes cause confusion, BTG3 and BRG3 are distinct proteins with different functions:
Feature | BTG3 | BRG3 |
---|---|---|
Full name | B-cell translocation gene 3 | BOI-related E3 ubiquitin-protein ligase 3 |
Primary function | Cell cycle regulation (G0/G1 to S phase transition) | E3 ubiquitin ligase activity |
Modification | Various post-translational modifications | Undergoes persulfidation at Cys 206 and Cys 212 |
Functional effect | Impairs cell cycle progression when overexpressed | Persulfidation reduces ubiquitination activity |
Research context | Cell proliferation, cancer research | Plant biology (e.g., tomato ripening) |
UniProt ID | Human: Q14201, Mouse: P50615 | Not specified in search results |
BRG3 has been studied in plant biology contexts, particularly regarding its role in tomato ripening processes. Research has shown that BRG3 can undergo persulfidation (addition of sulfhydryl groups) at specific cysteine residues (Cys 206 and Cys 212), which reduces its ubiquitination activity and affects its interactions with transcription factors like WRKY71 . This mechanism plays a role in delaying tomato fruit ripening through complex regulatory pathways involving protein-protein interactions and transcriptional regulation.
BTG3 antibodies serve various research applications, with the most common being:
Western Blotting (WB): BTG3 antibodies are frequently used at dilutions of approximately 1:1000 for detecting endogenous levels of BTG3 protein in cell and tissue lysates . This application allows researchers to determine protein expression levels and validate knockout or overexpression models.
Immunohistochemistry-Paraffin (IHC-P): At dilutions ranging from 1:10 to 1:50, BTG3 antibodies can visualize the spatial distribution of BTG3 protein in fixed, paraffin-embedded tissue sections . This application is particularly valuable for studying expression patterns in developmental contexts or disease states.
Flow Cytometry: Though less commonly referenced in the provided materials, properly validated BTG3 antibodies can be used to measure BTG3 expression in individual cells within heterogeneous populations .
Mechanistic Studies: BTG3 antibodies are essential tools for investigating the protein's role in cell cycle regulation, particularly regarding the G0/G1 to S phase transition .
Protein Interaction Studies: Through techniques like co-immunoprecipitation, BTG3 antibodies help identify binding partners and protein complexes that regulate or are regulated by BTG3.
The choice of application should determine which specific BTG3 antibody format is selected, as not all antibodies perform equally across different experimental platforms.
Validation of BTG3 antibodies follows comprehensive protocols aligned with the "five pillars" recommendations from the International Working Group for Antibody Validation (IWGAV), addressing the broader scientific reproducibility challenges . Proper validation typically includes:
Genetic Strategies:
CRISPR-Cas9 gene editing: Creating BTG3 knockouts to confirm antibody specificity
siRNA knockdown: Reducing BTG3 expression to verify corresponding signal reduction
Independent Antibody Validation: Using different antibodies targeting distinct epitopes of BTG3 to confirm consistent results
Orthogonal Validation: Correlating protein detection with mRNA expression data
Expression Validation: Testing in cell lines with known BTG3 expression profiles (e.g., HeLa cells, which are documented to express BTG3)
Biochemical Validation:
Immunoprecipitation followed by mass spectrometry
Western blotting to confirm expected molecular weight
Application-Specific Validation: For flow cytometry applications, testing on relevant cell lines or primary cells known to express BTG3 under appropriate conditions
This multi-faceted approach ensures that antibodies specifically recognize BTG3 protein without cross-reactivity to other proteins, thereby generating reliable and reproducible research results.
Proper storage of BTG3 antibodies is crucial for maintaining their specificity and activity over time. Based on manufacturer recommendations, researchers should follow these guidelines:
Short-term Storage: Maintain refrigerated at 2-8°C for up to 2 weeks . This temperature range prevents protein denaturation while allowing convenient access for ongoing experiments.
Long-term Storage: Store at -20°C in small aliquots to prevent freeze-thaw cycles . Repeated freezing and thawing significantly diminishes antibody activity and specificity over time.
Aliquoting Strategy: Upon receipt, divide the antibody into small single-use aliquots before freezing to minimize freeze-thaw cycles. This practice is particularly important for polyclonal antibodies, such as the rabbit polyclonal BTG3 antibodies described in the search results.
Buffer Considerations: Most commercial BTG3 antibodies are supplied in phosphate-buffered saline (PBS) with 0.09% (W/V) sodium azide as a preservative . This formulation helps maintain antibody stability during storage.
Reconstitution Practices: For lyophilized antibodies, reconstitute according to manufacturer instructions immediately prior to use, and store any unused reconstituted antibody following the guidelines above.
Adhering to these storage practices ensures optimal antibody performance and contributes significantly to experimental reproducibility across different research projects and timeframes.
Ensuring antibody specificity in complex tissue samples requires a multi-faceted approach that combines careful experimental design with appropriate controls:
Epitope Mapping Validation: Select antibodies targeting well-characterized epitopes in the central region of human BTG3 (amino acids 100-129) . This region tends to have lower homology with related proteins, reducing cross-reactivity concerns.
Cross-Species Reactivity Assessment: When working with non-human samples, confirm whether the BTG3 antibody has been validated for your species of interest. Most commercial BTG3 antibodies are optimized for human samples, with predicted homology-based reactivity in other species requiring empirical validation .
Multi-method Verification: Apply complementary techniques to confirm findings:
Perform protein detection using antibodies recognizing different BTG3 epitopes
Correlate protein expression with mRNA levels (RT-qPCR or in situ hybridization)
Use mass spectrometry to verify pulled-down proteins from immunoprecipitation
Negative Controls: Include tissue samples where BTG3 expression is known to be absent or minimal, or tissues from BTG3 knockout models if available.
Absorption Controls: Pre-incubate the antibody with recombinant BTG3 protein or the immunizing peptide prior to staining to confirm that signal elimination occurs when the antibody binding sites are occupied.
Isotype Controls: Use matched isotype control antibodies at the same concentration to identify non-specific binding.
Dilution Optimization: Perform careful titration experiments to identify the optimal antibody concentration that maximizes specific signal while minimizing background, particularly for immunohistochemistry applications where tissue autofluorescence can be problematic.
By implementing these rigorous controls, researchers can generate more reliable data when using BTG3 antibodies in complex tissue environments where multiple potential cross-reactive targets may be present.
BTG3's role in cell cycle regulation involves several molecular mechanisms that collectively impair G0/G1 to S phase progression:
Interaction with Cell Cycle Regulators: BTG3 interacts with critical cell cycle machinery, particularly components involved in the G1/S transition. These interactions prevent the formation of active complexes needed for S phase entry.
CDK Inhibition: Evidence suggests BTG3 can directly or indirectly influence cyclin-dependent kinase (CDK) activity, potentially through:
Physical association with CDK/cyclin complexes
Modulation of CDK inhibitors (CKIs) like p21 and p27
Alteration of CDK phosphorylation status
E2F Transcription Factor Regulation: BTG3 appears to impact the E2F family of transcription factors, which are crucial for activating genes required for DNA synthesis and S phase entry. This regulation may occur through:
Direct protein-protein interactions
Influencing upstream regulators like Rb (Retinoblastoma protein)
Affecting post-translational modifications of E2F proteins
Cellular Localization Dynamics: The nuclear-cytoplasmic shuttling of BTG3 may be regulated in a cell cycle-dependent manner, with nuclear localization coinciding with its antiproliferative effects.
To study these mechanisms, researchers employ BTG3 antibodies in various experimental approaches:
Experimental Approach | Application of BTG3 Antibodies | Key Insights Gained |
---|---|---|
Co-immunoprecipitation | Pull down BTG3 and identify interacting proteins | Maps BTG3's protein interaction network |
Chromatin immunoprecipitation (ChIP) | Detect BTG3 association with chromatin | Identifies genomic regions affected by BTG3 |
Immunofluorescence | Visualize BTG3 subcellular localization | Determines spatial dynamics during cell cycle |
Flow cytometry | Correlate BTG3 levels with cell cycle phases | Links expression patterns to cell cycle position |
Western blotting | Monitor BTG3 expression in synchronized cells | Establishes temporal dynamics during cell cycle |
Understanding these molecular mechanisms has significant implications for cancer research and developmental biology, given BTG3's expression in the ventricular zone of the developing central nervous system and its potential tumor suppressor functions .
