AKT2 is a protein-coding gene located on chromosome 19q13.2 in humans that encodes the serine/threonine-protein kinase AKT2, also known as protein kinase B beta (PKBβ) or RAC-beta serine/threonine-protein kinase, a key member of the AGC family of kinases and one of three AKT isoforms (alongside AKT1 and AKT3).[1][2] The AKT2 protein, consisting of 481 amino acids and approximately 56 kDa, features a pleckstrin homology (PH) domain, a kinase domain, and a regulatory domain, enabling its activation via phosphorylation downstream of phosphoinositide 3-kinase (PI3K) signaling in response to growth factors like insulin.[2][3] It plays a central role in mediating insulin's metabolic effects by regulating glucose uptake through the translocation of the GLUT4glucose transporter to the cell membrane, thereby influencing glycogen synthesis, protein synthesis, and lipid metabolism.[1][4]In cellular physiology, AKT2 promotes cell survival by inhibiting pro-apoptotic proteins such as BAD and FOXO transcription factors, while also facilitating cell proliferation, migration, and adhesion through pathways involving NF-κB and CREB1 activation.[1][4] Unlike AKT1, which is more associated with cell survival and growth, AKT2 is particularly implicated in metabolic homeostasis, including skeletal muscle development and hepatic insulin signaling via inhibition of PGC-1α.[1] Dysregulation of AKT2, often through amplification or overexpression, contributes to oncogenesis; it is amplified in approximately 10-20% of pancreatic and ovarian cancers and overexpressed in a subset of breast cancers, driving tumor progression, invasion, and metastasis by upregulating β1 integrins and enhancing glycolysis.[2][5]Genetically, mutations in AKT2 are linked to metabolic disorders, including autosomal dominant type 2 diabetes mellitus due to variants like R274H that impair insulin signaling and glucose homeostasis, and hypoinsulinemic hypoglycemia with hemihypertrophy caused by activating mutations such as E17K, leading to excessive insulin-independent glucose uptake.[2][6][7] As a therapeutic target, AKT2 inhibitors are under investigation for cancers with PI3K/AKT pathway hyperactivation, highlighting its dual role in metabolism and malignancy.[5]
Discovery and Nomenclature
Gene Identification
The AKT2 gene was first identified in 1987 as one of two human homologues of the v-akt oncogene derived from the AKT8 retrovirus, which induces murine T-cell lymphomas.[8] Researchers cloned AKT1 and AKT2 by screening a human genomic DNA library with a probe specific to the v-akt sequence under low-stringency hybridization conditions, revealing high conservation between the viral oncogene and its cellular counterparts. This discovery established AKT2 as part of the AKT family of genes, alongside AKT1, with early evidence of potential oncogenic roles through amplification of AKT1 in a primary human gastric adenocarcinoma.[8]In 1992, the full-length cDNA of AKT2 was cloned from a humanthymuscDNA library, providing the complete coding sequence for a 481-amino-acid protein.[9]Sequence analysis confirmed its identity as a serine/threoninekinase, with the open reading frame spanning 1,443 base pairs, and in vitro translation yielding a 56-kDa protein product.[9]Fluorescence in situ hybridization mapped the AKT2 gene to the chromosomal region 19q13.1-q13.2.[9]The official gene symbol AKT2 was assigned by the HUGO Gene Nomenclature Committee, with common aliases including PKBB and RAC-BETA.[10] Initial characterization highlighted AKT2's homology to the protein kinase C (PKC) family, particularly in its catalytic domain, which shares similarity with PKC isoforms, suggesting shared functional motifs in kinase activity and substrate phosphorylation.[9]
Isoform Distinctions
