The ATM monoclonal antibody refers to laboratory-generated antibodies that specifically target the Ataxia Telangiectasia Mutated (ATM) protein, a serine/threonine kinase critical for DNA damage response. These antibodies are primarily used in research and diagnostics to study ATM’s role in DNA repair, apoptosis, and cancer biology. While most sources focus on antibodies detecting ATM expression or activity, one therapeutic monoclonal antibody (ATM-027) targeting T cells has been explored in clinical trials for multiple sclerosis .
ATM is activated in response to DNA double-strand breaks (DSBs), phosphorylating downstream targets (e.g., Chk2, p53) to induce cell cycle arrest or apoptosis . Monoclonal antibodies against ATM are designed to:
Detect protein expression: Used in Western blot (WB), immunohistochemistry (IHC), and immunofluorescence (IF) to localize ATM in nuclear compartments .
Monitor phosphorylation status: Antibodies like 200-301-400 detect ATM’s autophosphorylated S1981 site, a marker of activation .
Study functional interactions: For example, 5C2 (mouse monoclonal) binds to ATM’s catalytic domain and is used to assess DNA damage responses in HeLa cells .
Cancer biomarker: Loss of ATM expression correlates with aggressive progression in breast, lung, and gastric cancers .
Therapeutic stratification: ATM-deficient tumors (e.g., chronic lymphocytic leukemia, mantle cell lymphoma) show sensitivity to PARP inhibitors like olaparib .
While most ATM antibodies are research tools, one therapeutic antibody—ATM-027—targeted Vβ5.2/5.3+ T cells in multiple sclerosis. Key findings:
Pharmacokinetics: Followed a two-compartment model with a terminal half-life of ~22 days .
Efficacy: Reduced target T cells to 25–28% of baseline levels in phase I/II trials .
Limitations: Development halted due to lack of clinical benefit .
In contrast, non-antibody therapies for ATM-deficient cancers include:
ATM protein demonstrates predominant cytoplasmic localization in specific neuronal cells. Studies using anti-ATM monoclonal antibodies show that in mouse cerebellum, ATM is exclusively expressed in the cytoplasm of Purkinje cells (PCs) in the cerebellar cortex. Similar cytoplasmic ATM immunoreactivity has been observed in a subset of neurons in dorsal root ganglia. This localization pattern in mouse cerebellum resembles that reported in human adult cerebellum . When conducting immunohistochemical studies, it is essential to include appropriate negative controls, such as ATM-knockout tissues (Atm−/−), to validate antibody specificity.
ATM expression has been documented across various tissues through antibody-based detection methods. According to validation studies, ATM is expressed in:
When designing experiments to detect ATM in these tissues, researchers should consider tissue-specific optimization of antibody concentrations and incubation conditions.
For long-term storage, ATM monoclonal antibodies should be maintained at -20°C for up to one year. For short-term storage and frequent use, store at 4°C for up to one month. It is crucial to avoid repeated freeze-thaw cycles as these can significantly degrade antibody performance . When working with these antibodies, proper handling includes maintaining a cold chain during experiments and avoiding contamination.
Validating antibody specificity requires multiple complementary approaches:
Western blotting validation: Load protein extracts from both wild-type and ATM-knockout (Atm−/−) tissues on 7.5% low crosslinking acrylamide gels (121:1 acrylamide:bisacrylamide). The absence of signal in knockout tissues confirms specificity .
Immunoprecipitation assays: Perform pull-down experiments followed by mass spectrometry to confirm target enrichment.
Multi-application testing: Validate across different applications (WB, IHC, ICC, IF, Flow Cytometry) to ensure consistent specificity .
Cross-reactivity testing: If working with non-human samples, test the antibody against multiple species to determine cross-reactivity boundaries.
For therapeutic applications of ATM monoclonal antibodies (such as ATM-027 in multiple sclerosis studies), a two-compartment pharmacokinetic model is most appropriate. Key parameters to consider include:
Total volume of distribution: approximately 5.9 liters
Terminal half-life: approximately 22.3 days in typical patients
EC50 for receptor expression: 138-148 μg/L using inhibitory sigmoidal Emax-models
Target cell reduction: treatment with ATM-027 decreases target T cell numbers to approximately 25.7-28.9% of baseline values
When designing dosing regimens, these parameters enable prediction of antibody concentrations and biological effects over time.
Performance varies significantly across experimental platforms:
Application | Optimal Dilution | Incubation Time | Expected Results |
---|---|---|---|
Western Blot | 1:500-1:1000 | Overnight at 4°C | Single band at ~370 kDa |
IHC | 1:100-1:200 | 1-2 hours at RT | Cytoplasmic staining in neurons |
ICC/IF | 1:100-1:200 | 1-2 hours at RT | Primarily cytoplasmic pattern |
Flow Cytometry | 1:50-1:100 | 30-45 minutes on ice | Cell population-specific signal |
These parameters should be optimized for each specific experimental setup and cell/tissue type .
For detecting ATM in fibroblast samples, the following optimized protocol is recommended:
For Western Blotting:
Lyse fibroblasts in RIPA buffer supplemented with protease inhibitors
Load 30-50 μg protein per lane on 7.5% low crosslinking acrylamide gels (121:1 acrylamide:bisacrylamide)
Transfer to PVDF membrane at 100V for 2 hours
Block with 5% non-fat milk for 1 hour at room temperature
Incubate with anti-ATM antibody (1:500 dilution) overnight at 4°C
Wash and incubate with HRP-conjugated secondary antibody
For Immunocytochemistry:
Fix fibroblasts with 4% paraformaldehyde for 15 minutes
Permeabilize with 0.1% Triton X-100 for 10 minutes
Block with 1% BSA in PBS for 30 minutes
Incubate with anti-ATM antibody (1:100 dilution) for 2 hours at room temperature
Follow with fluorophore-conjugated secondary antibody incubation
For neuronal tissue analysis:
Tissue preparation:
Fix tissues in 4% paraformaldehyde
Create paraffin-embedded or frozen sections (10-12 μm thickness)
For paraffin sections, perform antigen retrieval using citrate buffer (pH 6.0)
Staining procedure:
Block endogenous peroxidase with 0.3% H₂O₂
Apply protein block (5% normal serum)
Incubate with anti-ATM antibody overnight at 4°C
Use appropriate detection system (e.g., avidin-biotin complex)
Develop with DAB and counterstain with hematoxylin
Analysis focus:
For quantitative analysis of cell surface ATM:
Flow cytometry approach:
Prepare single-cell suspensions
Block Fc receptors with appropriate blocking solution
Stain with anti-ATM antibody (1:50-1:100 dilution)
Apply fluorophore-conjugated secondary antibody
Analyze using standard flow cytometry protocols
Quantify using mean fluorescence intensity (MFI)
Modeling receptor expression:
Inconsistent staining may result from several factors:
Antibody specificity issues:
Validate antibody using ATM-knockout tissues or cells
Confirm expression patterns align with known tissue distribution
Test multiple antibody lots for consistency
Technical considerations:
Optimize antigen retrieval methods for fixed tissues
Adjust antibody concentration and incubation times
Ensure proper blocking to reduce non-specific binding
Control temperature during all incubation steps
Sample preparation factors:
A comprehensive control strategy includes:
Genetic controls:
ATM-knockout (Atm−/−) tissues/cells as negative controls
Tissues with known high expression (e.g., cerebellum) as positive controls
Antibody controls:
Isotype control antibodies to assess non-specific binding
Secondary antibody-only controls to evaluate background
Peptide competition assays to confirm binding specificity
Disease-specific controls:
When facing contradictory results between methods:
Methodological considerations:
Western blotting detects denatured proteins; conformation-specific antibodies may not work
IHC/ICC preserves spatial information but may have accessibility limitations
Flow cytometry only detects surface or permeabilized cellular epitopes
Resolution approaches:
Use multiple antibodies targeting different epitopes of ATM
Employ complementary techniques (e.g., mRNA analysis, reporter assays)
Consider post-translational modifications that might affect epitope recognition
Evaluate subcellular fractionation to resolve localization discrepancies
Data integration:
ATM's unique cytoplasmic localization in neurons makes it valuable for neurodegenerative research:
Cerebellar ataxia studies:
Monitor Purkinje cell ATM expression and localization changes
Correlate ATM levels with disease progression
Investigate ATM's role in neuronal survival pathways
DNA damage response in neurons:
Evaluate nuclear versus cytoplasmic ATM distribution in response to oxidative stress
Assess post-translational modifications of ATM in neurodegenerative conditions
Monitor ATM activation status using phospho-specific antibodies
Therapeutic targeting:
When developing ATM-based biomarkers:
Assay development:
Select antibodies with high specificity and sensitivity
Establish standardized protocols with defined cut-off values
Validate across multiple sample types and disease states
Clinical correlation:
Correlate ATM expression/activity with clinical outcomes
Determine temporal relationships between ATM changes and disease progression
Establish minimally important differences for intervention studies
Monitoring parameters:
The discovery of ATM's cytoplasmic localization has important implications:
Epitope selection:
Choose antibodies recognizing epitopes accessible in both nuclear and cytoplasmic environments
Consider antibodies specific to different ATM conformational states
Select clones validated for the appropriate subcellular compartment
Functional studies:
Design experiments that differentiate between ATM's DNA damage response and cytoplasmic signaling roles
Include cytoplasmic signaling partners in interaction studies
Develop assays specific to cytoplasmic ATM activation states
Disease relevance: