ATIC Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Overview of ATIC and Its Biological Role

ATIC (5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inosine monophosphate cyclohydrolase) is a bifunctional enzyme critical to the de novo purine biosynthesis pathway. It catalyzes the final two steps of purine synthesis: the formylation of AICAR (5-aminoimidazole-4-carboxamide ribonucleotide) and the cyclization of FAICAR (5-formamidoimidazole-4-carboxamide ribonucleotide) to inosine monophosphate (IMP). Dysregulation of ATIC is implicated in metabolic disorders and cancer due to its role in nucleotide metabolism .

Diagnostic and Basic Research Uses

  • Cancer Research: ATIC overexpression is observed in malignancies with dysregulated nucleotide metabolism. The antibody enables detection of ATIC in tumor tissues, aiding studies on metabolic reprogramming in cancers like leukemia and pancreatic carcinoma .

  • Developmental Biology: Used to study ATIC expression patterns in model organisms (e.g., Xenopus, Drosophila), providing insights into purine synthesis during embryogenesis .

  • Protein Interaction Studies: Facilitates mapping ATIC’s role in enzyme complexes within the purine biosynthesis pathway .

Technical Validation

  • Specificity: No cross-reactivity with unrelated proteins, confirmed via knockout cell lines .

  • Sensitivity: Detects ATIC at concentrations as low as 0.1 µg/mL in Western blot assays .

  • Reproducibility: Validated across multiple laboratories using standardized protocols, ensuring consistent performance in inter-study comparisons .

Clinical and Therapeutic Implications

While no ATIC-targeted therapies are currently FDA-approved, research highlights its potential as a biomarker:

  • Metabolic Disorders: ATIC inhibition is explored for gout and hyperuricemia treatment, leveraging its role in uric acid production .

  • Cancer Therapeutics: Preclinical studies investigate ATIC inhibitors (e.g., L-Alanosine) to disrupt nucleotide synthesis in rapidly dividing cancer cells .

Product Specs

Form
Purified mouse monoclonal antibody in 0.1 M Tris-Glycine buffer (pH 7.4), containing 150 mM NaCl, 0.2% sodium azide, and 50% glycerol.
Lead Time
Orders typically ship within 1-3 business days of receipt. Delivery times may vary depending on shipping method and destination. Please contact your local distributor for precise delivery estimates.
Synonyms
PURH, AICAR, AICARFT, IMPCHASE

Q&A

What is the optimal method for producing ATIC monoclonal antibodies?

The most effective approach involves using hybridoma technology, where mice are immunized with ATIC protein or specific epitopes, followed by fusion of B lymphocytes with myeloma cells. Based on comparative studies of monoclonal antibody production, mouse ascites typically yields significantly higher concentrations than hybridoma cell culture supernatants (3×10^-5 mol/L from ascites compared to 3.75×10^-6 mol/L from supernatants) .

The recommended production workflow includes:

  • Immunization of Balb/C mice with purified ATIC or synthetic ATIC peptides

  • Isolation of spleen lymphocytes and fusion with myeloma cells

  • Selection of hybridomas secreting ATIC-specific antibodies via ELISA

  • Expansion of selected hybridoma clones

  • Intraperitoneal injection of 1×10^7 hybridoma cells for ascites production

  • Harvesting and purification of antibodies using protein A/G affinity chromatography

This approach yields antibodies of IgG class, with heavy chains of approximately 55 kDa and light chains of 25 kDa as confirmed by SDS-PAGE analysis .

How can I validate the specificity of my ATIC monoclonal antibody?

Comprehensive validation requires multiple complementary approaches:

Validation MethodPurposeControls Required
Western blottingConfirms size-specific recognitionATIC-expressing and knockout cells
Immunoprecipitation-MSVerifies target identityIgG isotype control
ImmunohistochemistryConfirms expression patternKnown ATIC-positive tissues
ELISA binding studiesQuantifies affinityBlocking peptides
Cross-reactivity testingAssesses specificityRelated protein family members

For optimal specificity validation, perform Western blot analysis on cellular extracts from tissues/cells known to express varying levels of ATIC. The antibody should recognize a single band at approximately 64 kDa (ATIC's molecular weight). Always include blocking experiments with the immunizing peptide to confirm specificity, which should abolish signal if the antibody is truly specific .

What are the critical factors affecting ATIC antibody performance in different experimental systems?

Several factors can significantly impact experimental outcomes:

  • Epitope accessibility: ATIC's bifunctional nature means certain epitopes may be obscured in different conformational states or when involved in protein-protein interactions.

