ITM2A belongs to the BRICHOS superfamily and is encoded by the ITM2A gene on the X chromosome. Its expression is prominent in osteogenic, lymphoid, and brain tissues . The protein exhibits a molecular weight of approximately 30–39 kDa, depending on post-translational modifications .
| Property | Details |
|---|---|
| Molecular Weight | 30–39 kDa (observed) |
| Subcellular Location | Integral membrane protein |
| Tissue Expression | Osteogenic, lymphoid, brain microvessels |
| Gene Family | BRICHOS superfamily (ITM2 family) |
The ITM2A antibody is primarily used in research settings for protein detection and functional studies. Key applications include:
Western Blotting (WB): Validated for detecting endogenous ITM2A in human, mouse, and rat tissues .
Immunohistochemistry (IHC): Utilized to study ITM2A localization in tumor microenvironments .
Enzyme-Linked Immunosorbent Assay (ELISA): Employed for quantitative analysis of ITM2A expression .
Brain Drug Delivery: ITM2A has been explored as a transcytosis target for crossing the blood-brain barrier (BBB), with antibodies showing selective binding to ITM2A-overexpressing cells .
Cancer Immunotherapy: ITM2A’s role in modulating PD-L1 expression and tumor-infiltrating lymphocytes (TILs) suggests potential utility in immuno-oncology research .
Recent studies highlight ITM2A’s role in cancer biology and BBB transcytosis:
Antibodies targeting ITM2A’s extracellular domain show selective uptake in ITM2A-overexpressing cells, though in vivo transcytosis remains inconclusive .
ITM2A (Integral Membrane Protein 2A) is a type II transmembrane protein that belongs to the ITM2 family. It is also known as BRICD2A, E25A, and BRICHOS domain containing 2A. The protein has a molecular weight of approximately 29.7 kilodaltons in its native form, though it may appear larger (~39-45 kDa) in some experimental conditions due to post-translational modifications .
ITM2A is of significant research interest because it is expressed in several important cell types, including chondrocytes involved in endochondrial ossification, skeletal muscle, adipose tissue-derived stem cells, and CD3-activated CD4+ and CD8+ T cells . Recent studies have also identified ITM2A as a potential tumor suppressor in breast cancer, with high expression correlating with better patient outcomes .
Most commercially available ITM2A antibodies are designed to detect human ITM2A, but many also cross-react with orthologs in other species. Based on gene homology, reactivity has been reported with mouse, rat, porcine, and monkey ITM2A .
When selecting an antibody for your research, it's important to verify the specific species reactivity. For example, some antibodies like the rabbit polyclonal antibody ab203620 have been specifically validated for rat samples in applications such as IHC-P and ICC/IF . Other antibodies may show broader reactivity across human, mouse, and rat samples .
Always review the technical specifications and validation data for each antibody to ensure compatibility with your experimental model.
Based on validated protocols, the following methods have shown good results for ITM2A immunohistochemistry:
For formalin-fixed, paraffin-embedded (FFPE) tissues:
Fix tissues in 10% neutral buffered formalin for 24-48 hours
Process and embed in paraffin following standard protocols
Section tissues at 4-6 μm thickness
Perform heat-induced epitope retrieval (HIER) using either:
Citrate buffer (pH 6.0) for 20 minutes at 95-100°C
EDTA buffer (pH 9.0) for 20 minutes at 95-100°C
Cool sections to room temperature gradually
Block endogenous peroxidase activity with 3% H₂O₂
Apply ITM2A antibody at optimized dilution (e.g., 1:200 for ab203620)
For frozen sections or cultured cells:
Fix with 4% paraformaldehyde (PFA) for 10-15 minutes at room temperature
Permeabilize with 0.1-0.5% Triton X-100 for 5-10 minutes
Apply ITM2A antibody at appropriate dilution (e.g., 1:100 for immunofluorescence)
These protocols have been successfully used to detect ITM2A in rat brain tissue and rat brain vascular endothelial cells (RBE4) .
Proper storage and handling of ITM2A antibodies is crucial for maintaining their activity and specificity. Based on manufacturer recommendations:
For lyophilized antibodies:
Store unopened vials at -20 to -70°C for up to 12 months from the date of receipt
Reconstitute following manufacturer's instructions, typically using sterile buffer
After reconstitution, aliquot to avoid repeated freeze-thaw cycles
Store reconstituted antibodies at:
For liquid antibodies:
Store at 2-8°C for short-term (1-2 weeks)
For long-term storage, aliquot and freeze at -20°C
Avoid more than 2-3 freeze-thaw cycles as this can degrade antibody performance
When thawing, allow the antibody to equilibrate to room temperature before opening the vial
Centrifuge the vial briefly before use to collect all material
General handling recommendations:
Use sterile technique when handling antibodies
Return antibodies to appropriate storage conditions immediately after use
Work with antibodies on ice when possible
Including appropriate controls is essential for validating results obtained with ITM2A antibodies:
Positive Controls:
Cell lines known to express ITM2A, such as:
Tissues with confirmed ITM2A expression, such as rat brain tissue
Negative Controls:
Primary antibody omission control (substitute antibody diluent for primary antibody)
Isotype control (use non-specific antibody of the same isotype and concentration)
Cell lines with confirmed low/no ITM2A expression
ITM2A-knockout or knockdown samples (if available)
Additional Technical Controls:
Secondary antibody only control (to assess non-specific binding)
Blocking peptide competition (pre-incubate antibody with immunizing peptide)
Parallel detection with alternative antibody clones targeting different epitopes of ITM2A
Validation by orthogonal methods (e.g., confirm protein detection with RNA expression data)
These controls help ensure specificity of detection and rule out technical artifacts or non-specific binding.
Recent research has identified ITM2A as a potential tumor suppressor in breast cancer. Analysis of gene expression profiles from multiple datasets (GSE29413, GSE61304, and TCGA) has demonstrated that ITM2A is frequently downregulated in breast cancer compared to normal tissues .
The relationship between ITM2A expression and patient outcomes includes:
These findings suggest that ITM2A may function as a tumor suppressor in breast cancer, potentially through modulation of immune responses in the tumor microenvironment. Further research is needed to determine if similar relationships exist in other cancer types.
To investigate ITM2A's role in cellular differentiation, researchers can employ several complementary experimental approaches:
1. Expression Analysis During Differentiation:
qRT-PCR to quantify ITM2A mRNA expression at different time points during differentiation
Western blot with ITM2A antibodies to track protein expression changes
Immunofluorescence to visualize subcellular localization changes during differentiation
Flow cytometry to quantify ITM2A expression at the single-cell level
2. Gain-of-Function Studies:
Stable or transient overexpression of ITM2A using plasmid vectors
Inducible expression systems (e.g., Tet-On) to control timing of ITM2A expression
Analysis of differentiation markers before and after ITM2A induction
3. Loss-of-Function Studies:
siRNA or shRNA-mediated knockdown of ITM2A
CRISPR-Cas9 gene editing to create ITM2A knockout cell lines
Analysis of differentiation potential in ITM2A-depleted cells
4. Protein Interaction Studies:
Co-immunoprecipitation with ITM2A antibodies to identify interaction partners
Proximity labeling approaches (BioID, APEX) to identify proteins in close proximity to ITM2A
Mass spectrometry analysis of ITM2A-containing protein complexes
5. Functional Assays:
Cell proliferation assays in ITM2A-manipulated cells
Migration and invasion assays to assess cellular behavior
Lineage-specific differentiation assays (osteogenic, chondrogenic, myogenic, etc.)
Analysis of signaling pathway activation using phospho-specific antibodies
6. In vivo Models:
Conditional knockout mouse models to study tissue-specific roles of ITM2A
Xenograft studies with ITM2A-overexpressing or -depleted cells
Analysis of developmental phenotypes in model organisms
These approaches can be combined to build a comprehensive understanding of ITM2A's functional role in cellular differentiation across different cellular contexts.
The relationship between ITM2A expression and immune responses in cancer, particularly breast cancer, involves several key aspects:
1. PD-L1 Expression Regulation:
ITM2A has been found to facilitate the expression of PD-L1 (Programmed Death-Ligand 1) in breast cancer cells. This relationship was verified through both qRT-PCR assays and public database analysis. PD-L1 is a critical immune checkpoint molecule that can inhibit T cell function, but its expression in certain contexts can also be associated with active immune responses .
2. Association with Tumor-Infiltrating Lymphocytes (TILs):
Breast cancers with higher ITM2A expression were found to have more tumor-infiltrating lymphocytes. This correlation suggests that ITM2A may play a role in modulating the immune microenvironment of tumors .
3. Differential Gene Expression:
RNA-sequencing analysis of breast cancer cells overexpressing ITM2A revealed that differentially expressed genes were associated with immunity responses, further supporting ITM2A's role in immune modulation .
