MTA3 antibody targets the MTA3 protein, encoded by the MTA3 gene located on human chromosome 2p21. MTA3 is a chromatin-modifying protein involved in transcriptional regulation, with roles in epithelial-mesenchymal transition (EMT), hormone response, and cancer metastasis .
Parameter | MTA3-Negative (%) | MTA3-Positive (%) | P-value |
---|---|---|---|
Poor differentiation | 82.6 | 17.4 | 0.002 |
Lymph node metastasis | 75.3 | 24.7 | <0.001 |
Distant metastasis | 89.2 | 10.8 | 0.006 |
Advanced TNM stage | 68.9 | 31.1 | <0.001 |
Western blotting: Identifies MTA3 in HepG2, MCF-7, and Raji cell lysates .
Immunohistochemistry (IHC): Localizes MTA3 in mouse brain and human tumor tissues .
Functional studies: Links MTA3 loss to increased Snail expression and EMT in breast cancer .
Upregulation: Estrogen receptor signaling, β-elemene (traditional Chinese medicine) .
Downregulation: microRNA-495 in non-small cell lung cancer .
MTA3 antibodies are pivotal for:
Applications : WB
Review: Western blot analysis of MAT3 and LBR in BHK cells infected with Eimeria tenella compared with mock-infected cells.
MTA3 is a component of the Metastasis-associated protein family (MTA) that functions as part of the histone deacetylase NuRD complex involved in chromatin remodeling . Unlike other MTA family members that typically promote tumor progression, MTA3 often demonstrates tumor-suppressive properties in various cancers. MTA3 plays a crucial role in maintaining normal epithelial architecture by repressing SNAI1 transcription in a histone deacetylase-dependent manner, thereby regulating E-cadherin levels . This makes MTA3 particularly significant in cancer research, as its expression is frequently downregulated in malignancies including colorectal, breast, and ovarian cancer . The molecular mechanisms through which MTA3 regulates epithelial-to-mesenchymal transition (EMT) position it as an important research target for understanding cancer progression and potential therapeutic interventions.
MTA3 antibodies can be utilized across multiple experimental applications:
Application | Recommended Dilution | Notes |
---|---|---|
Western Blot (WB) | 1:5000-1:50000 | Detects 62kDa and 68kDa bands |
Immunohistochemistry (IHC) | Application-dependent | Effective for formalin-fixed paraffin-embedded tissues |
Immunofluorescence (IF/ICC) | 1:50-1:500 | Suitable for cellular localization studies |
Flow Cytometry (FC) | 0.25 μg per 10^6 cells | For intracellular detection |
ELISA | Assay-dependent | For quantitative analysis |
These applications enable researchers to detect, quantify, and localize MTA3 protein in various experimental models and clinical samples . Optimal dilutions should be determined empirically for each specific application and sample type.
When selecting an MTA3 antibody, consider the following factors:
Target region specificity: Determine whether you need an antibody targeting the C-terminal, N-terminal, or internal region of MTA3. For instance, some antibodies specifically target the C-terminal region (amino acids 400-450) .
Species reactivity: Verify the antibody's cross-reactivity with your model organism. Available antibodies react with various species including human, mouse, rat, and even cow, chicken, and Xenopus laevis .
Clonality: Choose between polyclonal antibodies (broader epitope recognition, higher sensitivity) and monoclonal antibodies (greater specificity, consistency between lots) based on your experimental needs .
Conjugation: Determine whether you need an unconjugated antibody or one conjugated to a detection molecule, based on your detection method .
Validation data: Review available validation data for your application to ensure the antibody has been verified for your specific usage scenario .
For optimal Western blot detection of MTA3:
Sample preparation:
Use RIPA buffer with protease inhibitors for cell lysis
Include phosphatase inhibitors if studying phosphorylation status
Heat samples at 95°C for 5 minutes in reducing sample buffer
Gel electrophoresis:
Transfer and detection:
Expected results:
Remember that optimization may be necessary for your specific experimental system.
