MICAL1 (microtubule associated monoxygenase, calponin and LIM domain containing 1) is a multidomain flavoprotein monooxygenase with diverse cellular functions. It has four conserved domains: an N-terminal flavin adenine dinucleotide (FAD) binding domain, a calponin homology (CH) domain, a Lin11, Isl-1 and Mec-3 (LIM) domain, and a C-terminal coiled-coil (CC) domain . MICAL1 regulates actin stress fibers and is required for normal actin organization by promoting depolymerization of F-actin through oxidation of specific methionine residues. It may also be involved in apoptosis through binding with NDR (nuclear Dbf2-related) kinases . Recent research has implicated MICAL1 in cancer progression, cell migration, and viral processes, making it an important target for antibody-based studies.
MICAL1 antibodies are available in both monoclonal and polyclonal forms, with various host species including mouse and rabbit. The primary applications include:
These antibodies have been validated in multiple species, with most showing reactivity to human and mouse MICAL1, while some also react with rat samples .
Storage conditions vary depending on the specific antibody formulation:
Unconjugated antibodies in PBS only should be stored at -80°C for maximum stability
Antibodies in PBS with 0.02% sodium azide and 50% glycerol should be stored at -20°C for long-term storage
For frequent use over short periods (up to one month), 4°C storage may be suitable for some formulations
Avoid repeated freeze-thaw cycles as they can compromise antibody quality and performance
Always refer to the manufacturer's specific recommendations for your particular antibody.
When performing Western blot analysis for MICAL1:
Sample preparation: Use appropriate lysis buffers containing protease inhibitors to prevent degradation
Gel selection: Since MICAL1 has a high molecular weight (~118-120 kDa), use lower percentage gels (8-10% SDS-PAGE) for better resolution
Transfer conditions: Longer transfer times or higher voltage may be necessary for efficient transfer of this high molecular weight protein
Blocking: 5% non-fat milk or BSA in TBST is typically effective
Antibody dilution: Start with manufacturer's recommended dilution (often 1:2000-1:16000) , but optimize as needed
Positive controls: Include lysates from cells known to express MICAL1 (e.g., Jurkat cells, HEK-293 cells, T-47D cells, HeLa cells)
Detection: Enhanced chemiluminescence (ECL) is usually sufficient, but for lower expression levels, more sensitive detection systems may be required
Importantly, the observed molecular weight of MICAL1 is typically around 120 kDa, slightly higher than the calculated 118 kDa , which is common for many proteins due to post-translational modifications.
Proper validation of MICAL1 antibody specificity is critical for reliable results:
Positive and negative controls:
Multiple detection methods:
Compare results across different applications (WB, IHC, IF) to ensure consistent detection patterns
Use antibodies targeting different epitopes of MICAL1 to confirm specificity
Immunogen competition:
Multiple antibody validation:
Compare results using antibodies from different sources or clones
Look for consistent results across monoclonal and polyclonal antibodies
Molecular weight verification:
Confirm that the detected band matches the expected molecular weight (~120 kDa)
Be aware of potential splice variants or post-translational modifications
To investigate MICAL1 protein-protein interactions:
Co-immunoprecipitation:
Use anti-MICAL1 antibodies for IP followed by Western blotting for suspected binding partners
Alternatively, IP the suspected partner and blot for MICAL1
Include appropriate controls (IgG, lysate inputs)
Consider crosslinking for transient interactions
Proximity ligation assay (PLA):
Useful for detecting proteins that interact in situ
Requires antibodies raised in different species
Provides spatial information about interactions
Pull-down assays:
FRET/BRET approaches:
For real-time interaction studies in living cells
Requires fluorescent/luminescent protein tagging
Yeast two-hybrid screening:
For discovery of novel interaction partners
Follow up with biochemical validation
When studying MICAL1 interactions, consider its domain structure and known binding regions. For example, MICAL1 binds to MyoVa through a GTBM (GTD-binding motif), which is present in MICAL1 but not in MICAL2 or MICAL3 .
MICAL1 promotes F-actin disassembly through oxidation of specific methionine residues. To study this function:
Immunofluorescence co-localization:
Use anti-MICAL1 antibodies alongside F-actin staining (phalloidin)
Analyze co-localization patterns in various cellular contexts
Time-course experiments can reveal dynamics of recruitment
Live-cell imaging with MICAL1 knockdown/rescue:
In vitro actin disassembly assays:
Immunoprecipitate MICAL1 from cells under different conditions
Test activity on purified F-actin
Compare with recombinant MICAL1
F-actin quantification following manipulation:
Research has demonstrated that MICAL1 knockdown significantly increases F-actin levels at structures like the midbody during cytokinesis, while overexpression of wild-type MICAL1 (but not catalytically inactive MICAL1-3G3W) reduces F-actin levels .
MICAL1 exists in an autoinhibited state that can be activated by various mechanisms:
Domain deletion/mutation analysis:
Create constructs lacking specific domains (MO, CH, LIM, CC)
Test activity using actin depolymerization assays
Use antibodies specific to different domains to track conformational changes
Study of activator interactions:
Structural studies supported by antibody validation:
Phosphorylation analysis:
Evidence suggests MICAL1 activity may be regulated by phosphorylation
Use phospho-specific antibodies if available, or general phospho-antibodies after IP
Mass spectrometry analysis of immunoprecipitated MICAL1 can identify modification sites
Recent research indicates that "MICAL1 autoinhibition hinges on the binding of the CH and LIM domains, facilitated by a helical region of the long linker, to the CC domain" . During Rab-induced activation, Rab binding to the CC domain likely triggers conformational changes that destabilize this autoinhibitory interaction.
MICAL1 has been implicated in multiple cancer types, including pancreatic cancer and renal clear cell carcinoma. To investigate its role:
Expression analysis in patient samples:
IHC staining of tissue microarrays with anti-MICAL1 antibodies
Compare expression between tumor and adjacent normal tissues
Correlate with clinical parameters and survival data
Functional studies in cancer cell lines:
Generate stable MICAL1-overexpressing or MICAL1-silencing cells
Assess effects on:
Proliferation (CCK-8, colony formation, EdU assays)
Migration and invasion (wound healing, Transwell assays)
In vivo tumor growth and metastasis models
Mechanism investigation:
Therapeutic targeting:
Test effects of inhibiting MICAL1 on cancer cell sensitivity to treatments
Explore combination approaches
Develop screening assays for MICAL1 inhibitors
Multiple bands in MICAL1 Western blots could result from:
Alternative splicing:
MICAL1 may have splice variants
Consult databases for known splice variants and their predicted molecular weights
Use antibodies targeting different epitopes to confirm identity
Post-translational modifications:
Phosphorylation, ubiquitination, or other modifications can alter mobility
Use phosphatase treatment to test for phosphorylation
Mass spectrometry analysis can identify modifications
Proteolytic degradation:
MICAL1 may be sensitive to proteolysis during sample preparation
Use fresh samples with complete protease inhibitor cocktails
Optimize lysis and sample handling conditions
Cross-reactivity:
Antibodies may recognize related proteins (MICAL2, MICAL3, MICAL-L1, MICAL-L2)
Validate using knockout/knockdown controls
Compare results with different antibodies
Non-specific binding:
Optimize blocking conditions and antibody dilutions
Try different blocking agents (milk vs. BSA)
Increase washing stringency
The full-length MICAL1 protein is expected at ~120 kDa, with potential fragments at lower molecular weights depending on the epitope recognized by your antibody.
When facing inconsistent results between different MICAL1 antibodies:
Epitope mapping:
Determine the epitope regions recognized by each antibody
Different domains may have different accessibility in various experimental conditions
Some antibodies target N-terminal regions (e.g., FAD domain) while others target C-terminal regions (e.g., CC domain)
Validation strategy:
Use MICAL1 knockdown/knockout samples as negative controls
Test all antibodies against the same positive and negative controls
Include recombinant MICAL1 as a standard when possible
Application optimization:
Each antibody may require specific optimization for different applications
Adjust fixation methods for IF/IHC (aldehyde vs. organic solvent)
Try different antigen retrieval methods for IHC
Modify blocking and incubation conditions
Conformational considerations:
MICAL1 has an autoinhibited conformation where some epitopes may be masked
Activation (e.g., by RAB proteins) may expose additional epitopes
Denaturing vs. native conditions may affect epitope accessibility
Cross-validation approaches:
Use alternative detection methods (e.g., mass spectrometry)
Tag endogenous MICAL1 using CRISPR knock-in strategies
Employ proximity labeling approaches
For example, when studying MICAL1's role in cytokinesis, researchers validated their findings using both siRNA knockdown and rescue experiments with siRNA-resistant MICAL1 constructs .
MICAL1 shows distinct localization patterns depending on cellular context:
Typical localization patterns:
Factors affecting localization:
Cell type and physiological state
Cell cycle stage (particularly evident during cytokinesis)
Activation status of MICAL1
Fixation and permeabilization methods
Validation approaches:
Compare multiple antibodies targeting different epitopes
Use tagged MICAL1 constructs (being careful about potential artifacts)
Include domain deletion mutants to map localization determinants
Perform co-localization with known markers and partners
Advanced imaging techniques:
Super-resolution microscopy for detailed localization studies
Live-cell imaging with fluorescently tagged MICAL1
FRAP (Fluorescence Recovery After Photobleaching) for dynamics studies
Research has shown that "MICAL1 does not directly interact with Rab11a" but "drives the accumulation of Rab11a-positive vesicles at the midbody by binding to MyoVa" , highlighting the importance of careful interpretation of co-localization data.
Recent research has implicated MICAL1 in viral infection processes, particularly in HIV-1 budding:
Virus budding studies:
Use MICAL1 antibodies to track localization during viral budding
Compare infected vs. uninfected cells
Co-localization with viral components
Super-resolution microscopy to visualize structural details
Mechanistic investigations:
RAB35-MICAL1 pathway:
Branched actin networks:
Research shows that "viral release can be restored in MICAL1-depleted cells by inhibiting Arp2/3-dependent branched actin networks" , suggesting a specific role in remodeling branched actin during viral budding.
MICAL1's role in immune responses is an emerging area of research:
Expression analysis in immune cell subsets:
Use flow cytometry with anti-MICAL1 antibodies to profile expression across immune cell types
Examine changes in expression upon activation/differentiation
Compare expression in healthy vs. disease states
T cell exhaustion studies:
Immune infiltration analysis:
Signaling pathway analysis:
Bioinformatic analyses have revealed that "MICAL1 expression had strong relationships with various T cell exhaustion markers" and is "positively associated with CD8+/Treg cell infiltration levels" , suggesting important but not fully characterized roles in immune function.
Development and application of specialized MICAL1 antibodies could provide unprecedented insights:
Phospho-specific antibodies:
Conformation-specific antibodies:
Domain-specific antibodies:
Target individual domains (MO, CH, LIM, CC) to study domain-specific functions
Use to track domain accessibility during activation/inactivation
Combine with functional studies using domain deletion/mutation constructs
Applications in high-throughput screening:
Develop ELISA or flow cytometry-based assays using conformation-specific antibodies
Screen for compounds that modulate MICAL1 activation
Potential therapeutic applications in cancer or viral infections
The recent cryo-EM structure showing that "MICAL1 autoinhibition hinges on the binding of the CH and LIM domains... to the CC domain" provides structural insights that could guide the development of such specialized antibodies.