Abstract
The Ser/Thr protein kinase Akt regulates essential biological processes such as cell survival, growth, and metabolism. Upon growth factor stimulation, Akt is phosphorylated at Ser474; however, how this phosphorylation contributes to Akt activation remains controversial. Previous studies, which induced loss of Ser474 phosphorylation by ablating its upstream kinase mTORC2, have implicated Ser474 phosphorylation as a driver of Akt substrate specificity. Here we directly studied the role of Akt2 Ser474 phosphorylation in 3T3-L1 adipocytes by preventing Ser474 phosphorylation without perturbing mTORC2 activity. This was achieved by utilizing a chemical genetics approach, where ectopically expressed S474A Akt2 was engineered with a W80A mutation to confer resistance to the Akt inhibitor MK2206, and thus allow its activation independent of endogenous Akt. We found that insulin-stimulated phosphorylation of four bona fide Akt substrates (TSC2, PRAS40, FOXO1/3a, and AS160) was reduced by ∼50% in the absence of Ser474 phosphorylation. Accordingly, insulin-stimulated mTORC1 activation, protein synthesis, FOXO nuclear exclusion, GLUT4 translocation, and glucose uptake were attenuated upon loss of Ser474 phosphorylation. We propose a model where Ser474 phosphorylation is required for maximal Akt2 kinase activity in adipocytes.
Keywords: Akt PKB, serine/threonine protein kinase, phosphorylation, cell signaling, adipocyte, insulin, substrate specificity, mTOR complex (mTORC), protein synthesis, glucose transport, Akt Ser474 phosphorylation, Akt W80A, chemical genetics, GLUT4, MK2206
Introduction
The serine/threonine kinase Akt (protein kinase B) is a key regulatory node in a range of cell signaling networks that control essential biological processes such as cell survival, proliferation, and metabolism (1). Upon growth factor stimulation, there is an increase in plasma membrane phosphatidylinositol (3,4,5) triphosphate (PIP3)7 (2, 3). Akt then binds PIP3 via its pleckstrin homology (PH) domain and accumulates at the plasma membrane (4, 5). This interaction causes a conformational change, allowing PDK1 to phosphorylate Akt within its kinase domain at Thr309 (in Akt2; Thr308 in Akt1 and Thr305 in Akt3) and mTORC2 to phosphorylate Akt within its hydrophobic motif (HM) at Ser474 (in Akt2; Ser473 in Akt1 and Ser472 in Akt3) (6, 7). These phosphorylation events enhance Akt kinase activity, allowing Akt to phosphorylate a number of substrates and facilitate several biological processes. For example, phosphorylation of TSC2 and PRAS40 activates mTORC1 and protein synthesis (8–10), phosphorylation of AS160/TBC1D4 facilitates GLUT4 translocation and glucose uptake (11, 12), and phosphorylation of FOXO proteins regulates their transcriptional activity via nuclear exclusion (13, 14).
Although there is consensus that Akt Thr309 phosphorylation is required for its activation (6, 15–17), the role of Ser474 phosphorylation remains controversial. Initial studies that assessed Akt activity in vitro reported that loss of Ser474 phosphorylation severely diminished Akt kinase activity (6, 17). Accordingly, Akt crystal structures show that an active kinase conformation is only possible by mimicking Ser474 phosphorylation with a S474D substitution (18, 19). From this, a model arose whereby Ser474 phosphorylation was integral to a high level of Akt kinase activity.
However, the accuracy of this model has been disputed in more recent studies in cells, where Ser474 phosphorylation was required for Akt activity only toward specific substrates. These studies abolished Ser474 phosphorylation through genetic or pharmacological ablation of its upstream kinase, mTORC2. For example, in mouse embryonic fibroblasts lacking SIN1, one of the subunits of mTORC2, there is loss of FOXO1/3a phosphorylation, but other Akt substrates (TSC2 and GSK3) and mTORC1 effectors (S6K and 4EBP1) are unaffected (20). Similarly, phosphorylation of FOXO3a and AS160 is impaired, but GSK3β phosphorylation is unaltered, in adipocytes from mice lacking the mTORC2 subunit RICTOR (21). Further, rat adipocytes treated with small-molecule inhibitors of mTORC2 exhibit decreased phosphorylation of PRAS40 but not GSK3β (22). These studies have implicated Ser474 phosphorylation as a driver of Akt substrate specificity. Alternatively, a recent study using RICTOR knockout mice indicates that Ser474 phosphorylation is dispensable for maximal phosphorylation of AS160, PRAS40, FOXO1, and GSK3β in adipose tissue (23). Nevertheless, use of mTORC2 deletion or inhibition to study the role of Akt Ser474 phosphorylation may be confounded by several factors, including the induction of compensatory mechanisms because of a chronic absence of mTORC2 (24), lack of specificity of mTORC2 inhibitors (25), and interference from other downstream actions of mTORC2 that are independent of Akt Ser474 phosphorylation, such as other phosphorylation sites on Akt (26–28).
A chemical genetics approach originally designed by Green et al. (29) overcomes these disadvantages by facilitating acute prevention of Ser474 phosphorylation in cells without perturbing mTORC2 activity. An Akt phosphomutant can be engineered to contain a W80A mutation in its PH domain, which confers resistance to the allosteric pan-Akt inhibitor MK2206 without compromising its activation. Treatment of cells ectopically expressing this mutant with MK2206 acutely eliminates endogenous Akt activity, allowing specific analysis of the W80A Akt mutant (29–31). We utilized this system to address how loss of Ser474 phosphorylation influences Akt2 activity toward a number of substrates and insulin-regulated processes in 3T3-L1 adipocytes. We show that insulin-stimulated phosphorylation of four Akt substrates was reduced by ∼50% in the absence of Ser474 phosphorylation. Similarly, several insulin-regulated processes were partially impaired following loss of Ser474 phosphorylation. These findings support a model where Ser474 phosphorylation is essential for maximal Akt2 kinase activity in adipocytes.
Results
The Akt2 W80A mutation confers MK2206 resistance and is a powerful tool to study Akt2 mutants
Studying the activity of ectopically expressed mutant proteins in cells can be confounded by the presence of their endogenous counterparts. To circumvent this and examine the role of Akt2 Ser474 phosphorylation in 3T3-L1 adipocytes (hereafter referred to as adipocytes), we employed a chemical genetics approach originally designed by Green et al. (29). MK2206 is an Akt inhibitor that works by preventing Akt recruitment to the plasma membrane (PM), which is essential for its activation (5, 32, 33). However, Akt with the W80A mutation is resistant to MK2206 inhibition. Treatment of cells ectopically expressing W80A Akt with MK2206 prevents activation of endogenous Akt, allowing specific assessment of the W80A Akt mutant upon insulin stimulation (Fig. 1A) (29–31).
Figure 1.

The Akt2 W80A mutation confers MK2206 resistance and has negligible impact on Akt2 activation at the plasma membrane in 3T3-L1 adipocytes. A, the Akt W80A mutation confers resistance to the Akt inhibitor MK2206. Treatment of cells ectopically expressing W80A Akt with MK2206 prevents activation of endogenous Akt, allowing specific activation of the W80A Akt mutant upon insulin stimulation. IR, insulin receptor; P, phosphorylation site. B, 3T3-L1 adipocytes were electroporated with TagRFP-T-WT or W80A Akt2, and plasma membrane recruitment was assessed using live-cell TIRF microscopy. Adipocytes were exposed to 1 nm insulin. Representative images for 3–7 independent experiments are presented. C, quantification of B (WT, 41 cells from three independent experiments; W80A, 60 cells from seven independent experiments; mean ± S.E.). D, 3T3-L1 adipocytes were electroporated with TagRFP-T WT or W80A Akt2, and plasma membrane recruitment was assessed using live-cell TIRF microscopy. Adipocytes were exposed to 10 μm MK2206 for 5 min, followed by 1 nm insulin. Representative images for two independent experiments are presented. E, quantification of D (WT, 15 cells from two independent experiments; W80A, 59 cells from two independent experiments; mean ± S.E.).
To validate the W80A system, we assessed insulin-stimulated PM recruitment of TagRFP-T-WT and W80A Akt2 in adipocytes using live-cell total internal reflection fluorescence (TIRF) microscopy. There was no significant difference in the kinetics of insulin-stimulated WT and W80A Akt2 PM recruitment (Fig. 1, B and C). Aligning with previous studies (34, 35), both WT and W80A Akt2 exhibited an overshoot in PM localization in response to insulin. However, pretreatment with 10 μm MK2206 abolished PM accumulation of WT Akt2, whereas robust accumulation of W80A Akt2 was observed under identical conditions (Fig. 1, D and E). These data are consistent with the Akt2 W80A mutation conferring MK2206 resistance but having negligible impact on Akt2 recruitment to the PM.
We next generated a series of N-terminally FLAG-tagged Akt2 constructs that were engineered to express Akt2 with Thr309 or Ser474 mutated to alanine to prevent phosphorylation; WT Akt2 (WT), Akt2 with the W80A mutation (W80A), Akt2 with W80A and T309A mutations (W80A-T309A), and Akt2 with W80A and S474A mutations (W80A-S474A) (Fig. 2A). These were stably expressed in adipocytes to similar degrees at ∼75% of endogenous Akt2 (Fig. 2, B–D). Ectopic FLAG-Akt2 could be separated from endogenous Akt2 (Fig. S1). As expected, treatment of adipocytes expressing WT Akt2 with 10 μm MK2206 blocked insulin-stimulated phosphorylation of Akt at Thr309 and Ser474 and phosphorylation of its substrates TSC2, PRAS40, FOXO1/3a, and AS160 (Fig. 2, E and F). MK2206 did not completely block insulin-stimulated phosphorylation of the Akt substrate GSK3β at Ser9 (Fig. S2), suggesting GSK3β is also phosphorylated by another kinase at this site. Therefore, we did not use GSK3β phosphorylation as a readout of Akt activity in this study because we could not confidently attribute changes in GSK3β phosphorylation to Akt. In contrast, adipocytes expressing W80A Akt2 displayed robust insulin-stimulated phosphorylation of Akt and its substrates in the presence of 10 μm MK2206, again implicating W80A Akt2 as being MK2206-resistant (Fig. 2, E and F). Consistent with previous reports (6, 15–17), Thr309 phosphorylation was required for Akt2 activity because loss of phosphorylation at this site abolished insulin-stimulated phosphorylation of Akt substrates (Fig. 2, E and F). Intriguingly, loss of Thr309 phosphorylation resulted in hyperphosphorylation of Akt at Ser474 upon insulin stimulation, indicating interdependence between the two phosphorylation sites. These data highlight that the Akt2 W80A mutation confers resistance to MK2206 and that this can be used to analyze the activity of Akt2 mutants in the absence of endogenous Akt activity in adipocytes.
Figure 2.
W80A Akt2 is resistant to MK2206 and can be used to study Akt2 mutants in 3T3-L1 adipocytes independent of endogenous Akt. A, FLAG-tagged Akt2 constructs were generated: WT, W80A, W80A-T309A, and W80A-S474A Akt2. Each consisted of mutations in the PH domain, kinase domain (KD), and HM. B, lysates from 3T3-L1 adipocytes stably expressing empty vector (EV), WT, W80A, W80A-T309A, or W80A-S474A Akt2 were immunoblotted using antibodies as specified, with 14-3-3 as a loading control. A representative blot for three independent experiments is shown. C, quantification of FLAG expression in B (n = 3, mean ± S.E.). AU, arbitrary unit. D, quantification of Akt2 expression in B relative to the empty vector cell line, which represents endogenous Akt2 (n = 3, mean ± S.E.). E, 3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were treated with 10 μm MK2206 for 5 min followed by 1 nm insulin for 10 min. Lysates were immunoblotted with antibodies as specified, with 14-3-3 as a loading control. A representative blot for three independent experiments is presented. Band 1 and Band 2 indicate shifts in Akt2 gel migration. p, phosphorylated. F, quantification of E (n = 3, mean ± S.E.).
Akt2 Ser474 phosphorylation is required for maximal phosphorylation of TSC2, PRAS40, FOXO1/3a, and AS160
How Ser474 phosphorylation regulates Akt kinase activity remains controversial. MK2206-resistant W80A Akt was a valuable tool to assess the role of Ser474 phosphorylation in the activation of Akt2 in adipocytes through direct mutation of this phosphorylation site. We first assessed the role of Ser474 phosphorylation in Akt2 activity toward its canonical substrates TSC2, PRAS40, FOXO1/3a, and AS160. We did so by measuring the temporal dynamics by which these substrates were phosphorylated following insulin stimulation in adipocytes stably expressing W80A Akt2 or W80A-S474A Akt2. This was conducted in the presence of MK2206 to specifically activate the ectopic mutant. We have reported previously that a small degree of Akt phosphorylation is sufficient to achieve maximal substrate phosphorylation (36, 37); thus, we utilized a submaximal insulin dose (1 nm) to ensure that changes in Akt kinase activity were detectable.
In adipocytes expressing W80A Akt2, there was a time-dependent increase in phosphorylation of all substrates following insulin stimulation (Fig. 3, A and B). Distinct phosphorylation kinetics were observed between each substrate, aligning with previous reports of a lack of concordance in the behavior of Akt substrates (36, 38). Nevertheless, in adipocytes expressing W80A-S474A Akt2, substrate phosphorylation plateaued at ∼50% of that observed in adipocytes expressing W80A Akt2 (Fig. 3, A and B). These data suggest that Ser474 phosphorylation plays an important role in maximizing Akt2 kinase activity toward TSC2, PRAS40, FOXO1/3a, and AS160 in adipocytes, with no evidence for substrate specificity.
Figure 3.
Akt2 Ser474 phosphorylation is required for maximal insulin-stimulated phosphorylation of PRAS40, AS160, FOXO1/3a, and TSC2 in 3T3-L1 adipocytes. 3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were treated with 10 μm MK2206 for 5 min, followed by 1 nm insulin for the times specified. A, lysates were immunoblotted with antibodies as specified, with 14-3-3 as a loading control. A representative blot for three to four independent experiments is presented. Band 1 and Band 2 indicate shifts in Akt2 gel migration. p, phosphorylated. B, quantification of A (n = 3–4, mean ± S.E., two-way analysis of variance; *, p < 0.05; ns, not significant). AU, arbitrary unit; p, phosphorylated.
Throughout the insulin time course, total phosphorylation of Akt at Thr309 was not significantly different between adipocytes expressing W80A and W80A-S474A Akt2 (Fig. 3, A and B). This finding is corroborated by previous studies where loss of Ser474 phosphorylation did not affect Thr309 phosphorylation (6, 20), supporting the notion that defective Akt2 substrate phosphorylation upon loss of Ser474 phosphorylation is not due to decreased Thr309 phosphorylation.
Akt2 Ser474 phosphorylation is required for maximal insulin-stimulated mTORC1 activation and protein synthesis
We next assessed whether loss of Akt2 Ser474 phosphorylation alters the activation of the insulin signaling network beyond the immediate phosphorylation of Akt substrates. In response to insulin, Akt activates mTORC1 via phosphorylation of its substrates TSC2 and PRAS40 (8, 9). mTORC1 then phosphorylates its substrates, which facilitate a variety of responses, including an increase in the rate of protein synthesis (Fig. 4A) (10, 39, 40). Thus, we first assessed whether loss of Ser474 phosphorylation affects insulin-stimulated activation of mTORC1 and protein synthesis in adipocytes. We assessed mTORC1 activation by the phosphorylation status of its substrate, P70 S6K. In adipocytes expressing WT or kinase-dead W80A-T309A Akt2, MK2206 blocked the ability of Akt2 to activate mTORC1 in response to insulin (Fig. 4, B and C). Similarly, following MK2206 treatment, adipocytes expressing WT Akt2 could not facilitate insulin-stimulated protein synthesis (Fig. 4D), aligning with these events being activated downstream of Akt. In the presence of MK2206, adipocytes expressing W80A-S474A Akt2 exhibited partially impaired P70 S6K phosphorylation, S6 phosphorylation and protein synthesis compared with adipocytes expressing W80A Akt2 (Fig. 4, B–D). These data, together with diminished TSC2 and PRAS40 phosphorylation (Fig. 3, A and B), suggest that Akt2 Ser474 phosphorylation is essential for maximal insulin-stimulated mTORC1 activation and protein synthesis.
Figure 4.
Akt2 Ser474 phosphorylation is required for maximal insulin-stimulated mTORC1 activation, protein synthesis, FOXO nuclear exclusion, GLUT4 translocation, and glucose uptake in 3T3-L1 adipocytes. A, Akt facilitates insulin-stimulated GLUT4 translocation, glucose uptake, mTORC1 activation, protein synthesis, and FOXO nuclear exclusion. B, 3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were treated with 10 μm MK2206 for 5 min, followed by 1 nm insulin for 10 min. Lysates were immunoblotted with antibodies as specified, with 14-3-3 as a loading control. A representative blot for three independent experiments is presented. Band 1 and Band 2 indicate shifts in Akt2 gel migration. p, phosphorylated. C, quantification of B (n = 3, mean ± S.E., two-tailed paired t test; *, p < 0.05). AU, arbitrary unit. D, 3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were treated with 10 μm MK2206 for 5 min followed by 1 nm insulin for 1 h and assessed for [3H]leucine incorporation for protein synthesis (n = 4, mean ± S.E., two-tailed paired t test; *, p < 0.05). E, 3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were electroporated with FOXO1-mNeonGreen. Cells were treated with 10 μm MK2206 for 5 min, followed by 1 nm insulin and nuclear exclusion assessed using live-cell spinning-disk confocal microscopy. Representative images for three independent experiments are presented. F, quantification of E (WT, 45 cells from three independent experiments; W80A, 63 cells from three independent experiments; W80A-S474A, 63 cells from three independent experiments; mean ± S.E.). G, 3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were electroporated with pHluorin-GLUT4-mRuby3. Cells were treated with 10 μm MK2206 for 5 min followed by 1 nm insulin and GLUT4 translocation to the plasma membrane and assessed using live-cell TIRF microscopy. Representative images for three independent experiments are presented. H, quantification of G (WT, 78 cells from three independent experiments; W80A, 138 cells from three independent experiments; W80A-S474A, 135 cells from three independent experiments, mean ± S.E.). I, 3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were treated with 10 μm MK2206 for 5 min followed by 1 nm insulin for 20 min and assessed for [3H]2-deoxyglucose uptake (n = 4, mean ± S.E., two-tailed paired t test; *, p < 0.05).
Akt2 Ser474 phosphorylation is required for maximal insulin-stimulated FOXO nuclear exclusion
Akt phosphorylates FOXO proteins to regulate their transcriptional activity via nuclear exclusion (Fig. 4A) (13, 14). Therefore, we next assessed the functional consequence of Akt-mediated FOXO phosphorylation by examining whether Akt2 Ser474 phosphorylation is required for insulin-stimulated FOXO1 nuclear exclusion. In adipocytes expressing WT Akt2, MK2206 inhibited insulin-stimulated FOXO1 nuclear exclusion (Fig. 4, E and F), consistent with it being Akt-dependent. In adipocytes expressing W80A-S474A Akt2, the rate of FOXO1 nuclear exclusion was reduced compared to adipocytes expressing W80A Akt2 (Fig. 4, E and F). These observations, together with impaired FOXO1/3a phosphorylation (Fig. 3, A and B), suggest that Akt2 Ser474 phosphorylation is required for maximal insulin-stimulated FOXO nuclear exclusion.
Akt2 Ser474 phosphorylation is required for maximal insulin-stimulated GLUT4 translocation and glucose uptake
Finally, we assessed whether Akt2 Ser474 phosphorylation is required for insulin-stimulated GLUT4 translocation and glucose uptake in adipocytes. Akt activation is both necessary and sufficient for insulin-stimulated glucose uptake (41). This is predominantly achieved through the phosphorylation of AS160, which is required for translocation of GLUT4 to the PM (Fig. 4A) (30, 42). Consistent with this, MK2206 blocked insulin-stimulated GLUT4 translocation and glucose uptake in adipocytes expressing WT Akt2 (Fig. 4, G–I). In the presence of MK2206, both GLUT4 translocation and glucose uptake were activated by insulin in adipocytes expressing W80A Akt2; however, only partial activation was achieved in adipocytes expressing W80A-S474A Akt2 (Fig. 4, G–I). These observations, together with impaired AS160 phosphorylation (Fig. 3, A and B), suggest that Akt2 Ser474 phosphorylation is essential for maximal insulin-stimulated GLUT4 translocation and glucose uptake.
Discussion
Akt is a major regulatory node for numerous biological processes, but the mechanism by which it is activated remains controversial. It is widely accepted that Thr309 phosphorylation is essential for Akt activation (6, 15–17); however, the role of Ser474 phosphorylation is unclear. Here we examined how loss of Ser474 phosphorylation influences Akt2 activity toward a number of substrates and insulin-regulated processes in 3T3-L1 adipocytes. Insulin-stimulated phosphorylation of four canonical Akt substrates was reduced by ∼50% in the absence of Ser474 phosphorylation. Accordingly, we measured reduced efficiency of several insulin-regulated processes in the absence of Ser474 phosphorylation. Overall, our findings suggest that Ser474 phosphorylation regulates the full activation of Akt2 toward a cadre of substrates and biological processes in adipocytes.
Previous studies have proposed an alternate model where Ser474 phosphorylation is required for Akt activity only toward select substrates. Evidence for this model is largely derived from studies where Ser474 phosphorylation was prevented via ablation of its upstream kinase, mTORC2 (20–22). However, by interfering with the upstream kinase of Ser474, these studies have several disadvantages, such as possible induction of long-term compensatory mechanisms and interference with other downstream actions of mTORC2 independent of Akt Ser474 phosphorylation. MK2206-resistant W80A Akt offered a valuable tool to overcome these disadvantages and interrogate the role of Akt2 Ser474 phosphorylation in cells through direct mutation of Ser474. Despite examining a cadre of Akt substrates and downstream biological processes, many of which were identical to those examined in prior studies, our data do not illustrate selective signaling in the absence of Ser474 phosphorylation but, rather, a global decrease in Akt2 activity. We cannot exclude that these differences between our data and previous reports are due to cell-specific effects, as each study was undertaken in a different cell line, or that the role of Ser474 phosphorylation differs for each Akt isoform. However, we speculate that these disparities are more likely reflective of other events downstream of mTORC2 conferring Akt substrate specificity rather than Ser474 phosphorylation itself. For example, mTORC2 has been shown to phosphorylate other sites on Akt, such as Thr450 on its turn motif (27, 28), and Ser477/Thr479 on its hydrophobic motif (26). This and other reported mechanisms of Akt substrate specificity (43) deserve further investigation largely because of their applicability to cancer treatments. Akt inhibitors have been tested in clinical trials; however, although they can successfully inhibit cancer growth, hyperglycemia often accompanies treatment (44, 45). This is not surprising considering Akt controls glucose metabolism. To design cancer therapeutics without these metabolic toxicities, it is of interest to therapeutically target specific actions of Akt rather than using pan-Akt inhibitors.
Akt2 undergoes differential SDS-PAGE migration upon insulin stimulation as a result of changes in its phosphorylation status (Fig. S3). FLAG-W80A, FLAG-W80A-T309A, and FLAG-W80A-S474A Akt2 did not differ in their gel migration in the basal state (Fig. 2B; indicated as Band 2 in Figs. 2E, 3A, and 4B). However, upon insulin stimulation, there was decreased migration of FLAG-W80A and FLAG-W80A-T309A Akt2 (indicated as Band 1 in Figs. 2E, 3A, and 4B) but not FLAG-W80A-S474A Akt2. These data suggest that Ser474 phosphorylation, but not Thr309 phosphorylation, is required for the insulin-stimulated band shift in Akt2. It is well established that some but not all phosphorylation events can result in a band shift (46). This can be attributed to the charge of the residues surrounding the phosphosite; only in the appropriate local environment does addition of a phosphate group decrease SDS binding and, consequently, migration (47). Nevertheless, this, coupled with the requirement for Ser474 phosphorylation for the band shift, leads us to propose that the insulin-stimulated band shift in Akt2 is caused by either Ser474 phosphorylation itself or other Ser474 phosphorylation–dependent phosphorylation sites. For example, we speculate that phosphorylation of Akt2 at Ser478 may be dependent on Ser474 phosphorylation as they are closely localized, increase with similar kinetics upon insulin stimulation (38), and interdependence between the corresponding sites on Akt1 has been reported (26). Elucidating whether this is the case may help dissect why Ser474 phosphorylation is required for maximal Akt2 kinase activity, particularly as phosphorylation events on Akt other than that at Thr309 and Ser474 are increasingly being shown to have substantial impact on Akt kinase activity (26, 48).
A major advantage of the W80A system used in this study is that it allowed investigation of Akt mutants in cells rather than in vitro. Our work in cells suggests that Akt can achieve half of its maximal kinase activity without Ser474 phosphorylation, which is in striking contrast to in vitro studies showing no or very little kinase activity in the absence of Ser474 phosphorylation (6, 17, 49). This difference is likely reflective of in vitro systems not reproducing complex cellular control points, comprising signal amplification, macromolecular crowding, interacting proteins, protein localization, and positive/negative feedback (36, 49, 50). As substantial Akt activation occurs without Ser474 phosphorylation in the cell, our data align with studies reporting that Ser474 phosphorylation poorly correlated with Akt activity (51); however, Ser474 phosphorylation is used in countless studies as a proxy of Akt activity. Rather, we suggest using phosphorylation of Akt at Thr309 or its substrates as more accurate indices of Akt activation (52).
Although we show a global defect in insulin-stimulated Akt2 activity upon loss of Ser474 phosphorylation, a recent study by Beg et al. (30), using the same W80A system in 3T3-L1 adipocytes, reported that insulin-stimulated GLUT4 translocation and mTORC1 activation were unaffected by loss of Akt2 Ser474 phosphorylation. There are differences between our studies that may explain these disparities. Most importantly, Beg et al. (30) used a dose of MK2206 (1 μm) they and others have shown to be insufficient to completely impair substrate phosphorylation by endogenous Akt (53). Having residual endogenous Akt activity will likely alter the behavior of the ectopic W80A Akt mutant, as Akt activity is extremely sensitive to temporal changes in its activation (54–56). Even a minute amount of residual endogenous Akt activity may skew signaling outcomes, as only a small percentage of the total Akt pool is required to achieve a maximal response for many downstream processes (36, 37). In contrast, we utilized 10 μm MK2206, as it was sufficient to block endogenous Akt activation, allowing specific analysis of ectopic W80A Akt2 (Figs. 1, D and E, and 2, E and F). Furthermore, to control for endogenous Akt inhibition, Beg et al. (30) used untransfected 3T3-L1 adipocytes. However, based on our studies, it is important to use adipocytes expressing FLAG-WT Akt2 to ensure effective comparison with adipocytes expressing FLAG-W80A Akt2. Nevertheless, Beg et al. (30) also reported that insulin-stimulated GLUT1 translocation requires Akt2 Ser474 phosphorylation, which we support (Fig. S4). These data suggest that further inquiry may reveal Akt substrates that control GLUT1 translocation and require Ser474-phosphorylated Akt for activation. However, an alternate explanation is that maximal Akt activity may be required to trigger insulin-stimulated GLUT1 translocation; this result may not reflect Akt substrate specificity per se but, rather, a difference in the dose–response relationship between Akt activation and various downstream processes.
The mechanism by which substantial Akt activity is possible in the cell without Ser474 phosphorylation remains elusive. Akt crystal structures suggest that HM binding to the N-lobe of the kinase domain is required for an active conformation and that Ser474 phosphorylation is essential for this interaction (18, 19). We speculate that this interaction can occur in the absence of Ser474 phosphorylation in cells; this weak interaction could be more favorable in cells than in vitro because of macromolecular crowding or stabilization by interacting proteins (50). Intriguingly, other AGC kinases, such as protein kinase A, lack a phosphosite within their HM, yet it retains the ability to interact with its kinase domain and induce activity (18, 19, 57). Thus, Ser474 phosphorylation may increase the affinity of the HM for the kinase domain but not be a prerequisite for the interaction or Akt activation. Alternatively, it has recently been found that the HM interacts with the PH–kinase domain linker to relieve PH domain–mediated autoinhibition and increase Akt activity (58). Whether this interaction can occur without Ser474 phosphorylation remains to be elucidated. Uncovering the mechanism by which substantial Akt kinase activity is possible without Ser474 phosphorylation is essential, as there is interest in developing therapeutic agents that inhibit Akt by targeting its hydrophobic motif (59).
Experimental procedures
Cloning
TagRFP-T-Akt2 has been described previously (34). TagRFP-T-Akt2 with the W80A mutation was generated using site-directed mutagenesis (60). For FLAG-Akt2 constructs, Akt2 was amplified by PCR from human cDNA with addition of attB sites at either end to be compatible with gateway cloning. Akt2 cDNA was inserted into pDONR221 (Thermo Fisher Scientific) using GatewayTM BP ClonaseTM II Enzyme Mix (Invitrogen) and then into the retroviral vector pMIG-t-sapphire-puromycin (pMIG-tsap-puro) using GatewayTM LR ClonaseTM II Enzyme Mix (Invitrogen). A FLAG tag was then added to the N terminus of Akt2 using Gibson assembly cloning (61). Site-directed mutagenesis (60) was used to create FLAG-tagged W80A, W80A-T309A, and W80A-S474A Akt2 plasmids. For FOXO1-mNeonGreen, the N-terminal region of FOXO1 (1–380) containing the nuclear localization and nuclear export sequences was codon-optimized and synthesized as a gene block (IDT Technologies). An mNeonGreen tag was placed at its C terminus, and this was cloned into the mammalian expression vector pcDNA3.1. The mutations S209A, H212A, and S215A were introduced to impair DNA binding and phosphorylation by STK4 as described previously (62). pHluorin-GLUT4-mRuby3 was generated from pHluorin-GLUT4-tdTomato (63) by replacing tdTomato with mammalian expression-optimized mRuby3 synthesized as a gene block (IDT Technologies). Primer and construct sequences are available upon request. All constructs were confirmed by Sanger sequencing.
Cell culture
3T3-L1 fibroblasts obtained from the Howard Green Laboratory (Harvard Medical School) were cultured in high glucose DMEM (Gibco by Life Technologies) supplemented with 10% (v/v) FBS (Gibco by Life Technologies), and 1× GlutaMAX (Gibco by Life Technologies) at 37 °C and 10% CO2. All cell lines were mycoplasma-free. For the generation of 3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants, fibroblasts were transduced in the presence of 8 μg/ml hexadimethrine bromide (Polybrene, Sigma-Aldrich) with pMIG-tsap-puro (empty vector control) or pMIG-tsap-puro-FLAG-Akt2 retrovirus generated from Plat-E cells (Cell Biolabs, San Diego, CA) as described previously (64). Transduced cells were then selected using 2 μg/ml puromycin. Cells were differentiated into adipocytes as described previously (65), and used for experiments 7–12 days after initiation of differentiation. To minimize differences between cells expressing each Akt2 mutant, retroviral transduction was performed simultaneously using the same population of 3T3-L1 fibroblasts. Cells were infected with high virus titers so that there was minimal cell loss upon antibiotic selection. Then, to minimize differences in genetic drift, each cell population was subject to identical culturing conditions, used for experiments at the same passage, and not passaged more than 10 times. Macroscopically, we observed no morphological differences between cell populations at any stage throughout their differentiation into adipocytes. Furthermore, we performed the central experiments comprising Figs. 3, A and B, and 4, B–D, with three independently derived cell populations. Each achieved identical results; Ser474 phosphorylation was required for maximal Akt2 kinase activity.
Western blotting
3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were serum-starved with DMEM containing 1× GlutaMAX and 0.2% BSA (w/v) for 2 h. Cells were then exposed to 10 μm MK2206 (Selleckchem) or DMSO (Sigma-Aldrich) vehicle control for 5 min, followed by 1 nm insulin. We utilized 10 μm MK2206 to inhibit endogenous Akt, as we have shown previously that this dose is required to completely inhibit Akt activation (53). Cells were then placed on ice, washed with cold PBS, lysed with 1% (w/v) SDS in PBS containing protease inhibitors (Roche Applied Science) and phosphatase inhibitors (2 mm Na3VO4, 1 mm Na4O7P2, and 10 mm NaF), and tip probe–sonicated. Lysates were centrifuged at 13,000 × g for 15 min at 4 °C. The lipid layer was removed, and protein content was quantified using the PierceTM BCA Protein Assay Kit (Thermo Scientific). 10 μg of lysate was then resolved by SDS-PAGE, transferred to PVDF membranes, and immunoblotted as described previously (39). All primary antibodies were from Cell Signaling Technology, but 14-3-3 was from Santa Cruz Biotechnology. Densitometry analysis was performed using ImageStudioLite version 5.2.5 (LI-COR). Band intensities were normalized to the loading control (14-3-3). Statistical tests were performed using GraphPad Prism version 7.0.
Live-cell TIRF microscopy
Matrigel-coated coverslips were prepared as described previously (34). To assess insulin-stimulated GLUT4 translocation to the plasma membrane, 3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were electroporated 7 days post-differentiation with 6 μg of pHluorin-GLUT4-mRuby3. To assess insulin-stimulated Akt2 recruitment to the plasma membrane, 3T3-L1 adipocytes were electroporated 6–8 days post-differentiation with 6 μg of WT or W80A TagRFP-T-Akt2. Then cells were placed onto the Matrigel-coated coverslips as described previously (34). 24–48 h after electroporation, adipocytes were serum-starved for 2 h and then incubated at 37 °C with Krebs–Ringer–phosphate–HEPES (KRPH) buffer (0.6 mm Na2HPO4, 0.4 mm NaH2PO4, 120 mm NaCl, 6 mm KCl, 1 mm CaCl2, 1.2 mm MgSO4, and 12.5 mm HEPES (pH 7.4)) supplemented with 10 mm glucose, 1× minimum Eagle's medium amino acids (Gibco by Life Technologies), 1× GlutaMAX, and 0.2% (w/v) BSA. The cells were then treated with 10 μm MK2206 and/or 1 nm insulin using a custom-made perfusion system. Images were acquired using the Nikon Ti-Lapps H-TIRF module angled to image ∼90 nm into cells. Temperature and humidity were maintained using an Okolab cage incubator and temperature control. The change in pixel intensity over time was determined for each individual cell using Fiji (66). The average pixel intensity over time for each cell was normalized to its average intensity over the basal period.
[3H]2-deoxyglucose uptake assay
3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were serum-starved for 2 h, washed with PBS, and incubated at 37 °C with KRPH buffer. Cells were stimulated with 10 μm MK2206 for 5 min or DMSO vehicle control, followed by 1 nm insulin. To determine background radiation, 25 μm cytochalasin B (Sigma-Aldrich) as a glucose transport inhibitor was added to a well of cells following 10-min insulin stimulation. After 15 min of insulin stimulation, 50 μm unlabeled 2-deoxyglucose and 0.25 μCi/ml [3H]2-deoxyglucose (Perkin-Elmer Life Sciences) was added to all cells for 5 min. Glucose uptake was terminated by placing the plate on ice, washing with cold PBS, and lysing the cells with 1% (w/v) Triton X-100 in PBS. Lysates were added to a scintillation vial with 3 ml of Ultima GoldTM XR liquid scintillation mixture (Perkin-Elmer Life Sciences). The radioactivity of each sample in disintegrations per minute was determined using a Tri-Carb liquid scintillation counter (Perkin-Elmer Life Sciences). The protein content of each sample was assessed using the Pierce BCA Protein Assay Kit (Thermo Scientific). Assays were performed in duplicate, and the average of the duplicate was considered one biological replicate. The disintegrations per minute for each condition were converted to nanomoles of 2-deoxyglucose, and data were normalized to protein levels. The value for cytochalasin B–treated cells was subtracted from all other values as a background control. Statistical tests were performed using GraphPad Prism version 7.0.
[3H]leucine incorporation assay for protein synthesis
3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were serum-starved in leucine-free DMEM supplemented with 0.2% BSA and 20 mm HEPES (pH 7.4) for 2 h. Cells were then treated with 10 μm MK2206 for 5 min, followed by 5 μCi/ml [3H]leucine (Perkin-Elmer Life Sciences) and 1 nm insulin for 1 h. To determine background radiation, 10 μm cycloheximide, a translation inhibitor, was added 30 min before [3H]leucine to a single well. Leucine incorporation was terminated by placing the plate on ice, washing with cold PBS, and then incubating cells with cold 10% (v/v) TCA for 10 min to lyse cells and precipitate protein. Pellets were washed three times in cold 10% (v/v) TCA to remove free [3H]leucine. Pellets were resuspended in 50 mm NaOH with 1% (w/v) Triton X-100 in PBS at 65 °C for 30 min and then added to a scintillation vial with 3 ml of Ultima GoldTM XR liquid scintillation mixture (Perkin-Elmer Life Sciences). The radioactivity of each sample in disintegrations per minute was determined using a Tri-Carb liquid scintillation counter (Perkin-Elmer Life Sciences). The protein content of each sample was assessed using the Pierce BCA Protein Assay Kit (Thermo Scientific). Assays were performed in triplicate, and the average of the triplicate was considered one biological replicate. The disintegrations per minute for each condition were converted to nanomoles leucine, and data were normalized to protein levels. The value for cycloheximide-treated cells was subtracted from all other values to account for background radiation. Statistical tests were performed using GraphPad Prism version 7.0.
Live-cell spinning-disk confocal microscopy
Matrigel-coated coverslips were prepared as described previously (34). To assess insulin-stimulated FOXO nuclear exclusion, 3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were electroporated 6 days post-differentiation with 6 μg of FOXO1(1–380; S209A, H212A, S215A)-mNeonGreen. Then cells were placed onto the Matrigel-coated coverslips as described previously (34). 24 h after electroporation, adipocytes were serum-starved for 2 h and then stained with Hoechst 33342 for 10 min. Adipocytes were then incubated at 37 °C with KRPH buffer supplemented with 10 mm glucose, 1× minimum Eagle's medium amino acids, 1× GlutaMAX, and 0.2% (w/v) BSA. The cells were then treated with 10 μm MK2206 and 1 nm insulin using a custom-made perfusion system. Images were acquired on a Nikon Ti inverted confocal microscope with an Andor Diskovery spinning-disk system and an Andor Ixon 888 EmCCD camera (Andor). Temperature and humidity were maintained using an Okolab cage incubator and temperature control. For each individual cell, the median pixel intensity of the cytosol was divided by the median pixel intensity of the nucleus using Fiji (66).
λ protein phosphatase assay
3T3-L1 adipocytes were serum-starved for 2 h and then treated with 100 nm insulin for 10 min. Cells were placed on ice, washed with cold PBS, and harvested in 1% (w/v) Triton X-100 in PBS. Cells were homogenized by passing through a 22-gauge needle six times and a 27-gauge needle six times prior to centrifugation at 12,000 × g for 15 min. 100 μg of the supernatant was treated with 1 mm MnCl2 (New England Biolabs), 1× NEBuffer for protein metallophosphatases (New England Biolabs), and either 800 units of λ protein phosphatase (New England Biolabs) or phosphatase inhibitors (10 mm Na3VO4, 5 mm Na4O7P2, or 50 mm NaF) at 30 °C for 15 min. Reactions were stopped with 2% (w/v) SDS and then resolved by SDS-PAGE.
Subcellular fractionation
3T3-L1 adipocytes stably expressing FLAG-Akt2 mutants were serum-starved for 2 h and then exposed to 10 μm MK2206 for 5 min, followed by 0.5 nm insulin for 20 min. Cells were placed on ice, washed with cold PBS, and harvested in cold HES buffer (20 mm HEPES, 1 mm EDTA, and 250 mm sucrose (pH 7.4)) containing phosphatase and protease inhibitors. All subsequent steps were carried out at 4 °C as described previously (67). Briefly, cells were homogenized using a Dounce tissue grinder prior to centrifugation at 500 × g for 10 min. The supernatant was centrifuged at 13,550 × g for 12 min to pellet the plasma membrane and mitochondria/nuclei. This pellet was resuspended in HES buffer and again centrifuged at 13,550 × g for 12 min. The pellet was then resuspended in HES buffer, layered over high-sucrose HES buffer (20 mm HEPES, 1 mm EDTA, and 1.12 m sucrose (pH 7.4)), and centrifuged at 111,160 × g for 60 min in a swing-out rotor. The PM fraction was collected at the interface between the sucrose layers and pelleted by centrifugation at 235,000 × g for 75 min. The PM pellet was resuspended in HES buffer containing phosphatase and protease inhibitors. Protein concentrations for each fraction were determined using the Pierce BCA Protein Assay Kit (Thermo Scientific).
Author contributions
A. L. K. data curation; A. L. K. formal analysis; A. L. K. validation; A. L. K., K. C. C., D. M. N., and A. Z. investigation; A. L. K. visualization; A. L. K., K. C. C., and D. M. N. methodology; A. L. K. writing-original draft; A. L. K., J. R. K., D. J. F., J. G. B., and D. E. J. writing-review and editing; K. C. C., J. R. K., D. J. F., J. G. B., and D. E. J. supervision; K. C. C. project administration; J. R. K., D. J. F., J. G. B., and D. E. J. funding acquisition; D. J. F., J. G. B., and D. E. J. conceptualization; D. E. J. resources.
Supplementary Material
Acknowledgments
We acknowledge the facilities of and scientific and technical assistance from the Australian Microscopy and Microanalysis Research Facility at the Charles Perkins Centre, University of Sydney.
This work was supported by Australian Research Council project grant DP180103482 (to D. E. J. and J. G. B.), National Health and Medical Research Council project grant GNT1120201 (to D. E. J.), and Diabetes Australia research grant Y19G-FAZD to (D. J. F., J. G. B., and J. R. K.). The authors declare that they have no conflicts of interest with the contents of this article.
This article contains Figs. S1–S4.
- PIP3
- phosphatidylinositol (3,4,5) triphosphate
- PH
- pleckstrin homology
- HM
- hydrophobic motif
- PM
- plasma membrane
- TIRF
- total internal reflection fluorescence
- cDNA
- complementary DNA.
References
- 1. Manning B. D., and Cantley L. C. (2007) AKT/PKB signaling: navigating downstream. Cell 129, 1261–1274 10.1016/j.cell.2007.06.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Czech M. P. (2000) PIP2 and PIP3: complex roles at the cell surface. Cell 100, 603–606 10.1016/S0092-8674(00)80696-0 [DOI] [PubMed] [Google Scholar]
- 3. Oatey P. B., Venkateswarlu K., Williams A. G., Fletcher L. M., Foulstone E. J., Cullen P. J., and Tavaré J. M. (1999) Confocal imaging of the subcellular distribution of phosphatidylinositol 3,4,5-trisphosphate in insulin- and PDGF-stimulated 3T3-L1 adipocytes. Biochem. J. 344, 511–518 10.1042/bj3440511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Kumar C. C., and Madison V. (2005) AKT crystal structure and AKT-specific inhibitors. Oncogene 24, 7493–7501 10.1038/sj.onc.1209087 [DOI] [PubMed] [Google Scholar]
- 5. Andjelković M., Alessi D. R., Meier R., Fernandez A., Lamb N. J., Frech M., Cron P., Cohen P., Lucocq J. M., and Hemmings B. A. (1997) Role of translocation in the activation and function of protein kinase B. J. Biol. Chem. 272, 31515–31524 10.1074/jbc.272.50.31515 [DOI] [PubMed] [Google Scholar]
- 6. Alessi D. R., Andjelkovic M., Caudwell B., Cron P., Morrice N., Cohen P., and Hemmings B. A. (1996) Mechanism of activation of protein kinase B by insulin and IGF-1. EMBO J. 15, 6541–6551 10.1002/j.1460-2075.1996.tb01045.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Sarbassov D. D., Guertin D. A., Ali S. M., and Sabatini D. M. (2005) Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 307, 1098–1101 10.1126/science.1106148 [DOI] [PubMed] [Google Scholar]
- 8. Vander Haar E., Lee S. I., Bandhakavi S, Griffin T. J., and Kim D.-H. (2007) Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40. Nat. Cell Biol. 9, 316–323 10.1038/ncb1547 [DOI] [PubMed] [Google Scholar]
- 9. Inoki K., Li Y., Zhu T., Wu J., and Guan K. L. (2002) TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat. Cell Biol. 4, 648–657 10.1038/ncb839 [DOI] [PubMed] [Google Scholar]
- 10. Ma X. M., and Blenis J. (2009) Molecular mechanisms of mTOR-mediated translational control. Nat. Rev. Mol. Cell Biol. 10, 307–318 10.1038/nrm2672 [DOI] [PubMed] [Google Scholar]
- 11. Tan S. X., Ng Y., Burchfield J. G., Ramm G., Lambright D. G., Stöckli J., and James D. E. (2012) The Rab GTPase-activating protein TBC1D4/AS160 contains an atypical phosphotyrosine-binding domain that interacts with plasma membrane phospholipids to facilitate GLUT4 trafficking in adipocytes. Mol. Cell. Biol. 32, 4946–4959 10.1128/MCB.00761-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Kane S., Sano H., Liu S. C., Asara J. M., Lane W. S., Garner C. C., and Lienhard G. E. (2002) A method to identify serine kinase substrates. Akt phosphorylates a novel adipocyte protein with a Rab GTPase-activating protein (GAP) domain. J. Biol. Chem. 277, 22115–22118 10.1074/jbc.C200198200 [DOI] [PubMed] [Google Scholar]
- 13. Brunet A., Bonni A., Zigmond M. J., Lin M. Z., Juo P., Hu L. S., Anderson M. J., Arden K. C., Blenis J., and Greenberg M. E. (1999) Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96, 857–868 10.1016/S0092-8674(00)80595-4 [DOI] [PubMed] [Google Scholar]
- 14. Czech M. P. (2003) Insulin's expanding control of forkheads. Proc. Natl. Acad. Sci. U.S.A. 100, 11198–11200 10.1073/pnas.2135308100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Mora A., Lipina C., Tronche F., Sutherland C., and Alessi D. R. (2005) Deficiency of PDK1 in liver results in glucose intolerance, impairment of insulin-regulated gene expression and liver failure. Biochem. J. 385, 639–648 10.1042/BJ20041782 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 16. Dangelmaier C., Manne B. K., Liverani E., Jin J., Bray P., and Kunapuli S. P. (2014) PDK1 selectively phosphorylates Thr308 on Akt and contributes to human platelet functional responses. Thromb. Haemost. 111, 508–517 10.1160/TH13-06-0484 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Meier R., Alessi D. R., Cron P., Andjelković M., and Hemmings B. A. (1997) Mitogenic activation, phosphorylation, and nuclear translocation of protein kinase Bβ. J. Biol. Chem. 272, 30491–30497 10.1074/jbc.272.48.30491 [DOI] [PubMed] [Google Scholar]
