92 results on '"mTORC1"'
Search Results
2. SPOP negatively regulates mTORC1 activity by ubiquitinating Sec13.
- Author
-
Yang, Yong, Han, Yan-Chun, Cao, Qi, Wang, Xi, Wei, Xiao-Dan, Shang, Meng-Di, Zhang, Xiao-Gang, Li, Xiao, Hu, Bin, Tian, Cheng-Yang, Yang, Zhen-Lin, Liu, Ke-Hui, and Wang, Jiu-Qiang
- Subjects
- *
AMINO acid metabolism , *CANCER cell migration , *CANCER cell proliferation , *TUMOR suppressor genes , *CELL metabolism , *CELLULAR signal transduction - Abstract
The mammalian target of rapamycin complex1 (mTORC1) can response to amino acid to regulate metabolism and cell growth. GATOR2 act as important role in amino acid mediated mTORC1 signaling pathway by repressing GTPase activity (GAP) of GATOR1. However, it is still unclear how GATOR2 regulates mTORC1 signaling pathway. Here, we found that K63-ubiquitination of Sce13, one component of GATOR2, suppresses the mTORC1 activity by lessening the inter-interaction of GATOR2. Mechanistically, the ubiquitination of Sec13 was mediated by SPOP. Subsequently, the ubiquitination of Sec13 attenuated its interaction with the other component of GATOR2, thus suppressing the activity of mTORC1. Importantly, the deficiency of SPOP promoted the faster proliferation and migration of breast cancer cells, which was attenuated by knocking down of Sec13. Therefore, SPOP can act as a tumor suppressor gene by negatively regulating mTORC1 signaling pathway. • SPOP promote the K63 linked ubiquitination of Sec13. • The ubiquitinated Sec13 destabilized the GATOR2 complex. • The deficiency of SPOP promotes the faster migration and proliferation of breast cancer cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Recent progress in the study of the Rheb family GTPases
- Author
-
Heard, Jeffrey J, Fong, Valerie, Bathaie, S Zahra, and Tamanoi, Fuyuhiko
- Subjects
Cancer ,Brain Disorders ,1.1 Normal biological development and functioning ,Underpinning research ,Animals ,Cardiovascular System ,Humans ,Lysosomes ,Mechanistic Target of Rapamycin Complex 1 ,Monomeric GTP-Binding Proteins ,Multiprotein Complexes ,Protein Structure ,Tertiary ,TOR Serine-Threonine Kinases ,Rheb GTPase ,mTORC1 ,Lysosome ,Mouse study ,Structure ,Mutants ,Biochemistry and Cell Biology ,Medical Physiology ,Biochemistry & Molecular Biology - Abstract
In this review we highlight recent progress in the study of Rheb family GTPases. Structural studies using X-ray crystallography and NMR have given us insight into unique features of this GTPase. Combined with mutagenesis studies, these works have expanded our understanding of residues that affect Rheb GTP/GDP bound ratios, effector protein interactions, and stimulation of mTORC1 signaling. Analysis of cancer genome databases has revealed that several human carcinomas contain activating mutations of the protein. Rheb's role in activating mTORC1 signaling at the lysosome in response to stimuli has been further elucidated. Rheb has also been suggested to play roles in other cellular pathways including mitophagy and peroxisomal ROS response. A number of studies in mice have demonstrated the importance of Rheb in development, as well as in a variety of functions including cardiac protection and myelination. We conclude with a discussion of future prospects in the study of Rheb family GTPases.
- Published
- 2014
4. The amino acid transporter PAT1 regulates mTORC1 in a nutrient-sensitive manner that requires its transport activity.
- Author
-
Zhao, Lingling, Zhang, Xiangxiang, Ji, Xin, Jin, Yaping, and Liu, Wei
- Subjects
- *
AMINO acids , *PROTONS , *CELL proliferation , *LYSOSOMES , *KIDNEYS - Abstract
Abstract The proton-coupled amino acid transporter PAT1 has been postulated to regulate the amino acid-stimulated mTORC1 through two different mechanisms, either it activates mTORC1 by sensing and transducing the lysosomal amino acid signal to mTORC1, or it inhibits mTORC1 by decreasing the signal level, as increased PAT1 has been shown to either activate or inactivate mTORC1 in the human embryonic kidney HEK293 cells. The current study aims to clarify the cause of these controversial observations, which is promoted by the recent discovery that the lysosomal PAT1 can be induced by starvation. Here, we show that under the normal culture condition, overexpression of PAT1 did not apparently change the mTORC1 activity in the fast proliferating cells. However when these cells were synchronized by starvation, followed by nutrient replenishment for a short period of time, the mTORC1 activity was decreased by PAT1 overexpression; if the nutrient stimulation lasted for longer time, the mTORC1 activities could be recovered in the PAT1-overexpressing cells. In addition, we showed the starvation-induced lysosomal PAT1 was gradually decreased during the nutrient replenishment. These results reveal that the influence of PAT1 on mTORC1 seems to be affected by the nutrient condition and the level of lysosomal PAT1. We further demonstrate that suppressing the transport activity of PAT1 abolished its inhibitory effect on mTORC1. Our data support a mechanism that PAT1 can negatively regulate mTORC1 by controlling the cellular nutrient signal level. Highlights • Increasing the PAT1 level does not readily increase the mTORC1 activity. • The lysosomal PAT1 was increased by starvation and decreased by nutrient replenishment. • The lysosomal PAT1 plays a negative role on the mTORC1 activity. • The transport activity is required for PAT1 to regulate mTORC1. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
5. Direct and indirect activation of eukaryotic elongation factor 2 kinase by AMP-activated protein kinase.
- Author
-
Johanns, M., Pyr dit Ruys, S., Houddane, A., Vertommen, D., Herinckx, G., Hue, L., Proud, C.G., and Rider, M.H.
- Subjects
- *
EUKARYOTIC cell genetics , *PROTEIN synthesis , *PROTEIN kinases , *MUTAGENESIS , *PHOSPHORYLATION - Abstract
Background Eukaryotic elongation factor 2 (eEF2) kinase (eEF2K) is a key regulator of protein synthesis in mammalian cells. It phosphorylates and inhibits eEF2, the translation factor necessary for peptide translocation during the elongation phase of protein synthesis. When cellular energy demand outweighs energy supply, AMP-activated protein kinase (AMPK) and eEF2K become activated, leading to eEF2 phosphorylation, which reduces the rate of protein synthesis, a process that consumes a large proportion of cellular energy under optimal conditions. Aim The goal of the present study was to elucidate the mechanisms by which AMPK activation leads to increased eEF2 phosphorylation to decrease protein synthesis. Methods Using genetically modified mouse embryo fibroblasts (MEFs), effects of treatments with commonly used AMPK activators to increase eEF2 phosphorylation were compared with that of the novel compound 991. Bacterially expressed recombinant eEF2K was phosphorylated in vitro by recombinant activated AMPK for phosphorylation site-identification by mass spectrometry followed by site-directed mutagenesis of the identified sites to alanine residues to study effects on the kinetic properties of eEF2K. Wild-type eEF2K and a Ser491/Ser492 mutant were retrovirally re-introduced in eEF2K-deficient MEFs and effects of 991 treatment on eEF2 phosphorylation and protein synthesis rates were studied in these cells. Results & conclusions AMPK activation leads to increased eEF2 phosphorylation in MEFs mainly by direct activation of eEF2K and partly by inhibition of mammalian target of rapamycin complex 1 (mTORC1) signaling. Treatment of MEFs with AMPK activators can also lead to eEF2K activation independently of AMPK probably via a rise in intracellular Ca 2+ . AMPK activates eEF2K by multi-site phosphorylation and the newly identified Ser491/Ser492 is important for activation, leading to mTOR-independent inhibition of protein synthesis. Our study provides new insights into the control of eEF2K by AMPK, with implications for linking metabolic stress to decreased protein synthesis to conserve energy reserves, a pathway that is of major importance in cancer cell survival. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
6. PAQR3 augments amino acid deprivation-induced autophagy by inhibiting mTORC1 signaling.
- Author
-
Wang, Lin, Pan, Yi, Huang, Meiqin, You, Xue, Guo, Feifan, and Chen, Yan
- Subjects
- *
MTOR protein , *AMINO acids , *PROGESTATIONAL hormones , *AUTOPHAGY , *CELLULAR signal transduction - Abstract
Amino acids are the key activators of the mTOR complex 1 (mTORC1, mainly composed of mTOR, Raptor and mLST8) required for cell growth and proliferation. On the other hand, deprivation of amino acids induces autophagy via inhibition of mTORC1 signaling. We report here that amino acid-induced mTORC1 activity and amino acid deprivation-induced autophagy are regulated by PAQR3, a newly found tumor suppressor. At the cellular level, PAQR3 negatively regulates amino acid-induced activation of mTORC1. The N-terminal end of PAQR3 interacts with the WD domains of Raptor and mLST8 directly. PAQR3 reduces the interaction of mTOR with Raptor and mLST8, thus disrupts formation of intact mTORC1 complex. PAQR3 modulates leucine-induced alteration in cell size. In addition, PAQR3 knockdown reduces amino acid deprivation-induced autophagy. The inhibitory effect of PAQR3 knockdown on autophagy is abrogated by rapamycin treatment, indicating that PAQR3 modulates autophagy via its regulation on mTORC1 signaling. In conclusion, our finding reveals a new mode of regulation of mTORC1 signaling and autophagy by PAQR3 in response to alterations of amino acids. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
7. A novel mechanism of mTORC1-mediated serine/glycine metabolism in osteosarcoma development.
- Author
-
Wang, Da-wei, Wu, Liwen, Cao, Yang, Yang, Lei, Liu, Wei, E, Xiao-qiang, Ji, Guangrong, and Bi, Zheng-gang
- Subjects
- *
OSTEOSARCOMA , *TOR proteins , *DRUG resistance in cancer cells , *GENE expression , *CELLULAR signal transduction - Abstract
Osteosarcoma is the major malignant primary bone cancer in children and adolescents, which is highly aggressive with frequent acquisition of chemoresistance phenotypes. Although much progress has been made, mechanisms of osteosarcoma rapid growth and chemoresistance are still not well elucidated. Generally, alternated metabolic characterization has been proposed to be a hallmark of cancer, yet it is lack of a systematic characterization of cancer metabolic networks. In the present study, we aim to characterize osteosarcoma metabolism and key regulators to reveal mechanisms of how osteosarcoma grows and resists apoptosis under stress conditions. The results demonstrate that mTORC1 pathway is hyperactivated in clinical osteosarcoma samples. However, inhibition of mTORC1 may not be enough to induce significant death of osteosarcoma cells. Results of GC-TOFMS suggested that inhibition of mTORC1 reduce one-carbon amino acids, serine and glycine, in osteosarcoma cells. Moreover, mTORC1 regulates serine/glycine de novo synthesis via modulating glycolysis and serine/glycine synthesis gene expressions. Further, mTORC1/serine/glycine metabolic axis promotes osteosarcoma proliferation and antioxidant ability to environmental stress, which finally leads to cell survival. Our results identify a novel mechanism of mTORC1-mediated serine/glycine metabolism as a significant protective system in osteosarcoma cells. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
8. LysM-positive neurons drive Tuberous Sclerosis Complex (TSC)-associated brain lesions.
- Author
-
Zhang, Jiahuan, Xu, Song, Liang, Kangyan, Cao, Xiong, Ye, Zhixin, Huang, Wenlan, Bai, Xiaochun, and Zhang, Yue
- Subjects
- *
TUBEROUS sclerosis , *BRAIN damage , *CENTRAL nervous system , *MYELOID cells , *NEURONS , *MICROGLIA , *CEREBRAL cortex - Abstract
Mutations of Tsc1 or Tsc2 can lead to excessive activation of mTORC1 and cause Tuberous Sclerosis Complex (TSC), which is an autosomal dominant genetic disease prominently characterized by seizures, mental retardation and multiorgan hamartoma. In TSC, pathological changes in the central nervous system are the leading cause of death and disability. In decades, series of rodent models have been established by mutating Tsc1 or Tsc2 genes in diverse neural cell lineages to investigate the underlying cellular and molecular mechanisms, however, the cellular origin triggering neural pathological changes in TSC is undetermined. In this study, we generated a novel mouse model involving conditional deletion of Tsc1 in lysozyme 2 (Lyz2)-positive cells which replicated several features of brain lesions including epileptic seizures, megalencephaly, highly enlarged pS6-positive neurons and astrogliosis. In addition, we confirmed that bone marrow-derived myeloid cells including microglia with Tsc1 deficiency are not the decisive lineage in the cerebral pathologies in TSC. These histological assays in our murine model indicate an essential contribution of Lyz2-positive neurons to TSC progression. The Lyz2-positive neural population-specific onset of Tsc1 loss in murine postnatal brain might be the key to pathological phenotypes. Our findings thus provided evidences supporting new insights into the role of Lyz2-positive neurons in TSC events. • A population of Lyz2-positive neurons presents in murine cerebral cortex. • Loss of Tsc1 in Lyz2-positive neuronal cells generate a mouse model mimicking TSC. • Lyz2-positive neuronal cells are expressing GAD67. • Tsc1 deficiency in myeloid cells has less impact on TSC pathology. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. The amino acid transporter PAT1 regulates mTORC1 in a nutrient-sensitive manner that requires its transport activity
- Author
-
Lingling Zhao, Wei Liu, Xiangxiang Zhang, Xin Ji, and Yaping Jin
- Subjects
0301 basic medicine ,Amino Acid Transport Systems ,Period (gene) ,Stimulation ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,03 medical and health sciences ,0302 clinical medicine ,Lysosome ,medicine ,Humans ,Amino acid transporter ,chemistry.chemical_classification ,Symporters ,fungi ,HEK 293 cells ,Epithelial Cells ,Nutrients ,Cell Biology ,Embryonic stem cell ,Amino acid ,Cell biology ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,biological phenomena, cell phenomena, and immunity ,Lysosomes - Abstract
The proton-coupled amino acid transporter PAT1 has been postulated to regulate the amino acid-stimulated mTORC1 through two different mechanisms, either it activates mTORC1 by sensing and transducing the lysosomal amino acid signal to mTORC1, or it inhibits mTORC1 by decreasing the signal level, as increased PAT1 has been shown to either activate or inactivate mTORC1 in the human embryonic kidney HEK293 cells. The current study aims to clarify the cause of these controversial observations, which is promoted by the recent discovery that the lysosomal PAT1 can be induced by starvation. Here, we show that under the normal culture condition, overexpression of PAT1 did not apparently change the mTORC1 activity in the fast proliferating cells. However when these cells were synchronized by starvation, followed by nutrient replenishment for a short period of time, the mTORC1 activity was decreased by PAT1 overexpression; if the nutrient stimulation lasted for longer time, the mTORC1 activities could be recovered in the PAT1-overexpressing cells. In addition, we showed the starvation-induced lysosomal PAT1 was gradually decreased during the nutrient replenishment. These results reveal that the influence of PAT1 on mTORC1 seems to be affected by the nutrient condition and the level of lysosomal PAT1. We further demonstrate that suppressing the transport activity of PAT1 abolished its inhibitory effect on mTORC1. Our data support a mechanism that PAT1 can negatively regulate mTORC1 by controlling the cellular nutrient signal level.
- Published
- 2019
10. Ergothioneine-induced neuronal differentiation is mediated through activation of S6K1 and neurotrophin 4/5-TrkB signaling in murine neural stem cells
- Author
-
Takahiro Ishimoto, Yukio Kato, Noritaka Nakamichi, and Yusuke Masuo
- Subjects
0301 basic medicine ,Neurogenesis ,P70-S6 Kinase 1 ,mTORC1 ,Tropomyosin receptor kinase B ,Ribosomal Protein S6 Kinases, 90-kDa ,Antioxidants ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,Neurotrophic factors ,Animals ,Nerve Growth Factors ,Cells, Cultured ,PI3K/AKT/mTOR pathway ,Mice, Inbred ICR ,Membrane Glycoproteins ,biology ,Chemistry ,Ergothioneine ,Cell Biology ,Protein-Tyrosine Kinases ,Neural stem cell ,Cell biology ,030104 developmental biology ,nervous system ,030220 oncology & carcinogenesis ,biology.protein ,Signal Transduction ,Neurotrophin - Abstract
The promotion of neurogenesis is considered to be an effective therapeutic strategy for neuropsychiatric disorders because impairment of neurogenesis is associated with the onset and progression of these disorders. We have previously demonstrated that orally ingested ergothioneine (ERGO), a naturally occurring antioxidant and hydrophilic amino acid, promotes neurogenesis in the hippocampal dentate gyrus (DG) with its abundant neural stem cells (NSCs) and exerts antidepressant-like effects in mice. Independent of its antioxidant activities, ERGO induces in cultured NSCs this differentiation through induction of the basic helix-loop-helix transcription factor Math1. However, the upstream signaling of Math1 in the mechanisms underlying ERGO-induced neuronal differentiation remains unclear. The purpose of the present study was to elucidate the upstream signaling with the aim of discovering novel targets for the treatment of neuropsychiatric disorders. We focused on neurotrophic factor signaling, as it is important for the promotion of neurogenesis and the induction of antidepressant effects. We also focused on the signaling of the mammalian target of rapamycin (mTOR) complex 1 (mTORC1), a known amino acid sensor, and the members of this signaling pathway, mTOR and p70 ribosomal protein S6 kinase 1 (S6K1). Exposure of cultured NSCs to ERGO significantly increased the expression of phosphorylated S6K1 (p-S6K1) at Thr389 in only 1 h, of phosphorylated mTOR (p-mTOR) in 6 h, and of the gene product of neurotrophin 4/5 (NT5) which activates tropomyosin receptor kinase B (TrkB) in 24 h. ERGO increased the population of βIII-tubulin-positive neurons, and this effect was suppressed by the inhibitors of S6K1 (PF4708671), mTORC1 (rapamycin), and TrkB (GNF5837). Oral administration of ERGO to mice significantly increased in the DG the expression of p-S6K1 at Thr389, the gene product of NT5, and phosphorylated TrkB but not that of p-mTOR. Thus, neuronal differentiation of NSCs induced by ERGO is mediated, at least in part, through phosphorylation of S6K1 at Thr389 and subsequent activation of TrkB signaling through the induction of NT5. Thus, S6K1 and NT5 might be promising target molecules for the treatment of neuropsychiatric disorders.
- Published
- 2019
11. BAFF inhibits autophagy promoting cell proliferation and survival by activating Ca2+-CaMKII-dependent Akt/mTOR signaling pathway in normal and neoplastic B-lymphoid cells
- Author
-
Chunxiao Liu, Chong Xu, Long Chen, Ruijie Zhang, Qingyu Zeng, Xiaoqing Dong, Hai Zhang, Jing Ma, Shile Huang, Shuangquan Zhang, and Jiamin Qin
- Subjects
0301 basic medicine ,Cell growth ,Chemistry ,Autophagy ,Cell Biology ,mTORC1 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Ca2+/calmodulin-dependent protein kinase ,Cancer research ,Phosphorylation ,biological phenomena, cell phenomena, and immunity ,B-cell activating factor ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
B cell activating factor from the TNF family (BAFF) is implicated in not only the physiology of normal B cells, but also the pathophysiology of aggressive B cells related to malignant and autoimmune diseases. Autophagy plays a crucial role in balancing the beneficial and detrimental effects of immunity and inflammation. However, little is known about whether and how excessive BAFF mediates autophagy contributing to B-cell proliferation and survival. Here, we show that excessive human soluble BAFF (hsBAFF) inhibited autophagy with a concomitant reduction of LC3-II in normal and B-lymphoid (Raji) cells. Knockdown of LC3 not only potentiated hsBAFF inhibition of autophagy, but also attenuated hsBAFF activation of Akt/mTOR pathway, thereby diminishing hsBAFF-induced B-cell proliferation/viability. Further, we found that hsBAFF inhibition of autophagy was Akt/mTOR-dependent. This is supported by the findings that hsBAFF increased mTORC1-mediated phosphorylation of ULK1 (Ser757); Akt inhibitor X, mTORC1 inhibitor rapamycin, mTORC1/2 inhibitor PP242, expression of dominant negative Akt, or knockdown of mTOR attenuated hsBAFF-induced phosphorylation of ULK1, decrease of LC3-II level, and increase of cell proliferation/viability. Chelating intracellular free Ca2+ ([Ca2+]i) with BAPTA/AM or preventing [Ca2+]i elevation using EGTA or 2-APB profoundly blocked hsBAFF-induced activation of Akt/mTOR, phosphorylation of ULK1 and decrease of LC3-II, as well as increase of cell proliferation/viability. Similar effects were observed in the cells where CaMKII was inhibited by KN93 or knocked down by CaMKII shRNA. Collectively, these results indicate that hsBAFF inhibits autophagy promoting cell proliferation and survival through activating Ca2+-CaMKII-dependent Akt/mTOR signaling pathway in normal and neoplastic B-lymphoid cells. Our findings suggest that manipulation of intracellular Ca2+ level or CaMKII, Akt, or mTOR activity to promote autophagy may be exploited for prevention of excessive BAFF-induced aggressive B lymphocyte disorders and autoimmune diseases.
- Published
- 2019
12. High glucose-stimulated enhancer of zeste homolog-2 (EZH2) forces suppression of deptor to cause glomerular mesangial cell pathology
- Author
-
Nandini Ghosh-Choudhury, Amrita Kamat, Falguni Das, Kavitha Sataranatarajan, Goutam Ghosh Choudhury, Balakuntalam S. Kasinath, and Amit Bera
- Subjects
biology ,Mesangial cell ,Chemistry ,Glomerular Mesangial Cell ,Intracellular Signaling Peptides and Proteins ,macromolecular substances ,Cell Biology ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,Mechanistic Target of Rapamycin Complex 1 ,DEPTOR ,mTORC2 ,Diabetes Mellitus, Experimental ,Fibronectin ,Mice ,Glucose ,Downregulation and upregulation ,Mesangial Cells ,Cancer research ,biology.protein ,Animals ,Enhancer of Zeste Homolog 2 Protein ,PI3K/AKT/mTOR pathway - Abstract
Function of mTORC1 and mTORC2 has emerged as a driver of mesangial cell pathologies in diabetic nephropathy. The mechanism of mTOR activation is poorly understood in this disease. Deptor is a constitutive subunit and a negative regulator of both mTOR complexes. Mechanistic investigation in mesangial cells revealed that high glucose decreased the expression of deptor concomitant with increased mTORC1 and mTORC2 activities, induction of hypertrophy and, expression of fibronectin and PAI-1. shRNAs against deptor mimicked these pathologic outcomes of high glucose. Conversely, overexpression of deptor significantly inhibited all effects of high glucose. To determine the mechanism of deptor suppression, we found that high glucose significantly increased the expression of EZH2, resulting in lysine-27 tri-methylation of histone H3 (H3K27Me3). Employing approaches including pharmacological inhibition, shRNA-mediated downregulation and overexpression of EZH2, we found that EZH2 regulates high glucose-induced deptor suppression along with activation of mTOR, mesangial cell hypertrophy and fibronectin/PAI-1 expression. Moreover, expression of hyperactive mTORC1 reversed shEZH2-mediated inhibition of hypertrophy and expression of fibronectin and PAI-1 by high glucose. Finally, in renal cortex of diabetic mice, we found that enhanced expression of EZH2 is associated with decreased deptor levels and increased mTOR activity and, expression of fibronectin and PAI-1. Together, our findings provide a novel mechanism for mTOR activation via EZH2 to induce mesangial cell hypertrophy and matrix expansion during early progression of diabetic nephropathy. These results suggest a strategy for leveraging the intrinsic effect of deptor to suppress mTOR activity via reducing EZH2 as a novel therapy for diabetic nephropathy.
