The antibody is a recombinant monoclonal rabbit IgG purified via affinity chromatography. It is generated using synthetic phosphopeptides corresponding to the phosphorylated T71 residue of human ATF2 (P15336) . Key structural features include:
Immunogen: Synthetic phosphorylated peptide around T71 (sequence: TPTP T) .
Clonality: Monoclonal, ensuring high specificity for phosphorylated ATF2 .
Target Specificity: Binds exclusively to ATF2 phosphorylated at T71, distinguishing it from non-phosphorylated forms .
Phosphorylation at T71 activates ATF2’s transcriptional functions, enabling interactions with AP-1 proteins (e.g., c-Jun) and nuclear translocation to regulate genes involved in apoptosis, proliferation, and DNA repair .
This antibody is validated for diverse experimental techniques:
Phosphorylation at T71 is induced by stress-activated kinases (e.g., p38 MAPK, JNK) in response to UV radiation, cytokines, or chemotoxic agents like anisomycin . This modification enables ATF2 to:
Regulate Apoptosis: Promote cell death by disrupting mitochondrial complexes (e.g., HK1-VDAC1) .
DNA Damage Response: Mediate S-phase checkpoint control and recruit repair proteins (e.g., MRN complex) .
ATF2 exhibits dual roles depending on cellular context:
Activity | Mechanism | Implications |
---|---|---|
Oncogenic | Hyperactivation in cancer cells, promoting proliferation . | |
Tumor-Suppressive | Induction of apoptosis in stressed cells; degradation of acetyltransferase KAT5 . |
Anisomycin Treatment: Western blots show robust T71 phosphorylation in NIH/3T3 and HeLa cells post-treatment .
Dot Blot Specificity: No cross-reactivity with non-phosphorylated ATF2 peptides .
This phospho-ATF2 (T71) recombinant monoclonal antibody was generated by cloning the coding sequence of a phospho-ERN1 (S724) monoclonal antibody (produced via immunization with a synthetic phosphopeptide of ERN1) into expression plasmids and transfecting these into suitable cell lines. The resulting antibody, a rabbit IgG, was purified using affinity chromatography. This antibody specifically detects endogenous human ATF2 only when phosphorylated at threonine 71 (T71). It is suitable for ELISA, Western blotting (WB), and immunofluorescence (IF) applications.
ATF2 is ubiquitously expressed and regulates the transcription of genes involved in diverse biological processes, including cell growth, development, and stress response. It functions as a key mediator of extracellular signaling pathways, transducing signals to the nucleus and initiating appropriate transcriptional responses. A variety of stimuli, such as growth factors, UV radiation, and cytokines, activate ATF2. Specifically, stress-activated protein kinases (SAPKs), such as p38, phosphorylate ATF2 at threonine 69 (T69) and threonine 71 (T71), leading to its activation and enhanced transcriptional activity. Following phosphorylation at T69/T71, ATF2 interacts with other activator protein 1 (AP-1) family members and translocates to the nucleus to modulate gene expression.
Activating transcription factor 2 (ATF2) is a transcriptional regulator controlling the expression of numerous genes, including those implicated in anti-apoptosis, cell growth, and DNA damage repair. Its binding specificity varies depending on its interacting partner; it can bind to cAMP response element (CRE) consensus sequences (5'-TGACGTCA-3') or AP-1 consensus sequences (5'-TGACTCA-3'). Within the nucleus, ATF2 contributes to global transcription and DNA damage response, in addition to specific transcriptional regulation of genes involved in cell development, proliferation, and apoptosis. In the cytoplasm, ATF2 interacts with and disrupts hexokinase 1 (HK1) and voltage-dependent anion channel 1 (VDAC1) complexes at the mitochondrial outer membrane, consequently impairing mitochondrial membrane potential, causing mitochondrial dysfunction, and promoting apoptosis. ATM-mediated phosphorylation of ATF2 plays a critical role in DNA damage response, contributing to the ionizing radiation (IR)-induced S phase checkpoint and recruitment of the MRN complex to IR-induced foci (IRIF). ATF2 possesses histone acetyltransferase (HAT) activity, specifically acetylating histones H2B and H4 in vitro. Furthermore, in conjunction with Cullin 3 (CUL3) and RING-box protein 1 (RBX1), ATF2 promotes the degradation of histone acetyltransferase KAT5, thereby attenuating its activation of ataxia-telangiectasia mutated (ATM). The ultimate biological outcome of ATF2 activity (oncogenic or tumor suppressive) is context-dependent, varying with tissue and cell type.
Phospho-ATF2 (T71) refers to the Activating Transcription Factor 2 (ATF2) protein that has been phosphorylated at threonine 71. ATF2 functions as a critical transcriptional activator that regulates the expression of various genes involved in anti-apoptosis, cell growth, and DNA damage response pathways . The phosphorylation at threonine 71 is a key post-translational modification that activates ATF2, enabling it to perform its diverse cellular functions. This phosphorylation is mediated by several kinases, including stress-activated protein kinases (SAPKs), p38, and JNK . The importance of this phosphorylation lies in its role as a molecular switch that alters ATF2's activity, subcellular localization, and interaction partners. In particular, the phosphorylated form (mediated by ATM) plays a crucial role in the DNA damage response and is involved in ionizing radiation-induced S phase checkpoint control . Understanding ATF2 phosphorylation status provides insights into cellular responses to stress and various signaling pathways that regulate cell survival, proliferation, and death.
Phosphorylation of ATF2 at threonine 71 (and often in conjunction with threonine 69) creates significant functional differences compared to its non-phosphorylated form. When phosphorylated, ATF2 exhibits enhanced DNA binding affinity to specific consensus sequences, including CRE (cAMP response element) sequences (5'-TGACGTCA-3') and AP-1 (activator protein 1) sequences (5'-TGACTCA-3') . This phosphorylation-dependent binding allows for selective gene transcription regulation.
The functional differences include:
Parameter | Non-phosphorylated ATF2 | Phosphorylated ATF2 (T71) |
---|---|---|
Transcriptional activity | Limited | Enhanced activation of target genes |
DNA damage response | Minimal involvement | Active participation in S-phase checkpoint control |
Protein interactions | Restricted partner binding | Expanded interaction with MRN complex and other DNA repair factors |
Subcellular localization | May be sequestered in cytoplasm | Nuclear translocation enhanced |
Role in viral replication | Limited effect | Promotes viral replication (e.g., PRV) |
Additionally, phosphorylated ATF2 has been shown to facilitate the recruitment of the MRN complex into ionizing radiation-induced foci (IRIF), a critical step in DNA damage repair processes . Research has also demonstrated that the phosphorylation of ATF2 can promote viral replication, as evidenced in studies with pseudorabies virus (PRV), where increased phosphorylation levels of ATF2 enhanced viral replication efficiency .
Recommended Western Blot protocol elements:
Sample preparation:
Extract proteins under phosphatase inhibitor protection
Use fresh samples when possible to minimize phospho-epitope degradation
Include positive controls (cells treated with stress inducers like UV or anisomycin)
Gel electrophoresis:
Use 10-12% SDS-PAGE gels for optimal resolution
Load 20-40 μg of total protein per lane
Include molecular weight markers (ATF2 appears at approximately 70 kDa)
Transfer and blocking:
Transfer to PVDF membrane (preferred over nitrocellulose for phospho-epitopes)
Block with 5% BSA in TBST (not milk, which contains phospho-proteins)
Incubate with primary antibody overnight at 4°C
Detection:
Use HRP-conjugated secondary antibody (typically anti-rabbit IgG)
Consider enhanced chemiluminescence detection systems
Validate specificity using phosphatase-treated control samples
The antibody is most effective when samples are freshly prepared and stored appropriately, as repeated freeze-thaw cycles should be avoided to maintain phospho-epitope integrity .
For immunofluorescence (IF) applications, optimization of several parameters is crucial to achieve specific and sensitive detection of Phospho-ATF2 (T71). The recommended dilution range for IF applications is 1:20-1:200 , though this should be empirically determined for each experimental system.
Detailed immunofluorescence optimization strategy:
Fixation and permeabilization:
Test both paraformaldehyde (4%) and methanol fixation methods
For paraformaldehyde fixation, permeabilize with 0.1-0.5% Triton X-100
Methanol fixation often preserves phospho-epitopes better but test both
Blocking conditions:
Use 5-10% normal serum (different from antibody host species) with 1% BSA
Include 0.1% Triton X-100 in blocking buffer for nuclear antigens
Block for minimum 1 hour at room temperature
Antibody incubation:
Start with 1:50 dilution and optimize
Incubate primary antibody overnight at 4°C
Use gentle rocking/rotation during incubation
Signal enhancement strategies:
Consider tyramide signal amplification for low abundance targets
Use high-sensitivity fluorophore-conjugated secondary antibodies
Include DAPI counterstain for nuclear visualization (ATF2 has nuclear localization)
Controls:
Phosphatase-treated negative control
Stimulated cell positive control (e.g., UV radiation, JNK activators)
Secondary antibody-only control
This antibody is compatible with fluorescent detection systems and can be used to visualize the subcellular localization of phosphorylated ATF2, which is predominantly nuclear when activated .
