AKT3 Human Recombinant produced in HEK293 Cells is a single, glycosylated polypeptide chain containing 485 amino acids (1-479a.a) and having a molecular mass of 56.5kDa.
AKT3 is fused to a 6 amino acid His-tag at C-terminus and is purified by proprietary chromatographic techniques.
AKT Serine/Threonine Kinase 3 (Akt3) is a member of the Akt subfamily of serine/threonine protein kinases. Akt3 is one of three closely related serine/threonine-protein kinases (Akt1, Akt2, and Akt3) known as the Akt kinase, which regulate various cellular processes including proliferation, metabolism, cell survival, growth, and angiogenesis. This regulation is mediated through the phosphorylation of serine and/or threonine residues on a range of downstream substrates. Akt3 plays a role in brain development and is essential for the survival of malignant glioma cells.
Recombinant human AKT3, expressed in HEK293 cells, is a single, glycosylated polypeptide chain comprising 485 amino acids (1-479a.a) with a molecular weight of 56.5 kDa.
A 6-amino acid His-tag is fused to the C-terminus of AKT3. The protein is purified using proprietary chromatographic techniques.
Sterile, colorless solution.
The AKT3 solution (0.25 mg/ml) is supplied in PBS (pH 7.4) with 10% glycerol.
For short-term storage (2-4 weeks), store at 4°C.
For long-term storage, freeze at -20°C.
Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage.
Avoid repeated freeze-thaw cycles.
Greater than 90% purity as determined by SDS-PAGE analysis.
RAC-gamma serine/threonine-protein kinase, Protein kinase Akt-3, Protein kinase B gamma, PKB gamma, RAC-PK-gamma, STK-2, AKT3, PKBG.
HEK293 Cells.
MSDVTIVKEG WVQKRGEYIK NWRPRYFLLK TDGSFIGYKE KPQDVDLPYP LNNFSVAKCQ LMKTERPKPN TFIIRCLQWT TVIERTFHVD TPEEREEWTE AIQAVADRLQ RQEEERMNCS PTSQIDNIGE EEMDASTTHH KRKTMNDFDY LKLLGKGTFG KVILVREKAS GKYYAMKILK KEVIIAKDEV AHTLTESRVL KNTRHPFLTS LKYSFQTKDR LCFVMEYVNG GELFFHLSRE RVFSEDRTRF YGAEIVSALD YLHSGKIVYR DLKLENLMLD KDGHIKITDF GLCKEGITDA ATMKTFCGTP EYLAPEVLED NDYGRAVDWW GLGVVMYEMM CGRLPFYNQD HEKLFELILM EDIKFPRTLS SDAKSLLSGL LIKDPNKRLG GGPDDAKEIM RHSFFSGVNW QDVYDKKLVP PFKPQVTSET DTRYFDEEFT AQTITITPPE KYDEDGMDCM DNERRPHFPQ FSYSASGREH HHHHH.
AKT3, also known as protein kinase B gamma (PKB gamma) and RAC-gamma serine/threonine kinase (RAC-PK-gamma), is part of the AKT family that includes AKT1 (PKB alpha) and AKT2 (PKB beta). While structurally similar with an amino-terminal pleckstrin homology domain, a central kinase domain, and a carboxyl-terminal regulatory domain, AKT3 has unique functions. Most notably, AKT3 preferably phosphorylates p47phox and activates NADPH oxidase, resulting in robust induction of reactive oxygen species (ROS). This distinctive property leads to activation of the DNA damage response pathway and upregulation of p53 expression, differentiating AKT3 functionally from its family members . The protein is detected at approximately 60 kDa in human samples, with specific expression patterns across different tissues and cancer types .
For reliable detection of AKT3 in human samples, researchers should employ multiple complementary approaches. Western blotting using specific monoclonal antibodies (such as Clone #199864) can detect AKT3 at approximately 60 kDa in human cell lines like DU145 (prostate carcinoma) and MDA-MB-231 (breast cancer). For optimal results, use 0.5 μg/mL antibody concentration with HRP-conjugated secondary antibodies and perform experiments under reducing conditions . RNA-sequencing provides comprehensive expression analysis, as demonstrated by studies using The Cancer Genome Atlas database to compare AKT3 levels between tumor and normal tissues . For phosphorylation state analysis, phospho-specific antibodies targeting key regulatory sites (e.g., Ser472) are essential. Importantly, researchers must consider potential splice variants, which may have distinct functions but similar molecular weights, requiring additional verification through RT-PCR or mass spectrometry .
