AKT1 antibodies are immunoglobulins designed to bind specifically to AKT1. They are categorized based on their target epitopes, isoform specificity, and phosphorylation state:
AKT1 antibodies are pivotal in deciphering AKT1’s roles in oncology, cell biology, and signaling pathways.
Purpose: Quantify AKT1 protein levels or phosphorylation status.
Example:
Workflow: Lysate preparation → SDS-PAGE → Transfer to PVDF membrane → Antibody probing → Detection via HRP-conjugated secondary antibodies.
Purpose: Identify AKT1 interactors or assess phosphorylation-dependent binding partners.
Example:
Purpose: Localize AKT1 in tissue sections.
Example:
AKT1 activation requires dual phosphorylation at Thr308 and Ser473.
Role: Critical for full kinase activity; phosphorylation at Ser473 enhances substrate targeting.
Antibodies:
Role: Initial activation step mediated by PDK1.
Antibodies:
sc-33437 (Santa Cruz): Validates Thr308 phosphorylation in insulin-stimulated cells.
AKT1 Interactome: SILAC-based mass spectrometry identified 213 AKT1 interactors in HEK293 cells, including CDK1 and CCNB1 .
Specificity Issues: Pan-AKT antibodies may cross-react with AKT2/3. Use isoform-specific reagents (e.g., MAB1775) to avoid ambiguity .
Phospho-Specific Validation: Confirm phosphorylation sites using orthogonal methods (e.g., kinase assays) alongside antibodies.
Cancer Therapeutics: Targeting AKT1 with isoform-specific antibodies may reduce off-target effects in cancer therapy .
V-Akt Murine Thymoma Viral Oncogene Homolog 1, Protein Kinase B Alpha, Proto-Oncogene C-Akt, RAC-PK-Alpha, EC 2.7.11.1, PKB Alpha, CWS6, PKB, RAC, RAC-Alpha Serine/Threonine-Protein Kinase, Rac Protein Kinase Alpha, Protein Kinase B, PKB-ALPHA, RAC-ALPHA, EC 2.7.11, AKT1m, PRKBA, AKT, RAC-alpha serine/threonine-protein kinase.
AKT1 antibody was purified from mouse ascetic fluids by protein-A affinity chromatography.
PAT6D5AT.
Anti-human AKT1 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human AKT1 amino acids 1-480 purified from E. coli.
Mouse IgG2a heavy chain and κ light chain.
AKT1 (also known as Protein Kinase B alpha, RAC-alpha serine/threonine-protein kinase, or PKB alpha) is a 56 kDa serine/threonine kinase that mediates multiple cellular processes including apoptosis, angiogenesis, metabolism, and cell proliferation in both normal and cancerous cells . As one of three AKT isoforms (AKT1, AKT2, and AKT3, also known as PKBα, β, and γ), AKT1 has unique tissue-specific functions, including cardioprotective effects supporting physiological heart growth and function . At the subcellular level, AKT1 can be found in both cytoplasmic and nuclear compartments , facilitating its diverse signaling roles across multiple cellular pathways.
Phospho-specific AKT1 antibodies (such as those targeting phospho-Serine 473) recognize AKT1 only when phosphorylated at specific amino acid residues, indicating its activated state. These antibodies are designed to have minimal reactivity against non-phosphorylated AKT . They are generated using phospho-peptide immunogens and often undergo affinity purification to remove antibodies recognizing non-phosphorylated epitopes.
In contrast, total AKT1 antibodies detect the protein regardless of its phosphorylation status, binding to epitopes that are accessible in both active and inactive conformations. These antibodies are valuable for normalizing phospho-AKT1 levels against total protein expression in quantitative analyses. When using both antibody types in parallel experiments, researchers can assess both AKT1 expression levels and activation status simultaneously .
Reliable differentiation between AKT isoforms requires antibodies with demonstrated specificity. High-quality isoform-specific antibodies show no cross-reactivity with other AKT family members, as validated by Western blot analysis against recombinant proteins. For example, some commercial antibodies have been tested against recombinant human AKT1, AKT2, and AKT3, confirming specificity for AKT1 with no cross-reactivity to the other isoforms .
For definitive validation, knockdown/knockout models provide the gold standard. Western blot analysis comparing parental cell lines with AKT1 knockout lines demonstrates antibody specificity when a band appears in the wild-type sample but is absent in the knockout sample . When designing isoform-specific experiments, choose antibodies validated through both recombinant protein testing and knockout cell line verification to ensure accurate targeting of your specific AKT isoform of interest.
Selecting the appropriate AKT1 antibody requires consideration of multiple factors:
Robust experimental design requires multiple controls when studying AKT1 signaling:
Positive controls: Include cell lines or tissues known to express AKT1, such as HeLa cells, MCF-7 cells, or C2C12 myoblasts . For phospho-AKT1 studies, use samples treated with pathway activators (e.g., insulin, EGF, or serum stimulation).
Negative controls: For Western blot specificity, include AKT1 knockout cell lines when available . For immunostaining, use secondary antibody-only controls to assess background.
Isoform specificity controls: When available, include recombinant AKT1, AKT2, and AKT3 proteins to confirm antibody specificity .
Phosphorylation state controls: For phospho-AKT1 studies, include:
Untreated/serum-starved samples (low phosphorylation)
Phosphatase-treated samples (dephosphorylated negative control)
Pathway inhibitor controls (e.g., PI3K inhibitors like LY294002 or Wortmannin)
Loading controls: Include housekeeping proteins (GAPDH, β-actin) for Western blot normalization .
These controls collectively ensure the reliability of your data and facilitate the accurate interpretation of AKT1 signaling dynamics in your experimental system.
Preserving AKT1 phosphorylation requires careful attention to sample preparation:
Rapid processing: Phosphorylation states can change rapidly after sample collection. Process samples immediately on ice to minimize phosphatase activity.
Phosphatase inhibitors: Include comprehensive phosphatase inhibitor cocktails in all lysis buffers. Common components include sodium fluoride, sodium orthovanadate, β-glycerophosphate, and pyrophosphate.
Lysis buffer composition: Use RIPA or modified RIPA buffers containing 1% NP-40 or Triton X-100, 0.1-0.5% sodium deoxycholate, and 0.1% SDS, supplemented with protease inhibitors.
Temperature considerations: Maintain samples at 4°C throughout processing to minimize enzymatic activity that might alter phosphorylation.
Fixation for microscopy: For immunofluorescence studies, rapid fixation with paraformaldehyde (typically 0.5-4%) helps preserve phosphorylation states . Some epitopes may require specific fixation protocols—for example, certain phospho-AKT1 antibodies have been validated with 0.5% PFA fixation .
Storage: Aliquot lysates to avoid freeze-thaw cycles, and store at -80°C for long-term preservation of phosphorylation status.
These precautions are critical for obtaining accurate data on AKT1 activation state, particularly when studying dynamic signaling events that involve transient phosphorylation.
Optimal Western blot conditions for AKT1 antibodies require careful optimization:
Sample preparation: Lyse cells in RIPA buffer supplemented with protease and phosphatase inhibitors. Load 15-25 μg of total protein per lane (20 μg is commonly used) .
Gel selection: Use 7.5% to 10% SDS-PAGE gels to optimize separation around the 56-60 kDa range where AKT1 migrates .
Transfer conditions: Transfer to PVDF membranes (preferred over nitrocellulose for phospho-epitopes) using standard wet transfer protocols.
Blocking: Block with 5% non-fat dry milk in TBST for total AKT1 detection. For phospho-specific antibodies, use 5% BSA in TBST to avoid phosphatase contamination in milk.
Antibody dilutions: Working concentrations vary by antibody and manufacturer:
Incubation conditions: Incubate primary antibodies overnight at 4°C with gentle agitation for optimal signal-to-noise ratio.
Detection system: Use HRP-conjugated secondary antibodies with ECL detection systems. For low abundance targets, consider enhanced sensitivity substrates .
Expected results: AKT1 typically appears as a distinct band at approximately 56-60 kDa .
Adjusting these parameters for your specific experimental system will help achieve clear, specific detection of AKT1 and its phosphorylated forms.
Optimizing AKT1 antibodies for immunostaining requires attention to several critical variables:
Fixation methods:
Antigen retrieval (for FFPE tissues):
Blocking:
5-10% normal serum (from the species of secondary antibody)
1-3% BSA in PBS or TBS
Antibody dilutions:
Incubation conditions:
Primary antibody: 1 hour at room temperature or overnight at 4°C
Secondary antibody: 30-60 minutes at room temperature
Detection systems:
Expected localization:
Counterstaining:
Single-cell analysis of AKT1 activation requires techniques that preserve spatial information and allow for quantitative assessment:
Quantitative immunofluorescence microscopy:
Use dual staining with phospho-specific and total AKT1 antibodies on fixed cells
Apply ratio imaging to calculate phospho-AKT1/total AKT1 on a per-cell basis
Measure nuclear-to-cytoplasmic ratios as an indicator of AKT1 activation and translocation
Employ high-content imaging systems for automated quantification across large cell populations
Flow cytometry:
Fix and permeabilize cells using methanol or commercial permeabilization kits
Stain with fluorophore-conjugated phospho-AKT1 antibodies
Analyze fluorescence intensity distributions to identify responding subpopulations
Combine with surface markers to correlate AKT1 activation with cell phenotypes
Time-lapse imaging with biosensors:
Utilize FRET-based reporters to monitor AKT1 activation dynamics in living cells
Complement with immunostaining of fixed timepoints using validated phospho-AKT1 antibodies
Single-cell western blotting:
Apply microfluidic platforms that enable Western blot analysis of individual cells
Probe with AKT1 and phospho-AKT1 antibodies to assess activation at the single-cell level
Mass cytometry (CyTOF):
Label with metal-conjugated antibodies against AKT1 pathway components
Simultaneously measure multiple phosphorylation sites to map signaling networks
These approaches can reveal heterogeneity in AKT1 activation within seemingly homogeneous populations, providing insights into differential responses to treatments and correlation with other cellular phenotypes.
Troubleshooting non-specific binding and high background requires systematic optimization:
Antibody validation:
Western blot optimization:
Increase blocking time/concentration (5% milk or BSA for 1-2 hours)
Reduce primary antibody concentration (perform titration experiments)
Increase washing duration and number of washes (5-6 washes of 5-10 minutes each)
Add 0.1-0.5% Tween-20 to washing buffers
Consider alternative membranes (PVDF vs. nitrocellulose)
Immunostaining optimization:
Implement additional blocking steps (e.g., avidin/biotin blocking for biotin-based detection)
Pre-absorb antibodies with acetone powder from relevant tissues
Include detergent (0.1-0.3% Triton X-100) in antibody diluents
Reduce autofluorescence using Sudan Black B (for fluorescence applications)
Optimize fixation conditions which can affect epitope accessibility
Sample preparation considerations:
Ensure complete cell lysis to avoid aggregate formation
Pre-clear lysates by centrifugation to remove insoluble material
Consider using TCA precipitation to concentrate proteins while removing interfering compounds
Application-specific approaches:
For IHC: Implement hydrogen peroxide blocking to reduce endogenous peroxidase activity
For ICC/IF: Use phalloidin counterstaining to visualize cell boundaries and assess non-specific binding patterns
Carefully documenting optimization steps and including appropriate negative controls in each experiment will help distinguish specific from non-specific signals.
Conflicting results between phospho-AKT1 antibodies require careful analysis and validation:
Numerous factors can influence the reproducibility of AKT1 phosphorylation measurements:
Biological variables:
Cell density and confluency (affects contact inhibition and growth factor signaling)
Passage number and cellular senescence
Serum batch variations affecting growth factor content
Circadian rhythm effects on signaling pathway activity
Genomic instability in cancer cell lines causing population drift
Technical variables:
Sample handling time affecting phosphorylation decay rates
Variations in lysis buffer composition and effectiveness
Phosphatase inhibitor freshness and concentration
Freeze-thaw cycles degrading phospho-epitopes
Antibody lot-to-lot variations in specificity and sensitivity
Analytical considerations:
Normalization methods (total AKT1 vs. housekeeping proteins)
Quantification approaches (densitometry settings, dynamic range limitations)
Exposure times for chemiluminescent Western blots
Detection system linearity and saturation
Image processing methods affecting signal quantification
Experimental design factors:
Timing of stimulation/inhibition relative to sample collection
Variability in drug or stimulant preparation and administration
Temperature fluctuations during experimental procedures
Operator-to-operator variations in technique
To maximize reproducibility:
Standardize protocols with detailed SOPs
Process samples in parallel whenever possible
Include internal reference samples across experiments
Consider multiparametric measurements when feasible
Document all reagents, including lot numbers and preparation dates
AKT1 antibodies can be powerful tools for studying disease-associated mutations through several approaches:
Mutation-specific antibodies:
Custom antibodies can be generated against specific mutant epitopes (e.g., the common E17K mutation)
These allow direct detection of mutant AKT1 in patient samples by IHC or Western blot
Validation requires parallel testing in samples with confirmed genotypes
Functional phospho-site analysis:
Subcellular localization studies:
Some mutations (like E17K) affect membrane recruitment and subcellular distribution
Immunofluorescence with AKT1 antibodies can reveal altered localization patterns
Co-localization with membrane markers provides additional functional insights
Pathway interaction analysis:
Immunoprecipitation with AKT1 antibodies followed by mass spectrometry
Comparison of wild-type vs. mutant AKT1 interactomes
Identification of altered binding partners specific to disease-associated mutations
Patient stratification applications:
IHC with phospho-AKT1 antibodies on patient tissues can identify activated AKT1 signaling
Correlation with genomic data to link specific mutations with protein expression/activation patterns
Potential prognostic and predictive biomarker applications in cancer and other diseases
These approaches are particularly relevant for cancer research, where AKT1 mutations have been identified in breast, colorectal, and other cancers, as well as for Proteus syndrome and Cowden syndrome studies .
AKT1 antibodies are enabling several innovative applications in cancer research:
Single-cell profiling of tumor heterogeneity:
Mass cytometry with AKT1 and phospho-AKT1 antibodies to profile thousands of individual cells
Identification of therapy-resistant subpopulations with distinct AKT1 activation states
Spatial analysis of AKT1 activation in tumor microenvironment using multiplexed IHC/IF
Drug resistance mechanisms:
Monitoring AKT1 phosphorylation dynamics during treatment and relapse
Identifying compensatory activation of AKT1 following inhibition of parallel pathways
Correlation of phospho-AKT1 levels with response to targeted therapies
Companion diagnostics for AKT pathway inhibitors:
IHC-based measurement of AKT1 activation as predictive biomarkers
Phospho-AKT1 quantification for patient selection and response monitoring
Integration with genomic analysis to correlate mutations with protein activity
Circulating tumor cell (CTC) analysis:
Phospho-AKT1 immunostaining of CTCs as liquid biopsy approach
Monitoring treatment response through sequential phospho-AKT1 measurement
Correlation with clinical outcomes and drug resistance
In vitro diagnostic applications:
Highly specific AKT1 antibodies for distinguishing liver cancer and other malignancies
Prognostic assessment based on phospho-AKT1 levels in tumor samples
Multiparameter profiling of AKT pathway activation combined with other cancer markers
These applications are critical for advancing personalized medicine approaches in cancer, where AKT1 status may determine therapeutic strategies and predict treatment outcomes.
Integration of AKT1 antibodies with complementary technologies enables comprehensive pathway analysis:
Multi-omics integration:
Combining phospho-AKT1 immunoprecipitation with phosphoproteomics to identify substrates
Correlating transcriptomics data with AKT1 activation states measured by antibody-based methods
Integrating genomic mutation data with protein-level AKT1 activation profiles
Live-cell imaging combined with fixed-cell validation:
Real-time monitoring of AKT1 activity using fluorescent biosensors
Validation of key timepoints with phospho-specific antibodies
Correlation of dynamic signaling behaviors with cellular outcomes
Spatial analysis technologies:
Combining AKT1 antibodies with digital spatial profiling platforms
Mapping AKT1 activation gradients in tissue microenvironments
Co-localization of phospho-AKT1 with cell type-specific markers in complex tissues
High-throughput screening applications:
Automated immunofluorescence with AKT1 antibodies for drug screening
Identifying compounds that modulate AKT1 phosphorylation or localization
Multiplexed readouts combining AKT1 with downstream effectors
Proximity-based interaction methods:
Proximity ligation assays to detect AKT1 interactions with binding partners
BioID or APEX2 proximity labeling with AKT1 fusions followed by antibody validation
FRET-based approaches to measure direct protein-protein interactions
In vivo imaging:
Radiolabeled AKT1 antibodies for PET imaging of pathway activation
Optical imaging with near-infrared fluorophore-conjugated antibodies in preclinical models
Correlation of imaging findings with ex vivo tissue analysis
These integrated approaches provide a more complete understanding of AKT1 signaling dynamics and context-dependent functions across different physiological and pathological states.
The relationship between AKT1 expression/activation and clinical outcomes exhibits disease-specific patterns:
Liver cancer correlations:
Analysis of AKT1 gene expression in liver hepatocellular carcinoma (LIHC) reveals significant differences compared to normal tissue
Patient stratification based on AKT1 expression levels (high vs. low) shows correlation with survival outcomes
Specific mutations, such as R273Q, have been associated with liver cancer development
Breast cancer subtypes:
Methodological considerations for clinical correlation studies:
Importance of standardized staining protocols for clinical samples
Quantitative scoring systems for phospho-AKT1 immunohistochemistry
Integration with other biomarkers and clinicopathological factors
Multivariate analysis to establish independent prognostic value
Therapeutic implications:
Potential for AKT1 expression/activation as predictive biomarkers for PI3K/AKT/mTOR inhibitors
Monitoring phospho-AKT1 levels during treatment to assess target engagement
Identification of compensatory mechanisms in resistance development
Researchers investigating AKT1 as a prognostic biomarker should employ well-validated antibodies, standardized quantification methods, and appropriate statistical approaches to establish clinically meaningful correlations.
Despite their utility, AKT1 antibodies face several important limitations in translational research:
Isoform specificity challenges:
Epitope accessibility issues:
Protein-protein interactions or conformational changes may mask epitopes
Fixation and processing artifacts in clinical samples can affect antibody binding
Non-standardized sample preparation across institutions limits comparability
Quantification limitations:
Semi-quantitative nature of IHC scoring systems
Dynamic range limitations in Western blot densitometry
Challenges in absolute quantification of phosphorylation stoichiometry
Temporal considerations:
Phosphorylation states represent snapshots of dynamic processes
Pre-analytical variables (ischemia time, fixation delay) affect phospho-epitope preservation
Limited ability to capture signaling dynamics in fixed clinical specimens
Reproducibility concerns:
Lot-to-lot variability in antibody performance
Non-standardized protocols across laboratories
Limited cross-validation between different antibody clones
Inadequate reporting of validation metrics in publications
Technical barriers to multiplexing:
Challenges in combining multiple rabbit-derived antibodies on single samples
Limited spectral separation in conventional fluorescence microscopy
Cost and complexity of advanced multiplexing platforms
Addressing these limitations requires coordinated efforts between researchers, antibody manufacturers, and clinical laboratories to establish standardized protocols, validation criteria, and reporting standards.
Computational methods are increasingly enhancing antibody-based AKT1 research:
In silico epitope analysis:
Image analysis algorithms:
Automated quantification of AKT1 staining intensity and subcellular localization
Machine learning approaches for pattern recognition in complex tissues
Deep learning models for cell classification based on AKT1 activation states
Network analysis integration:
Mapping antibody-derived AKT1 activation data onto known signaling networks
Identification of context-specific feedback mechanisms and crosstalk
Prediction of pathway vulnerabilities based on AKT1 activation patterns
Virtual screening for AKT1 modulators:
Structure-based drug design targeting specific AKT1 conformations
Computational prediction of compounds that affect AKT1 phosphorylation
Integration with high-content screening data from antibody-based assays
Predictive biomarker models:
Development of multivariate models incorporating AKT1 antibody data
Integration of phospho-AKT1 levels with genomic alterations for patient stratification
Machine learning algorithms to predict treatment response based on AKT1 pathway activation These computational approaches substantially enhance the value of antibody-generated data by providing deeper insights into AKT1 biology, identifying novel therapeutic targets, and supporting personalized medicine applications.
Protein Kinase B was first cloned by three independent groups in 1991, following the identification of its viral homolog, the v-Akt proto-oncogene, expressed by a transforming retrovirus (AKT-8) isolated from a spontaneous thymic lymphoma of an AKR mouse . The three mammalian isoforms, Akt1, Akt2, and Akt3, share a high degree of similarity but have distinct physiological roles .
Protein Kinase B Alpha is a key mediator of the phosphoinositide 3-kinase (PI3K) signaling pathway . Upon activation by PI3K, Protein Kinase B Alpha is phosphorylated at two key residues, T308 and S473, by PDK1 and mTORC2 or DNA-PK, respectively . This phosphorylation is essential for its full kinase activity, allowing it to regulate various downstream targets involved in cell survival, growth, and metabolism .
In the context of cancer, Protein Kinase B Alpha has been shown to regulate tumor growth, survival, and invasiveness of tumor cells . It increases cell proliferation through cell cycle proteins like p21, p27, and cyclin D1 and impairs apoptosis via p53 . However, it also decreases the migration of cancer cells by regulating proteins such as TSC2, palladin, and EMT-proteins .
The Mouse Anti Human Protein Kinase B Alpha antibody is a monoclonal antibody used in research to detect and study the human form of Protein Kinase B Alpha. This antibody is produced by immunizing mice with human Protein Kinase B Alpha and then isolating the specific antibody-producing cells. The resulting monoclonal antibody can specifically bind to human Protein Kinase B Alpha, allowing researchers to investigate its expression, localization, and function in various biological samples.
The Mouse Anti Human Protein Kinase B Alpha antibody is widely used in various research applications, including: