PMC:3585731 JSONTXT 30 Projects

Annnotations TAB TSV DIC JSON TextAE

Id Subject Object Predicate Lexical cue Negation Speculation
T728 0-3 Protein denotes Akt
T744 14-17 Protein denotes TNF
T753 43-47 Protein denotes RIP1
T741 43-54 Protein denotes RIP1 Kinase
T765 43-84 Positive_regulation denotes RIP1 Kinase Activation during Necroptosis
T748 415-487 Protein denotes by the necroptosis mediator receptor interacting protein-1 (RIP1) kinase
T736 443-480 Protein denotes receptor interacting protein-1 (RIP1)
T746 475-479 Protein denotes RIP1
T743 502-505 Protein denotes Akt
T742 502-512 Protein denotes Akt kinase
T734 588-592 Protein denotes TNFα
T764 588-603 Gene_expression denotes TNFα production
T758 605-707 Phosphorylation denotes During necroptosis, Akt is activated in a RIP1 dependent fashion through its phosphorylation on Thr308
T757 605-707 Positive_regulation denotes During necroptosis, Akt is activated in a RIP1 dependent fashion through its phosphorylation on Thr308
T729 625-628 Protein denotes Akt
T745 647-661 Protein denotes RIP1 dependent
T737 698-707 Entity denotes on Thr308
T760 709-811 Regulation denotes In L929 cells, this activation requires independent signaling inputs from both growth factors and RIP1
T732 807-811 Protein denotes RIP1
T733 813-816 Protein denotes Akt
T735 870-899 Protein denotes mammalian Target of Rapamycin
T730 900-918 Entity denotes complex 1 (mTORC1)
T738 911-917 Entity denotes mTORC1
T750 920-923 Protein denotes Akt
T751 959-965 Entity denotes mTORC1
T763 959-1028 Binding denotes mTORC1, links RIP1 to JNK activation and autocrine production of TNFα
T752 973-977 Protein denotes RIP1
T740 981-984 Protein denotes JNK
T762 981-995 Positive_regulation denotes JNK activation
T761 1000-1028 Gene_expression denotes autocrine production of TNFα
T756 1021-1028 Protein denotes of TNFα
T731 1033-1058 Entity denotes other cell types, such as
T739 1033-1081 Entity denotes other cell types, such as mouse lung fibroblasts
T755 1086-1098 Entity denotes macrophages,
T749 1099-1102 Protein denotes Akt
T759 1126-1164 Gene_expression denotes necroptosis-associated TNFα production
T747 1149-1153 Protein denotes TNFα
T754 1290-1303 Protein denotes of Akt kinase
T727 1293-1296 Protein denotes Akt
T2758 1601-1708 Entity denotes retinal ischemia-reperfusion injury, acute pancreatitis, brain trauma, retinal detachment, and Huntington’s
T2805 1768-1786 Binding denotes linked necroptosis
T2779 2079-2088 Entity denotes A complex
T2745 2115-2122 Entity denotes Ser/Thr
T2742 2115-2122 Entity denotes Ser/Thr
T2776 2115-2146 Protein denotes Ser/Thr kinases, RIP1 and RIP3,
T2783 2119-2122 Entity denotes Thr
T2729 2132-2136 Protein denotes RIP1
T2732 2141-2145 Protein denotes RIP3
T2762 2308-2345 Entity denotes by the pan-caspase inhibitor zVAD.fmk
T2737 2308-2345 Protein denotes by the pan-caspase inhibitor zVAD.fmk
T2756 2327-2336 Entity denotes inhibitor
T2780 2349-2378 Entity denotes mouse fibrosarcoma L929 cells
T2744 2349-2378 Entity denotes mouse fibrosarcoma L929 cells
T2736 2429-2474 Protein denotes 432 genes that may regulate this process [10]
T2740 2757-2761 Protein denotes RIP1
T2775 2766-2770 Protein denotes RIP3
T2739 2869-2872 Protein denotes JNK
T2785 2869-2879 Protein denotes JNK kinase
T2799 2869-2890 Positive_regulation denotes JNK kinase activation
T2730 2964-2968 Protein denotes RIP1
T2760 2964-2975 Protein denotes RIP1 kinase
T2767 2990-3060 Protein denotes the transcription factor c-Jun, a key cellular target of JNK activity,
T2782 2994-3014 Protein denotes transcription factor
T2753 3047-3050 Protein denotes JNK
T2755 3102-3105 Protein denotes RNA
T2796 3119-3150 Positive_regulation denotes Activation of JNK in L929 cells
T2795 3119-3254 Binding denotes Activation of JNK in L929 cells has been linked to autocrine TNFα synthesis, activation of oxidative stress and induction of autophagy,
T2750 3130-3136 Protein denotes of JNK
T2733 3167-3254 Protein denotes to autocrine TNFα synthesis, activation of oxidative stress and induction of autophagy,
T7185 3308-3312 Protein denotes RIP1
T2771 3308-3312 Protein denotes RIP1
T7201 3308-10171 Protein denotes RIP1 kinase dependent activation of JNK and TNFα production has recently been described to be independent of its role in necroptosis [15]. Curiously, Akt kinase, a key pro-survival molecule and a well-established inhibitor of apoptotic cell death, has also recently been linked to necroptosis in L929 cells [16], where insulin-dependent activation of Akt was suggested to promote necroptosis by suppressing autophagy. This conclusion was unexpected, since several reports from different groups, including ours, have established that autophagy promotes, rather than suppresses, zVAD.fmk-induced necroptosis in L929 cells [11], [14], [17]. This raised the possibility that Akt controls more general mechanisms that contribute to the execution of necroptosis. Furthermore, the key question of whether insulin-dependent Akt activity solely provides an environment conducive for necroptosis or if Akt activation is an intrinsic component of necroptosis signaling that is linked to RIP1 kinase has not been explored. In this study, we expanded these observations to delineate the specific contributions and molecular ordering of the Akt and JNK pathways downstream from RIP1 kinase during necroptosis. Our data reveal that Akt is activated through RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis
T2791 3313-3329 Protein denotes kinase dependent
T2734 3344-3347 Protein denotes JNK
T2746 3352-3356 Protein denotes TNFα
T2800 3352-3367 Gene_expression denotes TNFα production
T2770 3458-3461 Protein denotes Akt
T2728 3458-3469 Protein denotes Akt kinase,
T2757 3489-3497 Entity denotes molecule
T2747 3521-3555 Entity denotes inhibitor of apoptotic cell death,
T2790 3627-3644 Protein denotes insulin-dependent
T2807 3627-3662 Positive_regulation denotes insulin-dependent activation of Akt
T2798 3627-3662 Regulation denotes insulin-dependent activation of Akt
T2788 3656-3662 Protein denotes of Akt
T2772 3885-3901 Entity denotes zVAD.fmk-induced
T2748 3979-3982 Protein denotes Akt
T2763 4106-4123 Protein denotes insulin-dependent
T2806 4106-4136 Regulation denotes insulin-dependent Akt activity
T2754 4124-4127 Protein denotes Akt
T2777 4200-4203 Protein denotes Akt
T2801 4200-4214 Positive_regulation denotes Akt activation
T2741 4281-4295 Protein denotes to RIP1 kinase
T2759 4284-4288 Protein denotes RIP1
T2781 4435-4438 Protein denotes Akt
T2731 4443-4446 Protein denotes JNK
T2792 4472-4476 Protein denotes RIP1
T2786 4472-4483 Protein denotes RIP1 kinase
T2803 4520-4636 Positive_regulation denotes that Akt is activated through RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types
T2735 4525-4528 Protein denotes Akt
T15670 4550-4554 Protein denotes RIP1
T2793 4550-4554 Protein denotes RIP1
T15674 4550-4571 Protein denotes RIP1 kinase-dependent
T2804 4550-4636 Regulation denotes RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types
T15756 4550-30854 Phosphorylation denotes RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis Given our observation that growth factors are important for zVAD.fmk induced death, we examined the contribution of several pathways, including MAPK pathways and Akt, which are known to be activated following growth factor receptor activation (Fig. 2A). Inhibition of Akt (Akt inhibitor VIII) strongly protected the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16] as well as cell death triggered by bFGF or IGF-1/zVAD.fmk under serum free conditions (Fig. 2B). Inhibition of Akt also protected the cells from growth-factor insensitive death by caused by TNFα (Fig. 2A). Consistent with previous reports, the JNK inhibitor SP600125 protected the cells from both zVAD.fmk and TNFα induced death (Fig. 2A,B and Fig. S2A) [12], [14]. In contrast, inhibition of two other MAPKs, p38 and ERK, previously reported not to be activated during necroptosis [14], did not protect from either zVAD.fmk or TNFα induced death (Fig. 2A). 10.1371/journal.pone.0056576.g002 Figure 2 Akt contributes to necroptosis induced by zVAD.fmk and TNFα. (A,B) Necroptosis was induced by zVAD.fmk or TNFα (full serum, A) or growth factors/zVAD.fmk (serum free, B) in the presence of inhibitors of Akt (Akt inhibitor VIII), JNK (SP600125), p38 (PD169316), and Erk (UO126). Cell viability was determined after 24 hrs. (C) L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs were treated with zVAD.fmk or TNFα for 9 hrs. Cell viability and Akt expression levels were determined after 24 hrs. In all graphs, average±SD was plotted. Next, we used two approaches to further validate the role of Akt in necroptotic cell death. First, two additional Akt inhibitors, a highly specific, allosteric kinase inhibitor MK-2206 [25] and triciribine (TCN) [26], which blocks membrane translocation of Akt, both attenuated cell death (Fig. S2B). Secondly, simultaneous knockdown of Akt isoforms Akt1 and Akt2 using siRNAs protected cells from necroptosis induced by both zVAD.fmk and TNFα (Fig. 2C). No expression of Akt3 was seen in L929 cells (Fig. S2C) and, consistently, Akt3 siRNA had no additional effect on necroptosis. Our results confirmed that Akt plays a key role in necroptosis induced by multiple stimuli in L929 cells. To understand the activation of Akt and JNK under necroptotic conditions, we examined the changes in Akt and JNK phosphorylation at 9 hrs post zVAD.fmk and TNFα stimulation. This time point was chosen because it reflects the early stage of cell death in our system (Fig. S3A, B). Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation at a known major activation site, Thr308 (Fig. 3A). Interestingly, we did not observe concomitant phosphorylation changes in the second major activation site of Akt, Ser473. We also observed an increase in the phosphorylation of both the p46 and p54 isoforms of JNK and its major substrate c-Jun (Fig. 3A). These data indicate that both Akt and JNK are activated under necroptotic conditions. 10.1371/journal.pone.0056576.g003 Figure 3 RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr, followed by western blotting with indicated antibodies. (B,C) L929 cells were treated with zVAD.fmk (B) or bFGF/zVAD.fmk (serum free conditions, C) and samples were collected at the indicated time points for western blot. (D) Nec-1 was added to the cells stimulated with bFGF or bFGF/zVAD (serum free conditions) for 15 min or 9 hr followed by western blot with the indicated antibodies. The RIP1 kinase inhibitor, Nec-1, completely prevented the increase in Thr308 Akt phosphorylation, while Nec-1i did not (Fig. 3A, Fig. S1D). Similarly, Nec-1 prevented the induction of JNK phosphorylation in response to zVAD.fmk and substantially reduced this change after TNFα addition. We observed some changes in total protein levels of JNK and c-Jun following necroptotic stimulation. Some of these changes, e.g. zVAD.fmk-induced increase in c-Jun, were also attenuated by Nec-1. Importantly, Nec-1 did not alter the basal phosphorylation levels of either Akt or JNK (Fig. 3A). This established that Akt Thr308 and JNK phosphorylation during necroptosis is RIP1 dependent. Interestingly, we discovered that the phosphorylation of Akt Thr308, JNK and Jun are late events following zVAD.fmk stimulation (Fig. 3B) that coincide with the onset of necroptosis at 6 hr post-stimulation (Fig. S3A). To better understand the contributions of growth factors and RIP1 kinase to necroptotic regulation of Akt, we next analyzed the time course of these phosphorylation changes under serum free conditions. We found that the addition of bFGF alone or in combination with zVAD.fmk led to a substantial rapid and transient increase in both Thr308 and Ser473 phosphorylation of Akt as well as JNK and c-Jun at 15 minutes, reflecting the expected response to growth factor stimulation (Fig. 3C). Significantly, the combination of bFGF/zVAD.fmk, but not bFGF alone, also caused a robust, second, delayed increase in the phosphorylation of Thr308, but not Ser473, of Akt as well as a delayed increase in the phosphorylation of JNK and Jun. Furthermore, Nec-1 had no significant effect on the early increase in both Akt and JNK/c-Jun phosphorylation triggered by both bFGF and bFGF/zVAD, while Nec-1, but not its inactive analog Nec-1i (Fig. S1E), efficiently blocked the bFGF/zVAD increase at 6–9 hr (Fig. 3D), suggesting that only the delayed activation of Akt and JNK is specific for necroptosis and dependent on RIP1 kinase activity. Similarly, IGF/zVAD, which also promoted cell death under serum free conditions, produced a delayed increase in Thr308 phosphorylation on Akt, while IGF alone caused solely an early, transient increase in phosphorylation (Fig. S3C). We confirmed the kinetics of the Akt Thr308 and Ser473 phosphorylation changes using a quantitative ELISA assay, which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation (Fig. S3D, E). Taken together, these results indicate that the observed delayed increases in Akt and JNK phosphorylation, preceding the onset of cell death, represent specific consequences of necroptotic signaling downstream from RIP1 kinase. TNFα Induces Delayed Akt Thr308 Phosphorylation and Necroptosis Independent of Growth Factor Stimulation Consistent with TNFα inducing necroptosis independently of growth factors (Fig. 1A), FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation, while efficiently preventing these changes in response to zVAD.fmk (Fig. S4A). Furthermore, addition of TNFα led to comparable late activation of Akt p308 signal under both normal and serum free conditions (Fig. S4B, C), indicating that TNFα signaling to Akt Thr308 is growth factor-independent. In contrast, activation of JNK by TNFα followed different kinetics from zVAD.fmk-induced changes. TNFα treatment caused an early and robust increase in the phosphorylation of JNK and c-Jun. Nec-1 did not affect this early increase, however, it reduced levels of pJNK/Jun at the late, 9 hr time point (Fig. S4B, C). This again separated early RIP1-independent changes, which likely reflect the ability of additional upstream kinases, such as Ask1 to activate JNK [27], from the late RIP1 kinase-dependent necroptotic signaling. Late Increase in Akt Thr308 Phosphorylation Contributes to the Induction of Necroptotic Cell Death We next investigated if the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation functionally contributes to the execution of necroptotic cell death. Firstly, PDGF/zVAD.fmk, which cannot induce necroptosis (Fig. 2A), triggered only the initial, rapid Akt and JNK phosphorylation changes and not the delayed activation (Fig. 4A), indicating that late, rather than early Akt phosphorylation correlates with necroptosis. Secondly, we saw that the ability of the Akt inhibitor to protect cells from necroptosis rapidly declined after 6 hrs of stimulation with zVAD.fmk, TNFα or bFGF/zVAD.fmk and no protection was observed when the inhibitor was added at 9 hrs (Fig. 4B,C). This time frame coincides with the timing of the secondary Akt Thr308 phosphorylation. Finally, we terminated the bFGF signal one hour after addition of bFGF by the addition of PD173074. This allowed us to retain early Akt activation, but to suppress the secondary increase (Fig. 4D). Both pre-addition and delayed addition of PD173074 fully prevented necroptosis (Fig. 4E). Overall, these data, while correlative, indicate that early Akt activation is insufficient to promote necroptosis and are strongly supportive of an important role for the delayed activation of Akt in the induction of necroptotic cell death. 10.1371/journal.pone.0056576.g004 Figure 4 Late Thr308 phosphorylation of Akt contributes to necroptosis. (A) L929 cells were treated with zVAD.fmk and bFGF or PDGF, with or without Nec-1, for the indicated periods of time. (B,C) L929 cells were stimulated by zVAD.fmk or TNFα (B) or bFGF/zVAD.fmk under serum free conditions (C). Akt inh. VIII was added 15 min before necroptotic stimulation (Pre) or at indicated times after stimulation. Viability was measured 24 hr after activation of necroptosis. (D) L929 cells were stimulated with bFGF/zVAD under serum free conditions. PD173074 was added 15 min before or 1 hr after FGF/zVAD. Samples for western blot were collected at 15 min and 9 hr time points. (E) Cells were pretreated with PD173074 or it was added 1 hr after bFGF/zVAD.fmk, followed by viability assessment at 24 hr. In all graphs, average±SD was plotted. The Akt Signaling Pathway Contributes to the Regulation of Necroptosis We next determined whether the necroptosis-associated increase in Thr308 phosphorylation results in an increase in Akt kinase activity. Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates (Forkhead box class O (FoxO) proteins, GSK-3 kinases and mouse double minute 2 (MDM2)) as well as downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6) (Fig. 5A). In some cases (FoxO1, FoxO4, MDM2), a robust increase was observed. In other cases (FoxO3a, GSK-3α/β, p70S6K and its substrate S6), the changes were less pronounced (Fig. 5A). The timing of the phosphorylation changes paralleled the increase in Akt phosphorylation (Fig. 5B, S5A, B). In the case of pFoxO1 we occasionally observed a shift in migration rather than an increase in band intensity (e.g. comparing Fig. 5A and B), suggesting that phosphorylation events in addition to Thr24 take place during necroptosis. Notably, in all cases the necroptosis-associated increases in Akt substrates were abrogated by Nec-1 (Fig. 5A, Fig. S5A, B). Overall, these data suggested that a significant part of the “canonical” Akt signaling network is activated at the onset of necroptotic cell death in a RIP1 dependent fashion. 10.1371/journal.pone.0056576.g005 Figure 5 mTORC1 contributes to the regulation of necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr and harvested for western blot. (B) Cell under serum free condition were treated with bFGF or bFGF/zVAD.fmk for the indicated amounts of time, followed by western blotting using the indicated antibodies. (C) Necroptosis was induced by zVAD.fmk or TNFα in L929 cell in the presence of inhibitors of Akt(Akt inh. VIII) and mTOR (rapamycin, Torin-1 and PI-103). (D) L929 cells with mTOR siRNA knockdown were harvested for western blot or treated with zVAD.fmk or TNFα for 24 hrs. Cell viability was determined 24 hr after activation of necroptosis. In all graphs, average±SD was plotted. Akt kinase is considered to be a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]. An important question is whether these same molecules reverse their pro-survival roles during necroptosis. We found that inhibition of mTORC1 by rapamycin, an inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C). Similarly, the direct mTOR kinase inhibitor Torin1 [30] and the dual PI3K/mTOR inhibitor PI-103 [31] also efficiently inhibited necroptosis (Fig. 5C). Knockdown of mTOR using siRNA further validated the small-molecule inhibitor data indicating a role for mTOR in necroptosis by protecting cells from both zVAD.fmk and TNFα induced death (Fig. 5D). mTORC1 regulates translation through activation of p70S6 kinase and, subsequently, ribosomal protein S6 [32]. Notably, a genome-wide siRNA screen [10] suggested an important role for protein translation in necroptosis. Consistently, we found that the small molecule inhibitor of p70S6K PF-4708671 [33] attenuated necroptosis at the concentrations required to block S6 phosphorylation (Fig. S5C, D). Partial siRNA knockdown of S6 protein attenuated necroptosis as well (Fig. S5E), suggesting that translational control by p70S6K/S6 may play a role in necroptosis. Overall, while the full repertoire of Akt targets during necroptosis remains to be fully explored, our data provide evidence that the activity of an anti-apoptotic branch of Akt signaling can promote necroptosis. RIP1 kinase, Akt, mTORC1 and JNK control the upregulation of TNFα accompanying necroptosis. Hitomi et al. [10] have recently reported that the induction of necroptosis by zVAD.fmk in L929 cells is associated with increased synthesis of TNFα, which potentiates cell death. Therefore, we examined whether Akt and its effectors contribute to TNFα synthesis. Consistent with a RIP1-dependent increase in TNFα protein (Fig. S6A, B), we found that TNFα mRNA levels increased during necroptosis in L929 cells in a RIP1 (Fig. S6C. Under serum free conditions, bFGF alone triggered some induction of TNFα mRNA, while its combination with zVAD.fmk (but not zVAD.fmk alone) caused a pronounced further increase (Fig. 6A). Conversely, PDGF caused a modest upregulation of TNFα mRNA, which was not further increased in the presence of zVAD.fmk (Fig. 6A), demonstrating that activation of necroptosis is specifically accompanied by a marked increase in autocrine TNFα synthesis. 10.1371/journal.pone.0056576.g006 Figure 6 Akt and mTORC1 control autocrine TNFα synthesis and JNK activation during necroptosis. (A) Cells were treated under serum free conditions with bFGF or PDGF with or without zVAD.fmk for 9 hr, followed by qRT-PCR analysis of mTNFα. Data was normalized to mouse 18S RNA. (B) Necroptosis was induced by zVAD.fmk or TNFα in cells treated with Nec-1, rapamycin (rapa), or Akt inh. VIII inh. followed by qRT-PCR analysis of TNFα mRNA levels. (C-F) L929 cells with siRNA knockdown of Akt isoforms (C,E) or mTOR (D,F) were stimulated with zVAD.fmk or TNFα for 9 hr, followed by qRT-PCR analysis of mTNFα (C,D) or western blot (E,F). In all graphs, average±SD was plotted. Further analysis suggested that both Akt and mTORC1 contribute to the upregulation of TNFα mRNA during necroptosis as both small-molecule inhibition and siRNA knockdown of Akt and mTOR reduced TNFα mRNA levels in necroptotic cells (Fig. 6B,C,D). Notably, RIP1 and Akt inhibitors had no effect on the levels of TNFα mRNA in control cells or in the cells stimulated with bFGF alone (Fig. 6A,B, Fig. S6C), suggesting that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis. Akt and mTORC1 Control the Activation of JNK during Necroptosis JNK is a well-established regulator of TNFα synthesis in a variety of systems [13], [14], [15], [34]. Therefore, the ability of Akt and mTORC1 inhibitors to block the increase in TNFα mRNA lead us to examine their role in the activation of JNK during necroptosis. Knockdown of Akt isoforms Akt1 and Akt2 or inhibition of Akt prominently suppressed the necroptosis dependent increase in JNK and c-Jun phosphorylation (Fig. 6E, S6D,E) suggesting that Akt may provide a link between RIP1 and JNK activation. Importantly, inhibition of Akt only inhibited the delayed, but not the early, increase in bFGF/zVAD.fmk induced JNK and c-Jun phosphorylation (Fig. S6F). Knockdown of mTOR, rapamycin and the p70S6K inhibitor PF-4708671 also attenuated the necroptosis-associated increase in JNK and c-Jun phosphorylation (Fig. 6F, S6E,G, Fig. S5D). Overall, these data suggested that the Akt-mTORC1-S6K axis, acting downstream from RIP1 kinase, is required for the increase in JNK activity during necroptosis in L929 cells. PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under Necroptotic Conditions Typical regulation of Akt by growth factors involves its recruitment to the plasma membrane, which is mediated by the binding of the pleckstrin homology (PH) domain of Akt to the product of PI3K, phosphatidylinositol-3,4,5-triphosphate (PIP3). In the membrane, Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1) and mTORC2 (or DNA-PK), respectively [35]. Since our results showed that only Thr308 Akt phosphorylation is increased during necroptosis, we next examined whether it is still dependent on PI3K and PDK1. Inhibition of PI3K and PDK1 using the specific inhibitors LY249002 and BX912 [36] resulted in the efficient inhibition of cell death and Akt Thr308 phosphorylation (Fig. S7A–D). Likewise, siRNA knockdown of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation (Fig. S7E,F) Therefore, PI3K and PDK1 activity is still required for non-canonical Akt activation during necroptosis. Expression of Constitutively Active Akt, Rescues Necroptosis Under Serum Free Conditions We used L929 cells stably expressing constitutively active wild type Akt1 (Myr-Akt) or the catalytically inactive mutant K179M in order to further understand the contribution of growth factors and RIP1 kinase to Akt activation during necroptosis. Constitutively active Akt1 (Myr-Akt) was generated as previously described [37] by the addition of a myristoylation signal which provides constitutive localization to the plasma membrane and by the deletion of the auto-inhibitory PH domain (Fig. 7A) resulting in an Akt that is active under serum free. It is important to note that the cells expressing Myr-Akt were viable, grew in a manner indistinguishable from the empty vector control cells, and were not triggered to induce necroptosis by serum starvation (Fig. 7B). This indicates that active Akt alone is not sufficient to induce necroptotic cell death. Under serum free conditions Myr-Akt, but not the K179M mutant, fully restored zVAD.fmk-induced necroptosis (Fig. 7A,B). Nec-1 prevented both Myr-Akt dependent cell death and the necroptosis-specific delayed increase in Akt Thr308 phosphorylation (Fig. 7B,C). Myr-Akt also allowed other zVAD.fmk-dependent events, including activation of JNK and c-Jun phosphorylation (Fig. 7C) and upregulation of TNFα mRNA (Fig. 7D) to occur under serum free conditions, confirming an important role for Akt at the apex of necroptotic signaling. These data demonstrated that the presence of active and membrane localized Akt is sufficient to uncouple Akt activation during necroptosis from growth factor signaling. RIP1 kinase was still able to regulate Akt activation during necroptosis, suggesting that growth factors and RIP1 kinase provide two independent inputs required for Akt changes during necroptosis. 10.1371/journal.pone.0056576.g007 Figure 7 Over expression of constitutively active Akt restores necroptosis under serum free conditions. (A,B) L929 cells were stably infected with empty MSCV retrovirus or viruses encoding Myr-Akt or the catalytically inactive Myr-Akt K179M. Necroptosis was induced by the addition of zVAD.fmk under serum free conditions (A) or serum or serum free conditions with Nec-1 (B). Viability assays were performed after 24 hr. (C) Myr-Akt and Myr-Akt K179M cells were treated with zVAD.fmk and/or Nec-1 under serum free conditions for 9 hr, followed by western blot using the indicated antibodies. Endogenous Akt (∼) and Myr-Akt (*) bands are indicated. (D) L929 cells, stably infected with Myr-Akt and Myr-Akt K179KM, were stimulated with zVAD.fmk for 9 hr under serum free conditions. TNFα mRNA levels were determined by qRT-PCR and normalized using mouse 18S RNA. (E-G) L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473 were treated with zVAD.fmk under serum free conditions, followed by viability assay at 24 hr (E), western blot at 9 hr (F), or evaluation of TNFα mRNA levels by qRT-PCR at 9 hrs (G). In all graphs, average±SD was plotted. RIP1 kinase-dependent Thr308 phosphorylation of Myr-Akt during necroptosis
T15753 4550-30854 Regulation denotes RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis Given our observation that growth factors are important for zVAD.fmk induced death, we examined the contribution of several pathways, including MAPK pathways and Akt, which are known to be activated following growth factor receptor activation (Fig. 2A). Inhibition of Akt (Akt inhibitor VIII) strongly protected the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16] as well as cell death triggered by bFGF or IGF-1/zVAD.fmk under serum free conditions (Fig. 2B). Inhibition of Akt also protected the cells from growth-factor insensitive death by caused by TNFα (Fig. 2A). Consistent with previous reports, the JNK inhibitor SP600125 protected the cells from both zVAD.fmk and TNFα induced death (Fig. 2A,B and Fig. S2A) [12], [14]. In contrast, inhibition of two other MAPKs, p38 and ERK, previously reported not to be activated during necroptosis [14], did not protect from either zVAD.fmk or TNFα induced death (Fig. 2A). 10.1371/journal.pone.0056576.g002 Figure 2 Akt contributes to necroptosis induced by zVAD.fmk and TNFα. (A,B) Necroptosis was induced by zVAD.fmk or TNFα (full serum, A) or growth factors/zVAD.fmk (serum free, B) in the presence of inhibitors of Akt (Akt inhibitor VIII), JNK (SP600125), p38 (PD169316), and Erk (UO126). Cell viability was determined after 24 hrs. (C) L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs were treated with zVAD.fmk or TNFα for 9 hrs. Cell viability and Akt expression levels were determined after 24 hrs. In all graphs, average±SD was plotted. Next, we used two approaches to further validate the role of Akt in necroptotic cell death. First, two additional Akt inhibitors, a highly specific, allosteric kinase inhibitor MK-2206 [25] and triciribine (TCN) [26], which blocks membrane translocation of Akt, both attenuated cell death (Fig. S2B). Secondly, simultaneous knockdown of Akt isoforms Akt1 and Akt2 using siRNAs protected cells from necroptosis induced by both zVAD.fmk and TNFα (Fig. 2C). No expression of Akt3 was seen in L929 cells (Fig. S2C) and, consistently, Akt3 siRNA had no additional effect on necroptosis. Our results confirmed that Akt plays a key role in necroptosis induced by multiple stimuli in L929 cells. To understand the activation of Akt and JNK under necroptotic conditions, we examined the changes in Akt and JNK phosphorylation at 9 hrs post zVAD.fmk and TNFα stimulation. This time point was chosen because it reflects the early stage of cell death in our system (Fig. S3A, B). Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation at a known major activation site, Thr308 (Fig. 3A). Interestingly, we did not observe concomitant phosphorylation changes in the second major activation site of Akt, Ser473. We also observed an increase in the phosphorylation of both the p46 and p54 isoforms of JNK and its major substrate c-Jun (Fig. 3A). These data indicate that both Akt and JNK are activated under necroptotic conditions. 10.1371/journal.pone.0056576.g003 Figure 3 RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr, followed by western blotting with indicated antibodies. (B,C) L929 cells were treated with zVAD.fmk (B) or bFGF/zVAD.fmk (serum free conditions, C) and samples were collected at the indicated time points for western blot. (D) Nec-1 was added to the cells stimulated with bFGF or bFGF/zVAD (serum free conditions) for 15 min or 9 hr followed by western blot with the indicated antibodies. The RIP1 kinase inhibitor, Nec-1, completely prevented the increase in Thr308 Akt phosphorylation, while Nec-1i did not (Fig. 3A, Fig. S1D). Similarly, Nec-1 prevented the induction of JNK phosphorylation in response to zVAD.fmk and substantially reduced this change after TNFα addition. We observed some changes in total protein levels of JNK and c-Jun following necroptotic stimulation. Some of these changes, e.g. zVAD.fmk-induced increase in c-Jun, were also attenuated by Nec-1. Importantly, Nec-1 did not alter the basal phosphorylation levels of either Akt or JNK (Fig. 3A). This established that Akt Thr308 and JNK phosphorylation during necroptosis is RIP1 dependent. Interestingly, we discovered that the phosphorylation of Akt Thr308, JNK and Jun are late events following zVAD.fmk stimulation (Fig. 3B) that coincide with the onset of necroptosis at 6 hr post-stimulation (Fig. S3A). To better understand the contributions of growth factors and RIP1 kinase to necroptotic regulation of Akt, we next analyzed the time course of these phosphorylation changes under serum free conditions. We found that the addition of bFGF alone or in combination with zVAD.fmk led to a substantial rapid and transient increase in both Thr308 and Ser473 phosphorylation of Akt as well as JNK and c-Jun at 15 minutes, reflecting the expected response to growth factor stimulation (Fig. 3C). Significantly, the combination of bFGF/zVAD.fmk, but not bFGF alone, also caused a robust, second, delayed increase in the phosphorylation of Thr308, but not Ser473, of Akt as well as a delayed increase in the phosphorylation of JNK and Jun. Furthermore, Nec-1 had no significant effect on the early increase in both Akt and JNK/c-Jun phosphorylation triggered by both bFGF and bFGF/zVAD, while Nec-1, but not its inactive analog Nec-1i (Fig. S1E), efficiently blocked the bFGF/zVAD increase at 6–9 hr (Fig. 3D), suggesting that only the delayed activation of Akt and JNK is specific for necroptosis and dependent on RIP1 kinase activity. Similarly, IGF/zVAD, which also promoted cell death under serum free conditions, produced a delayed increase in Thr308 phosphorylation on Akt, while IGF alone caused solely an early, transient increase in phosphorylation (Fig. S3C). We confirmed the kinetics of the Akt Thr308 and Ser473 phosphorylation changes using a quantitative ELISA assay, which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation (Fig. S3D, E). Taken together, these results indicate that the observed delayed increases in Akt and JNK phosphorylation, preceding the onset of cell death, represent specific consequences of necroptotic signaling downstream from RIP1 kinase. TNFα Induces Delayed Akt Thr308 Phosphorylation and Necroptosis Independent of Growth Factor Stimulation Consistent with TNFα inducing necroptosis independently of growth factors (Fig. 1A), FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation, while efficiently preventing these changes in response to zVAD.fmk (Fig. S4A). Furthermore, addition of TNFα led to comparable late activation of Akt p308 signal under both normal and serum free conditions (Fig. S4B, C), indicating that TNFα signaling to Akt Thr308 is growth factor-independent. In contrast, activation of JNK by TNFα followed different kinetics from zVAD.fmk-induced changes. TNFα treatment caused an early and robust increase in the phosphorylation of JNK and c-Jun. Nec-1 did not affect this early increase, however, it reduced levels of pJNK/Jun at the late, 9 hr time point (Fig. S4B, C). This again separated early RIP1-independent changes, which likely reflect the ability of additional upstream kinases, such as Ask1 to activate JNK [27], from the late RIP1 kinase-dependent necroptotic signaling. Late Increase in Akt Thr308 Phosphorylation Contributes to the Induction of Necroptotic Cell Death We next investigated if the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation functionally contributes to the execution of necroptotic cell death. Firstly, PDGF/zVAD.fmk, which cannot induce necroptosis (Fig. 2A), triggered only the initial, rapid Akt and JNK phosphorylation changes and not the delayed activation (Fig. 4A), indicating that late, rather than early Akt phosphorylation correlates with necroptosis. Secondly, we saw that the ability of the Akt inhibitor to protect cells from necroptosis rapidly declined after 6 hrs of stimulation with zVAD.fmk, TNFα or bFGF/zVAD.fmk and no protection was observed when the inhibitor was added at 9 hrs (Fig. 4B,C). This time frame coincides with the timing of the secondary Akt Thr308 phosphorylation. Finally, we terminated the bFGF signal one hour after addition of bFGF by the addition of PD173074. This allowed us to retain early Akt activation, but to suppress the secondary increase (Fig. 4D). Both pre-addition and delayed addition of PD173074 fully prevented necroptosis (Fig. 4E). Overall, these data, while correlative, indicate that early Akt activation is insufficient to promote necroptosis and are strongly supportive of an important role for the delayed activation of Akt in the induction of necroptotic cell death. 10.1371/journal.pone.0056576.g004 Figure 4 Late Thr308 phosphorylation of Akt contributes to necroptosis. (A) L929 cells were treated with zVAD.fmk and bFGF or PDGF, with or without Nec-1, for the indicated periods of time. (B,C) L929 cells were stimulated by zVAD.fmk or TNFα (B) or bFGF/zVAD.fmk under serum free conditions (C). Akt inh. VIII was added 15 min before necroptotic stimulation (Pre) or at indicated times after stimulation. Viability was measured 24 hr after activation of necroptosis. (D) L929 cells were stimulated with bFGF/zVAD under serum free conditions. PD173074 was added 15 min before or 1 hr after FGF/zVAD. Samples for western blot were collected at 15 min and 9 hr time points. (E) Cells were pretreated with PD173074 or it was added 1 hr after bFGF/zVAD.fmk, followed by viability assessment at 24 hr. In all graphs, average±SD was plotted. The Akt Signaling Pathway Contributes to the Regulation of Necroptosis We next determined whether the necroptosis-associated increase in Thr308 phosphorylation results in an increase in Akt kinase activity. Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates (Forkhead box class O (FoxO) proteins, GSK-3 kinases and mouse double minute 2 (MDM2)) as well as downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6) (Fig. 5A). In some cases (FoxO1, FoxO4, MDM2), a robust increase was observed. In other cases (FoxO3a, GSK-3α/β, p70S6K and its substrate S6), the changes were less pronounced (Fig. 5A). The timing of the phosphorylation changes paralleled the increase in Akt phosphorylation (Fig. 5B, S5A, B). In the case of pFoxO1 we occasionally observed a shift in migration rather than an increase in band intensity (e.g. comparing Fig. 5A and B), suggesting that phosphorylation events in addition to Thr24 take place during necroptosis. Notably, in all cases the necroptosis-associated increases in Akt substrates were abrogated by Nec-1 (Fig. 5A, Fig. S5A, B). Overall, these data suggested that a significant part of the “canonical” Akt signaling network is activated at the onset of necroptotic cell death in a RIP1 dependent fashion. 10.1371/journal.pone.0056576.g005 Figure 5 mTORC1 contributes to the regulation of necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr and harvested for western blot. (B) Cell under serum free condition were treated with bFGF or bFGF/zVAD.fmk for the indicated amounts of time, followed by western blotting using the indicated antibodies. (C) Necroptosis was induced by zVAD.fmk or TNFα in L929 cell in the presence of inhibitors of Akt(Akt inh. VIII) and mTOR (rapamycin, Torin-1 and PI-103). (D) L929 cells with mTOR siRNA knockdown were harvested for western blot or treated with zVAD.fmk or TNFα for 24 hrs. Cell viability was determined 24 hr after activation of necroptosis. In all graphs, average±SD was plotted. Akt kinase is considered to be a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]. An important question is whether these same molecules reverse their pro-survival roles during necroptosis. We found that inhibition of mTORC1 by rapamycin, an inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C). Similarly, the direct mTOR kinase inhibitor Torin1 [30] and the dual PI3K/mTOR inhibitor PI-103 [31] also efficiently inhibited necroptosis (Fig. 5C). Knockdown of mTOR using siRNA further validated the small-molecule inhibitor data indicating a role for mTOR in necroptosis by protecting cells from both zVAD.fmk and TNFα induced death (Fig. 5D). mTORC1 regulates translation through activation of p70S6 kinase and, subsequently, ribosomal protein S6 [32]. Notably, a genome-wide siRNA screen [10] suggested an important role for protein translation in necroptosis. Consistently, we found that the small molecule inhibitor of p70S6K PF-4708671 [33] attenuated necroptosis at the concentrations required to block S6 phosphorylation (Fig. S5C, D). Partial siRNA knockdown of S6 protein attenuated necroptosis as well (Fig. S5E), suggesting that translational control by p70S6K/S6 may play a role in necroptosis. Overall, while the full repertoire of Akt targets during necroptosis remains to be fully explored, our data provide evidence that the activity of an anti-apoptotic branch of Akt signaling can promote necroptosis. RIP1 kinase, Akt, mTORC1 and JNK control the upregulation of TNFα accompanying necroptosis. Hitomi et al. [10] have recently reported that the induction of necroptosis by zVAD.fmk in L929 cells is associated with increased synthesis of TNFα, which potentiates cell death. Therefore, we examined whether Akt and its effectors contribute to TNFα synthesis. Consistent with a RIP1-dependent increase in TNFα protein (Fig. S6A, B), we found that TNFα mRNA levels increased during necroptosis in L929 cells in a RIP1 (Fig. S6C. Under serum free conditions, bFGF alone triggered some induction of TNFα mRNA, while its combination with zVAD.fmk (but not zVAD.fmk alone) caused a pronounced further increase (Fig. 6A). Conversely, PDGF caused a modest upregulation of TNFα mRNA, which was not further increased in the presence of zVAD.fmk (Fig. 6A), demonstrating that activation of necroptosis is specifically accompanied by a marked increase in autocrine TNFα synthesis. 10.1371/journal.pone.0056576.g006 Figure 6 Akt and mTORC1 control autocrine TNFα synthesis and JNK activation during necroptosis. (A) Cells were treated under serum free conditions with bFGF or PDGF with or without zVAD.fmk for 9 hr, followed by qRT-PCR analysis of mTNFα. Data was normalized to mouse 18S RNA. (B) Necroptosis was induced by zVAD.fmk or TNFα in cells treated with Nec-1, rapamycin (rapa), or Akt inh. VIII inh. followed by qRT-PCR analysis of TNFα mRNA levels. (C-F) L929 cells with siRNA knockdown of Akt isoforms (C,E) or mTOR (D,F) were stimulated with zVAD.fmk or TNFα for 9 hr, followed by qRT-PCR analysis of mTNFα (C,D) or western blot (E,F). In all graphs, average±SD was plotted. Further analysis suggested that both Akt and mTORC1 contribute to the upregulation of TNFα mRNA during necroptosis as both small-molecule inhibition and siRNA knockdown of Akt and mTOR reduced TNFα mRNA levels in necroptotic cells (Fig. 6B,C,D). Notably, RIP1 and Akt inhibitors had no effect on the levels of TNFα mRNA in control cells or in the cells stimulated with bFGF alone (Fig. 6A,B, Fig. S6C), suggesting that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis. Akt and mTORC1 Control the Activation of JNK during Necroptosis JNK is a well-established regulator of TNFα synthesis in a variety of systems [13], [14], [15], [34]. Therefore, the ability of Akt and mTORC1 inhibitors to block the increase in TNFα mRNA lead us to examine their role in the activation of JNK during necroptosis. Knockdown of Akt isoforms Akt1 and Akt2 or inhibition of Akt prominently suppressed the necroptosis dependent increase in JNK and c-Jun phosphorylation (Fig. 6E, S6D,E) suggesting that Akt may provide a link between RIP1 and JNK activation. Importantly, inhibition of Akt only inhibited the delayed, but not the early, increase in bFGF/zVAD.fmk induced JNK and c-Jun phosphorylation (Fig. S6F). Knockdown of mTOR, rapamycin and the p70S6K inhibitor PF-4708671 also attenuated the necroptosis-associated increase in JNK and c-Jun phosphorylation (Fig. 6F, S6E,G, Fig. S5D). Overall, these data suggested that the Akt-mTORC1-S6K axis, acting downstream from RIP1 kinase, is required for the increase in JNK activity during necroptosis in L929 cells. PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under Necroptotic Conditions Typical regulation of Akt by growth factors involves its recruitment to the plasma membrane, which is mediated by the binding of the pleckstrin homology (PH) domain of Akt to the product of PI3K, phosphatidylinositol-3,4,5-triphosphate (PIP3). In the membrane, Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1) and mTORC2 (or DNA-PK), respectively [35]. Since our results showed that only Thr308 Akt phosphorylation is increased during necroptosis, we next examined whether it is still dependent on PI3K and PDK1. Inhibition of PI3K and PDK1 using the specific inhibitors LY249002 and BX912 [36] resulted in the efficient inhibition of cell death and Akt Thr308 phosphorylation (Fig. S7A–D). Likewise, siRNA knockdown of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation (Fig. S7E,F) Therefore, PI3K and PDK1 activity is still required for non-canonical Akt activation during necroptosis. Expression of Constitutively Active Akt, Rescues Necroptosis Under Serum Free Conditions We used L929 cells stably expressing constitutively active wild type Akt1 (Myr-Akt) or the catalytically inactive mutant K179M in order to further understand the contribution of growth factors and RIP1 kinase to Akt activation during necroptosis. Constitutively active Akt1 (Myr-Akt) was generated as previously described [37] by the addition of a myristoylation signal which provides constitutive localization to the plasma membrane and by the deletion of the auto-inhibitory PH domain (Fig. 7A) resulting in an Akt that is active under serum free. It is important to note that the cells expressing Myr-Akt were viable, grew in a manner indistinguishable from the empty vector control cells, and were not triggered to induce necroptosis by serum starvation (Fig. 7B). This indicates that active Akt alone is not sufficient to induce necroptotic cell death. Under serum free conditions Myr-Akt, but not the K179M mutant, fully restored zVAD.fmk-induced necroptosis (Fig. 7A,B). Nec-1 prevented both Myr-Akt dependent cell death and the necroptosis-specific delayed increase in Akt Thr308 phosphorylation (Fig. 7B,C). Myr-Akt also allowed other zVAD.fmk-dependent events, including activation of JNK and c-Jun phosphorylation (Fig. 7C) and upregulation of TNFα mRNA (Fig. 7D) to occur under serum free conditions, confirming an important role for Akt at the apex of necroptotic signaling. These data demonstrated that the presence of active and membrane localized Akt is sufficient to uncouple Akt activation during necroptosis from growth factor signaling. RIP1 kinase was still able to regulate Akt activation during necroptosis, suggesting that growth factors and RIP1 kinase provide two independent inputs required for Akt changes during necroptosis. 10.1371/journal.pone.0056576.g007 Figure 7 Over expression of constitutively active Akt restores necroptosis under serum free conditions. (A,B) L929 cells were stably infected with empty MSCV retrovirus or viruses encoding Myr-Akt or the catalytically inactive Myr-Akt K179M. Necroptosis was induced by the addition of zVAD.fmk under serum free conditions (A) or serum or serum free conditions with Nec-1 (B). Viability assays were performed after 24 hr. (C) Myr-Akt and Myr-Akt K179M cells were treated with zVAD.fmk and/or Nec-1 under serum free conditions for 9 hr, followed by western blot using the indicated antibodies. Endogenous Akt (∼) and Myr-Akt (*) bands are indicated. (D) L929 cells, stably infected with Myr-Akt and Myr-Akt K179KM, were stimulated with zVAD.fmk for 9 hr under serum free conditions. TNFα mRNA levels were determined by qRT-PCR and normalized using mouse 18S RNA. (E-G) L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473 were treated with zVAD.fmk under serum free conditions, followed by viability assay at 24 hr (E), western blot at 9 hr (F), or evaluation of TNFα mRNA levels by qRT-PCR at 9 hrs (G). In all graphs, average±SD was plotted. RIP1 kinase-dependent Thr308 phosphorylation of Myr-Akt during necroptosis