In normal cellular physiology, AKT3 functions as a central regulator in diverse processes including glucose uptake, cell cycle progression, and apoptosis . Its activation follows the canonical PI3K pathway, where growth factors or hormones stimulate PI3K, generating PIP3 at the plasma membrane, which recruits AKT3 via its pleckstrin homology domain. Following membrane recruitment, AKT3 is activated through phosphorylation at two key regulatory sites. Once activated, AKT3 phosphorylates numerous downstream substrates, regulating metabolism, protein synthesis, and cell survival. Unlike other AKT isoforms, AKT3 has a particular role in modulating ROS levels through NADPH oxidase activation, which impacts cellular redox balance and stress responses . To study these normal functions experimentally, researchers should use tissue-specific knockout models or inducible systems that allow temporal control of AKT3 activity, as constitutive deletion may trigger compensatory mechanisms through other AKT isoforms .
AKT3 dysregulation varies significantly across cancer types, with distinct patterns and consequences:
In head and neck squamous cell carcinoma (HNSCC), AKT3 is specifically elevated compared to normal tissues and correlates with genes related to the immunosuppressive microenvironment more strongly than other AKT isoforms. Experimental knockdown of AKT3 in HNSCC cell lines impairs proliferation, shifts cell cycle distribution from G2/M to G1/G0 phase, increases apoptosis, and downregulates immunosuppressive gene expression .
For breast cancer, particularly triple-negative breast cancer (TNBC), the AKT3 gene is frequently amplified with a novel recurrent fusion oncogene MAGI3-AKT3 identified. High levels of AKT3 significantly correlate with patient survival duration. Interestingly, a splice variant lacking the Ser472 phosphorylation site induces apoptosis and suppresses TNBC growth by upregulating pro-apoptotic Bim and activating Bax and caspase-3 .
In glioblastoma (GBM), AKT3 overexpression promotes malignancy, with siRNA knockdown significantly decreasing cell viability, proliferation, invasion, and metastasis. This effect is regulated by tumor suppressor microRNAs including miR-610 .
These distinct patterns suggest context-dependent roles for AKT3, necessitating cancer-type specific therapeutic approaches.
Distinguishing AKT3-specific functions from general AKT family effects requires sophisticated experimental approaches:
Genetic manipulation: Employ isoform-specific siRNA/shRNA or CRISPR/Cas9 to target AKT3's unique sequences. This approach revealed that AKT3 knockdown in HNSCC cells specifically impaired proliferation, shifted cell cycle distribution, and increased apoptosis .
Substrate specificity analysis: Focus on AKT3-preferred substrates like p47phox. Studies showed that AKT3, more than other isoforms, phosphorylates p47phox to activate NADPH oxidase and generate ROS. This specificity was confirmed by expressing AKT3 in p47phox-/- MEFs, which failed to induce ROS .
Isogenic cell systems: Utilize isogenic cell lines expressing individual AKT isoforms to compare direct effects. This approach demonstrated AKT3's unique ability to induce ROS and activate DNA damage response pathways .
Readout selection: Measure distinct phenotypic outcomes like ROS levels, DNA damage markers, and p53 activation. The antioxidant N-acetylcysteine specifically rescued proliferation defects in AKT3-expressing cells, confirming ROS induction as an AKT3-specific mechanism .
Domain swapping experiments: Create chimeric proteins exchanging domains between AKT isoforms to identify regions responsible for functional specificity.
These approaches help delineate AKT3's unique contributions to cellular phenotypes and disease progression.
AKT3 exhibits context-dependent roles in cancer, with significant contradictions across different models:
These contradictions likely stem from:
Genetic background variations across models
Splice variant expression differences
Tissue-specific signaling networks
Downstream effector availability
Compensatory mechanisms through other AKT isoforms
Resolving these contradictions requires comprehensive profiling of AKT3 interactions, post-translational modifications, and splice variants in each specific context. Researchers should carefully consider these factors when designing experiments and interpreting results .
AKT3 exhibits a unique mechanism for ROS regulation that distinguishes it from other AKT isoforms. Studies using isogenic cell lines revealed that AKT3 is the most potent inducer of ROS among the three AKT proteins. The primary mechanism involves AKT3's preferential phosphorylation of p47phox, a regulatory subunit of NADPH oxidase. This phosphorylation activates the NADPH oxidase complex, triggering robust ROS production. The specificity of this mechanism was confirmed through multiple experimental approaches: expression of AKT3 in p47phox-/- MEFs failed to induce ROS, demonstrating the essential role of this substrate in AKT3-mediated oxidative stress .
Consequently, AKT3-expressing cells activate the DNA damage response pathway, increase p53 levels, and express high levels of p53's direct transcriptional target miR-34. This cascade results in proliferation defects that can be rescued by the antioxidant N-acetylcysteine, confirming ROS as the mediator of these effects. The importance of this pathway is further validated by observations in p53-/- and INK4a-/-/Arf-/- MEFs, where AKT3 overexpression failed to inhibit proliferation despite inducing high ROS levels . This unique signaling axis represents a potential vulnerability that could be exploited for therapeutic intervention in AKT3-driven cancers.
The relationship between AKT3 and p53 forms a critical axis in cancer biology with significant implications for tumor progression and treatment response. AKT3-induced ROS activates the DNA damage response, leading to upregulation of p53 expression. Consequently, AKT3-expressing cells exhibit elevated p53 levels and increased expression of p53 transcriptional targets like miR-34. This activation contributes to the reduced proliferation rate observed in cells with high AKT3 activity .
This adaptation is demonstrated experimentally by the observation that p53 loss reverses the proliferation defect in AKT3-expressing cells. Similarly, AKT3 overexpression in p53-/- MEFs failed to inhibit cell proliferation despite inducing high ROS levels . These findings suggest that the functional status of p53 is a crucial determinant of how cancer cells respond to AKT3 activation, with important implications for therapeutic strategies targeting the AKT3-p53 axis.
MicroRNAs and other non-coding RNAs form a complex regulatory network controlling AKT3 expression and function in cancer:
This regulatory network represents a promising target for therapeutic intervention. Experimental approaches to study these interactions include luciferase reporter assays to confirm direct binding between miRNAs and AKT3 mRNA, RNA immunoprecipitation to verify interactions between circRNAs and miRNAs, and rescue experiments where re-expression of AKT3 reverses phenotypes caused by microRNA overexpression. These complex interactions highlight the importance of considering the entire non-coding RNA landscape when targeting AKT3 in cancer .
Selecting appropriate experimental models is crucial for accurately studying AKT3 function in human pathologies:
Cell line models:
Isogenic cell lines expressing different AKT isoforms provide clean systems for comparing isoform-specific functions
Cancer cell lines with AKT3 amplification (e.g., MDA-MB-231 for breast cancer, T98G for glioblastoma) represent relevant disease contexts
CRISPR-engineered cell lines with endogenous tagging of AKT3 maintain physiological expression levels while enabling tracking
Animal models:
Tissue-specific conditional AKT3 knockout mice avoid developmental effects of constitutive deletion
Patient-derived xenografts maintain tumor heterogeneity and microenvironment interactions
Genetically engineered mouse models with tissue-specific AKT3 overexpression or mutation
Three-dimensional models:
Organoids derived from patient samples preserve tissue architecture and cellular heterogeneity
Spheroid co-culture systems incorporating immune and stromal components
Validation approaches:
Complementary use of pharmacological inhibition and genetic manipulation
Rescue experiments to confirm specificity (e.g., re-expression of wild-type or mutant AKT3)
Correlation of findings with clinical samples and patient outcomes
Accurately measuring AKT3-specific activity in complex biological samples requires sophisticated methodological approaches:
Phosphorylation-state analysis:
Phospho-specific antibodies targeting AKT3's Ser472 (equivalent to Ser473 in AKT1)
Mass spectrometry-based phosphoproteomics to detect isoform-specific phosphopeptides
Proximity ligation assays to visualize AKT3 interactions with specific substrates in situ
Substrate-focused approaches:
Genetic manipulation controls:
Parallel analysis of samples with AKT3 knockdown/knockout as specificity controls
Rescue experiments with catalytically active versus inactive AKT3
Comparison with pan-AKT inhibition to identify isoform-specific effects
Downstream readouts:
Single-cell approaches:
Single-cell phospho-flow cytometry to measure AKT3 activation in specific cell populations
Single-cell RNA-seq to correlate AKT3 expression with pathway activity signatures
Imaging-based approaches using biosensors for spatiotemporal activity measurement
Researchers should employ multiple complementary methods and include appropriate controls to ensure measurements reflect genuine AKT3-specific activity rather than general AKT pathway activation.
AKT3 contributes to therapeutic resistance through multiple mechanisms across different cancer contexts:
Endocrine therapy resistance in breast cancer:
Radiation resistance:
Chemotherapy resistance:
In triple-negative breast cancer, AKT3 decreases sensitivity to the pan-Akt inhibitor GSK690693
Circular RNAs interacting with AKT3 signaling (e.g., hsa_circ_0000199) promote chemoresistance in multiple cancer types
In gastric cancer, hsa_circ_0000199 enhances DNA damage repair and cell survival by modulating the miR-198/AKT3 axis
Immunosuppressive effects:
Experimentally, these resistance mechanisms can be studied using resistant cell line models, acquired resistance protocols where sensitive cells are exposed to escalating drug concentrations, and combination treatment strategies targeting both AKT3 and the primary therapy. Clinical correlation studies examining AKT3 expression or activation in resistant versus responsive patient samples provide valuable translational insights .
Developing AKT3-specific therapeutic strategies presents unique challenges but offers potential precision medicine opportunities:
Small molecule inhibitor approaches:
Structure-guided design targeting non-conserved regions of AKT3
Allosteric inhibitors that exploit conformational differences between AKT isoforms
Covalent inhibitors targeting AKT3-specific cysteine residues
RNA-based therapeutics:
Substrate-directed approaches:
Context-dependent combination strategies:
Biomarker-guided patient selection:
These approaches require rigorous validation in preclinical models with careful consideration of potential toxicities in tissues where AKT3 has essential functions. Therapeutic window assessment is crucial given AKT3's roles in normal physiology alongside its disease-promoting functions.
The unique relationship between AKT3 and oxidative stress presents novel therapeutic opportunities:
Synthetic lethality approaches:
AKT3-high cancer cells already have elevated ROS levels, making them potentially vulnerable to further oxidative stress
Combining AKT3 inhibitors with pro-oxidant therapies could push cancer cells beyond their adaptive capacity
Targeting antioxidant defense mechanisms (e.g., glutathione synthesis inhibitors) may be selectively toxic to AKT3-high cells
DNA damage response targeting:
p53 status-dependent strategies:
NADPH oxidase modulation:
Experimental validation of these approaches requires careful assessment of redox status before and after treatment, measurement of DNA damage markers, and evaluation of cell death mechanisms in both cancer and normal cells to establish therapeutic windows.
Predicting patient response to AKT3-targeted therapies requires comprehensive biomarker strategies:
Genomic biomarkers:
Expression biomarkers:
Activation biomarkers:
Pathway context assessment:
Functional testing:
Ex vivo drug sensitivity testing of patient-derived cells
Organoid-based drug response profiling
Pathway activation analysis in circulating tumor cells
A multiparameter approach combining these biomarkers will likely provide the most accurate prediction of response. For example, a patient with AKT3 amplification, wild-type p53, high ROS levels, and intact DNA damage response might be an ideal candidate for AKT3 inhibition, while different biomarker patterns might suggest alternative targeting strategies or combinations .
The AKT3 gene is located on chromosome 1q43-q44 in humans . The protein encoded by this gene is one of three closely related AKT isoforms, the others being AKT1 and AKT2 . AKT3 is the least studied among the AKT isoforms but is known to be essential for brain development and the viability of malignant glioma cells .
AKT3 is involved in regulating cell signaling in response to insulin and growth factors such as platelet-derived growth factor (PDGF) and insulin-like growth factor 1 (IGF1) . It mediates its effects through serine and/or threonine phosphorylation of a range of downstream substrates . This kinase has been shown to play a role in various biological processes, including:
AKT3 has been implicated in several critical biological processes: