scholarly journals A cell cycle kinase-phosphatase module restrains PI3K-Akt activity in an mTORC1-dependent manner

2020 ◽  
Author(s):  
Belén Sanz-Castillo ◽  
Begoña Hurtado ◽  
Aicha El Bakkali ◽  
Dario Hermida ◽  
Beatriz Salvador-Barbero ◽  
...  

AbstractThe AKT-mTOR pathway is a central regulator of cell growth and metabolism. Upon sustained mTOR activity, AKT activity is attenuated by a feedback loop that restrains upstream signaling. However, how cells control the signals that limit AKT activity is not fully understood. Here we show that MASTL/Greatwall, a cell-cycle kinase that supports mitosis by phosphorylating the PP2A/B55 inhibitors ENSA/ARPP19, inhibits PI3K-AKT activity by sustaining mTORC1- and S6K1-dependent phosphorylation of IRS1 and GRB10. Genetic depletion of MASTL results in an inefficient feedback loop and AKT hyperactivity. These defects are rescued by expression of phospho-mimetic ENSA/ARPP19 or inhibition of PP2A/B55 phosphatases. MASTL is directly phosphorylated by mTORC1, thereby limiting the PP2A/B55-dependent dephosphorylation of IRS1 and GRB10 downstream of mTORC1. Downregulation of MASTL results in increased glucose uptake in vitro and increased glucose tolerance in adult mice, suggesting the relevance of the MASTL-PP2A/B55 kinase-phosphatase module in controlling AKT and maintaining metabolic homeostasis.

Development ◽  
1995 ◽  
Vol 121 (10) ◽  
pp. 3259-3266 ◽  
Author(s):  
K.T. Jones ◽  
J. Carroll ◽  
J.A. Merriman ◽  
D.G. Whittingham ◽  
T. Kono

Mature mouse oocytes are arrested at metaphase of the second meiotic division. Completion of meiosis and a block to polyspermy is caused by a series of repetitive Ca2+ transients triggered by the sperm at fertilization. These Ca2+ transients have been widely reported to last for a number of hours but when, or why, they cease is not known. Here we show that Ca2+ transients cease during entry into interphase, at the time when pronuclei are forming. In fertilized oocytes arrested at metaphase using colcemid, Ca2+ transients continued for as long as measurements were made, up to 18 hours after fertilization. Therefore sperm is able to induce Ca2+ transients during metaphase but not during interphase. In addition metaphase II oocytes, but not pronuclear stage 1-cell embryos showed highly repetitive Ca2+ oscillations in response to microinjection of inositol trisphosphate. This was explored further by treating in vitro maturing oocytes at metaphase I for 4–5 hours with cycloheximide, which induced nuclear progression to interphase (nucleus formation) and subsequent re-entry to metaphase (nuclear envelope breakdown). Fertilization of cycloheximide-treated oocytes revealed that continuous Ca2+ oscillations in response to sperm were observed after nuclear envelope breakdown but not during interphase. However interphase oocytes were able to generate Ca2+ transients in response to thimerosal. This data suggests that the ability of the sperm to trigger repetitive Ca2+ transients in oocytes is modulated in a cell cycle-dependent manner.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3367-3367 ◽  
Author(s):  
Weiguo Zhang ◽  
Marina Konopleva ◽  
Teresa McQueen ◽  
Jorje Cortes ◽  
James McCubrey ◽  
...  