T15763 4550-30891 Positive_regulation denotes RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis Given our observation that growth factors are important for zVAD.fmk induced death, we examined the contribution of several pathways, including MAPK pathways and Akt, which are known to be activated following growth factor receptor activation (Fig. 2A). Inhibition of Akt (Akt inhibitor VIII) strongly protected the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16] as well as cell death triggered by bFGF or IGF-1/zVAD.fmk under serum free conditions (Fig. 2B). Inhibition of Akt also protected the cells from growth-factor insensitive death by caused by TNFα (Fig. 2A). Consistent with previous reports, the JNK inhibitor SP600125 protected the cells from both zVAD.fmk and TNFα induced death (Fig. 2A,B and Fig. S2A) [12], [14]. In contrast, inhibition of two other MAPKs, p38 and ERK, previously reported not to be activated during necroptosis [14], did not protect from either zVAD.fmk or TNFα induced death (Fig. 2A). 10.1371/journal.pone.0056576.g002 Figure 2 Akt contributes to necroptosis induced by zVAD.fmk and TNFα. (A,B) Necroptosis was induced by zVAD.fmk or TNFα (full serum, A) or growth factors/zVAD.fmk (serum free, B) in the presence of inhibitors of Akt (Akt inhibitor VIII), JNK (SP600125), p38 (PD169316), and Erk (UO126). Cell viability was determined after 24 hrs. (C) L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs were treated with zVAD.fmk or TNFα for 9 hrs. Cell viability and Akt expression levels were determined after 24 hrs. In all graphs, average±SD was plotted. Next, we used two approaches to further validate the role of Akt in necroptotic cell death. First, two additional Akt inhibitors, a highly specific, allosteric kinase inhibitor MK-2206 [25] and triciribine (TCN) [26], which blocks membrane translocation of Akt, both attenuated cell death (Fig. S2B). Secondly, simultaneous knockdown of Akt isoforms Akt1 and Akt2 using siRNAs protected cells from necroptosis induced by both zVAD.fmk and TNFα (Fig. 2C). No expression of Akt3 was seen in L929 cells (Fig. S2C) and, consistently, Akt3 siRNA had no additional effect on necroptosis. Our results confirmed that Akt plays a key role in necroptosis induced by multiple stimuli in L929 cells. To understand the activation of Akt and JNK under necroptotic conditions, we examined the changes in Akt and JNK phosphorylation at 9 hrs post zVAD.fmk and TNFα stimulation. This time point was chosen because it reflects the early stage of cell death in our system (Fig. S3A, B). Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation at a known major activation site, Thr308 (Fig. 3A). Interestingly, we did not observe concomitant phosphorylation changes in the second major activation site of Akt, Ser473. We also observed an increase in the phosphorylation of both the p46 and p54 isoforms of JNK and its major substrate c-Jun (Fig. 3A). These data indicate that both Akt and JNK are activated under necroptotic conditions. 10.1371/journal.pone.0056576.g003 Figure 3 RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr, followed by western blotting with indicated antibodies. (B,C) L929 cells were treated with zVAD.fmk (B) or bFGF/zVAD.fmk (serum free conditions, C) and samples were collected at the indicated time points for western blot. (D) Nec-1 was added to the cells stimulated with bFGF or bFGF/zVAD (serum free conditions) for 15 min or 9 hr followed by western blot with the indicated antibodies. The RIP1 kinase inhibitor, Nec-1, completely prevented the increase in Thr308 Akt phosphorylation, while Nec-1i did not (Fig. 3A, Fig. S1D). Similarly, Nec-1 prevented the induction of JNK phosphorylation in response to zVAD.fmk and substantially reduced this change after TNFα addition. We observed some changes in total protein levels of JNK and c-Jun following necroptotic stimulation. Some of these changes, e.g. zVAD.fmk-induced increase in c-Jun, were also attenuated by Nec-1. Importantly, Nec-1 did not alter the basal phosphorylation levels of either Akt or JNK (Fig. 3A). This established that Akt Thr308 and JNK phosphorylation during necroptosis is RIP1 dependent. Interestingly, we discovered that the phosphorylation of Akt Thr308, JNK and Jun are late events following zVAD.fmk stimulation (Fig. 3B) that coincide with the onset of necroptosis at 6 hr post-stimulation (Fig. S3A). To better understand the contributions of growth factors and RIP1 kinase to necroptotic regulation of Akt, we next analyzed the time course of these phosphorylation changes under serum free conditions. We found that the addition of bFGF alone or in combination with zVAD.fmk led to a substantial rapid and transient increase in both Thr308 and Ser473 phosphorylation of Akt as well as JNK and c-Jun at 15 minutes, reflecting the expected response to growth factor stimulation (Fig. 3C). Significantly, the combination of bFGF/zVAD.fmk, but not bFGF alone, also caused a robust, second, delayed increase in the phosphorylation of Thr308, but not Ser473, of Akt as well as a delayed increase in the phosphorylation of JNK and Jun. Furthermore, Nec-1 had no significant effect on the early increase in both Akt and JNK/c-Jun phosphorylation triggered by both bFGF and bFGF/zVAD, while Nec-1, but not its inactive analog Nec-1i (Fig. S1E), efficiently blocked the bFGF/zVAD increase at 6–9 hr (Fig. 3D), suggesting that only the delayed activation of Akt and JNK is specific for necroptosis and dependent on RIP1 kinase activity. Similarly, IGF/zVAD, which also promoted cell death under serum free conditions, produced a delayed increase in Thr308 phosphorylation on Akt, while IGF alone caused solely an early, transient increase in phosphorylation (Fig. S3C). We confirmed the kinetics of the Akt Thr308 and Ser473 phosphorylation changes using a quantitative ELISA assay, which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation (Fig. S3D, E). Taken together, these results indicate that the observed delayed increases in Akt and JNK phosphorylation, preceding the onset of cell death, represent specific consequences of necroptotic signaling downstream from RIP1 kinase. TNFα Induces Delayed Akt Thr308 Phosphorylation and Necroptosis Independent of Growth Factor Stimulation Consistent with TNFα inducing necroptosis independently of growth factors (Fig. 1A), FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation, while efficiently preventing these changes in response to zVAD.fmk (Fig. S4A). Furthermore, addition of TNFα led to comparable late activation of Akt p308 signal under both normal and serum free conditions (Fig. S4B, C), indicating that TNFα signaling to Akt Thr308 is growth factor-independent. In contrast, activation of JNK by TNFα followed different kinetics from zVAD.fmk-induced changes. TNFα treatment caused an early and robust increase in the phosphorylation of JNK and c-Jun. Nec-1 did not affect this early increase, however, it reduced levels of pJNK/Jun at the late, 9 hr time point (Fig. S4B, C). This again separated early RIP1-independent changes, which likely reflect the ability of additional upstream kinases, such as Ask1 to activate JNK [27], from the late RIP1 kinase-dependent necroptotic signaling. Late Increase in Akt Thr308 Phosphorylation Contributes to the Induction of Necroptotic Cell Death We next investigated if the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation functionally contributes to the execution of necroptotic cell death. Firstly, PDGF/zVAD.fmk, which cannot induce necroptosis (Fig. 2A), triggered only the initial, rapid Akt and JNK phosphorylation changes and not the delayed activation (Fig. 4A), indicating that late, rather than early Akt phosphorylation correlates with necroptosis. Secondly, we saw that the ability of the Akt inhibitor to protect cells from necroptosis rapidly declined after 6 hrs of stimulation with zVAD.fmk, TNFα or bFGF/zVAD.fmk and no protection was observed when the inhibitor was added at 9 hrs (Fig. 4B,C). This time frame coincides with the timing of the secondary Akt Thr308 phosphorylation. Finally, we terminated the bFGF signal one hour after addition of bFGF by the addition of PD173074. This allowed us to retain early Akt activation, but to suppress the secondary increase (Fig. 4D). Both pre-addition and delayed addition of PD173074 fully prevented necroptosis (Fig. 4E). Overall, these data, while correlative, indicate that early Akt activation is insufficient to promote necroptosis and are strongly supportive of an important role for the delayed activation of Akt in the induction of necroptotic cell death. 10.1371/journal.pone.0056576.g004 Figure 4 Late Thr308 phosphorylation of Akt contributes to necroptosis. (A) L929 cells were treated with zVAD.fmk and bFGF or PDGF, with or without Nec-1, for the indicated periods of time. (B,C) L929 cells were stimulated by zVAD.fmk or TNFα (B) or bFGF/zVAD.fmk under serum free conditions (C). Akt inh. VIII was added 15 min before necroptotic stimulation (Pre) or at indicated times after stimulation. Viability was measured 24 hr after activation of necroptosis. (D) L929 cells were stimulated with bFGF/zVAD under serum free conditions. PD173074 was added 15 min before or 1 hr after FGF/zVAD. Samples for western blot were collected at 15 min and 9 hr time points. (E) Cells were pretreated with PD173074 or it was added 1 hr after bFGF/zVAD.fmk, followed by viability assessment at 24 hr. In all graphs, average±SD was plotted. The Akt Signaling Pathway Contributes to the Regulation of Necroptosis We next determined whether the necroptosis-associated increase in Thr308 phosphorylation results in an increase in Akt kinase activity. Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates (Forkhead box class O (FoxO) proteins, GSK-3 kinases and mouse double minute 2 (MDM2)) as well as downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6) (Fig. 5A). In some cases (FoxO1, FoxO4, MDM2), a robust increase was observed. In other cases (FoxO3a, GSK-3α/β, p70S6K and its substrate S6), the changes were less pronounced (Fig. 5A). The timing of the phosphorylation changes paralleled the increase in Akt phosphorylation (Fig. 5B, S5A, B). In the case of pFoxO1 we occasionally observed a shift in migration rather than an increase in band intensity (e.g. comparing Fig. 5A and B), suggesting that phosphorylation events in addition to Thr24 take place during necroptosis. Notably, in all cases the necroptosis-associated increases in Akt substrates were abrogated by Nec-1 (Fig. 5A, Fig. S5A, B). Overall, these data suggested that a significant part of the “canonical” Akt signaling network is activated at the onset of necroptotic cell death in a RIP1 dependent fashion. 10.1371/journal.pone.0056576.g005 Figure 5 mTORC1 contributes to the regulation of necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr and harvested for western blot. (B) Cell under serum free condition were treated with bFGF or bFGF/zVAD.fmk for the indicated amounts of time, followed by western blotting using the indicated antibodies. (C) Necroptosis was induced by zVAD.fmk or TNFα in L929 cell in the presence of inhibitors of Akt(Akt inh. VIII) and mTOR (rapamycin, Torin-1 and PI-103). (D) L929 cells with mTOR siRNA knockdown were harvested for western blot or treated with zVAD.fmk or TNFα for 24 hrs. Cell viability was determined 24 hr after activation of necroptosis. In all graphs, average±SD was plotted. Akt kinase is considered to be a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]. An important question is whether these same molecules reverse their pro-survival roles during necroptosis. We found that inhibition of mTORC1 by rapamycin, an inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C). Similarly, the direct mTOR kinase inhibitor Torin1 [30] and the dual PI3K/mTOR inhibitor PI-103 [31] also efficiently inhibited necroptosis (Fig. 5C). Knockdown of mTOR using siRNA further validated the small-molecule inhibitor data indicating a role for mTOR in necroptosis by protecting cells from both zVAD.fmk and TNFα induced death (Fig. 5D). mTORC1 regulates translation through activation of p70S6 kinase and, subsequently, ribosomal protein S6 [32]. Notably, a genome-wide siRNA screen [10] suggested an important role for protein translation in necroptosis. Consistently, we found that the small molecule inhibitor of p70S6K PF-4708671 [33] attenuated necroptosis at the concentrations required to block S6 phosphorylation (Fig. S5C, D). Partial siRNA knockdown of S6 protein attenuated necroptosis as well (Fig. S5E), suggesting that translational control by p70S6K/S6 may play a role in necroptosis. Overall, while the full repertoire of Akt targets during necroptosis remains to be fully explored, our data provide evidence that the activity of an anti-apoptotic branch of Akt signaling can promote necroptosis. RIP1 kinase, Akt, mTORC1 and JNK control the upregulation of TNFα accompanying necroptosis. Hitomi et al. [10] have recently reported that the induction of necroptosis by zVAD.fmk in L929 cells is associated with increased synthesis of TNFα, which potentiates cell death. Therefore, we examined whether Akt and its effectors contribute to TNFα synthesis. Consistent with a RIP1-dependent increase in TNFα protein (Fig. S6A, B), we found that TNFα mRNA levels increased during necroptosis in L929 cells in a RIP1 (Fig. S6C. Under serum free conditions, bFGF alone triggered some induction of TNFα mRNA, while its combination with zVAD.fmk (but not zVAD.fmk alone) caused a pronounced further increase (Fig. 6A). Conversely, PDGF caused a modest upregulation of TNFα mRNA, which was not further increased in the presence of zVAD.fmk (Fig. 6A), demonstrating that activation of necroptosis is specifically accompanied by a marked increase in autocrine TNFα synthesis. 10.1371/journal.pone.0056576.g006 Figure 6 Akt and mTORC1 control autocrine TNFα synthesis and JNK activation during necroptosis. (A) Cells were treated under serum free conditions with bFGF or PDGF with or without zVAD.fmk for 9 hr, followed by qRT-PCR analysis of mTNFα. Data was normalized to mouse 18S RNA. (B) Necroptosis was induced by zVAD.fmk or TNFα in cells treated with Nec-1, rapamycin (rapa), or Akt inh. VIII inh. followed by qRT-PCR analysis of TNFα mRNA levels. (C-F) L929 cells with siRNA knockdown of Akt isoforms (C,E) or mTOR (D,F) were stimulated with zVAD.fmk or TNFα for 9 hr, followed by qRT-PCR analysis of mTNFα (C,D) or western blot (E,F). In all graphs, average±SD was plotted. Further analysis suggested that both Akt and mTORC1 contribute to the upregulation of TNFα mRNA during necroptosis as both small-molecule inhibition and siRNA knockdown of Akt and mTOR reduced TNFα mRNA levels in necroptotic cells (Fig. 6B,C,D). Notably, RIP1 and Akt inhibitors had no effect on the levels of TNFα mRNA in control cells or in the cells stimulated with bFGF alone (Fig. 6A,B, Fig. S6C), suggesting that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis. Akt and mTORC1 Control the Activation of JNK during Necroptosis JNK is a well-established regulator of TNFα synthesis in a variety of systems [13], [14], [15], [34]. Therefore, the ability of Akt and mTORC1 inhibitors to block the increase in TNFα mRNA lead us to examine their role in the activation of JNK during necroptosis. Knockdown of Akt isoforms Akt1 and Akt2 or inhibition of Akt prominently suppressed the necroptosis dependent increase in JNK and c-Jun phosphorylation (Fig. 6E, S6D,E) suggesting that Akt may provide a link between RIP1 and JNK activation. Importantly, inhibition of Akt only inhibited the delayed, but not the early, increase in bFGF/zVAD.fmk induced JNK and c-Jun phosphorylation (Fig. S6F). Knockdown of mTOR, rapamycin and the p70S6K inhibitor PF-4708671 also attenuated the necroptosis-associated increase in JNK and c-Jun phosphorylation (Fig. 6F, S6E,G, Fig. S5D). Overall, these data suggested that the Akt-mTORC1-S6K axis, acting downstream from RIP1 kinase, is required for the increase in JNK activity during necroptosis in L929 cells. PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under Necroptotic Conditions Typical regulation of Akt by growth factors involves its recruitment to the plasma membrane, which is mediated by the binding of the pleckstrin homology (PH) domain of Akt to the product of PI3K, phosphatidylinositol-3,4,5-triphosphate (PIP3). In the membrane, Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1) and mTORC2 (or DNA-PK), respectively [35]. Since our results showed that only Thr308 Akt phosphorylation is increased during necroptosis, we next examined whether it is still dependent on PI3K and PDK1. Inhibition of PI3K and PDK1 using the specific inhibitors LY249002 and BX912 [36] resulted in the efficient inhibition of cell death and Akt Thr308 phosphorylation (Fig. S7A–D). Likewise, siRNA knockdown of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation (Fig. S7E,F) Therefore, PI3K and PDK1 activity is still required for non-canonical Akt activation during necroptosis. Expression of Constitutively Active Akt, Rescues Necroptosis Under Serum Free Conditions We used L929 cells stably expressing constitutively active wild type Akt1 (Myr-Akt) or the catalytically inactive mutant K179M in order to further understand the contribution of growth factors and RIP1 kinase to Akt activation during necroptosis. Constitutively active Akt1 (Myr-Akt) was generated as previously described [37] by the addition of a myristoylation signal which provides constitutive localization to the plasma membrane and by the deletion of the auto-inhibitory PH domain (Fig. 7A) resulting in an Akt that is active under serum free. It is important to note that the cells expressing Myr-Akt were viable, grew in a manner indistinguishable from the empty vector control cells, and were not triggered to induce necroptosis by serum starvation (Fig. 7B). This indicates that active Akt alone is not sufficient to induce necroptotic cell death. Under serum free conditions Myr-Akt, but not the K179M mutant, fully restored zVAD.fmk-induced necroptosis (Fig. 7A,B). Nec-1 prevented both Myr-Akt dependent cell death and the necroptosis-specific delayed increase in Akt Thr308 phosphorylation (Fig. 7B,C). Myr-Akt also allowed other zVAD.fmk-dependent events, including activation of JNK and c-Jun phosphorylation (Fig. 7C) and upregulation of TNFα mRNA (Fig. 7D) to occur under serum free conditions, confirming an important role for Akt at the apex of necroptotic signaling. These data demonstrated that the presence of active and membrane localized Akt is sufficient to uncouple Akt activation during necroptosis from growth factor signaling. RIP1 kinase was still able to regulate Akt activation during necroptosis, suggesting that growth factors and RIP1 kinase provide two independent inputs required for Akt changes during necroptosis. 10.1371/journal.pone.0056576.g007 Figure 7 Over expression of constitutively active Akt restores necroptosis under serum free conditions. (A,B) L929 cells were stably infected with empty MSCV retrovirus or viruses encoding Myr-Akt or the catalytically inactive Myr-Akt K179M. Necroptosis was induced by the addition of zVAD.fmk under serum free conditions (A) or serum or serum free conditions with Nec-1 (B). Viability assays were performed after 24 hr. (C) Myr-Akt and Myr-Akt K179M cells were treated with zVAD.fmk and/or Nec-1 under serum free conditions for 9 hr, followed by western blot using the indicated antibodies. Endogenous Akt (∼) and Myr-Akt (*) bands are indicated. (D) L929 cells, stably infected with Myr-Akt and Myr-Akt K179KM, were stimulated with zVAD.fmk for 9 hr under serum free conditions. TNFα mRNA levels were determined by qRT-PCR and normalized using mouse 18S RNA. (E-G) L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473 were treated with zVAD.fmk under serum free conditions, followed by viability assay at 24 hr (E), western blot at 9 hr (F), or evaluation of TNFα mRNA levels by qRT-PCR at 9 hrs (G). In all graphs, average±SD was plotted. RIP1 kinase-dependent Thr308 phosphorylation of Myr-Akt during necroptosis increased Myr-Akt activity as it did
T2773 4555-4571 Protein denotes kinase-dependent
T15647 4572-4578 Entity denotes Thr308
T2765 4572-4578 Entity denotes Thr308
T2764 4617-4636 Entity denotes multiple cell types
T2794 4651-4689 Positive_regulation denotes we found that downstream Akt signaling
T2787 4676-4679 Protein denotes Akt
T2778 4699-4704 Entity denotes TORC1
T2738 4709-4711 Protein denotes S6
T2769 4761-4765 Protein denotes TNFα
T2802 4761-4776 Gene_expression denotes TNFα production
T2749 4792-4807 Protein denotes the Akt pathway
T2743 4842-4846 Protein denotes RIP1
T2727 4842-4853 Protein denotes RIP1 kinase
T2752 4858-4861 Protein denotes JNK
T2808 4858-4886 Positive_regulation denotes JNK activation in L929 cells
T2784 4919-4983 Entity denotes multiple other cell types including FADD deficient Jurkat cells,
T2774 4955-4959 Protein denotes FADD
T2768 4955-4982 Entity denotes FADD deficient Jurkat cells
T2751 5026-5048 Entity denotes mouse lung fibroblasts
T2789 5049-5052 Protein denotes Akt
T2766 5076-5080 Protein denotes TNFα
T2797 5076-5091 Gene_expression denotes TNFα production
T2761 5193-5208 Protein denotes the Akt pathway
T4918 5289-5319 Protein denotes Basic Fibroblast Growth Factor
T4984 5289-5320 Localization denotes Basic Fibroblast Growth Factor
T4976 5295-5305 Entity denotes Fibroblast
T4974 5472-5487 Protein denotes TNFα [10], [17]
T4985 5492-5534 Negative_regulation denotes addition, inhibition of caspase-8 activity
T4951 5516-5525 Protein denotes caspase-8
T4933 5585-5611 Protein denotes the pan-caspase inhibitor,
T4964 5585-5611 Entity denotes the pan-caspase inhibitor,
T4934 5612-5621 Entity denotes zVAD.fmk,
T4978 5662-5684 Entity denotes these cells [10], [14]
T4958 5827-5855 Entity denotes of apoptosis inhibitors [17]
T4950 5987-5996 Protein denotes caspase-8
T4961 6001-6016 Protein denotes FADD [18], [19]
T4915 6239-6246 Protein denotes caspase
T4924 6259-6272 Entity denotes healthy cells
T4954 6411-6420 Protein denotes caspase-8
T4991 6431-6500 Negative_regulation denotes which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase
T4990 6431-6500 Negative_regulation denotes which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase
T4963 6461-6465 Protein denotes RIP1
T4931 6461-6472 Protein denotes RIP1 kinase
T4967 6477-6500 Protein denotes the RIP1 deubiquitinase
T4968 6501-6517 Protein denotes CYLD [21], [22],
T4986 6501-6542 Negative_regulation denotes CYLD [21], [22], is removed in L929 cells
T4932 6666-6674 Entity denotes zVAD.fmk
T4981 6699-6749 Entity denotes of growth factors, such as bFGF, restored zVAD.fmk
T4945 6726-6731 Protein denotes bFGF,
T4952 6866-6879 Protein denotes growth factor
T4983 6866-6889 Positive_regulation denotes growth factor signaling
T4928 6920-6924 Protein denotes bFGF
T4973 6929-6934 Protein denotes IGF-1
T4941 6944-6947 Protein denotes EGF
T4965 6997-7020 Protein denotes growth factor-dependent
T4992 7042-7077 Negative_regulation denotes the inhibition of caspase activity,
T4977 7060-7067 Protein denotes caspase
T4966 7081-7085 Protein denotes bFGF
T4923 7142-7146 Protein denotes TNFα
T4917 7266-7274 Entity denotes zVAD.fmk
T4938 7278-7282 Protein denotes TNFα
T28893 7377-7381 Protein denotes bFGF
T28890 7386-7391 Protein denotes IGF-1
T28888 7428-7436 Entity denotes zVAD.fmk
T28905 7471-7475 Protein denotes TNFα
T28898 7479-7487 Entity denotes zVAD.fmk
T28896 7508-7515 Protein denotes 10% FBS
T28902 7591-7624 Protein denotes the CellTiter-Glo Viability assay
T28891 7595-7599 Entity denotes Cell
T28906 7763-7768 Entity denotes Cells
T28900 7787-7796 Entity denotes zVAD.fmk,
T28903 7831-7836 Protein denotes Nec-1
T28892 7869-7917 Protein denotes 24 hrs followed by measurement of cell viability
T28894 7900-7917 Entity denotes of cell viability
T28887 7923-7956 Entity denotes Cells under serum free conditions
T28889 7975-7978 Entity denotes FGF
T28904 7980-7988 Entity denotes zVAD.fmk
T28899 8002-8008 Protein denotes 24 hrs
T28897 8068-8076 Entity denotes zVAD.fmk
T28895 8080-8084 Protein denotes TNFα
T28901 8133-8144 Entity denotes µM PD173074
T4960 8261-8266 Protein denotes Nec-1
T4946 8273-8356 Entity denotes potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]
T4936 8307-8311 Protein denotes RIP1
T4943 8307-8318 Protein denotes RIP1 kinase
T4937 8430-8462 Protein denotes more than 400 human kinases [15]
T4982 8464-8478 Entity denotes This inhibitor
T4975 8593-8601 Entity denotes zVAD.fmk
T4957 8606-8618 Protein denotes TNFα-induced
T4920 8660-8663 Protein denotes Fig
T4962 8704-8711 Protein denotes of RIP1
T4921 8791-8884 Entity denotes an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity
T4980 8853-8857 Protein denotes RIP1
T4925 8853-8875 Protein denotes RIP1 kinase inhibitory
T4929 8929-8933 Entity denotes L929
T4916 8980-8988 Entity denotes zVAD.fmk
T4949 8992-8996 Protein denotes TNFα
T4935 9046-9050 Protein denotes bFGF
T4955 9051-9059 Entity denotes zVAD.fmk
T4942 9091-9095 Protein denotes RIP1
T4972 9091-9102 Protein denotes RIP1 kinase
T4948 9217-9221 Protein denotes bFGF
T4930 9237-9253 Entity denotes zVAD.fmk-induced
T4927 9297-9304 Protein denotes 10% FBS
T4989 9307-9433 Negative_regulation denotes We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling
T4953 9315-9384 Protein denotes two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866),
T4947 9315-9384 Entity denotes two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866),
T4939 9319-9323 Protein denotes bFGF
T4987 9416-9433 Positive_regulation denotes of bFGF signaling
T4922 9419-9423 Protein denotes bFGF
T4970 9453-9461 Entity denotes zVAD.fmk
T4956 9513-9516 Protein denotes Fig
T4959 9544-9548 Protein denotes bFGF
T4979 9549-9557 Protein denotes receptor
T4944 9549-9567 Entity denotes receptor inhibitor
T4919 9590-9602 Protein denotes TNFα-induced
T4971 9616-9619 Protein denotes Fig
T4969 9765-9776 Entity denotes by zVAD.fmk
T4940 9789-9796 Protein denotes by bFGF
T4988 9908-9934 Positive_regulation denotes of growth factor signaling
T4926 9911-9924 Protein denotes growth factor
T7294 10127-10144 Positive_regulation denotes Activation of Akt
T7204 10138-10144 Protein denotes of Akt
T7169 10178-10240 Entity denotes our observation that growth factors are important for zVAD.fmk
T7238 10316-10329 Protein denotes MAPK pathways
T7194 10334-10337 Protein denotes Akt
T7302 10361-10414 Positive_regulation denotes activated following growth factor receptor activation
T7265 10395-10403 Protein denotes receptor
T7225 10416-10419 Protein denotes Fig
T7293 10426-10464 Negative_regulation denotes Inhibition of Akt (Akt inhibitor VIII)
T7268 10437-10443 Protein denotes of Akt
T7262 10445-10463 Entity denotes Akt inhibitor VIII
T7264 10484-10559 Entity denotes the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16]
T7207 10499-10522 Protein denotes growth factor-sensitive
T7182 10543-10559 Entity denotes by zVAD.fmk [16]
T7234 10595-10599 Protein denotes bFGF
T7274 10647-10650 Protein denotes Fig
T7291 10657-10674 Negative_regulation denotes Inhibition of Akt
T7271 10668-10674 Protein denotes of Akt
T7203 10690-10699 Entity denotes the cells
T7239 10705-10730 Protein denotes growth-factor insensitive
T7221 10747-10764 Protein denotes by TNFα (Fig. 2A)
T7183 10800-10807 Protein denotes the JNK
T7155 10808-10817 Entity denotes inhibitor
T7226 10808-10826 Entity denotes inhibitor SP600125
T7151 10837-10846 Entity denotes the cells
T7260 10857-10865 Entity denotes zVAD.fmk
T7275 10870-10874 Protein denotes TNFα
T7259 10904-10907 Protein denotes Fig
T7282 10939-10968 Negative_regulation denotes inhibition of two other MAPKs
T7224 10950-10968 Protein denotes of two other MAPKs
T7250 10970-10973 Protein denotes p38
T7187 10978-11046 Protein denotes ERK, previously reported not to be activated during necroptosis [14]
T7191 11076-11084 Entity denotes zVAD.fmk
T7272 11088-11092 Protein denotes TNFα
T29360 11162-11165 Protein denotes Akt
T29358 11204-11212 Entity denotes zVAD.fmk
T29346 11217-11221 Protein denotes TNFα
T29350 11256-11264 Entity denotes zVAD.fmk
T29362 11268-11272 Protein denotes TNFα
T29368 11307-11315 Entity denotes zVAD.fmk
T29353 11351-11394 Entity denotes inhibitors of Akt (Akt inhibitor VIII), JNK
T29361 11365-11390 Protein denotes Akt (Akt inhibitor VIII),
T29365 11365-11394 Protein denotes Akt (Akt inhibitor VIII), JNK
T29356 11370-11388 Entity denotes Akt inhibitor VIII
T29359 11407-11410 Protein denotes p38
T29345 11427-11430 Protein denotes Erk
T29357 11476-11482 Protein denotes 24 hrs
T29348 11488-11492 Entity denotes L929
T29367 11488-11554 Entity denotes L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs
T29351 11516-11520 Protein denotes Akt1
T29364 11522-11526 Protein denotes Akt2
T29349 11532-11543 Protein denotes Akt3 siRNAs
T29363 11548-11554 Protein denotes 72 hrs
T29352 11573-11581 Entity denotes zVAD.fmk
T29347 11585-11599 Protein denotes TNFα for 9 hrs
T29369 11594-11599 Protein denotes 9 hrs
T29366 11601-11615 Entity denotes Cell viability
T29355 11620-11623 Protein denotes Akt
T29370 11620-11641 Gene_expression denotes Akt expression levels
T29354 11664-11670 Protein denotes 24 hrs
T7177 11769-11775 Protein denotes of Akt
T7180 11810-11839 Entity denotes two additional Akt inhibitors
T7273 11825-11828 Protein denotes Akt
T7252 11850-11900 Entity denotes specific, allosteric kinase inhibitor MK-2206 [25]
T7161 11850-11900 Protein denotes specific, allosteric kinase inhibitor MK-2206 [25]
T7200 11878-11887 Entity denotes inhibitor
T7186 11905-11922 Entity denotes triciribine (TCN)
T7220 11918-11921 Entity denotes TCN
T7289 11929-11972 Negative_regulation denotes which blocks membrane translocation of Akt,
T7184 11942-11950 Entity denotes membrane
T7202 11965-11971 Protein denotes of Akt
T7150 12048-12051 Protein denotes Akt
T7233 12048-12065 Protein denotes Akt isoforms Akt1
T7256 12070-12087 Protein denotes Akt2 using siRNAs
T7217 12098-12103 Entity denotes cells
T7158 12137-12145 Entity denotes zVAD.fmk
T7249 12150-12154 Protein denotes TNFα
T7215 12156-12159 Protein denotes Fig
T7279 12166-12221 Gene_expression denotes No expression of Akt3 was seen in L929 cells (Fig. S2C)
T7176 12180-12187 Protein denotes of Akt3
T7195 12241-12251 Protein denotes Akt3 siRNA
T7228 12320-12323 Protein denotes Akt
T7306 12413-12442 Positive_regulation denotes the activation of Akt and JNK
T7305 12413-12442 Positive_regulation denotes the activation of Akt and JNK
T7190 12431-12434 Protein denotes Akt
T7162 12439-12442 Protein denotes JNK
T7247 12500-12503 Protein denotes Akt
T7165 12508-12511 Protein denotes JNK
T7287 12508-12527 Phosphorylation denotes JNK phosphorylation
T7153 12531-12536 Protein denotes 9 hrs
T7214 12542-12550 Entity denotes zVAD.fmk
T7276 12555-12559 Protein denotes TNFα
T7281 12555-12571 Positive_regulation denotes TNFα stimulation
T7166 12670-12674 Protein denotes S3A,
T7299 12679-12782 Positive_regulation denotes Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation
T7198 12713-12721 Entity denotes zVAD.fmk
T7211 12725-12729 Protein denotes TNFα
T7286 12760-12782 Phosphorylation denotes in Akt phosphorylation
T7240 12763-12766 Protein denotes Akt
T7251 12811-12815 Entity denotes site
T7197 12817-12823 Entity denotes Thr308
T7245 12941-12955 Entity denotes of Akt, Ser473
T7179 12944-12948 Protein denotes Akt,
T7196 13021-13024 Protein denotes p46
T7178 13029-13032 Protein denotes p54
T7181 13045-13048 Protein denotes JNK
T7193 13057-13072 Entity denotes major substrate
T7237 13073-13078 Protein denotes c-Jun
T7160 13080-13083 Protein denotes Fig
T7298 13090-13174 Positive_regulation denotes These data indicate that both Akt and JNK are activated under necroptotic conditions
T7297 13090-13174 Positive_regulation denotes These data indicate that both Akt and JNK are activated under necroptotic conditions
T7241 13120-13123 Protein denotes Akt
T7218 13128-13131 Protein denotes JNK
T29707 13220-13224 Protein denotes RIP1
T29720 13220-13291 Phosphorylation denotes RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
T29719 13220-13291 Phosphorylation denotes RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
T29718 13220-13291 Regulation denotes RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
T29717 13220-13291 Regulation denotes RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
T29709 13225-13241 Protein denotes kinase-dependent
T29706 13261-13264 Protein denotes Akt
T29705 13269-13272 Protein denotes JNK
T29708 13326-13334 Entity denotes zVAD.fmk
T29714 13338-13381 Protein denotes TNFα for 9 hr, followed by western blotting
T29711 13397-13407 Entity denotes antibodies
T29712 13444-13456 Entity denotes zVAD.fmk (B)
T29704 13460-13464 Protein denotes bFGF
T29710 13465-13473 Entity denotes zVAD.fmk
T29716 13579-13584 Protein denotes Nec-1
T29702 13595-13636 Entity denotes to the cells stimulated with bFGF or bFGF
T29713 13624-13628 Protein denotes bFGF
T29715 13632-13636 Protein denotes bFGF
T29703 13729-13739 Entity denotes antibodies
T7210 13741-13767 Protein denotes The RIP1 kinase inhibitor,
T7199 13741-13767 Entity denotes The RIP1 kinase inhibitor,
T7159 13741-13767 Protein denotes The RIP1 kinase inhibitor,
T7253 13768-13773 Protein denotes Nec-1
T7307 13768-13839 Negative_regulation denotes Nec-1, completely prevented the increase in Thr308 Akt phosphorylation,
T7301 13796-13839 Positive_regulation denotes the increase in Thr308 Akt phosphorylation,
T7303 13809-13838 Phosphorylation denotes in Thr308 Akt phosphorylation
T7175 13812-13818 Entity denotes Thr308
T7156 13812-13822 Protein denotes Thr308 Akt
T7267 13893-13898 Protein denotes Nec-1
T7292 13923-13945 Phosphorylation denotes of JNK phosphorylation
T7216 13926-13929 Protein denotes JNK
T7170 13961-13969 Entity denotes zVAD.fmk
T7223 14014-14018 Protein denotes TNFα
T7173 14081-14084 Protein denotes JNK
T7232 14089-14128 Protein denotes c-Jun following necroptotic stimulation
T7255 14158-14174 Entity denotes zVAD.fmk-induced
T7263 14184-14193 Protein denotes in c-Jun,
T7257 14215-14223 Protein denotes by Nec-1
T7154 14238-14243 Protein denotes Nec-1
T7213 14301-14304 Protein denotes Akt
T7269 14308-14321 Protein denotes JNK (Fig. 3A)
T7209 14345-14379 Protein denotes Akt Thr308 and JNK phosphorylation
T7258 14349-14355 Entity denotes Thr308
T7219 14360-14363 Protein denotes JNK
T7296 14360-14379 Phosphorylation denotes JNK phosphorylation
T7231 14402-14416 Protein denotes RIP1 dependent
T7308 14452-14507 Phosphorylation denotes the phosphorylation of Akt Thr308, JNK and Jun are late
T7147 14472-14486 Entity denotes of Akt Thr308,
T7189 14475-14478 Protein denotes Akt
T7235 14487-14490 Protein denotes JNK
T7157 14495-14498 Protein denotes Jun
T7152 14525-14533 Entity denotes zVAD.fmk
T7261 14698-14702 Protein denotes RIP1
T7148 14698-14709 Protein denotes RIP1 kinase
T7304 14713-14742 Regulation denotes necroptotic regulation of Akt
T7254 14736-14742 Protein denotes of Akt
T7244 14866-14873 Protein denotes of bFGF
T7208 14903-14911 Entity denotes zVAD.fmk
T7248 14970-14976 Entity denotes Thr308
T7229 14981-14987 Entity denotes Ser473
T7188 15007-15010 Protein denotes Akt
T7230 15022-15025 Protein denotes JNK
T7172 15030-15050 Protein denotes c-Jun at 15 minutes,
T7285 15658-15695 Positive_regulation denotes the delayed activation of Akt and JNK
T7284 15658-15695 Positive_regulation denotes the delayed activation of Akt and JNK
T7278 15658-15695 Negative_regulation denotes the delayed activation of Akt and JNK
T7277 15658-15695 Negative_regulation denotes the delayed activation of Akt and JNK
T7227 15684-15687 Protein denotes Akt
T7174 15692-15695 Protein denotes JNK
T7246 15728-15761 Protein denotes dependent on RIP1 kinase activity
T7192 15741-15745 Protein denotes RIP1
T7290 15853-15905 Negative_regulation denotes a delayed increase in Thr308 phosphorylation on Akt,
T7280 15853-15905 Positive_regulation denotes a delayed increase in Thr308 phosphorylation on Akt,
T7270 15875-15881 Entity denotes Thr308
T7212 15901-15905 Protein denotes Akt,
T7171 15906-15915 Protein denotes while IGF
T7222 16022-16039 Entity denotes of the Akt Thr308
T7266 16029-16032 Protein denotes Akt
T7242 16044-16050 Entity denotes Ser473
T7300 16109-16218 Positive_regulation denotes which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation
T7295 16127-16218 Regulation denotes a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation
T7283 16127-16218 Negative_regulation denotes a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation
T7164 16165-16179 Protein denotes RIP1-dependent
T7288 16189-16218 Phosphorylation denotes in Akt Thr308 phosphorylation
T7205 16192-16195 Protein denotes Akt
T7236 16192-16202 Entity denotes Akt Thr308
T7206 16220-16223 Protein denotes Fig
T7149 16312-16315 Protein denotes Akt
T7243 16320-16323 Protein denotes JNK
T7309 16320-16340 Phosphorylation denotes JNK phosphorylation,
T7163 16361-16368 Entity denotes of cell
T7167 16449-16453 Protein denotes RIP1
T7168 16449-16460 Protein denotes RIP1 kinase
T8045 16463-16467 Protein denotes TNFα
T8075 16476-16510 Phosphorylation denotes Delayed Akt Thr308 Phosphorylation
T8064 16476-16510 Negative_regulation denotes Delayed Akt Thr308 Phosphorylation
T8042 16476-16510 Protein denotes Delayed Akt Thr308 Phosphorylation
T8032 16488-16494 Entity denotes Thr308
T8071 16539-16567 Positive_regulation denotes of Growth Factor Stimulation
T8061 16542-16555 Protein denotes Growth Factor
T8046 16584-16587 Protein denotes TNF
T8033 16653-16668 Entity denotes FGFR inhibitors
T8073 16653-16737 Negative_regulation denotes FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation true
T8050 16687-16699 Protein denotes TNFα-induced
T8029 16711-16714 Protein denotes Akt
T8053 16718-16721 Protein denotes JNK
T8068 16718-16737 Phosphorylation denotes JNK phosphorylation
T8037 16797-16816 Entity denotes zVAD.fmk (Fig. S4A)
T8035 16840-16847 Protein denotes of TNFα
T8067 16871-16888 Positive_regulation denotes activation of Akt
T8049 16882-16888 Protein denotes of Akt
T8055 16889-16893 Entity denotes p308
T8041 16946-16949 Protein denotes Fig
T8036 16976-16980 Protein denotes TNFα
T8065 16976-16990 Positive_regulation denotes TNFα signaling
T8076 16976-17033 Regulation denotes TNFα signaling to Akt Thr308 is growth factor-independent true
T8056 16994-16997 Protein denotes Akt
T8031 16994-17004 Entity denotes Akt Thr308
T8062 17008-17033 Protein denotes growth factor-independent
T8069 17048-17073 Positive_regulation denotes activation of JNK by TNFα
T8047 17059-17065 Protein denotes of JNK
T8059 17066-17073 Protein denotes by TNFα
T8043 17107-17115 Entity denotes zVAD.fmk
T8057 17133-17137 Protein denotes TNFα
T8077 17133-17154 Regulation denotes TNFα treatment caused
T8063 17168-17213 Positive_regulation denotes robust increase in the phosphorylation of JNK
T8066 17184-17213 Phosphorylation denotes in the phosphorylation of JNK
T8038 17207-17213 Protein denotes of JNK
T8034 17225-17230 Protein denotes Nec-1
T8048 17297-17305 Entity denotes pJNK/Jun
T8040 17298-17305 Protein denotes JNK/Jun
T8039 17298-17305 Protein denotes JNK/Jun
T8030 17302-17305 Protein denotes Jun
T8060 17377-17393 Protein denotes RIP1-independent
T8058 17436-17496 Protein denotes of additional upstream kinases, such as Ask1 to activate JNK
T8054 17459-17475 Protein denotes kinases, such as
T8070 17481-17496 Positive_regulation denotes to activate JNK
T8052 17493-17496 Protein denotes JNK
T8051 17517-17521 Protein denotes RIP1
T8074 17517-17560 Positive_regulation denotes RIP1 kinase-dependent necroptotic signaling
T8072 17517-17560 Regulation denotes RIP1 kinase-dependent necroptotic signaling
T8044 17522-17538 Protein denotes kinase-dependent
T8827 17577-17606 Phosphorylation denotes in Akt Thr308 Phosphorylation
T8793 17580-17583 Protein denotes Akt
T8798 17580-17590 Entity denotes Akt Thr308
T8830 17686-17758 Regulation denotes the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation
T8825 17686-17758 Positive_regulation denotes the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation
T8822 17686-17758 Negative_regulation denotes the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation
T8818 17698-17702 Protein denotes RIP1
T8794 17698-17719 Protein denotes RIP1 kinase-dependent
T8831 17729-17758 Phosphorylation denotes in Akt Thr308 phosphorylation
T8802 17732-17735 Protein denotes Akt
T8797 17732-17742 Entity denotes Akt Thr308
T8799 17910-17932 Protein denotes the initial, rapid Akt
T8796 17937-17940 Protein denotes JNK
T8823 17937-17964 Phosphorylation denotes JNK phosphorylation changes
T8824 17973-18005 Negative_regulation denotes the delayed activation (Fig. 4A)
T8815 18047-18094 Protein denotes Akt phosphorylation correlates with necroptosis
T8807 18130-18150 Entity denotes of the Akt inhibitor
T8795 18137-18140 Protein denotes Akt
T8808 18162-18167 Entity denotes cells
T8821 18208-18228 Protein denotes 6 hrs of stimulation
T8820 18234-18242 Entity denotes zVAD.fmk
T8817 18244-18248 Protein denotes TNFα
T8806 18252-18256 Protein denotes bFGF
T8800 18257-18265 Entity denotes zVAD.fmk
T8812 18302-18315 Entity denotes the inhibitor
T8803 18329-18334 Protein denotes 9 hrs
T8801 18336-18339 Protein denotes Fig
T8810 18393-18417 Entity denotes the secondary Akt Thr308
T8828 18393-18433 Phosphorylation denotes the secondary Akt Thr308 phosphorylation
T8814 18407-18410 Protein denotes Akt
T8804 18458-18466 Protein denotes the bFGF
T8811 18498-18505 Protein denotes of bFGF
T8819 18522-18533 Entity denotes of PD173074
T8805 18567-18570 Protein denotes Akt
T8832 18567-18581 Positive_regulation denotes Akt activation
T8834 18587-18631 Negative_regulation denotes to suppress the secondary increase (Fig. 4D)
T8835 18633-18683 Negative_regulation denotes Both pre-addition and delayed addition of PD173074
T8813 18672-18683 Entity denotes of PD173074
T8816 18783-18786 Protein denotes Akt
T8826 18783-18797 Positive_regulation denotes Akt activation
T8833 18890-18962 Negative_regulation denotes the delayed activation of Akt in the induction of necroptotic cell death
T8829 18890-18962 Positive_regulation denotes the delayed activation of Akt in the induction of necroptotic cell death
T8809 18913-18919 Protein denotes of Akt
T30192 19013-19069 Entity denotes Thr308 phosphorylation of Akt contributes to necroptosis
T30203 19020-19042 Phosphorylation denotes phosphorylation of Akt
T30199 19036-19042 Protein denotes of Akt
T30190 19104-19112 Entity denotes zVAD.fmk
T30189 19104-19121 Entity denotes zVAD.fmk and bFGF
T30187 19125-19129 Entity denotes PDGF
T30201 19147-19152 Protein denotes Nec-1
T30195 19225-19233 Entity denotes zVAD.fmk
T30194 19237-19245 Protein denotes TNFα (B)
T30200 19249-19253 Protein denotes bFGF
T30198 19254-19262 Entity denotes zVAD.fmk
T30196 19296-19299 Protein denotes Akt
T30197 19296-19303 Entity denotes Akt inh
T30191 19305-19309 Protein denotes VIII
T30202 19542-19550 Entity denotes PD173074
T30193 19675-19680 Entity denotes Cells
T30188 19702-19710 Entity denotes PD173074
T10833 19840-19843 Protein denotes Akt
T10932 19934-19995 Positive_regulation denotes the necroptosis-associated increase in Thr308 phosphorylation
T10836 19973-19979 Entity denotes Thr308
T10939 20004-20041 Positive_regulation denotes in an increase in Akt kinase activity
T10881 20022-20025 Protein denotes Akt
T10915 20022-20032 Protein denotes Akt kinase
T10928 20043-20152 Positive_regulation denotes Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates
T10813 20138-20141 Protein denotes Akt
T10852 20176-20180 Entity denotes FoxO
T10883 20182-20239 Protein denotes proteins, GSK-3 kinases and mouse double minute 2 (MDM2))
T10910 20192-20197 Protein denotes GSK-3
T10860 20192-20205 Protein denotes GSK-3 kinases
T10805 20210-20238 Protein denotes mouse double minute 2 (MDM2)
T10844 20233-20237 Protein denotes MDM2
T10847 20251-20318 Entity denotes downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6)
T10827 20273-20314 Protein denotes p70 ribosomal protein S6 Kinase (p70S6K),
T10921 20273-20317 Protein denotes p70 ribosomal protein S6 Kinase (p70S6K), S6
T10808 20306-20312 Protein denotes p70S6K
T10863 20345-20358 Protein denotes FoxO1, FoxO4,
T10895 20345-20363 Protein denotes FoxO1, FoxO4, MDM2
T10911 20352-20357 Protein denotes FoxO4
T10879 20414-20420 Protein denotes FoxO3a
T10893 20422-20430 Protein denotes GSK-3α/β
T10810 20422-20430 Protein denotes GSK-3α/β
T10797 20429-20430 Protein denotes β
T10871 20432-20438 Protein denotes p70S6K
T10865 20443-20459 Protein denotes its substrate S6
T10814 20443-20459 Entity denotes its substrate S6
T10899 20575-20578 Protein denotes Akt
T10926 20575-20604 Phosphorylation denotes Akt phosphorylation (Fig. 5B,
T10896 20629-20635 Protein denotes pFoxO1
T10884 20740-20743 Protein denotes Fig
T10840 20810-20815 Entity denotes Thr24
T10892 20909-20912 Protein denotes Akt
T10902 20909-20923 Entity denotes Akt substrates
T10907 20939-20957 Protein denotes by Nec-1 (Fig. 5A,
T10812 20958-20961 Protein denotes Fig
T10838 21045-21048 Protein denotes Akt
T10886 21124-21128 Protein denotes RIP1
T30618 21193-21198 Entity denotes TORC1
T30611 21278-21286 Entity denotes zVAD.fmk
T30617 21290-21303 Protein denotes TNFα for 9 hr
T30616 21340-21371 Entity denotes Cell under serum free condition
T30629 21390-21394 Protein denotes bFGF
T30623 21398-21402 Protein denotes bFGF
T30613 21403-21411 Entity denotes zVAD.fmk
T30627 21496-21506 Entity denotes antibodies
T30625 21539-21547 Entity denotes zVAD.fmk
T30610 21551-21555 Protein denotes TNFα
T30608 21588-21598 Entity denotes inhibitors
T30621 21602-21605 Protein denotes Akt
T30609 21606-21609 Protein denotes Akt
T30606 21606-21613 Entity denotes Akt inh
T30612 21615-21619 Protein denotes VIII
T30619 21625-21661 Protein denotes mTOR (rapamycin, Torin-1 and PI-103)
T30624 21631-21640 Entity denotes rapamycin
T30614 21642-21649 Entity denotes Torin-1
T30620 21654-21660 Entity denotes PI-103
T30605 21667-21671 Entity denotes L929
T30615 21667-21703 Entity denotes L929 cells with mTOR siRNA knockdown
T30622 21683-21687 Protein denotes mTOR
T30628 21752-21760 Entity denotes zVAD.fmk
T30626 21764-21779 Protein denotes TNFα for 24 hrs
T30607 21773-21779 Protein denotes 24 hrs
T10834 21889-21892 Protein denotes Akt
T10861 21889-21899 Protein denotes Akt kinase
T10839 21920-22048 Protein denotes a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]
T10876 22019-22025 Entity denotes mTORC1
T10822 22027-22032 Protein denotes GSK-3
T10811 22083-22103 Entity denotes these same molecules
T10909 22182-22191 Entity denotes of mTORC1
T10817 22192-22205 Entity denotes by rapamycin,
T10906 22209-22296 Entity denotes inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C)
T10845 22222-22296 Entity denotes the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C)
T10866 22226-22230 Protein denotes mTOR
T10870 22241-22252 Protein denotes Raptor [29]
T10835 22309-22331 Protein denotes the direct mTOR kinase
T10855 22320-22324 Protein denotes mTOR
T10856 22332-22341 Entity denotes inhibitor
T10841 22332-22353 Entity denotes inhibitor Torin1 [30]
T10867 22358-22398 Protein denotes the dual PI3K/mTOR inhibitor PI-103 [31]
T10802 22358-22398 Entity denotes the dual PI3K/mTOR inhibitor PI-103 [31]
T10830 22377-22386 Entity denotes inhibitor
T10816 22459-22475 Entity denotes of mTOR using si
T10848 22462-22466 Protein denotes mTOR
T10920 22475-22478 Protein denotes RNA
T10821 22516-22525 Entity denotes inhibitor
T10882 22553-22557 Protein denotes mTOR
T10831 22587-22592 Entity denotes cells
T10796 22603-22611 Entity denotes zVAD.fmk
T10820 22616-22620 Protein denotes TNFα
T10857 22636-22639 Protein denotes Fig
T10875 22647-22652 Entity denotes TORC1
T10924 22647-22709 Regulation denotes TORC1 regulates translation through activation of p70S6 kinase
T10929 22683-22709 Positive_regulation denotes activation of p70S6 kinase
T10905 22694-22709 Protein denotes of p70S6 kinase
T10917 22729-22754 Protein denotes ribosomal protein S6 [32]
T10903 22829-22836 Protein denotes protein
T10937 22829-22863 Translation denotes protein translation in necroptosis
T10853 22893-22947 Entity denotes the small molecule inhibitor of p70S6K PF-4708671 [33]
T10794 22893-22947 Entity denotes the small molecule inhibitor of p70S6K PF-4708671 [33]
T10919 22925-22931 Protein denotes p70S6K
T10837 22925-22942 Entity denotes p70S6K PF-4708671
T10935 23005-23029 Negative_regulation denotes block S6 phosphorylation
T10823 23011-23013 Protein denotes S6
T10931 23011-23029 Phosphorylation denotes S6 phosphorylation
T10874 23031-23034 Protein denotes Fig
T10916 23069-23124 Protein denotes of S6 protein attenuated necroptosis as well (Fig. S5E)
T10815 23075-23082 Protein denotes protein
T10888 23167-23176 Protein denotes p70S6K/S6
T10798 23167-23176 Protein denotes p70S6K/S6
T10878 23174-23176 Protein denotes S6
T10829 23247-23250 Protein denotes Akt
T10927 23380-23396 Positive_regulation denotes of Akt signaling
T10804 23383-23386 Protein denotes Akt
T10894 23422-23426 Protein denotes RIP1
T10868 23422-23433 Protein denotes RIP1 kinase
T10908 23435-23439 Protein denotes Akt,
T10803 23435-23446 Entity denotes Akt, mTORC1
T10795 23451-23454 Protein denotes JNK
T10933 23463-23512 Positive_regulation denotes the upregulation of TNFα accompanying necroptosis
T10826 23480-23512 Protein denotes of TNFα accompanying necroptosis
T10904 23590-23601 Entity denotes by zVAD.fmk
T10825 23658-23692 Protein denotes TNFα, which potentiates cell death
T10807 23725-23728 Protein denotes Akt
T10862 23761-23775 Protein denotes TNFα synthesis
T10930 23788-23834 Regulation denotes with a RIP1-dependent increase in TNFα protein
T10850 23795-23809 Protein denotes RIP1-dependent
T10887 23819-23834 Protein denotes in TNFα protein
T10901 23822-23826 Protein denotes TNFα
T10889 23859-23923 Protein denotes that TNFα mRNA levels increased during necroptosis in L929 cells
T10828 23869-23873 Protein denotes mRNA
T10824 23929-23933 Protein denotes RIP1
T10880 23935-23938 Protein denotes Fig
T10914 23974-23978 Protein denotes bFGF
T10923 23995-24022 Gene_expression denotes some induction of TNFα mRNA
T10922 23995-24022 Positive_regulation denotes some induction of TNFα mRNA
T10859 24010-24022 Protein denotes of TNFα mRNA
T10854 24013-24017 Protein denotes TNFα
T10897 24051-24059 Entity denotes zVAD.fmk
T10918 24069-24077 Entity denotes zVAD.fmk
T10925 24157-24192 Positive_regulation denotes a modest upregulation of TNFα mRNA,
T10818 24179-24192 Protein denotes of TNFα mRNA,
T10898 24182-24186 Protein denotes TNFα
T10877 24244-24263 Entity denotes zVAD.fmk (Fig. 6A),
T10799 24358-24385 Protein denotes in autocrine TNFα synthesis
T31129 24431-24434 Protein denotes Akt
T31120 24439-24445 Entity denotes mTORC1
T31143 24454-24478 Protein denotes autocrine TNFα synthesis
T31121 24483-24486 Protein denotes JNK
T31152 24483-24516 Positive_regulation denotes JNK activation during necroptosis
T31150 24522-24527 Entity denotes Cells
T31140 24574-24578 Protein denotes bFGF
T31135 24582-24586 Entity denotes PDGF
T31148 24603-24611 Entity denotes zVAD.fmk
T31144 24638-24641 Protein denotes PCR
T31142 24651-24659 Protein denotes of mTNFα
T31147 24681-24697 Protein denotes to mouse 18S RNA
T31123 24690-24693 Protein denotes 18S
T31125 24730-24738 Entity denotes zVAD.fmk
T31131 24742-24746 Protein denotes TNFα
T31128 24750-24793 Entity denotes cells treated with Nec-1, rapamycin (rapa),
T31137 24769-24793 Protein denotes Nec-1, rapamycin (rapa),
T31134 24776-24792 Entity denotes rapamycin (rapa)
T31127 24787-24791 Protein denotes rapa
T31122 24797-24800 Protein denotes Akt
T31145 24797-24804 Entity denotes Akt inh
T31149 24806-24810 Protein denotes VIII
T31136 24806-24814 Entity denotes VIII inh
T31119 24828-24835 Protein denotes qRT-PCR
T31133 24845-24864 Protein denotes of TNFα mRNA levels
T31118 24848-24852 Protein denotes TNFα
T31132 24888-24890 Entity denotes si
T31138 24890-24893 Protein denotes RNA
T31151 24907-24910 Protein denotes Akt
T31141 24929-24937 Protein denotes mTOR (D,
T31139 24929-24938 Entity denotes mTOR (D,F
T31146 24961-24969 Entity denotes zVAD.fmk
T31126 24973-24977 Protein denotes TNFα
T31130 25004-25007 Protein denotes PCR
T31124 25017-25025 Protein denotes of mTNFα
T10846 25131-25134 Protein denotes Akt
T10793 25140-25145 Entity denotes TORC1
T10934 25160-25208 Positive_regulation denotes the upregulation of TNFα mRNA during necroptosis
T10842 25177-25189 Protein denotes of TNFα mRNA
T10858 25180-25184 Protein denotes TNFα
T10806 25217-25231 Entity denotes small-molecule
T10912 25266-25269 Protein denotes Akt
T10872 25274-25278 Protein denotes mTOR
T10832 25287-25291 Protein denotes TNFα
T10851 25287-25303 Protein denotes TNFα mRNA levels
T10819 25307-25324 Entity denotes necroptotic cells
T10843 25326-25329 Protein denotes Fig
T10864 25349-25353 Protein denotes RIP1
T10849 25358-25361 Protein denotes Akt
T10809 25358-25372 Entity denotes Akt inhibitors
T10801 25401-25413 Protein denotes of TNFα mRNA
T10873 25404-25408 Protein denotes TNFα
T10891 25417-25430 Entity denotes control cells
T10869 25437-25457 Entity denotes the cells stimulated
T10890 25463-25467 Protein denotes bFGF
T10913 25475-25478 Protein denotes Fig
T10800 25486-25489 Protein denotes Fig
T10938 25508-25596 Regulation denotes that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis
T10885 25513-25526 Protein denotes these kinases
T10936 25548-25596 Regulation denotes necroptosis-dependent increase in TNFα synthesis
T10900 25579-25596 Protein denotes in TNFα synthesis
T12658 25599-25602 Protein denotes Akt
T12657 25607-25613 Entity denotes mTORC1
T12661 25622-25662 Positive_regulation denotes the Activation of JNK during Necroptosis
T12641 25637-25643 Protein denotes of JNK
T12630 25663-25666 Protein denotes JNK
T12633 25702-25716 Protein denotes TNFα synthesis
T12645 25788-25794 Protein denotes of Akt
T12637 25806-25816 Entity denotes inhibitors
T12666 25820-25851 Negative_regulation denotes block the increase in TNFα mRNA
T12626 25839-25851 Protein denotes in TNFα mRNA
T12631 25842-25846 Protein denotes TNFα
T12668 25885-25925 Positive_regulation denotes the activation of JNK during necroptosis
T12648 25900-25906 Protein denotes of JNK
T12635 25937-25943 Protein denotes of Akt
T12653 25953-25957 Protein denotes Akt1
T12652 25962-25966 Protein denotes Akt2
T12670 25970-25987 Negative_regulation denotes inhibition of Akt
T12640 25981-25987 Protein denotes of Akt
T12647 26049-26052 Protein denotes JNK
T12628 26057-26062 Protein denotes c-Jun
T12663 26057-26078 Phosphorylation denotes c-Jun phosphorylation
T12655 26112-26115 Protein denotes Akt
T12660 26143-26147 Protein denotes RIP1
T12642 26152-26155 Protein denotes JNK
T12669 26152-26166 Positive_regulation denotes JNK activation
T12667 26181-26198 Negative_regulation denotes inhibition of Akt
T12625 26192-26198 Protein denotes of Akt
T12638 26258-26262 Protein denotes bFGF
T12639 26263-26271 Entity denotes zVAD.fmk
T12654 26280-26283 Protein denotes JNK
T12632 26288-26293 Protein denotes c-Jun
T12664 26288-26309 Phosphorylation denotes c-Jun phosphorylation
T12643 26335-26339 Protein denotes mTOR
T12629 26341-26350 Entity denotes rapamycin
T12651 26355-26386 Entity denotes the p70S6K inhibitor PF-4708671
T12636 26355-26386 Entity denotes the p70S6K inhibitor PF-4708671
T12659 26359-26365 Protein denotes p70S6K
T12646 26442-26445 Protein denotes JNK
T12627 26450-26455 Protein denotes c-Jun
T12662 26450-26471 Phosphorylation denotes c-Jun phosphorylation
T12656 26473-26476 Protein denotes Fig
T12644 26486-26492 Protein denotes G, Fig
T12649 26583-26587 Protein denotes RIP1
T12650 26583-26595 Protein denotes RIP1 kinase,
T12665 26612-26673 Positive_regulation denotes the increase in JNK activity during necroptosis in L929 cells
T12634 26628-26631 Protein denotes JNK
T13372 26676-26686 Protein denotes PI3-kinase
T13397 26676-26752 Regulation denotes PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under
T13396 26676-26752 Regulation denotes PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under
T13391 26691-26695 Protein denotes PDK1
T13383 26717-26752 Entity denotes in Akt Thr308 Phosphorylation Under
T13366 26720-26723 Protein denotes Akt
T13368 26795-26801 Protein denotes of Akt
T13381 26909-26919 Protein denotes pleckstrin
T13384 26930-26932 Protein denotes PH
T13386 26934-26971 Entity denotes domain of Akt to the product of PI3K,
T13377 26944-26947 Protein denotes Akt
T13365 26963-26971 Protein denotes of PI3K,
T13364 27013-27017 Entity denotes PIP3
T13362 27023-27035 Entity denotes the membrane
T13373 27037-27040 Protein denotes Akt
T13398 27037-27135 Phosphorylation denotes Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1)
T13361 27062-27068 Entity denotes Thr308
T13385 27073-27079 Entity denotes Ser473
T13379 27080-27135 Protein denotes by 3-phosphoinositide dependent protein kinase-1 (PDK1)
T13369 27130-27134 Protein denotes PDK1
T13393 27214-27220 Entity denotes Thr308
T13370 27214-27224 Protein denotes Thr308 Akt
T13399 27214-27240 Phosphorylation denotes Thr308 Akt phosphorylation
T13367 27324-27328 Protein denotes PI3K
T13378 27333-27337 Protein denotes PDK1
T13403 27339-27366 Negative_regulation denotes Inhibition of PI3K and PDK1
T13402 27339-27366 Negative_regulation denotes Inhibition of PI3K and PDK1
T13382 27353-27357 Protein denotes PI3K
T13392 27362-27366 Protein denotes PDK1
T13376 27476-27479 Protein denotes Akt
T13388 27476-27486 Entity denotes Akt Thr308
T13400 27476-27502 Phosphorylation denotes Akt Thr308 phosphorylation
T13371 27504-27507 Protein denotes Fig
T13389 27527-27529 Entity denotes si
T13359 27529-27532 Protein denotes RNA
T13363 27543-27618 Entity denotes of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation
T13380 27546-27550 Protein denotes PDK1
T13395 27582-27618 Phosphorylation denotes inhibited Akt Thr308 phosphorylation
T13394 27582-27618 Negative_regulation denotes inhibited Akt Thr308 phosphorylation
T13374 27592-27595 Protein denotes Akt
T13390 27592-27602 Entity denotes Akt Thr308
T13375 27620-27623 Protein denotes Fig
T13387 27652-27656 Protein denotes PDK1
T13401 27688-27735 Positive_regulation denotes non-canonical Akt activation during necroptosis
T13360 27702-27705 Protein denotes Akt
T15681 27879-27910 Protein denotes active wild type Akt1 (Myr-Akt)
T15633 27902-27909 Protein denotes Myr-Akt
T15653 27932-27953 Protein denotes inactive mutant K179M
T15624 28024-28028 Protein denotes RIP1
T15637 28024-28035 Protein denotes RIP1 kinase
T15702 28039-28042 Protein denotes Akt
T15757 28039-28053 Positive_regulation denotes Akt activation
T15748 28074-28124 Gene_expression denotes Constitutively active Akt1 (Myr-Akt) was generated
T15634 28089-28110 Protein denotes active Akt1 (Myr-Akt)
T15616 28102-28109 Protein denotes Myr-Akt
T15767 28157-28189 Positive_regulation denotes the addition of a myristoylation
T15743 28241-28260 Entity denotes the plasma membrane
T15693 28268-28306 Protein denotes the deletion of the auto-inhibitory PH
T15722 28307-28323 Entity denotes domain (Fig. 7A)
T15613 28340-28375 Protein denotes Akt that is active under serum free
T15759 28406-28434 Gene_expression denotes the cells expressing Myr-Akt
T15730 28406-28434 Entity denotes the cells expressing Myr-Akt
T15672 28427-28434 Protein denotes Myr-Akt
T15659 28488-28518 Entity denotes the empty vector control cells
T15729 28611-28626 Protein denotes that active Akt
T15667 28713-28747 Entity denotes Myr-Akt, but not the K179M mutant,
T15715 28713-28747 Entity denotes Myr-Akt, but not the K179M mutant,
T15717 28717-28720 Protein denotes Akt
T15704 28730-28746 Protein denotes the K179M mutant
T15657 28763-28779 Entity denotes zVAD.fmk-induced
T15682 28826-28843 Protein denotes Myr-Akt dependent
T15752 28901-28930 Phosphorylation denotes in Akt Thr308 phosphorylation
T15643 28904-28907 Protein denotes Akt
T15640 28904-28914 Entity denotes Akt Thr308
T15627 28932-28935 Protein denotes Fig
T15740 28944-28951 Protein denotes Myr-Akt
T15745 28965-29035 Positive_regulation denotes other zVAD.fmk-dependent events, including activation of JNK and c-Jun
T15744 28965-29035 Positive_regulation denotes other zVAD.fmk-dependent events, including activation of JNK and c-Jun
T15699 28971-28989 Entity denotes zVAD.fmk-dependent
T15660 29022-29025 Protein denotes JNK
T15668 29030-29035 Protein denotes c-Jun
T15651 29053-29056 Protein denotes Fig
T15766 29066-29091 Positive_regulation denotes upregulation of TNFα mRNA
T15697 29079-29091 Protein denotes of TNFα mRNA
T15683 29082-29086 Protein denotes TNFα
T15724 29093-29096 Protein denotes Fig
T15619 29173-29176 Protein denotes Akt
T15655 29180-29213 Entity denotes the apex of necroptotic signaling
T15712 29257-29293 Protein denotes of active and membrane localized Akt
T15735 29271-29289 Entity denotes membrane localized
T15725 29320-29323 Protein denotes Akt
T15765 29320-29334 Positive_regulation denotes Akt activation
T15694 29359-29372 Protein denotes growth factor
T15749 29359-29382 Positive_regulation denotes growth factor signaling
T15690 29384-29388 Protein denotes RIP1
T15641 29384-29395 Protein denotes RIP1 kinase
T15746 29411-29456 Regulation denotes to regulate Akt activation during necroptosis
T15606 29423-29426 Protein denotes Akt
T15758 29423-29437 Positive_regulation denotes Akt activation
T15710 29493-29497 Protein denotes RIP1
T15721 29493-29504 Protein denotes RIP1 kinase
T15649 29549-29552 Protein denotes Akt
T31749 29805-29812 Entity denotes Myr-Akt
T31747 29805-29812 Protein denotes Myr-Akt
T31764 29834-29856 Protein denotes inactive Myr-Akt K179M
T31754 29834-29856 Entity denotes inactive Myr-Akt K179M
T31763 29898-29909 Entity denotes of zVAD.fmk
T31743 29981-29990 Protein denotes Nec-1 (B)
T31778 30041-30048 Entity denotes Myr-Akt
T31739 30041-30048 Protein denotes Myr-Akt
T31776 30053-30072 Entity denotes Myr-Akt K179M cells
T31774 30053-30072 Protein denotes Myr-Akt K179M cells
T31767 30053-30072 Entity denotes Myr-Akt K179M cells
T31755 30091-30099 Entity denotes zVAD.fmk
T31765 30107-30149 Protein denotes Nec-1 under serum free conditions for 9 hr
T31751 30196-30206 Entity denotes antibodies
T31769 30208-30226 Protein denotes Endogenous Akt (∼)
T31760 30231-30238 Protein denotes Myr-Akt
T31748 30231-30238 Entity denotes Myr-Akt
T31761 30301-30308 Protein denotes Myr-Akt
T31746 30301-30308 Entity denotes Myr-Akt
T31780 30313-30328 Entity denotes Myr-Akt K179KM,
T31759 30313-30328 Entity denotes Myr-Akt K179KM,
T31750 30313-30328 Protein denotes Myr-Akt K179KM,
T31741 30350-30367 Entity denotes zVAD.fmk for 9 hr
T31770 30397-30401 Protein denotes TNFα
T31758 30397-30413 Protein denotes TNFα mRNA levels
T31775 30462-30475 Protein denotes mouse 18S RNA
T31773 30468-30471 Protein denotes 18S
T31757 30478-30481 Entity denotes E-G
T31762 30483-30487 Entity denotes L929
T31756 30483-30557 Entity denotes L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473
T31772 30505-30512 Entity denotes Myr-Akt
T31742 30505-30512 Protein denotes Myr-Akt
T31740 30517-30520 Entity denotes Ala
T31745 30525-30528 Protein denotes Asp
T31753 30540-30546 Entity denotes Thr308
T31766 30551-30557 Entity denotes Ser473
T31752 30576-30584 Entity denotes zVAD.fmk
T31768 30678-30679 Entity denotes F
T31779 30696-30726 Protein denotes of TNFα mRNA levels by qRT-PCR
T31777 30699-30703 Protein denotes TNFα
T31744 30719-30726 Protein denotes qRT-PCR
T31771 30730-30739 Protein denotes 9 hrs (G)
T15708 30825-30835 Protein denotes of Myr-Akt
T15612 30865-30872 Protein denotes Myr-Akt
T15685 30908-30911 Protein denotes Akt
T15677 30967-30970 Protein denotes Akt
T15684 30967-30981 Entity denotes Akt substrates
T15764 31001-31053 Gene_expression denotes the expression of Myr-Akt, but not the K179M mutant,
T15610 31016-31026 Protein denotes of Myr-Akt
T15644 31036-31052 Protein denotes the K179M mutant
T15632 31070-31085 Entity denotes these molecules
T15609 31090-31093 Protein denotes Akt
T15630 31090-31104 Entity denotes Akt substrates
T15707 31141-31152 Entity denotes of zVAD.fmk
T15761 31200-31239 Positive_regulation denotes the increased basal activity of Myr-Akt
T15614 31229-31239 Protein denotes of Myr-Akt
T15675 31241-31296 Entity denotes Some substrates, including p70S6K, S6, GSK-3 and FoxO4,
T15714 31268-31274 Protein denotes p70S6K
T15741 31276-31278 Protein denotes S6
T15663 31280-31285 Protein denotes GSK-3
T15698 31290-31295 Protein denotes FoxO4
T15666 31343-31354 Entity denotes of zVAD.fmk
T15751 31375-31408 Phosphorylation denotes phosphorylation of FoxO1 and MDM2
T15750 31375-31408 Phosphorylation denotes phosphorylation of FoxO1 and MDM2
T15755 31375-31465 Positive_regulation denotes phosphorylation of FoxO1 and MDM2 was significantly increased in the presence of zVAD.fmk,
T15754 31375-31465 Positive_regulation denotes phosphorylation of FoxO1 and MDM2 was significantly increased in the presence of zVAD.fmk,
T15656 31394-31399 Protein denotes FoxO1
T15718 31404-31408 Protein denotes MDM2
T15638 31453-31464 Entity denotes of zVAD.fmk
T15771 31482-31527 Phosphorylation denotes necroptotic Thr308 phosphorylation of Myr-Akt
T15706 31494-31500 Entity denotes Thr308
T15631 31517-31527 Protein denotes of Myr-Akt
T15703 31589-31605 Entity denotes zVAD.fmk-induced
T15618 31625-31657 Protein denotes cell death, JNK activation, TNFα
T15747 31625-31668 Gene_expression denotes cell death, JNK activation, TNFα production
T15620 31637-31640 Protein denotes JNK
T15760 31637-31651 Positive_regulation denotes JNK activation
T15648 31707-31714 Protein denotes Myr-Akt
T15701 31764-31767 Protein denotes Akt
T15662 31810-31846 Entity denotes that membrane localization of Akt is
T15692 31837-31843 Protein denotes of Akt
T15686 31857-31861 Protein denotes Full
T15688 31869-31872 Protein denotes Akt
T15671 31898-31911 Entity denotes the PH domain
T15719 31902-31904 Protein denotes PH
T15720 31920-31923 Entity denotes Myr
T15642 31986-31992 Entity denotes Thr308
T15628 32019-32027 Entity denotes zVAD.fmk
T15713 32066-32069 Protein denotes Fig
T15669 32129-32135 Entity denotes Thr308
T15625 32199-32205 Protein denotes of Akt
T15676 32233-32236 Entity denotes Ala
T15652 32250-32256 Entity denotes Thr308
T15673 32261-32267 Entity denotes Ser473
T15608 32312-32318 Protein denotes of Akt
T15691 32320-32337 Protein denotes while Asp mutants
T15654 32388-32391 Entity denotes Ala
T15734 32396-32421 Protein denotes Asp mutants at both sites
T15709 32456-32472 Protein denotes both Asp mutants
T15711 32503-32520 Protein denotes to wild type Akt,
T15696 32532-32543 Entity denotes Ala mutants
T15621 32637-32643 Entity denotes Thr308
T15695 32648-32654 Entity denotes Ser473
T15762 32711-32738 Positive_regulation denotes promote necroptotic changes
T15716 32767-32784 Protein denotes the S473D mutant,
T15733 32813-32819 Entity denotes Thr308
T15639 32890-32901 Protein denotes while S473A
T15617 32963-32968 Protein denotes S473A
T15727 33016-33022 Entity denotes Thr308
T15680 33056-33063 Entity denotes the Ala
T15623 33072-33080 Entity denotes 473 site
T15678 33114-33128 Entity denotes a docking site
T15626 33133-33137 Protein denotes PDK1
T15629 33155-33166 Entity denotes Thr308 [39]
T15664 33185-33188 Entity denotes Ala
T15728 33193-33214 Protein denotes Asp mutants of Thr308
T15700 33208-33214 Entity denotes Thr308
T15769 33271-33303 Phosphorylation denotes phosphorylation of JNK and c-Jun
T15768 33271-33303 Phosphorylation denotes phosphorylation of JNK and c-Jun
T15689 33290-33293 Protein denotes JNK
T15687 33298-33303 Protein denotes c-Jun
T15636 33309-33313 Protein denotes TNFα
T15742 33309-33318 Protein denotes TNFα mRNA
T15665 33339-33388 Protein denotes T308D, in spite of being an active Akt construct,
T15739 33374-33377 Protein denotes Akt
T15736 33402-33434 Entity denotes perfect mimic of phosphorylation
T15723 33456-33469 Protein denotes of the kinase
T15737 33542-33555 Entity denotes of substrates
T15738 33559-33568 Entity denotes the cells
T15622 33583-33588 Protein denotes T308D
T15607 33639-33772 Entity denotes several substrates that were phosphorylated by the Myr-Akt construct in the presence of zVAD including FoxO1, Foxo4, MDM2, and p70S6K
T15646 33690-33697 Protein denotes Myr-Akt
T15611 33742-33747 Protein denotes FoxO1
T15645 33749-33754 Protein denotes Foxo4
T15679 33756-33760 Protein denotes MDM2
T15705 33766-33772 Protein denotes p70S6K
T15658 33774-33777 Protein denotes Fig
T15635 33849-33855 Entity denotes Thr308
T15731 33931-33934 Protein denotes Akt
T15650 33931-33941 Protein denotes Akt kinase
T15732 33952-33955 Protein denotes Akt
T15615 33973-33983 Entity denotes substrates
T15726 34012-34026 Protein denotes TNFα synthesis
T15661 34028-34031 Protein denotes JNK
T15770 34028-34042 Positive_regulation denotes JNK activation
T18066 34069-34072 Protein denotes Akt
T18100 34069-34117 Regulation denotes Akt Controls TNFα Production in Other Cell Types
T18064 34082-34086 Protein denotes TNFα
T18098 34082-34117 Gene_expression denotes TNFα Production in Other Cell Types
T18099 34101-34117 Localization denotes Other Cell Types
T18056 34101-34117 Protein denotes Other Cell Types
T18051 34107-34111 Entity denotes Cell
T18103 34146-34180 Regulation denotes of RIP1 kinase-dependent signaling
T18095 34146-34180 Positive_regulation denotes of RIP1 kinase-dependent signaling
T18035 34149-34153 Protein denotes RIP1
T18038 34154-34170 Protein denotes kinase-dependent
T18062 34184-34201 Protein denotes Akt in L929 cells
T18061 34233-34301 Entity denotes to other cell types that are known to undergo necroptotic cell death
T18082 34303-34306 Protein denotes Fas
T18096 34303-34350 Binding denotes Fas-associated protein with death domain (FADD)
T18039 34318-34325 Protein denotes protein
T18055 34326-34350 Entity denotes with death domain (FADD)
T18084 34345-34349 Protein denotes FADD
T18086 34350-34381 Entity denotes -deficient Jurkat T lymphocytes
T18080 34390-34400 Entity denotes macrophage
T18049 34509-34513 Protein denotes TNFα
T18070 34517-34525 Entity denotes zVAD.fmk
T18106 34569-34641 Regulation denotes a RIP1 kinase dependent increase in the phosphorylation of Thr308 on Akt
T18104 34569-34641 Positive_regulation denotes a RIP1 kinase dependent increase in the phosphorylation of Thr308 on Akt
T18057 34571-34575 Protein denotes RIP1
T18069 34576-34592 Protein denotes kinase dependent
T18052 34625-34634 Entity denotes of Thr308
T18044 34638-34641 Protein denotes Akt
T18058 34687-34703 Entity denotes these cell types
T18063 34718-34722 Protein denotes TNFα
T18092 34753-34777 Entity denotes each of these cell types
T18079 34758-34777 Entity denotes of these cell types
T18093 34831-34835 Protein denotes RIP1
T18077 34840-34843 Protein denotes Akt
T18088 34840-34854 Entity denotes Akt inhibitors
T18105 34879-34896 Negative_regulation denotes inhibition of Akt
T18090 34890-34896 Protein denotes of Akt
T18042 34913-34924 Entity denotes these cells
T18073 35004-35008 Protein denotes TNFα
T18040 35102-35108 Protein denotes of Akt
T18067 35128-35142 Protein denotes by RIP1 kinase
T18087 35131-35135 Protein denotes RIP1
T18083 35146-35165 Entity denotes multiple cell types
T32265 35283-35331 Gene_expression denotes autocrine TNFα production in multiple cell types
T32263 35293-35297 Protein denotes TNFα
T32236 35312-35331 Entity denotes multiple cell types
T32259 35333-35337 Protein denotes FADD
T32264 35333-35360 Entity denotes FADD deficient Jurkat cells
T32245 35379-35414 Protein denotes TNFα followed by measurement of (A)
T32235 35415-35430 Protein denotes human TNFα mRNA
T32253 35421-35425 Protein denotes TNFα
T32241 35441-35448 Protein denotes qRT-PCR
T32251 35470-35483 Protein denotes human 18S RNA
T32252 35476-35479 Protein denotes 18S
T32261 35558-35566 Entity denotes zVAD.fmk
T32247 35628-35658 Protein denotes of TNFα mRNA levels by qRT-PCR
T32244 35631-35635 Protein denotes TNFα
T32239 35651-35658 Protein denotes qRT-PCR
T32257 35665-35666 Entity denotes F
T32242 35694-35697 Protein denotes Akt
T32238 35709-35736 Entity denotes lung fibroblasts expressing
T32250 35737-35744 Entity denotes Myr-Akt
T32262 35737-35744 Protein denotes Myr-Akt
T32248 35772-35780 Entity denotes zVAD.fmk
T32240 35785-35844 Protein denotes TNFα followed by measurement of TNFα mRNA levels by qRT-PCR
T32232 35814-35833 Protein denotes of TNFα mRNA levels
T32256 35822-35826 Protein denotes mRNA
T32234 35837-35844 Protein denotes qRT-PCR
T32233 35855-35856 Entity denotes H
T32260 35869-35920 Entity denotes fibroblasts expressing only endogenous Akt1 or Akt2
T32267 35869-35920 Gene_expression denotes fibroblasts expressing only endogenous Akt1 or Akt2
T32266 35869-35920 Gene_expression denotes fibroblasts expressing only endogenous Akt1 or Akt2
T32258 35892-35912 Protein denotes only endogenous Akt1
T32255 35916-35920 Protein denotes Akt2
T32249 35939-35947 Entity denotes zVAD.fmk
T32246 35952-36011 Protein denotes TNFα followed by measurement of TNFα mRNA levels by qRT-PCR
T32254 35981-36000 Protein denotes of TNFα mRNA levels
T32243 35984-35988 Protein denotes TNFα
T32237 36004-36011 Protein denotes qRT-PCR
T18074 36055-36061 Protein denotes of Akt
T18048 36080-36102 Entity denotes mouse lung fibroblasts
T18050 36104-36186 Entity denotes Lung fibroblasts selected to survive after deletion of all three Akt isoforms [40]
T18071 36169-36172 Protein denotes Akt
T18037 36243-36247 Protein denotes TNFα
T18094 36252-36260 Entity denotes zVAD.fmk
T18078 36290-36310 Protein denotes active Akt (Myr-Akt)
T18089 36302-36309 Entity denotes Myr-Akt
T18059 36302-36309 Protein denotes Myr-Akt
T18041 36314-36325 Entity denotes these cells
T18053 36335-36339 Protein denotes TNFα
T18091 36335-36344 Protein denotes TNFα mRNA
T18101 36335-36355 Gene_expression denotes TNFα mRNA production
T18034 36371-36375 Protein denotes TNFα
T18032 36380-36388 Entity denotes zVAD.fmk
T18045 36390-36393 Protein denotes Fig
T18036 36435-36438 Protein denotes Fig
T18047 36474-36477 Protein denotes Akt
T18108 36499-36545 Gene_expression denotes fibroblasts expressing endogenous Akt1 or Akt2
T18107 36499-36545 Gene_expression denotes fibroblasts expressing endogenous Akt1 or Akt2
T18075 36499-36545 Entity denotes fibroblasts expressing endogenous Akt1 or Akt2
T18043 36522-36537 Protein denotes endogenous Akt1
T18060 36541-36545 Protein denotes Akt2
T18054 36569-36575 Entity denotes Thr308
T18033 36591-36595 Protein denotes TNFα
T18076 36600-36608 Entity denotes zVAD.fmk
T18072 36610-36613 Protein denotes Fig
T18081 36638-36669 Protein denotes robust RIP1-dependent TNFα mRNA
T18102 36638-36682 Positive_regulation denotes robust RIP1-dependent TNFα mRNA upregulation
T18031 36645-36659 Protein denotes RIP1-dependent
T18046 36660-36664 Protein denotes TNFα
T18085 36770-36778 Protein denotes that Akt
T18065 36800-36829 Protein denotes for autocrine TNFα synthesis,
T18068 36926-36938 Protein denotes Akt-mediated
T18097 36926-36990 Regulation denotes Akt-mediated inflammatory signaling under necroptotic conditions
T19845 37024-37028 Protein denotes RIP1
T19840 37030-37033 Protein denotes Akt
T19792 37038-37051 Protein denotes JNK Dependent
T19815 37065-37087 Entity denotes Necroptotic L929 Cells
T19857 37077-37081 Entity denotes L929
T19826 37118-37122 Protein denotes RIP1
T19812 37123-37139 Protein denotes kinase-dependent
T19788 37165-37194 Entity denotes mouse fibrosarcoma L929 cells
T19777 37165-37194 Entity denotes mouse fibrosarcoma L929 cells
T19830 37237-37271 Entity denotes the pan-caspase inhibitor zVAD.fmk
T19823 37237-37271 Protein denotes the pan-caspase inhibitor zVAD.fmk
T19863 37253-37262 Entity denotes inhibitor
T19865 37310-37313 Protein denotes Akt
T19838 37310-37320 Protein denotes Akt kinase
T19852 37374-37378 Protein denotes RIP1
T19822 37374-37386 Protein denotes RIP1 kinase,
T19794 37446-37455 Entity denotes on Thr308
T19804 37465-37471 Entity denotes Ser473
T19888 37506-37551 Phosphorylation denotes necroptosis-associated phosphorylation of Akt
T19786 37545-37551 Protein denotes of Akt
T19834 37645-37664 Entity denotes the plasma membrane
T19877 37678-37684 Protein denotes of Akt
T19841 37717-37734 Entity denotes membrane-targeted
T19839 37735-37738 Protein denotes Akt
T19868 37750-37757 Protein denotes Myr-Akt
T19881 37823-37844 Gene_expression denotes expression of Myr-Akt
T19861 37834-37844 Protein denotes of Myr-Akt
T19801 37913-37917 Protein denotes RIP1
T19785 37918-37934 Protein denotes kinase-dependent
T19835 37957-37963 Entity denotes Thr308
T19897 37957-37986 Phosphorylation denotes Thr308 phosphorylation of Akt
T19784 37980-37986 Protein denotes of Akt
T19781 38002-38009 Protein denotes caspase
T19886 38002-38020 Negative_regulation denotes caspase inhibition
T32557 38138-38142 Protein denotes RIP1
T32554 38144-38147 Protein denotes Akt
T32552 38152-38155 Protein denotes JNK
T32568 38152-38175 Positive_regulation denotes JNK dependent signaling
T32565 38179-38201 Entity denotes necroptotic L929 cells
T32563 38191-38195 Entity denotes L929
T32560 38203-38206 Protein denotes Akt
T32571 38203-38251 Phosphorylation denotes Akt phosphorylation at Thr308 during necroptosis
T32570 38203-38308 Regulation denotes Akt phosphorylation at Thr308 during necroptosis requires inputs from both growth factors and RIP1 kinase
T32559 38226-38232 Entity denotes Thr308
T32556 38297-38301 Protein denotes RIP1
T32553 38297-38308 Protein denotes RIP1 kinase
T32567 38326-38330 Protein denotes Akt,
T32555 38326-38345 Protein denotes Akt, JNK activation
T32561 38352-38369 Protein denotes to TNFα synthesis
T32569 38371-38407 Positive_regulation denotes Activation of Akt during necroptosis
T32564 38382-38388 Protein denotes of Akt
T32572 38419-38484 Phosphorylation denotes the phosphorylation of several known Akt substrates, such as mTOR
T32566 38439-38484 Protein denotes of several known Akt substrates, such as mTOR
T32562 38442-38479 Entity denotes several known Akt substrates, such as
T32558 38456-38459 Protein denotes Akt
T19793 38562-38565 Protein denotes Akt
T19846 38562-38577 Entity denotes Akt inhibitors,
T19800 38591-38594 Protein denotes Akt
T19827 38624-38638 Protein denotes of Akt mutants
T19898 38655-38684 Positive_regulation denotes necroptotic activation of Akt
T19774 38678-38684 Protein denotes of Akt
T19829 38777-38790 Protein denotes Akt-dependent
T19890 38860-38914 Phosphorylation denotes selective necroptotic phosphorylation of Thr308 of Akt
T19831 38898-38914 Entity denotes of Thr308 of Akt
T19776 38911-38914 Protein denotes Akt
T19849 38971-38990 Entity denotes of known substrates
T19795 38995-38998 Protein denotes Akt
T19858 38999-39024 Entity denotes effector pathways such as
T19802 39025-39043 Entity denotes the mTORC1 pathway
T19799 39115-39123 Protein denotes that Akt
T19851 39170-39174 Protein denotes RIP1
T19811 39170-39211 Protein denotes RIP1 kinase to known downstream signaling
T19783 39236-39258 Entity denotes necroptotic L929 cells
T19789 39248-39252 Entity denotes L929
T19810 39268-39271 Protein denotes JNK
T19884 39268-39282 Positive_regulation denotes JNK activation
T19874 39287-39311 Protein denotes autocrine TNFα synthesis
T19843 39348-39363 Entity denotes L929 cells [15]
T19889 39401-39465 Phosphorylation denotes we examined Akt phosphorylation after inhibition of a downstream
T19860 39413-39416 Protein denotes Akt
T19875 39466-39492 Protein denotes kinase in the pathway, JNK
T19796 39476-39492 Protein denotes the pathway, JNK
T19820 39512-39526 Entity denotes that SP600125,
T19893 39527-39594 Negative_regulation denotes which protected L929 cells from death and inhibited TNFα production
T19787 39579-39583 Protein denotes TNFα
T19892 39579-39594 Gene_expression denotes TNFα production
T19871 39596-39599 Protein denotes Fig
T19825 39683-39686 Protein denotes Akt
T19870 39695-39701 Entity denotes Ser473
T19814 39706-39712 Entity denotes Thr308
T19901 39789-39860 Negative_regulation denotes inhibitor that may inhibit the p110δ subunit of PI3K [41] and PDK1 [42] true
T19900 39789-39860 Negative_regulation denotes inhibitor that may inhibit the p110δ subunit of PI3K [41] and PDK1 [42] true
T19878 39789-39860 Entity denotes inhibitor that may inhibit the p110δ subunit of PI3K [41] and PDK1 [42]
T19790 39816-39846 Protein denotes the p110δ subunit of PI3K [41]
T19782 39837-39841 Protein denotes PI3K
T19854 39851-39860 Protein denotes PDK1 [42]
T19883 39909-39942 Phosphorylation denotes basal Akt phosphorylation levels,
T19844 39915-39918 Protein denotes Akt
T19856 39962-39973 Entity denotes of SP600125
T19779 40021-40027 Protein denotes of JNK
T19876 40044-40099 Entity denotes a more specific JNK inhibitor, JNK inhibitor VIII [43],
T19816 40060-40063 Protein denotes JNK
T19808 40075-40078 Protein denotes JNK
T19855 40075-40098 Entity denotes JNK inhibitor VIII [43]
T19813 40075-40098 Protein denotes JNK inhibitor VIII [43]
T19828 40119-40123 Protein denotes JNK1
T19819 40128-40132 Protein denotes JNK2
T19805 40134-40145 Protein denotes Fig. S10B–G
T19866 40197-40203 Protein denotes JNK1/2
T19842 40197-40203 Protein denotes JNK1/2
T19867 40202-40203 Protein denotes 2
T19896 40212-40234 Phosphorylation denotes phosphorylation of Akt
T19821 40228-40234 Protein denotes of Akt
T19859 40238-40244 Entity denotes Thr308
T19894 40262-40299 Phosphorylation denotes the phosphorylation of c-Jun at Ser63
T19837 40282-40290 Protein denotes of c-Jun
T19847 40294-40299 Entity denotes Ser63
T19850 40384-40388 Protein denotes TNFα
T19880 40384-40399 Gene_expression denotes TNFα production
T19832 40415-40418 Protein denotes Fig
T19873 40427-40431 Entity denotes F,G)
T19775 40488-40491 Protein denotes Fig
T19818 40532-40545 Entity denotes this molecule
T19824 40559-40562 Protein denotes JNK
T19887 40559-40573 Negative_regulation denotes JNK inhibition
T19862 40629-40693 Protein denotes Jun in necroptosis and autocrine TNFα synthesis [13], [14], [15]
T19833 40652-40693 Protein denotes autocrine TNFα synthesis [13], [14], [15]
T19848 40735-40740 Protein denotes c-Jun
T19817 40795-40801 Entity denotes Thr308
T19899 40872-40918 Regulation denotes autocrine TNFα production, dependent on c-Jun,
T19891 40872-40918 Gene_expression denotes autocrine TNFα production, dependent on c-Jun,
T19864 40882-40886 Protein denotes TNFα
T19791 40909-40917 Protein denotes on c-Jun
T19895 40966-40995 Positive_regulation denotes the delayed activation of Akt
T19882 40966-40995 Negative_regulation denotes the delayed activation of Akt
T19806 40989-40995 Protein denotes of Akt
T19778 41063-41077 Protein denotes in the protein
T19869 41087-41092 Protein denotes c-Jun
T19807 41128-41136 Entity denotes zVAD.fmk
T19798 41140-41144 Protein denotes TNFα
T19803 41161-41164 Protein denotes Akt
T19853 41169-41183 Protein denotes mTOR-dependent
T19879 41262-41272 Protein denotes that c-Jun
T19797 41308-41311 Protein denotes JNK
T19885 41407-41431 Phosphorylation denotes phosphorylation of c-Jun
T19872 41423-41431 Protein denotes of c-Jun
T19809 41435-41441 Entity denotes a site
T19780 41453-41458 Entity denotes Ser63
T19836 41560-41568 Entity denotes SP600125
T23552 41655-41658 Protein denotes Akt
T23517 41655-41665 Protein denotes Akt kinase
T23597 41669-41734 Protein denotes specifically engaged in the signaling downstream from RIP1 kinase
T23598 41723-41727 Protein denotes RIP1
T23634 41770-41833 Positive_regulation denotes promoting a selective increase in Akt phosphorylation on Thr308
T23632 41780-41833 Positive_regulation denotes a selective increase in Akt phosphorylation on Thr308
T23488 41804-41807 Protein denotes Akt
T23539 41824-41833 Entity denotes on Thr308
T23478 41867-41871 Protein denotes RIP1
T23568 41867-41878 Protein denotes RIP1 kinase
T23494 41968-41971 Protein denotes JNK
T23637 41968-41982 Positive_regulation denotes JNK activation
T23534 41984-42008 Protein denotes autocrine TNFα synthesis
T23625 42058-42080 Phosphorylation denotes phosphorylation of Akt
T23541 42074-42080 Protein denotes of Akt
T23521 42174-42193 Entity denotes the plasma membrane
T23503 42207-42213 Protein denotes of Akt
T23532 42244-42276 Protein denotes active membrane-targeted Myr-Akt
T23611 42251-42268 Entity denotes membrane-targeted
T23643 42323-42344 Gene_expression denotes expression of Myr-Akt
T23522 42334-42344 Protein denotes of Myr-Akt
T23512 42423-42426 Protein denotes JNK
T23479 42431-42435 Protein denotes TNFα
T23560 42457-42461 Protein denotes RIP1
T23557 42457-42478 Protein denotes RIP1 kinase-dependent
T23551 42501-42507 Entity denotes Thr308
T23642 42501-42531 Phosphorylation denotes Thr308 phosphorylation of Akt,
T23535 42524-42530 Protein denotes of Akt
T23639 42585-42629 Phosphorylation denotes Necroptotic phosphorylation of Thr308 of Akt
T23486 42613-42629 Entity denotes of Thr308 of Akt
T23476 42626-42629 Protein denotes Akt
T23602 42686-42705 Entity denotes of known substrates
T23601 42745-42769 Entity denotes the Akt effector pathway
T23586 42749-42752 Protein denotes Akt
T23579 42784-42790 Entity denotes mTORC1
T23564 42976-42979 Protein denotes Akt
T23633 43032-43089 Regulation denotes the RIP1-dependent increase in Akt Thr308 phosphorylation
T23626 43032-43089 Positive_regulation denotes the RIP1-dependent increase in Akt Thr308 phosphorylation
T23573 43036-43050 Protein denotes RIP1-dependent
T23645 43060-43089 Phosphorylation denotes in Akt Thr308 phosphorylation
T23506 43063-43066 Protein denotes Akt
T23544 43063-43073 Entity denotes Akt Thr308
T23577 43110-43169 Protein denotes that RIP1 kinase inhibits a phosphatase that targets Thr308
T23526 43110-43169 Protein denotes that RIP1 kinase inhibits a phosphatase that targets Thr308
T23628 43110-43169 Negative_regulation denotes that RIP1 kinase inhibits a phosphatase that targets Thr308
T23555 43120-43126 Protein denotes kinase
T23513 43163-43169 Entity denotes Thr308
T23587 43197-43274 Protein denotes the only enzyme established to specifically dephosphorylate this residue [45]
T23650 43197-43274 Gene_expression denotes the only enzyme established to specifically dephosphorylate this residue [45]
T23501 43257-43274 Entity denotes this residue [45]
T23547 43315-43325 Entity denotes of the PP2
T23473 43327-43351 Entity denotes inhibitor, okadaic acid,
T23529 43338-43350 Entity denotes okadaic acid
T23491 43355-43361 Entity denotes Thr308
T23537 43463-43472 Entity denotes in Thr308
T23514 43486-43490 Protein denotes RIP1
T23590 43486-43540 Protein denotes RIP1 kinase targeting PDK1, Akt or scaffolding factors
T23520 43508-43512 Protein denotes PDK1
T23594 43514-43517 Protein denotes Akt
T23508 43552-43569 Protein denotes these two kinases
T23641 43580-43693 Phosphorylation denotes Interestingly, we observed phosphorylation of Akt by recombinant RIP1 kinase in vitro on Thr146, 195/197, and 435
T23576 43623-43629 Protein denotes of Akt
T23543 43630-43656 Protein denotes by recombinant RIP1 kinase
T23495 43645-43649 Protein denotes RIP1
T23474 43669-43685 Entity denotes Thr146, 195/197,
T23490 43698-43704 Entity denotes Ser381
T23582 43698-43713 Entity denotes Ser381 residues
T23497 43737-43769 Entity denotes these residues to Ala in Myr-Akt
T23605 43755-43769 Entity denotes Ala in Myr-Akt
T23493 43762-43769 Protein denotes Myr-Akt
T23496 43880-43897 Entity denotes of these residues
T23482 43901-43915 Protein denotes endogenous Akt
T23500 44005-44013 Protein denotes antibody
T23619 44029-44035 Entity denotes Thr435
T23578 44029-44043 Entity denotes Thr435 peptide
T23621 44084-44090 Protein denotes of Akt
T23546 44094-44098 Protein denotes RIP1
T23481 44199-44271 Entity denotes the key substrates of Akt that promote necrotic death and TNFα synthesis
T23507 44221-44224 Protein denotes Akt
T23505 44257-44271 Protein denotes TNFα synthesis
T23566 44321-44324 Protein denotes Akt
T23572 44321-44342 Entity denotes Akt effector pathways
T23571 44356-44384 Entity denotes TORC1 in necroptotic control
T23565 44445-44451 Entity denotes mTORC1
T23504 44507-44532 Entity denotes additional Akt substrates
T23502 44518-44521 Protein denotes Akt
T23561 44603-44631 Entity denotes of additional Akt substrates
T23485 44617-44620 Protein denotes Akt
T23647 44729-44753 Binding denotes connecting mTORC1 to JNK
T23533 44740-44746 Entity denotes mTORC1
T23519 44747-44753 Protein denotes to JNK
T23652 44816-44853 Regulation denotes of mTORC1-dependent regulation of JNK
T23542 44847-44853 Protein denotes of JNK
T23531 44870-44884 Protein denotes ER stress [46]
T23644 44958-44979 Positive_regulation denotes the activation of JNK
T23518 44973-44979 Protein denotes of JNK
T23492 44983-44987 Protein denotes TNFα
T23615 45089-45095 Entity denotes mTORC1
T23635 45111-45134 Translation denotes the translation of TNFα
T23609 45127-45134 Protein denotes of TNFα
T23580 45179-45182 Protein denotes JNK
T23623 45200-45226 Entity denotes key inhibitor of apoptosis
T23640 45417-45452 Regulation denotes necrosis-specific regulation of Akt
T23498 45446-45452 Protein denotes of Akt
T23585 45456-45460 Protein denotes RIP1
T23606 45456-45467 Protein denotes RIP1 kinase
T23489 45540-45543 Protein denotes Akt
T23569 45613-45616 Protein denotes Akt
T23617 45635-45641 Entity denotes mTORC1
T23595 45660-45665 Protein denotes GSK-3
T23559 45667-45674 Protein denotes FoxO1/4
T23613 45667-45674 Protein denotes FoxO1/4
T23515 45673-45674 Protein denotes 4
T23575 45680-45684 Protein denotes MDM2
T23629 45722-45754 Positive_regulation denotes to assume that activation of Akt
T23530 45748-45754 Protein denotes of Akt
T23614 45939-45954 Protein denotes the Akt pathway
T23648 45971-46065 Positive_regulation denotes promote cell fates alternative to apoptosis, ranging from survival to non-apoptotic cell death
T23527 45979-46065 Entity denotes cell fates alternative to apoptosis, ranging from survival to non-apoptotic cell death
T23649 45979-46065 Localization denotes cell fates alternative to apoptosis, ranging from survival to non-apoptotic cell death
T23608 45979-46065 Protein denotes cell fates alternative to apoptosis, ranging from survival to non-apoptotic cell death
T23516 46139-46154 Protein denotes Akt-independent
T23525 46185-46189 Protein denotes RIP1
T23612 46185-46197 Protein denotes RIP1 kinase,
T23536 46375-46381 Protein denotes by Akt
T23511 46396-46399 Protein denotes Akt
T23630 46396-46410 Positive_regulation denotes Akt activation
T23651 46456-46479 Positive_regulation denotes activation of mTOR [48]
T23550 46467-46479 Protein denotes of mTOR [48]
T23558 46699-46710 Entity denotes by zVAD.fmk
T23545 46851-46860 Protein denotes TNFα [49]
T23588 46909-46915 Protein denotes by Akt
T23589 46919-46937 Protein denotes mTOR in our system
T23620 46976-46983 Protein denotes by TNFα
T23556 47018-47071 Entity denotes reconcile with the positive role of these proteins in
T23604 47033-47071 Protein denotes the positive role of these proteins in
T23599 47203-47218 Entity denotes mammalian cells
T23487 47294-47305 Protein denotes Akt pathway
T23510 47343-47347 Protein denotes RIP1
T23591 47343-47354 Protein denotes RIP1 kinase
T23646 47343-47364 Positive_regulation denotes RIP1 kinase signaling
T23596 47368-47371 Protein denotes Akt
T23618 47438-47457 Entity denotes multiple cell types
T23499 47525-47543 Entity denotes cell type specific
T23584 47569-47591 Entity denotes mouse lung fibroblasts
T23524 47593-47620 Entity denotes FADD-deficient Jurkat cells
T23592 47626-47638 Entity denotes macrophages,
T23603 47639-47642 Protein denotes Akt
T23636 47639-47738 Positive_regulation denotes Akt signaling contributed more prominently to an increase in TNFα synthesis, rather than cell death
T23549 47697-47714 Protein denotes in TNFα synthesis
T23616 47743-47776 Entity denotes se, unlike its role in L929 cells
T23528 47861-47865 Protein denotes RIP1
T23638 47893-47925 Regulation denotes mediating the production of TNFα
T23631 47903-47925 Gene_expression denotes the production of TNFα
T23600 47918-47925 Protein denotes of TNFα
T23553 48045-48051 Protein denotes of Akt
T23610 48512-48579 Entity denotes of plasma membrane integrity characteristic for necrotic cell death
T23570 48522-48530 Entity denotes membrane
T23554 48684-48701 Protein denotes of TNFα synthesis
T23574 48702-48721 Protein denotes by RIP1/Akt kinases
T23484 48705-48713 Protein denotes RIP1/Akt
T23622 48705-48713 Protein denotes RIP1/Akt
T23607 48710-48713 Protein denotes Akt
T23548 48949-49018 Protein denotes RIP1/RIP3 kinase activities by FADD and caspase-8 in epithelial cells
T23475 48959-48965 Protein denotes kinase
T23540 48980-48984 Protein denotes FADD
T23562 48989-49018 Protein denotes caspase-8 in epithelial cells
T23653 48989-49019 Localization denotes caspase-8 in epithelial cells
T23523 49002-49018 Entity denotes epithelial cells
T23480 49069-49073 Protein denotes TNFα
T23563 49145-49149 Protein denotes mice
T23654 49208-49260 Gene_expression denotes This increased production of TNFα during necroptosis
T23627 49208-49260 Positive_regulation denotes This increased production of TNFα during necroptosis
T23477 49234-49241 Protein denotes of TNFα
T23567 49582-49586 Protein denotes RIP1
T23509 49591-49595 Protein denotes RIP3
T23593 49638-49649 Protein denotes TNF-induced
T23538 49669-49683 Protein denotes of RIP1 kinase
T23483 49672-49676 Protein denotes RIP1
T23583 49762-49789 Protein denotes efficient and specific RIP1
T23581 49790-49807 Entity denotes kinase inhibitors
T23624 49790-49807 Protein denotes kinase inhibitors
T25560 49893-49901 Entity denotes Reagents
T25565 49989-50011 Entity denotes The following reagents
T25563 50717-50758 Protein denotes Pan-caspase inhibitor zVAD.fmk (20–30 µM)
T25562 50717-50758 Entity denotes Pan-caspase inhibitor zVAD.fmk (20–30 µM)
T25569 50717-50784 Binding denotes Pan-caspase inhibitor zVAD.fmk (20–30 µM) was purchased from Bachem
T25568 50729-50738 Entity denotes inhibitor
T25567 50796-50806 Protein denotes mouse TNFα
T25564 50819-50829 Protein denotes human bFGF
T25566 50842-50845 Protein denotes EGF
T25561 50858-50865 Entity denotes PDGF-BB
T25557 50882-50887 Protein denotes IGF-1
T25558 50909-50913 Entity denotes Cell
T25559 50937-50955 Entity denotes All other reagents
T25556 50966-50971 Protein denotes Sigma
T25665 50974-50977 Entity denotes DNA
T25661 50986-51020 Entity denotes of Myr-Akt1, containing c-terminal
T25660 50986-51020 Entity denotes of Myr-Akt1, containing c-terminal
T25662 50989-50992 Entity denotes Myr
T25666 50993-50997 Protein denotes Akt1
T25663 51021-51054 Protein denotes FLAG tag, has been described [52]
T25668 51056-51069 Protein denotes Myr-Akt1-FLAG
T25667 51084-51090 Protein denotes by PCR
T25669 51114-51119 Entity denotes BglII
T25670 51124-51129 Entity denotes EcoRI
T25664 51198-51209 Protein denotes of Myr-Akt1
T25746 51251-51261 Entity denotes Antibodies
T26173 52211-52221 Protein denotes Mouse TNFα
T26174 52270-52305 Protein denotes 5′-GCTACGACGTGGGCTACAG-3′;mouse 18S
T26170 52386-52396 Protein denotes human TNFα
T26175 52437-52447 Protein denotes human TNFα
T26171 52487-52496 Protein denotes human 18S
T26172 52506-52544 Protein denotes 5′- CAGCCACCCGAGATTGAGCA -3, human 18S
T26466 52594-52598 Entity denotes L929
T26468 52603-52630 Entity denotes FADD-deficient Jurkat cells
T26473 52656-52672 Entity denotes Lung fibroblasts
T26469 52742-52762 Entity denotes J774A.1 (ATCC) cells
T26467 52767-52788 Entity denotes RAW264.7 (ATCC) cells
T26461 52903-52908 Entity denotes Cells
T26462 52975-52978 Protein denotes FBS
T26460 52984-53030 Entity denotes 1% antibiotic-antimycotic mixture (Invitrogen)
T26465 53047-53057 Entity denotes fibroblast
T26463 53099-53110 Entity denotes L-glutamine
T26472 53112-53137 Entity denotes non-essential amino acids
T26471 53143-53158 Entity denotes sodium pyruvate
T26470 53160-53172 Entity denotes Jurkat cells
T26464 53247-53272 Entity denotes 1% antibiotic-antimycotic
T26685 53275-53301 Entity denotes Cell Viability Experiments
T26690 53449-53463 Entity denotes Cell viability
T26689 53485-53529 Entity denotes CellTiter-Glo Cell Viability Assay (Promega)
T26686 53596-53626 Entity denotes of the control untreated cells
T26687 53663-53671 Entity denotes of cells
T26688 53775-53780 Entity denotes cells
T26684 53844-53888 Entity denotes non-specific toxicity of the small molecules
T26691 53869-53888 Entity denotes the small molecules
T26924 53945-54001 Protein denotes Mouse ribosomal S6 protein (L-040893-00 and L-045791-00)
T26927 53961-53963 Entity denotes S6
T26928 54003-54027 Protein denotes mouse Akt1 (L-040709-00)
T26925 54278-54307 Entity denotes RNAiMAX reagent (Invitrogen),
T26922 54278-54307 Protein denotes RNAiMAX reagent (Invitrogen),
T26926 54366-54371 Entity denotes cells
T26920 54390-54398 Entity denotes zVAD.fmk
T26923 54402-54437 Protein denotes TNFα for 9 hr (RNA or Western blot)
T26921 54448-54462 Entity denotes cell viability
T27365 54479-54569 Binding denotes For Western blot, 4×105 adherent cells (1×106 Jurkat cells) were seeded into 35 mm2 dishes
T27359 54497-54538 Entity denotes 4×105 adherent cells (1×106 Jurkat cells)
T27345 54519-54537 Entity denotes 1×106 Jurkat cells
T27358 54587-54592 Entity denotes cells
T27342 54614-54628 Entity denotes 30 µM zVAD.fmk
T27349 54632-54651 Protein denotes 10 ng/ml mouse TNFα
T27363 54697-54702 Entity denotes cells
T27348 54773-54810 Protein denotes 20 µM zVAD.fmk or 10 ng/ml mouse TNFα
T27353 54779-54787 Entity denotes zVAD.fmk
T27356 54812-54817 Entity denotes Cells
T27366 54812-54842 Binding denotes Cells were harvested in 1×RIPA
T27354 54851-54855 Entity denotes Cell
T27352 54949-54961 Entity denotes cell lysates
T27344 55003-55025 Entity denotes Protein concentrations
T27364 55061-55086 Entity denotes nm Assay Reagent (Pierce)
T27339 55102-55113 Protein denotes of proteins
T27347 55218-55231 Entity denotes SDS-PAGE gels
T27341 55252-55256 Entity denotes PVDF
T27361 55252-55265 Entity denotes PVDF membrane
T27340 55289-55318 Protein denotes 5% bovine serum albumin (BSA)
T27346 55314-55317 Protein denotes BSA
T27343 55366-55384 Entity denotes Primary antibodies
T27360 55432-55452 Entity denotes Secondary antibodies
T27351 55529-55532 Protein denotes ECL
T27362 55529-55541 Entity denotes ECL reagents
T27350 55591-55600 Entity denotes membranes
T27357 55649-55651 Entity denotes GM
T27355 55683-55697 Entity denotes new antibodies
T27693 55700-55707 Protein denotes qRT-PCR
T27689 55708-55713 Entity denotes Cells
T27692 55759-55768 Protein denotes Total RNA
T27690 55829-55832 Protein denotes RNA
T27691 55961-55975 Protein denotes qPCR reactions
T27877 56076-56104 Regulation denotes Stable Infection of Myr-Akt1
T27871 56093-56104 Protein denotes of Myr-Akt1
T27874 56136-56163 Entity denotes HEK293FT cells (Invitrogen)
T27876 56186-56203 Entity denotes 2 µg of viral DNA
T27870 56191-56203 Entity denotes of viral DNA
T27868 56276-56279 Protein denotes Gen
T27872 56276-56319 Entity denotes GenJet transfection reagent (Signagen Labs)
T27869 56434-56451 Entity denotes 8 µg/ml polybrene
T27873 56453-56458 Entity denotes Cells
T27875 56491-56509 Entity denotes 10 µg/ml puromycin
T28061 56527-56573 Entity denotes ELISAOne assays (TGRBio, Hindmarsh, Australia)
T28062 56544-56547 Protein denotes TGR
T28069 56660-56732 Entity denotes Cell lysates were prepared in RIPA buffer as described for Western blots
T28059 56828-56883 Entity denotes Primary antibodies to phopsho-Thr308 and phopsho-Ser473
T28060 56850-56864 Entity denotes phopsho-Thr308
T28067 56869-56883 Entity denotes phopsho-Ser473
T28064 56946-56962 Entity denotes Primary antibody
T28065 56966-56973 Protein denotes pan-Akt
T28068 57006-57013 Entity denotes Signals
T28063 57018-57036 Entity denotes phospho-antibodies
T28066 57062-57076 Protein denotes pan-Akt values
T28269 57079-57083 Protein denotes TNFα
T28268 57090-57111 Protein denotes Mouse TNFα Quantikine
T28270 57196-57208 Entity denotes Cell lysates
T28267 57228-57275 Entity denotes 3×106 cells plated and treated in a 10 cm2 dish
T28271 57262-57275 Protein denotes a 10 cm2 dish
T28403 57278-57303 Protein denotes In vitro Akt Kinase Assay
T28407 57287-57290 Protein denotes Akt
T28402 57304-57307 Protein denotes Akt
T28398 57304-57314 Protein denotes Akt kinase
T28408 57347-57350 Protein denotes Akt
T28400 57351-57357 Protein denotes kinase
T28401 57390-57415 Entity denotes Cell Signaling Technology
T28406 57427-57434 Protein denotes Myr-Akt
T28405 57490-57492 Entity denotes M2
T28399 57509-57514 Protein denotes Sigma
T28409 57582-57599 Protein denotes protein substrate
T28397 57582-57599 Entity denotes protein substrate
T28404 57617-57642 Protein denotes of the GSK fusion protein
R10210 T15606 T15758 themeOf Akt,Akt activation
R10211 T15610 T15764 themeOf of Myr-Akt,"the expression of Myr-Akt, but not the K179M mutant,"
R10212 T15612 T15763 themeOf Myr-Akt,"RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis Given our observation that growth factors are important for zVAD.fmk induced death, we examined the contribution of several pathways, including MAPK pathways and Akt, which are known to be activated following growth factor receptor activation (Fig. 2A). Inhibition of Akt (Akt inhibitor VIII) strongly protected the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16] as well as cell death triggered by bFGF or IGF-1/zVAD.fmk under serum free conditions (Fig. 2B). Inhibition of Akt also protected the cells from growth-factor insensitive death by caused by TNFα (Fig. 2A). Consistent with previous reports, the JNK inhibitor SP600125 protected the cells from both zVAD.fmk and TNFα induced death (Fig. 2A,B and Fig. S2A) [12], [14]. In contrast, inhibition of two other MAPKs, p38 and ERK, previously reported not to be activated during necroptosis [14], did not protect from either zVAD.fmk or TNFα induced death (Fig. 2A). 10.1371/journal.pone.0056576.g002 Figure 2 Akt contributes to necroptosis induced by zVAD.fmk and TNFα. (A,B) Necroptosis was induced by zVAD.fmk or TNFα (full serum, A) or growth factors/zVAD.fmk (serum free, B) in the presence of inhibitors of Akt (Akt inhibitor VIII), JNK (SP600125), p38 (PD169316), and Erk (UO126). Cell viability was determined after 24 hrs. (C) L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs were treated with zVAD.fmk or TNFα for 9 hrs. Cell viability and Akt expression levels were determined after 24 hrs. In all graphs, average±SD was plotted. Next, we used two approaches to further validate the role of Akt in necroptotic cell death. First, two additional Akt inhibitors, a highly specific, allosteric kinase inhibitor MK-2206 [25] and triciribine (TCN) [26], which blocks membrane translocation of Akt, both attenuated cell death (Fig. S2B). Secondly, simultaneous knockdown of Akt isoforms Akt1 and Akt2 using siRNAs protected cells from necroptosis induced by both zVAD.fmk and TNFα (Fig. 2C). No expression of Akt3 was seen in L929 cells (Fig. S2C) and, consistently, Akt3 siRNA had no additional effect on necroptosis. Our results confirmed that Akt plays a key role in necroptosis induced by multiple stimuli in L929 cells. To understand the activation of Akt and JNK under necroptotic conditions, we examined the changes in Akt and JNK phosphorylation at 9 hrs post zVAD.fmk and TNFα stimulation. This time point was chosen because it reflects the early stage of cell death in our system (Fig. S3A, B). Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation at a known major activation site, Thr308 (Fig. 3A). Interestingly, we did not observe concomitant phosphorylation changes in the second major activation site of Akt, Ser473. We also observed an increase in the phosphorylation of both the p46 and p54 isoforms of JNK and its major substrate c-Jun (Fig. 3A). These data indicate that both Akt and JNK are activated under necroptotic conditions. 10.1371/journal.pone.0056576.g003 Figure 3 RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr, followed by western blotting with indicated antibodies. (B,C) L929 cells were treated with zVAD.fmk (B) or bFGF/zVAD.fmk (serum free conditions, C) and samples were collected at the indicated time points for western blot. (D) Nec-1 was added to the cells stimulated with bFGF or bFGF/zVAD (serum free conditions) for 15 min or 9 hr followed by western blot with the indicated antibodies. The RIP1 kinase inhibitor, Nec-1, completely prevented the increase in Thr308 Akt phosphorylation, while Nec-1i did not (Fig. 3A, Fig. S1D). Similarly, Nec-1 prevented the induction of JNK phosphorylation in response to zVAD.fmk and substantially reduced this change after TNFα addition. We observed some changes in total protein levels of JNK and c-Jun following necroptotic stimulation. Some of these changes, e.g. zVAD.fmk-induced increase in c-Jun, were also attenuated by Nec-1. Importantly, Nec-1 did not alter the basal phosphorylation levels of either Akt or JNK (Fig. 3A). This established that Akt Thr308 and JNK phosphorylation during necroptosis is RIP1 dependent. Interestingly, we discovered that the phosphorylation of Akt Thr308, JNK and Jun are late events following zVAD.fmk stimulation (Fig. 3B) that coincide with the onset of necroptosis at 6 hr post-stimulation (Fig. S3A). To better understand the contributions of growth factors and RIP1 kinase to necroptotic regulation of Akt, we next analyzed the time course of these phosphorylation changes under serum free conditions. We found that the addition of bFGF alone or in combination with zVAD.fmk led to a substantial rapid and transient increase in both Thr308 and Ser473 phosphorylation of Akt as well as JNK and c-Jun at 15 minutes, reflecting the expected response to growth factor stimulation (Fig. 3C). Significantly, the combination of bFGF/zVAD.fmk, but not bFGF alone, also caused a robust, second, delayed increase in the phosphorylation of Thr308, but not Ser473, of Akt as well as a delayed increase in the phosphorylation of JNK and Jun. Furthermore, Nec-1 had no significant effect on the early increase in both Akt and JNK/c-Jun phosphorylation triggered by both bFGF and bFGF/zVAD, while Nec-1, but not its inactive analog Nec-1i (Fig. S1E), efficiently blocked the bFGF/zVAD increase at 6–9 hr (Fig. 3D), suggesting that only the delayed activation of Akt and JNK is specific for necroptosis and dependent on RIP1 kinase activity. Similarly, IGF/zVAD, which also promoted cell death under serum free conditions, produced a delayed increase in Thr308 phosphorylation on Akt, while IGF alone caused solely an early, transient increase in phosphorylation (Fig. S3C). We confirmed the kinetics of the Akt Thr308 and Ser473 phosphorylation changes using a quantitative ELISA assay, which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation (Fig. S3D, E). Taken together, these results indicate that the observed delayed increases in Akt and JNK phosphorylation, preceding the onset of cell death, represent specific consequences of necroptotic signaling downstream from RIP1 kinase. TNFα Induces Delayed Akt Thr308 Phosphorylation and Necroptosis Independent of Growth Factor Stimulation Consistent with TNFα inducing necroptosis independently of growth factors (Fig. 1A), FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation, while efficiently preventing these changes in response to zVAD.fmk (Fig. S4A). Furthermore, addition of TNFα led to comparable late activation of Akt p308 signal under both normal and serum free conditions (Fig. S4B, C), indicating that TNFα signaling to Akt Thr308 is growth factor-independent. In contrast, activation of JNK by TNFα followed different kinetics from zVAD.fmk-induced changes. TNFα treatment caused an early and robust increase in the phosphorylation of JNK and c-Jun. Nec-1 did not affect this early increase, however, it reduced levels of pJNK/Jun at the late, 9 hr time point (Fig. S4B, C). This again separated early RIP1-independent changes, which likely reflect the ability of additional upstream kinases, such as Ask1 to activate JNK [27], from the late RIP1 kinase-dependent necroptotic signaling. Late Increase in Akt Thr308 Phosphorylation Contributes to the Induction of Necroptotic Cell Death We next investigated if the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation functionally contributes to the execution of necroptotic cell death. Firstly, PDGF/zVAD.fmk, which cannot induce necroptosis (Fig. 2A), triggered only the initial, rapid Akt and JNK phosphorylation changes and not the delayed activation (Fig. 4A), indicating that late, rather than early Akt phosphorylation correlates with necroptosis. Secondly, we saw that the ability of the Akt inhibitor to protect cells from necroptosis rapidly declined after 6 hrs of stimulation with zVAD.fmk, TNFα or bFGF/zVAD.fmk and no protection was observed when the inhibitor was added at 9 hrs (Fig. 4B,C). This time frame coincides with the timing of the secondary Akt Thr308 phosphorylation. Finally, we terminated the bFGF signal one hour after addition of bFGF by the addition of PD173074. This allowed us to retain early Akt activation, but to suppress the secondary increase (Fig. 4D). Both pre-addition and delayed addition of PD173074 fully prevented necroptosis (Fig. 4E). Overall, these data, while correlative, indicate that early Akt activation is insufficient to promote necroptosis and are strongly supportive of an important role for the delayed activation of Akt in the induction of necroptotic cell death. 10.1371/journal.pone.0056576.g004 Figure 4 Late Thr308 phosphorylation of Akt contributes to necroptosis. (A) L929 cells were treated with zVAD.fmk and bFGF or PDGF, with or without Nec-1, for the indicated periods of time. (B,C) L929 cells were stimulated by zVAD.fmk or TNFα (B) or bFGF/zVAD.fmk under serum free conditions (C). Akt inh. VIII was added 15 min before necroptotic stimulation (Pre) or at indicated times after stimulation. Viability was measured 24 hr after activation of necroptosis. (D) L929 cells were stimulated with bFGF/zVAD under serum free conditions. PD173074 was added 15 min before or 1 hr after FGF/zVAD. Samples for western blot were collected at 15 min and 9 hr time points. (E) Cells were pretreated with PD173074 or it was added 1 hr after bFGF/zVAD.fmk, followed by viability assessment at 24 hr. In all graphs, average±SD was plotted. The Akt Signaling Pathway Contributes to the Regulation of Necroptosis We next determined whether the necroptosis-associated increase in Thr308 phosphorylation results in an increase in Akt kinase activity. Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates (Forkhead box class O (FoxO) proteins, GSK-3 kinases and mouse double minute 2 (MDM2)) as well as downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6) (Fig. 5A). In some cases (FoxO1, FoxO4, MDM2), a robust increase was observed. In other cases (FoxO3a, GSK-3α/β, p70S6K and its substrate S6), the changes were less pronounced (Fig. 5A). The timing of the phosphorylation changes paralleled the increase in Akt phosphorylation (Fig. 5B, S5A, B). In the case of pFoxO1 we occasionally observed a shift in migration rather than an increase in band intensity (e.g. comparing Fig. 5A and B), suggesting that phosphorylation events in addition to Thr24 take place during necroptosis. Notably, in all cases the necroptosis-associated increases in Akt substrates were abrogated by Nec-1 (Fig. 5A, Fig. S5A, B). Overall, these data suggested that a significant part of the “canonical” Akt signaling network is activated at the onset of necroptotic cell death in a RIP1 dependent fashion. 10.1371/journal.pone.0056576.g005 Figure 5 mTORC1 contributes to the regulation of necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr and harvested for western blot. (B) Cell under serum free condition were treated with bFGF or bFGF/zVAD.fmk for the indicated amounts of time, followed by western blotting using the indicated antibodies. (C) Necroptosis was induced by zVAD.fmk or TNFα in L929 cell in the presence of inhibitors of Akt(Akt inh. VIII) and mTOR (rapamycin, Torin-1 and PI-103). (D) L929 cells with mTOR siRNA knockdown were harvested for western blot or treated with zVAD.fmk or TNFα for 24 hrs. Cell viability was determined 24 hr after activation of necroptosis. In all graphs, average±SD was plotted. Akt kinase is considered to be a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]. An important question is whether these same molecules reverse their pro-survival roles during necroptosis. We found that inhibition of mTORC1 by rapamycin, an inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C). Similarly, the direct mTOR kinase inhibitor Torin1 [30] and the dual PI3K/mTOR inhibitor PI-103 [31] also efficiently inhibited necroptosis (Fig. 5C). Knockdown of mTOR using siRNA further validated the small-molecule inhibitor data indicating a role for mTOR in necroptosis by protecting cells from both zVAD.fmk and TNFα induced death (Fig. 5D). mTORC1 regulates translation through activation of p70S6 kinase and, subsequently, ribosomal protein S6 [32]. Notably, a genome-wide siRNA screen [10] suggested an important role for protein translation in necroptosis. Consistently, we found that the small molecule inhibitor of p70S6K PF-4708671 [33] attenuated necroptosis at the concentrations required to block S6 phosphorylation (Fig. S5C, D). Partial siRNA knockdown of S6 protein attenuated necroptosis as well (Fig. S5E), suggesting that translational control by p70S6K/S6 may play a role in necroptosis. Overall, while the full repertoire of Akt targets during necroptosis remains to be fully explored, our data provide evidence that the activity of an anti-apoptotic branch of Akt signaling can promote necroptosis. RIP1 kinase, Akt, mTORC1 and JNK control the upregulation of TNFα accompanying necroptosis. Hitomi et al. [10] have recently reported that the induction of necroptosis by zVAD.fmk in L929 cells is associated with increased synthesis of TNFα, which potentiates cell death. Therefore, we examined whether Akt and its effectors contribute to TNFα synthesis. Consistent with a RIP1-dependent increase in TNFα protein (Fig. S6A, B), we found that TNFα mRNA levels increased during necroptosis in L929 cells in a RIP1 (Fig. S6C. Under serum free conditions, bFGF alone triggered some induction of TNFα mRNA, while its combination with zVAD.fmk (but not zVAD.fmk alone) caused a pronounced further increase (Fig. 6A). Conversely, PDGF caused a modest upregulation of TNFα mRNA, which was not further increased in the presence of zVAD.fmk (Fig. 6A), demonstrating that activation of necroptosis is specifically accompanied by a marked increase in autocrine TNFα synthesis. 10.1371/journal.pone.0056576.g006 Figure 6 Akt and mTORC1 control autocrine TNFα synthesis and JNK activation during necroptosis. (A) Cells were treated under serum free conditions with bFGF or PDGF with or without zVAD.fmk for 9 hr, followed by qRT-PCR analysis of mTNFα. Data was normalized to mouse 18S RNA. (B) Necroptosis was induced by zVAD.fmk or TNFα in cells treated with Nec-1, rapamycin (rapa), or Akt inh. VIII inh. followed by qRT-PCR analysis of TNFα mRNA levels. (C-F) L929 cells with siRNA knockdown of Akt isoforms (C,E) or mTOR (D,F) were stimulated with zVAD.fmk or TNFα for 9 hr, followed by qRT-PCR analysis of mTNFα (C,D) or western blot (E,F). In all graphs, average±SD was plotted. Further analysis suggested that both Akt and mTORC1 contribute to the upregulation of TNFα mRNA during necroptosis as both small-molecule inhibition and siRNA knockdown of Akt and mTOR reduced TNFα mRNA levels in necroptotic cells (Fig. 6B,C,D). Notably, RIP1 and Akt inhibitors had no effect on the levels of TNFα mRNA in control cells or in the cells stimulated with bFGF alone (Fig. 6A,B, Fig. S6C), suggesting that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis. Akt and mTORC1 Control the Activation of JNK during Necroptosis JNK is a well-established regulator of TNFα synthesis in a variety of systems [13], [14], [15], [34]. Therefore, the ability of Akt and mTORC1 inhibitors to block the increase in TNFα mRNA lead us to examine their role in the activation of JNK during necroptosis. Knockdown of Akt isoforms Akt1 and Akt2 or inhibition of Akt prominently suppressed the necroptosis dependent increase in JNK and c-Jun phosphorylation (Fig. 6E, S6D,E) suggesting that Akt may provide a link between RIP1 and JNK activation. Importantly, inhibition of Akt only inhibited the delayed, but not the early, increase in bFGF/zVAD.fmk induced JNK and c-Jun phosphorylation (Fig. S6F). Knockdown of mTOR, rapamycin and the p70S6K inhibitor PF-4708671 also attenuated the necroptosis-associated increase in JNK and c-Jun phosphorylation (Fig. 6F, S6E,G, Fig. S5D). Overall, these data suggested that the Akt-mTORC1-S6K axis, acting downstream from RIP1 kinase, is required for the increase in JNK activity during necroptosis in L929 cells. PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under Necroptotic Conditions Typical regulation of Akt by growth factors involves its recruitment to the plasma membrane, which is mediated by the binding of the pleckstrin homology (PH) domain of Akt to the product of PI3K, phosphatidylinositol-3,4,5-triphosphate (PIP3). In the membrane, Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1) and mTORC2 (or DNA-PK), respectively [35]. Since our results showed that only Thr308 Akt phosphorylation is increased during necroptosis, we next examined whether it is still dependent on PI3K and PDK1. Inhibition of PI3K and PDK1 using the specific inhibitors LY249002 and BX912 [36] resulted in the efficient inhibition of cell death and Akt Thr308 phosphorylation (Fig. S7A–D). Likewise, siRNA knockdown of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation (Fig. S7E,F) Therefore, PI3K and PDK1 activity is still required for non-canonical Akt activation during necroptosis. Expression of Constitutively Active Akt, Rescues Necroptosis Under Serum Free Conditions We used L929 cells stably expressing constitutively active wild type Akt1 (Myr-Akt) or the catalytically inactive mutant K179M in order to further understand the contribution of growth factors and RIP1 kinase to Akt activation during necroptosis. Constitutively active Akt1 (Myr-Akt) was generated as previously described [37] by the addition of a myristoylation signal which provides constitutive localization to the plasma membrane and by the deletion of the auto-inhibitory PH domain (Fig. 7A) resulting in an Akt that is active under serum free. It is important to note that the cells expressing Myr-Akt were viable, grew in a manner indistinguishable from the empty vector control cells, and were not triggered to induce necroptosis by serum starvation (Fig. 7B). This indicates that active Akt alone is not sufficient to induce necroptotic cell death. Under serum free conditions Myr-Akt, but not the K179M mutant, fully restored zVAD.fmk-induced necroptosis (Fig. 7A,B). Nec-1 prevented both Myr-Akt dependent cell death and the necroptosis-specific delayed increase in Akt Thr308 phosphorylation (Fig. 7B,C). Myr-Akt also allowed other zVAD.fmk-dependent events, including activation of JNK and c-Jun phosphorylation (Fig. 7C) and upregulation of TNFα mRNA (Fig. 7D) to occur under serum free conditions, confirming an important role for Akt at the apex of necroptotic signaling. These data demonstrated that the presence of active and membrane localized Akt is sufficient to uncouple Akt activation during necroptosis from growth factor signaling. RIP1 kinase was still able to regulate Akt activation during necroptosis, suggesting that growth factors and RIP1 kinase provide two independent inputs required for Akt changes during necroptosis. 10.1371/journal.pone.0056576.g007 Figure 7 Over expression of constitutively active Akt restores necroptosis under serum free conditions. (A,B) L929 cells were stably infected with empty MSCV retrovirus or viruses encoding Myr-Akt or the catalytically inactive Myr-Akt K179M. Necroptosis was induced by the addition of zVAD.fmk under serum free conditions (A) or serum or serum free conditions with Nec-1 (B). Viability assays were performed after 24 hr. (C) Myr-Akt and Myr-Akt K179M cells were treated with zVAD.fmk and/or Nec-1 under serum free conditions for 9 hr, followed by western blot using the indicated antibodies. Endogenous Akt (∼) and Myr-Akt (*) bands are indicated. (D) L929 cells, stably infected with Myr-Akt and Myr-Akt K179KM, were stimulated with zVAD.fmk for 9 hr under serum free conditions. TNFα mRNA levels were determined by qRT-PCR and normalized using mouse 18S RNA. (E-G) L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473 were treated with zVAD.fmk under serum free conditions, followed by viability assay at 24 hr (E), western blot at 9 hr (F), or evaluation of TNFα mRNA levels by qRT-PCR at 9 hrs (G). In all graphs, average±SD was plotted. RIP1 kinase-dependent Thr308 phosphorylation of Myr-Akt during necroptosis increased Myr-Akt activity as it did"
R10213 T15614 T15761 themeOf of Myr-Akt,the increased basal activity of Myr-Akt
R10214 T15618 T15747 themeOf "cell death, JNK activation, TNFα","cell death, JNK activation, TNFα production"
R10215 T15620 T15760 themeOf JNK,JNK activation
R10216 T15631 T15771 themeOf of Myr-Akt,necroptotic Thr308 phosphorylation of Myr-Akt
R10217 T15634 T15748 themeOf active Akt1 (Myr-Akt),Constitutively active Akt1 (Myr-Akt) was generated
R10218 T15640 T15643 partOf Akt Thr308,Akt
R10219 T15640 T15752 siteOf Akt Thr308,in Akt Thr308 phosphorylation
R10220 T15643 T15752 themeOf Akt,in Akt Thr308 phosphorylation
R10221 T15647 T15708 partOf Thr308,of Myr-Akt
R10222 T15647 T15756 siteOf Thr308,"RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis Given our observation that growth factors are important for zVAD.fmk induced death, we examined the contribution of several pathways, including MAPK pathways and Akt, which are known to be activated following growth factor receptor activation (Fig. 2A). Inhibition of Akt (Akt inhibitor VIII) strongly protected the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16] as well as cell death triggered by bFGF or IGF-1/zVAD.fmk under serum free conditions (Fig. 2B). Inhibition of Akt also protected the cells from growth-factor insensitive death by caused by TNFα (Fig. 2A). Consistent with previous reports, the JNK inhibitor SP600125 protected the cells from both zVAD.fmk and TNFα induced death (Fig. 2A,B and Fig. S2A) [12], [14]. In contrast, inhibition of two other MAPKs, p38 and ERK, previously reported not to be activated during necroptosis [14], did not protect from either zVAD.fmk or TNFα induced death (Fig. 2A). 10.1371/journal.pone.0056576.g002 Figure 2 Akt contributes to necroptosis induced by zVAD.fmk and TNFα. (A,B) Necroptosis was induced by zVAD.fmk or TNFα (full serum, A) or growth factors/zVAD.fmk (serum free, B) in the presence of inhibitors of Akt (Akt inhibitor VIII), JNK (SP600125), p38 (PD169316), and Erk (UO126). Cell viability was determined after 24 hrs. (C) L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs were treated with zVAD.fmk or TNFα for 9 hrs. Cell viability and Akt expression levels were determined after 24 hrs. In all graphs, average±SD was plotted. Next, we used two approaches to further validate the role of Akt in necroptotic cell death. First, two additional Akt inhibitors, a highly specific, allosteric kinase inhibitor MK-2206 [25] and triciribine (TCN) [26], which blocks membrane translocation of Akt, both attenuated cell death (Fig. S2B). Secondly, simultaneous knockdown of Akt isoforms Akt1 and Akt2 using siRNAs protected cells from necroptosis induced by both zVAD.fmk and TNFα (Fig. 2C). No expression of Akt3 was seen in L929 cells (Fig. S2C) and, consistently, Akt3 siRNA had no additional effect on necroptosis. Our results confirmed that Akt plays a key role in necroptosis induced by multiple stimuli in L929 cells. To understand the activation of Akt and JNK under necroptotic conditions, we examined the changes in Akt and JNK phosphorylation at 9 hrs post zVAD.fmk and TNFα stimulation. This time point was chosen because it reflects the early stage of cell death in our system (Fig. S3A, B). Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation at a known major activation site, Thr308 (Fig. 3A). Interestingly, we did not observe concomitant phosphorylation changes in the second major activation site of Akt, Ser473. We also observed an increase in the phosphorylation of both the p46 and p54 isoforms of JNK and its major substrate c-Jun (Fig. 3A). These data indicate that both Akt and JNK are activated under necroptotic conditions. 10.1371/journal.pone.0056576.g003 Figure 3 RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr, followed by western blotting with indicated antibodies. (B,C) L929 cells were treated with zVAD.fmk (B) or bFGF/zVAD.fmk (serum free conditions, C) and samples were collected at the indicated time points for western blot. (D) Nec-1 was added to the cells stimulated with bFGF or bFGF/zVAD (serum free conditions) for 15 min or 9 hr followed by western blot with the indicated antibodies. The RIP1 kinase inhibitor, Nec-1, completely prevented the increase in Thr308 Akt phosphorylation, while Nec-1i did not (Fig. 3A, Fig. S1D). Similarly, Nec-1 prevented the induction of JNK phosphorylation in response to zVAD.fmk and substantially reduced this change after TNFα addition. We observed some changes in total protein levels of JNK and c-Jun following necroptotic stimulation. Some of these changes, e.g. zVAD.fmk-induced increase in c-Jun, were also attenuated by Nec-1. Importantly, Nec-1 did not alter the basal phosphorylation levels of either Akt or JNK (Fig. 3A). This established that Akt Thr308 and JNK phosphorylation during necroptosis is RIP1 dependent. Interestingly, we discovered that the phosphorylation of Akt Thr308, JNK and Jun are late events following zVAD.fmk stimulation (Fig. 3B) that coincide with the onset of necroptosis at 6 hr post-stimulation (Fig. S3A). To better understand the contributions of growth factors and RIP1 kinase to necroptotic regulation of Akt, we next analyzed the time course of these phosphorylation changes under serum free conditions. We found that the addition of bFGF alone or in combination with zVAD.fmk led to a substantial rapid and transient increase in both Thr308 and Ser473 phosphorylation of Akt as well as JNK and c-Jun at 15 minutes, reflecting the expected response to growth factor stimulation (Fig. 3C). Significantly, the combination of bFGF/zVAD.fmk, but not bFGF alone, also caused a robust, second, delayed increase in the phosphorylation of Thr308, but not Ser473, of Akt as well as a delayed increase in the phosphorylation of JNK and Jun. Furthermore, Nec-1 had no significant effect on the early increase in both Akt and JNK/c-Jun phosphorylation triggered by both bFGF and bFGF/zVAD, while Nec-1, but not its inactive analog Nec-1i (Fig. S1E), efficiently blocked the bFGF/zVAD increase at 6–9 hr (Fig. 3D), suggesting that only the delayed activation of Akt and JNK is specific for necroptosis and dependent on RIP1 kinase activity. Similarly, IGF/zVAD, which also promoted cell death under serum free conditions, produced a delayed increase in Thr308 phosphorylation on Akt, while IGF alone caused solely an early, transient increase in phosphorylation (Fig. S3C). We confirmed the kinetics of the Akt Thr308 and Ser473 phosphorylation changes using a quantitative ELISA assay, which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation (Fig. S3D, E). Taken together, these results indicate that the observed delayed increases in Akt and JNK phosphorylation, preceding the onset of cell death, represent specific consequences of necroptotic signaling downstream from RIP1 kinase. TNFα Induces Delayed Akt Thr308 Phosphorylation and Necroptosis Independent of Growth Factor Stimulation Consistent with TNFα inducing necroptosis independently of growth factors (Fig. 1A), FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation, while efficiently preventing these changes in response to zVAD.fmk (Fig. S4A). Furthermore, addition of TNFα led to comparable late activation of Akt p308 signal under both normal and serum free conditions (Fig. S4B, C), indicating that TNFα signaling to Akt Thr308 is growth factor-independent. In contrast, activation of JNK by TNFα followed different kinetics from zVAD.fmk-induced changes. TNFα treatment caused an early and robust increase in the phosphorylation of JNK and c-Jun. Nec-1 did not affect this early increase, however, it reduced levels of pJNK/Jun at the late, 9 hr time point (Fig. S4B, C). This again separated early RIP1-independent changes, which likely reflect the ability of additional upstream kinases, such as Ask1 to activate JNK [27], from the late RIP1 kinase-dependent necroptotic signaling. Late Increase in Akt Thr308 Phosphorylation Contributes to the Induction of Necroptotic Cell Death We next investigated if the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation functionally contributes to the execution of necroptotic cell death. Firstly, PDGF/zVAD.fmk, which cannot induce necroptosis (Fig. 2A), triggered only the initial, rapid Akt and JNK phosphorylation changes and not the delayed activation (Fig. 4A), indicating that late, rather than early Akt phosphorylation correlates with necroptosis. Secondly, we saw that the ability of the Akt inhibitor to protect cells from necroptosis rapidly declined after 6 hrs of stimulation with zVAD.fmk, TNFα or bFGF/zVAD.fmk and no protection was observed when the inhibitor was added at 9 hrs (Fig. 4B,C). This time frame coincides with the timing of the secondary Akt Thr308 phosphorylation. Finally, we terminated the bFGF signal one hour after addition of bFGF by the addition of PD173074. This allowed us to retain early Akt activation, but to suppress the secondary increase (Fig. 4D). Both pre-addition and delayed addition of PD173074 fully prevented necroptosis (Fig. 4E). Overall, these data, while correlative, indicate that early Akt activation is insufficient to promote necroptosis and are strongly supportive of an important role for the delayed activation of Akt in the induction of necroptotic cell death. 10.1371/journal.pone.0056576.g004 Figure 4 Late Thr308 phosphorylation of Akt contributes to necroptosis. (A) L929 cells were treated with zVAD.fmk and bFGF or PDGF, with or without Nec-1, for the indicated periods of time. (B,C) L929 cells were stimulated by zVAD.fmk or TNFα (B) or bFGF/zVAD.fmk under serum free conditions (C). Akt inh. VIII was added 15 min before necroptotic stimulation (Pre) or at indicated times after stimulation. Viability was measured 24 hr after activation of necroptosis. (D) L929 cells were stimulated with bFGF/zVAD under serum free conditions. PD173074 was added 15 min before or 1 hr after FGF/zVAD. Samples for western blot were collected at 15 min and 9 hr time points. (E) Cells were pretreated with PD173074 or it was added 1 hr after bFGF/zVAD.fmk, followed by viability assessment at 24 hr. In all graphs, average±SD was plotted. The Akt Signaling Pathway Contributes to the Regulation of Necroptosis We next determined whether the necroptosis-associated increase in Thr308 phosphorylation results in an increase in Akt kinase activity. Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates (Forkhead box class O (FoxO) proteins, GSK-3 kinases and mouse double minute 2 (MDM2)) as well as downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6) (Fig. 5A). In some cases (FoxO1, FoxO4, MDM2), a robust increase was observed. In other cases (FoxO3a, GSK-3α/β, p70S6K and its substrate S6), the changes were less pronounced (Fig. 5A). The timing of the phosphorylation changes paralleled the increase in Akt phosphorylation (Fig. 5B, S5A, B). In the case of pFoxO1 we occasionally observed a shift in migration rather than an increase in band intensity (e.g. comparing Fig. 5A and B), suggesting that phosphorylation events in addition to Thr24 take place during necroptosis. Notably, in all cases the necroptosis-associated increases in Akt substrates were abrogated by Nec-1 (Fig. 5A, Fig. S5A, B). Overall, these data suggested that a significant part of the “canonical” Akt signaling network is activated at the onset of necroptotic cell death in a RIP1 dependent fashion. 10.1371/journal.pone.0056576.g005 Figure 5 mTORC1 contributes to the regulation of necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr and harvested for western blot. (B) Cell under serum free condition were treated with bFGF or bFGF/zVAD.fmk for the indicated amounts of time, followed by western blotting using the indicated antibodies. (C) Necroptosis was induced by zVAD.fmk or TNFα in L929 cell in the presence of inhibitors of Akt(Akt inh. VIII) and mTOR (rapamycin, Torin-1 and PI-103). (D) L929 cells with mTOR siRNA knockdown were harvested for western blot or treated with zVAD.fmk or TNFα for 24 hrs. Cell viability was determined 24 hr after activation of necroptosis. In all graphs, average±SD was plotted. Akt kinase is considered to be a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]. An important question is whether these same molecules reverse their pro-survival roles during necroptosis. We found that inhibition of mTORC1 by rapamycin, an inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C). Similarly, the direct mTOR kinase inhibitor Torin1 [30] and the dual PI3K/mTOR inhibitor PI-103 [31] also efficiently inhibited necroptosis (Fig. 5C). Knockdown of mTOR using siRNA further validated the small-molecule inhibitor data indicating a role for mTOR in necroptosis by protecting cells from both zVAD.fmk and TNFα induced death (Fig. 5D). mTORC1 regulates translation through activation of p70S6 kinase and, subsequently, ribosomal protein S6 [32]. Notably, a genome-wide siRNA screen [10] suggested an important role for protein translation in necroptosis. Consistently, we found that the small molecule inhibitor of p70S6K PF-4708671 [33] attenuated necroptosis at the concentrations required to block S6 phosphorylation (Fig. S5C, D). Partial siRNA knockdown of S6 protein attenuated necroptosis as well (Fig. S5E), suggesting that translational control by p70S6K/S6 may play a role in necroptosis. Overall, while the full repertoire of Akt targets during necroptosis remains to be fully explored, our data provide evidence that the activity of an anti-apoptotic branch of Akt signaling can promote necroptosis. RIP1 kinase, Akt, mTORC1 and JNK control the upregulation of TNFα accompanying necroptosis. Hitomi et al. [10] have recently reported that the induction of necroptosis by zVAD.fmk in L929 cells is associated with increased synthesis of TNFα, which potentiates cell death. Therefore, we examined whether Akt and its effectors contribute to TNFα synthesis. Consistent with a RIP1-dependent increase in TNFα protein (Fig. S6A, B), we found that TNFα mRNA levels increased during necroptosis in L929 cells in a RIP1 (Fig. S6C. Under serum free conditions, bFGF alone triggered some induction of TNFα mRNA, while its combination with zVAD.fmk (but not zVAD.fmk alone) caused a pronounced further increase (Fig. 6A). Conversely, PDGF caused a modest upregulation of TNFα mRNA, which was not further increased in the presence of zVAD.fmk (Fig. 6A), demonstrating that activation of necroptosis is specifically accompanied by a marked increase in autocrine TNFα synthesis. 10.1371/journal.pone.0056576.g006 Figure 6 Akt and mTORC1 control autocrine TNFα synthesis and JNK activation during necroptosis. (A) Cells were treated under serum free conditions with bFGF or PDGF with or without zVAD.fmk for 9 hr, followed by qRT-PCR analysis of mTNFα. Data was normalized to mouse 18S RNA. (B) Necroptosis was induced by zVAD.fmk or TNFα in cells treated with Nec-1, rapamycin (rapa), or Akt inh. VIII inh. followed by qRT-PCR analysis of TNFα mRNA levels. (C-F) L929 cells with siRNA knockdown of Akt isoforms (C,E) or mTOR (D,F) were stimulated with zVAD.fmk or TNFα for 9 hr, followed by qRT-PCR analysis of mTNFα (C,D) or western blot (E,F). In all graphs, average±SD was plotted. Further analysis suggested that both Akt and mTORC1 contribute to the upregulation of TNFα mRNA during necroptosis as both small-molecule inhibition and siRNA knockdown of Akt and mTOR reduced TNFα mRNA levels in necroptotic cells (Fig. 6B,C,D). Notably, RIP1 and Akt inhibitors had no effect on the levels of TNFα mRNA in control cells or in the cells stimulated with bFGF alone (Fig. 6A,B, Fig. S6C), suggesting that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis. Akt and mTORC1 Control the Activation of JNK during Necroptosis JNK is a well-established regulator of TNFα synthesis in a variety of systems [13], [14], [15], [34]. Therefore, the ability of Akt and mTORC1 inhibitors to block the increase in TNFα mRNA lead us to examine their role in the activation of JNK during necroptosis. Knockdown of Akt isoforms Akt1 and Akt2 or inhibition of Akt prominently suppressed the necroptosis dependent increase in JNK and c-Jun phosphorylation (Fig. 6E, S6D,E) suggesting that Akt may provide a link between RIP1 and JNK activation. Importantly, inhibition of Akt only inhibited the delayed, but not the early, increase in bFGF/zVAD.fmk induced JNK and c-Jun phosphorylation (Fig. S6F). Knockdown of mTOR, rapamycin and the p70S6K inhibitor PF-4708671 also attenuated the necroptosis-associated increase in JNK and c-Jun phosphorylation (Fig. 6F, S6E,G, Fig. S5D). Overall, these data suggested that the Akt-mTORC1-S6K axis, acting downstream from RIP1 kinase, is required for the increase in JNK activity during necroptosis in L929 cells. PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under Necroptotic Conditions Typical regulation of Akt by growth factors involves its recruitment to the plasma membrane, which is mediated by the binding of the pleckstrin homology (PH) domain of Akt to the product of PI3K, phosphatidylinositol-3,4,5-triphosphate (PIP3). In the membrane, Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1) and mTORC2 (or DNA-PK), respectively [35]. Since our results showed that only Thr308 Akt phosphorylation is increased during necroptosis, we next examined whether it is still dependent on PI3K and PDK1. Inhibition of PI3K and PDK1 using the specific inhibitors LY249002 and BX912 [36] resulted in the efficient inhibition of cell death and Akt Thr308 phosphorylation (Fig. S7A–D). Likewise, siRNA knockdown of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation (Fig. S7E,F) Therefore, PI3K and PDK1 activity is still required for non-canonical Akt activation during necroptosis. Expression of Constitutively Active Akt, Rescues Necroptosis Under Serum Free Conditions We used L929 cells stably expressing constitutively active wild type Akt1 (Myr-Akt) or the catalytically inactive mutant K179M in order to further understand the contribution of growth factors and RIP1 kinase to Akt activation during necroptosis. Constitutively active Akt1 (Myr-Akt) was generated as previously described [37] by the addition of a myristoylation signal which provides constitutive localization to the plasma membrane and by the deletion of the auto-inhibitory PH domain (Fig. 7A) resulting in an Akt that is active under serum free. It is important to note that the cells expressing Myr-Akt were viable, grew in a manner indistinguishable from the empty vector control cells, and were not triggered to induce necroptosis by serum starvation (Fig. 7B). This indicates that active Akt alone is not sufficient to induce necroptotic cell death. Under serum free conditions Myr-Akt, but not the K179M mutant, fully restored zVAD.fmk-induced necroptosis (Fig. 7A,B). Nec-1 prevented both Myr-Akt dependent cell death and the necroptosis-specific delayed increase in Akt Thr308 phosphorylation (Fig. 7B,C). Myr-Akt also allowed other zVAD.fmk-dependent events, including activation of JNK and c-Jun phosphorylation (Fig. 7C) and upregulation of TNFα mRNA (Fig. 7D) to occur under serum free conditions, confirming an important role for Akt at the apex of necroptotic signaling. These data demonstrated that the presence of active and membrane localized Akt is sufficient to uncouple Akt activation during necroptosis from growth factor signaling. RIP1 kinase was still able to regulate Akt activation during necroptosis, suggesting that growth factors and RIP1 kinase provide two independent inputs required for Akt changes during necroptosis. 10.1371/journal.pone.0056576.g007 Figure 7 Over expression of constitutively active Akt restores necroptosis under serum free conditions. (A,B) L929 cells were stably infected with empty MSCV retrovirus or viruses encoding Myr-Akt or the catalytically inactive Myr-Akt K179M. Necroptosis was induced by the addition of zVAD.fmk under serum free conditions (A) or serum or serum free conditions with Nec-1 (B). Viability assays were performed after 24 hr. (C) Myr-Akt and Myr-Akt K179M cells were treated with zVAD.fmk and/or Nec-1 under serum free conditions for 9 hr, followed by western blot using the indicated antibodies. Endogenous Akt (∼) and Myr-Akt (*) bands are indicated. (D) L929 cells, stably infected with Myr-Akt and Myr-Akt K179KM, were stimulated with zVAD.fmk for 9 hr under serum free conditions. TNFα mRNA levels were determined by qRT-PCR and normalized using mouse 18S RNA. (E-G) L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473 were treated with zVAD.fmk under serum free conditions, followed by viability assay at 24 hr (E), western blot at 9 hr (F), or evaluation of TNFα mRNA levels by qRT-PCR at 9 hrs (G). In all graphs, average±SD was plotted. RIP1 kinase-dependent Thr308 phosphorylation of Myr-Akt during necroptosis"
R10223 T15656 T15751 themeOf FoxO1,phosphorylation of FoxO1 and MDM2
R10224 T15660 T15744 themeOf JNK,"other zVAD.fmk-dependent events, including activation of JNK and c-Jun"
R10225 T15661 T15770 themeOf JNK,JNK activation
R10226 T15668 T15745 themeOf c-Jun,"other zVAD.fmk-dependent events, including activation of JNK and c-Jun"
R10227 T15672 T15759 themeOf Myr-Akt,the cells expressing Myr-Akt
R10228 T15674 T15753 causeOf RIP1 kinase-dependent,"RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis Given our observation that growth factors are important for zVAD.fmk induced death, we examined the contribution of several pathways, including MAPK pathways and Akt, which are known to be activated following growth factor receptor activation (Fig. 2A). Inhibition of Akt (Akt inhibitor VIII) strongly protected the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16] as well as cell death triggered by bFGF or IGF-1/zVAD.fmk under serum free conditions (Fig. 2B). Inhibition of Akt also protected the cells from growth-factor insensitive death by caused by TNFα (Fig. 2A). Consistent with previous reports, the JNK inhibitor SP600125 protected the cells from both zVAD.fmk and TNFα induced death (Fig. 2A,B and Fig. S2A) [12], [14]. In contrast, inhibition of two other MAPKs, p38 and ERK, previously reported not to be activated during necroptosis [14], did not protect from either zVAD.fmk or TNFα induced death (Fig. 2A). 10.1371/journal.pone.0056576.g002 Figure 2 Akt contributes to necroptosis induced by zVAD.fmk and TNFα. (A,B) Necroptosis was induced by zVAD.fmk or TNFα (full serum, A) or growth factors/zVAD.fmk (serum free, B) in the presence of inhibitors of Akt (Akt inhibitor VIII), JNK (SP600125), p38 (PD169316), and Erk (UO126). Cell viability was determined after 24 hrs. (C) L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs were treated with zVAD.fmk or TNFα for 9 hrs. Cell viability and Akt expression levels were determined after 24 hrs. In all graphs, average±SD was plotted. Next, we used two approaches to further validate the role of Akt in necroptotic cell death. First, two additional Akt inhibitors, a highly specific, allosteric kinase inhibitor MK-2206 [25] and triciribine (TCN) [26], which blocks membrane translocation of Akt, both attenuated cell death (Fig. S2B). Secondly, simultaneous knockdown of Akt isoforms Akt1 and Akt2 using siRNAs protected cells from necroptosis induced by both zVAD.fmk and TNFα (Fig. 2C). No expression of Akt3 was seen in L929 cells (Fig. S2C) and, consistently, Akt3 siRNA had no additional effect on necroptosis. Our results confirmed that Akt plays a key role in necroptosis induced by multiple stimuli in L929 cells. To understand the activation of Akt and JNK under necroptotic conditions, we examined the changes in Akt and JNK phosphorylation at 9 hrs post zVAD.fmk and TNFα stimulation. This time point was chosen because it reflects the early stage of cell death in our system (Fig. S3A, B). Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation at a known major activation site, Thr308 (Fig. 3A). Interestingly, we did not observe concomitant phosphorylation changes in the second major activation site of Akt, Ser473. We also observed an increase in the phosphorylation of both the p46 and p54 isoforms of JNK and its major substrate c-Jun (Fig. 3A). These data indicate that both Akt and JNK are activated under necroptotic conditions. 10.1371/journal.pone.0056576.g003 Figure 3 RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr, followed by western blotting with indicated antibodies. (B,C) L929 cells were treated with zVAD.fmk (B) or bFGF/zVAD.fmk (serum free conditions, C) and samples were collected at the indicated time points for western blot. (D) Nec-1 was added to the cells stimulated with bFGF or bFGF/zVAD (serum free conditions) for 15 min or 9 hr followed by western blot with the indicated antibodies. The RIP1 kinase inhibitor, Nec-1, completely prevented the increase in Thr308 Akt phosphorylation, while Nec-1i did not (Fig. 3A, Fig. S1D). Similarly, Nec-1 prevented the induction of JNK phosphorylation in response to zVAD.fmk and substantially reduced this change after TNFα addition. We observed some changes in total protein levels of JNK and c-Jun following necroptotic stimulation. Some of these changes, e.g. zVAD.fmk-induced increase in c-Jun, were also attenuated by Nec-1. Importantly, Nec-1 did not alter the basal phosphorylation levels of either Akt or JNK (Fig. 3A). This established that Akt Thr308 and JNK phosphorylation during necroptosis is RIP1 dependent. Interestingly, we discovered that the phosphorylation of Akt Thr308, JNK and Jun are late events following zVAD.fmk stimulation (Fig. 3B) that coincide with the onset of necroptosis at 6 hr post-stimulation (Fig. S3A). To better understand the contributions of growth factors and RIP1 kinase to necroptotic regulation of Akt, we next analyzed the time course of these phosphorylation changes under serum free conditions. We found that the addition of bFGF alone or in combination with zVAD.fmk led to a substantial rapid and transient increase in both Thr308 and Ser473 phosphorylation of Akt as well as JNK and c-Jun at 15 minutes, reflecting the expected response to growth factor stimulation (Fig. 3C). Significantly, the combination of bFGF/zVAD.fmk, but not bFGF alone, also caused a robust, second, delayed increase in the phosphorylation of Thr308, but not Ser473, of Akt as well as a delayed increase in the phosphorylation of JNK and Jun. Furthermore, Nec-1 had no significant effect on the early increase in both Akt and JNK/c-Jun phosphorylation triggered by both bFGF and bFGF/zVAD, while Nec-1, but not its inactive analog Nec-1i (Fig. S1E), efficiently blocked the bFGF/zVAD increase at 6–9 hr (Fig. 3D), suggesting that only the delayed activation of Akt and JNK is specific for necroptosis and dependent on RIP1 kinase activity. Similarly, IGF/zVAD, which also promoted cell death under serum free conditions, produced a delayed increase in Thr308 phosphorylation on Akt, while IGF alone caused solely an early, transient increase in phosphorylation (Fig. S3C). We confirmed the kinetics of the Akt Thr308 and Ser473 phosphorylation changes using a quantitative ELISA assay, which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation (Fig. S3D, E). Taken together, these results indicate that the observed delayed increases in Akt and JNK phosphorylation, preceding the onset of cell death, represent specific consequences of necroptotic signaling downstream from RIP1 kinase. TNFα Induces Delayed Akt Thr308 Phosphorylation and Necroptosis Independent of Growth Factor Stimulation Consistent with TNFα inducing necroptosis independently of growth factors (Fig. 1A), FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation, while efficiently preventing these changes in response to zVAD.fmk (Fig. S4A). Furthermore, addition of TNFα led to comparable late activation of Akt p308 signal under both normal and serum free conditions (Fig. S4B, C), indicating that TNFα signaling to Akt Thr308 is growth factor-independent. In contrast, activation of JNK by TNFα followed different kinetics from zVAD.fmk-induced changes. TNFα treatment caused an early and robust increase in the phosphorylation of JNK and c-Jun. Nec-1 did not affect this early increase, however, it reduced levels of pJNK/Jun at the late, 9 hr time point (Fig. S4B, C). This again separated early RIP1-independent changes, which likely reflect the ability of additional upstream kinases, such as Ask1 to activate JNK [27], from the late RIP1 kinase-dependent necroptotic signaling. Late Increase in Akt Thr308 Phosphorylation Contributes to the Induction of Necroptotic Cell Death We next investigated if the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation functionally contributes to the execution of necroptotic cell death. Firstly, PDGF/zVAD.fmk, which cannot induce necroptosis (Fig. 2A), triggered only the initial, rapid Akt and JNK phosphorylation changes and not the delayed activation (Fig. 4A), indicating that late, rather than early Akt phosphorylation correlates with necroptosis. Secondly, we saw that the ability of the Akt inhibitor to protect cells from necroptosis rapidly declined after 6 hrs of stimulation with zVAD.fmk, TNFα or bFGF/zVAD.fmk and no protection was observed when the inhibitor was added at 9 hrs (Fig. 4B,C). This time frame coincides with the timing of the secondary Akt Thr308 phosphorylation. Finally, we terminated the bFGF signal one hour after addition of bFGF by the addition of PD173074. This allowed us to retain early Akt activation, but to suppress the secondary increase (Fig. 4D). Both pre-addition and delayed addition of PD173074 fully prevented necroptosis (Fig. 4E). Overall, these data, while correlative, indicate that early Akt activation is insufficient to promote necroptosis and are strongly supportive of an important role for the delayed activation of Akt in the induction of necroptotic cell death. 10.1371/journal.pone.0056576.g004 Figure 4 Late Thr308 phosphorylation of Akt contributes to necroptosis. (A) L929 cells were treated with zVAD.fmk and bFGF or PDGF, with or without Nec-1, for the indicated periods of time. (B,C) L929 cells were stimulated by zVAD.fmk or TNFα (B) or bFGF/zVAD.fmk under serum free conditions (C). Akt inh. VIII was added 15 min before necroptotic stimulation (Pre) or at indicated times after stimulation. Viability was measured 24 hr after activation of necroptosis. (D) L929 cells were stimulated with bFGF/zVAD under serum free conditions. PD173074 was added 15 min before or 1 hr after FGF/zVAD. Samples for western blot were collected at 15 min and 9 hr time points. (E) Cells were pretreated with PD173074 or it was added 1 hr after bFGF/zVAD.fmk, followed by viability assessment at 24 hr. In all graphs, average±SD was plotted. The Akt Signaling Pathway Contributes to the Regulation of Necroptosis We next determined whether the necroptosis-associated increase in Thr308 phosphorylation results in an increase in Akt kinase activity. Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates (Forkhead box class O (FoxO) proteins, GSK-3 kinases and mouse double minute 2 (MDM2)) as well as downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6) (Fig. 5A). In some cases (FoxO1, FoxO4, MDM2), a robust increase was observed. In other cases (FoxO3a, GSK-3α/β, p70S6K and its substrate S6), the changes were less pronounced (Fig. 5A). The timing of the phosphorylation changes paralleled the increase in Akt phosphorylation (Fig. 5B, S5A, B). In the case of pFoxO1 we occasionally observed a shift in migration rather than an increase in band intensity (e.g. comparing Fig. 5A and B), suggesting that phosphorylation events in addition to Thr24 take place during necroptosis. Notably, in all cases the necroptosis-associated increases in Akt substrates were abrogated by Nec-1 (Fig. 5A, Fig. S5A, B). Overall, these data suggested that a significant part of the “canonical” Akt signaling network is activated at the onset of necroptotic cell death in a RIP1 dependent fashion. 10.1371/journal.pone.0056576.g005 Figure 5 mTORC1 contributes to the regulation of necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr and harvested for western blot. (B) Cell under serum free condition were treated with bFGF or bFGF/zVAD.fmk for the indicated amounts of time, followed by western blotting using the indicated antibodies. (C) Necroptosis was induced by zVAD.fmk or TNFα in L929 cell in the presence of inhibitors of Akt(Akt inh. VIII) and mTOR (rapamycin, Torin-1 and PI-103). (D) L929 cells with mTOR siRNA knockdown were harvested for western blot or treated with zVAD.fmk or TNFα for 24 hrs. Cell viability was determined 24 hr after activation of necroptosis. In all graphs, average±SD was plotted. Akt kinase is considered to be a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]. An important question is whether these same molecules reverse their pro-survival roles during necroptosis. We found that inhibition of mTORC1 by rapamycin, an inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C). Similarly, the direct mTOR kinase inhibitor Torin1 [30] and the dual PI3K/mTOR inhibitor PI-103 [31] also efficiently inhibited necroptosis (Fig. 5C). Knockdown of mTOR using siRNA further validated the small-molecule inhibitor data indicating a role for mTOR in necroptosis by protecting cells from both zVAD.fmk and TNFα induced death (Fig. 5D). mTORC1 regulates translation through activation of p70S6 kinase and, subsequently, ribosomal protein S6 [32]. Notably, a genome-wide siRNA screen [10] suggested an important role for protein translation in necroptosis. Consistently, we found that the small molecule inhibitor of p70S6K PF-4708671 [33] attenuated necroptosis at the concentrations required to block S6 phosphorylation (Fig. S5C, D). Partial siRNA knockdown of S6 protein attenuated necroptosis as well (Fig. S5E), suggesting that translational control by p70S6K/S6 may play a role in necroptosis. Overall, while the full repertoire of Akt targets during necroptosis remains to be fully explored, our data provide evidence that the activity of an anti-apoptotic branch of Akt signaling can promote necroptosis. RIP1 kinase, Akt, mTORC1 and JNK control the upregulation of TNFα accompanying necroptosis. Hitomi et al. [10] have recently reported that the induction of necroptosis by zVAD.fmk in L929 cells is associated with increased synthesis of TNFα, which potentiates cell death. Therefore, we examined whether Akt and its effectors contribute to TNFα synthesis. Consistent with a RIP1-dependent increase in TNFα protein (Fig. S6A, B), we found that TNFα mRNA levels increased during necroptosis in L929 cells in a RIP1 (Fig. S6C. Under serum free conditions, bFGF alone triggered some induction of TNFα mRNA, while its combination with zVAD.fmk (but not zVAD.fmk alone) caused a pronounced further increase (Fig. 6A). Conversely, PDGF caused a modest upregulation of TNFα mRNA, which was not further increased in the presence of zVAD.fmk (Fig. 6A), demonstrating that activation of necroptosis is specifically accompanied by a marked increase in autocrine TNFα synthesis. 10.1371/journal.pone.0056576.g006 Figure 6 Akt and mTORC1 control autocrine TNFα synthesis and JNK activation during necroptosis. (A) Cells were treated under serum free conditions with bFGF or PDGF with or without zVAD.fmk for 9 hr, followed by qRT-PCR analysis of mTNFα. Data was normalized to mouse 18S RNA. (B) Necroptosis was induced by zVAD.fmk or TNFα in cells treated with Nec-1, rapamycin (rapa), or Akt inh. VIII inh. followed by qRT-PCR analysis of TNFα mRNA levels. (C-F) L929 cells with siRNA knockdown of Akt isoforms (C,E) or mTOR (D,F) were stimulated with zVAD.fmk or TNFα for 9 hr, followed by qRT-PCR analysis of mTNFα (C,D) or western blot (E,F). In all graphs, average±SD was plotted. Further analysis suggested that both Akt and mTORC1 contribute to the upregulation of TNFα mRNA during necroptosis as both small-molecule inhibition and siRNA knockdown of Akt and mTOR reduced TNFα mRNA levels in necroptotic cells (Fig. 6B,C,D). Notably, RIP1 and Akt inhibitors had no effect on the levels of TNFα mRNA in control cells or in the cells stimulated with bFGF alone (Fig. 6A,B, Fig. S6C), suggesting that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis. Akt and mTORC1 Control the Activation of JNK during Necroptosis JNK is a well-established regulator of TNFα synthesis in a variety of systems [13], [14], [15], [34]. Therefore, the ability of Akt and mTORC1 inhibitors to block the increase in TNFα mRNA lead us to examine their role in the activation of JNK during necroptosis. Knockdown of Akt isoforms Akt1 and Akt2 or inhibition of Akt prominently suppressed the necroptosis dependent increase in JNK and c-Jun phosphorylation (Fig. 6E, S6D,E) suggesting that Akt may provide a link between RIP1 and JNK activation. Importantly, inhibition of Akt only inhibited the delayed, but not the early, increase in bFGF/zVAD.fmk induced JNK and c-Jun phosphorylation (Fig. S6F). Knockdown of mTOR, rapamycin and the p70S6K inhibitor PF-4708671 also attenuated the necroptosis-associated increase in JNK and c-Jun phosphorylation (Fig. 6F, S6E,G, Fig. S5D). Overall, these data suggested that the Akt-mTORC1-S6K axis, acting downstream from RIP1 kinase, is required for the increase in JNK activity during necroptosis in L929 cells. PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under Necroptotic Conditions Typical regulation of Akt by growth factors involves its recruitment to the plasma membrane, which is mediated by the binding of the pleckstrin homology (PH) domain of Akt to the product of PI3K, phosphatidylinositol-3,4,5-triphosphate (PIP3). In the membrane, Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1) and mTORC2 (or DNA-PK), respectively [35]. Since our results showed that only Thr308 Akt phosphorylation is increased during necroptosis, we next examined whether it is still dependent on PI3K and PDK1. Inhibition of PI3K and PDK1 using the specific inhibitors LY249002 and BX912 [36] resulted in the efficient inhibition of cell death and Akt Thr308 phosphorylation (Fig. S7A–D). Likewise, siRNA knockdown of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation (Fig. S7E,F) Therefore, PI3K and PDK1 activity is still required for non-canonical Akt activation during necroptosis. Expression of Constitutively Active Akt, Rescues Necroptosis Under Serum Free Conditions We used L929 cells stably expressing constitutively active wild type Akt1 (Myr-Akt) or the catalytically inactive mutant K179M in order to further understand the contribution of growth factors and RIP1 kinase to Akt activation during necroptosis. Constitutively active Akt1 (Myr-Akt) was generated as previously described [37] by the addition of a myristoylation signal which provides constitutive localization to the plasma membrane and by the deletion of the auto-inhibitory PH domain (Fig. 7A) resulting in an Akt that is active under serum free. It is important to note that the cells expressing Myr-Akt were viable, grew in a manner indistinguishable from the empty vector control cells, and were not triggered to induce necroptosis by serum starvation (Fig. 7B). This indicates that active Akt alone is not sufficient to induce necroptotic cell death. Under serum free conditions Myr-Akt, but not the K179M mutant, fully restored zVAD.fmk-induced necroptosis (Fig. 7A,B). Nec-1 prevented both Myr-Akt dependent cell death and the necroptosis-specific delayed increase in Akt Thr308 phosphorylation (Fig. 7B,C). Myr-Akt also allowed other zVAD.fmk-dependent events, including activation of JNK and c-Jun phosphorylation (Fig. 7C) and upregulation of TNFα mRNA (Fig. 7D) to occur under serum free conditions, confirming an important role for Akt at the apex of necroptotic signaling. These data demonstrated that the presence of active and membrane localized Akt is sufficient to uncouple Akt activation during necroptosis from growth factor signaling. RIP1 kinase was still able to regulate Akt activation during necroptosis, suggesting that growth factors and RIP1 kinase provide two independent inputs required for Akt changes during necroptosis. 10.1371/journal.pone.0056576.g007 Figure 7 Over expression of constitutively active Akt restores necroptosis under serum free conditions. (A,B) L929 cells were stably infected with empty MSCV retrovirus or viruses encoding Myr-Akt or the catalytically inactive Myr-Akt K179M. Necroptosis was induced by the addition of zVAD.fmk under serum free conditions (A) or serum or serum free conditions with Nec-1 (B). Viability assays were performed after 24 hr. (C) Myr-Akt and Myr-Akt K179M cells were treated with zVAD.fmk and/or Nec-1 under serum free conditions for 9 hr, followed by western blot using the indicated antibodies. Endogenous Akt (∼) and Myr-Akt (*) bands are indicated. (D) L929 cells, stably infected with Myr-Akt and Myr-Akt K179KM, were stimulated with zVAD.fmk for 9 hr under serum free conditions. TNFα mRNA levels were determined by qRT-PCR and normalized using mouse 18S RNA. (E-G) L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473 were treated with zVAD.fmk under serum free conditions, followed by viability assay at 24 hr (E), western blot at 9 hr (F), or evaluation of TNFα mRNA levels by qRT-PCR at 9 hrs (G). In all graphs, average±SD was plotted. RIP1 kinase-dependent Thr308 phosphorylation of Myr-Akt during necroptosis"
R10229 T15687 T15768 themeOf c-Jun,phosphorylation of JNK and c-Jun
R10230 T15689 T15769 themeOf JNK,phosphorylation of JNK and c-Jun
R10231 T15694 T15749 causeOf growth factor,growth factor signaling
R10232 T15697 T15766 themeOf of TNFα mRNA,upregulation of TNFα mRNA
R10233 T15702 T15757 themeOf Akt,Akt activation
R10234 T15706 T15631 partOf Thr308,of Myr-Akt
R10235 T15706 T15771 siteOf Thr308,necroptotic Thr308 phosphorylation of Myr-Akt
R10236 T15708 T15756 themeOf of Myr-Akt,"RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis Given our observation that growth factors are important for zVAD.fmk induced death, we examined the contribution of several pathways, including MAPK pathways and Akt, which are known to be activated following growth factor receptor activation (Fig. 2A). Inhibition of Akt (Akt inhibitor VIII) strongly protected the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16] as well as cell death triggered by bFGF or IGF-1/zVAD.fmk under serum free conditions (Fig. 2B). Inhibition of Akt also protected the cells from growth-factor insensitive death by caused by TNFα (Fig. 2A). Consistent with previous reports, the JNK inhibitor SP600125 protected the cells from both zVAD.fmk and TNFα induced death (Fig. 2A,B and Fig. S2A) [12], [14]. In contrast, inhibition of two other MAPKs, p38 and ERK, previously reported not to be activated during necroptosis [14], did not protect from either zVAD.fmk or TNFα induced death (Fig. 2A). 10.1371/journal.pone.0056576.g002 Figure 2 Akt contributes to necroptosis induced by zVAD.fmk and TNFα. (A,B) Necroptosis was induced by zVAD.fmk or TNFα (full serum, A) or growth factors/zVAD.fmk (serum free, B) in the presence of inhibitors of Akt (Akt inhibitor VIII), JNK (SP600125), p38 (PD169316), and Erk (UO126). Cell viability was determined after 24 hrs. (C) L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs were treated with zVAD.fmk or TNFα for 9 hrs. Cell viability and Akt expression levels were determined after 24 hrs. In all graphs, average±SD was plotted. Next, we used two approaches to further validate the role of Akt in necroptotic cell death. First, two additional Akt inhibitors, a highly specific, allosteric kinase inhibitor MK-2206 [25] and triciribine (TCN) [26], which blocks membrane translocation of Akt, both attenuated cell death (Fig. S2B). Secondly, simultaneous knockdown of Akt isoforms Akt1 and Akt2 using siRNAs protected cells from necroptosis induced by both zVAD.fmk and TNFα (Fig. 2C). No expression of Akt3 was seen in L929 cells (Fig. S2C) and, consistently, Akt3 siRNA had no additional effect on necroptosis. Our results confirmed that Akt plays a key role in necroptosis induced by multiple stimuli in L929 cells. To understand the activation of Akt and JNK under necroptotic conditions, we examined the changes in Akt and JNK phosphorylation at 9 hrs post zVAD.fmk and TNFα stimulation. This time point was chosen because it reflects the early stage of cell death in our system (Fig. S3A, B). Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation at a known major activation site, Thr308 (Fig. 3A). Interestingly, we did not observe concomitant phosphorylation changes in the second major activation site of Akt, Ser473. We also observed an increase in the phosphorylation of both the p46 and p54 isoforms of JNK and its major substrate c-Jun (Fig. 3A). These data indicate that both Akt and JNK are activated under necroptotic conditions. 10.1371/journal.pone.0056576.g003 Figure 3 RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr, followed by western blotting with indicated antibodies. (B,C) L929 cells were treated with zVAD.fmk (B) or bFGF/zVAD.fmk (serum free conditions, C) and samples were collected at the indicated time points for western blot. (D) Nec-1 was added to the cells stimulated with bFGF or bFGF/zVAD (serum free conditions) for 15 min or 9 hr followed by western blot with the indicated antibodies. The RIP1 kinase inhibitor, Nec-1, completely prevented the increase in Thr308 Akt phosphorylation, while Nec-1i did not (Fig. 3A, Fig. S1D). Similarly, Nec-1 prevented the induction of JNK phosphorylation in response to zVAD.fmk and substantially reduced this change after TNFα addition. We observed some changes in total protein levels of JNK and c-Jun following necroptotic stimulation. Some of these changes, e.g. zVAD.fmk-induced increase in c-Jun, were also attenuated by Nec-1. Importantly, Nec-1 did not alter the basal phosphorylation levels of either Akt or JNK (Fig. 3A). This established that Akt Thr308 and JNK phosphorylation during necroptosis is RIP1 dependent. Interestingly, we discovered that the phosphorylation of Akt Thr308, JNK and Jun are late events following zVAD.fmk stimulation (Fig. 3B) that coincide with the onset of necroptosis at 6 hr post-stimulation (Fig. S3A). To better understand the contributions of growth factors and RIP1 kinase to necroptotic regulation of Akt, we next analyzed the time course of these phosphorylation changes under serum free conditions. We found that the addition of bFGF alone or in combination with zVAD.fmk led to a substantial rapid and transient increase in both Thr308 and Ser473 phosphorylation of Akt as well as JNK and c-Jun at 15 minutes, reflecting the expected response to growth factor stimulation (Fig. 3C). Significantly, the combination of bFGF/zVAD.fmk, but not bFGF alone, also caused a robust, second, delayed increase in the phosphorylation of Thr308, but not Ser473, of Akt as well as a delayed increase in the phosphorylation of JNK and Jun. Furthermore, Nec-1 had no significant effect on the early increase in both Akt and JNK/c-Jun phosphorylation triggered by both bFGF and bFGF/zVAD, while Nec-1, but not its inactive analog Nec-1i (Fig. S1E), efficiently blocked the bFGF/zVAD increase at 6–9 hr (Fig. 3D), suggesting that only the delayed activation of Akt and JNK is specific for necroptosis and dependent on RIP1 kinase activity. Similarly, IGF/zVAD, which also promoted cell death under serum free conditions, produced a delayed increase in Thr308 phosphorylation on Akt, while IGF alone caused solely an early, transient increase in phosphorylation (Fig. S3C). We confirmed the kinetics of the Akt Thr308 and Ser473 phosphorylation changes using a quantitative ELISA assay, which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation (Fig. S3D, E). Taken together, these results indicate that the observed delayed increases in Akt and JNK phosphorylation, preceding the onset of cell death, represent specific consequences of necroptotic signaling downstream from RIP1 kinase. TNFα Induces Delayed Akt Thr308 Phosphorylation and Necroptosis Independent of Growth Factor Stimulation Consistent with TNFα inducing necroptosis independently of growth factors (Fig. 1A), FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation, while efficiently preventing these changes in response to zVAD.fmk (Fig. S4A). Furthermore, addition of TNFα led to comparable late activation of Akt p308 signal under both normal and serum free conditions (Fig. S4B, C), indicating that TNFα signaling to Akt Thr308 is growth factor-independent. In contrast, activation of JNK by TNFα followed different kinetics from zVAD.fmk-induced changes. TNFα treatment caused an early and robust increase in the phosphorylation of JNK and c-Jun. Nec-1 did not affect this early increase, however, it reduced levels of pJNK/Jun at the late, 9 hr time point (Fig. S4B, C). This again separated early RIP1-independent changes, which likely reflect the ability of additional upstream kinases, such as Ask1 to activate JNK [27], from the late RIP1 kinase-dependent necroptotic signaling. Late Increase in Akt Thr308 Phosphorylation Contributes to the Induction of Necroptotic Cell Death We next investigated if the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation functionally contributes to the execution of necroptotic cell death. Firstly, PDGF/zVAD.fmk, which cannot induce necroptosis (Fig. 2A), triggered only the initial, rapid Akt and JNK phosphorylation changes and not the delayed activation (Fig. 4A), indicating that late, rather than early Akt phosphorylation correlates with necroptosis. Secondly, we saw that the ability of the Akt inhibitor to protect cells from necroptosis rapidly declined after 6 hrs of stimulation with zVAD.fmk, TNFα or bFGF/zVAD.fmk and no protection was observed when the inhibitor was added at 9 hrs (Fig. 4B,C). This time frame coincides with the timing of the secondary Akt Thr308 phosphorylation. Finally, we terminated the bFGF signal one hour after addition of bFGF by the addition of PD173074. This allowed us to retain early Akt activation, but to suppress the secondary increase (Fig. 4D). Both pre-addition and delayed addition of PD173074 fully prevented necroptosis (Fig. 4E). Overall, these data, while correlative, indicate that early Akt activation is insufficient to promote necroptosis and are strongly supportive of an important role for the delayed activation of Akt in the induction of necroptotic cell death. 10.1371/journal.pone.0056576.g004 Figure 4 Late Thr308 phosphorylation of Akt contributes to necroptosis. (A) L929 cells were treated with zVAD.fmk and bFGF or PDGF, with or without Nec-1, for the indicated periods of time. (B,C) L929 cells were stimulated by zVAD.fmk or TNFα (B) or bFGF/zVAD.fmk under serum free conditions (C). Akt inh. VIII was added 15 min before necroptotic stimulation (Pre) or at indicated times after stimulation. Viability was measured 24 hr after activation of necroptosis. (D) L929 cells were stimulated with bFGF/zVAD under serum free conditions. PD173074 was added 15 min before or 1 hr after FGF/zVAD. Samples for western blot were collected at 15 min and 9 hr time points. (E) Cells were pretreated with PD173074 or it was added 1 hr after bFGF/zVAD.fmk, followed by viability assessment at 24 hr. In all graphs, average±SD was plotted. The Akt Signaling Pathway Contributes to the Regulation of Necroptosis We next determined whether the necroptosis-associated increase in Thr308 phosphorylation results in an increase in Akt kinase activity. Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates (Forkhead box class O (FoxO) proteins, GSK-3 kinases and mouse double minute 2 (MDM2)) as well as downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6) (Fig. 5A). In some cases (FoxO1, FoxO4, MDM2), a robust increase was observed. In other cases (FoxO3a, GSK-3α/β, p70S6K and its substrate S6), the changes were less pronounced (Fig. 5A). The timing of the phosphorylation changes paralleled the increase in Akt phosphorylation (Fig. 5B, S5A, B). In the case of pFoxO1 we occasionally observed a shift in migration rather than an increase in band intensity (e.g. comparing Fig. 5A and B), suggesting that phosphorylation events in addition to Thr24 take place during necroptosis. Notably, in all cases the necroptosis-associated increases in Akt substrates were abrogated by Nec-1 (Fig. 5A, Fig. S5A, B). Overall, these data suggested that a significant part of the “canonical” Akt signaling network is activated at the onset of necroptotic cell death in a RIP1 dependent fashion. 10.1371/journal.pone.0056576.g005 Figure 5 mTORC1 contributes to the regulation of necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr and harvested for western blot. (B) Cell under serum free condition were treated with bFGF or bFGF/zVAD.fmk for the indicated amounts of time, followed by western blotting using the indicated antibodies. (C) Necroptosis was induced by zVAD.fmk or TNFα in L929 cell in the presence of inhibitors of Akt(Akt inh. VIII) and mTOR (rapamycin, Torin-1 and PI-103). (D) L929 cells with mTOR siRNA knockdown were harvested for western blot or treated with zVAD.fmk or TNFα for 24 hrs. Cell viability was determined 24 hr after activation of necroptosis. In all graphs, average±SD was plotted. Akt kinase is considered to be a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]. An important question is whether these same molecules reverse their pro-survival roles during necroptosis. We found that inhibition of mTORC1 by rapamycin, an inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C). Similarly, the direct mTOR kinase inhibitor Torin1 [30] and the dual PI3K/mTOR inhibitor PI-103 [31] also efficiently inhibited necroptosis (Fig. 5C). Knockdown of mTOR using siRNA further validated the small-molecule inhibitor data indicating a role for mTOR in necroptosis by protecting cells from both zVAD.fmk and TNFα induced death (Fig. 5D). mTORC1 regulates translation through activation of p70S6 kinase and, subsequently, ribosomal protein S6 [32]. Notably, a genome-wide siRNA screen [10] suggested an important role for protein translation in necroptosis. Consistently, we found that the small molecule inhibitor of p70S6K PF-4708671 [33] attenuated necroptosis at the concentrations required to block S6 phosphorylation (Fig. S5C, D). Partial siRNA knockdown of S6 protein attenuated necroptosis as well (Fig. S5E), suggesting that translational control by p70S6K/S6 may play a role in necroptosis. Overall, while the full repertoire of Akt targets during necroptosis remains to be fully explored, our data provide evidence that the activity of an anti-apoptotic branch of Akt signaling can promote necroptosis. RIP1 kinase, Akt, mTORC1 and JNK control the upregulation of TNFα accompanying necroptosis. Hitomi et al. [10] have recently reported that the induction of necroptosis by zVAD.fmk in L929 cells is associated with increased synthesis of TNFα, which potentiates cell death. Therefore, we examined whether Akt and its effectors contribute to TNFα synthesis. Consistent with a RIP1-dependent increase in TNFα protein (Fig. S6A, B), we found that TNFα mRNA levels increased during necroptosis in L929 cells in a RIP1 (Fig. S6C. Under serum free conditions, bFGF alone triggered some induction of TNFα mRNA, while its combination with zVAD.fmk (but not zVAD.fmk alone) caused a pronounced further increase (Fig. 6A). Conversely, PDGF caused a modest upregulation of TNFα mRNA, which was not further increased in the presence of zVAD.fmk (Fig. 6A), demonstrating that activation of necroptosis is specifically accompanied by a marked increase in autocrine TNFα synthesis. 10.1371/journal.pone.0056576.g006 Figure 6 Akt and mTORC1 control autocrine TNFα synthesis and JNK activation during necroptosis. (A) Cells were treated under serum free conditions with bFGF or PDGF with or without zVAD.fmk for 9 hr, followed by qRT-PCR analysis of mTNFα. Data was normalized to mouse 18S RNA. (B) Necroptosis was induced by zVAD.fmk or TNFα in cells treated with Nec-1, rapamycin (rapa), or Akt inh. VIII inh. followed by qRT-PCR analysis of TNFα mRNA levels. (C-F) L929 cells with siRNA knockdown of Akt isoforms (C,E) or mTOR (D,F) were stimulated with zVAD.fmk or TNFα for 9 hr, followed by qRT-PCR analysis of mTNFα (C,D) or western blot (E,F). In all graphs, average±SD was plotted. Further analysis suggested that both Akt and mTORC1 contribute to the upregulation of TNFα mRNA during necroptosis as both small-molecule inhibition and siRNA knockdown of Akt and mTOR reduced TNFα mRNA levels in necroptotic cells (Fig. 6B,C,D). Notably, RIP1 and Akt inhibitors had no effect on the levels of TNFα mRNA in control cells or in the cells stimulated with bFGF alone (Fig. 6A,B, Fig. S6C), suggesting that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis. Akt and mTORC1 Control the Activation of JNK during Necroptosis JNK is a well-established regulator of TNFα synthesis in a variety of systems [13], [14], [15], [34]. Therefore, the ability of Akt and mTORC1 inhibitors to block the increase in TNFα mRNA lead us to examine their role in the activation of JNK during necroptosis. Knockdown of Akt isoforms Akt1 and Akt2 or inhibition of Akt prominently suppressed the necroptosis dependent increase in JNK and c-Jun phosphorylation (Fig. 6E, S6D,E) suggesting that Akt may provide a link between RIP1 and JNK activation. Importantly, inhibition of Akt only inhibited the delayed, but not the early, increase in bFGF/zVAD.fmk induced JNK and c-Jun phosphorylation (Fig. S6F). Knockdown of mTOR, rapamycin and the p70S6K inhibitor PF-4708671 also attenuated the necroptosis-associated increase in JNK and c-Jun phosphorylation (Fig. 6F, S6E,G, Fig. S5D). Overall, these data suggested that the Akt-mTORC1-S6K axis, acting downstream from RIP1 kinase, is required for the increase in JNK activity during necroptosis in L929 cells. PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under Necroptotic Conditions Typical regulation of Akt by growth factors involves its recruitment to the plasma membrane, which is mediated by the binding of the pleckstrin homology (PH) domain of Akt to the product of PI3K, phosphatidylinositol-3,4,5-triphosphate (PIP3). In the membrane, Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1) and mTORC2 (or DNA-PK), respectively [35]. Since our results showed that only Thr308 Akt phosphorylation is increased during necroptosis, we next examined whether it is still dependent on PI3K and PDK1. Inhibition of PI3K and PDK1 using the specific inhibitors LY249002 and BX912 [36] resulted in the efficient inhibition of cell death and Akt Thr308 phosphorylation (Fig. S7A–D). Likewise, siRNA knockdown of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation (Fig. S7E,F) Therefore, PI3K and PDK1 activity is still required for non-canonical Akt activation during necroptosis. Expression of Constitutively Active Akt, Rescues Necroptosis Under Serum Free Conditions We used L929 cells stably expressing constitutively active wild type Akt1 (Myr-Akt) or the catalytically inactive mutant K179M in order to further understand the contribution of growth factors and RIP1 kinase to Akt activation during necroptosis. Constitutively active Akt1 (Myr-Akt) was generated as previously described [37] by the addition of a myristoylation signal which provides constitutive localization to the plasma membrane and by the deletion of the auto-inhibitory PH domain (Fig. 7A) resulting in an Akt that is active under serum free. It is important to note that the cells expressing Myr-Akt were viable, grew in a manner indistinguishable from the empty vector control cells, and were not triggered to induce necroptosis by serum starvation (Fig. 7B). This indicates that active Akt alone is not sufficient to induce necroptotic cell death. Under serum free conditions Myr-Akt, but not the K179M mutant, fully restored zVAD.fmk-induced necroptosis (Fig. 7A,B). Nec-1 prevented both Myr-Akt dependent cell death and the necroptosis-specific delayed increase in Akt Thr308 phosphorylation (Fig. 7B,C). Myr-Akt also allowed other zVAD.fmk-dependent events, including activation of JNK and c-Jun phosphorylation (Fig. 7C) and upregulation of TNFα mRNA (Fig. 7D) to occur under serum free conditions, confirming an important role for Akt at the apex of necroptotic signaling. These data demonstrated that the presence of active and membrane localized Akt is sufficient to uncouple Akt activation during necroptosis from growth factor signaling. RIP1 kinase was still able to regulate Akt activation during necroptosis, suggesting that growth factors and RIP1 kinase provide two independent inputs required for Akt changes during necroptosis. 10.1371/journal.pone.0056576.g007 Figure 7 Over expression of constitutively active Akt restores necroptosis under serum free conditions. (A,B) L929 cells were stably infected with empty MSCV retrovirus or viruses encoding Myr-Akt or the catalytically inactive Myr-Akt K179M. Necroptosis was induced by the addition of zVAD.fmk under serum free conditions (A) or serum or serum free conditions with Nec-1 (B). Viability assays were performed after 24 hr. (C) Myr-Akt and Myr-Akt K179M cells were treated with zVAD.fmk and/or Nec-1 under serum free conditions for 9 hr, followed by western blot using the indicated antibodies. Endogenous Akt (∼) and Myr-Akt (*) bands are indicated. (D) L929 cells, stably infected with Myr-Akt and Myr-Akt K179KM, were stimulated with zVAD.fmk for 9 hr under serum free conditions. TNFα mRNA levels were determined by qRT-PCR and normalized using mouse 18S RNA. (E-G) L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473 were treated with zVAD.fmk under serum free conditions, followed by viability assay at 24 hr (E), western blot at 9 hr (F), or evaluation of TNFα mRNA levels by qRT-PCR at 9 hrs (G). In all graphs, average±SD was plotted. RIP1 kinase-dependent Thr308 phosphorylation of Myr-Akt during necroptosis"
R10237 T15718 T15750 themeOf MDM2,phosphorylation of FoxO1 and MDM2
R10238 T15725 T15765 themeOf Akt,Akt activation
R10239 T15750 T15754 themeOf phosphorylation of FoxO1 and MDM2,"phosphorylation of FoxO1 and MDM2 was significantly increased in the presence of zVAD.fmk,"
R10240 T15751 T15755 themeOf phosphorylation of FoxO1 and MDM2,"phosphorylation of FoxO1 and MDM2 was significantly increased in the presence of zVAD.fmk,"
R10241 T15756 T15753 themeOf "RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis Given our observation that growth factors are important for zVAD.fmk induced death, we examined the contribution of several pathways, including MAPK pathways and Akt, which are known to be activated following growth factor receptor activation (Fig. 2A). Inhibition of Akt (Akt inhibitor VIII) strongly protected the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16] as well as cell death triggered by bFGF or IGF-1/zVAD.fmk under serum free conditions (Fig. 2B). Inhibition of Akt also protected the cells from growth-factor insensitive death by caused by TNFα (Fig. 2A). Consistent with previous reports, the JNK inhibitor SP600125 protected the cells from both zVAD.fmk and TNFα induced death (Fig. 2A,B and Fig. S2A) [12], [14]. In contrast, inhibition of two other MAPKs, p38 and ERK, previously reported not to be activated during necroptosis [14], did not protect from either zVAD.fmk or TNFα induced death (Fig. 2A). 10.1371/journal.pone.0056576.g002 Figure 2 Akt contributes to necroptosis induced by zVAD.fmk and TNFα. (A,B) Necroptosis was induced by zVAD.fmk or TNFα (full serum, A) or growth factors/zVAD.fmk (serum free, B) in the presence of inhibitors of Akt (Akt inhibitor VIII), JNK (SP600125), p38 (PD169316), and Erk (UO126). Cell viability was determined after 24 hrs. (C) L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs were treated with zVAD.fmk or TNFα for 9 hrs. Cell viability and Akt expression levels were determined after 24 hrs. In all graphs, average±SD was plotted. Next, we used two approaches to further validate the role of Akt in necroptotic cell death. First, two additional Akt inhibitors, a highly specific, allosteric kinase inhibitor MK-2206 [25] and triciribine (TCN) [26], which blocks membrane translocation of Akt, both attenuated cell death (Fig. S2B). Secondly, simultaneous knockdown of Akt isoforms Akt1 and Akt2 using siRNAs protected cells from necroptosis induced by both zVAD.fmk and TNFα (Fig. 2C). No expression of Akt3 was seen in L929 cells (Fig. S2C) and, consistently, Akt3 siRNA had no additional effect on necroptosis. Our results confirmed that Akt plays a key role in necroptosis induced by multiple stimuli in L929 cells. To understand the activation of Akt and JNK under necroptotic conditions, we examined the changes in Akt and JNK phosphorylation at 9 hrs post zVAD.fmk and TNFα stimulation. This time point was chosen because it reflects the early stage of cell death in our system (Fig. S3A, B). Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation at a known major activation site, Thr308 (Fig. 3A). Interestingly, we did not observe concomitant phosphorylation changes in the second major activation site of Akt, Ser473. We also observed an increase in the phosphorylation of both the p46 and p54 isoforms of JNK and its major substrate c-Jun (Fig. 3A). These data indicate that both Akt and JNK are activated under necroptotic conditions. 10.1371/journal.pone.0056576.g003 Figure 3 RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr, followed by western blotting with indicated antibodies. (B,C) L929 cells were treated with zVAD.fmk (B) or bFGF/zVAD.fmk (serum free conditions, C) and samples were collected at the indicated time points for western blot. (D) Nec-1 was added to the cells stimulated with bFGF or bFGF/zVAD (serum free conditions) for 15 min or 9 hr followed by western blot with the indicated antibodies. The RIP1 kinase inhibitor, Nec-1, completely prevented the increase in Thr308 Akt phosphorylation, while Nec-1i did not (Fig. 3A, Fig. S1D). Similarly, Nec-1 prevented the induction of JNK phosphorylation in response to zVAD.fmk and substantially reduced this change after TNFα addition. We observed some changes in total protein levels of JNK and c-Jun following necroptotic stimulation. Some of these changes, e.g. zVAD.fmk-induced increase in c-Jun, were also attenuated by Nec-1. Importantly, Nec-1 did not alter the basal phosphorylation levels of either Akt or JNK (Fig. 3A). This established that Akt Thr308 and JNK phosphorylation during necroptosis is RIP1 dependent. Interestingly, we discovered that the phosphorylation of Akt Thr308, JNK and Jun are late events following zVAD.fmk stimulation (Fig. 3B) that coincide with the onset of necroptosis at 6 hr post-stimulation (Fig. S3A). To better understand the contributions of growth factors and RIP1 kinase to necroptotic regulation of Akt, we next analyzed the time course of these phosphorylation changes under serum free conditions. We found that the addition of bFGF alone or in combination with zVAD.fmk led to a substantial rapid and transient increase in both Thr308 and Ser473 phosphorylation of Akt as well as JNK and c-Jun at 15 minutes, reflecting the expected response to growth factor stimulation (Fig. 3C). Significantly, the combination of bFGF/zVAD.fmk, but not bFGF alone, also caused a robust, second, delayed increase in the phosphorylation of Thr308, but not Ser473, of Akt as well as a delayed increase in the phosphorylation of JNK and Jun. Furthermore, Nec-1 had no significant effect on the early increase in both Akt and JNK/c-Jun phosphorylation triggered by both bFGF and bFGF/zVAD, while Nec-1, but not its inactive analog Nec-1i (Fig. S1E), efficiently blocked the bFGF/zVAD increase at 6–9 hr (Fig. 3D), suggesting that only the delayed activation of Akt and JNK is specific for necroptosis and dependent on RIP1 kinase activity. Similarly, IGF/zVAD, which also promoted cell death under serum free conditions, produced a delayed increase in Thr308 phosphorylation on Akt, while IGF alone caused solely an early, transient increase in phosphorylation (Fig. S3C). We confirmed the kinetics of the Akt Thr308 and Ser473 phosphorylation changes using a quantitative ELISA assay, which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation (Fig. S3D, E). Taken together, these results indicate that the observed delayed increases in Akt and JNK phosphorylation, preceding the onset of cell death, represent specific consequences of necroptotic signaling downstream from RIP1 kinase. TNFα Induces Delayed Akt Thr308 Phosphorylation and Necroptosis Independent of Growth Factor Stimulation Consistent with TNFα inducing necroptosis independently of growth factors (Fig. 1A), FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation, while efficiently preventing these changes in response to zVAD.fmk (Fig. S4A). Furthermore, addition of TNFα led to comparable late activation of Akt p308 signal under both normal and serum free conditions (Fig. S4B, C), indicating that TNFα signaling to Akt Thr308 is growth factor-independent. In contrast, activation of JNK by TNFα followed different kinetics from zVAD.fmk-induced changes. TNFα treatment caused an early and robust increase in the phosphorylation of JNK and c-Jun. Nec-1 did not affect this early increase, however, it reduced levels of pJNK/Jun at the late, 9 hr time point (Fig. S4B, C). This again separated early RIP1-independent changes, which likely reflect the ability of additional upstream kinases, such as Ask1 to activate JNK [27], from the late RIP1 kinase-dependent necroptotic signaling. Late Increase in Akt Thr308 Phosphorylation Contributes to the Induction of Necroptotic Cell Death We next investigated if the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation functionally contributes to the execution of necroptotic cell death. Firstly, PDGF/zVAD.fmk, which cannot induce necroptosis (Fig. 2A), triggered only the initial, rapid Akt and JNK phosphorylation changes and not the delayed activation (Fig. 4A), indicating that late, rather than early Akt phosphorylation correlates with necroptosis. Secondly, we saw that the ability of the Akt inhibitor to protect cells from necroptosis rapidly declined after 6 hrs of stimulation with zVAD.fmk, TNFα or bFGF/zVAD.fmk and no protection was observed when the inhibitor was added at 9 hrs (Fig. 4B,C). This time frame coincides with the timing of the secondary Akt Thr308 phosphorylation. Finally, we terminated the bFGF signal one hour after addition of bFGF by the addition of PD173074. This allowed us to retain early Akt activation, but to suppress the secondary increase (Fig. 4D). Both pre-addition and delayed addition of PD173074 fully prevented necroptosis (Fig. 4E). Overall, these data, while correlative, indicate that early Akt activation is insufficient to promote necroptosis and are strongly supportive of an important role for the delayed activation of Akt in the induction of necroptotic cell death. 10.1371/journal.pone.0056576.g004 Figure 4 Late Thr308 phosphorylation of Akt contributes to necroptosis. (A) L929 cells were treated with zVAD.fmk and bFGF or PDGF, with or without Nec-1, for the indicated periods of time. (B,C) L929 cells were stimulated by zVAD.fmk or TNFα (B) or bFGF/zVAD.fmk under serum free conditions (C). Akt inh. VIII was added 15 min before necroptotic stimulation (Pre) or at indicated times after stimulation. Viability was measured 24 hr after activation of necroptosis. (D) L929 cells were stimulated with bFGF/zVAD under serum free conditions. PD173074 was added 15 min before or 1 hr after FGF/zVAD. Samples for western blot were collected at 15 min and 9 hr time points. (E) Cells were pretreated with PD173074 or it was added 1 hr after bFGF/zVAD.fmk, followed by viability assessment at 24 hr. In all graphs, average±SD was plotted. The Akt Signaling Pathway Contributes to the Regulation of Necroptosis We next determined whether the necroptosis-associated increase in Thr308 phosphorylation results in an increase in Akt kinase activity. Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates (Forkhead box class O (FoxO) proteins, GSK-3 kinases and mouse double minute 2 (MDM2)) as well as downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6) (Fig. 5A). In some cases (FoxO1, FoxO4, MDM2), a robust increase was observed. In other cases (FoxO3a, GSK-3α/β, p70S6K and its substrate S6), the changes were less pronounced (Fig. 5A). The timing of the phosphorylation changes paralleled the increase in Akt phosphorylation (Fig. 5B, S5A, B). In the case of pFoxO1 we occasionally observed a shift in migration rather than an increase in band intensity (e.g. comparing Fig. 5A and B), suggesting that phosphorylation events in addition to Thr24 take place during necroptosis. Notably, in all cases the necroptosis-associated increases in Akt substrates were abrogated by Nec-1 (Fig. 5A, Fig. S5A, B). Overall, these data suggested that a significant part of the “canonical” Akt signaling network is activated at the onset of necroptotic cell death in a RIP1 dependent fashion. 10.1371/journal.pone.0056576.g005 Figure 5 mTORC1 contributes to the regulation of necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr and harvested for western blot. (B) Cell under serum free condition were treated with bFGF or bFGF/zVAD.fmk for the indicated amounts of time, followed by western blotting using the indicated antibodies. (C) Necroptosis was induced by zVAD.fmk or TNFα in L929 cell in the presence of inhibitors of Akt(Akt inh. VIII) and mTOR (rapamycin, Torin-1 and PI-103). (D) L929 cells with mTOR siRNA knockdown were harvested for western blot or treated with zVAD.fmk or TNFα for 24 hrs. Cell viability was determined 24 hr after activation of necroptosis. In all graphs, average±SD was plotted. Akt kinase is considered to be a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]. An important question is whether these same molecules reverse their pro-survival roles during necroptosis. We found that inhibition of mTORC1 by rapamycin, an inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C). Similarly, the direct mTOR kinase inhibitor Torin1 [30] and the dual PI3K/mTOR inhibitor PI-103 [31] also efficiently inhibited necroptosis (Fig. 5C). Knockdown of mTOR using siRNA further validated the small-molecule inhibitor data indicating a role for mTOR in necroptosis by protecting cells from both zVAD.fmk and TNFα induced death (Fig. 5D). mTORC1 regulates translation through activation of p70S6 kinase and, subsequently, ribosomal protein S6 [32]. Notably, a genome-wide siRNA screen [10] suggested an important role for protein translation in necroptosis. Consistently, we found that the small molecule inhibitor of p70S6K PF-4708671 [33] attenuated necroptosis at the concentrations required to block S6 phosphorylation (Fig. S5C, D). Partial siRNA knockdown of S6 protein attenuated necroptosis as well (Fig. S5E), suggesting that translational control by p70S6K/S6 may play a role in necroptosis. Overall, while the full repertoire of Akt targets during necroptosis remains to be fully explored, our data provide evidence that the activity of an anti-apoptotic branch of Akt signaling can promote necroptosis. RIP1 kinase, Akt, mTORC1 and JNK control the upregulation of TNFα accompanying necroptosis. Hitomi et al. [10] have recently reported that the induction of necroptosis by zVAD.fmk in L929 cells is associated with increased synthesis of TNFα, which potentiates cell death. Therefore, we examined whether Akt and its effectors contribute to TNFα synthesis. Consistent with a RIP1-dependent increase in TNFα protein (Fig. S6A, B), we found that TNFα mRNA levels increased during necroptosis in L929 cells in a RIP1 (Fig. S6C. Under serum free conditions, bFGF alone triggered some induction of TNFα mRNA, while its combination with zVAD.fmk (but not zVAD.fmk alone) caused a pronounced further increase (Fig. 6A). Conversely, PDGF caused a modest upregulation of TNFα mRNA, which was not further increased in the presence of zVAD.fmk (Fig. 6A), demonstrating that activation of necroptosis is specifically accompanied by a marked increase in autocrine TNFα synthesis. 10.1371/journal.pone.0056576.g006 Figure 6 Akt and mTORC1 control autocrine TNFα synthesis and JNK activation during necroptosis. (A) Cells were treated under serum free conditions with bFGF or PDGF with or without zVAD.fmk for 9 hr, followed by qRT-PCR analysis of mTNFα. Data was normalized to mouse 18S RNA. (B) Necroptosis was induced by zVAD.fmk or TNFα in cells treated with Nec-1, rapamycin (rapa), or Akt inh. VIII inh. followed by qRT-PCR analysis of TNFα mRNA levels. (C-F) L929 cells with siRNA knockdown of Akt isoforms (C,E) or mTOR (D,F) were stimulated with zVAD.fmk or TNFα for 9 hr, followed by qRT-PCR analysis of mTNFα (C,D) or western blot (E,F). In all graphs, average±SD was plotted. Further analysis suggested that both Akt and mTORC1 contribute to the upregulation of TNFα mRNA during necroptosis as both small-molecule inhibition and siRNA knockdown of Akt and mTOR reduced TNFα mRNA levels in necroptotic cells (Fig. 6B,C,D). Notably, RIP1 and Akt inhibitors had no effect on the levels of TNFα mRNA in control cells or in the cells stimulated with bFGF alone (Fig. 6A,B, Fig. S6C), suggesting that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis. Akt and mTORC1 Control the Activation of JNK during Necroptosis JNK is a well-established regulator of TNFα synthesis in a variety of systems [13], [14], [15], [34]. Therefore, the ability of Akt and mTORC1 inhibitors to block the increase in TNFα mRNA lead us to examine their role in the activation of JNK during necroptosis. Knockdown of Akt isoforms Akt1 and Akt2 or inhibition of Akt prominently suppressed the necroptosis dependent increase in JNK and c-Jun phosphorylation (Fig. 6E, S6D,E) suggesting that Akt may provide a link between RIP1 and JNK activation. Importantly, inhibition of Akt only inhibited the delayed, but not the early, increase in bFGF/zVAD.fmk induced JNK and c-Jun phosphorylation (Fig. S6F). Knockdown of mTOR, rapamycin and the p70S6K inhibitor PF-4708671 also attenuated the necroptosis-associated increase in JNK and c-Jun phosphorylation (Fig. 6F, S6E,G, Fig. S5D). Overall, these data suggested that the Akt-mTORC1-S6K axis, acting downstream from RIP1 kinase, is required for the increase in JNK activity during necroptosis in L929 cells. PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under Necroptotic Conditions Typical regulation of Akt by growth factors involves its recruitment to the plasma membrane, which is mediated by the binding of the pleckstrin homology (PH) domain of Akt to the product of PI3K, phosphatidylinositol-3,4,5-triphosphate (PIP3). In the membrane, Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1) and mTORC2 (or DNA-PK), respectively [35]. Since our results showed that only Thr308 Akt phosphorylation is increased during necroptosis, we next examined whether it is still dependent on PI3K and PDK1. Inhibition of PI3K and PDK1 using the specific inhibitors LY249002 and BX912 [36] resulted in the efficient inhibition of cell death and Akt Thr308 phosphorylation (Fig. S7A–D). Likewise, siRNA knockdown of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation (Fig. S7E,F) Therefore, PI3K and PDK1 activity is still required for non-canonical Akt activation during necroptosis. Expression of Constitutively Active Akt, Rescues Necroptosis Under Serum Free Conditions We used L929 cells stably expressing constitutively active wild type Akt1 (Myr-Akt) or the catalytically inactive mutant K179M in order to further understand the contribution of growth factors and RIP1 kinase to Akt activation during necroptosis. Constitutively active Akt1 (Myr-Akt) was generated as previously described [37] by the addition of a myristoylation signal which provides constitutive localization to the plasma membrane and by the deletion of the auto-inhibitory PH domain (Fig. 7A) resulting in an Akt that is active under serum free. It is important to note that the cells expressing Myr-Akt were viable, grew in a manner indistinguishable from the empty vector control cells, and were not triggered to induce necroptosis by serum starvation (Fig. 7B). This indicates that active Akt alone is not sufficient to induce necroptotic cell death. Under serum free conditions Myr-Akt, but not the K179M mutant, fully restored zVAD.fmk-induced necroptosis (Fig. 7A,B). Nec-1 prevented both Myr-Akt dependent cell death and the necroptosis-specific delayed increase in Akt Thr308 phosphorylation (Fig. 7B,C). Myr-Akt also allowed other zVAD.fmk-dependent events, including activation of JNK and c-Jun phosphorylation (Fig. 7C) and upregulation of TNFα mRNA (Fig. 7D) to occur under serum free conditions, confirming an important role for Akt at the apex of necroptotic signaling. These data demonstrated that the presence of active and membrane localized Akt is sufficient to uncouple Akt activation during necroptosis from growth factor signaling. RIP1 kinase was still able to regulate Akt activation during necroptosis, suggesting that growth factors and RIP1 kinase provide two independent inputs required for Akt changes during necroptosis. 10.1371/journal.pone.0056576.g007 Figure 7 Over expression of constitutively active Akt restores necroptosis under serum free conditions. (A,B) L929 cells were stably infected with empty MSCV retrovirus or viruses encoding Myr-Akt or the catalytically inactive Myr-Akt K179M. Necroptosis was induced by the addition of zVAD.fmk under serum free conditions (A) or serum or serum free conditions with Nec-1 (B). Viability assays were performed after 24 hr. (C) Myr-Akt and Myr-Akt K179M cells were treated with zVAD.fmk and/or Nec-1 under serum free conditions for 9 hr, followed by western blot using the indicated antibodies. Endogenous Akt (∼) and Myr-Akt (*) bands are indicated. (D) L929 cells, stably infected with Myr-Akt and Myr-Akt K179KM, were stimulated with zVAD.fmk for 9 hr under serum free conditions. TNFα mRNA levels were determined by qRT-PCR and normalized using mouse 18S RNA. (E-G) L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473 were treated with zVAD.fmk under serum free conditions, followed by viability assay at 24 hr (E), western blot at 9 hr (F), or evaluation of TNFα mRNA levels by qRT-PCR at 9 hrs (G). In all graphs, average±SD was plotted. RIP1 kinase-dependent Thr308 phosphorylation of Myr-Akt during necroptosis","RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis Given our observation that growth factors are important for zVAD.fmk induced death, we examined the contribution of several pathways, including MAPK pathways and Akt, which are known to be activated following growth factor receptor activation (Fig. 2A). Inhibition of Akt (Akt inhibitor VIII) strongly protected the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16] as well as cell death triggered by bFGF or IGF-1/zVAD.fmk under serum free conditions (Fig. 2B). Inhibition of Akt also protected the cells from growth-factor insensitive death by caused by TNFα (Fig. 2A). Consistent with previous reports, the JNK inhibitor SP600125 protected the cells from both zVAD.fmk and TNFα induced death (Fig. 2A,B and Fig. S2A) [12], [14]. In contrast, inhibition of two other MAPKs, p38 and ERK, previously reported not to be activated during necroptosis [14], did not protect from either zVAD.fmk or TNFα induced death (Fig. 2A). 10.1371/journal.pone.0056576.g002 Figure 2 Akt contributes to necroptosis induced by zVAD.fmk and TNFα. (A,B) Necroptosis was induced by zVAD.fmk or TNFα (full serum, A) or growth factors/zVAD.fmk (serum free, B) in the presence of inhibitors of Akt (Akt inhibitor VIII), JNK (SP600125), p38 (PD169316), and Erk (UO126). Cell viability was determined after 24 hrs. (C) L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs were treated with zVAD.fmk or TNFα for 9 hrs. Cell viability and Akt expression levels were determined after 24 hrs. In all graphs, average±SD was plotted. Next, we used two approaches to further validate the role of Akt in necroptotic cell death. First, two additional Akt inhibitors, a highly specific, allosteric kinase inhibitor MK-2206 [25] and triciribine (TCN) [26], which blocks membrane translocation of Akt, both attenuated cell death (Fig. S2B). Secondly, simultaneous knockdown of Akt isoforms Akt1 and Akt2 using siRNAs protected cells from necroptosis induced by both zVAD.fmk and TNFα (Fig. 2C). No expression of Akt3 was seen in L929 cells (Fig. S2C) and, consistently, Akt3 siRNA had no additional effect on necroptosis. Our results confirmed that Akt plays a key role in necroptosis induced by multiple stimuli in L929 cells. To understand the activation of Akt and JNK under necroptotic conditions, we examined the changes in Akt and JNK phosphorylation at 9 hrs post zVAD.fmk and TNFα stimulation. This time point was chosen because it reflects the early stage of cell death in our system (Fig. S3A, B). Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation at a known major activation site, Thr308 (Fig. 3A). Interestingly, we did not observe concomitant phosphorylation changes in the second major activation site of Akt, Ser473. We also observed an increase in the phosphorylation of both the p46 and p54 isoforms of JNK and its major substrate c-Jun (Fig. 3A). These data indicate that both Akt and JNK are activated under necroptotic conditions. 