4. Prognostic Implications:
Interestingly, PD-L1 expression associated with tumor-infiltrating lymphocytes has been found to be a positive prognostic feature in breast cancer. Given ITM2A's relationship with both PD-L1 expression and TILs, this could explain in part why high ITM2A expression correlates with better patient outcomes .
This emerging understanding of ITM2A's immunomodulatory roles presents new research directions, including:
Investigating whether ITM2A could serve as a biomarker for immunotherapy response
Exploring whether modulating ITM2A expression could enhance anti-tumor immune responses
Determining the molecular mechanisms by which ITM2A regulates PD-L1 expression
Examining how ITM2A affects different immune cell populations within the tumor microenvironment
When detecting ITM2A by Western blot, researchers may encounter several challenges. Here are common issues and their solutions:
Expected observation: ITM2A should appear as a band at approximately 39 kDa , though the theoretical molecular weight is 29.7 kDa .
Potential causes and solutions:
Post-translational modifications: ITM2A may undergo glycosylation or other modifications
Solution: Treat samples with deglycosylation enzymes (PNGase F) to confirm glycosylation
Incomplete protein denaturation
Solution: Increase SDS concentration or boiling time in sample buffer
Protein degradation
Solution: Use fresh samples and add protease inhibitors during extraction
Antibody cross-reactivity
Solution: Test multiple antibodies targeting different epitopes of ITM2A
Potential causes and solutions:
Low protein expression
Solution: Load more protein (50-100 μg) or use enrichment techniques
Inefficient transfer
Solution: Optimize transfer conditions (time, voltage, buffer composition)
Suboptimal antibody concentration
Incompatible blocking agent
Solution: Test different blocking agents (BSA vs. non-fat milk)
Potential causes and solutions:
Excessive antibody concentration
Solution: Dilute primary and secondary antibodies
Insufficient blocking
Solution: Increase blocking time or concentration
Inadequate washing
Solution: Add additional washing steps with 0.1% Tween-20 in TBS
Secondary antibody cross-reactivity
Solution: Use highly cross-adsorbed secondary antibodies
Technical Tips for Optimal ITM2A Detection:
Use PVDF membranes, which have shown good results for ITM2A detection
Perform electrophoresis under reducing conditions for optimal ITM2A resolution
Consider using specialized Western blot buffer systems, such as Western Blot Buffer Group 1, which has been validated for ITM2A detection
Include positive control lysates, such as A-673 human Ewing's sarcoma cell line
Validating antibody specificity is crucial for obtaining reliable results. For ITM2A antibodies, consider these comprehensive validation approaches:
1. Genetic Validation:
siRNA knockdown: Demonstrate reduction in signal intensity following ITM2A knockdown
CRISPR-Cas9 knockout: Show complete absence of signal in ITM2A knockout cells
Overexpression: Show increased signal in cells transfected with ITM2A expression vectors
These genetic validations should be performed in relevant cell types that express detectable levels of endogenous ITM2A
2. Orthogonal Methods Validation:
Correlation with mRNA levels: Compare protein detection with qRT-PCR results across multiple samples
Mass spectrometry validation: Confirm the identity of the detected protein band
In situ hybridization: Compare antibody staining patterns with mRNA localization
Verify results with multiple antibodies: Use antibodies targeting different epitopes of ITM2A
3. Technical Validation:
Peptide competition: Pre-incubate the antibody with the immunizing peptide to demonstrate signal reduction
Isotype controls: Use matched isotype control antibodies to evaluate non-specific binding
Signal titration: Show concentration-dependent signal with increasing amounts of target protein
Cell type specificity: Confirm expected expression patterns across cell types known to express or lack ITM2A
4. Application-Specific Validation:
For IHC/ICC: Compare staining patterns across multiple fixation and antigen retrieval methods
For flow cytometry: Compare surface vs. intracellular staining protocols
For IP experiments: Confirm pulldown of correctly sized protein by Western blot
For functional assays: Demonstrate that antibody binding affects expected biological functions
5. Cross-reactivity Testing:
Test the antibody against related family members (e.g., ITM2B, ITM2C)
Evaluate species cross-reactivity using samples from different organisms
Examine antibody performance in tissues with complex protein mixtures
Implementing multiple validation strategies provides strong evidence for antibody specificity and increases confidence in experimental results.
When facing contradictory findings regarding ITM2A function, researchers should employ a systematic approach to reconcile these discrepancies:
Cell type specificity: ITM2A may function differently in various cell types (chondrocytes vs. T cells vs. cancer cells)
Developmental stage: Function may vary across developmental or differentiation stages
Pathological state: Normal vs. disease state may alter protein function
Experimental methodology: Different techniques may reveal distinct aspects of function
Create a comparison table of contradictory studies, analyzing:
Cell/tissue systems used
Expression levels (physiological vs. overexpression)
Knockdown/knockout strategies
Readout methods
Time points examined
ITM2A function may depend on:
Specific protein-protein interactions that vary by context
Post-translational modifications affecting function
Subcellular localization differences
Cleavage or processing events
Direct comparison experiments using:
Multiple cell types in parallel
Range of expression levels
Both gain- and loss-of-function approaches
Multiple functional readouts
Time-course studies
5. Example Reconciliation Framework for ITM2A:
Recent studies suggest ITM2A functions as a tumor suppressor in breast cancer , but earlier work implicated it in other processes like chondrogenesis. To reconcile these findings:
Investigate whether ITM2A's tumor suppressive effects are mediated through:
Differentiation pathways shared with developmental contexts
Novel cancer-specific mechanisms
Immune modulatory functions
Examine whether ITM2A's relationship with PD-L1 expression exists across different cellular contexts
Is this relationship cancer-specific?
Does it depend on the immune microenvironment?
Is it influenced by other signaling pathways?
Determine whether ITM2A's effects on cell proliferation, invasion, and migration are universal or context-dependent
By systematically analyzing contradictory findings through these approaches, researchers can develop more nuanced models of ITM2A function that accommodate seemingly disparate results within a coherent framework.
Several emerging applications of ITM2A antibodies show significant promise for deepening our understanding of disease mechanisms:
1. Cancer Prognostic Biomarker Development:
Using ITM2A antibodies for tissue microarray analysis to correlate expression with patient outcomes across cancer types
Developing standardized immunohistochemical scoring systems for ITM2A expression in tumors
Exploring whether ITM2A expression patterns can predict response to immunotherapies, given its relationship with PD-L1 and TILs
2. Single-Cell Analysis Applications:
Combining ITM2A antibodies with other markers for multi-parameter flow cytometry to identify specific cell populations
Applying CyTOF (mass cytometry) with metal-conjugated ITM2A antibodies for high-dimensional phenotyping of tumor and immune cells
Using ITM2A antibodies in single-cell Western blot techniques to examine protein heterogeneity
3. Spatial Biology and Tissue Architecture:
Employing ITM2A antibodies in multiplex immunofluorescence to map spatial relationships with immune cells in the tumor microenvironment
Using imaging mass cytometry with ITM2A antibodies to preserve spatial context while examining multiple markers
Correlating ITM2A expression patterns with tissue architecture in normal development and disease
4. Functional Screening Applications:
Using ITM2A antibodies in high-content screening to identify modulators of ITM2A expression or localization
Developing ITM2A-targeted proximity labeling approaches to identify context-specific interaction partners
Employing antibody-based techniques to modulate ITM2A function in cellular models
5. Translational Medicine Applications:
Investigating whether ITM2A antibodies could be used therapeutically to modulate immune responses in cancer
Developing companion diagnostic approaches using ITM2A antibodies to stratify patients for targeted therapies
Creating circulating tumor cell detection methods incorporating ITM2A antibodies
These emerging applications leverage the specificity of ITM2A antibodies to advance both basic understanding of ITM2A biology and its potential clinical applications, particularly in cancer research and immunology.
The study of ITM2A offers several promising avenues for novel therapeutic development:
1. Cancer Immunotherapy Enhancement:
Given ITM2A's association with PD-L1 expression and tumor-infiltrating lymphocytes , therapeutic approaches could include:
Modulating ITM2A expression to enhance response to existing immune checkpoint inhibitors
Developing combination therapies targeting both ITM2A signaling and other immune pathways
Using ITM2A expression as a biomarker to identify patients likely to respond to immunotherapies
2. Targeted Cancer Therapies:
Restoring ITM2A expression in cancers where it is downregulated could potentially suppress tumor growth and invasiveness
Developing small molecules or biologics that mimic ITM2A's tumor-suppressive functions
Creating synthetic biology approaches that link ITM2A expression to therapeutic effectors
3. Regenerative Medicine Applications:
Based on ITM2A's role in cellular differentiation:
Modulating ITM2A to enhance chondrogenic differentiation for cartilage regeneration
Controlling ITM2A expression to direct stem cell fate in tissue engineering
Developing tissue-specific delivery systems for ITM2A-targeting therapeutics
4. Diagnostic and Prognostic Tools:
Creating standardized ITM2A immunohistochemistry protocols for cancer prognosis
Developing circulating biomarker assays that include ITM2A assessment
Integrating ITM2A status into multi-parameter prognostic models
5. Drug Development Strategies:
Screening for compounds that modulate ITM2A expression or activity
Identifying the molecular mechanisms of ITM2A-mediated tumor suppression to reveal novel drug targets
Developing antibody-drug conjugates targeting cells with specific ITM2A expression patterns
6. Experimental Therapeutic Approaches:
Engineered T cells with enhanced ITM2A signaling for adoptive cell therapy
mRNA or gene therapy approaches to restore ITM2A expression
Nanotechnology-based delivery of ITM2A modulators to specific tissues
As research continues to clarify ITM2A's functions in normal physiology and disease, these therapeutic directions will likely expand and become more precisely defined. The connection between ITM2A and immune regulation is particularly promising for near-term therapeutic development.