AKT2 encodes a serine/threonine kinase that shares approximately 81% amino acid sequence identity with AKT1 and about 78% with AKT3, reflecting their common evolutionary origin within the AGC kinase family while allowing for functional specialization.[11][12] These isoforms exhibit high homology in their overall domain architecture, including the pleckstrin homology (PH), kinase, and regulatory domains, but diverge in specific regions that influence substrate specificity and regulatory interactions. For instance, in the kinase domain, subtle amino acid variations, such as the conserved arginine at position 274 in AKT2 (corresponding to arginine 273 in AKT1), contribute to differences in activation dynamics and resistance to dephosphorylation, with mutations at this residue (e.g., R274H) known to impair AKT2 activity in insulin signaling contexts.[13]In terms of expression patterns, AKT2 displays a distinct tissue bias compared to its isoforms, with predominant expression in insulin-responsive tissues such as skeletal muscle, liver, and adipose tissue, where it plays a key role in glucose homeostasis.[12] In contrast, AKT1 is more ubiquitously expressed but present in neuronal tissues like the brain, supporting cell survival and growth processes, while AKT3 shows restricted expression primarily in the brain, testis, and kidney.[14] This differential distribution underscores non-overlapping roles, as evidenced by isoform-specific responses to stimuli like insulin, which preferentially activate AKT2 in metabolic tissues over AKT1 or AKT3.[15]The AKT2 gene is highly conserved across mammalian species, with orthologs in humans, mice, and other mammals sharing over 95% amino acid identity in the protein-coding regions, indicating strong selective pressure for its metabolic functions.[16] In humans, the AKT2 gene spans 14 exons on chromosome 19q13.2, similar to the 14-exon structure of AKT1 on chromosome 14q32.3, though subtle differences exist in exon lengths and intron sizes that may influence alternative splicing patterns unique to AKT2, such as rare variants lacking the hydrophobic motif.[17][18]Non-redundant functions of AKT2 are clearly demonstrated by isoform-specific knockout studies in mice. AKT2-null mice develop severe insulin resistance, hyperglycemia, glucose intolerance, and age-dependent lipoatrophy, phenotypes absent in AKT1-null mice, which instead exhibit only mild growth retardation and no metabolic dysregulation.[19][20] Double AKT1/AKT2 knockouts amplify these effects, resulting in perinatal lethality and profound developmental defects, further highlighting AKT2's specialized role in insulin-mediated signaling distinct from AKT1's contributions to cellular proliferation.[21]
Gene and Expression
Genomic Location
The AKT2 gene is located on the long (q) arm of human chromosome 19 at cytogenetic band 19q13.2. In the GRCh38.p14 reference genome assembly, it spans genomic coordinates 19:40,230,317-40,285,345 (strand: minus), encompassing approximately 55 kb of DNA sequence and comprising 14 exons that encode the serine/threonine kinase AKT2 protein.[1]The promoter region of AKT2, situated upstream of the transcription start site, contains regulatory elements that influence gene transcription, including multiple binding sites for transcription factors. Analysis of the AKT2 promoter has identified nine putative E-box motifs (CANNTG sequences), which serve as binding sites for basic helix-loop-helix transcription factors such as MyoD, facilitating muscle-specific regulation. Additionally, variants in the 5' regulatory region, such as single nucleotide polymorphisms, have been associated with modulation of susceptibility to insulin resistance and related metabolic traits in human populations, though these do not commonly alter basal expression levels.[22][23]Intronic sequences within AKT2 harbor variants that may contribute to alternative splicing or regulatory functions, including a novel splice variant (AKT2-13a) that skips the final exon, resulting in a protein isoform lacking the hydrophobic motif essential for activation. The 3' untranslated region (UTR) of AKT2 transcripts includes conserved binding sites for several microRNAs that post-transcriptionally repress expression; notable examples include miR-149-3p, which targets a site in the 3' UTR to inhibit proliferation in oral squamous cell carcinoma cells, miR-296, which suppresses AKT2 in pancreatic cancer, and miR-200c, which regulates chemosensitivity in non-small cell lung cancer. These miRNA interactions highlight the role of the 3' UTR in fine-tuning AKT2 levels across cellular contexts.