Rigorous validation of antibody specificity is essential for reliable research results, particularly for phospho-specific antibodies. For Phospho-ATF2 (T71) antibody, several controls should be implemented:
Control Type | Implementation Method | Purpose |
---|---|---|
Positive control | Cells treated with known ATF2-T71 phosphorylation inducers (UV radiation, anisomycin, JNK activators) | Confirms antibody can detect increased phosphorylation |
Negative control | Samples treated with lambda phosphatase | Verifies signal dependence on phosphorylation status |
Knockdown/knockout control | siRNA against ATF2 or CRISPR/Cas9 ATF2 knockout cells | Confirms antibody specificity for ATF2 protein |
Peptide competition | Pre-incubation with phospho-T71 peptide vs. non-phospho peptide | Demonstrates phospho-epitope specificity |
Cross-reactivity assessment | Testing against related proteins (e.g., other CREB family members) | Determines potential off-target binding |
Additionally, researchers should consider:
Testing the antibody across multiple techniques (WB, IF, ELISA) to confirm consistent specificity
Using stimulation time-course experiments to correlate signal with known ATF2 phosphorylation kinetics
Comparing results with alternative phospho-ATF2 (T71) antibodies from different suppliers or clones
When possible, implementing mass spectrometry validation of the phosphorylation site in immunoprecipitated samples
For advanced validation, consider using cells expressing ATF2 with T71A mutation (preventing phosphorylation) as a definitive negative control, though this requires special preparation and is not commercially available .
Phospho-ATF2 (T71) antibody serves as a valuable tool for investigating DNA damage response (DDR) pathways, given ATF2's significant role in these processes. The phosphorylated form of ATF2 (mediated by ATM) is specifically involved in the ionizing radiation (IR)-induced S phase checkpoint control and in recruiting the MRN complex to IR-induced foci (IRIF) . Researchers can leverage this antibody to explore multiple aspects of the DDR pathway:
Temporal dynamics of ATF2 activation:
Treat cells with DNA damaging agents (ionizing radiation, genotoxic chemicals)
Harvest at various time points (5 min to 24 hours)
Quantify phospho-ATF2 levels via Western blot or flow cytometry
Correlate with activation of upstream kinases (ATM, JNK, p38)
Spatial organization of DDR components:
Perform co-immunofluorescence with phospho-ATF2 (T71) antibody and other DDR proteins
Visualize recruitment to damage foci using confocal microscopy
Quantify colocalization with MRN complex components (MRE11, RAD50, NBS1)
Analyze nuclear redistribution patterns during DDR activation
Pathway interaction studies:
Combine with inhibitors of ATM, ATR, DNA-PK, JNK, or p38 pathways
Assess changes in ATF2 phosphorylation status
Determine pathway dependencies for different damage types
Analyze feedback mechanisms between ATF2 and other DDR components
Chromatin immunoprecipitation (ChIP) applications:
Use phospho-ATF2 (T71) antibody for ChIP experiments
Map binding sites on damage-responsive genes
Quantify recruitment to specific genomic loci following damage
Compare binding patterns between different damage types
This antibody enables researchers to discern the specific roles of phosphorylated ATF2 versus total ATF2 protein in DDR processes, providing insights into the molecular mechanisms underlying genome stability maintenance .
The relationship between ATF2 phosphorylation and viral replication represents an emerging area of research with significant implications for understanding host-pathogen interactions. Studies have demonstrated that ATF2 phosphorylation plays a critical role in promoting viral replication, particularly for viruses like pseudorabies virus (PRV) . This relationship can be systematically investigated using Phospho-ATF2 (T71) antibody through several experimental approaches:
Viral infection models:
Infect appropriate cell lines with virus of interest
Monitor changes in ATF2 phosphorylation status over the course of infection
Correlate phosphorylation with viral replication markers
Compare responses across different viral strains and multiplicities of infection
Mechanistic investigation:
Signaling pathway analysis:
Use inhibitors of JNK pathway components (demonstrated to mediate ATF2 activation during infection)
Monitor effects on both ATF2 phosphorylation and viral replication
Identify viral proteins that may activate kinases upstream of ATF2
Perform phosphoproteomics to identify additional targets in the pathway
Transcriptional regulation studies:
Use ChIP-seq with phospho-ATF2 (T71) antibody in infected vs. uninfected cells
Identify viral and host genes regulated by phosphorylated ATF2
Analyze promoter regions for ATF2 binding motifs (CRE or AP-1 sites)
Validate findings with reporter assays and site-directed mutagenesis
Research has specifically shown that ATF2-knockdown reduces both PRV titer and viral genome copy number, while overexpression of ATF2 and phosphorylation at positions 69/71 promotes PRV replication . These findings suggest that targeting ATF2 phosphorylation could provide a potential therapeutic strategy for inhibiting viral infections.
ATF2 exhibits distinct functions depending on its subcellular localization. In the nucleus, it contributes to transcriptional regulation and DNA damage response, while in the cytoplasm, it interacts with mitochondrial membrane proteins like HK1 and VDAC1 to influence mitochondrial membrane potential and cell death . Distinguishing between these compartment-specific functions requires sophisticated experimental approaches:
Subcellular fractionation combined with Western blotting:
Perform careful subcellular fractionation to separate nuclear, cytoplasmic, and mitochondrial fractions
Use Phospho-ATF2 (T71) antibody to quantify distribution across compartments
Validate fraction purity with compartment-specific markers (e.g., Lamin B for nucleus, GAPDH for cytoplasm)
Compare phosphorylation status across compartments
High-resolution microscopy approaches:
Experimental manipulation of localization:
Generate ATF2 constructs with mutated nuclear localization signals or nuclear export signals
Create fusion proteins with compartment-targeting sequences
Compare phosphorylation-dependent activities in different compartments
Implement inducible translocation systems for temporal control
Proximity-based labeling methods:
Implement BioID or APEX2 proximity labeling fused to ATF2
Identify compartment-specific interaction partners of phosphorylated ATF2
Compare interactomes between wild-type and phospho-mutant versions
Validate key interactions with co-immunoprecipitation using Phospho-ATF2 (T71) antibody
Compartment-specific functional assays:
For nuclear functions: ChIP-seq, transcriptional reporter assays
For cytoplasmic/mitochondrial functions: membrane potential assays, cytochrome c release, interaction with HK1/VDAC1
These approaches, when used in combination, allow researchers to parse the complex dual functionality of phosphorylated ATF2 and determine how phosphorylation at T71 impacts its compartment-specific roles in both normal cellular processes and disease states .
Accurate quantification and normalization of Phospho-ATF2 (T71) signals in Western blot experiments are essential for reliable interpretation of results. This process requires careful attention to several methodological considerations:
Image acquisition parameters:
Use a wide dynamic range detection system (digital imaging preferred over film)
Ensure exposure is within linear range (no saturated pixels)
Maintain consistent acquisition settings across experimental replicates
Capture both phospho-ATF2 and normalization controls in same image when possible
Quantification approach:
Use dedicated image analysis software (ImageJ, Image Lab, etc.)
Perform densitometry on defined regions of interest
Subtract local background for each band
Generate integrated density values (area × mean intensity)
Normalization strategies:
Normalization Method | Advantages | Limitations | Best Use Case |
---|---|---|---|
Total ATF2 | Accounts for variations in ATF2 expression | Requires stripping and reprobing or parallel gels | Comparing phosphorylation status independent of expression |
Housekeeping proteins (β-actin, GAPDH) | Simple, widely accepted | May not reflect ATF2-specific variations | General loading control, especially with stable ATF2 expression |
Phosphorylation ratio (pATF2/total ATF2) | Most accurate measure of phosphorylation status | Requires reliable total ATF2 detection | Studies focused on activation state rather than absolute levels |
Internal reference samples | Enables cross-gel/cross-experiment comparison | Requires consistent reference preparation | Long-term studies or comparisons across multiple experiments |
Statistical analysis:
Perform experiments in biological triplicate (minimum)
Apply appropriate statistical tests (t-test, ANOVA) based on experimental design
Report both fold changes and p-values
Present data with error bars representing standard deviation or standard error
Special considerations for phospho-epitopes:
Always include positive controls (cells treated with known activators)
Consider phosphorylation kinetics (time-course experiments)
Be aware of potential rapid dephosphorylation during sample preparation
Use phosphatase inhibitors consistently
When working with the Phospho-ATF2 (T71) antibody at the recommended dilutions (1:500-1:5000) , researchers should optimize exposure times to ensure signals fall within the linear detection range of their imaging system, which is critical for accurate quantification of subtle changes in phosphorylation status.
ATF2 function is regulated by a complex network of post-translational modifications (PTMs), with phosphorylation at T71 being a critical regulatory site that often works in concert with other modifications. Understanding this interplay requires specialized experimental approaches:
Sequential and combinatorial phosphorylation analysis:
Investigate the relationship between T71 phosphorylation and T69 phosphorylation
Perform time-course experiments with various stimuli
Use site-specific phospho-antibodies (including Phospho-ATF2 (T71) antibody)
Implement phospho-mimetic and phospho-resistant mutations (T71E/D vs. T71A)
Cross-talk with other modification types:
Examine interactions between phosphorylation and other PTMs (acetylation, SUMOylation, ubiquitination)
Use deacetylase inhibitors, proteasome inhibitors, or SUMO pathway modulators
Implement mass spectrometry-based approaches for global PTM mapping
Develop or acquire modification-specific antibodies for co-detection studies
PTM-dependent protein interactions:
Perform co-immunoprecipitation with Phospho-ATF2 (T71) antibody
Compare interactome of phosphorylated vs. non-phosphorylated ATF2
Identify readers of the phospho-T71 mark
Implement proximity ligation assays to detect interactions in situ
Functional outcomes of modification patterns:
Assess DNA binding properties using electrophoretic mobility shift assays
Measure transcriptional activity with reporter constructs
Evaluate histone acetyltransferase (HAT) activity with in vitro assays
Determine effects on cellular processes (DNA damage response, apoptosis, cell cycle)
Pathway integration analysis:
Map kinase signaling pathways leading to different modification patterns
Use specific inhibitors of JNK, p38, ATM, and other relevant kinases
Implement mathematical modeling to predict modification interdependencies
Correlate phosphorylation patterns with biological outcomes
These approaches enable researchers to move beyond single-modification studies and develop a more comprehensive understanding of how T71 phosphorylation coordinates with other modifications to fine-tune ATF2 function in different cellular contexts and in response to various stimuli .
Contradictory findings regarding ATF2 phosphorylation across different experimental systems are not uncommon and can result from various methodological, biological, and technical factors. Resolving such discrepancies requires systematic troubleshooting and careful experimental design:
Methodological standardization:
Compare antibody performance across studies (clone, supplier, lot number)
Standardize application protocols (dilutions, incubation conditions)
Implement consistent sample preparation methods
Use recombinant phosphorylated standards for cross-laboratory calibration
Biological system variations:
Analyze cell type-specific effects (ATF2 can elicit oncogenic or tumor suppressor activities depending on tissue/cell type)
Consider species differences in ATF2 sequence and regulation
Evaluate the impact of culture conditions and passage number
Assess microenvironmental factors that might influence signaling
Technical approach diversification:
Validate findings across multiple techniques (WB, IF, flow cytometry, ELISA)
Implement complementary approaches (mass spectrometry for direct PTM detection)
Use genetic approaches (CRISPR/Cas9 editing, site-directed mutagenesis)
Conduct functional validation experiments
Systematic meta-analysis framework:
Analysis Parameter | Investigation Approach | Resolution Strategy |
---|---|---|
Stimulus conditions | Compare concentration, duration, delivery method | Create standardized response curves |
Antibody specificity | Perform side-by-side comparison of different antibodies | Identify consensus signals across antibodies |
Signaling context | Map activation status of upstream and downstream components | Define pathway-specific phosphorylation patterns |
Temporal dynamics | Conduct detailed time-course experiments | Identify system-specific kinetic profiles |
Subcellular compartmentalization | Analyze fraction-specific phosphorylation patterns | Develop compartment-specific models |
Collaborative validation approaches:
Implement multi-laboratory testing of identical samples
Share positive control materials and protocols
Develop consensus guidelines for ATF2 phosphorylation analysis
Create repository of validated reagents and protocols
When working with Phospho-ATF2 (T71) antibody specifically, researchers should ensure they are using validated antibodies such as the recombinant rabbit monoclonal antibody (clone 4F2) at appropriate dilutions for each application (WB:1:500-1:5000, IF:1:20-1:200) , while implementing appropriate controls for each experimental system to facilitate accurate cross-study comparisons.
ATF2 exhibits context-dependent roles in cancer biology, functioning as either an oncogene or tumor suppressor depending on the tissue or cell type . Phospho-ATF2 (T71) antibody provides a valuable tool for investigating these complex roles across various cancer types and experimental models:
Cancer type-specific phosphorylation patterns:
Compare phospho-ATF2 levels across cancer cell lines and patient samples
Correlate with clinical outcomes and tumor characteristics
Develop tissue microarray analysis protocols using the antibody (typically at 1:50-1:100 dilution)
Establish prognostic significance of T71 phosphorylation in specific cancer types
Functional studies in cancer models:
Manipulate ATF2 phosphorylation through kinase modulation or mutation
Assess impacts on hallmark cancer phenotypes (proliferation, invasion, metastasis)
Implement xenograft models with phosphorylation-dependent ATF2 variants
Correlate phosphorylation status with therapy response
Mechanistic investigation of dual functionality:
Determine phosphorylation-dependent target gene specificity in cancer cells
Investigate compartment-specific functions (nuclear vs. cytoplasmic)
Analyze interaction with oncogenic and tumor suppressor pathways
Map cross-talk with other transcription factors and signaling molecules
Therapeutic targeting opportunities:
Screen for compounds that modulate ATF2 phosphorylation
Assess synergistic effects with established cancer therapies
Develop phosphorylation-specific intervention strategies
Identify biomarkers for phosphorylation-dependent therapeutic responses
Single-cell analysis approaches:
The antibody's ability to specifically detect the phosphorylated form enables researchers to distinguish active ATF2 from total protein, providing crucial insights into its activation state in cancer cells. This is particularly important given ATF2's involvement in key cancer-related processes, including cell growth regulation, DNA damage response, and apoptosis pathways .
Investigating the dynamic phosphorylation of ATF2 in response to cellular stress requires careful methodological considerations to capture the often rapid and transient nature of these signaling events:
Temporal resolution optimization:
Design fine-grained time-course experiments (seconds to hours)
Implement rapid sample collection and preservation techniques
Use phosphatase inhibitors consistently across all samples
Consider live-cell imaging with phospho-specific biosensors as complementary approach
Stress stimulus parameters:
Calibrate stimulus intensity to avoid oversaturation of response
Compare different stress types (oxidative, genotoxic, inflammatory, metabolic)
Implement dose-response studies alongside time-course experiments
Consider physiologically relevant stress conditions alongside standard laboratory stimuli
Single-cell vs. population analysis:
Use flow cytometry with phospho-specific antibodies for heterogeneity assessment
Implement immunofluorescence microscopy for spatial information
Consider single-cell Western techniques for protein-limited samples
Correlate with single-cell transcriptomics when possible
Pathway crosstalk analysis:
Simultaneously monitor multiple phosphorylation sites (T69, T71)
Track activation of upstream kinases (JNK, p38, ATM)
Use specific pathway inhibitors to dissect contribution to ATF2 phosphorylation
Implement systems biology approaches to model signaling networks
Technical considerations for Phospho-ATF2 (T71) antibody use:
Optimize antibody concentration based on signal intensity and specificity
Include both phosphorylation-positive and negative controls in each experiment
Consider multiple detection methods (colorimetric, chemiluminescent, fluorescent)
Quantification and normalization approaches:
Experimental Approach | Optimal Quantification Method | Normalization Strategy |
---|---|---|
Western blot time-course | Densitometry with linear range detection | Ratio to total ATF2 at each timepoint |
Immunofluorescence | Integrated nuclear intensity measurements | Cell-by-cell ratio to DAPI or total ATF2 |
Flow cytometry | Median fluorescence intensity | Unstimulated control population |
ELISA-based methods | Standard curve-based absolute quantification | Total protein or total ATF2 normalization |
By carefully implementing these methodological considerations, researchers can achieve high-quality, reproducible data on the dynamics of ATF2 phosphorylation in response to various cellular stressors, providing insights into the temporal regulation of stress response pathways and their dysregulation in disease states .