Abstract We have previously demonstrated constitutive activation of MAPK signaling in 70% of primary AML samples (Millela et al, JCI108:851–859, 2001), suggesting that upstream kinases (Raf and MEK) may play a role in the leukemic transformation of myeloid cells. BAY 43-9006 is a small molecule Raf kinase inhibitor that has demonstrated potent anti-tumor activity against solid human tumors in xenograft models. In this study, we tested the hypothesis that BAY 43-9006 inhibits leukemia cell growth and/or induces apoptosis by suppressing the activity of the MAPK pathway. In the in vitro kinase assay, BAY 43-9006 inhibited both Raf-1 and B-Raf-mediated MEK1 phosphorylation in a dose-dependent manner, with Raf-1 kinase being more sensitive to the inhibitory effects of BAY 43-9006 (IC50Raf-1, 1.37 μM vs. IC50B-Raf, 4.64 μM). BAY 43-9006 suppressed MEK1/2 and ERK phosphorylation in the AML cell lines OCI-AML3, HL-60, U937 and KG-1 in a dose-dependent manner after 24 hr treatment. Unexpectedly, BAY 43-9006 also inhibited AKT phosphorylation on Ser473 (after 4.5 hrs). BAY 43-9006 inhibited growth of AML cells in a dose- and time-dependent manner. The 50% inhibitory concentration (IC50) of BAY 43-9006 was 0.39, 1.14, 2.86 and 2.80 μM, respectively in OCI-AML3, HL-60, U937 and KG-1 cells after 72 hrs. This growth-inhibitory effect was mediated by a dose-dependent induction of cell cycle arrest in G1 mediated by the down-regulation of the cell cycle-related proteins cyclin E, cdk2 and cdc2, followed by induction of apoptosis after 72 hrs. In primary AML patient samples, BAY 43-9006 not only inhibited cell growth and induced apoptosis after 48–72 hrs in vitro, but also preferentially inhibited colony formation of AML progenitor cells compared to normal bone marrow cells [IC50: 2.33 μM vs. 9.34μM (CFU-GM), 5.69 μM (Erythroid) and 3.75 μM (Mixed), respectively]. Time-course analyses demonstrated that BAY 43-9006 suppressed phosphorylation of the pro-apoptotic protein Bim (at 4.5 hrs), caused loss of the mitochondrial membrane potential and cytochrome c release (at 6 hrs) followed by cleavage of caspases-3 and -9 but not of caspase-8, suggesting primary involvement of the intrinsic mitochondrial pathway. Furthermore, the pro-apoptotic proteins Bim and Bax were up-regulated after 48 hrs of BAY 43-9006 treatment, and the level of the inhibitor-of-apoptosis protein Survivin was down-regulated after 48 hrs. In summary, our data demonstrates that BAY 43-9006 inhibits Raf-MEK-ERK signaling and induces apoptosis in AML via Bim de-phosphorylation and activation of the intrinsic apoptotic pathway. The potential of BAY 43-9006 in the therapy of AML patients will be tested in a Phase I clinical trial.


2021 ◽  
Author(s):  
Alexander Keller ◽  
Yingnan Lei ◽  
Nuša Krivec ◽  
Edouard Couvreu De Deckersberg ◽  
Dominika Dziedzicka ◽  
...  

Though gains of chromosome 12p13.31 are highly recurrent in hPSC, their impact on differentiation is poorly understood. We identify a reduction in differentiation capacity towards all three germ layers and a subpopulation of residual pluripotent cells that appear during hepatic specification. These cells form as a result of the overexpression of NANOG and GDF3, whereby NANOG as the primary driver delays activation of WNT signaling, partly as a result of a direct physical interaction with TCF7. Entry into the residual state is determined by cell cycle position at the onset of differentiation and is maintained by a feedback loop between NANOG and GDF3. These findings highlight the ability of genetically abnormal hPSC to escape correct differentiation and to form residual pluripotent cells, an important risk in the safe clinical translation of hPSC. Our results further refine the molecular mechanisms that underpin the exit from pluripotency and onset of differentiation.


2017 ◽  
Vol 37 (6) ◽  
Author(s):  
Jie Ma ◽  
Xian-Bin Wang ◽  
Rui Li ◽  
Shu-Hong Xuan ◽  
Fang Wang ◽  
...  

Esophageal cancer (EC) remains an important health problem in China. In the present study, through the use of siRNA, specific gene knockdown of transcription factor 3 gene (TCF-3) was achieved in vitro and the effect of TCF-3 gene on human EC Eca-109 cell proliferation and apoptosis. Eca-109 cells were treated using negative control (NC) of siRNA against TCF-3 (siTCF-3) and siTCF-3 group. Colony formation assay was used to detect the colony formation ability in Eca-109 cells. MTT assay was used to measure the cell growth and viability, whereas BrDU assay was used to evaluate cell proliferation, and flow cytometry (FCM) to assess cell apoptosis. Reverse-transcription quantitative PCR (RT-qPCR) was applied to measure TCF-3 gene expression. Protein expressions of TCF-3, apoptosis-related proteins, Bcl-2, Bax, and caspase-3 were determined using Western blotting. Transfection of siTCF-3 successfully down-regulated TCF-3 gene expression. In addition, siTCF-3, reduced Eca-109 cell viability and proliferation, in a time-dependent manner, and inhibited progression of cell cycle from G0/G1 to S-stage. When treated with siTCF-3, the Eca-109 cells exhibited increased apoptosis, with up-regulated cleaved caspase and Bax expressions, whereas Bcl-2 expression was down-regulated. The present study shows that TCF-3 gene silencing inhibits Eca-109 cell growth and proliferation, suppresses cell cycle progression, and promotes apoptosis, which might serve as a new objective for EC treatment.


1998 ◽  
Vol 333 (3) ◽  
pp. 543-548 ◽  
Author(s):  
Takeo USUI ◽  
Masuo KONDOH ◽  
Cheng-Bin CUI ◽  
Tadanori MAYUMI ◽  
Hiroyuki OSADA

We have investigated the cell cycle inhibition mechanism and primary target of tryprostatin A (TPS-A) purified from Aspergillus fumigatus. TPS-A inhibited cell cycle progression of asynchronously cultured 3Y1 cells in the M phase in a dose- and time-dependent manner. In contrast, TPS-B (the demethoxy analogue of TPS-A) showed cell-cycle non-specific inhibition on cell growth even though it inhibited cell growth at lower concentrations than TPS-A. TPS-A treatment induced the reversible disruption of the cytoplasmic microtubules of 3Y1 cells as observed by indirect immunofluorescence microscopy in the range of concentrations that specifically inhibited M-phase progression. TPS-A inhibited the assembly in vitro of microtubules purified from bovine brains (40% inhibition at 250 µM); however, there was little or no effect on the self-assembly of purified tubulin when polymerization was induced by glutamate even at 250 µM TPS-A. TPS-A did not inhibit assembly promoted by taxol or by digestion of the C-terminal domain of tubulin. However, TPS-A blocked the tubulin assembly induced by inducers interacting with the C-terminal domain, microtubule-associated protein 2 (MAP2), tau and poly-(l-lysine). These results indicate that TPS-A is a novel inhibitor of MAP-dependent microtubule assembly and, through the disruption of the microtubule spindle, specifically inhibits cell cycle progression at the M phase.


2015 ◽  
Vol 35 (23) ◽  
pp. 4043-4052 ◽  
Author(s):  
Junyue Xing ◽  
Jie Yi ◽  
Xiaoyu Cai ◽  
Hao Tang ◽  
Zhenyun Liu ◽  
...  

The tRNA methytransferase NSun2 promotes cell proliferation, but the molecular mechanism has not been elucidated. Here, we report that NSun2 regulates cyclin-dependent kinase 1 (CDK1) expression in a cell cycle-dependent manner. Knockdown of NSun2 decreased the CDK1 protein level, while overexpression of NSun2 elevated it without alteringCDK1mRNA levels. Further studies revealed that NSun2 methylatedCDK1mRNAin vitroand in cells and that methylation by NSun2 enhanced CDK1 translation. Importantly, NSun2-mediated regulation of CDK1 expression had an impact on the cell division cycle. These results provide new insight into the regulation of CDK1 during the cell division cycle.


2018 ◽  
Vol 19 (12) ◽  
pp. 3935 ◽  
Author(s):  
Linfu He ◽  
Wenjing Shi ◽  
Xiaocui Liu ◽  
Xiaohuan Zhao ◽  
Zhicai Zhang

Isaria cicadae, a medicinal food fungus, is a fruit from Paecilomyces cicadae. In this study, we purified ergosterol peroxide (EP) from the fermentation broth of P. cicadae and investigated its effects on renal cell carcinoma (RCC) cells, in vitro. EP was purified from P. cicadae fermentation broth. The human RCC cell line 786-0 was used to analyze the anticancer mechanism of EP and inhibit its effect on cancer cell proliferation, in vitro. EP with a validated structure showed a yield rate of 20.1 mg/L and a purity of 96%. EP significantly inhibited RCC cell growth and clone formation in vitro. In addition, EP suppressed the migration and invasion, triggered the apoptosis, and modulated the cell cycle of RCC cells, in a dose-dependent manner. It also downregulated β-catenin expression. EP could be routinely produced through P. cicadae. It fights RCC cells in vitro through multiple mechanisms, including suppressing cell growth, colonization, migration, and invasion, arresting the cell cycle, attenuating β-catenin pathways, and triggering apoptosis.


1999 ◽  
Vol 19 (1) ◽  
pp. 646-656 ◽  
Author(s):  
Christian Voitenleitner ◽  
Christoph Rehfuess ◽  
Melissa Hilmes ◽  
Lynda O’Rear ◽  
Pao-Chi Liao ◽  
...  

ABSTRACT DNA polymerase α-primase is known to be phosphorylated in human and yeast cells in a cell cycle-dependent manner on the p180 and p68 subunits. Here we show that phosphorylation of purified human DNA polymerase α-primase by purified cyclin A/cdk2 in vitro reduced its ability to initiate simian virus 40 (SV40) DNA replication in vitro, while phosphorylation by cyclin E/cdk2 stimulated its initiation activity. Tryptic phosphopeptide mapping revealed a family of p68 peptides that was modified well by cyclin A/cdk2 and poorly by cyclin E/cdk2. The p180 phosphopeptides were identical with both kinases. By mass spectrometry, the p68 peptide family was identified as residues 141 to 160. Cyclin A/cdk2- and cyclin A/cdc2-modified p68 also displayed a phosphorylation-dependent shift to slower electrophoretic mobility. Mutation of the four putative phosphorylation sites within p68 peptide residues 141 to 160 prevented its phosphorylation by cyclin A/cdk2 and the inhibition of replication activity. Phosphopeptide maps of the p68 subunit of DNA polymerase α-primase from human cells, synchronized and labeled in G1/S and in G2, revealed a cyclin E/cdk2-like pattern in G1/S and a cyclin A/cdk2-like pattern in G2. The slower-electrophoretic-mobility form of p68 was absent in human cells in G1/S and appeared as the cells entered G2/M. Consistent with this, the ability of DNA polymerase α-primase isolated from synchronized human cells to initiate SV40 replication was maximal in G1/S, decreased as the cells completed S phase, and reached a minimum in G2/M. These results suggest that the replication activity of DNA polymerase α-primase in human cells is regulated by phosphorylation in a cell cycle-dependent manner.


1999 ◽  
Vol 146 (6) ◽  
pp. 1265-1276 ◽  
Author(s):  
Stephen L. Rogers ◽  
Ryan L. Karcher ◽  
Joseph T. Roland ◽  
Alexander A. Minin ◽  
Walter Steffen ◽  
...  

Previously, we have shown that melanosomes of Xenopus laevis melanophores are transported along both microtubules and actin filaments in a coordinated manner, and that myosin V is bound to purified melanosomes (Rogers, S., and V.I. Gelfand. 1998. Curr. Biol. 8:161–164). In the present study, we have demonstrated that myosin V is the actin-based motor responsible for melanosome transport. To examine whether myosin V was regulated in a cell cycle-dependent manner, purified melanosomes were treated with interphase- or metaphase-arrested Xenopus egg extracts and assayed for in vitro motility along Nitella actin filaments. Motility of organelles treated with mitotic extract was found to decrease dramatically, as compared with untreated or interphase extract-treated melanosomes. This mitotic inhibition of motility correlated with the dissociation of myosin V from melanosomes, but the activity of soluble motor remained unaffected. Furthermore, we find that myosin V heavy chain is highly phosphorylated in metaphase extracts versus interphase extracts. We conclude that organelle transport by myosin V is controlled by a cell cycle-regulated association of this motor to organelles, and that this binding is likely regulated by phosphorylation of myosin V during mitosis.


Sign in / Sign up

Export Citation Format

Share Document