ATIC (5-Aminoimidazole-4-Carboxamide Ribonucleotide Formyltransferase/IMP Cyclohydrolase) is a bifunctional enzyme critical for de novo purine biosynthesis. ATIC antibodies are specialized immunoglobulins designed to detect and quantify this enzyme in research and clinical contexts. These antibodies play pivotal roles in studying ATIC's involvement in cancer progression, metabolic regulation, and therapeutic targeting .
Protein Structure: A 65 kDa enzyme with two functional domains:
Post-Translational Modifications: Glycosylation sites in the Fc region influence effector functions .
Commercial ATIC antibodies (e.g., Proteintech 10726-1-AP, Abcam ab33520) target regions such as the C-terminus (AA 301–592) or middle domains (AA 379–428) .
ATIC antibodies are validated for multiple techniques:
Mechanism: ATIC promotes tumor growth by suppressing AMPK and activating mTOR-S6K1 signaling, enhancing proliferation and migration .
Prognostic Value: High ATIC expression correlates with poor survival (HR = 1.82, P < 0.001) .
Regulatory Role: ATIC upregulates Myc expression, driving cell cycle progression and metastasis .
Therapeutic Target: Knockdown reduces tumor growth by 60% in xenograft models .
ATIC inhibits autophagy in HCC via the AKT/FOXO3 pathway, increasing chemoresistance .
HCC Progression (PMC5732395):
LUAD Mechanisms (PMC8895470):
Immune Microenvironment:
ATIC (5-Aminoimidazole-4-Carboxamide Ribonucleotide Formyltransferase/IMP Cyclohydrolase) is a bifunctional enzyme that catalyzes the last two steps of de novo purine biosynthesis. This 65 kDa protein acts as a transformylase that incorporates a formyl group to the AMP analog AICAR (5-amino-1-(5-phospho-beta-D-ribosyl)imidazole-4-carboxamide) to produce formyl-AICAR (FAICAR), and subsequently catalyzes the cyclization of FAICAR to inosine monophosphate (IMP) .
ATIC has been implicated in several cellular processes beyond purine biosynthesis:
It can convert thio-AICAR to 6-mercaptopurine ribonucleotide, a purine biosynthesis inhibitor used in leukemia treatment
It promotes insulin receptor (INSR) autophosphorylation and is involved in INSR internalization
It has been reported to participate in myeloma and hepatocellular carcinoma progression
Selecting the right ATIC antibody requires careful consideration of several factors:
Determine your application: Different ATIC antibodies are validated for specific applications such as Western blot (WB), immunohistochemistry (IHC), immunofluorescence (IF), flow cytometry (FC), and ELISA. Review the validation data for your specific application .
Species reactivity: Verify that the antibody recognizes ATIC in your species of interest. Some ATIC antibodies show reactivity across multiple species with high sequence homology, like human, mouse, rat, and other mammals .
Antibody format: Consider whether a monoclonal or polyclonal antibody better suits your needs:
Validation evidence: Review published literature citations and validation data including Western blot images, ICC/IF images, or flow cytometry data .
Epitope information: Understanding the binding region (e.g., N-terminal, C-terminal, or middle region) can be important, especially if studying specific domains or protein fragments .
Proper validation of ATIC antibodies should include multiple approaches:
Optimizing ATIC antibody dilution for Western blotting requires systematic testing:
Start with manufacturer recommendations: Begin with the suggested dilution range (e.g., 1:2000-1:12000 for some ATIC antibodies) .
Perform a dilution series: Test 3-4 dilutions across the recommended range using the same sample.
Control selection: Include positive controls (cell lines known to express ATIC, such as HeLa, HCT116, or Jurkat cells) and negative controls (if available) .
Blocking optimization: Use 5% non-fat dry milk or BSA in TBS-T for blocking; optimize if background is high.
Incubation conditions: Test both 1-hour room temperature and overnight 4°C primary antibody incubations to determine optimal conditions.
Evaluation criteria: Select the dilution that provides:
Strong specific bands at the expected molecular weight (65 kDa for ATIC)
Minimal background
Optimal signal-to-noise ratio
Some ATIC antibodies may require specific optimization based on their formulation and the protein abundance in your samples .
Successful immunofluorescence with ATIC antibodies requires attention to several factors:
Fixation method: Test both paraformaldehyde (4%) and methanol fixation to determine which best preserves ATIC epitopes while maintaining cellular architecture.
Permeabilization: Since ATIC is primarily cytoplasmic, mild permeabilization with 0.1-0.3% Triton X-100 is typically sufficient.
Blocking: Use 5-10% normal serum from the same species as the secondary antibody to reduce non-specific binding.
Antibody dilution: Begin with the manufacturer's recommended dilution range (e.g., 1:300-1:1200) and optimize as needed .
Incubation time and temperature: Test both 1-hour room temperature and overnight 4°C incubations for primary antibody.