10.1371/journal.pone.0056576.g003 Figure 3 RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr, followed by western blotting with indicated antibodies. (B,C) L929 cells were treated with zVAD.fmk (B) or bFGF/zVAD.fmk (serum free conditions, C) and samples were collected at the indicated time points for western blot. (D) Nec-1 was added to the cells stimulated with bFGF or bFGF/zVAD (serum free conditions) for 15 min or 9 hr followed by western blot with the indicated antibodies. The RIP1 kinase inhibitor, Nec-1, completely prevented the increase in Thr308 Akt phosphorylation, while Nec-1i did not (Fig. 3A, Fig. S1D). Similarly, Nec-1 prevented the induction of JNK phosphorylation in response to zVAD.fmk and substantially reduced this change after TNFα addition. We observed some changes in total protein levels of JNK and c-Jun following necroptotic stimulation. Some of these changes, e.g. zVAD.fmk-induced increase in c-Jun, were also attenuated by Nec-1. Importantly, Nec-1 did not alter the basal phosphorylation levels of either Akt or JNK (Fig. 3A). This established that Akt Thr308 and JNK phosphorylation during necroptosis is RIP1 dependent. Interestingly, we discovered that the phosphorylation of Akt Thr308, JNK and Jun are late events following zVAD.fmk stimulation (Fig. 3B) that coincide with the onset of necroptosis at 6 hr post-stimulation (Fig. S3A). To better understand the contributions of growth factors and RIP1 kinase to necroptotic regulation of Akt, we next analyzed the time course of these phosphorylation changes under serum free conditions. We found that the addition of bFGF alone or in combination with zVAD.fmk led to a substantial rapid and transient increase in both Thr308 and Ser473 phosphorylation of Akt as well as JNK and c-Jun at 15 minutes, reflecting the expected response to growth factor stimulation (Fig. 3C). Significantly, the combination of bFGF/zVAD.fmk, but not bFGF alone, also caused a robust, second, delayed increase in the phosphorylation of Thr308, but not Ser473, of Akt as well as a delayed increase in the phosphorylation of JNK and Jun. Furthermore, Nec-1 had no significant effect on the early increase in both Akt and JNK/c-Jun phosphorylation triggered by both bFGF and bFGF/zVAD, while Nec-1, but not its inactive analog Nec-1i (Fig. S1E), efficiently blocked the bFGF/zVAD increase at 6–9 hr (Fig. 3D), suggesting that only the delayed activation of Akt and JNK is specific for necroptosis and dependent on RIP1 kinase activity. Similarly, IGF/zVAD, which also promoted cell death under serum free conditions, produced a delayed increase in Thr308 phosphorylation on Akt, while IGF alone caused solely an early, transient increase in phosphorylation (Fig. S3C). We confirmed the kinetics of the Akt Thr308 and Ser473 phosphorylation changes using a quantitative ELISA assay, which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation (Fig. S3D, E). Taken together, these results indicate that the observed delayed increases in Akt and JNK phosphorylation, preceding the onset of cell death, represent specific consequences of necroptotic signaling downstream from RIP1 kinase. TNFα Induces Delayed Akt Thr308 Phosphorylation and Necroptosis Independent of Growth Factor Stimulation Consistent with TNFα inducing necroptosis independently of growth factors (Fig. 1A), FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation, while efficiently preventing these changes in response to zVAD.fmk (Fig. S4A). Furthermore, addition of TNFα led to comparable late activation of Akt p308 signal under both normal and serum free conditions (Fig. S4B, C), indicating that TNFα signaling to Akt Thr308 is growth factor-independent. In contrast, activation of JNK by TNFα followed different kinetics from zVAD.fmk-induced changes. TNFα treatment caused an early and robust increase in the phosphorylation of JNK and c-Jun. Nec-1 did not affect this early increase, however, it reduced levels of pJNK/Jun at the late, 9 hr time point (Fig. S4B, C). This again separated early RIP1-independent changes, which likely reflect the ability of additional upstream kinases, such as Ask1 to activate JNK [27], from the late RIP1 kinase-dependent necroptotic signaling. Late Increase in Akt Thr308 Phosphorylation Contributes to the Induction of Necroptotic Cell Death We next investigated if the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation functionally contributes to the execution of necroptotic cell death. Firstly, PDGF/zVAD.fmk, which cannot induce necroptosis (Fig. 2A), triggered only the initial, rapid Akt and JNK phosphorylation changes and not the delayed activation (Fig. 4A), indicating that late, rather than early Akt phosphorylation correlates with necroptosis. Secondly, we saw that the ability of the Akt inhibitor to protect cells from necroptosis rapidly declined after 6 hrs of stimulation with zVAD.fmk, TNFα or bFGF/zVAD.fmk and no protection was observed when the inhibitor was added at 9 hrs (Fig. 4B,C). This time frame coincides with the timing of the secondary Akt Thr308 phosphorylation. Finally, we terminated the bFGF signal one hour after addition of bFGF by the addition of PD173074. This allowed us to retain early Akt activation, but to suppress the secondary increase (Fig. 4D). Both pre-addition and delayed addition of PD173074 fully prevented necroptosis (Fig. 4E). Overall, these data, while correlative, indicate that early Akt activation is insufficient to promote necroptosis and are strongly supportive of an important role for the delayed activation of Akt in the induction of necroptotic cell death. 10.1371/journal.pone.0056576.g004 Figure 4 Late Thr308 phosphorylation of Akt contributes to necroptosis. (A) L929 cells were treated with zVAD.fmk and bFGF or PDGF, with or without Nec-1, for the indicated periods of time. (B,C) L929 cells were stimulated by zVAD.fmk or TNFα (B) or bFGF/zVAD.fmk under serum free conditions (C). Akt inh. VIII was added 15 min before necroptotic stimulation (Pre) or at indicated times after stimulation. Viability was measured 24 hr after activation of necroptosis. (D) L929 cells were stimulated with bFGF/zVAD under serum free conditions. PD173074 was added 15 min before or 1 hr after FGF/zVAD. Samples for western blot were collected at 15 min and 9 hr time points. (E) Cells were pretreated with PD173074 or it was added 1 hr after bFGF/zVAD.fmk, followed by viability assessment at 24 hr. In all graphs, average±SD was plotted. The Akt Signaling Pathway Contributes to the Regulation of Necroptosis We next determined whether the necroptosis-associated increase in Thr308 phosphorylation results in an increase in Akt kinase activity. Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates (Forkhead box class O (FoxO) proteins, GSK-3 kinases and mouse double minute 2 (MDM2)) as well as downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6) (Fig. 5A). In some cases (FoxO1, FoxO4, MDM2), a robust increase was observed. In other cases (FoxO3a, GSK-3α/β, p70S6K and its substrate S6), the changes were less pronounced (Fig. 5A). The timing of the phosphorylation changes paralleled the increase in Akt phosphorylation (Fig. 5B, S5A, B). In the case of pFoxO1 we occasionally observed a shift in migration rather than an increase in band intensity (e.g. comparing Fig. 5A and B), suggesting that phosphorylation events in addition to Thr24 take place during necroptosis. Notably, in all cases the necroptosis-associated increases in Akt substrates were abrogated by Nec-1 (Fig. 5A, Fig. S5A, B). Overall, these data suggested that a significant part of the “canonical” Akt signaling network is activated at the onset of necroptotic cell death in a RIP1 dependent fashion. 10.1371/journal.pone.0056576.g005 Figure 5 mTORC1 contributes to the regulation of necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr and harvested for western blot. (B) Cell under serum free condition were treated with bFGF or bFGF/zVAD.fmk for the indicated amounts of time, followed by western blotting using the indicated antibodies. (C) Necroptosis was induced by zVAD.fmk or TNFα in L929 cell in the presence of inhibitors of Akt(Akt inh. VIII) and mTOR (rapamycin, Torin-1 and PI-103). (D) L929 cells with mTOR siRNA knockdown were harvested for western blot or treated with zVAD.fmk or TNFα for 24 hrs. Cell viability was determined 24 hr after activation of necroptosis. In all graphs, average±SD was plotted. Akt kinase is considered to be a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]. An important question is whether these same molecules reverse their pro-survival roles during necroptosis. We found that inhibition of mTORC1 by rapamycin, an inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C). Similarly, the direct mTOR kinase inhibitor Torin1 [30] and the dual PI3K/mTOR inhibitor PI-103 [31] also efficiently inhibited necroptosis (Fig. 5C). Knockdown of mTOR using siRNA further validated the small-molecule inhibitor data indicating a role for mTOR in necroptosis by protecting cells from both zVAD.fmk and TNFα induced death (Fig. 5D). mTORC1 regulates translation through activation of p70S6 kinase and, subsequently, ribosomal protein S6 [32]. Notably, a genome-wide siRNA screen [10] suggested an important role for protein translation in necroptosis. Consistently, we found that the small molecule inhibitor of p70S6K PF-4708671 [33] attenuated necroptosis at the concentrations required to block S6 phosphorylation (Fig. S5C, D). Partial siRNA knockdown of S6 protein attenuated necroptosis as well (Fig. S5E), suggesting that translational control by p70S6K/S6 may play a role in necroptosis. Overall, while the full repertoire of Akt targets during necroptosis remains to be fully explored, our data provide evidence that the activity of an anti-apoptotic branch of Akt signaling can promote necroptosis. RIP1 kinase, Akt, mTORC1 and JNK control the upregulation of TNFα accompanying necroptosis. Hitomi et al. [10] have recently reported that the induction of necroptosis by zVAD.fmk in L929 cells is associated with increased synthesis of TNFα, which potentiates cell death. Therefore, we examined whether Akt and its effectors contribute to TNFα synthesis. Consistent with a RIP1-dependent increase in TNFα protein (Fig. S6A, B), we found that TNFα mRNA levels increased during necroptosis in L929 cells in a RIP1 (Fig. S6C. Under serum free conditions, bFGF alone triggered some induction of TNFα mRNA, while its combination with zVAD.fmk (but not zVAD.fmk alone) caused a pronounced further increase (Fig. 6A). Conversely, PDGF caused a modest upregulation of TNFα mRNA, which was not further increased in the presence of zVAD.fmk (Fig. 6A), demonstrating that activation of necroptosis is specifically accompanied by a marked increase in autocrine TNFα synthesis. 10.1371/journal.pone.0056576.g006 Figure 6 Akt and mTORC1 control autocrine TNFα synthesis and JNK activation during necroptosis. (A) Cells were treated under serum free conditions with bFGF or PDGF with or without zVAD.fmk for 9 hr, followed by qRT-PCR analysis of mTNFα. Data was normalized to mouse 18S RNA. (B) Necroptosis was induced by zVAD.fmk or TNFα in cells treated with Nec-1, rapamycin (rapa), or Akt inh. VIII inh. followed by qRT-PCR analysis of TNFα mRNA levels. (C-F) L929 cells with siRNA knockdown of Akt isoforms (C,E) or mTOR (D,F) were stimulated with zVAD.fmk or TNFα for 9 hr, followed by qRT-PCR analysis of mTNFα (C,D) or western blot (E,F). In all graphs, average±SD was plotted. Further analysis suggested that both Akt and mTORC1 contribute to the upregulation of TNFα mRNA during necroptosis as both small-molecule inhibition and siRNA knockdown of Akt and mTOR reduced TNFα mRNA levels in necroptotic cells (Fig. 6B,C,D). Notably, RIP1 and Akt inhibitors had no effect on the levels of TNFα mRNA in control cells or in the cells stimulated with bFGF alone (Fig. 6A,B, Fig. S6C), suggesting that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis. Akt and mTORC1 Control the Activation of JNK during Necroptosis JNK is a well-established regulator of TNFα synthesis in a variety of systems [13], [14], [15], [34]. Therefore, the ability of Akt and mTORC1 inhibitors to block the increase in TNFα mRNA lead us to examine their role in the activation of JNK during necroptosis. Knockdown of Akt isoforms Akt1 and Akt2 or inhibition of Akt prominently suppressed the necroptosis dependent increase in JNK and c-Jun phosphorylation (Fig. 6E, S6D,E) suggesting that Akt may provide a link between RIP1 and JNK activation. Importantly, inhibition of Akt only inhibited the delayed, but not the early, increase in bFGF/zVAD.fmk induced JNK and c-Jun phosphorylation (Fig. S6F). Knockdown of mTOR, rapamycin and the p70S6K inhibitor PF-4708671 also attenuated the necroptosis-associated increase in JNK and c-Jun phosphorylation (Fig. 6F, S6E,G, Fig. S5D). Overall, these data suggested that the Akt-mTORC1-S6K axis, acting downstream from RIP1 kinase, is required for the increase in JNK activity during necroptosis in L929 cells. PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under Necroptotic Conditions Typical regulation of Akt by growth factors involves its recruitment to the plasma membrane, which is mediated by the binding of the pleckstrin homology (PH) domain of Akt to the product of PI3K, phosphatidylinositol-3,4,5-triphosphate (PIP3). In the membrane, Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1) and mTORC2 (or DNA-PK), respectively [35]. Since our results showed that only Thr308 Akt phosphorylation is increased during necroptosis, we next examined whether it is still dependent on PI3K and PDK1. Inhibition of PI3K and PDK1 using the specific inhibitors LY249002 and BX912 [36] resulted in the efficient inhibition of cell death and Akt Thr308 phosphorylation (Fig. S7A–D). Likewise, siRNA knockdown of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation (Fig. S7E,F) Therefore, PI3K and PDK1 activity is still required for non-canonical Akt activation during necroptosis. Expression of Constitutively Active Akt, Rescues Necroptosis Under Serum Free Conditions We used L929 cells stably expressing constitutively active wild type Akt1 (Myr-Akt) or the catalytically inactive mutant K179M in order to further understand the contribution of growth factors and RIP1 kinase to Akt activation during necroptosis. Constitutively active Akt1 (Myr-Akt) was generated as previously described [37] by the addition of a myristoylation signal which provides constitutive localization to the plasma membrane and by the deletion of the auto-inhibitory PH domain (Fig. 7A) resulting in an Akt that is active under serum free. It is important to note that the cells expressing Myr-Akt were viable, grew in a manner indistinguishable from the empty vector control cells, and were not triggered to induce necroptosis by serum starvation (Fig. 7B). This indicates that active Akt alone is not sufficient to induce necroptotic cell death. Under serum free conditions Myr-Akt, but not the K179M mutant, fully restored zVAD.fmk-induced necroptosis (Fig. 7A,B). Nec-1 prevented both Myr-Akt dependent cell death and the necroptosis-specific delayed increase in Akt Thr308 phosphorylation (Fig. 7B,C). Myr-Akt also allowed other zVAD.fmk-dependent events, including activation of JNK and c-Jun phosphorylation (Fig. 7C) and upregulation of TNFα mRNA (Fig. 7D) to occur under serum free conditions, confirming an important role for Akt at the apex of necroptotic signaling. These data demonstrated that the presence of active and membrane localized Akt is sufficient to uncouple Akt activation during necroptosis from growth factor signaling. RIP1 kinase was still able to regulate Akt activation during necroptosis, suggesting that growth factors and RIP1 kinase provide two independent inputs required for Akt changes during necroptosis. 10.1371/journal.pone.0056576.g007 Figure 7 Over expression of constitutively active Akt restores necroptosis under serum free conditions. (A,B) L929 cells were stably infected with empty MSCV retrovirus or viruses encoding Myr-Akt or the catalytically inactive Myr-Akt K179M. Necroptosis was induced by the addition of zVAD.fmk under serum free conditions (A) or serum or serum free conditions with Nec-1 (B). Viability assays were performed after 24 hr. (C) Myr-Akt and Myr-Akt K179M cells were treated with zVAD.fmk and/or Nec-1 under serum free conditions for 9 hr, followed by western blot using the indicated antibodies. Endogenous Akt (∼) and Myr-Akt (*) bands are indicated. (D) L929 cells, stably infected with Myr-Akt and Myr-Akt K179KM, were stimulated with zVAD.fmk for 9 hr under serum free conditions. TNFα mRNA levels were determined by qRT-PCR and normalized using mouse 18S RNA. (E-G) L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473 were treated with zVAD.fmk under serum free conditions, followed by viability assay at 24 hr (E), western blot at 9 hr (F), or evaluation of TNFα mRNA levels by qRT-PCR at 9 hrs (G). In all graphs, average±SD was plotted. RIP1 kinase-dependent Thr308 phosphorylation of Myr-Akt during necroptosis"
R10242 T15756 T15763 causeOf "RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis Given our observation that growth factors are important for zVAD.fmk induced death, we examined the contribution of several pathways, including MAPK pathways and Akt, which are known to be activated following growth factor receptor activation (Fig. 2A). Inhibition of Akt (Akt inhibitor VIII) strongly protected the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16] as well as cell death triggered by bFGF or IGF-1/zVAD.fmk under serum free conditions (Fig. 2B). Inhibition of Akt also protected the cells from growth-factor insensitive death by caused by TNFα (Fig. 2A). Consistent with previous reports, the JNK inhibitor SP600125 protected the cells from both zVAD.fmk and TNFα induced death (Fig. 2A,B and Fig. S2A) [12], [14]. In contrast, inhibition of two other MAPKs, p38 and ERK, previously reported not to be activated during necroptosis [14], did not protect from either zVAD.fmk or TNFα induced death (Fig. 2A). 10.1371/journal.pone.0056576.g002 Figure 2 Akt contributes to necroptosis induced by zVAD.fmk and TNFα. (A,B) Necroptosis was induced by zVAD.fmk or TNFα (full serum, A) or growth factors/zVAD.fmk (serum free, B) in the presence of inhibitors of Akt (Akt inhibitor VIII), JNK (SP600125), p38 (PD169316), and Erk (UO126). Cell viability was determined after 24 hrs. (C) L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs were treated with zVAD.fmk or TNFα for 9 hrs. Cell viability and Akt expression levels were determined after 24 hrs. In all graphs, average±SD was plotted. Next, we used two approaches to further validate the role of Akt in necroptotic cell death. First, two additional Akt inhibitors, a highly specific, allosteric kinase inhibitor MK-2206 [25] and triciribine (TCN) [26], which blocks membrane translocation of Akt, both attenuated cell death (Fig. S2B). Secondly, simultaneous knockdown of Akt isoforms Akt1 and Akt2 using siRNAs protected cells from necroptosis induced by both zVAD.fmk and TNFα (Fig. 2C). No expression of Akt3 was seen in L929 cells (Fig. S2C) and, consistently, Akt3 siRNA had no additional effect on necroptosis. Our results confirmed that Akt plays a key role in necroptosis induced by multiple stimuli in L929 cells. To understand the activation of Akt and JNK under necroptotic conditions, we examined the changes in Akt and JNK phosphorylation at 9 hrs post zVAD.fmk and TNFα stimulation. This time point was chosen because it reflects the early stage of cell death in our system (Fig. S3A, B). Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation at a known major activation site, Thr308 (Fig. 3A). Interestingly, we did not observe concomitant phosphorylation changes in the second major activation site of Akt, Ser473. We also observed an increase in the phosphorylation of both the p46 and p54 isoforms of JNK and its major substrate c-Jun (Fig. 3A). These data indicate that both Akt and JNK are activated under necroptotic conditions. 10.1371/journal.pone.0056576.g003 Figure 3 RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr, followed by western blotting with indicated antibodies. (B,C) L929 cells were treated with zVAD.fmk (B) or bFGF/zVAD.fmk (serum free conditions, C) and samples were collected at the indicated time points for western blot. (D) Nec-1 was added to the cells stimulated with bFGF or bFGF/zVAD (serum free conditions) for 15 min or 9 hr followed by western blot with the indicated antibodies. The RIP1 kinase inhibitor, Nec-1, completely prevented the increase in Thr308 Akt phosphorylation, while Nec-1i did not (Fig. 3A, Fig. S1D). Similarly, Nec-1 prevented the induction of JNK phosphorylation in response to zVAD.fmk and substantially reduced this change after TNFα addition. We observed some changes in total protein levels of JNK and c-Jun following necroptotic stimulation. Some of these changes, e.g. zVAD.fmk-induced increase in c-Jun, were also attenuated by Nec-1. Importantly, Nec-1 did not alter the basal phosphorylation levels of either Akt or JNK (Fig. 3A). This established that Akt Thr308 and JNK phosphorylation during necroptosis is RIP1 dependent. Interestingly, we discovered that the phosphorylation of Akt Thr308, JNK and Jun are late events following zVAD.fmk stimulation (Fig. 3B) that coincide with the onset of necroptosis at 6 hr post-stimulation (Fig. S3A). To better understand the contributions of growth factors and RIP1 kinase to necroptotic regulation of Akt, we next analyzed the time course of these phosphorylation changes under serum free conditions. We found that the addition of bFGF alone or in combination with zVAD.fmk led to a substantial rapid and transient increase in both Thr308 and Ser473 phosphorylation of Akt as well as JNK and c-Jun at 15 minutes, reflecting the expected response to growth factor stimulation (Fig. 3C). Significantly, the combination of bFGF/zVAD.fmk, but not bFGF alone, also caused a robust, second, delayed increase in the phosphorylation of Thr308, but not Ser473, of Akt as well as a delayed increase in the phosphorylation of JNK and Jun. Furthermore, Nec-1 had no significant effect on the early increase in both Akt and JNK/c-Jun phosphorylation triggered by both bFGF and bFGF/zVAD, while Nec-1, but not its inactive analog Nec-1i (Fig. S1E), efficiently blocked the bFGF/zVAD increase at 6–9 hr (Fig. 3D), suggesting that only the delayed activation of Akt and JNK is specific for necroptosis and dependent on RIP1 kinase activity. Similarly, IGF/zVAD, which also promoted cell death under serum free conditions, produced a delayed increase in Thr308 phosphorylation on Akt, while IGF alone caused solely an early, transient increase in phosphorylation (Fig. S3C). We confirmed the kinetics of the Akt Thr308 and Ser473 phosphorylation changes using a quantitative ELISA assay, which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation (Fig. S3D, E). Taken together, these results indicate that the observed delayed increases in Akt and JNK phosphorylation, preceding the onset of cell death, represent specific consequences of necroptotic signaling downstream from RIP1 kinase. TNFα Induces Delayed Akt Thr308 Phosphorylation and Necroptosis Independent of Growth Factor Stimulation Consistent with TNFα inducing necroptosis independently of growth factors (Fig. 1A), FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation, while efficiently preventing these changes in response to zVAD.fmk (Fig. S4A). Furthermore, addition of TNFα led to comparable late activation of Akt p308 signal under both normal and serum free conditions (Fig. S4B, C), indicating that TNFα signaling to Akt Thr308 is growth factor-independent. In contrast, activation of JNK by TNFα followed different kinetics from zVAD.fmk-induced changes. TNFα treatment caused an early and robust increase in the phosphorylation of JNK and c-Jun. Nec-1 did not affect this early increase, however, it reduced levels of pJNK/Jun at the late, 9 hr time point (Fig. S4B, C). This again separated early RIP1-independent changes, which likely reflect the ability of additional upstream kinases, such as Ask1 to activate JNK [27], from the late RIP1 kinase-dependent necroptotic signaling. Late Increase in Akt Thr308 Phosphorylation Contributes to the Induction of Necroptotic Cell Death We next investigated if the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation functionally contributes to the execution of necroptotic cell death. Firstly, PDGF/zVAD.fmk, which cannot induce necroptosis (Fig. 2A), triggered only the initial, rapid Akt and JNK phosphorylation changes and not the delayed activation (Fig. 4A), indicating that late, rather than early Akt phosphorylation correlates with necroptosis. Secondly, we saw that the ability of the Akt inhibitor to protect cells from necroptosis rapidly declined after 6 hrs of stimulation with zVAD.fmk, TNFα or bFGF/zVAD.fmk and no protection was observed when the inhibitor was added at 9 hrs (Fig. 4B,C). This time frame coincides with the timing of the secondary Akt Thr308 phosphorylation. Finally, we terminated the bFGF signal one hour after addition of bFGF by the addition of PD173074. This allowed us to retain early Akt activation, but to suppress the secondary increase (Fig. 4D). Both pre-addition and delayed addition of PD173074 fully prevented necroptosis (Fig. 4E). Overall, these data, while correlative, indicate that early Akt activation is insufficient to promote necroptosis and are strongly supportive of an important role for the delayed activation of Akt in the induction of necroptotic cell death. 10.1371/journal.pone.0056576.g004 Figure 4 Late Thr308 phosphorylation of Akt contributes to necroptosis. (A) L929 cells were treated with zVAD.fmk and bFGF or PDGF, with or without Nec-1, for the indicated periods of time. (B,C) L929 cells were stimulated by zVAD.fmk or TNFα (B) or bFGF/zVAD.fmk under serum free conditions (C). Akt inh. VIII was added 15 min before necroptotic stimulation (Pre) or at indicated times after stimulation. Viability was measured 24 hr after activation of necroptosis. (D) L929 cells were stimulated with bFGF/zVAD under serum free conditions. PD173074 was added 15 min before or 1 hr after FGF/zVAD. Samples for western blot were collected at 15 min and 9 hr time points. (E) Cells were pretreated with PD173074 or it was added 1 hr after bFGF/zVAD.fmk, followed by viability assessment at 24 hr. In all graphs, average±SD was plotted. The Akt Signaling Pathway Contributes to the Regulation of Necroptosis We next determined whether the necroptosis-associated increase in Thr308 phosphorylation results in an increase in Akt kinase activity. Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates (Forkhead box class O (FoxO) proteins, GSK-3 kinases and mouse double minute 2 (MDM2)) as well as downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6) (Fig. 5A). In some cases (FoxO1, FoxO4, MDM2), a robust increase was observed. In other cases (FoxO3a, GSK-3α/β, p70S6K and its substrate S6), the changes were less pronounced (Fig. 5A). The timing of the phosphorylation changes paralleled the increase in Akt phosphorylation (Fig. 5B, S5A, B). In the case of pFoxO1 we occasionally observed a shift in migration rather than an increase in band intensity (e.g. comparing Fig. 5A and B), suggesting that phosphorylation events in addition to Thr24 take place during necroptosis. Notably, in all cases the necroptosis-associated increases in Akt substrates were abrogated by Nec-1 (Fig. 5A, Fig. S5A, B). Overall, these data suggested that a significant part of the “canonical” Akt signaling network is activated at the onset of necroptotic cell death in a RIP1 dependent fashion. 10.1371/journal.pone.0056576.g005 Figure 5 mTORC1 contributes to the regulation of necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr and harvested for western blot. (B) Cell under serum free condition were treated with bFGF or bFGF/zVAD.fmk for the indicated amounts of time, followed by western blotting using the indicated antibodies. (C) Necroptosis was induced by zVAD.fmk or TNFα in L929 cell in the presence of inhibitors of Akt(Akt inh. VIII) and mTOR (rapamycin, Torin-1 and PI-103). (D) L929 cells with mTOR siRNA knockdown were harvested for western blot or treated with zVAD.fmk or TNFα for 24 hrs. Cell viability was determined 24 hr after activation of necroptosis. In all graphs, average±SD was plotted. Akt kinase is considered to be a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]. An important question is whether these same molecules reverse their pro-survival roles during necroptosis. We found that inhibition of mTORC1 by rapamycin, an inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C). Similarly, the direct mTOR kinase inhibitor Torin1 [30] and the dual PI3K/mTOR inhibitor PI-103 [31] also efficiently inhibited necroptosis (Fig. 5C). Knockdown of mTOR using siRNA further validated the small-molecule inhibitor data indicating a role for mTOR in necroptosis by protecting cells from both zVAD.fmk and TNFα induced death (Fig. 5D). mTORC1 regulates translation through activation of p70S6 kinase and, subsequently, ribosomal protein S6 [32]. Notably, a genome-wide siRNA screen [10] suggested an important role for protein translation in necroptosis. Consistently, we found that the small molecule inhibitor of p70S6K PF-4708671 [33] attenuated necroptosis at the concentrations required to block S6 phosphorylation (Fig. S5C, D). Partial siRNA knockdown of S6 protein attenuated necroptosis as well (Fig. S5E), suggesting that translational control by p70S6K/S6 may play a role in necroptosis. Overall, while the full repertoire of Akt targets during necroptosis remains to be fully explored, our data provide evidence that the activity of an anti-apoptotic branch of Akt signaling can promote necroptosis. RIP1 kinase, Akt, mTORC1 and JNK control the upregulation of TNFα accompanying necroptosis. Hitomi et al. [10] have recently reported that the induction of necroptosis by zVAD.fmk in L929 cells is associated with increased synthesis of TNFα, which potentiates cell death. Therefore, we examined whether Akt and its effectors contribute to TNFα synthesis. Consistent with a RIP1-dependent increase in TNFα protein (Fig. S6A, B), we found that TNFα mRNA levels increased during necroptosis in L929 cells in a RIP1 (Fig. S6C. Under serum free conditions, bFGF alone triggered some induction of TNFα mRNA, while its combination with zVAD.fmk (but not zVAD.fmk alone) caused a pronounced further increase (Fig. 6A). Conversely, PDGF caused a modest upregulation of TNFα mRNA, which was not further increased in the presence of zVAD.fmk (Fig. 6A), demonstrating that activation of necroptosis is specifically accompanied by a marked increase in autocrine TNFα synthesis. 10.1371/journal.pone.0056576.g006 Figure 6 Akt and mTORC1 control autocrine TNFα synthesis and JNK activation during necroptosis. (A) Cells were treated under serum free conditions with bFGF or PDGF with or without zVAD.fmk for 9 hr, followed by qRT-PCR analysis of mTNFα. Data was normalized to mouse 18S RNA. (B) Necroptosis was induced by zVAD.fmk or TNFα in cells treated with Nec-1, rapamycin (rapa), or Akt inh. VIII inh. followed by qRT-PCR analysis of TNFα mRNA levels. (C-F) L929 cells with siRNA knockdown of Akt isoforms (C,E) or mTOR (D,F) were stimulated with zVAD.fmk or TNFα for 9 hr, followed by qRT-PCR analysis of mTNFα (C,D) or western blot (E,F). In all graphs, average±SD was plotted. Further analysis suggested that both Akt and mTORC1 contribute to the upregulation of TNFα mRNA during necroptosis as both small-molecule inhibition and siRNA knockdown of Akt and mTOR reduced TNFα mRNA levels in necroptotic cells (Fig. 6B,C,D). Notably, RIP1 and Akt inhibitors had no effect on the levels of TNFα mRNA in control cells or in the cells stimulated with bFGF alone (Fig. 6A,B, Fig. S6C), suggesting that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis. Akt and mTORC1 Control the Activation of JNK during Necroptosis JNK is a well-established regulator of TNFα synthesis in a variety of systems [13], [14], [15], [34]. Therefore, the ability of Akt and mTORC1 inhibitors to block the increase in TNFα mRNA lead us to examine their role in the activation of JNK during necroptosis. Knockdown of Akt isoforms Akt1 and Akt2 or inhibition of Akt prominently suppressed the necroptosis dependent increase in JNK and c-Jun phosphorylation (Fig. 6E, S6D,E) suggesting that Akt may provide a link between RIP1 and JNK activation. Importantly, inhibition of Akt only inhibited the delayed, but not the early, increase in bFGF/zVAD.fmk induced JNK and c-Jun phosphorylation (Fig. S6F). Knockdown of mTOR, rapamycin and the p70S6K inhibitor PF-4708671 also attenuated the necroptosis-associated increase in JNK and c-Jun phosphorylation (Fig. 6F, S6E,G, Fig. S5D). Overall, these data suggested that the Akt-mTORC1-S6K axis, acting downstream from RIP1 kinase, is required for the increase in JNK activity during necroptosis in L929 cells. PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under Necroptotic Conditions Typical regulation of Akt by growth factors involves its recruitment to the plasma membrane, which is mediated by the binding of the pleckstrin homology (PH) domain of Akt to the product of PI3K, phosphatidylinositol-3,4,5-triphosphate (PIP3). In the membrane, Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1) and mTORC2 (or DNA-PK), respectively [35]. Since our results showed that only Thr308 Akt phosphorylation is increased during necroptosis, we next examined whether it is still dependent on PI3K and PDK1. Inhibition of PI3K and PDK1 using the specific inhibitors LY249002 and BX912 [36] resulted in the efficient inhibition of cell death and Akt Thr308 phosphorylation (Fig. S7A–D). Likewise, siRNA knockdown of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation (Fig. S7E,F) Therefore, PI3K and PDK1 activity is still required for non-canonical Akt activation during necroptosis. Expression of Constitutively Active Akt, Rescues Necroptosis Under Serum Free Conditions We used L929 cells stably expressing constitutively active wild type Akt1 (Myr-Akt) or the catalytically inactive mutant K179M in order to further understand the contribution of growth factors and RIP1 kinase to Akt activation during necroptosis. Constitutively active Akt1 (Myr-Akt) was generated as previously described [37] by the addition of a myristoylation signal which provides constitutive localization to the plasma membrane and by the deletion of the auto-inhibitory PH domain (Fig. 7A) resulting in an Akt that is active under serum free. It is important to note that the cells expressing Myr-Akt were viable, grew in a manner indistinguishable from the empty vector control cells, and were not triggered to induce necroptosis by serum starvation (Fig. 7B). This indicates that active Akt alone is not sufficient to induce necroptotic cell death. Under serum free conditions Myr-Akt, but not the K179M mutant, fully restored zVAD.fmk-induced necroptosis (Fig. 7A,B). Nec-1 prevented both Myr-Akt dependent cell death and the necroptosis-specific delayed increase in Akt Thr308 phosphorylation (Fig. 7B,C). Myr-Akt also allowed other zVAD.fmk-dependent events, including activation of JNK and c-Jun phosphorylation (Fig. 7C) and upregulation of TNFα mRNA (Fig. 7D) to occur under serum free conditions, confirming an important role for Akt at the apex of necroptotic signaling. These data demonstrated that the presence of active and membrane localized Akt is sufficient to uncouple Akt activation during necroptosis from growth factor signaling. RIP1 kinase was still able to regulate Akt activation during necroptosis, suggesting that growth factors and RIP1 kinase provide two independent inputs required for Akt changes during necroptosis. 10.1371/journal.pone.0056576.g007 Figure 7 Over expression of constitutively active Akt restores necroptosis under serum free conditions. (A,B) L929 cells were stably infected with empty MSCV retrovirus or viruses encoding Myr-Akt or the catalytically inactive Myr-Akt K179M. Necroptosis was induced by the addition of zVAD.fmk under serum free conditions (A) or serum or serum free conditions with Nec-1 (B). Viability assays were performed after 24 hr. (C) Myr-Akt and Myr-Akt K179M cells were treated with zVAD.fmk and/or Nec-1 under serum free conditions for 9 hr, followed by western blot using the indicated antibodies. Endogenous Akt (∼) and Myr-Akt (*) bands are indicated. (D) L929 cells, stably infected with Myr-Akt and Myr-Akt K179KM, were stimulated with zVAD.fmk for 9 hr under serum free conditions. TNFα mRNA levels were determined by qRT-PCR and normalized using mouse 18S RNA. (E-G) L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473 were treated with zVAD.fmk under serum free conditions, followed by viability assay at 24 hr (E), western blot at 9 hr (F), or evaluation of TNFα mRNA levels by qRT-PCR at 9 hrs (G). In all graphs, average±SD was plotted. RIP1 kinase-dependent Thr308 phosphorylation of Myr-Akt during necroptosis","RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types. Furthermore, we found that downstream Akt signaling through mTORC1 and S6 contributes to the activation of necroptosis and TNFα production. We found that the Akt pathway serves as a critical link between RIP1 kinase and JNK activation in L929 cells. Further data suggested that in multiple other cell types including FADD deficient Jurkat cells, RAW and J774.1 macrophage cell lines, and mouse lung fibroblasts Akt provides a key link to TNFα production, but is dispensible for cell death per se. Overall, our results reveal a specific and novel role for the Akt pathway in regulated necrosis and necrosis-associated inflammatory signaling. Results Basic Fibroblast Growth Factor Promotes Necroptosis in L929 Cells It has been established that mouse fibrosarcoma L929 cells undergo necroptotic cell death following stimulation with TNFα [10], [17]. In addition, inhibition of caspase-8 activity alone, either through siRNA knockdown or by using the pan-caspase inhibitor, zVAD.fmk, is sufficient to trigger necroptosis in these cells [10], [14]. Interestingly, while necroptosis was initially identified as a back-up form of cell death triggered by pro-apoptotic stimuli in the presence of apoptosis inhibitors [17], recent analysis of physiological cell death during mouse development has suggested that the loss of apoptotic regulators, such as caspase-8 and FADD [18], [19], [20], leads to robust induction of necroptosis and death of E10.5 embryos even though apoptosis is not normally induced in wild type embryos. These data are reminiscent of the observations in L929 cells where the loss of caspase activity in healthy cells is sufficient to trigger necroptosis and prompted us to explore the extrinsic or intrinsic cellular factors that promote necroptosis once caspase-8 activity, which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase CYLD [21], [22], is removed in L929 cells. Consistent with a previous report [16], we found that serum starvation of L929 cells prevented necroptosis in response to zVAD.fmk (Fig. 1A). The addition of growth factors, such as bFGF, restored zVAD.fmk induced death under serum free conditions (Fig. 1B). Interestingly, this does not reflect a generic requirement for growth factor signaling, as only some growth factors (bFGF and IGF-1, but not EGF and PDGF) promoted death (Fig. 1B). Furthermore, growth factor-dependent necroptosis required the inhibition of caspase activity, as bFGF alone did not induce cell death (Fig. 1C). In contrast, TNFα triggered necroptosis equally efficiently in the absence of serum (Fig. 1A), suggesting that either growth factors and zVAD.fmk or TNFα are required for necroptotic death in L929 cells. 10.1371/journal.pone.0056576.g001 Figure 1 bFGF and IGF-1 promote necroptosis in concert with zVAD.fmk. (A) L929 cells were treated with TNFα or zVAD.fmk under normal serum (10% FBS) or serum free conditions. Cell viability was determined after 24 hr using the CellTiter-Glo Viability assay. The concentrations of all necroptosis-inducing agents are listed in the Materials and Methods section or indicated in the figures. (B) Cells were treated with zVAD.fmk, the indicated growth factors, and Nec-1 under serum free conditions for 24 hrs followed by measurement of cell viability. (C) Cells under serum free conditions were treated with FGF, zVAD.fmk, or both for 24 hrs followed by viability assay. (D) Cell death was induced by zVAD.fmk or TNFα under full serum condition in the presence of 2 µM PD173074 and 20 µM PD166866. In all graphs, average±SD was plotted. Previously we described the development of 7-Cl-O-Nec-1 (Nec-1) as a potent and selective inhibitor of RIP1 kinase and necroptosis (Fig. S1A) [23], [24]. Recently, its selectivity has been further validated against a panel of more than 400 human kinases [15]. This inhibitor efficiently blocked growth factor/zVAD.fmk-induced necroptosis under serum free conditions in L929 cells and both zVAD.fmk and TNFα-induced necroptosis under full serum conditions (Fig. 1B, S1B). To further validate the role of RIP1, we used an inactive analog, 7-Cl-O-Nec-1i (Nec-1i) (Fig. S1A), which contains an extra N-methyl group that leads to almost complete loss of RIP1 kinase inhibitory activity in vitro [23]. Nec-1i was unable to protect L929 cell death under serum condtions treated with zVAD.fmk or TNFα (Fig. S1B) or serum free conditions treated with bFGF/zVAD.fmk (Fig. S1C). This confirms that RIP1 kinase is responsible for necroptosis in L929 cells under both serum and serum free conditions. We next examined whether bFGF contributes to zVAD.fmk-induced necroptosis under normal serum conditions (10% FBS). We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling strongly inhibited zVAD.fmk-induced necroptosis under normal serum conditions (Fig. 1D). In contrast, neither bFGF receptor inhibitor was able to attenuate TNFα-induced necroptosis (Fig. 1D), consistent with growth factors being dispensable for this pathway (Fig. 1A). Overall, these data suggest that the induction of necroptosis by zVAD.fmk is promoted by bFGF under both serum and serum free conditions. The induction of necroptosis, however, is not a simple consequence of growth factor signaling since not all growth factors allowed death to occur. Instead, specific signaling events mediated by particular growth factors appear to contribute to necroptotic death. RIP1 Kinase-dependent Activation of Akt Contributes to Necroptosis Given our observation that growth factors are important for zVAD.fmk induced death, we examined the contribution of several pathways, including MAPK pathways and Akt, which are known to be activated following growth factor receptor activation (Fig. 2A). Inhibition of Akt (Akt inhibitor VIII) strongly protected the cells from growth factor-sensitive necroptosis induced by zVAD.fmk [16] as well as cell death triggered by bFGF or IGF-1/zVAD.fmk under serum free conditions (Fig. 2B). Inhibition of Akt also protected the cells from growth-factor insensitive death by caused by TNFα (Fig. 2A). Consistent with previous reports, the JNK inhibitor SP600125 protected the cells from both zVAD.fmk and TNFα induced death (Fig. 2A,B and Fig. S2A) [12], [14]. In contrast, inhibition of two other MAPKs, p38 and ERK, previously reported not to be activated during necroptosis [14], did not protect from either zVAD.fmk or TNFα induced death (Fig. 2A). 10.1371/journal.pone.0056576.g002 Figure 2 Akt contributes to necroptosis induced by zVAD.fmk and TNFα. (A,B) Necroptosis was induced by zVAD.fmk or TNFα (full serum, A) or growth factors/zVAD.fmk (serum free, B) in the presence of inhibitors of Akt (Akt inhibitor VIII), JNK (SP600125), p38 (PD169316), and Erk (UO126). Cell viability was determined after 24 hrs. (C) L929 cells transfected with Akt1, Akt2, and Akt3 siRNAs for 72 hrs were treated with zVAD.fmk or TNFα for 9 hrs. Cell viability and Akt expression levels were determined after 24 hrs. In all graphs, average±SD was plotted. Next, we used two approaches to further validate the role of Akt in necroptotic cell death. First, two additional Akt inhibitors, a highly specific, allosteric kinase inhibitor MK-2206 [25] and triciribine (TCN) [26], which blocks membrane translocation of Akt, both attenuated cell death (Fig. S2B). Secondly, simultaneous knockdown of Akt isoforms Akt1 and Akt2 using siRNAs protected cells from necroptosis induced by both zVAD.fmk and TNFα (Fig. 2C). No expression of Akt3 was seen in L929 cells (Fig. S2C) and, consistently, Akt3 siRNA had no additional effect on necroptosis. Our results confirmed that Akt plays a key role in necroptosis induced by multiple stimuli in L929 cells. To understand the activation of Akt and JNK under necroptotic conditions, we examined the changes in Akt and JNK phosphorylation at 9 hrs post zVAD.fmk and TNFα stimulation. This time point was chosen because it reflects the early stage of cell death in our system (Fig. S3A, B). Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation at a known major activation site, Thr308 (Fig. 3A). Interestingly, we did not observe concomitant phosphorylation changes in the second major activation site of Akt, Ser473. We also observed an increase in the phosphorylation of both the p46 and p54 isoforms of JNK and its major substrate c-Jun (Fig. 3A). These data indicate that both Akt and JNK are activated under necroptotic conditions. 10.1371/journal.pone.0056576.g003 Figure 3 RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr, followed by western blotting with indicated antibodies. (B,C) L929 cells were treated with zVAD.fmk (B) or bFGF/zVAD.fmk (serum free conditions, C) and samples were collected at the indicated time points for western blot. (D) Nec-1 was added to the cells stimulated with bFGF or bFGF/zVAD (serum free conditions) for 15 min or 9 hr followed by western blot with the indicated antibodies. The RIP1 kinase inhibitor, Nec-1, completely prevented the increase in Thr308 Akt phosphorylation, while Nec-1i did not (Fig. 3A, Fig. S1D). Similarly, Nec-1 prevented the induction of JNK phosphorylation in response to zVAD.fmk and substantially reduced this change after TNFα addition. We observed some changes in total protein levels of JNK and c-Jun following necroptotic stimulation. Some of these changes, e.g. zVAD.fmk-induced increase in c-Jun, were also attenuated by Nec-1. Importantly, Nec-1 did not alter the basal phosphorylation levels of either Akt or JNK (Fig. 3A). This established that Akt Thr308 and JNK phosphorylation during necroptosis is RIP1 dependent. Interestingly, we discovered that the phosphorylation of Akt Thr308, JNK and Jun are late events following zVAD.fmk stimulation (Fig. 3B) that coincide with the onset of necroptosis at 6 hr post-stimulation (Fig. S3A). To better understand the contributions of growth factors and RIP1 kinase to necroptotic regulation of Akt, we next analyzed the time course of these phosphorylation changes under serum free conditions. We found that the addition of bFGF alone or in combination with zVAD.fmk led to a substantial rapid and transient increase in both Thr308 and Ser473 phosphorylation of Akt as well as JNK and c-Jun at 15 minutes, reflecting the expected response to growth factor stimulation (Fig. 3C). Significantly, the combination of bFGF/zVAD.fmk, but not bFGF alone, also caused a robust, second, delayed increase in the phosphorylation of Thr308, but not Ser473, of Akt as well as a delayed increase in the phosphorylation of JNK and Jun. Furthermore, Nec-1 had no significant effect on the early increase in both Akt and JNK/c-Jun phosphorylation triggered by both bFGF and bFGF/zVAD, while Nec-1, but not its inactive analog Nec-1i (Fig. S1E), efficiently blocked the bFGF/zVAD increase at 6–9 hr (Fig. 3D), suggesting that only the delayed activation of Akt and JNK is specific for necroptosis and dependent on RIP1 kinase activity. Similarly, IGF/zVAD, which also promoted cell death under serum free conditions, produced a delayed increase in Thr308 phosphorylation on Akt, while IGF alone caused solely an early, transient increase in phosphorylation (Fig. S3C). We confirmed the kinetics of the Akt Thr308 and Ser473 phosphorylation changes using a quantitative ELISA assay, which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation (Fig. S3D, E). Taken together, these results indicate that the observed delayed increases in Akt and JNK phosphorylation, preceding the onset of cell death, represent specific consequences of necroptotic signaling downstream from RIP1 kinase. TNFα Induces Delayed Akt Thr308 Phosphorylation and Necroptosis Independent of Growth Factor Stimulation Consistent with TNFα inducing necroptosis independently of growth factors (Fig. 1A), FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation, while efficiently preventing these changes in response to zVAD.fmk (Fig. S4A). Furthermore, addition of TNFα led to comparable late activation of Akt p308 signal under both normal and serum free conditions (Fig. S4B, C), indicating that TNFα signaling to Akt Thr308 is growth factor-independent. In contrast, activation of JNK by TNFα followed different kinetics from zVAD.fmk-induced changes. TNFα treatment caused an early and robust increase in the phosphorylation of JNK and c-Jun. Nec-1 did not affect this early increase, however, it reduced levels of pJNK/Jun at the late, 9 hr time point (Fig. S4B, C). This again separated early RIP1-independent changes, which likely reflect the ability of additional upstream kinases, such as Ask1 to activate JNK [27], from the late RIP1 kinase-dependent necroptotic signaling. Late Increase in Akt Thr308 Phosphorylation Contributes to the Induction of Necroptotic Cell Death We next investigated if the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation functionally contributes to the execution of necroptotic cell death. Firstly, PDGF/zVAD.fmk, which cannot induce necroptosis (Fig. 2A), triggered only the initial, rapid Akt and JNK phosphorylation changes and not the delayed activation (Fig. 4A), indicating that late, rather than early Akt phosphorylation correlates with necroptosis. Secondly, we saw that the ability of the Akt inhibitor to protect cells from necroptosis rapidly declined after 6 hrs of stimulation with zVAD.fmk, TNFα or bFGF/zVAD.fmk and no protection was observed when the inhibitor was added at 9 hrs (Fig. 4B,C). This time frame coincides with the timing of the secondary Akt Thr308 phosphorylation. Finally, we terminated the bFGF signal one hour after addition of bFGF by the addition of PD173074. This allowed us to retain early Akt activation, but to suppress the secondary increase (Fig. 4D). Both pre-addition and delayed addition of PD173074 fully prevented necroptosis (Fig. 4E). Overall, these data, while correlative, indicate that early Akt activation is insufficient to promote necroptosis and are strongly supportive of an important role for the delayed activation of Akt in the induction of necroptotic cell death. 10.1371/journal.pone.0056576.g004 Figure 4 Late Thr308 phosphorylation of Akt contributes to necroptosis. (A) L929 cells were treated with zVAD.fmk and bFGF or PDGF, with or without Nec-1, for the indicated periods of time. (B,C) L929 cells were stimulated by zVAD.fmk or TNFα (B) or bFGF/zVAD.fmk under serum free conditions (C). Akt inh. VIII was added 15 min before necroptotic stimulation (Pre) or at indicated times after stimulation. Viability was measured 24 hr after activation of necroptosis. (D) L929 cells were stimulated with bFGF/zVAD under serum free conditions. PD173074 was added 15 min before or 1 hr after FGF/zVAD. Samples for western blot were collected at 15 min and 9 hr time points. (E) Cells were pretreated with PD173074 or it was added 1 hr after bFGF/zVAD.fmk, followed by viability assessment at 24 hr. In all graphs, average±SD was plotted. The Akt Signaling Pathway Contributes to the Regulation of Necroptosis We next determined whether the necroptosis-associated increase in Thr308 phosphorylation results in an increase in Akt kinase activity. Under necroptotic conditions, we observed an increase in the phosphorylation of multiple known Akt substrates (Forkhead box class O (FoxO) proteins, GSK-3 kinases and mouse double minute 2 (MDM2)) as well as downstream molecules (p70 ribosomal protein S6 Kinase (p70S6K), S6) (Fig. 5A). In some cases (FoxO1, FoxO4, MDM2), a robust increase was observed. In other cases (FoxO3a, GSK-3α/β, p70S6K and its substrate S6), the changes were less pronounced (Fig. 5A). The timing of the phosphorylation changes paralleled the increase in Akt phosphorylation (Fig. 5B, S5A, B). In the case of pFoxO1 we occasionally observed a shift in migration rather than an increase in band intensity (e.g. comparing Fig. 5A and B), suggesting that phosphorylation events in addition to Thr24 take place during necroptosis. Notably, in all cases the necroptosis-associated increases in Akt substrates were abrogated by Nec-1 (Fig. 5A, Fig. S5A, B). Overall, these data suggested that a significant part of the “canonical” Akt signaling network is activated at the onset of necroptotic cell death in a RIP1 dependent fashion. 10.1371/journal.pone.0056576.g005 Figure 5 mTORC1 contributes to the regulation of necroptosis. (A) L929 cells were treated with zVAD.fmk or TNFα for 9 hr and harvested for western blot. (B) Cell under serum free condition were treated with bFGF or bFGF/zVAD.fmk for the indicated amounts of time, followed by western blotting using the indicated antibodies. (C) Necroptosis was induced by zVAD.fmk or TNFα in L929 cell in the presence of inhibitors of Akt(Akt inh. VIII) and mTOR (rapamycin, Torin-1 and PI-103). (D) L929 cells with mTOR siRNA knockdown were harvested for western blot or treated with zVAD.fmk or TNFα for 24 hrs. Cell viability was determined 24 hr after activation of necroptosis. In all graphs, average±SD was plotted. Akt kinase is considered to be a pro-survival protein that inhibits apoptosis through the control of multiple effectors including mTORC1, GSK-3 and others [28]. An important question is whether these same molecules reverse their pro-survival roles during necroptosis. We found that inhibition of mTORC1 by rapamycin, an inhibitor of the mTOR co-factor Raptor [29], protected cells from necroptosis (Fig. 5C). Similarly, the direct mTOR kinase inhibitor Torin1 [30] and the dual PI3K/mTOR inhibitor PI-103 [31] also efficiently inhibited necroptosis (Fig. 5C). Knockdown of mTOR using siRNA further validated the small-molecule inhibitor data indicating a role for mTOR in necroptosis by protecting cells from both zVAD.fmk and TNFα induced death (Fig. 5D). mTORC1 regulates translation through activation of p70S6 kinase and, subsequently, ribosomal protein S6 [32]. Notably, a genome-wide siRNA screen [10] suggested an important role for protein translation in necroptosis. Consistently, we found that the small molecule inhibitor of p70S6K PF-4708671 [33] attenuated necroptosis at the concentrations required to block S6 phosphorylation (Fig. S5C, D). Partial siRNA knockdown of S6 protein attenuated necroptosis as well (Fig. S5E), suggesting that translational control by p70S6K/S6 may play a role in necroptosis. Overall, while the full repertoire of Akt targets during necroptosis remains to be fully explored, our data provide evidence that the activity of an anti-apoptotic branch of Akt signaling can promote necroptosis. RIP1 kinase, Akt, mTORC1 and JNK control the upregulation of TNFα accompanying necroptosis. Hitomi et al. [10] have recently reported that the induction of necroptosis by zVAD.fmk in L929 cells is associated with increased synthesis of TNFα, which potentiates cell death. Therefore, we examined whether Akt and its effectors contribute to TNFα synthesis. Consistent with a RIP1-dependent increase in TNFα protein (Fig. S6A, B), we found that TNFα mRNA levels increased during necroptosis in L929 cells in a RIP1 (Fig. S6C. Under serum free conditions, bFGF alone triggered some induction of TNFα mRNA, while its combination with zVAD.fmk (but not zVAD.fmk alone) caused a pronounced further increase (Fig. 6A). Conversely, PDGF caused a modest upregulation of TNFα mRNA, which was not further increased in the presence of zVAD.fmk (Fig. 6A), demonstrating that activation of necroptosis is specifically accompanied by a marked increase in autocrine TNFα synthesis. 10.1371/journal.pone.0056576.g006 Figure 6 Akt and mTORC1 control autocrine TNFα synthesis and JNK activation during necroptosis. (A) Cells were treated under serum free conditions with bFGF or PDGF with or without zVAD.fmk for 9 hr, followed by qRT-PCR analysis of mTNFα. Data was normalized to mouse 18S RNA. (B) Necroptosis was induced by zVAD.fmk or TNFα in cells treated with Nec-1, rapamycin (rapa), or Akt inh. VIII inh. followed by qRT-PCR analysis of TNFα mRNA levels. (C-F) L929 cells with siRNA knockdown of Akt isoforms (C,E) or mTOR (D,F) were stimulated with zVAD.fmk or TNFα for 9 hr, followed by qRT-PCR analysis of mTNFα (C,D) or western blot (E,F). In all graphs, average±SD was plotted. Further analysis suggested that both Akt and mTORC1 contribute to the upregulation of TNFα mRNA during necroptosis as both small-molecule inhibition and siRNA knockdown of Akt and mTOR reduced TNFα mRNA levels in necroptotic cells (Fig. 6B,C,D). Notably, RIP1 and Akt inhibitors had no effect on the levels of TNFα mRNA in control cells or in the cells stimulated with bFGF alone (Fig. 6A,B, Fig. S6C), suggesting that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis. Akt and mTORC1 Control the Activation of JNK during Necroptosis JNK is a well-established regulator of TNFα synthesis in a variety of systems [13], [14], [15], [34]. Therefore, the ability of Akt and mTORC1 inhibitors to block the increase in TNFα mRNA lead us to examine their role in the activation of JNK during necroptosis. Knockdown of Akt isoforms Akt1 and Akt2 or inhibition of Akt prominently suppressed the necroptosis dependent increase in JNK and c-Jun phosphorylation (Fig. 6E, S6D,E) suggesting that Akt may provide a link between RIP1 and JNK activation. Importantly, inhibition of Akt only inhibited the delayed, but not the early, increase in bFGF/zVAD.fmk induced JNK and c-Jun phosphorylation (Fig. S6F). Knockdown of mTOR, rapamycin and the p70S6K inhibitor PF-4708671 also attenuated the necroptosis-associated increase in JNK and c-Jun phosphorylation (Fig. 6F, S6E,G, Fig. S5D). Overall, these data suggested that the Akt-mTORC1-S6K axis, acting downstream from RIP1 kinase, is required for the increase in JNK activity during necroptosis in L929 cells. PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under Necroptotic Conditions Typical regulation of Akt by growth factors involves its recruitment to the plasma membrane, which is mediated by the binding of the pleckstrin homology (PH) domain of Akt to the product of PI3K, phosphatidylinositol-3,4,5-triphosphate (PIP3). In the membrane, Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1) and mTORC2 (or DNA-PK), respectively [35]. Since our results showed that only Thr308 Akt phosphorylation is increased during necroptosis, we next examined whether it is still dependent on PI3K and PDK1. Inhibition of PI3K and PDK1 using the specific inhibitors LY249002 and BX912 [36] resulted in the efficient inhibition of cell death and Akt Thr308 phosphorylation (Fig. S7A–D). Likewise, siRNA knockdown of PDK1 protected cells from death and inhibited Akt Thr308 phosphorylation (Fig. S7E,F) Therefore, PI3K and PDK1 activity is still required for non-canonical Akt activation during necroptosis. Expression of Constitutively Active Akt, Rescues Necroptosis Under Serum Free Conditions We used L929 cells stably expressing constitutively active wild type Akt1 (Myr-Akt) or the catalytically inactive mutant K179M in order to further understand the contribution of growth factors and RIP1 kinase to Akt activation during necroptosis. Constitutively active Akt1 (Myr-Akt) was generated as previously described [37] by the addition of a myristoylation signal which provides constitutive localization to the plasma membrane and by the deletion of the auto-inhibitory PH domain (Fig. 7A) resulting in an Akt that is active under serum free. It is important to note that the cells expressing Myr-Akt were viable, grew in a manner indistinguishable from the empty vector control cells, and were not triggered to induce necroptosis by serum starvation (Fig. 7B). This indicates that active Akt alone is not sufficient to induce necroptotic cell death. Under serum free conditions Myr-Akt, but not the K179M mutant, fully restored zVAD.fmk-induced necroptosis (Fig. 7A,B). Nec-1 prevented both Myr-Akt dependent cell death and the necroptosis-specific delayed increase in Akt Thr308 phosphorylation (Fig. 7B,C). Myr-Akt also allowed other zVAD.fmk-dependent events, including activation of JNK and c-Jun phosphorylation (Fig. 7C) and upregulation of TNFα mRNA (Fig. 7D) to occur under serum free conditions, confirming an important role for Akt at the apex of necroptotic signaling. These data demonstrated that the presence of active and membrane localized Akt is sufficient to uncouple Akt activation during necroptosis from growth factor signaling. RIP1 kinase was still able to regulate Akt activation during necroptosis, suggesting that growth factors and RIP1 kinase provide two independent inputs required for Akt changes during necroptosis. 10.1371/journal.pone.0056576.g007 Figure 7 Over expression of constitutively active Akt restores necroptosis under serum free conditions. (A,B) L929 cells were stably infected with empty MSCV retrovirus or viruses encoding Myr-Akt or the catalytically inactive Myr-Akt K179M. Necroptosis was induced by the addition of zVAD.fmk under serum free conditions (A) or serum or serum free conditions with Nec-1 (B). Viability assays were performed after 24 hr. (C) Myr-Akt and Myr-Akt K179M cells were treated with zVAD.fmk and/or Nec-1 under serum free conditions for 9 hr, followed by western blot using the indicated antibodies. Endogenous Akt (∼) and Myr-Akt (*) bands are indicated. (D) L929 cells, stably infected with Myr-Akt and Myr-Akt K179KM, were stimulated with zVAD.fmk for 9 hr under serum free conditions. TNFα mRNA levels were determined by qRT-PCR and normalized using mouse 18S RNA. (E-G) L929 cells expressing Myr-Akt and Ala and Asp mutants of Thr308 and Ser473 were treated with zVAD.fmk under serum free conditions, followed by viability assay at 24 hr (E), western blot at 9 hr (F), or evaluation of TNFα mRNA levels by qRT-PCR at 9 hrs (G). In all graphs, average±SD was plotted. RIP1 kinase-dependent Thr308 phosphorylation of Myr-Akt during necroptosis increased Myr-Akt activity as it did"
R10243 T15758 T15746 themeOf Akt activation,to regulate Akt activation during necroptosis
R12061 T18035 T18095 causeOf RIP1,of RIP1 kinase-dependent signaling
R12062 T18038 T18103 causeOf kinase-dependent,of RIP1 kinase-dependent signaling
R12063 T18039 T18096 themeOf protein,Fas-associated protein with death domain (FADD)
R12064 T18043 T18107 themeOf endogenous Akt1,fibroblasts expressing endogenous Akt1 or Akt2
R12065 T18051 T18099 locationOf Cell,Other Cell Types
R12066 T18055 T18039 partOf with death domain (FADD),protein
R12067 T18055 T18096 siteOf with death domain (FADD),Fas-associated protein with death domain (FADD)
R12068 T18056 T18099 themeOf Other Cell Types,Other Cell Types
R12069 T18057 T18104 causeOf RIP1,a RIP1 kinase dependent increase in the phosphorylation of Thr308 on Akt
R12070 T18060 T18108 themeOf Akt2,fibroblasts expressing endogenous Akt1 or Akt2
R12071 T18064 T18098 themeOf TNFα,TNFα Production in Other Cell Types
R12072 T18066 T18100 causeOf Akt,Akt Controls TNFα Production in Other Cell Types
R12073 T18068 T18097 causeOf Akt-mediated,Akt-mediated inflammatory signaling under necroptotic conditions
R12074 T18069 T18106 causeOf kinase dependent,a RIP1 kinase dependent increase in the phosphorylation of Thr308 on Akt
R12075 T18081 T18102 themeOf robust RIP1-dependent TNFα mRNA,robust RIP1-dependent TNFα mRNA upregulation
R12076 T18082 T18096 themeOf Fas,Fas-associated protein with death domain (FADD)
R12077 T18090 T18105 themeOf of Akt,inhibition of Akt
R12078 T18091 T18101 themeOf TNFα mRNA,TNFα mRNA production
R12079 T18095 T18103 themeOf of RIP1 kinase-dependent signaling,of RIP1 kinase-dependent signaling
R12080 T18098 T18100 themeOf TNFα Production in Other Cell Types,Akt Controls TNFα Production in Other Cell Types
R12081 T18104 T18106 themeOf a RIP1 kinase dependent increase in the phosphorylation of Thr308 on Akt,a RIP1 kinase dependent increase in the phosphorylation of Thr308 on Akt
R13122 T19774 T19898 themeOf of Akt,necroptotic activation of Akt
R13123 T19776 T19890 themeOf Akt,selective necroptotic phosphorylation of Thr308 of Akt
R13124 T19781 T19886 themeOf caspase,caspase inhibition
R13125 T19784 T19897 themeOf of Akt,Thr308 phosphorylation of Akt
R13126 T19786 T19888 themeOf of Akt,necroptosis-associated phosphorylation of Akt
R13127 T19787 T19892 themeOf TNFα,TNFα production
R13128 T19790 T19900 themeOf the p110δ subunit of PI3K [41],inhibitor that may inhibit the p110δ subunit of PI3K [41] and PDK1 [42]
R13129 T19791 T19899 causeOf on c-Jun,"autocrine TNFα production, dependent on c-Jun,"
R13130 T19806 T19895 themeOf of Akt,the delayed activation of Akt
R13131 T19810 T19884 themeOf JNK,JNK activation
R13132 T19821 T19896 themeOf of Akt,phosphorylation of Akt
R13133 T19824 T19887 themeOf JNK,JNK inhibition
R13134 T19831 T19776 partOf of Thr308 of Akt,Akt
R13135 T19831 T19890 siteOf of Thr308 of Akt,selective necroptotic phosphorylation of Thr308 of Akt
R13136 T19835 T19784 partOf Thr308,of Akt
R13137 T19835 T19897 siteOf Thr308,Thr308 phosphorylation of Akt
R13138 T19837 T19894 themeOf of c-Jun,the phosphorylation of c-Jun at Ser63
R13139 T19844 T19883 themeOf Akt,"basal Akt phosphorylation levels,"
R13140 T19847 T19837 partOf Ser63,of c-Jun
R13141 T19847 T19894 siteOf Ser63,the phosphorylation of c-Jun at Ser63
R13142 T19850 T19880 themeOf TNFα,TNFα production
R13143 T19854 T19901 themeOf PDK1 [42],inhibitor that may inhibit the p110δ subunit of PI3K [41] and PDK1 [42]
R13144 T19860 T19889 themeOf Akt,we examined Akt phosphorylation after inhibition of a downstream
R13145 T19861 T19881 themeOf of Myr-Akt,expression of Myr-Akt
R13146 T19864 T19891 themeOf TNFα,"autocrine TNFα production, dependent on c-Jun,"
R13147 T19872 T19885 themeOf of c-Jun,phosphorylation of c-Jun
R13148 T19878 T19900 causeOf inhibitor that may inhibit the p110δ subunit of PI3K [41] and PDK1 [42],inhibitor that may inhibit the p110δ subunit of PI3K [41] and PDK1 [42]
R13149 T19878 T19901 causeOf inhibitor that may inhibit the p110δ subunit of PI3K [41] and PDK1 [42],inhibitor that may inhibit the p110δ subunit of PI3K [41] and PDK1 [42]
R13150 T19891 T19899 themeOf "autocrine TNFα production, dependent on c-Jun,","autocrine TNFα production, dependent on c-Jun,"
R13151 T19892 T19893 themeOf TNFα production,which protected L929 cells from death and inhibited TNFα production
R13152 T19895 T19882 themeOf the delayed activation of Akt,the delayed activation of Akt
R15643 T23476 T23639 themeOf Akt,Necroptotic phosphorylation of Thr308 of Akt
R15644 T23477 T23654 themeOf of TNFα,This increased production of TNFα during necroptosis
R15645 T23486 T23476 partOf of Thr308 of Akt,Akt
R15646 T23486 T23639 siteOf of Thr308 of Akt,Necroptotic phosphorylation of Thr308 of Akt
R15647 T23494 T23637 themeOf JNK,JNK activation
R15648 T23498 T23640 themeOf of Akt,necrosis-specific regulation of Akt
R15649 T23506 T23645 themeOf Akt,in Akt Thr308 phosphorylation
R15650 T23511 T23630 themeOf Akt,Akt activation
R15651 T23518 T23644 themeOf of JNK,the activation of JNK
R15652 T23519 T23647 themeOf to JNK,connecting mTORC1 to JNK
R15653 T23522 T23643 themeOf of Myr-Akt,expression of Myr-Akt
R15654 T23523 T23653 locationOf epithelial cells,caspase-8 in epithelial cells
R15655 T23527 T23649 locationOf "cell fates alternative to apoptosis, ranging from survival to non-apoptotic cell death","cell fates alternative to apoptosis, ranging from survival to non-apoptotic cell death"
R15656 T23530 T23629 themeOf of Akt,to assume that activation of Akt
R15657 T23533 T23647 themeOf mTORC1,connecting mTORC1 to JNK
R15658 T23535 T23642 themeOf of Akt,"Thr308 phosphorylation of Akt,"
R15659 T23541 T23625 themeOf of Akt,phosphorylation of Akt
R15660 T23542 T23652 themeOf of JNK,of mTORC1-dependent regulation of JNK
R15661 T23543 T23641 causeOf by recombinant RIP1 kinase,"Interestingly, we observed phosphorylation of Akt by recombinant RIP1 kinase in vitro on Thr146, 195/197, and 435"
R15662 T23544 T23506 partOf Akt Thr308,Akt
R15663 T23544 T23645 siteOf Akt Thr308,in Akt Thr308 phosphorylation
R15664 T23550 T23651 themeOf of mTOR [48],activation of mTOR [48]
R15665 T23551 T23535 partOf Thr308,of Akt
R15666 T23551 T23642 siteOf Thr308,"Thr308 phosphorylation of Akt,"
R15667 T23555 T23628 causeOf kinase,that RIP1 kinase inhibits a phosphatase that targets Thr308
R15668 T23562 T23653 themeOf caspase-8 in epithelial cells,caspase-8 in epithelial cells
R15669 T23573 T23633 causeOf RIP1-dependent,the RIP1-dependent increase in Akt Thr308 phosphorylation
R15670 T23576 T23641 themeOf of Akt,"Interestingly, we observed phosphorylation of Akt by recombinant RIP1 kinase in vitro on Thr146, 195/197, and 435"
R15671 T23577 T23628 themeOf that RIP1 kinase inhibits a phosphatase that targets Thr308,that RIP1 kinase inhibits a phosphatase that targets Thr308
R15672 T23587 T23650 themeOf the only enzyme established to specifically dephosphorylate this residue [45],the only enzyme established to specifically dephosphorylate this residue [45]
R15673 T23591 T23646 causeOf RIP1 kinase,RIP1 kinase signaling
R15674 T23600 T23631 themeOf of TNFα,the production of TNFα
R15675 T23603 T23636 causeOf Akt,"Akt signaling contributed more prominently to an increase in TNFα synthesis, rather than cell death"
R15676 T23608 T23648 themeOf "cell fates alternative to apoptosis, ranging from survival to non-apoptotic cell death","promote cell fates alternative to apoptosis, ranging from survival to non-apoptotic cell death"
R15677 T23608 T23649 themeOf "cell fates alternative to apoptosis, ranging from survival to non-apoptotic cell death","cell fates alternative to apoptosis, ranging from survival to non-apoptotic cell death"
R15678 T23609 T23635 themeOf of TNFα,the translation of TNFα
R15679 T23626 T23633 themeOf the RIP1-dependent increase in Akt Thr308 phosphorylation,the RIP1-dependent increase in Akt Thr308 phosphorylation
R15680 T23631 T23638 themeOf the production of TNFα,mediating the production of TNFα
R15681 T23632 T23634 themeOf a selective increase in Akt phosphorylation on Thr308,promoting a selective increase in Akt phosphorylation on Thr308
R15682 T23645 T23626 themeOf in Akt Thr308 phosphorylation,the RIP1-dependent increase in Akt Thr308 phosphorylation
R15683 T23654 T23627 themeOf This increased production of TNFα during necroptosis,This increased production of TNFα during necroptosis
R17464 T25562 T25569 themeOf Pan-caspase inhibitor zVAD.fmk (20–30 µM),Pan-caspase inhibitor zVAD.fmk (20–30 µM) was purchased from Bachem
R19285 T27871 T27877 themeOf of Myr-Akt1,Stable Infection of Myr-Akt1
R1953 T2733 T2795 themeOf "to autocrine TNFα synthesis, activation of oxidative stress and induction of autophagy,","Activation of JNK in L929 cells has been linked to autocrine TNFα synthesis, activation of oxidative stress and induction of autophagy,"
R1955 T2735 T2803 themeOf Akt,that Akt is activated through RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types
R1959 T2746 T2800 themeOf TNFα,TNFα production
R1962 T2750 T2796 themeOf of JNK,Activation of JNK in L929 cells
R1964 T2752 T2808 themeOf JNK,JNK activation in L929 cells
R1969 T2763 T2806 causeOf insulin-dependent,insulin-dependent Akt activity
R1971 T2766 T2797 themeOf TNFα,TNFα production
R1973 T2769 T2802 themeOf TNFα,TNFα production
R1976 T2773 T2804 causeOf kinase-dependent,RIP1 kinase-dependent Thr308 phosphorylation during necroptosis in multiple cell types
R1978 T2777 T2801 themeOf Akt,Akt activation
R1982 T2807 T2798 themeOf insulin-dependent activation of Akt,insulin-dependent activation of Akt
R1983 T2785 T2799 themeOf JNK kinase,JNK kinase activation
R1984 T2787 T2794 causeOf Akt,we found that downstream Akt signaling
R1985 T2788 T2807 themeOf of Akt,insulin-dependent activation of Akt
R1986 T2790 T2798 causeOf insulin-dependent,insulin-dependent activation of Akt
R20359 T29355 T29370 themeOf Akt,Akt expression levels
R20582 T29705 T29719 themeOf JNK,RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
R20583 T29706 T29720 themeOf Akt,RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
R20584 T29707 T29719 causeOf RIP1,RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
R20585 T29707 T29720 causeOf RIP1,RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
R20586 T29709 T29717 causeOf kinase-dependent,RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
R20587 T29709 T29718 causeOf kinase-dependent,RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
R20588 T29719 T29717 themeOf RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis,RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
R20589 T29720 T29718 themeOf RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis,RIP1 kinase-dependent phosphorylation of Akt and JNK during necroptosis
R20932 T30199 T30203 themeOf of Akt,phosphorylation of Akt
R21540 T31121 T31152 themeOf JNK,JNK activation during necroptosis
R22250 T32255 T32266 themeOf Akt2,fibroblasts expressing only endogenous Akt1 or Akt2
R22251 T32258 T32267 themeOf only endogenous Akt1,fibroblasts expressing only endogenous Akt1 or Akt2
R22252 T32263 T32265 themeOf TNFα,autocrine TNFα production in multiple cell types
R22423 T32552 T32568 causeOf JNK,JNK dependent signaling
R22424 T32559 T32560 partOf Thr308,Akt
R22425 T32559 T32571 siteOf Thr308,Akt phosphorylation at Thr308 during necroptosis
R22426 T32560 T32571 themeOf Akt,Akt phosphorylation at Thr308 during necroptosis
R22427 T32564 T32569 themeOf of Akt,Activation of Akt during necroptosis
R22428 T32566 T32572 themeOf "of several known Akt substrates, such as mTOR","the phosphorylation of several known Akt substrates, such as mTOR"
R22429 T32571 T32570 themeOf Akt phosphorylation at Thr308 during necroptosis,Akt phosphorylation at Thr308 during necroptosis requires inputs from both growth factors and RIP1 kinase
R3458 T4918 T4984 themeOf Basic Fibroblast Growth Factor,Basic Fibroblast Growth Factor
R3461 T4922 T4987 causeOf bFGF,of bFGF signaling
R3463 T4926 T4988 causeOf growth factor,of growth factor signaling
R3466 T4931 T4991 themeOf RIP1 kinase,which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase
R3476 T4952 T4983 causeOf growth factor,growth factor signaling
R3486 T4967 T4990 themeOf the RIP1 deubiquitinase,which cleaves and inactivates RIP1 kinase and the RIP1 deubiquitinase
R3488 T4968 T4986 themeOf "CYLD [21], [22],","CYLD [21], [22], is removed in L929 cells"
R3492 T4976 T4984 locationOf Fibroblast,Basic Fibroblast Growth Factor
R3495 T4987 T4989 themeOf of bFGF signaling,"We used two bFGF receptor tyrosine kinase inhibitors (PD173074 and PD166866), and determined that inhibition of bFGF signaling"
R429 T729 T757 themeOf Akt,"During necroptosis, Akt is activated in a RIP1 dependent fashion through its phosphorylation on Thr308"
R430 T729 T758 themeOf Akt,"During necroptosis, Akt is activated in a RIP1 dependent fashion through its phosphorylation on Thr308"
R431 T734 T764 themeOf TNFα,TNFα production
R432 T737 T729 partOf on Thr308,Akt
R433 T737 T758 siteOf on Thr308,"During necroptosis, Akt is activated in a RIP1 dependent fashion through its phosphorylation on Thr308"
R434 T740 T762 themeOf JNK,JNK activation
R435 T741 T765 themeOf RIP1 Kinase,RIP1 Kinase Activation during Necroptosis
R436 T747 T759 themeOf TNFα,necroptosis-associated TNFα production
R437 T751 T763 themeOf mTORC1,"mTORC1, links RIP1 to JNK activation and autocrine production of TNFα"
R438 T752 T763 themeOf RIP1,"mTORC1, links RIP1 to JNK activation and autocrine production of TNFα"
R439 T756 T761 themeOf of TNFα,autocrine production of TNFα
R4700 T7147 T7189 partOf "of Akt Thr308,",Akt
R4701 T7147 T7308 siteOf "of Akt Thr308,","the phosphorylation of Akt Thr308, JNK and Jun are late"
R4704 T7156 T7303 themeOf Thr308 Akt,in Thr308 Akt phosphorylation
R4706 T7162 T7306 themeOf JNK,the activation of Akt and JNK
R4707 T7164 T7295 causeOf RIP1-dependent,a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation
R4709 T7165 T7287 themeOf JNK,JNK phosphorylation
R4714 T7174 T7285 themeOf JNK,the delayed activation of Akt and JNK
R4719 T7175 T7156 partOf Thr308,Thr308 Akt
R4720 T7175 T7303 siteOf Thr308,in Thr308 Akt phosphorylation
R4721 T7176 T7279 themeOf of Akt3,No expression of Akt3 was seen in L929 cells (Fig. S2C)
R4727 T7189 T7308 themeOf Akt,"the phosphorylation of Akt Thr308, JNK and Jun are late"
R4728 T7190 T7305 themeOf Akt,the activation of Akt and JNK
R4732 T7204 T7294 themeOf of Akt,Activation of Akt
R4733 T7205 T7288 themeOf Akt,in Akt Thr308 phosphorylation
R4738 T7216 T7292 themeOf JNK,of JNK phosphorylation
R4740 T7218 T7297 themeOf JNK,These data indicate that both Akt and JNK are activated under necroptotic conditions
R4741 T7219 T7296 themeOf JNK,JNK phosphorylation
R4743 T7224 T7282 themeOf of two other MAPKs,inhibition of two other MAPKs
R4746 T7227 T7284 themeOf Akt,the delayed activation of Akt and JNK
R4749 T7236 T7205 partOf Akt Thr308,Akt
R4750 T7236 T7288 siteOf Akt Thr308,in Akt Thr308 phosphorylation
R4753 T7240 T7286 themeOf Akt,in Akt phosphorylation
R4754 T7241 T7298 themeOf Akt,These data indicate that both Akt and JNK are activated under necroptotic conditions
R4755 T7243 T7309 themeOf JNK,"JNK phosphorylation,"
R4759 T7253 T7307 causeOf Nec-1,"Nec-1, completely prevented the increase in Thr308 Akt phosphorylation,"
R4760 T7254 T7304 themeOf of Akt,necroptotic regulation of Akt
R4763 T7265 T7302 themeOf receptor,activated following growth factor receptor activation
R4766 T7268 T7293 themeOf of Akt,Inhibition of Akt (Akt inhibitor VIII)
R4768 T7271 T7291 themeOf of Akt,Inhibition of Akt
R4770 T7280 T7290 themeOf "a delayed increase in Thr308 phosphorylation on Akt,","a delayed increase in Thr308 phosphorylation on Akt,"
R4771 T7276 T7281 themeOf TNFα,TNFα stimulation
R4772 T7284 T7277 themeOf the delayed activation of Akt and JNK,the delayed activation of Akt and JNK
R4773 T7285 T7278 themeOf the delayed activation of Akt and JNK,the delayed activation of Akt and JNK
R4774 T7286 T7299 themeOf in Akt phosphorylation,Following stimulation with either zVAD.fmk or TNFα we observed a robust increase in Akt phosphorylation
R4775 T7288 T7300 themeOf in Akt Thr308 phosphorylation,which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation
R4776 T7300 T7283 themeOf which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation,a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation
R4777 T7300 T7295 themeOf which also showed a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation,a robust delayed necroptosis-specific RIP1-dependent increase in Akt Thr308 phosphorylation
R4778 T7301 T7307 themeOf "the increase in Thr308 Akt phosphorylation,","Nec-1, completely prevented the increase in Thr308 Akt phosphorylation,"
R4779 T7303 T7301 themeOf in Thr308 Akt phosphorylation,"the increase in Thr308 Akt phosphorylation,"
R5232 T8032 T8042 partOf Thr308,Delayed Akt Thr308 Phosphorylation
R5233 T8032 T8075 siteOf Thr308,Delayed Akt Thr308 Phosphorylation
R5234 T8033 T8073 causeOf FGFR inhibitors,FGFR inhibitors did not attenuate TNFα-induced changes in Akt or JNK phosphorylation
R5235 T8036 T8065 causeOf TNFα,TNFα signaling
R5236 T8038 T8066 themeOf of JNK,in the phosphorylation of JNK
R5237 T8042 T8075 themeOf Delayed Akt Thr308 Phosphorylation,Delayed Akt Thr308 Phosphorylation
R5238 T8044 T8072 causeOf kinase-dependent,RIP1 kinase-dependent necroptotic signaling
R5239 T8047 T8069 themeOf of JNK,activation of JNK by TNFα
R5240 T8049 T8067 themeOf of Akt,activation of Akt
R5241 T8051 T8074 causeOf RIP1,RIP1 kinase-dependent necroptotic signaling
R5242 T8052 T8070 themeOf JNK,to activate JNK
R5243 T8053 T8068 themeOf JNK,JNK phosphorylation
R5244 T8057 T8077 themeOf TNFα,TNFα treatment caused
R5245 T8059 T8069 causeOf by TNFα,activation of JNK by TNFα
R5246 T8061 T8071 themeOf Growth Factor,of Growth Factor Stimulation
R5247 T8062 T8076 causeOf growth factor-independent,TNFα signaling to Akt Thr308 is growth factor-independent
R5248 T8065 T8076 themeOf TNFα signaling,TNFα signaling to Akt Thr308 is growth factor-independent
R5249 T8066 T8063 themeOf in the phosphorylation of JNK,robust increase in the phosphorylation of JNK
R5250 T8074 T8072 themeOf RIP1 kinase-dependent necroptotic signaling,RIP1 kinase-dependent necroptotic signaling
R5251 T8075 T8064 themeOf Delayed Akt Thr308 Phosphorylation,Delayed Akt Thr308 Phosphorylation
R5781 T8793 T8827 themeOf Akt,in Akt Thr308 Phosphorylation
R5782 T8794 T8830 causeOf RIP1 kinase-dependent,the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation
R5783 T8796 T8823 themeOf JNK,JNK phosphorylation changes
R5784 T8797 T8802 partOf Akt Thr308,Akt
R5785 T8797 T8831 siteOf Akt Thr308,in Akt Thr308 phosphorylation
R5786 T8798 T8793 partOf Akt Thr308,Akt
R5787 T8798 T8827 siteOf Akt Thr308,in Akt Thr308 Phosphorylation
R5788 T8802 T8831 themeOf Akt,in Akt Thr308 phosphorylation
R5789 T8805 T8832 themeOf Akt,Akt activation
R5790 T8809 T8829 themeOf of Akt,the delayed activation of Akt in the induction of necroptotic cell death
R5791 T8810 T8814 partOf the secondary Akt Thr308,Akt
R5792 T8810 T8828 siteOf the secondary Akt Thr308,the secondary Akt Thr308 phosphorylation
R5793 T8814 T8828 themeOf Akt,the secondary Akt Thr308 phosphorylation
R5794 T8816 T8826 themeOf Akt,Akt activation
R5795 T8825 T8822 themeOf the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation,the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation
R5796 T8825 T8830 themeOf the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation,the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation
R5797 T8829 T8833 themeOf the delayed activation of Akt in the induction of necroptotic cell death,the delayed activation of Akt in the induction of necroptotic cell death
R5798 T8831 T8825 themeOf in Akt Thr308 phosphorylation,the delayed RIP1 kinase-dependent increase in Akt Thr308 phosphorylation
R6962 T10804 T10927 causeOf Akt,of Akt signaling
R6963 T10818 T10925 themeOf "of TNFα mRNA,","a modest upregulation of TNFα mRNA,"
R6964 T10823 T10931 themeOf S6,S6 phosphorylation
R6965 T10826 T10933 themeOf of TNFα accompanying necroptosis,the upregulation of TNFα accompanying necroptosis
R6966 T10842 T10934 themeOf of TNFα mRNA,the upregulation of TNFα mRNA during necroptosis
R6967 T10850 T10930 causeOf RIP1-dependent,with a RIP1-dependent increase in TNFα protein
R6968 T10859 T10922 themeOf of TNFα mRNA,some induction of TNFα mRNA
R6969 T10859 T10923 themeOf of TNFα mRNA,some induction of TNFα mRNA
R6970 T10875 T10924 causeOf TORC1,TORC1 regulates translation through activation of p70S6 kinase
R6971 T10885 T10938 causeOf these kinases,that these kinases specifically mediate necroptosis-dependent increase in TNFα synthesis
R6972 T10899 T10926 themeOf Akt,"Akt phosphorylation (Fig. 5B,"
R6973 T10903 T10937 themeOf protein,protein translation in necroptosis
R6974 T10905 T10929 themeOf of p70S6 kinase,activation of p70S6 kinase
R6975 T10915 T10939 themeOf Akt kinase,in an increase in Akt kinase activity
R6976 T10931 T10935 themeOf S6 phosphorylation,block S6 phosphorylation
R8381 T12625 T12667 themeOf of Akt,inhibition of Akt
R8382 T12627 T12662 themeOf c-Jun,c-Jun phosphorylation
R8383 T12628 T12663 themeOf c-Jun,c-Jun phosphorylation
R8384 T12632 T12664 themeOf c-Jun,c-Jun phosphorylation
R8385 T12634 T12665 themeOf JNK,the increase in JNK activity during necroptosis in L929 cells
R8386 T12640 T12670 themeOf of Akt,inhibition of Akt
R8387 T12641 T12661 themeOf of JNK,the Activation of JNK during Necroptosis
R8388 T12642 T12669 themeOf JNK,JNK activation
R8389 T12648 T12668 themeOf of JNK,the activation of JNK during necroptosis
R8868 T13360 T13401 themeOf Akt,non-canonical Akt activation during necroptosis
R8869 T13361 T13373 partOf Thr308,Akt
R8870 T13361 T13398 siteOf Thr308,Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1)
R8871 T13370 T13399 themeOf Thr308 Akt,Thr308 Akt phosphorylation
R8872 T13372 T13396 causeOf PI3-kinase,PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under
R8873 T13373 T13398 themeOf Akt,Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1)
R8874 T13374 T13395 themeOf Akt,inhibited Akt Thr308 phosphorylation
R8875 T13376 T13400 themeOf Akt,Akt Thr308 phosphorylation
R8876 T13379 T13398 causeOf by 3-phosphoinositide dependent protein kinase-1 (PDK1),Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1)
R8877 T13382 T13403 themeOf PI3K,Inhibition of PI3K and PDK1
R8878 T13385 T13373 partOf Ser473,Akt
R8879 T13385 T13398 siteOf Ser473,Akt is phosphorylated on Thr308 and Ser473 by 3-phosphoinositide dependent protein kinase-1 (PDK1)
R8880 T13388 T13376 partOf Akt Thr308,Akt
R8881 T13388 T13400 siteOf Akt Thr308,Akt Thr308 phosphorylation
R8882 T13390 T13374 partOf Akt Thr308,Akt
R8883 T13390 T13395 siteOf Akt Thr308,inhibited Akt Thr308 phosphorylation
R8884 T13391 T13397 causeOf PDK1,PI3-kinase and PDK1 Mediate the Increase in Akt Thr308 Phosphorylation Under
R8885 T13392 T13402 themeOf PDK1,Inhibition of PI3K and PDK1
R8886 T13393 T13370 partOf Thr308,Thr308 Akt
R8887 T13393 T13399 siteOf Thr308,Thr308 Akt phosphorylation
R8888 T13395 T13394 themeOf inhibited Akt Thr308 phosphorylation,inhibited Akt Thr308 phosphorylation