Robust flow cytometry experiments with BTG3 antibodies require comprehensive controls to ensure validity and interpretability of results:
Antibody Validation Controls:
Positive Cell Control: Include cell lines with documented BTG3 expression (such as HeLa cells) to confirm antibody performance
Negative Cell Control: Use cell lines known to lack BTG3 expression or BTG3 knockout/knockdown cells
Antibody Titration: Perform serial dilutions to determine optimal antibody concentration that maximizes signal-to-noise ratio
Technical Controls:
Unstained Control: Cells processed identically but without any antibodies to establish baseline autofluorescence
Secondary-Only Control: For indirect staining methods, include samples with only secondary antibody to assess non-specific binding
Isotype Control: Include matched isotype antibody at the same concentration to identify non-specific binding due to Fc receptors
Fluorescence Minus One (FMO) Control: Include all antibodies in the panel except BTG3 antibody to assess spillover from other fluorochromes
Biological Controls:
Cell Cycle-Specific Controls: Since BTG3 regulates cell cycle progression from G0/G1 to S phase , include samples synchronized at different cell cycle stages to correlate BTG3 expression with cell cycle position
Stimulation Controls: If studying conditions that might alter BTG3 expression, include both stimulated and unstimulated samples
Procedural Controls:
Fixation/Permeabilization Controls: Since BTG3 may require intracellular staining, optimize fixation and permeabilization protocols with appropriate controls
Viability Dye: Include a viability marker to exclude dead cells, which can bind antibodies non-specifically
Data Analysis Controls:
Compensation Controls: Single-stained controls for each fluorochrome to correct for spectral overlap
Gating Strategy Validation: Parallel analysis with alternative methods (e.g., imaging) to confirm subcellular localization
Recommended Control Panel for BTG3 Flow Cytometry:
Control Type | Purpose | Analysis Implication |
---|---|---|
Unstained | Establish autofluorescence | Set negative population boundaries |
Isotype Control | Identify non-specific binding | Differentiate specific from non-specific signal |
BTG3 Antibody in positive control cells | Confirm antibody performance | Establish positive signal parameters |
BTG3 Antibody in BTG3-knockdown cells | Verify specificity | Confirm signal reduction with decreased target |
Cell cycle marker + BTG3 | Correlate with cell cycle | Link expression to biological function |
By implementing this comprehensive control strategy, researchers can generate reliable flow cytometry data using BTG3 antibodies while avoiding common pitfalls in interpretation .
Post-translational modifications (PTMs) of BTG3 represent an important yet understudied aspect of its biology that can significantly impact both protein function and antibody recognition:
Types of PTMs Affecting BTG3:
Phosphorylation: Likely regulates BTG3 activity, stability, and interactions with binding partners
Ubiquitination: Controls protein turnover and potentially influences subcellular localization
SUMOylation: May alter BTG3's interaction with transcription factors and other nuclear proteins
Acetylation: Potentially regulates chromatin association and nuclear functions
Impact on BTG3 Function:
Cell Cycle Regulation: PTMs likely modulate BTG3's ability to impair G0/G1 to S phase progression
Protein-Protein Interactions: Modifications can create or disrupt binding interfaces
Subcellular Localization: PTMs may regulate nuclear-cytoplasmic shuttling
Protein Stability: Modifications often determine protein half-life and degradation pathways
Implications for Antibody Recognition:
Epitope Masking: PTMs can physically block antibody access to recognition sites
Conformational Changes: Modifications may alter protein folding, affecting discontinuous epitopes
Charge Alterations: PTMs like phosphorylation change local charge, potentially disrupting antibody binding
Western Blot Migration: Modified BTG3 may show altered migration patterns, appearing as multiple bands
Experimental Approaches to Address PTM Challenges:
Challenge | Experimental Approach | Expected Outcome |
---|---|---|
Multiple bands in Western blot | Phosphatase treatment of lysates | Band consolidation if due to phosphorylation |
Weak antibody binding | Epitope retrieval methods | Improved signal if PTMs are masking epitopes |
Tissue-specific recognition | Use antibodies targeting different epitopes | Different detection patterns may reveal tissue-specific PTMs |
Variable detection across cell cycle | Synchronize cells at different stages | Identification of cell cycle-specific modifications |
Research Strategy for PTM-Aware Antibody Selection:
Choose antibodies raised against synthetic peptides from regions less likely to undergo PTMs
Consider using multiple antibodies targeting different regions of BTG3
For PTM-specific studies, use antibodies specifically developed against modified forms of BTG3
When studying BRG3 (the E3 ligase), be aware that persulfidation at specific cysteine residues (Cys 206 and Cys 212) significantly impacts its ubiquitination activity
Understanding the interplay between PTMs, BTG3 function, and antibody recognition is crucial for accurate interpretation of experimental results, particularly when studying BTG3's cell cycle regulatory functions in different cellular contexts.
Despite significant progress in BTG3 research, several important knowledge gaps remain. Advanced antibody-based techniques offer promising approaches to address these outstanding questions:
Tissue-Specific Functions:
Knowledge Gap: While BTG3 is known to be ubiquitously expressed with higher levels in certain tissues (central nervous system, reproductive organs, lung) , the functional significance of this differential expression remains poorly understood.
Antibody-Based Approach: Spatial proteomics using highly specific BTG3 antibodies in multiplex immunofluorescence can map expression at cellular resolution across tissues, correlating with functional markers.
Cell Cycle Regulation Mechanisms:
Knowledge Gap: The precise molecular mechanisms by which BTG3 impairs cell cycle progression from G0/G1 to S phase remain incompletely characterized.
Antibody-Based Approach: Proximity ligation assays (PLA) with BTG3 antibodies paired with antibodies against cell cycle regulators can identify direct interactions in situ, while ChIP-seq can map genomic binding sites.
Protein Interaction Networks:
Knowledge Gap: The complete interactome of BTG3 across different cellular contexts is not fully mapped.
Antibody-Based Approach: BioID or APEX proximity labeling combined with BTG3 antibody-based purification can identify context-specific protein interactions with spatial resolution.
Post-Translational Regulation:
Knowledge Gap: The types, sites, and functional consequences of BTG3 post-translational modifications remain largely uncharacterized.
Antibody-Based Approach: Development of modification-specific antibodies, coupled with mass spectrometry validation, can identify regulated modification sites and their functional significance.
Disease Associations:
Knowledge Gap: While BTG3 has potential tumor suppressor functions, its role in specific diseases is not well established.
Antibody-Based Approach: Tissue microarray analysis with validated BTG3 antibodies can establish expression patterns across disease states. Single-cell CyTOF with BTG3 antibodies can identify cellular subpopulations with altered expression in disease contexts.
Developmental Dynamics:
Knowledge Gap: Despite high expression in the ventricular zone of the developing central nervous system , BTG3's role in neurodevelopment remains poorly characterized.
Antibody-Based Approach: Temporal immunohistochemistry studies with BTG3 antibodies during development, combined with lineage markers, can clarify developmental functions.
Research Priority Matrix for BTG3 Knowledge Gaps:
Knowledge Gap | Technical Approach | Potential Impact | Research Challenge |
---|---|---|---|
Tissue-specific functions | Spatial proteomics with multiplexed IF | Clarify context-dependent roles | Requires highly specific antibodies |
Cell cycle regulation mechanisms | PLA and ChIP-seq | Define molecular pathways | Need antibodies compatible with crosslinking |
Protein interaction networks | BioID with antibody purification | Map contextual interactome | Potential for false positives |
Post-translational regulation | PTM-specific antibodies | Identify regulatory mechanisms | Difficult epitope targeting |
Disease associations | TMA analysis and CyTOF | Clinical biomarker potential | Population heterogeneity |
Developmental dynamics | Temporal IHC with lineage markers | Developmental biology insights | Limited sample availability |
By strategically applying these advanced antibody-based approaches, researchers can address fundamental questions about BTG3 biology and potentially uncover new therapeutic targets related to its cell cycle regulatory functions.
Immunoprecipitation followed by mass spectrometry (IP-MS) represents a powerful approach for identifying and characterizing BTG3 protein interactions. This methodology requires careful experimental design and execution:
Experimental Design Considerations:
Cell/Tissue Selection: Choose models with verified BTG3 expression; HeLa cells are documented to express BTG3 and serve as a good starting point
Experimental Conditions: Consider both basal and stimulated conditions that might regulate BTG3 interactions
Controls: Include IgG control IPs and, ideally, BTG3 knockout/knockdown samples
Crosslinking Options: Evaluate whether crosslinking is needed to capture transient interactions
BTG3 Antibody Selection Criteria:
Epitope Location: Select antibodies targeting regions unlikely to interfere with protein-protein interactions
Validation Status: Use antibodies validated for immunoprecipitation applications
Format Considerations: Consider whether native or denatured IP conditions are optimal
Species Compatibility: Ensure compatibility with downstream MS detection systems
Optimized IP-MS Protocol Framework:
Cell Lysis: Use gentle lysis conditions to preserve protein complexes
Pre-clearing: Remove non-specific binding proteins with control beads
Immunocapture: Incubate lysates with BTG3 antibody pre-bound to beads
Washing: Perform stringent washes while preserving specific interactions
Elution: Choose between native elution (competition) or denaturing conditions
MS Analysis: Implement appropriate MS/MS methods for protein identification
Data Analysis Strategy:
Filtering Criteria: Compare to IgG control and exclude common contaminants
Quantification Approach: Use label-free or labeling methods (TMT, SILAC) for quantification
Interaction Confidence: Assign confidence scores based on peptide counts, specificity, and reproducibility
Network Analysis: Map interactions to known pathways and protein complexes
Validation of Identified Interactions:
Reciprocal IP: Confirm key interactions by IP with antibodies against identified partners
Proximity Ligation Assay: Visualize interactions in situ
Functional Studies: Test biological relevance through knockdown or mutation of interaction interfaces
Sample Workflow for BTG3 IP-MS:
Stage | Critical Steps | Quality Control Measures |
---|---|---|
Sample Preparation | Optimization of lysis buffers | Western blot to confirm BTG3 solubilization |
Immunoprecipitation | Antibody-bead coupling efficiency | SDS-PAGE with silver stain to visualize pulled-down proteins |
MS Sample Processing | In-gel or on-bead digestion | Peptide recovery assessment |
LC-MS/MS Analysis | Gradient optimization for peptide separation | Quality metrics for chromatography and MS performance |
Data Analysis | Appropriate filtering of non-specific binders | Comparison across biological replicates |
Interaction Validation | Selection of top candidates for confirmation | Statistical analysis of detection confidence |
This approach allows researchers to move beyond candidate-based interaction studies to unbiased discovery of the BTG3 interactome, particularly important for understanding BTG3's role in cell cycle regulation from G0/G1 to S phase .
BTG3's ubiquitous expression with tissue-specific enrichment presents both challenges and opportunities for contextual functional studies. Researchers can employ several sophisticated approaches to elucidate tissue-specific roles:
Spatial Expression Mapping Strategies:
Multiplexed Immunohistochemistry: Use BTG3 antibodies in conjunction with tissue-specific markers to identify cell populations with high expression
Single-Cell Protein Analysis: Apply techniques like CITE-seq or CyTOF with BTG3 antibodies to correlate expression with cell identity
In Situ Hybridization + Immunofluorescence: Combine BTG3 mRNA detection with protein localization to assess transcriptional vs. post-transcriptional regulation
Spatial Transcriptomics: Correlate BTG3 expression with tissue architecture and microenvironment
Tissue-Specific Functional Assessment:
Conditional Knockout Models: Generate tissue-specific BTG3 deletion models focusing on high-expression tissues (ventricular zone of CNS, reproductive tissues, thymus, lung)
Ex Vivo Tissue Cultures: Manipulate BTG3 expression in tissue explants to assess acute functional consequences
Organoid Systems: Derive organoids from tissues with high BTG3 expression to study function in 3D culture
Patient-Derived Xenografts: Examine BTG3 function in humanized models maintaining tissue architecture
Protein Interaction Network Analysis:
Tissue-Specific Interactome Mapping: Perform BTG3 immunoprecipitation followed by mass spectrometry across different tissues
Cell Type-Resolved Proximity Labeling: Use tissue-specific promoters to drive expression of BTG3-BioID fusions
Comparative Network Analysis: Identify tissue-specific vs. universal BTG3 interaction partners
Functional Readouts in Tissue Context:
Cell Cycle Analysis in Tissue Sections: Combine BTG3 antibody staining with proliferation markers (Ki67, BrdU) to assess correlation with cell cycle across tissues
Lineage Tracing with BTG3 Modulation: Track developmental consequences of BTG3 manipulation in tissues with high expression
Primary Cell Isolation and Culture: Compare BTG3 function in primary cells derived from different tissues
Comparative Analysis Framework for Tissue-Specific BTG3 Studies:
Technological Approaches for Tissue-Specific Studies:
Antibody-Based Tissue Cytometry: Quantitative assessment of BTG3 levels across tissue sections
In Vivo CRISPR Screens: Tissue-specific delivery of BTG3-targeting guides to assess function
Spatial Multi-omics: Integrate proteomic, transcriptomic and epigenomic data with spatial resolution
By systematically applying these approaches across tissues with differential BTG3 expression, researchers can deconvolute the general cell cycle regulatory functions of BTG3 from tissue-specific roles that may involve unique protein interactions or regulatory mechanisms.