- Published
- 2021
13. Parkin ubiquitinates mTOR to regulate mTORC1 activity under mitochondrial stress.
- Author
-
Park, Dohyun, Lee, Mi Nam, Jeong, Heeyoon, Koh, Ara, Yang, Yong Ryoul, Suh, Pann-Ghill, and Ryu, Sung Ho
- Subjects
- *
MTOR protein , *MITOCHONDRIA , *PHYSIOLOGICAL stress , *CELL growth , *CELL proliferation , *CELLULAR signal transduction , *PARKIN (Protein) - Abstract
mTORC1, a kinase complex that is considered a master regulator of cellular growth and proliferation, is regulated by many extra- and intracellular signals. Among these signals, mitochondrial status is known to have an impact on the effects of mTORC1 on cell growth and survival. However, how mitochondrial status affects mTORC1 activity, notably the molecular link, is not fully elucidated. Here, we found that Parkin can interact with and ubiquitinate mTOR. We also identified K2066 and K2306 as Parkin-dependent and mitochondrial stress-induced mTOR ubiquitination residues. This ubiquitination by Parkin is required for maintenance of mTORC1 activity under mitochondrial stress. With regard to the physiological meaning of mTORC1 activity under mitochondrial stress, we suggest that mTORC1 plays a pro-survival role. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
14. Requirement for lysosomal localization of mTOR for its activation differs between leucine and other amino acids.
- Author
-
Averous, Julien, Lambert-Langlais, Sarah, Carraro, Valérie, Gourbeyre, Ophélie, Parry, Laurent, B'Chir, Wafa, Muranishi, Yuki, Jousse, Céline, Bruhat, Alain, Maurin, Anne-Catherine, Proud, Christopher G., and Fafournoux, Pierre
- Subjects
- *
LEUCINE , *TARGETED drug delivery , *RAPAMYCIN , *CELL growth , *CELL metabolism , *GROWTH factors , *AMINO acids , *CELLULAR signal transduction - Abstract
The mammalian target of rapamycin complex 1 (mTORC1) is a master regulator of cell growth and metabolism. It controls many cell functions by integrating nutrient availability and growth factor signals. Amino acids, and in particular leucine, are among the main positive regulators of mTORC1 signaling. The current model for the regulation of mTORC1 by amino acids involves the movement of mTOR to the lysosome mediated by the Rag-GTPases. Here, we have examined the control of mTORC1 signaling and mTOR localization by amino acids and leucine in serum-fed cells, because both serum growth factors (or, e.g., insulin) and amino acids are required for full activation of mTORC1 signaling. We demonstrate that mTORC1 activity does not closely correlate with the lysosomal localization of mTOR. In particular, leucine controls mTORC1 activity without any detectable modification of the lysosomal localization of mTOR, indicating that the signal(s) exerted by leucine is likely distinct from those exerted by other amino acids. In addition, knock-down of the Rag-GTPases attenuated the inhibitory effect of amino acid- or leucine-starvation on the phosphorylation of mTORC1 targets. Furthermore, data from cells where Rag expression has been knocked down revealed that leucine can promote mTORC1 signaling independently of the lysosomal localization of mTOR. Our data complement existing models for the regulation of mTORC1 by amino acids and provide new insights into this important topic. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
15. Ceramide inhibits insulin-stimulated Akt phosphorylation through activation of Rheb/mTORC1/S6K signaling in skeletal muscle.
- Author
-
Hsieh, Chang-Ting, Chuang, Jen-Hua, Yang, Wen-Chin, Yin, Yi, and Lin, Yenshou
- Subjects
- *
CERAMIDES , *PROTEIN kinase B , *PHOSPHORYLATION , *CELLULAR signal transduction , *SKELETAL muscle , *RAPAMYCIN - Abstract
Abstract: Ceramide is a negative regulator of insulin activity. At the molecular level, it causes a decrease in insulin-stimulated Akt Ser473 phosphorylation in C2C12 myotubes. Interestingly, we found that the phosphorylation of S6K at Thr389 was increased under the same conditions. Utilizing both rapamycin to inhibit mTORC1 activity and shRNA to knock down Rheb, we demonstrated that the decrease in Akt Ser473 phosphorylation stimulated by insulin after C2-ceramide incubation can be prevented. The mechanism by which C2-ceramide impairs signaling would seem to involve a negative feedback of activated S6K via phosphorylation of insulin receptor substrate-1 at Ser636/639, since S6K inhibitor can block this phenomenon. Finally, rapamycin treatment was found not to affect C2-ceramide-induced PKCζ activation, suggesting that the pathway revealed in this study is parallel to the one involving PKCζ activation. We proposed a novel pathway/mechanism involving Rheb/mTORC1/S6K signaling to explain how C2-ceramide impairs insulin signaling via Akt phosphorylation. The existence of multiple pathways involved in insulin signaling impairment by C2-ceramide treatment implies that different strategies might be needed to ameliorate insulin resistance caused by C2-ceramide. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
16. mTOR modulates resistance to gemcitabine in lung cancer in an MTORC2 dependent mechanism
- Author
-
Philip R. Dash and Mahmoud A. Chawsheen
- Subjects
0301 basic medicine ,Programmed cell death ,Lung Neoplasms ,MAP Kinase Signaling System ,mTORC1 ,Mechanistic Target of Rapamycin Complex 2 ,DEPTOR ,mTORC2 ,Deoxycytidine ,03 medical and health sciences ,0302 clinical medicine ,Pancreatic cancer ,medicine ,Humans ,Lung cancer ,PI3K/AKT/mTOR pathway ,business.industry ,TOR Serine-Threonine Kinases ,Cell Biology ,medicine.disease ,Gemcitabine ,Neoplasm Proteins ,030104 developmental biology ,A549 Cells ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,MCF-7 Cells ,business ,medicine.drug - Abstract
Lung cancer has a poor prognosis partly due to a lack of response to treatments such as the chemotherapy drug gemcitabine. Combinations of chemotherapy drugs with signal transduction inhibitors may be more effective treatments. In this study we have investigated the impact of targeting the mTOR signalling pathway on the efficacy of gemcitabine in different cancer cell lines. Time-lapse microscopy, immuno-staining, and western blot techniques were used to evaluate the efficacy of applied treatments either in measuring phosphorylation levels of mTOR down-stream targets or in tracking down the fate of targeted cells. Reactive oxygen species and relative levels of protein phosphorylation were also quantified. For comparison between treated groups t-test and analysis of variance test were applied. Our data showed that mTORC1 has no role in sensitising A549 lung cancer cells to gemcitabine. However, targeting mTORC1/2 with the pharmacological inhibitor torin1 or by over-expressing Deptor, the negative regulator of mTOR signalling, sensitised these cells to gemcitabine. Silencing mTORC2, but not mTORC1, induced apoptosis and significantly improved the apoptosis-inducing effects of gemcitabine. Results also suggest that Rictor is required to maintain cell survival through modulating p38α, ERK1/2, RSK1/2/3 and the transcription factor STAT3. Multiple cell line comparisons revealed that PANC-1 pancreatic cancer cells were also sensitive to mTOR inhibition, but MCF7 breast cancer, MCF10A breast epithelial and H727 lung cancer cell lines were more resistant to the treatment. Inhibition of mTORC2 may have benefits in the treatment of gemcitabine resistant cancers, and the genetic background of the cell line may determine its response to mTOR inhibition. [Abstract copyright: Copyright © 2021 Elsevier Inc. All rights reserved.]
- Published
- 2020
17. Autophagy participants in the dedifferentiation of mouse 3T3-L1 adipocytes triggered by hypofunction of insulin signaling
- Author
-
Liwei Dong, Laihao Liu, Jie Pan, Suchart Kothan, Yiyi Yang, Aye Thidar Moe Moe, and Yanan Sun
- Subjects
0301 basic medicine ,medicine.medical_treatment ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,3T3-L1 Cells ,medicine ,Adipocytes ,Autophagy ,Animals ,Insulin ,White Adipocytes ,Insulin-Like Growth Factor I ,Insulin signalling ,PI3K/AKT/mTOR pathway ,Sirolimus ,biology ,Chemistry ,Imidazoles ,3T3-L1 ,Cell Biology ,Cell Dedifferentiation ,Cell biology ,Insulin receptor ,030104 developmental biology ,030220 oncology & carcinogenesis ,Pyrazines ,biology.protein ,Macrolides ,Proto-Oncogene Proteins c-akt ,Signal Transduction - Abstract
Our previous data indicate that both insulin and IGF-1 signallings dysfunction promotes the dedifferentiation of primary human and mouse white adipocytes. Based on the fact that insulin activates mTOR and inhibits autophagy, and autophagy deficiency can inhibit the differentiation of white adipocytes, we speculate that autophagy may be related to the dedifferentiation of white adipocytes. We investigated the underlying mechanism of autophagy during dedifferentiation of mouse 3T3-L1 adipocytes. After incomplete inhibition of insulin and IGF-1 signallings, 3T3-L1 adipocytes manifest dedifferentiation accompanied with an increase of autophagy level. If induction only of autophagy in the adipocytes, then the cells also occur somewhat dedifferentiation, and with a slight decrease of insulin signal, while its degree was weaker than insulin signal inhibited cells. Notably, after inhibition of the insulin and IGF-1 signallings and simultaneously inducing autophagy, the dedifferentiation of 3T3-L1 adipocytes was the most obvious compared with other groups, and the insulin and IGF-1 signallings decreases was greater than the cells with inhibition only of insulin signalling. If inhibition of both insulin signal and autophagy simultaneously, the dedifferentiation of the adipocytes reveals similar tendencies to the cells that insulin signal was inhibited. No significant dedifferentiation occurs of 3T3-L1 cells if only inhibition of autophagy. Taken all together, in this study, we proved that autophagy is positively related to the dedifferentiation of 3T3-L1 adipocytes and is regulated through the insulin-PI3K-AKT-mTOCR1-autophagy pathway. Autophagy may also has a certain degree of negative feedback affect on the insulin signalling of 3T3-L1 cells. Our work may help to better understand the biological properties of mature adipocytes and may help formulate anti-obesity strategies by regulating insulin and insulin signaling level.
- Published
- 2020
18. Curcumin represses mTORC1 signaling in Caco-2 cells by a two-sided mechanism involving the loss of IRS-1 and activation of AMPK
- Author
-
Régis Moreau and Harleen Kaur
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Curcumin ,MAP Kinase Signaling System ,mTORC1 ,AMP-Activated Protein Kinases ,Mechanistic Target of Rapamycin Complex 1 ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Humans ,Kinase activity ,Protein kinase B ,PI3K/AKT/mTOR pathway ,AMPK ,Cell Biology ,Cell biology ,Enzyme Activation ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Insulin Receptor Substrate Proteins ,Phosphorylation ,biological phenomena, cell phenomena, and immunity ,Caco-2 Cells - Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is a central modulator of inflammation and tumorigenesis in the gastrointestinal tract. Growth factors upregulate mTORC1 via the PI3K/AKT and/or Ras/MAPK signal pathways. Curcumin (CUR), a polyphenol found in turmeric roots (Curcuma longa) can repress mTORC1 kinase activity in colon cancer cell lines; however, key aspects of CUR mechanism of action remain to be elucidated including its primary cellular target. We investigated the molecular effects of physiologically attainable concentration of CUR (20 μM) in the intestinal lumen on mTORC1 signaling in Caco-2 cells. CUR markedly inhibited mTORC1 kinase activity as determined by the decreased phosphorylation of p70S6K (Thr389, -99%, P 0.0001) and S6 (Ser235/236, -92%, P 0.0001). Mechanistically, CUR decreased IRS-1 protein abundance (-80%, P 0.0001) thereby downregulating AKT phosphorylation (Ser473, -94%, P 0.0001) and in turn PRAS40 phosphorylation (Thr246, -99%, P 0.0001) while total PRAS40 abundance was unchanged. The use of proteasome inhibitor MG132 showed that CUR-mediated loss of IRS-1 involved proteasomal degradation. CUR lowered Raptor protein abundance, which combined with PRAS40 hypophosphorylation, suggests CUR repressed mTORC1 activity by inducing compositional changes that hinder the complex assembly. In addition, CUR activated AMPK (Thr172 phosphorylation, P 0.0001), a recognized repressor of mTORC1, and AMPK upstream regulator LKB1. Although cargo adapter protein p62 was decreased by CUR (-49%, P 0.004), CUR did not significantly induce autophagy. Inhibition of AKT/mTORC1 signaling by CUR may have lifted the cross-inhibition onto MAPK signaling, which became induced; p-ERK1/2 (+670%, P 0.0001), p-p38 (+1433%, P 0.0001). By concomitantly targeting IRS-1 and AMPK, CUR's mechanism of mTORC1 inhibition is distinct from that of rapamycin.
- Published
- 2020
19. Increased mTOR and suppressed autophagic flux in the heart of a hypomorphic Pkd1 mouse model of autosomal dominant polycystic kidney disease
- Author
-
Deepak Pokhrel, Andrew Thorburn, Sara J. Holditch, Charles L. Edelstein, Katharina Hopp, Daniel J. Atwood, Dheevena M. Bachu, and Carolyn N. Brown
- Subjects
0301 basic medicine ,Autophagosome ,medicine.medical_specialty ,Autosomal dominant polycystic kidney disease ,Cardiomegaly ,mTORC1 ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Lysosome ,Internal medicine ,medicine ,Autophagy ,Animals ,PI3K/AKT/mTOR pathway ,PKD1 ,TOR Serine-Threonine Kinases ,Cell Biology ,BECN1 ,medicine.disease ,Polycystic Kidney, Autosomal Dominant ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis - Abstract
Cardiac hypertrophy is common in autosomal dominant polycystic kidney disease (ADPKD) patients. We found increased heart weight in Pkd1RC/RC and Pkd2WS25/+ mouse models of ADPKD. As there is a link between increased heart weight and mammalian target of rapamycin (mTOR), the aim of the study was to determine mTOR complex 1 and 2 signaling proteins in the heart in the Pkd1RC/RC mouse model of PKD. In 70 day old Pkd1RC/RC hearts, on immunoblot analysis, there was a large increase in p-AMPKThr172, a known autophagy inducer, and an increase in p-AktSer473 and p-AktThr308, but no increase in other mTORC1/2 proteins (p-S6Ser240/244, p-mTORSer2448). In 150 day old Pkd1RC/RC hearts, there was an increase in mTORC1 (p-S6Ser240/244) and mTOR-related proteins (p-AktThr308, p-GSK3βSer9, p-AMPKThr172). As the mTOR pathway is the master regulator of autophagy, autophagy proteins were measured. There was an increase in p-Beclin-1 (BECN1), an autophagy regulator and activating molecule in Beclin-1-regulated autophagy (AMBRA1), a regulator of Beclin that play a role in autophagosome formation, an early stage of autophagy. There was a defect in the later stage of autophagy, the fusion of the autophagosome with the lysosome, known as autophagic flux, as evidenced by the lack of an increase in LC3-II, a marker of autophagosomes, with the lysosomal inhibitor bafilomycin, in both 70 day old and 150 day old hearts. To determine the role of autophagy in causing increased heart weight, Pkd1RC/RC were treated with 2-deoxyglucose (2-DG) or Tat-Beclin1 peptide, agents known to induce autophagy. 2-DG treatment from 150 to 350 days of age, a time period when increased heart weight developed, did not reduce the increased heart weight. Unexpectedly, Tat-Beclin 1 peptide treatment from 70 to 120 days of age resulted in increased heart weight. In summary, there is suppressed autophagic flux in the heart at an early age in Pkd1RC/RC mice. Increased mTOR signaling in older mice is associated suppressed autophagic flux. There was a large increase in p-AMPKThr172, a known autophagy inducer, in both young and old mice. 2-DG treatment did not impact increased heart weight and Tat-Beclin1 peptide increased heart weight.
- Published
- 2020
20. Targeting AMP-activated protein kinase (AMPK) for treatment of autosomal dominant polycystic kidney disease
- Author
-
York Pei, Xuewen Song, Evelyn Tsakiridis, and Gregory R. Steinberg
- Subjects
0301 basic medicine ,Autosomal dominant polycystic kidney disease ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,03 medical and health sciences ,Cystic kidney disease ,0302 clinical medicine ,AMP-activated protein kinase ,AMP-Activated Protein Kinase Kinases ,medicine ,Polycystic kidney disease ,Animals ,Humans ,Cell Proliferation ,biology ,PKD1 ,urogenital system ,business.industry ,AMPK ,Cell Biology ,medicine.disease ,Polycystic Kidney, Autosomal Dominant ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,business ,Energy Metabolism ,Protein Kinases ,Kidney disease ,Signal Transduction - Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenetic kidney disease worldwide and an important cause of chronic kidney disease. Multiple experimental studies have highlighted the role of increased mammalian target of rapamycin complex 1 (mTORC1) and reduced AMP-activated protein kinase (AMPK) signaling in modulating cyst growth in ADPKD. Notably, mTORC1 and AMPK are two diametrically opposing sensors of energy metabolism which regulate cell growth and proliferation. Although pharmacological mTORC1 inhibition was highly effective in experimental studies of ADPKD, clinical trials of mTORC1 inhibitors showed a lack of efficacy with low-dose treatment and poor tolerability with high-dose treatment. Therapeutic AMPK activation has been shown to attenuate cystic kidney disease severity in Pkd1 mutant animal models by improving mitochondrial biogenesis and reducing tissue inflammation. This review summarizes the current knowledge on the function of AMPK as a regulator of cellular energy metabolism and how AMPK activation by pharmacological and non-pharmacological means can potentially be exploited to treat ADPKD in the clinical settings.
- Published
- 2020
21. Post-translational regulation of mTOR complex 1 in hypoxia and reoxygenation
- Author
-
Tan, Chia Yee and Hagen, Thilo
- Subjects
- *
MTOR protein , *POST-translational modification , *HYPOXEMIA , *OXYGENATION (Chemistry) , *RAPAMYCIN , *CELL proliferation , *GENETIC transcription - Abstract
Abstract: The mechanistic target of rapamycin complex 1 (mTORC1) is a key regulator of cell growth and proliferation in response to various upstream signals. Hypoxia has been shown to exert a strong inhibitory effect on mTORC1 activity. Various mechanisms involving gene transcription have been proposed to mediate the effect of hypoxia on mTORC1 activity. Here we show that oxygen concentrations regulate mTORC1 activity in a highly dynamic manner. The rapid response of mTORC1 to changes in oxygen concentrations was not mediated by the HIF transcription factor or its transcriptional targets, REDD1 and BNIP3. Interestingly, we observed that the rapid response of mTORC1 activity to changes in oxygen concentrations is independent of transcription and new protein synthesis. This suggests a post-translational regulation mTORC1 activity in hypoxia and reoxygenation. We also provide evidence that hypoxia does not regulate mTORC1 via the TSC1/2 or Ragulator pathways but directly at the level of mTORC1. In conclusion, our results suggest that mTORC1 can respond rapidly to changes in oxygen concentrations via a post-translational mechanism that may involve a heme containing protein. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
22. Cell cycle control by anchorage signaling
- Author
-
Okayama, Hiroto
- Subjects
- *
CELL cycle , *CELLULAR signal transduction , *EXTRACELLULAR matrix , *APOPTOSIS , *CELL proliferation , *CELL death , *CELLULAR control mechanisms - Abstract
Abstract: Virtually all the cells constituting solid organs in adult animals require anchorage to the extracellular matrix for their proliferation and survival. When deprived of anchorage, those cells arrest in G1 phase of the cell cycle and die of apoptosis known as anoikis. However, if malignantly transformed, cells no longer require such an anchorage to proliferate and survive, and it is generally thought that the acquirement of this ability underlies the tumorigenic and metastatic capability of malignant cells. Therefore, for the past two decades, great efforts have been devoted to uncovering the nature of the anchorage signal and the mechanism by which this signal controls the G1–S transition in the cell cycle with little progress. However, several critical findings were recently made on anchorage signaling and the control of the cell cycle and cell death by this signaling. This review focuses on the newly emerging understanding and perspective of this highly important cell cycle and cell death regulation. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
23. Proline-rich Akt substrate of 40kDa (PRAS40): A novel downstream target of PI3k/Akt signaling pathway
- Author
-
Wang, Haitao, Zhang, Qishan, Wen, Qiang, Zheng, Yongxin, Philip, Lazarovici, Jiang, Hao, Lin, Jun, and Zheng, WenHua
- Subjects
- *
CELLULAR signal transduction , *PROTEIN kinases , *TYPE 2 diabetes , *PROLINE , *MELANOMA , *RAPAMYCIN , *GROWTH factors , *CARRIER proteins - Abstract
Abstract: Modifications in signaling of the proline-rich Akt substrate of 40-kDa (PRAS40) pathway is implicated in type 2 diabetes and melanoma. PRAS40 is known for its ability to regulate the mammalian target of rapamycin complex 1 (mTORC1) kinase activity, possessing a key regulatory role at the cross point of signal transduction pathways activated by growth factor receptors. Recently it has been found that PRAS40 is regulated by its upstream phosphatidylinositol 3-kinase/Akt (PI3K/Akt) which is activated by many tyrosine kinase receptors growth factors including insulin-like growth factor 1. Also, PRAS40 functions downstream of mTORC1 and upstream from its effectors ribosomal protein S6 kinase 1 (S6K1) and eukaryotic initiation factor 4E binding protein 1 (4E-BP1). Phosphorylation of PRAS40 by Akt and mTORC1 disrupts the binding between mTORC1 and PRAS40, and relieves the inhibitory constraint of PRAS40 on mTORC1 activity. This review summarizes the signaling regulating PRAS40 phosphorylation, as well as the dual function of PRAS40 as substrate and inhibitor of mTORC1 upon growth factor stimulation and under pathophysiological conditions. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
24. cAMP inhibits mammalian target of rapamycin complex-1 and -2 (mTORC1 and 2) by promoting complex dissociation and inhibiting mTOR kinase activity
- Author
-
Xie, Jianling, Ponuwei, Godwin A., Moore, Claire E., Willars, Gary B., Tee, Andrew R., and Herbert, Terence P.