[24][25][26][27]Copy number variations (CNVs) involving AKT2 have been reported in specific populations and disease states, such as amplification observed in 12-27% of epithelial ovarian cancers, potentially driving oncogenesis through increased AKT2 dosage. However, in general human populations, no common CNVs or polymorphisms significantly impact basal AKT2 expression, and the gene shows no evidence of haploinsufficiency or triplosensitivity based on current assessments.[28][1]
Expression Patterns
AKT2 displays high basal expression at the mRNA and protein levels in insulin-sensitive tissues, including skeletal muscle, liver, adipose tissue, and pancreas. Data from the Genotype-Tissue Expression (GTEx) project and the Human Protein Atlas indicate elevated mRNA transcripts per million (TPM) in skeletal muscle (high expression category) and moderate levels in liver and adipose tissue, reflecting its role in glucose homeostasis in these sites. In the pancreas, overall tissue expression is lower, but AKT2 is expressed in beta cells, where it supports insulin secretion and cell survival.[29][30][31]Expression of AKT2 is dynamically regulated by growth factors such as insulin and insulin-like growth factor 1 (IGF-1), which activate the PI3K pathway to enhance AKT2 phosphorylation and activity, particularly in metabolic tissues. This stimulation occurs rapidly in response to nutrient signals, promoting downstream effects on glucose uptake without necessarily altering steady-state mRNA levels. Additionally, AKT phosphorylates CLOCK, influencing circadian gene expression in insulin-responsive tissues like skeletal muscle and liver, aligning with daily metabolic demands.[32]AKT2 is expressed during pancreatic development, contributing to beta cell maturation, and remains prominent in adult beta cells to maintain endocrine function. In pathological contexts, such as obesity, AKT2 activation is often impaired in adipose and muscle tissues, contributing to insulin resistance, as shown by reduced phosphorylation in human samples via Western blot analyses, though total levels may vary.[31][33]
Protein Structure
Domain Architecture
The AKT2 protein, encoded by the human AKT2 gene, is a 481-amino acid serine/threonine kinase with a calculated molecular weight of approximately 55 kDa.[3] Its domain architecture follows the conserved organization typical of AGC kinases, comprising an N-terminal pleckstrin homology (PH) domain, a central catalytic kinase domain, and a C-terminal regulatory domain. This modular structure enables AKT2 to integrate lipid signaling with kinase activity and regulatory phosphorylation. The PH domain spans residues 1–106 and serves as the primary site for binding phosphoinositides, such as phosphatidylinositol (3,4,5)-trisphosphate (PIP3), facilitating membrane recruitment in response to upstream signals like PI3K activation.[3][34]The core kinase domain, encompassing residues 144–427, houses the ATP-binding pocket and the activation loop, which includes the critical threonine residue Thr309 targeted for phosphorylation by PDK1 to initiate catalytic competence.[35] This domain adopts a bilobal fold characteristic of eukaryotic protein kinases, with an N-lobe for nucleotide binding and a C-lobe for substraterecognition, but in its inactive state, structural elements such as a disordered α-helix C and steric hindrance in the activation loop obstruct ATP and substrate access.00937-1) The linker region connecting the PH and kinase domains (approximately residues 107–143) exhibits significant flexibility, as evidenced by crystallographic studies, allowing the PH domain to swing away from the kinase core upon lipid binding to relieve autoinhibition.00550-7)The short C-terminal regulatory domain (residues 428–481) contains a hydrophobic motif (F/Y)XDF(Y/F), which is phosphorylated at Ser474 by mTORC2 (functioning as PDK2) to fully activate the kinase by stabilizing the activation loop conformation.[36] This motif promotes intramolecular interactions that enhance kinase activity and substrate access. Crystal structures, including PDB entry 1O6K of the activated AKT2 kinase domain in complex with a GSK3β peptidesubstrate and AMP-PNP, highlight how phosphorylation at Ser474 induces closure of the activation loop, enabling productive ATP orientation and substrate binding.00550-7) Overall, AKT2 lacks isoform-specific structural folds unique from AKT1 and AKT3, sharing over 80% sequence identity and identical domain organization across the family, which underlies their functional redundancy in many contexts.[37]
Post-Translational Modifications
K63-linked polyubiquitination at Lys8 and Lys14 within the PH domain, mediated by TRAF6, promotes membrane recruitment and activation of AKT2.[38] Additionally, K48-linked polyubiquitination at Lys284, facilitated by the E3 ligase MULAN, particularly in response to phosphorylated AKT2, targets it for proteasomal degradation, contributing to the regulation of its protein levels in cellular homeostasis.[39][40]Similar to AKT1, acetylation likely occurs on conserved Lys14 and Lys20 in the PH domain of AKT2, mediated by histone acetyltransferases like EP300 and KAT2B, which reduces the affinity of the PH domain for phosphatidylinositol lipids and thereby inhibits AKT2 activity by impairing membrane localization. This modification is reversed by deacetylases such as SIRT1, allowing for dynamic control of AKT2 function.[41] Additionally, SUMOylation at Lys276 (and Lys301) modulates nuclear import of AKT2; this process, catalyzed by PIAS family E3 ligases and reversed by SENP1, influences AKT2's subcellular distribution and its role in nuclear events like alternative splicing and cell cycle regulation.[42]O-GlcNAc modifications occur at Thr306 and Thr313 in the kinase domain of AKT2, inhibiting its activation by reducing phosphorylation at Thr309 and disrupting interaction with PDK1. These modifications, part of the broader O-GlcNAcylation pathway, help regulate AKT2 in response to nutrient conditions without isoform-specific variations unique to AKT2 compared to AKT1 or AKT3. While many PTMs are conserved across AKT isoforms, AKT2-specific modifications, such as enhanced O-GlcNAc sensitivity in metabolic contexts, contribute to its distinct roles in glucose homeostasis.[5] The half-life of AKT2 is stabilized by binding to the chaperone HSP90, protecting it against ubiquitination and proteasomal degradation, thereby maintaining steady-state levels during basal conditions.02644-3/fulltext)[43]
Activation and Regulation
Upstream Signaling
The activation of AKT2 is primarily initiated through receptor tyrosine kinases (RTKs), such as the insulin receptor, which upon ligand binding undergo autophosphorylation and recruit adapter proteins like the insulin receptor substrates (IRS-1 and IRS-2).[44] These adapters activate class I phosphoinositide 3-kinase (PI3K) by relieving its autoinhibitory conformation, leading to the phosphorylation of phosphatidylinositol-4,5-bisphosphate (PIP2) to generate phosphatidylinositol-3,4,5-trisphosphate (PIP3) at the plasmamembrane.[45] The pleckstrin homology (PH) domain of AKT2 specifically binds PIP3 (and to a lesser extent PIP2), facilitating rapid recruitment and translocation of AKT2 from the cytosol to the plasmamembrane, a process that occurs within 30-60 seconds following insulin stimulation in insulin-responsive cells like adipocytes.[46][47]This membrane localization induces a conformational change in AKT2, relieving autoinhibition and exposing key phosphorylation sites in the activation loop (Thr309) and hydrophobic motif (Ser474).[48] Thr309 is phosphorylated by 3-phosphoinositide-dependent protein kinase 1 (PDK1), which is also recruited to PIP3 via its PH domain, while Ser474 is phosphorylated by the mTORC2 complex, completing full AKT2 activation.[49] The affinity of the AKT2 PH domain for PIP3, characterized by an activation constant (Km) of approximately 1-10 μM, underscores the efficiency of this lipid-mediated recruitment under physiological PIP3 concentrations generated by PI3K.[50]AKT2 activation also establishes a negative feedback loop to fine-tune upstream signaling: activated AKT2 phosphorylates IRS-1 at inhibitory serine residues, such as Ser302 (mouse) or the equivalent Ser307 (human), which disrupts IRS-1's interaction with the insulin receptor and reduces further PI3K recruitment to the plasma membrane.[47] This phosphorylation event, observed in insulin-stimulated adipocytes and hepatocytes, limits excessive PIP3 production and prevents sustained hyperactivation of the pathway.[51]
Inhibitory Mechanisms
AKT2 activity is tightly regulated by several inhibitory mechanisms that prevent unwarranted signaling. One primary mode of inhibition involves dephosphorylation at key activation sites. The phosphatases PHLPP1 and PHLPP2 specifically target the hydrophobic motif phosphorylation at Ser474, with PHLPP1 preferentially dephosphorylating AKT2 and AKT3 isoforms, thereby attenuating their activity and promoting apoptosis in cancer cells.[52] Additionally, protein phosphatase 2A (PP2A) dephosphorylates AKT2 at Thr309 in the activation loop, reversing PDK1-mediated activation and terminating downstream signaling.[53] PTEN further inhibits AKT2 by hydrolyzing phosphatidylinositol (3,4,5)-trisphosphate (PIP3) to PIP2, reducing membrane recruitment and preventing full activation of the kinase.[54]In its inactive state, AKT2 maintains autoinhibition through an intramolecular interaction between its pleckstrin homology (PH) domain and the kinase domain, forming a "clasp" that blocks substrate access and ATP binding. This autoinhibitory conformation is relieved only upon binding to PIP3 at the plasma membrane, which disrupts the PH-kinase interaction and allows conformational opening for phosphorylation.[55][48]Post-activation, AKT2 undergoes proteasomal degradation to limit sustained signaling. The E3 ubiquitin ligase TTC3 binds to phosphorylated AKT2, facilitating its ubiquitination and subsequent proteasomal breakdown, particularly in the nucleus following activation.[56] Isoform-specific differences in regulation are evident, as AKT2 exhibits heightened sensitivity to dephosphorylation by PHLPP1 compared to AKT1, contributing to distinct roles in cellular processes such as insulin signaling.[52]Pharmacological inhibition of AKT2 has been achieved through allosteric modulators targeting the PH domain. For instance, MK-2206 binds selectively to the PH domain of AKT2, stabilizing the autoinhibited conformation and inhibiting kinase activity with an IC50 of approximately 12 nM.[57] This approach disrupts lipid-induced activation without competing at the ATP-binding site, offering isoform-nonselective but potent suppression across AKT family members.[58]
Biological Functions
Metabolic Roles
AKT2 serves as a key mediator in insulin-stimulated glucose uptake, primarily through its phosphorylation of the Rab GTPase-activating protein AS160 (also known as TBC1D4) at threonine 642. This phosphorylation inhibits AS160's GAP activity toward Rab proteins, facilitating the translocation of GLUT4 glucose transporters to the plasma membrane in skeletal muscle and adipose tissue, thereby enhancing glucose disposal during the postprandial state.[59][60]In carbohydrate and lipid metabolism, AKT2 phosphorylates glycogen synthase kinase 3β (GSK3β) at serine 9, inhibiting its activity and thereby relieving repression of glycogen synthase to promote insulin-dependent glycogen synthesis in liver and muscle.[61] AKT2 also drives de novo lipogenesis by activating the transcription factor SREBP1c in hepatocytes, which induces expression of lipogenic enzymes such as fatty acid synthase, contributing to triglyceride accumulation in response to nutrient excess.[62][63]AKT2 further regulates hepatic glucose production by phosphorylating the transcription factor FOXO1 at threonine 24, which promotes its binding to 14-3-3 proteins, nuclear exclusion, and subsequent downregulation of gluconeogenic genes like G6PC and PCK1, thereby suppressing gluconeogenesis under fed conditions.[64] This isoform-specific role is underscored by studies in knockout mice, where AKT2-null animals display postprandial hyperglycemia and age-dependent reduction in fat mass due to defective insulin-mediated metabolic responses in peripheral tissues, in contrast to AKT1-null mice, which maintain normal glucose homeostasis despite growth impairments.[19][65]
Cellular Processes
Members of the AKT family, including AKT2, support cellular growth by phosphorylating tuberous sclerosis complex 2 (TSC2) at threonine 1462 (Thr1462), which inactivates the TSC1-TSC2 complex and relieves inhibition of the mTORC1 pathway, thereby promoting ribosomal biogenesis and protein synthesis.[66] This phosphorylation event integrates upstream signals to enhance translation of growth-related proteins via downstream effectors like S6K1 and 4E-BP1. The resulting increase in protein synthesis drives anabolic processes essential for cell hypertrophy and proliferation, particularly in metabolic contexts such as insulin signaling.[66][67]AKT2 uniquely enhances cell migration, particularly in breast cancer cells, through isoform-specific upregulation of β1 integrins, which strengthens focal adhesion formation and extracellular matrix interactions to facilitate invasive motility.[68] Overexpression of AKT2 in these cells leads to elevated β1 integrin expression at the protein and mRNA levels, correlating with increased adhesion to collagen IV and heightened migratory potential without altering other integrin subtypes.[68] This mechanism distinguishes AKT2 from AKT1, as AKT2-driven integrin modulation promotes directional migration and tissue invasion.[69]Recent studies (as of 2024) have also identified AKT2's role in lysosomal biogenesis through the AKT2/SIRT5/TFEB pathway, where AKT2 regulates lysosomal function and autophagy, impacting cellular homeostasis and disease processes. Additionally, AKT2 modulates antiviral responses by phosphorylating IRF3 to reduce IFNβ1 production, thereby influencing innate immunity.[70][71]
Clinical Significance
Role in Cancer
AKT2 plays a significant oncogenic role in various malignancies through genomic amplification and overexpression, which occur in 12–20% of ovarian, pancreatic, and breast cancers.[5] In pancreatic cancer, AKT2 amplification is detected in approximately 10% of cases, contributing to tumor progression.[72] Overexpression of AKT2 in breast and ovarian cancers is associated with enhanced invasion, metastasis, and poor prognosis, including reduced survival rates.[73][74]Somatic mutations in AKT2 further drive oncogenesis, particularly in endometrial cancer where they are identified in about 12% of cases, often in the kinase domain such as R368C or D399N.[75] These mutations are considered oncogenic and likely enhance kinase activity, promoting aberrant signaling in PTEN-mutated backgrounds common to endometrial tumors.[75]AKT2 facilitates cancer metastasis by inducing epithelial-mesenchymal transition (EMT), a process essential for invasion and dissemination in breast and ovarian cancers; downregulation of AKT2 suppresses EMT-like morphological changes and reduces metastatic potential.[76] Additionally, AKT2 activation confers chemotherapy resistance through downstream NF-κB signaling, as seen in pancreatic cancer where AKT2 inhibition sensitizes cells to gemcitabine by suppressing NF-κB-mediated survival pathways.[77]Therapeutic targeting of AKT2 exploits these dependencies, with the pan-AKT inhibitor capivasertib, approved by the FDA in 2023, demonstrated efficacy in the phase III CAPItello-291 trial for hormone receptor-positive breast cancer with AKT pathway alterations. In the CAPItello-291 trial, capivasertib combined with fulvestrant achieved an objective response rate of 22.9% in the overall population (29.4% in patients with AKT pathway alterations), compared to 12.2% (8.3% in altered) with placebo plus fulvestrant.[78][79] As of 2025, AKT inhibitors like capivasertib are integrated into treatment guidelines for specific breast cancer subtypes, with emerging data on their use in endometrial and ovarian cancers.[80]
Metabolic Disorders