Based on collected evidence and technical considerations, here are best practices for designing experiments with ITM2A antibodies:
1. Antibody Selection:
Choose antibodies validated for your specific application (WB, IHC, ICC/IF, etc.)
Consider using monoclonal antibodies for higher specificity in complex applications
Verify species reactivity matches your experimental model
Review literature to identify antibody clones with proven performance in similar experiments
When possible, use antibodies targeting different epitopes to confirm findings
2. Experimental Controls:
Always include positive controls (e.g., A-673 human Ewing's sarcoma cell line)
Include appropriate negative controls (antibody omission, isotype controls)
Consider using genetic approaches (knockdown/knockout) to validate specificity
Include loading controls for Western blot experiments
Use blocking peptides when available to confirm specificity
3. Protocol Optimization:
Determine optimal antibody concentration through titration experiments
For Western blot, use PVDF membrane and reducing conditions as validated for ITM2A detection
For IHC, optimize fixation and antigen retrieval methods (4% PFA fixation for IF, formalin fixation with heat-induced epitope retrieval for IHC-P)
Adjust incubation times and temperatures based on signal strength and background
4. Data Analysis and Interpretation:
Quantify results using appropriate software and statistical methods
Consider ITM2A's molecular weight variation (theoretical 29.7 kDa vs. observed ~39-45 kDa) when interpreting Western blot results
Correlate protein expression with mRNA data when possible
Report detailed methods including antibody catalog numbers, dilutions, and incubation conditions
5. Documentation and Reporting:
Document all experimental conditions thoroughly
Include representative images with scale bars
Report both positive and negative results
Provide detailed materials and methods to enable reproducibility
Clearly state limitations of the antibodies used
By following these best practices, researchers can ensure reliable and reproducible results when using ITM2A antibodies, contributing to our collective understanding of this protein's functions in normal physiology and disease.
When interpreting ITM2A expression data across different experimental systems, researchers should consider several important factors:
1. Expression Level Variations:
Baseline expression levels vary significantly across tissue and cell types
Expression may be highly context-dependent (differentiation state, activation status)
Quantitative comparison across different systems requires standardization
Consider both absolute and relative expression changes when interpreting results
2. Technical Considerations:
Different detection methods (antibody-based vs. RNA-based) may not correlate perfectly
Various antibody clones may recognize different isoforms or post-translationally modified forms
Detection sensitivity varies across platforms (Western blot vs. IHC vs. flow cytometry)
Image acquisition settings significantly impact quantitative comparisons in microscopy
3. Biological Context Integration:
Interpret ITM2A expression in relation to known functions in specific tissue/cell types
Consider the developmental or disease stage when comparing expression patterns
Evaluate expression in the context of relevant signaling pathways
Assess whether expression changes are cause or consequence of observed phenotypes
4. Cross-Species Considerations:
ITM2A function may be conserved but regulation can differ across species
Antibody cross-reactivity may vary between human, mouse, rat, and other species
Expression patterns during development or disease progression may not be identical across species
Validate findings in multiple species when possible
5. Disease-Specific Interpretation:
In cancer research, consider tumor heterogeneity when interpreting ITM2A expression
Correlate ITM2A expression with clinical parameters and outcomes when available
Integrate findings with immune cell infiltration and activation data
Consider microenvironmental factors that may influence expression
6. Integration Framework:
When confronted with seemingly contradictory ITM2A expression data, consider this stepwise approach:
Verify technical aspects (antibody specificity, assay conditions)
Compare experimental conditions (cell types, treatments, time points)
Examine broader biological context (signaling pathways, cellular state)
Develop testable hypotheses to explain discrepancies
Design reconciliation experiments that directly compare systems