- Subjects
- *
GENE targeting , *ENZYME inhibitors , *RAPAMYCIN , *PROMOTERS (Genetics) , *CELLULAR signal transduction , *PROTEIN metabolism , *GENETIC regulation - Abstract
Abstract: cAMP and mTOR signalling pathways control a number of critical cellular processes including metabolism, protein synthesis, proliferation and cell survival and therefore understanding the signalling events which integrate these two signalling pathways is of particular interest. In this study, we show that the pharmacological elevation of [cAMP]i in mouse embryonic fibroblasts (MEFs) and human embryonic kidney 293 (HEK293) cells inhibits mTORC1 activation via a PKA-dependent mechanism. Although the inhibitory effect of cAMP on mTOR could be mediated by impinging on signalling cascades (i.e. PKB, MAPK and AMPK) that inhibit TSC1/2, an upstream negative regulator of mTORC1, we show that cAMP inhibits mTORC1 in TSC2 knockout (TSC2−/−) MEFs. We also show that cAMP inhibits insulin and amino acid-stimulated mTORC1 activation independently of Rheb, Rag GTPases, TSC2, PKB, MAPK and AMPK, indicating that cAMP may act independently of known regulatory inputs into mTOR. Moreover, we show that the prolonged elevation in [cAMP]i can also inhibit mTORC2. We provide evidence that this cAMP-dependent inhibition of mTORC1/2 is caused by the dissociation of mTORC1 and 2 and a reduction in mTOR catalytic activity, as determined by its auto-phosphorylation on Ser2481. Taken together, these results provide an important insight into how cAMP signals to mTOR and down-regulates its activity, which may lead to the identification of novel drug targets to inhibit mTOR that could be used for the treatment and prevention of human diseases such as cancer. [Copyright &y& Elsevier]
- Published
- 2011
- Full Text
- View/download PDF
25. Phosphorylation of PRAS40 on Thr246 by PKB/AKT facilitates efficient phosphorylation of Ser183 by mTORC1
- Author
-
Nascimento, Emmani B.M., Snel, Marieke, Guigas, Bruno, van der Zon, Gerard C.M., Kriek, Jan, Maassen, J. Antonie, Jazet, Ingrid M., Diamant, Michaela, and Ouwens, D. Margriet
- Subjects
- *
TYPE 2 diabetes , *PHOSPHORYLATION , *PROTEIN kinases , *RAPAMYCIN , *CELLULAR signal transduction , *INSULIN , *LABORATORY rats - Abstract
Abstract: Type 2 diabetes is associated with alterations in protein kinase B (PKB/Akt) and mammalian target of rapamycin complex 1 (mTORC1) signalling. The proline-rich Akt substrate of 40-kDa (PRAS40) is a component of mTORC1, which has a regulatory function at the intersection of the PKB/Akt and mTORC1 signalling pathway. Phosphorylation of PRAS40-Thr246 by PKB/Akt, and PRAS40-Ser183 and PRAS40-Ser221 by mTORC1 results in dissociation from mTORC1, and its binding to 14-3-3 proteins. Although all phosphorylation sites within PRAS40 have been implicated in 14-3-3 binding, substitution of Thr246 by Ala alone is sufficient to abolish 14-3-3 binding under conditions of intact mTORC1 signalling. This suggests that phosphorylation of PRAS40-Thr246 may facilitate efficient phosphorylation of PRAS40 on its mTORC1-dependent sites. In the present study, we investigated the mechanism of PRAS40-Ser183 phosphorylation in response to insulin. Insulin promoted PRAS40-Ser183 phosphorylation after a euglycaemic–hyperinsulinaemic clamp in human skeletal muscle. The insulin-induced PRAS40-Ser183 phosphorylation was further evidenced in vivo in rat skeletal and cardiac muscle, and in vitro in A14 fibroblasts, 3T3L1 adipocytes and L6 myotubes. Inhibition of mTORC1 by rapamycin or amino acid deprivation partially abrogated insulin-mediated PRAS40-Ser183 phosphorylation in cultured cell lines. However, lowering insulin-induced PRAS40-Thr246 phosphorylation using wortmannin or palmitate in cell lines, or by feeding rats a high-fat diet, completely abolished insulin-mediated PRAS40-Ser183 phosphorylation. In addition, replacement of Thr246 by Ala reduced insulin-mediated PRAS40-Ser183 phosphorylation. We conclude that PRAS40-Ser183 is a component of insulin action, and that efficient phosphorylation of PRAS40-Ser183 by mTORC1 requires the phosphorylation of PRAS40-Thr246 by PKB/Akt. [Copyright &y& Elsevier]
- Published
- 2010
- Full Text
- View/download PDF
26. Raptor-rictor axis in TGFβ-induced protein synthesis
- Author
-
Das, Falguni, Ghosh-Choudhury, Nandini, Mahimainathan, Lenin, Venkatesan, Balachandar, Feliers, Denis, Riley, Daniel J., Kasinath, Balakuntalam S., and Choudhury, Goutam Ghosh
- Subjects
- *
PROTEIN synthesis , *HYPERTROPHY , *PHOSPHORYLATION , *RAPAMYCIN - Abstract
Abstract: Transforming growth factor-β (TGFβ) stimulates pathological renal cell hypertrophy for which increased protein synthesis is critical. The mechanism of TGFβ-induced protein synthesis is not known, but PI 3 kinase-dependent Akt kinase activity is necessary. We investigated the contribution of downstream effectors of Akt in TGFβ-stimulated protein synthesis. TGFβ increased inactivating phosphorylation of Akt substrate tuberin in a PI 3 kinase/Akt dependent manner, resulting in activation of mTOR kinase. mTOR activity increased phosphorylation of S6 kinase and the translation repressor 4EBP-1, which were sensitive to inhibition of both PI 3 kinase and Akt. mTOR inhibitor rapamycin and a dominant negative mutant of mTOR suppressed TGFβ-induced phosphorylation of S6 kinase and 4EBP-1. PI 3 kinase/Akt and mTOR regulated dissociation of 4EBP-1 from eIF4E to make the latter available for binding to eIF4G. mTOR and 4EBP-1 modulated TGFβ-induced protein synthesis. mTOR is present in two multi protein complexes, mTORC1 and mTORC2. Raptor and rictor are part of mTORC1 and mTORC2, respectively. shRNA-mediated downregulation of raptor inhibited TGFβ-stimulated mTOR kinase activity, resulting in inhibition of phosphorylation of S6 kinase and 4EBP-1. Raptor shRNA also prevented protein synthesis in response to TGFβ. Downregulation of rictor inhibited serine 473 phosphorylation of Akt without any effect on phosphorylation of its substrate, tuberin. Furthermore, rictor shRNA increased phosphorylation of S6 kinase and 4EBP-1 in TGFβ-independent manner, resulting in increased protein synthesis. Thus mTORC1 function is essential for TGFβ-induced protein synthesis. Our data also provide novel evidence that rictor negatively regulates TORC1 activity to control basal protein synthesis, thus conferring tight control on cellular hypertrophy. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
27. Direct and indirect activation of eukaryotic elongation factor 2 kinase by AMP-activated protein kinase
- Author
-
Manuel Johanns, Louis Hue, Amina Houddane, Gaëtan Herinckx, Mark H. Rider, Christopher G. Proud, Didier Vertommen, S. Pyr dit Ruys, and UCL - PHOS
- Subjects
Elongation Factor 2 Kinase ,AMPK ,0301 basic medicine ,Translation ,Enzyme Activators ,mTORC1 ,AMP-Activated Protein Kinases ,Mechanistic Target of Rapamycin Complex 1 ,Mitogen-activated protein kinase kinase ,EEF2 ,Mice ,Phosphoserine ,03 medical and health sciences ,Compound 991 ,Animals ,Humans ,Phosphorylation ,Protein kinase A ,030102 biochemistry & molecular biology ,biology ,Cyclin-dependent kinase 2 ,Cell Biology ,eEF2K ,Autophagy-related protein 13 ,Cell biology ,Enzyme Activation ,030104 developmental biology ,Biochemistry ,Protein Biosynthesis ,eEF2 ,biology.protein ,Calcium ,cGMP-dependent protein kinase - Abstract
Background Eukaryotic elongation factor 2 (eEF2) kinase (eEF2K) is a key regulator of protein synthesis in mammalian cells. It phosphorylates and inhibits eEF2, the translation factor necessary for peptide translocation during the elongation phase of protein synthesis. When cellular energy demand outweighs energy supply, AMP-activated protein kinase (AMPK) and eEF2K become activated, leading to eEF2 phosphorylation, which reduces the rate of protein synthesis, a process that consumes a large proportion of cellular energy under optimal conditions. Aim The goal of the present study was to elucidate the mechanisms by which AMPK activation leads to increased eEF2 phosphorylation to decrease protein synthesis. Methods Using genetically modified mouse embryo fibroblasts (MEFs), effects of treatments with commonly used AMPK activators to increase eEF2 phosphorylation were compared with that of the novel compound 991. Bacterially expressed recombinant eEF2K was phosphorylated in vitro by recombinant activated AMPK for phosphorylation site-identification by mass spectrometry followed by site-directed mutagenesis of the identified sites to alanine residues to study effects on the kinetic properties of eEF2K. Wild-type eEF2K and a Ser491/Ser492 mutant were retrovirally re-introduced in eEF2K-deficient MEFs and effects of 991 treatment on eEF2 phosphorylation and protein synthesis rates were studied in these cells. Results & conclusions AMPK activation leads to increased eEF2 phosphorylation in MEFs mainly by direct activation of eEF2K and partly by inhibition of mammalian target of rapamycin complex 1 (mTORC1) signaling. Treatment of MEFs with AMPK activators can also lead to eEF2K activation independently of AMPK probably via a rise in intracellular Ca 2+ . AMPK activates eEF2K by multi-site phosphorylation and the newly identified Ser491/Ser492 is important for activation, leading to mTOR-independent inhibition of protein synthesis. Our study provides new insights into the control of eEF2K by AMPK, with implications for linking metabolic stress to decreased protein synthesis to conserve energy reserves, a pathway that is of major importance in cancer cell survival.
- Published
- 2017
28. T-cadherin promotes autophagy and survival in vascular smooth muscle cells through MEK1/2/Erk1/2 axis activation
- Author
-
Thérèse J. Resink, Klaus-Peter Lesch, Paul Erne, Olga Rivero, Boris Dasen, Maria Philippova, Dennis Pfaff, Agne Frismantiene, Emmanouil Kyriakakis, RS: MHeNs - R3 - Neuroscience, and Psychiatrie & Neuropsychologie
- Subjects
Transcriptional Activation ,0301 basic medicine ,MAP Kinase Signaling System ,MAP Kinase Kinase 2 ,Myocytes, Smooth Muscle ,CROSS-TALK ,MAP Kinase Kinase 1 ,Apoptosis ,mTORC1 ,Signal transduction ,ADHESION ,Biology ,Muscle, Smooth, Vascular ,Unfolded protein response ,SIGNALING PATHWAYS ,Mice ,03 medical and health sciences ,Downregulation and upregulation ,Vascular smooth muscle cells ,Autophagy ,Animals ,Humans ,STIMULATES AUTOPHAGY ,GLIOMA-CELLS ,OXIDATIVE STRESS ,Aorta ,PI3K/AKT/mTOR pathway ,Tissue homeostasis ,Flavonoids ,Cadherin ,DEATH ,EPITHELIAL-CELLS ,Cell Biology ,T-cadherin ,Cadherins ,Endoplasmic Reticulum Stress ,ENDOTHELIAL-CELLS ,Rats ,3. Good health ,Cell biology ,030104 developmental biology ,Gene Expression Regulation ,cardiovascular system ,Reactive Oxygen Species ,STRESS-INDUCED APOPTOSIS - Abstract
Autophagy is an evolutionary conserved intracellular catabolic process of vital importance to cell and tissue homeostasis. Autophagy is implicated in the pathogenesis of atherosclerosis but participating cells, molecular mechanisms and functional outcomes have not been fully elucidated. T-cadherin, an atypical glycosylpho-sphatidylinositol-anchored member of the cadherin superfamily of adhesion molecules, is upregulated on smooth muscle cells (SMCs)(1) in atherosclerotic lesions. Here, using rat and murine aortic SMCs as experimental models, we surveyed the ability of T-cadherin to regulate autophagy in SMCs during serum-starvation stress. Ectopic upregulation of T-cadherin in SMCs resulted in augmented autophagy characterized by increased autophagic flux, LC3-II abundance and autophagosome formation. Analysis of signal transduction pathway effectors and use of specific pharmacological inhibitors demonstrated that T-cadherin-associated enhancement of the autophagic response to serum-deprivation was dependent on MEK1/2/Erk1/2 activation and independent of PI3K/Akt/mTORC1, reactive oxygen species or endoplasmic reticulum stress. T-cadherin upregulation on SMCs conferred a survival advantage during prolonged serum-starvation which was sensitive to inhibition of MEK1/2/Erkl/2 by PD98059 or U0126 and to blockade of autophagy by chloroquine. Loss of T-cadherin expression in SMCs diminished autophagy responsiveness and compromised survival under conditions of serum starvation. Overall our findings have identified T-cadherin as a novel positive regulator of autophagy and survival in SMCs.
- Published
- 2017
29. PAQR3 augments amino acid deprivation-induced autophagy by inhibiting mTORC1 signaling
- Author
-
Xue You, Yi Pan, Feifan Guo, Meiqin Huang, Yan Chen, and Lin Wang
- Subjects
0301 basic medicine ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,BAG3 ,Cell Line ,Mice ,03 medical and health sciences ,Autophagy ,Animals ,Humans ,Amino Acids ,PI3K/AKT/mTOR pathway ,Cell Size ,chemistry.chemical_classification ,mTOR Associated Protein, LST8 Homolog ,Cell growth ,RPTOR ,Autophagosomes ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,Regulatory-Associated Protein of mTOR ,Cell Biology ,Cell biology ,Amino acid ,030104 developmental biology ,Biochemistry ,chemistry ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Protein Binding ,Signal Transduction - Abstract
Amino acids are the key activators of the mTOR complex 1 (mTORC1, mainly composed of mTOR, Raptor and mLST8) required for cell growth and proliferation. On the other hand, deprivation of amino acids induces autophagy via inhibition of mTORC1 signaling. We report here that amino acid-induced mTORC1 activity and amino acid deprivation-induced autophagy are regulated by PAQR3, a newly found tumor suppressor. At the cellular level, PAQR3 negatively regulates amino acid-induced activation of mTORC1. The N-terminal end of PAQR3 interacts with the WD domains of Raptor and mLST8 directly. PAQR3 reduces the interaction of mTOR with Raptor and mLST8, thus disrupts formation of intact mTORC1 complex. PAQR3 modulates leucine-induced alteration in cell size. In addition, PAQR3 knockdown reduces amino acid deprivation-induced autophagy. The inhibitory effect of PAQR3 knockdown on autophagy is abrogated by rapamycin treatment, indicating that PAQR3 modulates autophagy via its regulation on mTORC1 signaling. In conclusion, our finding reveals a new mode of regulation of mTORC1 signaling and autophagy by PAQR3 in response to alterations of amino acids.
- Published
- 2017
30. Nudix-type motif 2 contributes to cancer proliferation through the regulation of Rag GTPase-mediated mammalian target of rapamycin complex 1 localization
- Author
-
Pann-Ghill Suh, Mangeun Park, Hyeona Jeon, Sang Hoon Ha, Sung Ho Ryu, Yeonho Chang, Dongoh Kwak, and Ohman Kwon
- Subjects
0301 basic medicine ,Regulator ,GTPase ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Resting Phase, Cell Cycle ,03 medical and health sciences ,Downregulation and upregulation ,Cell Line, Tumor ,Neoplasms ,Humans ,Insulin ,Gene silencing ,Amino Acids ,Tumor Stem Cell Assay ,Cell Proliferation ,Monomeric GTP-Binding Proteins ,Regulation of gene expression ,Cell growth ,G1 Phase ,Cell Cycle Checkpoints ,Cell Biology ,Phosphoric Monoester Hydrolases ,Cell biology ,030104 developmental biology ,Gene Knockdown Techniques ,Protein Multimerization ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Lysosomes ,Protein Binding ,Signal Transduction - Abstract
Lysosomal localization of mammalian target of rapamycin complex 1 (mTORC1) is a critical step for activation of the molecule. Rag GTPases are essential for this translocation. Here, we demonstrate that Nudix-type motif 2 (NUDT2) is a novel positive regulator of mTORC1 activation. Activation of mTORC1 is impaired in NUDT2-silenced cells. Mechanistically, NUDT2 binds to Rag GTPase and controls mTORC1 translocation to the lysosomal membrane. Furthermore, NUDT2-dependent mTORC1 regulation is critical for proliferation of breast cancer cells, as NUDT2-silenced cells arrest in G0/G1 phases. Taken together, these results show that NUDT2 is a novel complex formation enhancing factor regulating mTORC1-Rag GTPase signaling that is crucial for cell growth control.
- Published
- 2017
31. Novel compounds for the modulation of mTOR and autophagy to treat neurodegenerative diseases
- Author
-
David Heras-Sandoval, José Pedraza-Chaverri, and Jazmin M. Pérez-Rojas
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Kinase ,TOR Serine-Threonine Kinases ,Neurodegeneration ,Autophagy ,Neurodegenerative Diseases ,Cell Biology ,mTORC1 ,Biology ,medicine.disease ,mTORC2 ,Models, Biological ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,medicine ,Animals ,Humans ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Signal Transduction - Abstract
Most neurodegenerative diseases show a disruption of autophagic function and display abnormal accumulation of toxic protein aggregates that promotes cellular stress and death. Therefore, induction of autophagy has been proposed as a reasonable strategy to help neurons clear abnormal protein aggregates and survive. The kinase mammalian target of rapamycin (mTOR) is a major regulator of the autophagic process and is regulated by starvation, growth factors, and cellular stressors. The phosphoinositide 3-kinase (PI3K)/ protein kinase B (Akt) pathway, which promotes cellular survival, is the main modulator upstream of mTOR, and alterations in this pathway are common in neurodegenerative diseases, e.g. Alzheimer’s disease (AD) and Parkinson’s disease (PD). In the present work we revised mammalian target of rapamycin complex 1 (mTORC1) pathway and mTORC2 as a complementary an important element in mTORC1 signaling. In addition, we revised the extracellular signal regulated kinase (ERK) pathway, which has become relevant in the regulation of the autophagic process and cellular survival through mTORC2 signaling. Finally, we summarize novel compounds that promote autophagy and neuronal protection in the last five years.
- Published
- 2019
32. High glucose-stimulated enhancer of zeste homolog-2 (EZH2) forces suppression of deptor to cause glomerular mesangial cell pathology.
- Author
-
Das, Falguni, Bera, Amit, Ghosh-Choudhury, Nandini, Sataranatarajan, Kavitha, Kamat, Amrita, Kasinath, Balakuntalam S., and Choudhury, Goutam Ghosh
- Subjects
- *
KIDNEY cortex , *DIABETIC nephropathies , *PATHOLOGY , *HYPERTROPHY , *GLUCOSE - Abstract
Function of mTORC1 and mTORC2 has emerged as a driver of mesangial cell pathologies in diabetic nephropathy. The mechanism of mTOR activation is poorly understood in this disease. Deptor is a constitutive subunit and a negative regulator of both mTOR complexes. Mechanistic investigation in mesangial cells revealed that high glucose decreased the expression of deptor concomitant with increased mTORC1 and mTORC2 activities, induction of hypertrophy and, expression of fibronectin and PAI-1. shRNAs against deptor mimicked these pathologic outcomes of high glucose. Conversely, overexpression of deptor significantly inhibited all effects of high glucose. To determine the mechanism of deptor suppression, we found that high glucose significantly increased the expression of EZH2, resulting in lysine-27 tri-methylation of histone H3 (H3K27Me3). Employing approaches including pharmacological inhibition, shRNA-mediated downregulation and overexpression of EZH2, we found that EZH2 regulates high glucose-induced deptor suppression along with activation of mTOR, mesangial cell hypertrophy and fibronectin/PAI-1 expression. Moreover, expression of hyperactive mTORC1 reversed shEZH2-mediated inhibition of hypertrophy and expression of fibronectin and PAI-1 by high glucose. Finally, in renal cortex of diabetic mice, we found that enhanced expression of EZH2 is associated with decreased deptor levels and increased mTOR activity and, expression of fibronectin and PAI-1. Together, our findings provide a novel mechanism for mTOR activation via EZH2 to induce mesangial cell hypertrophy and matrix expansion during early progression of diabetic nephropathy. These results suggest a strategy for leveraging the intrinsic effect of deptor to suppress mTOR activity via reducing EZH2 as a novel therapy for diabetic nephropathy. [Display omitted] • High glucose inhibits deptor to cause mesangial cell hypertrophy and matrix expansion. • High glucose increases EZH2 to downregulate deptor. • EZH2 regulates mTORC1 and mTORC2. • In diabetic kidney, enhanced EZH2 and mTOR activity are associated with pathology. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
33. Negative regulation of the FOXO3a transcription factor by mTORC2 induces a pro-survival response following exposure to ultraviolet-B irradiation
- Author
-
Robert P. Feehan and Lisa M. Shantz
- Subjects
0301 basic medicine ,Cell Survival ,Ultraviolet Rays ,Apoptosis ,P70-S6 Kinase 1 ,Mechanistic Target of Rapamycin Complex 2 ,IκB kinase ,mTORC1 ,Biology ,mTORC2 ,Article ,Cell Line ,03 medical and health sciences ,Humans ,Phosphorylation ,Transcription factor ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Nucleus ,Mitogen-Activated Protein Kinase Kinases ,integumentary system ,TOR Serine-Threonine Kinases ,Forkhead Box Protein O3 ,Cell Biology ,Cell biology ,030104 developmental biology ,Gene Knockdown Techniques ,Multiprotein Complexes ,Cancer research ,Signal transduction ,Gene Deletion ,Signal Transduction - Abstract
Exposure to ultraviolet-B (UVB) irradiation, the principal cause of non-melanoma skin cancer (NMSC), activates both the rapamycin-sensitive mammalian target of rapamycin complex 1 (mTORC1) and the rapamycin-resistant mTORC2. We have previously reported that UVB-induced keratinocyte survival is dependent on mTORC2, though the specific mechanism is not well understood. FOXO3a is an important transcription factor involved in regulating cell survival. The activity of FOXO3a is reduced as a result of protein kinase B (AKT/PKB) activation, which is downstream of mTORC2; however, the specific function of FOXO3a during UVB-induced apoptosis is unclear. In this study, we establish that in cells with wild-type mTORC2 activity, FOXO3a is quickly phosphorylated in response to UVB and sequestered in the cytoplasm. In contrast, loss of mTORC2 causes FOXO3a to be localized to the nucleus and sensitizes cells to UVB-induced apoptosis. Furthermore, this sensitization is rescued by knockdown of FOXO3a. Taken together, these studies provide strong evidence that inhibition of mTORC2 enhances UVB-induced apoptosis in a FOXO3a-dependent manner, and suggest that FOXO3a activation by mTORC2 inhibitors may be a valuable chemopreventive target in NMSC.
- Published
- 2016
34. Bridges between mitochondrial oxidative stress, ER stress and mTOR signaling in pancreatic β cells
- Author
-
Zhang Jingjing, Yang Xin, and Jing Wang
- Subjects
0301 basic medicine ,medicine.medical_specialty ,mTORC1 ,Oxidative phosphorylation ,Biology ,medicine.disease_cause ,03 medical and health sciences ,Insulin-Secreting Cells ,Internal medicine ,medicine ,Animals ,Humans ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell growth ,TOR Serine-Threonine Kinases ,Endoplasmic reticulum ,Cell Biology ,Endoplasmic Reticulum Stress ,Mitochondria ,Cell biology ,Oxidative Stress ,030104 developmental biology ,Endocrinology ,Unfolded protein response ,Oxidative stress ,Signal Transduction - Abstract
Pancreatic β cell dysfunction, i.e., failure to provide insulin in concentrations sufficient to control blood sugar, is central to the etiology of all types of diabetes. Current evidence implicates mitochondrial oxidative stress and endoplasmic reticulum (ER) stress in pancreatic β cell loss and impaired insulin secretion. Oxidative and ER stress are interconnected so that misfolded proteins induce reactive oxygen species (ROS) production; likewise, oxidative stress disturbs the ER redox state thereby disrupting correct disulfide bond formation and proper protein folding. mTOR signaling regulates many metabolic processes including protein synthesis, cell growth, survival and proliferation. Oxidative stress inhibits mTORC1, which is considered an important suppressor of mitochondrial oxidative stress in β cells, and ultimately, controls cell survival. The interplay between ER stress and mTORC1 is complicated, since the unfolded protein response (UPR) activation can occur upstream or downstream of mTORC1. Persistent activation of mTORC1 initiates protein synthesis and UPR activation, while in the later phase induces ER stress. Chronic activation of ER stress inhibits Akt/mTORC1 pathway, while under particular settings, acute activation of UPR activates Akt-mTOR signaling. Thus, modulating mitochondrial oxidative stress and ER stress via mTOR signaling may be an approach that will effectively suppress obesity- or glucolipotoxicity-induced metabolic disorders such as insulin resistance and type 2 diabetes mellitus (T2DM). In this review, we focus on the regulations between mTOR signaling and mitochondrial oxidative or ER stress in pancreatic β cells.
- Published
- 2016
35. Amino acid-dependent NPRL2 interaction with Raptor determines mTOR Complex 1 activation
- Author
-
Sang Su Kwak, Jeung-Hoon Lee, Jin Woo Kim, Kyung Hwa Kang, Gary G. Meadows, Cheol O. Joe, SeoEun Lee, Seyun Kim, and Boohyeong Byun
- Subjects
0301 basic medicine ,GTPase-activating protein ,GTP' ,Mutant ,mTORC1 ,Plasma protein binding ,GTPase ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Models, Biological ,03 medical and health sciences ,Humans ,Amino Acids ,Adaptor Proteins, Signal Transducing ,Monomeric GTP-Binding Proteins ,chemistry.chemical_classification ,TOR Serine-Threonine Kinases ,Tumor Suppressor Proteins ,GTPase-Activating Proteins ,HEK 293 cells ,Regulatory-Associated Protein of mTOR ,Cell Biology ,Amino acid ,HEK293 Cells ,030104 developmental biology ,chemistry ,Biochemistry ,Multiprotein Complexes ,biological phenomena, cell phenomena, and immunity ,Protein Binding ,Signal Transduction - Abstract
We assign a new function to a tumor suppressor NPRL2 that activates the mTOR complex 1 (mTORC1) activity. The positive regulation of mTORC1 activity by NPRL2 is mediated through NPRL2 interaction with Raptor. While NPRL2 interacts with Rag GTPases, RagD in particular, to interfere with mTORC1 activity in amino acid scarcity, NPRL2 interacts with Raptor in amino acid sufficiency to activate mTORC1. A reciprocal relationship exists between NPRL2 binding to Rag GTPases and Raptor. NPRL2 majorly locates in the lysosomal membranes and has a higher binding affinity to the dominant negative mutant heterodimer of RagA(GDP)/RagD(GTP) that inactivates mTORC1. However, the binding affinity of NPRL2 with Raptor is much less pronounced in cells expressing the dominant negative mutant heterodimer of RagA(GDP)/RagD(GTP) than in cells expressing the dominant positive mutant heterodimer, RagA(GTP)/RagD(GDP). The positive effect of NPRL2 on TORC1 pathway was also evidenced in Drosophila animal model. Here, we propose a 'seesaw' model in which the interactive behavior of NPRL2 with Raptor determines mTORC1 activation by amino acid signaling in animal cells.
- Published
- 2016
36. Iron depletion suppresses mTORC1-directed signalling in intestinal Caco-2 cells via induction of REDD1
- Author
-
Ailsa Watson, Christopher Lipina, Peter M. Taylor, Harry J McArdle, and Harinder S. Hundal
- Subjects
0301 basic medicine ,4E-BP1, eIF4E-binding protein 1 ,4E-BP1 ,mTORC1 ,Intestinal mucosa ,PP2A, protein phosphatase 2A ,Protein Phosphatase 2 ,S6, ribosomal protein S6 ,Amino Acids ,Intestinal Mucosa ,DFO, deferoxamine ,biology ,TOR Serine-Threonine Kinases ,REDD1, regulated in DNA damage and development 1 ,Ribosomal Protein S6 Kinases, 70-kDa ,PI3K, phosphatidylinositol 3-kinase ,S6K1 ,Iron Deficiencies ,Cell biology ,PP2A ,HIF, hypoxia-inducible factor ,IRS, insulin receptor substrate ,DMT-1, divalent metal transporter-1 ,biological phenomena, cell phenomena, and immunity ,Signal Transduction ,Rheb ,Down-Regulation ,Transferrin receptor ,P70-S6 Kinase 1 ,mTOR, mammalian target of rapamycin ,TfR, transferrin receptor ,Deferoxamine ,Mechanistic Target of Rapamycin Complex 1 ,Iron Chelating Agents ,Article ,03 medical and health sciences ,Humans ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Cell growth ,S6K1, ribosomal protein S6 kinase 1 ,Akt ,DMT1 ,Cell Biology ,TSC2 ,030104 developmental biology ,Multiprotein Complexes ,Protein Biosynthesis ,biology.protein ,Caco-2 Cells ,Proto-Oncogene Proteins c-akt ,Transcription Factors - Abstract
Iron is an indispensable micronutrient that regulates many aspects of cell function, including growth and proliferation. These processes are critically dependent upon signalling via the mammalian or mechanistic target of rapamycin complex 1 (mTORC1). Herein, we test whether iron depletion induced by cell incubation with the iron chelator, deferoxamine (DFO), mediates its effects on cell growth through mTORC1-directed signalling and protein synthesis. We have used Caco-2 cells, a well-established in vitro model of human intestinal epithelia. Iron depletion increased expression of iron-regulated proteins (TfR, transferrin receptor and DMT1, divalent metal transporter, as predicted, but it also promoted a marked reduction in growth and proliferation of Caco-2 cells. This was strongly associated with suppressed mTORC1 signalling, as judged by reduced phosphorylation of mTOR substrates, S6K1 and 4E-BP1, and diminished protein synthesis. The reduction in mTORC1 signalling was tightly coupled with increased expression and accumulation of REDD1 (regulated in DNA damage and development 1) and reduced phosphorylation of Akt and TSC2. The increase in REDD1 abundance was rapidly reversed upon iron repletion of cells but was also attenuated by inhibitors of gene transcription, protein phosphatase 2A (PP2A) and by REDD1 siRNA — strategies that also antagonised the loss in mTORC1 signalling associated with iron depletion. Our findings implicate REDD1 and PP2A as crucial regulators of mTORC1 activity in iron-depleted cells and indicate that their modulation may help mitigate atrophy of the intestinal mucosa that may occur in response to iron deficiency., Highlights • Cellular iron (Fe) depletion dramatically reduces growth of intestinal Caco-2 cells. • mTORC1-directed signalling and protein synthesis are reduced in Fe-depleted cells. • Fe deficiency induces expression and gain of REDD1 in a PP2A-dependent manner. • PP2A inhibition blocks REDD1 gain and restores mTORC1 activity in Fe-depleted cells.
- Published
- 2016
37. The multi-kinase inhibitor lenvatinib interacts with the HDAC inhibitor entinostat to kill liver cancer cells
- Author
-
Jane L. Roberts, Andrew Poklepovic, Paul Dent, and Laurence Booth
- Subjects
0301 basic medicine ,Sorafenib ,Cell Survival ,Pyridines ,ATG5 ,Antineoplastic Agents ,mTORC1 ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Humans ,Protein Kinase Inhibitors ,Entinostat ,Kinase ,Phenylurea Compounds ,Liver Neoplasms ,Drug Synergism ,Cell Biology ,Histone Deacetylase Inhibitors ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Benzamides ,Quinolines ,Cancer research ,Phosphorylation ,Histone deacetylase ,Reactive Oxygen Species ,Lenvatinib ,medicine.drug - Abstract
Prior studies from our group have combined the multi-kinase inhibitor sorafenib with HDAC inhibitors in GI tumor cells that resulted in the trials NCT02349867 and NCT01075113. The multi-kinase inhibitor lenvatinib, for the treatment of liver cancer, has fewer negative sequelae than sorafenib. We determined the mechanisms by which lenvatinib interacted with the HDAC inhibitor entinostat to kill hepatoma cells. Lenvatinib and entinostat interacted in an additive to greater-than-additive fashion to kill liver cancer cells. The drugs inactivated mTORC1 and mTORC2 and interacted to further increase the phosphorylation of ATM, ATG13 and eIF2α. Elevated eIF2α phosphorylation was responsible for reduced MCL-1 and BCL-XL expression and for increased Beclin1 and ATG5 expression. Over-expression of BCL-XL or knock down of Beclin1 or ATG5, significantly reduced killing. The drugs synergized to elevate ROS production; activation of ATM was ROS-dependent. ATM activation was required for enhanced phosphorylation of γH2AX, eIF2α and ATG13 S318. The drug combination reduced histone deacetylase protein expression which required autophagy. Knock down of HDACs1/2/3 prevented the lenvatinib and entinostat combination from regulating PD-L1 and MHCA expression. Collectively, our data demonstrate that lenvatinib and entinostat interact to kill liver cancer cells via ROS-dependent activation of ATM and inactivation of eIF2α, resulting in greater levels of toxic autophagosome formation and reduced expression of protective mitochondrial proteins.
- Published
- 2020
38. Metabolic reprogramming and the role of mitochondria in polycystic kidney disease
- Author
-
Laura Cassina, Christine Podrini, and Alessandra Boletta
- Subjects
0301 basic medicine ,Autosomal dominant polycystic kidney disease ,mTORC1 ,Oxidative phosphorylation ,Mitochondrion ,Biology ,Models, Biological ,Epigenesis, Genetic ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Polycystic kidney disease ,Animals ,Humans ,Beta oxidation ,Polycystic Kidney Diseases ,Cell Biology ,Lipid Metabolism ,medicine.disease ,Mitochondria ,Cell biology ,Glutamine ,030104 developmental biology ,Anaerobic glycolysis ,030220 oncology & carcinogenesis ,Signal Transduction - Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a slowly progressive disease characterized by the relentless growth of renal cysts throughout the life of affected individuals. Early evidence suggested that the epithelia lining the cysts share neoplastic features, leading to the definition of PKD as a "neoplasm in disguise". Recent work from our and other laboratories has identified a profound metabolic reprogramming in PKD, similar to the one reported in cancer and consistent with the reported increased proliferation. Multiple lines of evidence suggest that aerobic glycolysis (a Warburg-like effect) is present in the disease, along with other metabolic dysfunctions such as an increase in the pentose phosphate pathway, in glutamine anaplerosis and fatty acid biosynthesis, while fatty acid oxidation and oxidative phosphorylation (OXPHOS) are decreased. In addition to glutamine, other amino acid-related pathways appear altered, including asparagine and arginine. The precise origin of the metabolic alterations is not entirely clear, but two hypotheses can be formulated, not mutually exclusive. First, the polycystins have been recently shown to regulate directly mitochondrial function and structure either by regulating Ca2+ uptake in mitochondria at the Mitochondria Associated Membranes (MAMs) of the Endoplasmic Reticulum, or by a direct translocation of a small fragment of the protein into the matrix of mitochondria. One alternative possibility is that metabolic and mitochondrial dysfunctions in ADPKD are secondary to the de-regulation of proliferation, driven by the multiple signaling pathways identified in the disease, which include mTORC1 and AMPK among the most relevant. While the precise mechanisms underlying these novel alterations identified in ADPKD will need further investigation, it is evident that they offer a great opportunity for novel interventions in the disease.