AKT2 knockout mice exhibit partial insulin resistance characterized by impaired insulin-stimulated glucose uptake in skeletal muscle, with reductions in uptake efficiency observed under insulin stimulation conditions. These mice display hyperglycemia, hyperinsulinemia, and glucose intolerance, highlighting AKT2's critical role in insulin-mediated metabolic responses without complete abolition of glucose handling. In humans, rare loss-of-function variants in AKT2, such as the Finnish-enriched P50T allele, are associated with elevated fasting insulin levels, decreased insulin sensitivity, and increased risk of type 2 diabetes (odds ratio 1.05 per allele).[81] Additionally, the activating E17K variant is linked to hypoinsulinemic hypoglycemia, as it promotes excessive insulin-independent glucose uptake, underscoring AKT2's isoform-specific contributions to glucose homeostasis dysregulation.[82]In contrast, heterozygous loss-of-function mutations in AKT2 are established causes of familial partial lipodystrophy, where impaired adipogenesis leads to fat loss, hypertriglyceridemia, and severe insulin resistance, further emphasizing AKT2's dose-dependent role in adipose function.[83]AKT2 polymorphisms have been associated with polycystic ovary syndrome (PCOS) through dysregulation of hyperandrogenism signaling pathways, where variants influence insulin sensitivity and ovarian steroidogenesis.[84] For instance, specific AKT2 single nucleotide polymorphisms correlate with increased PCOS risk by modulating insulin signaling components that exacerbate androgen excess and ovulatory dysfunction.[84] Recent genome-wide association studies (GWAS) in metabolic syndrome cohorts have identified AKT2 SNPs linked to insulin resistance traits, reinforcing its genetic contributions to broader endocrine-metabolic disorders beyond isolated diabetes.[85]Therapeutic modulation of AKT2 activity holds promise for metabolic disorders, with metformin indirectly suppressing AKT2 signaling through activation of AMP-activated protein kinase (AMPK), which counters AKT2-mediated insulin resistance in adipose and hepatic tissues.[86] This AMPK-dependent mechanism enhances glucose uptake and reduces lipogenesis, providing a basis for metformin's efficacy in type 2 diabetes and related conditions without direct AKT2 targeting.[86]
Interactions
Key Protein Partners
AKT2 engages with key protein partners primarily through its pleckstrin homology (PH) domain and kinase domain, facilitating substrate recognition, membrane recruitment, and signal modulation. Among its direct substrates, insulin receptor substrate 1 (IRS-1) interacts with AKT2, with docking mediated by the PTB domain of IRS-1, enabling phosphorylation at multiple serine residues that regulate insulin signaling feedback.[87] Similarly, forkhead box O1 (FOXO1), a transcription factor, binds AKT2 directly via its forkhead domain, allowing AKT2-mediated phosphorylation at sites such as Thr24, Ser256, and Ser319, which promotes FOXO1 nuclear exclusion and inactivation.[88]Scaffold proteins further fine-tune AKT2 function. TCL1 binds the PH domain of AKT2, stabilizing its inactive conformation and enhancing kinase activity upon activation by preventing premature membrane dissociation. APPL1 serves as an adaptor protein that interacts with AKT2 to mediate endosomal signaling, tethering AKT2 to endocytic compartments for localized activation in response to growth factors like insulin.The PH domain of AKT2 exhibits a binding affinity for phosphatidylinositol 3,4,5-trisphosphate (PIP3) with a dissociation constant (Kd) of approximately 250 nM, which is crucial for its recruitment to the plasma membrane.[89] While AKT2 shares most binding partners with AKT1, it demonstrates differences in activation kinetics with phosphoinositide-dependent kinase 1 (PDK1).These interactions have been validated through methods such as yeast two-hybrid screening and co-immunoprecipitation in studies spanning the 1990s to the 2020s, including early identification of TCL1-AKT2 binding via co-IP and more recent Y2H screens confirming APPL1 associations.
Protein Partner
Binding Site on AKT2
Role in AKT2 Function
Validation Method
Reference
IRS-1
Kinase domain (substrate docking via IRS-1 PTB)
Direct phosphorylation substrate for feedback inhibition
Co-IP, phosphorylation assays
[87]
FOXO1
Kinase domain (via FOXO1 forkhead domain)
Direct substrate for nuclear export regulation
Co-IP, mutagenesis studies
[88]
TCL1
PH domain
Scaffold stabilizing inactive form and enhancing activity
AKT2 participates in the PI3K-AKT2 signaling axis localized to endosomal compartments, where it integrates insulin signaling to regulate metabolic processes. Specifically, the class II PI3K isoform PI3K-C2γ acts as a Rab5 effector on early endosomes, selectively activating AKT2 in response to insulin stimulation, independent of class I PI3K activity.[90] This endosomal AKT2 activation promotes downstream signaling, facilitating responses such as glycogensynthesis in insulin-responsive tissues like liver and muscle.[90] Disruption of this axis impairs hepatic AKT2 phosphorylation and glucose homeostasis, highlighting its role in compartmentalized signal propagation.[90]In stress responses, AKT2 negatively regulates the assembly of the POSH-MLK-JNK multiprotein complex, modulating JNK-mediated apoptosis and inflammation. POSH serves as a scaffold that recruits mixed-lineage kinases (MLKs), MAP kinase kinases (MKKs), and JNKs to form an active signaling module upon cellular stress. AKT2 binds this complex and phosphorylates MLK3 at inhibitory sites, preventing full JNK activation and thereby attenuating pro-apoptotic signals.[91] This regulation is evident in neuronal and epithelial cells, where reduced AKT2 activity enhances complex assembly and JNK-dependent stress responses.Nuclear translocation of dephosphorylated AKT2 influences transcription repression through complexes involving FOXO transcription factors. Inhibition of AKT2 activity allows FOXO3a to accumulate in the nucleus and exert repressive effects on estrogen receptor alpha (ERα)-driven transcription in breast cancer cells, forming co-repressor assemblies that suppress proliferative gene expression.[92] AKT2-mediated phosphorylation normally sequesters FOXO factors in the cytoplasm via 14-3-3 binding, but upon AKT2 inhibition, nuclear FOXO3a interacts with co-repressors to downregulate target genes, linking AKT2 to anti-proliferative transcriptional control.[93]At the plasma membrane, AKT2 localizes to lipid rafts in coordination with integrins to facilitate cell migration. Raft-associated AKT signaling is activated by chemokine receptors like CXCR4, promoting actin reorganization and directed motility in platelets and immune cells.[94] AKT phosphorylates effectors such as ACAP1, regulating the recycling of β1-integrins from endosomes back to rafts, which sustains focal adhesion turnover essential for migratory persistence.[95] This raft-integrin-AKT axis is critical for processes like wound healing and immune surveillance, with disruptions impairing directional migration.[96]The dynamic assembly and disassembly of AKT2-containing complexes are governed by feedback loops and phosphatase activity. Post-activation, AKT2 integrates into an mTORC2 feedback mechanism by phosphorylating the SIN1 subunit at Thr86, enhancing mTORC2 kinase activity and sustaining AKT2 phosphorylation at Ser473 for prolonged signaling.[97] Complex disassembly occurs via dephosphorylation by phosphatases like PHLPP1, which targets AKT2 at Ser473, and PP2A, which removes phosphates from multiple sites, terminating signaling and preventing aberrant activation.[98] This regulation ensures temporal control, with PHLPP1 inhibiting pancreatic cancer progression by dephosphorylating AKT2 at Ser473.[98]Structural studies using cryo-electron microscopy and other methods have provided insights into PIP3-mediated activation of AKT kinases, including interactions with PDK1 that induce conformational changes for Thr308 phosphorylation.[99] These analyses highlight PIP3 binding sites that stabilize the active conformation and explain activation in membrane microdomains.[100] Such insights underscore the role of PIP3 in coordinating multi-component assembly for precise signaling output.AKT2 also interacts with glycogen synthase kinase 3β (GSK3β), phosphorylating it to inhibit glycogen breakdown, and AS160 (TBC1D4), promoting GLUT4 translocation for glucose uptake—key to its metabolic functions.[101]