ACOT11 (Acyl-CoA Thioesterase 11) is an enzyme that hydrolyzes acyl-CoA thioesters to free fatty acids and coenzyme A, playing roles in lipid metabolism. Antibodies targeting ACOT11 are used to study its expression and function in metabolic pathways.
Validation: ACOT11 antibodies show specificity in detecting endogenous protein across tissues like liver and adipose .
Pathological Relevance: Dysregulation of ACOT11 is implicated in metabolic syndrome and cancer progression .
Annexin A11 is a calcium-dependent phospholipid-binding protein involved in vesicle trafficking, apoptosis, and RNA metabolism. Mutations in ANXA11 are linked to amyotrophic lateral sclerosis (ALS) and autoimmune disorders.
Validation: ANXA11 antibodies detect cytoplasmic inclusions in ALS patient neurons and validate protein aggregation in ANXA11-mutant models .
Mechanistic Insights:
Feature | ACOT11 Antibody | ANXA11 Antibody |
---|---|---|
Primary Function | Lipid metabolism regulation | Vesicle trafficking, RNA metabolism |
Disease Association | Metabolic disorders, cancer | ALS, autoimmune diseases, muscular dystrophy |
Key Applications | Metabolic studies, cancer research | Neurodegenerative disease research, autoimmune studies |
Commercial Clones | Boster Bio A11581, Aviva OAAB00136 | Proteintech 10479-2-AP, Biomatik CAU22689 |
Annexin A11 (ANXA11) is a member of the annexin family of calcium-dependent phospholipid-binding proteins that plays important roles in various cellular processes. Also known as ANX-A11, ANX11, or CAP50, it functions as a 56 kDa autoantigen and is identified as calcyclin-associated annexin 50 . ANXA11 is encoded by the gene ID 311 in humans with the accession number P50995 . The protein consists of amino acids from Met1 to Asp505 and participates in calcium-dependent vesicle trafficking, cell division, apoptosis, and calcium signaling pathways. ANXA11 has gained significant research interest due to its implications in autoimmune disorders and neurodegenerative diseases, making antibodies against this protein valuable research tools. The protein's structure includes calcium-binding domains that facilitate its interaction with membrane phospholipids, which is essential for its cellular functions.
Validation of ANXA11 antibodies should follow a systematic approach to ensure specificity and reproducibility in research applications. A comprehensive validation protocol includes:
Application-specific testing: ANXA11 antibodies should be validated for specific applications such as Western Blotting (WB), Immunohistochemistry (IHC), Immunocytochemistry (ICC), and Immunoprecipitation (IP) as indicated in the antibody specifications .
Cross-reactivity assessment: Evaluate species reactivity, particularly when working with human samples. Commercial ANXA11 antibodies, such as CAU22689, are often validated specifically for human (Homo sapiens) ANXA11 .
Positive and negative controls: Use cell lines or tissues known to express or lack ANXA11 to confirm specificity.
Molecular weight verification: Confirm that the detected protein band corresponds to the expected molecular weight of ANXA11 (approximately 56 kDa).
Antibody identity confirmation: For rigorous validation, consider using MALDI-TOF MS fingerprinting to confirm antibody identity, especially when obtaining antibodies from third parties . This method can circumvent lengthy denaturation, reduction, alkylation, and enzymatic digestion steps by using a simple formic acid hydrolysis approach that creates distinctive peptide fingerprints .
For optimal results across various experimental applications, researchers should follow these protocol guidelines:
Sample preparation: Lyse cells in a buffer containing protease inhibitors
Protein quantification: Determine concentration via Bradford Assay
SDS-PAGE: Load 10-20 μg protein per lane
Transfer: Use PVDF membrane for optimal protein binding
Blocking: 5% non-fat milk or BSA in TBST for 1 hour at room temperature
Primary antibody incubation: Dilute ANXA11 antibody (typically 1:1000) in blocking buffer and incubate overnight at 4°C
Secondary antibody: Use HRP-conjugated secondary antibody (1:5000) for 1 hour at room temperature
Detection: Develop using ECL substrate and imaging system
Tissue preparation: Fix tissues in 10% neutral buffered formalin and embed in paraffin
Sectioning: Cut 4-6 μm thick sections
Antigen retrieval: Heat-induced epitope retrieval in citrate buffer (pH 6.0)
Blocking: 10% normal serum from the species of secondary antibody
Primary antibody incubation: Apply diluted ANXA11 antibody and incubate overnight at 4°C
Detection: Use appropriate detection system (e.g., ABC method) with DAB as chromogen
Counterstaining: Hematoxylin for nuclear visualization
Cell fixation: 4% paraformaldehyde for 15 minutes at room temperature
Permeabilization: 0.2% Triton X-100 for 10 minutes
Blocking: 5% normal serum for 1 hour
Primary antibody: Incubate with diluted ANXA11 antibody overnight at 4°C
Secondary antibody: Fluorophore-conjugated secondary antibody for 1 hour at room temperature
Nuclear counterstain: DAPI or Hoechst
Mounting: Anti-fade mounting medium
Cell lysis: Use non-denaturing lysis buffer with protease inhibitors
Pre-clearing: Incubate lysate with protein A/G beads
Antibody binding: Add ANXA11 antibody to pre-cleared lysate and incubate overnight at 4°C
Immunoprecipitation: Add protein A/G beads and incubate for 2-4 hours
Washing: Wash beads 4-5 times with lysis buffer
Elution: Boil beads in SDS sample buffer
Analysis: Proceed with SDS-PAGE and Western blotting
Polyclonal and monoclonal ANXA11 antibodies each offer distinct advantages and limitations for research applications:
When selecting between polyclonal and monoclonal antibodies for ANXA11 research, consider the experimental requirements, especially regarding specificity, signal strength, and the nature of the application. For instance, commercial polyclonal ANXA11 antibodies like CAU22689 are validated for WB, IHC, ICC, and IP applications , making them versatile research tools.
Proper controls are essential for reliable ANXA11 antibody-based experiments. Researchers should implement the following controls:
Positive tissue/cell controls: Include samples known to express ANXA11 at detectable levels to validate antibody performance.
Negative tissue/cell controls: Use samples known to lack ANXA11 expression to confirm specificity.
Primary antibody omission control: Perform the immunoassay without the primary ANXA11 antibody to identify non-specific binding of secondary antibodies.
Isotype control: Use a non-specific antibody of the same isotype and concentration as the ANXA11 antibody to identify non-specific binding.
Blocking peptide control: Pre-incubate the ANXA11 antibody with the immunizing peptide (Met1~Asp505 for antibodies like CAU22689 ) before application to confirm specificity.
Concentration gradients: Test different antibody concentrations to determine optimal signal-to-noise ratios.
Cross-reactivity controls: When studying ANXA11 in multiple species, include samples from each species to confirm reactivity.
Loading controls: For Western blots, include housekeeping proteins (e.g., GAPDH, β-actin) to normalize ANXA11 expression.
Molecular weight markers: Include protein ladders to confirm the detected band is of the expected size for ANXA11 (approximately 56 kDa).
Reproducibility controls: Perform technical replicates to ensure consistent results across multiple experiments.
For researchers requiring rigorous confirmation of ANXA11 antibody identity and specificity, several advanced methodological approaches can be employed:
MALDI-TOF MS Fingerprinting:
This technique provides unambiguous antibody identification without lengthy sample preparation. The process involves:
Antibody enrichment and clean-up (approximately 10 μg per sample)
Comparison of fingerprint patterns to identify antibody-specific peak signatures
This method is especially valuable when obtaining antibodies from third parties, as it allows researchers to "undoubtedly identify an antibody to guarantee the traceability of any research activity" . The fingerprinting approach can detect unique peptide signatures (highlighted by the five most intense signals that only occur in the fingerprint spectrum of the respective antibody), enabling reliable antibody identification without requiring full peptide sequence assignment .
Software-Assisted Analysis:
Specialized software tools like "ABID" can assist in managing antibody fingerprint data:
Epitope Mapping:
For ANXA11 antibodies, determining the specific binding epitope can provide crucial information:
Use peptide arrays covering the ANXA11 sequence (Met1~Asp505)
Apply the antibody and detect binding to specific peptide fragments
Confirm results with mutational analysis of key residues
Compare epitope location with known functional domains of ANXA11
When encountering inconsistent results with ANXA11 antibodies, researchers should systematically address potential issues through the following troubleshooting approach:
Verify target specificity through appropriate controls
Assess lot-to-lot variability by comparing antibody performance across batches
Determine optimal antibody concentration through titration experiments
Sample preparation: Ensure complete protein extraction and prevent protein degradation with appropriate protease inhibitors
Antigen accessibility: Optimize fixation and antigen retrieval methods for IHC/ICC applications
Signal detection: Adjust exposure times and detection methods to capture the appropriate signal range
Blocking conditions: Test different blocking reagents to reduce background without compromising specific signal
Signal quantification: Implement appropriate normalization strategies for quantitative comparisons
Reproducibility: Perform technical and biological replicates to assess experimental variability
Positive and negative controls: Include appropriate controls in each experiment
Cross-validation: Confirm results using alternative methods (e.g., mRNA expression, mass spectrometry)
Post-translational modifications: Consider that modifications of ANXA11 might affect antibody recognition
Protein isoforms: Verify which ANXA11 isoforms are recognized by the antibody
Species differences: Ensure the antibody is validated for the species being studied
Tissue/cell-specific factors: Account for matrix effects or endogenous blocking factors
Current research on ANXA11's role in disease pathogenesis employs several sophisticated experimental approaches:
CRISPR/Cas9-mediated knockout or knockin of ANXA11 in cell lines
RNAi-mediated knockdown to study partial loss of function
Overexpression of wild-type or mutant ANXA11 to assess gain-of-function effects
Generation of transgenic animal models with altered ANXA11 expression
Co-immunoprecipitation using ANXA11 antibodies to identify binding partners
Proximity labeling techniques (BioID, APEX) to map the ANXA11 interactome
Yeast two-hybrid screening to identify novel interactions
Fluorescence resonance energy transfer (FRET) to study dynamic interactions in living cells
Calcium-dependent membrane binding assays to assess ANXA11 translocation
Vesicle trafficking analysis using live-cell imaging
Cell cycle progression and apoptosis assays to evaluate ANXA11's role in these processes
Stress response assays to determine ANXA11's function under various cellular stresses
Immunohistochemical analysis of ANXA11 expression in patient samples
Correlation of ANXA11 levels with disease severity or progression
Analysis of ANXA11 variants in patient cohorts
Evaluation of ANXA11 as a potential biomarker for disease diagnosis or prognosis
Integration of mass spectrometry with antibody-based detection creates powerful approaches for ANXA11 research:
Process samples for MS analysis through digestion with trypsin
Perform LC-MS/MS to identify ANXA11 and its interacting partners
Quantify relative abundances of proteins in the immunoprecipitated complex
This combined approach allows researchers to confirm ANXA11 antibody specificity while simultaneously identifying novel protein interactions and post-translational modifications.
Develop targeted MS assays for specific ANXA11 peptides
Use antibody-based enrichment to increase ANXA11 concentration prior to MS analysis
Quantify ANXA11 abundance with high specificity and sensitivity
Detect specific post-translational modifications on ANXA11
Antibody Validation by MS:
The MALDI-TOF MS fingerprinting methodology described in the literature provides a rapid approach for antibody validation:
Clean-up and enrichment of IgG antibodies
Formic acid hydrolysis (generating distinctive peptide patterns)
Comparison with reference spectra for identification
This method can be used to confirm the identity of ANXA11 antibodies obtained from third parties or to track antibody consistency across experiments .
For reliable quantitative analysis of ANXA11 expression, researchers should implement the following protocols:
Sample preparation: Standard lysis with protease inhibitors
Protein quantification: Bradford Assay to ensure equal loading
Standard curve generation: Include a dilution series of recombinant ANXA11
Loading controls: Include housekeeping proteins for normalization
Signal detection: Use digital imaging systems with appropriate dynamic range
Densitometric analysis: Measure band intensity using specialized software
Normalization: Calculate relative ANXA11 expression normalized to loading controls
Statistical analysis: Perform appropriate statistical tests for comparing groups
Tissue processing: Standardize fixation and antigen retrieval
Staining protocol: Use automated staining platforms for consistency
Controls: Include positive and negative controls on each slide
Digital image acquisition: Capture images under consistent conditions
Computer-assisted analysis: Use specialized software for quantifying staining intensity
Region of interest selection: Define consistent regions for analysis
Scoring system development: Implement H-score or Allred score for semi-quantitative analysis
Blind assessment: Have multiple observers score slides independently
Cell preparation: Optimize fixation and permeabilization for intracellular ANXA11 staining
Antibody titration: Determine optimal concentration for maximal signal-to-noise ratio
Multi-parameter analysis: Include markers to identify relevant cell populations
Controls: Incorporate isotype controls and unstained samples
Compensation: Adjust for spectral overlap when using multiple fluorophores
Data acquisition: Collect sufficient events for statistical analysis
Gating strategy: Implement consistent gating approach across samples
Quantification: Report median fluorescence intensity or percent positive cells
Plate coating: Use capture antibody at optimized concentration
Blocking: Reduce non-specific binding with appropriate blocking buffer
Standard curve: Include recombinant ANXA11 standards
Sample dilution: Test multiple dilutions to ensure measurements within linear range
Detection: Use conjugated detection antibody or secondary antibody system
Signal development: Optimize timing for consistent results
Data analysis: Generate standard curve using appropriate regression method
Quality control: Include internal controls to assess plate-to-plate variability
ANXA11 antibody-based research has revealed important connections between ANXA11 and neurodegenerative diseases. Immunohistochemical studies using ANXA11 antibodies have identified altered expression patterns in neurodegenerative diseases, particularly amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). These studies have demonstrated that ANXA11 participates in RNA granule dynamics and stress granule formation, processes critical to neuronal function and survival. Through immunoprecipitation experiments, researchers have identified ANXA11 interactions with proteins involved in RNA metabolism and vesicle trafficking, providing insight into disease mechanisms. Additionally, analysis of patient samples using ANXA11 antibodies has revealed that ANXA11 mutations can lead to protein aggregation and disruption of calcium homeostasis, contributing to neurodegeneration. ANXA11 antibody studies have also shown that the protein plays roles in autophagy and calcium-dependent vesicular transport, functions that may be compromised in neurodegenerative diseases.
Detection of post-translational modifications (PTMs) on ANXA11 is critical for understanding its functional regulation. Several emerging techniques combine antibody-based approaches with advanced analytical methods:
Phospho-specific antibodies: Development of antibodies recognizing specific phosphorylated residues on ANXA11 enables direct detection of this important regulatory modification.
Combined IP-MS approach: Immunoprecipitation with ANXA11 antibodies followed by mass spectrometry analysis allows comprehensive mapping of multiple PTMs simultaneously.
Proximity ligation assay (PLA): This technique can detect specific ANXA11 modifications in situ with high sensitivity by using antibodies against both ANXA11 and the modification of interest.
Phos-tag SDS-PAGE: When combined with ANXA11 antibodies for Western blotting, this specialized electrophoresis technique improves separation of phosphorylated ANXA11 forms.
ELISA-based PTM detection: Development of sandwich ELISA systems using ANXA11 capture antibodies and PTM-specific detection antibodies enables quantitative assessment of modified ANXA11.
MALDI-imaging mass spectrometry: This technique allows spatial visualization of ANXA11 PTMs in tissue sections when combined with antibody-based enrichment strategies.
ANXA11 antibodies can be adapted for various high-throughput screening approaches:
Culture cells in 96- or 384-well plates
Perform automated fixation, permeabilization, and staining with ANXA11 antibodies
Use high-content imaging systems for automated image acquisition
Implement image analysis algorithms to quantify ANXA11 expression, localization, or aggregation
Screen compounds for effects on ANXA11 biology
Spot cell or tissue lysates onto nitrocellulose-coated slides
Probe with validated ANXA11 antibodies
Use signal amplification systems for detection
Analyze large sample sets simultaneously for ANXA11 expression
Correlate findings with clinical outcomes or experimental conditions
Couple ANXA11 antibodies to uniquely coded microbeads
Incubate with samples and detection antibodies
Analyze using flow cytometry-based platforms
Simultaneously measure ANXA11 alongside other proteins of interest
Process hundreds of samples with minimal sample consumption
These high-throughput approaches enable efficient screening of drug candidates, genetic modifiers, or disease biomarkers related to ANXA11 function, significantly accelerating discovery in this field.