  • Sample preparation: For Western blotting, use RIPA buffer with protease inhibitors; for immunohistochemistry, optimize antigen retrieval methods (citrate buffer pH 6.0 often works well for ATIC detection).

  • Antibody concentration: Titration experiments should be performed for each application, with typical starting dilutions of 1:500-1:2000 for Western blots and 1:50-1:200 for IHC.

  • Detection systems: Amplification methods such as tyramide signal amplification can enhance sensitivity for detecting low ATIC expression levels.

  • Buffer composition: Presence of competing proteins, detergents, or salt concentrations can all affect binding kinetics and specificity.

Functional validation using ATIC enzymatic assays provides an additional verification method for antibody specificity, as demonstrated in similar validation approaches for other enzymatic targets .

How can I optimize ATIC monoclonal antibodies for studying protein-protein interactions?

When investigating ATIC's interactions with other proteins, consider these methodological approaches:

  • Epitope selection: Choose antibodies targeting regions away from known protein interaction domains to minimize interference with complex formation.

  • Co-immunoprecipitation optimization:

    • Use gentle lysis conditions (avoid strong detergents)

    • Cross-link protein complexes with DSP or formaldehyde before lysis

    • Include appropriate protease and phosphatase inhibitors

    • Consider native elution conditions to preserve complex integrity

  • Proximity ligation assays (PLA):

    • Combine ATIC antibody with antibodies against suspected interacting partners

    • Optimize antibody concentrations to minimize background

    • Include appropriate controls (single antibody, non-interacting protein pairs)

  • FRET/BRET approaches:

    • Use site-specific conjugation methods for fluorophore attachment

    • Target the Fc region for conjugation to preserve antigen binding

    • Validate that conjugation doesn't alter binding properties

Research in monoclonal antibody applications demonstrates that selecting the appropriate buffer conditions dramatically affects the preservation of protein-protein interactions during immunoprecipitation experiments .

What approaches should I use when facing contradictory results from different ATIC monoclonal antibodies?

When different antibodies yield conflicting results, implement this systematic troubleshooting approach:

  • Compare antibody characteristics:

    • Epitope location (N-terminal AICAR transformylase domain vs. C-terminal IMP cyclohydrolase domain)

    • Clonality and host species

    • Validation methods used by manufacturers

  • Perform side-by-side validation:

    • Western blot with positive and negative controls

    • Immunoprecipitation followed by mass spectrometry

    • Functional validation through enzyme activity assays

  • Consider biological variables:

    • Post-translational modifications affecting epitope accessibility

    • Alternative ATIC splice variants

    • Species-specific sequence variations

  • Technical considerations:

    • Sample preparation differences

    • Detection system sensitivities

    • Lot-to-lot antibody variability

  • Independent verification:

    • Use orthogonal methods (PCR, mass spectrometry)

    • CRISPR knockout validation

    • RNA interference experiments

This approach aligns with best practices documented in comprehensive monoclonal antibody validation studies, which emphasize the importance of multiple validation methods .

How can I design experiments to study ATIC localization changes under different cellular conditions?

For tracking ATIC localization dynamics:

  • Subcellular fractionation followed by Western blotting:

    • Use validated protocols for isolating cytoplasmic, nuclear, mitochondrial, and membrane fractions

    • Include compartment-specific markers (GAPDH for cytoplasm, Lamin B1 for nucleus)

    • Quantify relative ATIC distribution across fractions

  • Live-cell imaging approaches:

    • Develop cell lines expressing ATIC-fluorescent protein fusions

    • Validate that fusion proteins maintain normal localization and function

    • Use appropriate microscopy techniques (confocal, TIRF) based on expected localization patterns

  • Immunofluorescence optimization:

    • Compare fixation methods (paraformaldehyde vs. methanol)

    • Test permeabilization conditions (Triton X-100, saponin, digitonin)

    • Use z-stack imaging to capture complete distribution patterns

  • Quantitative analysis:

    • Employ digital image analysis software for quantifying colocalization

    • Use Pearson's or Mander's coefficients for colocalization metrics

    • Analyze sufficient cell numbers (typically >30 cells per condition)

  • Stimulus-response experiments:

    • Design time-course studies after cellular stimulation

    • Include appropriate controls (vehicle, inactive analogues)

    • Consider selective inhibitor treatments to confirm pathway specificity

Research on monoclonal antibody applications in cellular imaging demonstrates that optimization of fixation and permeabilization protocols is critical for preserving authentic protein localization patterns .

What are the best approaches for conjugating ATIC monoclonal antibodies to fluorophores or enzymes?

Strategic conjugation requires careful consideration of chemistry and target sites:

Conjugation MethodTarget SiteImpact on FunctionBest Applications
NHS ester chemistryLysine residuesMay affect binding if in CDRFluorophore attachment
Maleimide chemistryReduced cysteinesMinimal if targeting hinge regionSite-specific labeling
Enzymatic methodsQ295 in Fc regionNo impact on bindingReporter enzyme conjugation
Copper-free click chemistryModified glycansMinimal impactComplex conjugation
Photochemical crosslinkingSpatially controlledProximity-dependentInteraction studies

For optimal ATIC antibody conjugation:

  • Perform buffer exchange into non-amine containing buffer (PBS) at pH 8.3

  • Determine optimal dye-to-antibody ratio through titration (typically 2-4 molecules per antibody)

  • React at room temperature in the dark (2-4 hours)

  • Purify conjugated antibody using size exclusion chromatography

  • Validate conjugate functionality through binding assays

  • Determine degree of labeling spectrophotometrically

This approach aligns with established protocols for producing functional antibody conjugates with minimal impact on antigen recognition, similar to those used in the development of antibody-drug conjugates in clinical research .

How can I enhance detection sensitivity for ATIC in tissues with low expression levels?

For detecting low-abundance ATIC protein:

  • Signal amplification methods:

    • Tyramide signal amplification (TSA) - provides 10-100× signal enhancement

    • Polymer-based detection systems - offer 5-10× improved sensitivity

    • Quantum dot conjugates - higher quantum yield than conventional fluorophores

  • Sample preparation optimization:

    • Evaluate different antigen retrieval methods (heat-induced vs. enzymatic)

    • Test multiple buffer systems (citrate, EDTA, Tris) at varying pH values

    • Optimize retrieval times (15-40 minutes)

  • Background reduction strategies:

    • Block with appropriate proteins (5% BSA, normal serum)

    • Include detergents (0.1-0.3% Triton X-100) to reduce non-specific binding

    • Use monovalent Fab fragments to block endogenous Fc receptors

    • Apply Sudan Black B to reduce autofluorescence in tissues

  • Antibody incubation parameters:

    • Extend primary antibody incubation time (overnight at 4°C)

    • Optimize antibody concentration through titration experiments

    • Consider using antibody cocktails targeting different ATIC epitopes

Research on monoclonal antibody applications in challenging samples demonstrates that combining signal amplification with optimized sample preparation can achieve up to 50-fold improvement in detection sensitivity .

What are the best practices for quantifying ATIC protein levels in research samples?

Accurate quantification requires standardized approaches:

Quantification MethodPrincipleAdvantagesLimitations
Quantitative Western blotComparison to standard curveDirect visualizationLimited dynamic range
ELISASandwich or competitive bindingHigh-throughput capabilityRequires two non-competing antibodies
Flow cytometryPer-cell fluorescence intensitySingle-cell resolutionLimited to cells in suspension
IHC with digital analysisPixel intensity quantificationSpatial information preservedSubject to staining variability
Mass spectrometry with immunocapturePeptide quantificationAbsolute quantificationTechnically demanding

Implementation recommendations:

  • Include recombinant ATIC protein standards at known concentrations

  • Perform spike-recovery experiments to assess matrix effects

  • Use internal loading controls (housekeeping proteins, total protein stains)

  • Validate linear range of detection for each assay system

  • Include biological reference samples across experiments for normalization

For ATIC specifically, consider complementing antibody-based detection with enzymatic activity assays to correlate protein levels with functional activity, as demonstrated in studies with other enzyme-targeting antibodies .

How do post-translational modifications of ATIC affect antibody recognition and experimental outcomes?

ATIC undergoes several post-translational modifications that can impact antibody binding:

  • Phosphorylation:

    • ATIC can be phosphorylated at multiple serine and threonine residues

    • Phosphorylation may alter epitope accessibility or recognition

    • Phospho-specific antibodies might be required to study specific modification states

    • Use of phosphatase inhibitors during sample preparation is critical

  • SUMOylation:

    • ATIC can be SUMO-modified under certain cellular conditions

    • This large modification can mask epitopes

    • Denaturing conditions in Western blots may reveal epitopes hidden in native conformations

  • Experimental considerations:

    • Include phosphatase inhibitors (sodium orthovanadate, sodium fluoride) in lysis buffers

    • Consider parallel analysis with phospho-specific and total ATIC antibodies

    • Compare results under different denaturing conditions

    • Use recombinant ATIC with defined modification states as controls

  • Validation approaches:

    • Treat samples with phosphatases to confirm phosphorylation-dependent recognition

    • Use mass spectrometry to map modification sites

    • Compare antibody recognition before and after induction of specific modifications

This methodological approach follows established protocols for studying post-translationally modified proteins as documented in monoclonal antibody research applications .

What considerations are important when using ATIC monoclonal antibodies for therapeutic target validation?

When establishing ATIC as a therapeutic target:

  • Functional validation requirements:

    • Confirm antibody specificity through knockout/knockdown controls

    • Determine whether the antibody is inhibitory, stimulatory, or neutral to ATIC function

    • Assess the relationship between epitope binding and functional effects

  • Target biology considerations:

    • Evaluate ATIC expression levels across relevant tissues and disease states

    • Determine subcellular localization accessibility for therapeutic antibodies

    • Assess potential for on-target, off-tissue effects

  • Experimental design for validation:

    • Include isotype-matched control antibodies in all experiments

    • Design dose-response studies to establish EC50/IC50 values

    • Evaluate potential compensatory mechanisms after ATIC inhibition

    • Conduct time-course studies to determine onset and duration of effects

  • Translation to therapeutic development:

    • Consider antibody format requirements (full IgG vs. fragments)

    • Evaluate species cross-reactivity for preclinical model translation

    • Assess internalization potential for antibody-drug conjugate approaches

The methodological considerations align with current monoclonal antibody therapeutic development practices, where detailed target validation is critical before clinical advancement .

How can advanced antibody engineering techniques be applied to develop more specific ATIC monoclonal antibodies?

Cutting-edge approaches for next-generation ATIC antibodies:

  • Phage display technology:

    • Allows screening of billions of antibody variants

    • Can select for specific binding characteristics (affinity, specificity, pH-dependent binding)

    • Enables affinity maturation through directed evolution

  • Bispecific antibody engineering:

    • Targeting ATIC plus a second protein of interest

    • Useful for studying protein complexes or pathway intersections

    • Can enhance specificity through dual epitope recognition

  • Single-domain antibodies (nanobodies):

    • Smaller size allows access to epitopes unreachable by conventional antibodies

    • Better penetration into tissues and subcellular compartments

    • Easier genetic manipulation for fusion proteins

  • Rational design approaches:

    • Computational modeling of antibody-antigen interfaces

    • Structure-guided optimization of binding interface

    • In silico screening before experimental validation

  • Recombinant antibody production:

    • Allows precise control over antibody characteristics

    • Facilitates site-specific modifications

    • Reduces batch-to-batch variability seen with hybridoma-derived antibodies

These advanced technologies reflect the evolution of monoclonal antibody development described in recent WHO reports on the clinical research landscape of monoclonal antibodies .

What are the most promising research applications for ATIC monoclonal antibodies in cancer and inflammatory disease studies?

ATIC antibodies offer potential in several cutting-edge research areas:

  • Cancer metabolism studies:

    • Investigating ATIC's role in de novo purine synthesis in cancer cells

    • Studying metabolic adaptations in treatment-resistant tumors

    • Correlating ATIC expression with cancer aggressiveness and outcomes

  • AMPK pathway interaction analysis:

    • Exploring ATIC's relationship with AMPK signaling

    • Investigating how ATIC inhibition affects cellular energy homeostasis

    • Studying potential for metabolic synthetic lethality approaches

  • Inflammatory disease mechanisms:

    • Investigating ATIC's role in immune cell activation and proliferation

    • Studying purine metabolism in autoimmune disorders

    • Exploring connections between ATIC and inflammatory cytokine production

  • Methodology approaches:

    • Proximity labeling with ATIC antibodies to identify interaction partners

    • ChIP-seq applications to study potential transcriptional roles

    • Spatial transcriptomics combined with ATIC protein localization

  • Therapeutic target validation:

    • Using blocking antibodies to assess ATIC's potential as a drug target

    • Developing antibody-drug conjugates targeting ATIC-overexpressing cells

    • Creating conditionally active antibodies responsive to the tumor microenvironment

These applications build upon the growing trend of using monoclonal antibodies as both research tools and therapeutic agents, as documented in recent clinical research landscapes .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.