Controls:
Positive control: Cell lines with known ATIC expression (e.g., HeLa cells)
Negative control: Primary antibody omission
If available, siRNA knockdown or CRISPR knockout cells
Co-staining considerations: When performing double immunostaining, ensure secondary antibodies don't cross-react and choose primary antibodies raised in different species .
Signal amplification: For low-abundance detection, consider using tyramide signal amplification or other amplification methods.
Non-specific binding is a common challenge when working with antibodies. Here are systematic approaches to troubleshoot this issue with ATIC antibodies:
Increase blocking stringency:
Extend blocking time to 2 hours or overnight
Try different blocking agents (BSA, normal serum, commercial blockers)
Add 0.1-0.3% Triton X-100 to blocking buffer for membrane permeabilization
Optimize antibody conditions:
Further dilute primary antibody
Reduce incubation temperature (4°C instead of room temperature)
Add 0.1-0.5% Tween-20 to antibody dilution buffer
Try shorter incubation times
Washing optimization:
Increase number of wash steps
Extend washing duration
Add higher concentrations of detergent to wash buffer
Validate specificity:
Test on known positive and negative control samples
Perform peptide competition assay using the immunogen
If possible, test on ATIC knockdown/knockout samples
Secondary antibody considerations:
ATIC antibodies can serve as valuable tools in investigating purine metabolism disorders through several advanced applications:
Expression level analysis: Quantify ATIC protein levels in patient-derived samples compared to healthy controls using Western blotting or immunohistochemistry. Alterations in ATIC expression may correlate with disease severity or progression .
Enzyme activity correlation: Combine ATIC antibody-based detection with functional assays measuring ATIC enzymatic activity to establish relationships between protein levels and functional outcomes.
Protein-protein interaction studies:
Use ATIC antibodies for co-immunoprecipitation to identify interaction partners
Perform proximity ligation assays to visualize and quantify interactions in situ
Combine with mass spectrometry to identify novel interaction networks
Subcellular localization: Employ immunofluorescence with ATIC antibodies to track potential changes in subcellular distribution in disease states.
Post-translational modification analysis: Use modification-specific antibodies in conjunction with ATIC antibodies to assess how post-translational modifications affect ATIC function in disease contexts.
Therapeutic response monitoring: Measure changes in ATIC expression or localization following therapeutic interventions targeting purine metabolism.
Patient stratification: Develop immunohistochemistry-based assays using validated ATIC antibodies to potentially classify patients based on ATIC expression patterns .
Understanding the specific epitope recognized by an ATIC antibody is valuable for interpretation of results and experimental design. Several complementary approaches can be used:
Peptide array analysis:
Create an overlapping peptide array covering the ATIC sequence
Probe with the antibody of interest
Identify reactive peptides to narrow down binding regions
Truncation mutants:
Generate a series of ATIC truncation constructs
Express in mammalian or bacterial systems
Perform Western blotting to identify the minimal region required for antibody binding
Domain swapping:
Create chimeric proteins with domains from ATIC and an unrelated protein
Test antibody reactivity to identify the domain containing the epitope
Site-directed mutagenesis:
Once a potential epitope region is identified, introduce point mutations
Test mutants for altered antibody binding
Critical residues will significantly reduce binding when mutated
Hydrogen-deuterium exchange mass spectrometry (HDX-MS):
Compare HDX patterns of ATIC alone versus ATIC-antibody complex
Regions protected from exchange in the complex indicate binding sites
X-ray crystallography or cryo-EM: For definitive epitope mapping, determine the structure of the antibody-ATIC complex .
Some ATIC antibodies already have characterized epitopes, such as those targeting aa 379-428 or other specific regions, which can inform experimental design and interpretation .
Multiplexed detection allows simultaneous visualization of ATIC along with other proteins of interest. Advanced strategies include:
Multicolor immunofluorescence:
Select primary antibodies from different host species (e.g., rabbit anti-ATIC and mouse anti-target2)
Use spectrally distinct fluorophore-conjugated secondary antibodies
Include proper controls to ensure no cross-reactivity between secondary antibodies
Consider using directly conjugated primary antibodies for more than 3-4 targets
Sequential immunostaining:
Perform complete staining with first primary and secondary antibodies
Use elution buffer to remove antibodies without affecting tissue morphology
Repeat staining with subsequent antibody pairs
Useful when antibodies are from the same host species
Mass cytometry (CyTOF):
Conjugate ATIC antibodies with distinct metal isotopes
Allows for highly multiplexed detection (30+ parameters)
Particularly useful for analyzing complex cellular systems
Proximity ligation assay (PLA):
Use ATIC antibody in combination with antibodies against potential interaction partners
Generates fluorescent signals only when targets are in close proximity (<40 nm)
Enables visualization of protein-protein interactions in situ
Multiplex immunohistochemistry:
Optimization of each antibody individually before multiplexing is critical for successful outcomes.
Antibody degradation can compromise experimental results. These approaches help assess ATIC antibody stability:
Regular quality control testing:
Run Western blot on standard positive control (e.g., HeLa cell lysate)
Compare signal intensity and background to results from when antibody was new
Document with images for reference
Appearance assessment:
Check for visible precipitation, cloudiness, or color changes
Gently mix and observe if any particles remain undissolved
Stability measurements:
If equipped, use dynamic light scattering to check for aggregation
Analytical size exclusion chromatography can assess monomer content
Storage guidelines enforcement:
Activity monitoring timeline:
Test new batch immediately upon receipt to establish baseline
Retest after 3, 6, and 12 months to monitor stability
Document usage conditions and number of freeze-thaw cycles
Side-by-side comparison:
If possible, maintain a small reference aliquot from initial use
Compare performance of current working aliquot with reference sample
When different ATIC antibodies yield conflicting results, systematic investigation is required:
Epitope mapping comparison:
Validation with orthogonal methods:
Sample preparation assessment:
Test different lysis buffers, fixation methods, and antigen retrieval protocols
Some epitopes may be sensitive to specific preparation conditions
Antibody format consideration:
Independent validation:
Have different lab members perform experiments blindly
Collaborate with other labs to test antibodies under different conditions
Experimental design optimization:
Systematically modify blocking agents, incubation times, temperatures
Test different detection systems (chemiluminescence vs. fluorescence)
Literature reconciliation:
Validating antibody specificity in complex tissue samples requires rigorous approaches:
Genetic validation (gold standard):
Test on tissues from ATIC knockout/knockdown models
Compare with wild-type tissues processed identically
Specific signal should be absent or significantly reduced in knockout samples
Pre-absorption controls:
Pre-incubate antibody with excess purified ATIC protein or immunizing peptide
Apply to adjacent tissue sections
Specific binding should be blocked in pre-absorbed samples
Orthogonal method correlation:
Compare protein localization with mRNA expression using in situ hybridization
Patterns should generally correlate, though post-transcriptional regulation may cause differences
Multiple antibody verification:
Test multiple ATIC antibodies targeting different epitopes
Consistent staining patterns increase confidence in specificity
Western blot correlation:
Perform Western blot on tissue lysates from the same source
Confirm single band at expected molecular weight (65 kDa)
Compare relative expression levels across tissues with IHC staining intensity
Cell type-specific markers:
Machine learning is revolutionizing antibody research, including applications relevant to ATIC antibodies:
Binding prediction models:
Library-on-library approaches can screen many antibodies against many antigens
Machine learning models analyze relationships between antibodies and antigens
Models can predict binding even for antibodies and antigens not in training data
Active learning strategies can reduce experimental data needed by 35%
Epitope prediction:
Algorithms can predict likely binding epitopes based on protein structure and antibody sequences
Helps guide experimental design for epitope mapping
Can predict cross-reactivity with similar proteins
Developability prediction:
Application-specific performance prediction:
Models can predict which ATIC antibodies will perform best in specific applications (WB, IHC, IF)
Reduces time spent on empirical testing of multiple antibodies
Future developments:
ATIC antibodies serve as critical tools in cancer metabolism research:
Metabolic pathway analysis:
Therapeutic target validation:
Resistance mechanism investigation:
Changes in ATIC expression may contribute to therapy resistance
Antibodies allow monitoring of expression changes during treatment
Co-expression studies with other metabolic enzymes reveal compensatory pathways
Antibody-drug conjugates (ADCs):
Diagnostic applications:
Combination therapy development:
ATIC inhibition may sensitize cancers to other therapies
Antibodies help assess synergistic effects on metabolic pathways
Expression analysis guides rational combination strategies
Advanced structural techniques provide unprecedented insights into antibody-antigen interactions:
Single-particle analysis techniques:
Mass photometry and charge-detection mass spectrometry enable measurement of full IgG binding to target proteins
These techniques reveal that antibodies often bind at stoichiometries lower than expected from symmetry
Understanding actual binding stoichiometries is critical for interpreting biological effects
Cryo-electron microscopy (cryo-EM):
Provides high-resolution structures of antibody-antigen complexes
Reveals conformational epitopes not identifiable through sequence analysis
Helps understand steric constraints in complex formation
Can visualize structural changes induced by antibody binding
Hydrogen-deuterium exchange mass spectrometry (HDX-MS):
Maps regions of ATIC that become protected upon antibody binding
Identifies conformational changes in regions distant from binding site
Provides dynamic information not available from static structures
Surface plasmon resonance (SPR) and bio-layer interferometry (BLI):
Measures binding kinetics and affinity between ATIC and antibodies
Reveals how subtle changes in antibody sequence affect binding properties
Enables comparison of different antibodies targeting the same epitope
X-ray crystallography:
Provides atomic-level details of antibody-ATIC complexes
Identifies specific amino acid contacts at the binding interface
Guides rational antibody engineering efforts
Molecular dynamics simulations: