Host/Isotype: Rabbit/IgG
Reactivity: Human, mouse, rat
Applications: Western blot (WB), immunoprecipitation (IP), immunofluorescence (IF), immunohistochemistry (IHC), ELISA
Key Features:
Host/Isotype: Mouse/IgG1
Reactivity: Human, mouse, rat
Applications: WB, ELISA
Key Features:
The MITD1 Antibody has been instrumental in studying MITD1’s role in diverse biological contexts:
Key Finding: MITD1 interacts with ESCRT-III proteins (e.g., CHMP2A, IST1) to stabilize midbody formation during cytokinesis. Depletion of MITD1 via siRNA leads to multinucleated cells and abscission defects .
Antibody Use: Immunofluorescence with the polyclonal antibody confirmed MITD1 localization to midbodies in dividing HeLa cells .
Key Finding: MITD1 is upregulated in CC tissues and correlates with poor prognosis. Its knockdown inhibits cell proliferation, migration, and induces ferroptosis via lipid ROS accumulation and GSH depletion .
Antibody Use: IHC with the polyclonal antibody validated MITD1 overexpression in CC tissues .
Key Finding: MITD1 knockdown suppresses ccRCC cell growth and migration, suggesting its role as a therapeutic target .
Antibody Use: WB analysis using the monoclonal antibody confirmed MITD1 expression in ccRCC cell lines (e.g., 786-O, A498) .
Key Finding: Serine/arginine-rich splicing factor 1 (SRSF1) stabilizes MITD1 mRNA, enhancing its expression in CC cells .
Antibody Use: RIP assays with the polyclonal antibody demonstrated SRSF1-MITD1 mRNA binding .
| Study Focus | Key Findings | Antibody Used |
|---|---|---|
| Cytokinesis | MITD1 stabilizes midbody formation | Polyclonal (17264-1-AP) |
| Colorectal Cancer (CC) | MITD1 overexpression linked to poor prognosis | Polyclonal (17264-1-AP) |
| ccRCC | MITD1 knockdown inhibits tumor growth | Monoclonal (68367-1-Ig) |
| SRSF1 Regulation | SRSF1 stabilizes MITD1 mRNA | Polyclonal (17264-1-AP) |
MITD1 is a protein containing an N-terminal microtubule-interacting and trafficking (MIT) domain and a C-terminal phospholipase D-like (PLD) domain that binds membranes . It functions primarily in:
Abscission during cytokinesis, coordinating with ESCRT-III proteins to facilitate the final separation of daughter cells
Antiviral activity against flaviviruses including West Nile virus, Usutu virus, Zika virus, and dengue virus
Potential tumor suppression in certain cancers by inhibiting cell proliferation and migration
The protein exists as a homodimer and localizes to the midbody during the terminal stages of cell division, particularly when the midbody appears very thin .
MITD1 antibodies have been validated for multiple experimental applications:
Most commercially available MITD1 antibodies show reactivity with human, mouse, and rat samples, detecting the protein at approximately 29-30 kDa .
In the brain, MITD1 expression is specifically induced in microglial cells (the primary immune cells of the central nervous system)
MITD1 expression is not increased by type I interferon (IFN-I) in most human cells and mouse tissues examined, suggesting tissue-specific regulation mechanisms
At the single-cell level, MITD1 shows highest expression in T cells (87.7%) compared to other cell types
MITD1 expression varies significantly across cancer types, with upregulation in some cancers and downregulation in others compared to corresponding normal tissues
When using MITD1 antibodies for Western blot experiments, researchers should incorporate:
Positive controls: Use cell lines with confirmed MITD1 expression, such as MCF-7, HeLa, or HEK-293 cells
Negative controls: Consider MITD1 knockdown samples (siRNA-treated cells) to confirm antibody specificity
Loading controls: Standard housekeeping proteins such as GAPDH, β-actin, or α-tubulin
Molecular weight marker: To confirm the expected band size of approximately 29-30 kDa
Antibody validation: Test the antibody on multiple cell lines to ensure consistent detection (e.g., MCF-7, Saos-2, U2OS, LNCaP, HeLa, HEK-293, HSC-T6, NIH/3T3, 4T1 cells)
Always optimize antibody dilution (typically 1:500-1:10000 for Western blot) based on your specific experimental conditions .
For optimal IHC results with MITD1 antibodies:
Antigen retrieval: Perform microwave antigen retrieval with 10 mM PBS buffer pH 7.2 before commencing with the IHC staining protocol
Antibody dilution: Start with a dilution of 1:100 and optimize as needed
Tissue selection: For baseline studies, consider liver tissue which has been successfully used for MITD1 IHC
Controls: Include both positive control tissues with known MITD1 expression and negative controls (omitting primary antibody)
Detection method: Use appropriate secondary antibodies and visualization systems compatible with your primary antibody's host species (typically rabbit or mouse)
Counterstaining: Apply appropriate nuclear counterstain for context and cellular localization
Quantification: Consider digital image analysis for objective quantification of staining intensity and distribution
To investigate MITD1's antiviral properties:
Expression analysis: Use Western blot with MITD1 antibodies to measure MITD1 expression levels before and after viral infection or interferon treatment
Localization studies: Employ immunofluorescence with MITD1 antibodies to visualize subcellular localization during viral infection, particularly focusing on potential colocalization with viral replication factories
Functional studies: Combine MITD1 antibody detection with:
MITD1 overexpression or knockdown experiments
Viral load quantification (plaque assays, qPCR, etc.)
Assessment of viral replication factory formation
Interaction analysis: Use co-immunoprecipitation with MITD1 antibodies to identify interactions with ESCRT-III proteins and viral components
Tissue-specific expression: Apply MITD1 antibodies in IHC to examine expression in brain tissues, particularly in microglial cells during neurotropic flavivirus infections
Research indicates MITD1 inhibits flavivirus replication by sequestering specific ESCRT-III proteins involved in viral replication factory formation .
To study MITD1's role in cell division:
Temporal expression analysis: Use time-course Western blot or immunofluorescence with MITD1 antibodies to track expression throughout the cell cycle
Live-cell imaging: Combine MITD1 antibody staining with live-cell microscopy to observe MITD1 localization during cytokinesis
Colocalization studies: Perform dual immunofluorescence with MITD1 antibodies and markers for:
Loss-of-function analysis: Use siRNA knockdown of MITD1 followed by:
Structure-function analysis: Express wild-type and mutant MITD1 (MIT domain mutations or PLD domain mutations) and assess their localization and function using domain-specific antibodies
Research shows MITD1 depletion results in cytokinesis defects, indicated by increased multinucleated cells and membrane instabilities .
When facing contradictory findings about MITD1 in cancer research:
Cancer-specific expression analysis: Use MITD1 antibodies with tissue microarrays to systematically compare expression across multiple cancer types and corresponding normal tissues
Correlation with clinical parameters: Analyze MITD1 expression in relation to:
Cancer stage and grade
Patient survival
Treatment response
Molecular subtypes
Functional studies: Perform MITD1 overexpression and knockdown experiments in multiple cancer cell lines, followed by:
Context-dependent analysis: Consider the cellular context, including:
Molecular pathway analysis: Use MITD1 antibodies in combination with other markers to investigate associations with:
Research indicates MITD1 has dual roles: upregulated in renal cell carcinoma and associated with poor prognosis in some cancers (ACC, GBMLGG, LIHC, KIRC), but inhibits proliferation and migration in breast cancer and correlates with better prognosis in other cancers (BLCA, BRCA, OV, READ) .
To address antibody specificity concerns:
Validation in multiple systems:
Test the antibody across several cell lines with known MITD1 expression
Compare results between different application methods (WB, IHC, IF)
Use positive control recombinant MITD1 protein
Knockdown/knockout validation:
Cross-reactivity assessment:
Test for potential cross-reactivity with related proteins containing MIT domains
Use bioinformatic analysis to identify potential cross-reactive epitopes
Consider blocking peptide experiments if cross-reactivity is suspected
Antibody comparison:
Isotype controls:
When investigating MITD1 in immune contexts:
Cell type-specific analysis:
Activation state assessment:
Analyze MITD1 expression before and after immune cell activation
Correlate with activation markers and cytokine production
Assess the impact of interferon stimulation on MITD1 levels
Tissue-specific immune populations:
Tumor microenvironment analysis:
Functional consequences:
Assess how MITD1 expression affects immune cell function and communication
Investigate impact on cytokine production and cell-cell signaling
Consider potential roles in immune response to viral infection or cancer
To explore MITD1's potential as an immunotherapy biomarker:
Retrospective analysis:
Mechanism exploration:
Multiparameter analysis:
Combine MITD1 expression data with other clinical and molecular parameters
Develop and validate predictive models or nomograms
Assess model performance across different cancer types
Functional validation:
Prospective clinical studies:
Design prospective studies measuring MITD1 expression before immunotherapy
Establish standardized protocols for MITD1 detection and quantification
Define thresholds for "high" versus "low" expression in clinical contexts
Research indicates MITD1 expression is positively correlated with TMB and MSI in several cancers, suggesting potential predictive value for immunotherapy response .
To study MITD1-ESCRT-III interactions:
Co-immunoprecipitation approaches:
Use MITD1 antibodies to pull down MITD1 and associated proteins
Detect specific ESCRT-III components (CHMP1B, IST1) in immunoprecipitates
Perform reciprocal co-IP with antibodies against ESCRT-III proteins
Include appropriate controls (IgG control, MITD1-depleted samples)
Domain-specific interaction analysis:
Structural approaches:
Use purified proteins for crystallography studies
Perform site-directed mutagenesis of key interaction residues
Validate mutant effects using binding assays and functional studies
Localization studies:
Functional consequence assessment:
Research demonstrates that MITD1 interacts with a subset of ESCRT-III proteins through its MIT domain, and this interaction mediates MITD1 recruitment to the midbody during cytokinesis .
For comprehensive pan-cancer MITD1 analysis:
Standardized expression analysis:
Use consistent protocols for MITD1 antibody-based detection
Normalize expression data across different platforms and studies
Apply batch correction methods when integrating multiple datasets
Consider both mRNA and protein expression levels
Multi-omics integration:
Correlate MITD1 protein expression (detected by antibodies) with:
mRNA expression data
Genomic alterations affecting MITD1
Epigenetic modifications
Proteomic profiles
Use bioinformatic tools designed for multi-omics data integration
Clinical annotation correlation:
Maintain consistent clinical parameter definitions across datasets
Apply standardized statistical methods for survival analysis
Use multivariate models to account for confounding factors
Consider cancer subtype-specific analyses
Validation across cohorts:
Test findings from discovery datasets in independent validation cohorts
Assess consistency of results across different patient populations
Implement cross-validation strategies for predictive models
Address potential selection biases in retrospective datasets
Visualization and reporting:
Use consistent visualization methods for comparing MITD1 expression
Report detailed methodological approaches for data integration
Provide access to analysis code and procedures
Address limitations and heterogeneity in integrated datasets
Research demonstrates the value of this approach, revealing that MITD1 expression varies significantly across cancer types, with different prognostic implications depending on the specific cancer context .
When facing contradictory results:
Methodological comparison:
Evaluate differences in experimental approaches:
Antibody sources, clones, and validation methods
Cell lines and culture conditions
Knockdown/overexpression efficiency
Assay sensitivity and specificity
Replicate experiments using standardized protocols
Context-dependent analysis:
Consider cell type-specific effects:
Compare results across different cell lines
Evaluate primary cells versus established cell lines
Assess normal versus cancer cells
Examine microenvironmental influences
Temporal considerations:
Analyze time-dependent effects:
Acute versus chronic MITD1 modulation
Cell cycle phase-specific functions
Time course of cellular responses
Pathway interaction assessment:
Map MITD1 function within signaling networks:
Identify potential compensatory mechanisms
Assess pathway cross-talk
Evaluate feedback regulation
Consider combinatorial effects with other proteins
Systematic review approach:
Apply formal systematic review methodology:
Define clear inclusion/exclusion criteria
Extract data using standardized forms
Assess quality of evidence
Perform meta-analysis where appropriate
Address publication bias and selective reporting