- 18. Yang J., Cron P., Good V. M., Thompson V., Hemmings B. A., and Barford D. (2002) Crystal structure of an activated Akt/protein kinase B ternary complex with GSK3-peptide and AMP-PNP. Nat. Struct. Mol. Biol. 9, 940–944 10.1038/nsb870 [DOI] [PubMed] [Google Scholar]
- 19. Yang J., Cron P., Thompson V., Good V. M., Hess D., Hemmings B. A., and Barford D. (2002) Molecular mechanism for the regulation of protein kinase B/Akt by hydrophobic motif phosphorylation. Mol. Cell 9, 1227–1240 10.1016/S1097-2765(02)00550-6 [DOI] [PubMed] [Google Scholar]
- 20. Jacinto E., Facchinetti V., Liu D., Soto N., Wei S., Jung S. Y., Huang Q., Qin J., and Su B. (2006) SIN1/MIP1 maintains rictor-mTOR complex integrity and regulates Akt phosphorylation and substrate specificity. Cell 127, 125–137 10.1016/j.cell.2006.08.033 [DOI] [PubMed] [Google Scholar]
- 21. Kumar A., Lawrence J. C. Jr., Jung D. Y., Ko H. J., Keller S. R., Kim J. K., Magnuson M. A., and Harris T. E. (2010) Fat cell–specific ablation of Rictor in mice impairs insulin-regulated fat cell and whole-body glucose and lipid metabolism. Diabetes 59, 1397–1406 10.2337/db09-1061 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Moore S. F., Hunter R. W., and Hers I. (2011) mTORC2 protein complex-mediated Akt (protein kinase B) serine 473 phosphorylation is not required for Akt1 activity in human platelets [corrected]. J. Biol. Chem. 286, 24553–24560 10.1074/jbc.M110.202341 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Tang Y., Wallace M., Sanchez-Gurmaches J., Hsiao W.-Y., Li H., Lee P. L., Vernia S., Metallo C. M., and Guertin D. A. (2016) Adipose tissue mTORC2 regulates ChREBP-driven de novo lipogenesis and hepatic glucose metabolism. Nat. Commun. 7, 11365 10.1038/ncomms11365 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Barbaric I., Miller G., and Dear T. N. (2007) Appearances can be deceiving: phenotypes of knockout mice. Brief Funct. Genomic Proteomic. 6, 91–103 10.1093/bfgp/elm008 [DOI] [PubMed] [Google Scholar]
- 25. Murray E. R., and Cameron A. J. M. (2017) Towards specific inhibition of mTORC2. Aging 9, 2461–2462 10.18632/aging.101346 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Liu P., Begley M., Michowski W., Inuzuka H., Ginzberg M., Gao D., Tsou P., Gan W., Papa A., Kim B. M., Wan L., Singh A., Zhai B., Yuan M., Wang Z., et al. (2014) Cell-cycle-regulated activation of Akt kinase by phosphorylation at its carboxyl terminus. Nature 508, 541–545 10.1038/nature13079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Ikenoue T., Inoki K., Yang Q., Zhou X., and Guan K. L. (2008) Essential function of TORC2 in PKC and Akt turn motif phosphorylation, maturation and signalling. EMBO J. 27, 1919–1931 10.1038/emboj.2008.119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Facchinetti V., Ouyang W., Wei H., Soto N., Lazorchak A., Gould C., Lowry C., Newton A. C., Mao Y., Miao R. Q., Sessa W. C., Qin J., Zhang P., Su B., and Jacinto E. (2008) The mammalian target of rapamycin complex 2 controls folding and stability of Akt and protein kinase C. EMBO J. 27, 1932–1943 10.1038/emboj.2008.120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Green C. J., Göransson O., Kular G. S., Leslie N. R., Gray A., Alessi D. R., Sakamoto K., and Hundal H. S. (2008) Use of Akt inhibitor and a drug-resistant mutant validates a critical role for protein kinase B/Akt in the insulin-dependent regulation of glucose and system A amino acid uptake. J. Biol. Chem. 283, 27653–27667 10.1074/jbc.M802623200 [DOI] [PubMed] [Google Scholar]
- 30. Beg M., Abdullah N., Thowfeik F. S., Altorki N. K., and McGraw T. E. (2017) Distinct Akt phosphorylation states are required for insulin regulated Glut4 and Glut1-mediated glucose uptake. eLife 6, e26896 10.7554/eLife.26896 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Kajno E., McGraw T. E., and Gonzalez E. (2015) Development of a new model system to dissect isoform specific Akt signalling in adipocytes. Biochem. J. 468, 425–434 10.1042/BJ20150191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Kandel E. S., and Hay N. (1999) The regulation and activities of the multifunctional serine/threonine kinase Akt/PKB. Exp. Cell Res. 253, 210–229 10.1006/excr.1999.4690 [DOI] [PubMed] [Google Scholar]
- 33. Cherrin C., Haskell K., Howell B., Jones R., Leander K., Robinson R., Watkins A., Bilodeau M., Hoffman J., Sanderson P., Hartman G., Mahan E., Prueksaritanont T., Jiang G., She Q. B., et al. (2010) An allosteric Akt inhibitor effectively blocks Akt signaling and tumor growth with only transient effects on glucose and insulin levels in vivo. Cancer Biol. Ther. 9, 493–503 10.4161/cbt.9.7.11100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Norris D. M., Yang P., Krycer J. R., Fazakerley D. J., James D. E., and Burchfield J. G. (2017) Improved Akt reporter reveals intra- and inter-cellular heterogeneity and oscillations in signal transduction. J. Cell Sci. 130, 2757–2766 10.1242/jcs.205369 [DOI] [PubMed] [Google Scholar]
- 35. Ebner M., Lučić I., Leonard T. A., and Yudushkin I. (2017) PI(3,4,5)P3 engagement restricts Akt activity to cellular membranes. Mol. Cell 65, 416–431.e6 10.1016/j.molcel.2016.12.028 [DOI] [PubMed] [Google Scholar]
- 36. Tan S.-X., Ng Y., Meoli C. C., Kumar A., Khoo P.-S., Fazakerley D. J., Junutula J. R., Vali S., James D. E., and Stöckli J. (2012) Amplification and demultiplexing in insulin-regulated Akt protein kinase pathway in adipocytes. J. Biol. Chem. 287, 6128–6138 10.1074/jbc.M111.318238 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Hoehn K. L., Hohnen-Behrens C., Cederberg A., Wu L. E., Turner N., Yuasa T., Ebina Y., and James D. E. (2008) IRS1-independent defects define major nodes of insulin resistance. Cell Metab. 7, 421–433 10.1016/j.cmet.2008.04.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Humphrey S. J., Yang G., Yang P., Fazakerley D. J., Stöckli J., Yang J. Y., and James D. E. (2013) Dynamic adipocyte phosphoproteome reveals that Akt directly regulates mTORC2. Cell Metab. 17, 1009–1020 10.1016/j.cmet.2013.04.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Marshall S. (1989) Kinetics of insulin action on protein synthesis in isolated adipocytes: ability of glucose to selectively desensitize the glucose transport system without altering insulin stimulation of protein synthesis. J. Biol. Chem. 264, 2029–2036 [PubMed] [Google Scholar]
- 40. Marshall S., and Monzon R. (1989) Amino acid regulation of insulin action in isolated adipocytes: selective ability of amino acids to enhance both insulin sensitivity and maximal insulin responsiveness of the protein synthesis system. J. Biol. Chem. 264, 2037–2042 [PubMed] [Google Scholar]
- 41. Ng Y., Ramm G., Lopez J. A., and James D. E. (2008) Rapid activation of Akt2 is sufficient to stimulate GLUT4 translocation in 3T3-L1 adipocytes. Cell Metab. 7, 348–356 10.1016/j.cmet.2008.02.008 [DOI] [PubMed] [Google Scholar]
- 42. James D. E., Brown R., Navarro J., and Pilch P. F. (1988) Insulin-regulatable tissues express a unique insulin-sensitive glucose transport protein. Nature 333, 183–185 10.1038/333183a0 [DOI] [PubMed] [Google Scholar]
- 43. Balzano D., Fawal M. A., Velázquez J. V., Santiveri C. M., Yang J., Pastor J., Campos-Olivas R., Djouder N., and Lietha D. (2015) Alternative activation mechanisms of protein kinase B trigger distinct downstream signaling responses. J. Biol. Chem. 290, 24975–24985 10.1074/jbc.M115.651570 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Crouthamel M. C., Kahana J. A., Korenchuk S., Zhang S. Y., Sundaresan G., Eberwein D. J., Brown K. K., and Kumar R. (2009) Mechanism and management of AKT inhibitor-induced hyperglycemia. Clin. Cancer Res. 15, 217–225 10.1158/1078-0432.CCR-08-1253 [DOI] [PubMed] [Google Scholar]
- 45. Altomare D. A., and Testa J. R. (2005) Perturbations of the AKT signaling pathway in human cancer. Oncogene 24, 7455–7464 10.1038/sj.onc.1209085 [DOI] [PubMed] [Google Scholar]
- 46. Morton S., Davis R. J., McLaren A., and Cohen P. (2003) A reinvestigation of the multisite phosphorylation of the transcription factor c-Jun. EMBO J. 22, 3876–3886 10.1093/emboj/cdg388 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Shi Y., Mowery R. A., Ashley J., Hentz M., Ramirez A. J., Bilgicer B., Slunt-Brown H., Borchelt D. R., and Shaw B. F. (2012) Abnormal SDS-PAGE migration of cytosolic proteins can identify domains and mechanisms that control surfactant binding. Protein Sci. 21, 1197–1209 10.1002/pro.2107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Risso G., Blaustein M., Pozzi B., Mammi P., and Srebrow A. (2015) Akt/PKB: one kinase, many modifications. Biochem. J. 468, 203–214 10.1042/BJ20150041 [DOI] [PubMed] [Google Scholar]
- 49. Balasuriya N., Kunkel M. T., Liu X., Biggar K. K., Li S. S., Newton A. C., and O'Donoghue P. (2018) Genetic code expansion and live cell imaging reveal that Thr-308 phosphorylation is irreplaceable and sufficient for Akt1 activity. J. Biol. Chem. 293, 10744–10756 10.1074/jbc.RA118.002357 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Kuznetsova I. M., Turoverov K. K., and Uversky V. N. (2014) What macromolecular crowding can do to a protein. Int. J. Mol. Sci. 15, 23090–23140 10.3390/ijms151223090 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Vincent E. E., Elder D. J., Thomas E. C., Phillips L., Morgan C., Pawade J., Sohail M., May M. T., Hetzel M. R., and Tavaré J. M. (2011) Akt phosphorylation on Thr308 but not on Ser473 correlates with Akt protein kinase activity in human non-small cell lung cancer. Br. J. Cancer 104, 1755–1761 10.1038/bjc.2011.132 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Fazakerley D. J., Krycer J. R., Kearney A. L., Hocking S. L., and James D. E. (2018) Muscle and adipose tissue insulin resistance: malady without mechanism? J. Lipid Res. 10.1194/jlr.R087510 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Tan S. X., Fisher-Wellman K. H., Fazakerley D. J., Ng Y., Pant H., Li J., Meoli C. C., Coster A. C., Stöckli J., and James D. E. (2015) Selective insulin resistance in adipocytes. J. Biol. Chem. 290, 11337–11348 10.1074/jbc.M114.623686 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Fujita K. A., Toyoshima Y., Uda S., Ozaki Y., Kubota H., and Kuroda S. (2010) Decoupling of receptor and downstream signals in the Akt pathway by its low-pass filter characteristics. Sci. Signal. 3, ra56 [DOI] [PubMed] [Google Scholar]
- 55. Kubota H., Noguchi R., Toyoshima Y., Ozaki Y., Uda S., Watanabe K., Ogawa W., and Kuroda S. (2012) Temporal coding of insulin action through multiplexing of the AKT pathway. Mol. Cell 46, 820–832 10.1016/j.molcel.2012.04.018 [DOI] [PubMed] [Google Scholar]
- 56. Kubota H., Uda S., Matsuzaki F., Yamauchi Y., and Kuroda S. (2018) In vivo decoding mechanisms of the temporal patterns of blood insulin by the insulin-AKT pathway in the liver. Cell Syst. 7, 118–128.e3 10.1016/j.cels.2018.05.013 [DOI] [PubMed] [Google Scholar]
- 57. Knighton D. R., Zheng J. H., Ten Eyck L. F., Xuong N. H., Taylor S. S., and Sowadski J. M. (1991) Structure of a peptide inhibitor bound to the catalytic subunit of cyclic adenosine monophosphate-dependent protein kinase. Science 253, 414–420 10.1126/science.1862343 [DOI] [PubMed] [Google Scholar]
- 58. Chu N., Salguero A. L., Liu A. Z., Chen Z., Dempsey D. R., Ficarro S. B., Alexander W. M., Marto J. A., Li Y., Amzel L. M., Gabelli S. B., and Cole P. A. (2018) Akt Kinase activation mechanisms revealed using protein semisynthesis. Cell 174, 897–907.e14 10.1016/j.cell.2018.07.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Morrow J. K., Du-Cuny L., Chen L., Meuillet E. J., Mash E. A., Powis G., and Zhang S. (2011) Recent development of anticancer therapeutics targeting Akt. Recent Pat. Anticancer Drug Discov. 6, 146–159 10.2174/157489211793980079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Krycer J. R., and Brown A. J. (2011) Cross-talk between the androgen receptor and the liver X receptor: implications for cholesterol homeostasis. J. Biol. Chem. 286, 20637–20647 10.1074/jbc.M111.227082 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Gibson D. G., Young L., Chuang R.-Y., Venter J. C., Hutchison C. A. 3rd, and Smith H. O. (2009) Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat. Methods 6, 343–345 10.1038/nmeth.1318 [DOI] [PubMed] [Google Scholar]
- 62. Gross S. M., and Rotwein P. (2015) Akt signaling dynamics in individual cells. J. Cell Sci. 128, 2509–2519 10.1242/jcs.168773 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Burchfield J. G., Lu J., Fazakerley D. J., Tan S. X., Ng Y., Mele K., Buckley M. J., Han W., Hughes W. E., and James D. E. (2013) Novel systems for dynamically assessing insulin action in live cells reveals heterogeneity in the insulin response. Traffic 14, 259–273 10.1111/tra.12035 [DOI] [PubMed] [Google Scholar]
- 64. Stöckli J., Zadoorian A., Cooke K. C., Deshpande V., Yau B., Herrmann G., Kebede M. A., Humphrey S. J., and James D. E. (2019) ABHD15 regulates adipose tissue lipolysis and hepatic lipid accumulation. Mol. Metab. 25, 83–94 10.1016/j.molmet.2019.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Fazakerley D. J., Naghiloo S., Chaudhuri R., Koumanov F., Burchfield J. G., Thomas K. C., Krycer J. R., Prior M. J., Parker B. L., Murrow B. A., Stöckli J., Meoli C. C., Holman G. D., and James D. E. (2015) Proteomic analysis of GLUT4 storage vesicles reveals tumor suppressor candidate 5 (TUSC5) as a novel regulator of insulin action in adipocytes. J. Biol. Chem. 290, 23528–23542 10.1074/jbc.M115.657361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Schindelin J., Arganda-Carreras I., Frise E., Kaynig V., Longair M., Pietzsch T., Preibisch S., Rueden C., Saalfeld S., Schmid B., Tinevez J. Y., White D. J., Hartenstein V., Eliceiri K., Tomancak P., and Cardona A. (2012) Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 10.1038/nmeth.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Duan X., Krycer J. R., Cooke K. C., Yang G., James D. E., and Fazakerley D. J. (2018) Membrane topology of trafficking regulator of GLUT4 1 (TRARG1). Biochemistry 57, 3606–3615 10.1021/acs.biochem.8b00361 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



