AMOTL1 (angiomotin-like protein 1) is a peripheral membrane protein that functions as a component of tight junctions. It plays crucial roles in regulating cell polarity, paracellular permeability, and cell migration . AMOTL1 is particularly significant for research because:
It inhibits the Wnt/beta-catenin signaling pathway by recruiting CTNNB1 to recycling endosomes, preventing its nuclear translocation
It functions in the Hippo signaling pathway through interaction with YAP1, influencing cell proliferation and tissue growth
Mutations in AMOTL1, particularly affecting amino acids 157-161, are associated with a newly defined orofacial clefting syndrome with congenital heart disease and other manifestations
It has been implicated in various cancers, including gastric and cervical cancer, making it a potential therapeutic target
The protein has a calculated and observed molecular weight of 107 kDa, with the gene located at chromosome 11q21 .
For maintaining optimal antibody performance, follow these research-validated protocols:
Small volume options (20μl) often contain 0.1% BSA as a stabilizer . For experimental reproducibility, document the specific lot number and storage duration in your lab notebook.
AMOTL1 antibodies have been validated for multiple applications with specific optimal dilutions:
Remember that optimal dilutions may be sample-dependent. It is always recommended to titrate the antibody in your specific experimental system to obtain optimal results . Use positive controls such as HeLa cells, which are known to express AMOTL1, to validate your protocols .
To investigate the critical AMOTL1-YAP1 interaction that has been implicated in cancer progression, employ these methodological approaches:
Co-immunoprecipitation (Co-IP):
Cyclohexamide (CHX) chase assay for protein stability studies:
Ubiquitination assays:
Transfect cells with HA-tagged ubiquitin (HA-Ub)
Treat with proteasome inhibitor MG132
Perform immunoprecipitation with anti-YAP1 antibody
Detect ubiquitination levels by western blot using anti-HA antibody
Compare conditions with varying AMOTL1 expression levels to assess how AMOTL1 influences YAP1 ubiquitination
Subcellular fractionation:
Research has shown that AMOTL1 and YAP1 protect each other from ubiquitin-mediated degradation, and AMOTL1 promotes YAP1 translocation into the nucleus to activate downstream expression of genes like CTGF .
Based on published research methodologies, effective experimental approaches include:
In vitro functional assays following AMOTL1 knockdown/overexpression:
Cell proliferation: MTT assay or EdU incorporation assay (analyze at 0, 24, 48, 72, and 96 hours)
Colony formation: Monolayer colony formation assay to assess clonogenic potential
Cell invasion: Transwell invasion assays with Matrigel coating
Cell cycle analysis: Flow cytometry with PI staining; confirm with western blot for cell cycle markers (p21/p27, pRb)
Apoptosis assessment: 7AAD and Annexin V double staining to distinguish early and late apoptosis
Signaling pathway analysis:
In vivo tumor models:
Drug sensitivity testing:
Research has demonstrated that AMOTL1 knockdown significantly inhibits cancer cell proliferation, colony formation, and invasion capabilities while inducing cell cycle arrest and apoptosis .
To ensure high specificity and reliable results, implement these validation strategies:
Multiple antibody approach:
Genetic validation:
Species cross-reactivity assessment:
For studies using non-human models, conduct sequence alignment analysis between human AMOTL1 and your species of interest
Predicted reactivity based on sequence homology: Cow (93%), Dog (79%), Guinea Pig (100%), Mouse (100%), Rabbit (100%)
Perform pilot tests in your specific model organism if the antibody hasn't been validated for that species
Positive control selection:
Antibody titration:
For high-sensitivity applications, consider using recombinant monoclonal antibodies like EPR11803-97, which may offer greater consistency between lots compared to polyclonal antibodies .
For discriminating between closely related AMOTL family proteins or detecting specific AMOTL1 isoforms:
Selective antibody targeting:
Western blot optimization:
Use gradient gels (4-12%) for better separation of high molecular weight proteins
Extended run times can improve separation between AMOTL1 (107 kDa) and AMOTL2
Include positive controls expressing single family members for comparison
qRT-PCR validation:
RNA interference specificity:
Research indicates that in gastric cancer cell lines, AMOT is barely expressed, while AMOTL1 and AMOTL2 show relatively high expression levels, emphasizing the importance of discriminating between family members in cancer studies .
Recent research has identified AMOTL1 mutations associated with orofacial clefting syndrome and altered brain development. To investigate these roles:
Tissue-specific expression profiling:
Brain-specific analyses:
Mutation impact studies:
Metabolomic correlation:
Combine antibody-based detection of AMOTL1 expression with brain metabolite analyses
Focus on N-Acetylaspartylglutamic acid (NAAG)/N-Acetyl Aspartate (NAA) ratio and Glutamine (Gln) levels, which were altered in Amotl1-depleted brains
This approach can help connect AMOTL1 dysfunction to neurotransmitter metabolism
Research in Amotl1 mutant mice has revealed behavioral phenotypes including hyperactivity, reduced anxiety, altered social behavior, and episodes of backward walking and increased circling behavior, potentially modeling aspects of human neuropsychiatric disorders .
To investigate AMOTL1 regulation through post-translational modifications:
Phosphorylation studies:
Perform western blot using phospho-specific antibodies or phospho-staining
Alternatively, use mass spectrometry to identify phosphorylation sites
Compare phosphorylation status under different cellular conditions (e.g., growth factor stimulation, cell density)
Ubiquitination detection:
Protein stability assessment:
Subcellular localization changes:
Use cell fractionation followed by western blot or immunofluorescence
Monitor AMOTL1 localization changes in response to cell density, mechanical stress, or other stimuli
These approaches can reveal regulation through compartmentalization
Research indicates complex regulatory mechanisms where AMOTL1 and YAP1 reciprocally protect each other from degradation through the ubiquitin-proteasome system, suggesting that post-translational modifications significantly influence AMOTL1 function in signaling pathways .
When working with tissues where AMOTL1 expression is limited, implement these sensitivity-enhancing techniques:
Signal amplification methods:
Sample preparation optimization:
Antibody selection and concentration:
Detection system enhancement:
Implement fluorescence-based detection with sensitive cameras for IF
For colorimetric IHC, optimize DAB development times and consider alternate chromogens
Use enhanced chemiluminescence (ECL) kits with extended exposure times for western blots
Published protocols have successfully detected AMOTL1 in various tissue samples using techniques such as immunohistochemistry with DAB visualization and hematoxylin counterstaining .
When facing inconsistent results between different AMOTL1 antibodies or detection methods:
Epitope mapping analysis:
Multiple detection technique validation:
Experimental condition standardization:
Control cell density, passage number, and culture conditions
Standardize lysis buffers and protein extraction protocols
Document lot numbers of antibodies and reagents
Functional validation:
Species-specific considerations:
Remember that AMOTL1 function can be context-dependent, with different subcellular localizations (cytosolic in mesenchymal cells, tight junctions and adherens junctions in polarized epithelia) .
Based on recent findings, AMOTL1-targeted approaches offer promising avenues for therapeutic development:
Biomarker potential:
Therapeutic target validation:
Combination therapy assessment:
Circulating AMOTL1 detection:
Recent research demonstrates that AMOTL1 knockout in xenograft models significantly reduces tumor size and weight, while decreasing expression of proliferation markers and increasing apoptotic markers, supporting its potential as a therapeutic target .
Recent discoveries linking AMOTL1 mutations to a novel orofacial clefting syndrome open new research directions:
Genotype-phenotype correlation studies:
Use antibodies to assess how different AMOTL1 variants (particularly those affecting amino acids 157-161) impact protein expression, stability, and localization
Compare findings with patient phenotypes to establish mechanistic connections
Develop immunohistochemistry protocols for patient-derived samples
Developmental pathway analysis:
Animal model validation:
Tissue-specific effects:
The recent identification of heterozygous missense variants in AMOTL1 defining a new orofacial clefting syndrome highlights the importance of understanding this protein's role in human development and disease .