For optimal immunohistochemical detection of MTA3:
Tissue preparation:
Use 10% neutral-buffered formalin fixation (12-24 hours)
Process and embed in paraffin following standard protocols
Section tissues at 3-5μm thickness
Antigen retrieval:
Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) is recommended
Microwave or pressure cooker methods are effective
Blocking and antibody incubation:
Visualization and analysis:
Appropriate controls are essential for reliable results with MTA3 antibodies:
Positive tissue/cell controls:
Negative controls:
Technical validation controls:
Antibody specificity validation by peptide competition
siRNA knockdown of MTA3 to confirm antibody specificity
Recombinant MTA3 protein as a positive control in Western blots
Internal controls:
Housekeeping proteins (β-actin, GAPDH) for protein loading normalization in Western blots
Non-malignant adjacent tissue in tumor samples
MTA3 functions as a component of the histone deacetylase NuRD complex, making MTA3 antibodies valuable tools for studying chromatin remodeling:
Co-immunoprecipitation (Co-IP) applications:
Use MTA3 antibodies to precipitate the NuRD complex
Analyze co-precipitated proteins by mass spectrometry or Western blot
Target known NuRD components like HDAC1/2, Mi-2α/β, RbAp46/48
Optimize buffer conditions to maintain complex integrity
Chromatin immunoprecipitation (ChIP) assays:
Proximity ligation assays (PLA):
Visualize interactions between MTA3 and other NuRD complex components
Combine with immunofluorescence to assess cellular localization of interactions
Particularly useful for studying context-dependent associations in different cell types
Sequential ChIP (Re-ChIP):
These approaches provide insights into how MTA3-containing NuRD complexes regulate gene expression programs involved in cancer development and progression.
MTA3 plays a significant role in EMT regulation, particularly through SNAI1 repression and E-cadherin regulation:
Experimental models:
Select appropriate cell lines with intact EMT machinery
Consider breast cancer (MCF-7) or colorectal cancer cell lines
3D organoid culture systems provide physiologically relevant context
Analytical approaches:
Use immunofluorescence to co-localize MTA3 with E-cadherin and other EMT markers
Implement time-course experiments during EMT induction to track MTA3 dynamics
Combine MTA3 detection with analysis of SNAI1 and other EMT regulators
Functional studies:
Mechanistic investigations:
Analyze histone modifications at EMT gene promoters using ChIP
Assess MTA3 binding to SNAI1 promoter during EMT progression
Investigate cooperation with other transcription factors
Understanding MTA3's role in EMT could provide insights into cancer metastasis mechanisms and potential therapeutic strategies.
MTA3 expression has been associated with cancer prognosis, particularly in colorectal cancer:
Parameter | MTA3 Negative | MTA3 Positive | Clinical Association |
---|---|---|---|
Tumor Differentiation | More common in poorly differentiated tumors | More common in well-differentiated tumors | Poor differentiation associates with worse outcomes |
Lymph Node Metastasis | Higher frequency | Lower frequency | Lymph node involvement indicates disease progression |
Distant Metastasis | More common | Less common | Distant metastasis correlates with decreased survival |
TNM Stage | More advanced stages | Earlier stages | Advanced stages indicate poorer prognosis |
These applications provide valuable insights into the clinical relevance of MTA3 in cancer progression and patient outcomes .
When encountering weak or absent MTA3 signal in Western blots:
Sample preparation optimization:
Ensure complete lysis using appropriate buffers (RIPA with protease inhibitors)
Avoid repeated freeze-thaw cycles of protein samples
Prepare fresh samples or add additional protease inhibitors
Consider subcellular fractionation as MTA3 can be primarily nuclear
Technical adjustments:
Transfer optimization:
For 62-68 kDa proteins like MTA3, verify efficient transfer
Consider longer transfer times or semi-dry transfer systems
Use PVDF membranes for better protein retention
Antibody selection:
Positive controls:
Non-specific staining in MTA3 immunohistochemistry can be addressed by considering:
Antibody specificity issues:
Use antibodies with validated IHC applications
Titrate antibody concentration (perform dilution series)
Include peptide competition controls to confirm specificity
Consider monoclonal antibodies for higher specificity
Tissue preparation factors:
Optimize fixation times (over-fixation can mask epitopes)
Ensure complete deparaffinization and rehydration
Test different antigen retrieval methods (citrate vs. EDTA buffers)
Evaluate fresh vs. archived tissues (epitope degradation over time)
Blocking improvements:
Extend blocking time (60 minutes minimum)
Use higher concentrations of blocking serum (5-10%)
Add bovine serum albumin (BSA) to reduce background
Include avidin/biotin blocking for biotin-based detection systems
Detection system optimization:
Consider polymer-based detection systems to reduce background
Use species-specific secondary antibodies
Include additional washing steps with 0.1% Tween-20
Dilute chromogen appropriately to minimize non-specific reaction
Controls and verification:
MTA3 can be detected at both 62kDa and 68kDa molecular weights, representing different isoforms or post-translational modifications . To optimize detection of both forms:
Gel separation strategies:
Use 8-10% acrylamide gels for better separation in the 60-70kDa range
Consider gradient gels (4-15%) for optimal resolution
Extend electrophoresis time to improve band separation
Antibody selection:
Sample preparation considerations:
Test different lysis buffers to ensure complete extraction of both forms
Consider native vs. denaturing conditions
Evaluate the effects of phosphatase inhibitors on mobility shifts
Cell/tissue type selection:
Different cell types may express isoforms at varying levels
Compare expression patterns across multiple cell lines
Consider tissue-specific expression patterns
Analysis approaches:
Quantify both bands independently when assessing expression levels
Consider ratios between isoforms as potentially biologically relevant
Correlate isoform expression with functional assays
MTA3 can be integrated into multimarker panels for refined cancer classification:
Multiplex immunohistochemistry/immunofluorescence:
Combine MTA3 detection with EMT markers (E-cadherin, vimentin, SNAI1)
Include cell type-specific markers for contextual analysis
Implement spectral unmixing for simultaneous detection of multiple markers
Correlate MTA3 with established molecular subtype markers
Flow cytometry applications:
Digital pathology approaches:
Use image analysis software for quantitative co-localization studies
Implement machine learning algorithms for pattern recognition
Develop automated scoring systems for multiple markers
Create spatial maps of marker expression in tumor microenvironments
Clinical application considerations:
Test prognostic value of MTA3 in combination with established markers
Evaluate predictive value for therapy response
Develop standardized reporting frameworks for complex marker panels
These approaches can provide more nuanced classification of tumors and potentially reveal functional subtypes relevant to prognosis and treatment decisions.
Patient-derived xenograft models offer valuable systems for studying MTA3 in cancer biology:
Antibody cross-reactivity considerations:
Experimental design factors:
Compare MTA3 expression between primary patient samples and derived PDX
Track MTA3 expression across PDX passages to assess stability
Correlate MTA3 expression with PDX growth, invasion, and metastasis
Implement IHC protocols optimized for xenograft tissues
Functional investigations:
Test MTA3-targeting interventions on PDX growth and metastasis
Evaluate MTA3 as a biomarker for therapy response in PDX models
Combine with patient clinical data for translational relevance
Consider ex vivo manipulation of PDX-derived cells
Analysis approaches:
Implement spatial analysis of MTA3 expression within PDX tumors
Correlate with stromal infiltration patterns
Integrate with genomic and transcriptomic profiling
Compare expression patterns between primary tumor and metastatic PDX models
PDX models provide systems to study MTA3 biology in contexts that maintain tumor heterogeneity and microenvironment interactions.
MTA3's role in regulating EMT and differentiation suggests potential involvement in therapy resistance:
Expression analysis in resistant models:
Compare MTA3 levels between therapy-sensitive and resistant cell lines
Analyze MTA3 expression before and after treatment exposure
Correlate MTA3 with established resistance markers
Implement longitudinal sampling from patients during treatment course
Mechanistic investigations:
Assess MTA3 regulation of drug efflux pumps or detoxification enzymes
Investigate connections between MTA3-regulated EMT and therapy resistance
Explore MTA3's role in cancer stem cell maintenance
Evaluate interactions with therapy-induced signaling pathways
Manipulation approaches:
Test effects of MTA3 overexpression or knockdown on therapy sensitivity
Combine with pharmacological modulation of NuRD complex activity
Assess potential for MTA3-targeted interventions to overcome resistance
Evaluate synergistic approaches targeting MTA3-regulated pathways
Clinical correlation strategies:
Analyze MTA3 expression in pre- and post-treatment patient samples
Correlate expression patterns with therapy response and outcomes
Investigate value as a predictive biomarker
Consider differential expression across tumor regions (core vs. invasive front)
These investigations could identify novel mechanisms of therapy resistance and potentially reveal new therapeutic strategies to overcome resistance in cancer patients.