- Published
- 2020
39. Autophagy participants in the dedifferentiation of mouse 3T3-L1 adipocytes triggered by hypofunction of insulin signaling.
- Author
-
Pan, Jie, Kothan, Suchart, Liu, Laihao, Moe, Aye Thidar Moe, Dong, Liwei, Sun, Yanan, and Yang, Yiyi
- Subjects
- *
AUTOPHAGY , *FAT cells , *INSULIN , *INSULIN receptors , *MICE - Abstract
Our previous data indicate that both insulin and IGF-1 signallings dysfunction promotes the dedifferentiation of primary human and mouse white adipocytes. Based on the fact that insulin activates mTOR and inhibits autophagy, and autophagy deficiency can inhibit the differentiation of white adipocytes, we speculate that autophagy may be related to the dedifferentiation of white adipocytes. We investigated the underlying mechanism of autophagy during dedifferentiation of mouse 3T3-L1 adipocytes. After incomplete inhibition of insulin and IGF-1 signallings, 3T3-L1 adipocytes manifest dedifferentiation accompanied with an increase of autophagy level. If induction only of autophagy in the adipocytes, then the cells also occur somewhat dedifferentiation, and with a slight decrease of insulin signal, while its degree was weaker than insulin signal inhibited cells. Notably, after inhibition of the insulin and IGF-1 signallings and simultaneously inducing autophagy, the dedifferentiation of 3T3-L1 adipocytes was the most obvious compared with other groups, and the insulin and IGF-1 signallings decreases was greater than the cells with inhibition only of insulin signalling. If inhibition of both insulin signal and autophagy simultaneously, the dedifferentiation of the adipocytes reveals similar tendencies to the cells that insulin signal was inhibited. No significant dedifferentiation occurs of 3T3-L1 cells if only inhibition of autophagy. Taken all together, in this study, we proved that autophagy is positively related to the dedifferentiation of 3T3-L1 adipocytes and is regulated through the insulin-PI3K-AKT-mTOCR1-autophagy pathway. Autophagy may also has a certain degree of negative feedback affect on the insulin signalling of 3T3-L1 cells. Our work may help to better understand the biological properties of mature adipocytes and may help formulate anti-obesity strategies by regulating insulin and insulin signaling level. • Insulin-PI3K-AKT-mTORC1 signal regulates 3T3-L1 adipocyte dedifferentiation; • Induced autophagy alone could slightly promote the dedifferentiation of 3T3-L1 adipocytes; • Inhibiting insulin signal and promoting autophagy can accelerate the dedifferentiation of the cells. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
40. Growth-factor dependent expression of the translationally controlled tumour protein TCTP is regulated through the PI3-K/Akt/mTORC1 signalling pathway
- Author
-
Valentina Iadevaia, Bianca Knoch, Ulrich-Axel Bommer, Jiezhong Chen, Christopher G. Proud, and Martin Engel
- Subjects
Time Factors ,Uterine Cervical Neoplasms ,Cell Cycle Proteins ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Transfection ,Tuberous Sclerosis Complex 2 Protein ,Biomarkers, Tumor ,Humans ,RNA, Messenger ,Protein Kinase Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,biology ,Kinase ,TOR Serine-Threonine Kinases ,Tumor Suppressor Proteins ,EIF4E ,Tumor Protein, Translationally-Controlled 1 ,Translation (biology) ,Cell Biology ,Phosphoproteins ,Cell biology ,Gene Expression Regulation, Neoplastic ,Eukaryotic Initiation Factor-4E ,HEK293 Cells ,Multiprotein Complexes ,Polyribosomes ,Colonic Neoplasms ,Cancer cell ,biology.protein ,Female ,Translationally controlled tumour protein ,Phosphatidylinositol 3-Kinase ,biological phenomena, cell phenomena, and immunity ,HT29 Cells ,Proto-Oncogene Proteins c-akt ,HeLa Cells ,Signal Transduction - Abstract
Translationally controlled tumour protein TCTP (gene symbol: TPT1) is a highly-conserved, cyto-protective protein implicated in many physiological and disease processes, in particular cancer, where it is associated with poor patient outcomes. To understand the mechanisms underlying the accumulation of high TCTP levels in cancer cells, we studied the signalling pathways that control translation of TCTP mRNA, which contains a 5?-terminal oligopyrimidine tract (5?-TOP). In HT29 colon cancer cells and in HeLa cells, serum increases the expression of TCTP two- and four-fold, respectively, and this is inhibited by rapamycin or mTOR kinase inhibitors. Polysome profiling and mRNA quantification indicate that these effects occur at the level of mRNA translation. Blocking this pathway upstream of mTOR complex 1 (mTORC1) by inhibiting Akt also prevented increases in TCTP levels in both HeLa and HT29 colon cancer cells, whereas knockout of TSC2, a negative regulator of mTORC1, led to derepression of TCTP synthesis under serum starvation. Overexpression of eIF4E enhanced the polysomal association of the TCTP mRNA, although it did not protect its translation from inhibition by rapamycin. Conversely, expression of a constitutively-active mutant of the eIF4E inhibitor 4E-BP1, which is normally inactivated by mTORC1, inhibited of TCTP mRNA translation in HEK293 cells. Our results demonstrate that TCTP mRNA translation is regulated by signalling through the PI3-K/Akt/mTORC1 pathway. This explains why TCTP levels are frequently increased in cancers, since mTORC1 signalling is hyperactive in ~ 80% of tumours.
- Published
- 2015
41. High glucose enhances microRNA-26a to activate mTORC1 for mesangial cell hypertrophy and matrix protein expression
- Author
-
Falguni Das, Balakuntalam S. Kasinath, Amit Bera, Nandini Ghosh-Choudhury, Goutam Ghosh Choudhury, and Nirmalya Dey
- Subjects
Glomerular Mesangial Cell ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Article ,Collagen Type I ,Cell Line ,Tuberous Sclerosis Complex 2 Protein ,Animals ,Humans ,PTEN ,Phosphorylation ,3' Untranslated Regions ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Base Sequence ,biology ,Mesangial cell ,Kinase ,Ribosomal Protein S6 Kinases ,TOR Serine-Threonine Kinases ,Tumor Suppressor Proteins ,Intracellular Signaling Peptides and Proteins ,PTEN Phosphohydrolase ,Cell Biology ,Oligonucleotides, Antisense ,Phosphoproteins ,Fibronectins ,Rats ,Cell biology ,Fibronectin ,MicroRNAs ,Glucose ,Multiprotein Complexes ,Mesangial Cells ,biology.protein ,Cancer research ,Carrier Proteins ,Sequence Alignment - Abstract
High glucose milieu inhibits PTEN expression to activate Akt kinase and induces glomerular mesangial cell hypertrophy and matrix protein expression in diabetic nephropathy. Specific mechanism by which high glucose inhibits PTEN expression is not clear. We found that high glucose increased the expression of the microRNA-26a (miR-26a) in mesangial cells. Using a sensor plasmid with 3′UTR-driven luciferase, we showed PTEN as a target of miR-26a in response to high glucose. Overexpression of miR-26a reduced the PTEN protein levels resulting in increased Akt kinase activity similar to high glucose treatment. In contrast, anti-miR-26a reversed high glucose-induced suppression of PTEN with concomitant inhibition of Akt kinase activity. Akt-mediated phosphorylation of tuberin and PRAS40 regulates mTORC1, which is necessary for mesangial cell hypertrophy and matrix protein expression. Inhibition of high glucose-induced miR-26a blocked phosphorylation of tuberin and PRAS40, which lead to suppression of phosphorylation of S6 kinase and 4EBP-1, two substrates of mTORC1. Furthermore, we show that expression of miR-26a induced mesangial cell hypertrophy and increased fibronectin and collagen I (α2) expression similar to that observed with the cells incubated with high glucose. Anti-miR-26a inhibited these phenomena in response to high glucose. Together our results provide the first evidence for the involvement of miR-26a in high glucose-induced mesangial cell hypertrophy and matrix protein expression. These data indicate the potential therapeutic utility of anti-miR-26a for the complications of diabetic kidney disease.
- Published
- 2015
42. The roles of subcellularly located EGFR in autophagy
- Author
-
Hongsen Li, Hongming Pan, Weidong Han, Jiansheng Xie, and Liangkun You
- Subjects
0301 basic medicine ,Cytoplasm ,Endosome ,Autophagy-Related Proteins ,mTORC1 ,Endosomes ,BAG3 ,03 medical and health sciences ,0302 clinical medicine ,LAPTM4B ,Sodium-Glucose Transporter 1 ,Neoplasms ,Autophagy ,Humans ,PI3K/AKT/mTOR pathway ,Chemistry ,Cell Membrane ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,Cell biology ,Mitochondria ,ErbB Receptors ,030104 developmental biology ,Glucose ,030220 oncology & carcinogenesis ,Cancer research ,Beclin-1 ,Tyrosine kinase ,MAP1LC3A ,Signal Transduction - Abstract
The epidermal growth factor receptor (EGFR) is a well-studied receptor-tyrosine kinase that serves vital roles in regulation of organ development and cancer progression. EGFR not only exists on the plasma membrane, but also widely expressed in the nucleus, endosomes, and mitochondria. Most recently, several lines of evidences indicated that autophagy is regulated by EGFR in kinase-active and -independent manners. In this review, we summarized recent advances in our understanding of the functions of different subcellularly located EGFR on autophagy. Specifically, plasma membrane- and cytoplasm-located EGFR (pcEGFR) acts as a tyrosine kinase to regulate autophagy via the PI3K/AKT1/mTOR, RAS/MAPK1/3, and STAT3 signaling pathways. The kinase-independent function of pcEGFR inhibits autophagy by maintaining SLC5A1-regulated intracellular glucose level. Endosome-located EGFR phosphorylates and inhibits Beclin1 to suppress autophagy, while kinase-independent endosome-located EGFR releases Beclin1 from the Rubicon-Beclin1 complex to increase autophagy. Additionally, the nuclear EGFR activates PRKDC/PNPase/MYC signaling to inhibit autophagy. Although the role of mitochondria-located EGFR in autophagy is largely unexplored, the production of ATP and reactive oxygen species mediated by mitochondrial dynamics is most likely to influence autophagy.
- Published
- 2017
43. A role for Raptor phosphorylation in the mechanical activation of mTOR signaling
- Author
-
John Frey, Craig A. Goodman, Troy A. Hornberger, and Brittany L. Jacobs
- Subjects
Phosphopeptides ,Molecular Sequence Data ,P70-S6 Kinase 1 ,mTORC1 ,In Vitro Techniques ,Biology ,mTORC2 ,Article ,Mass Spectrometry ,Mice ,medicine ,Animals ,Amino Acid Sequence ,Phosphorylation ,Muscle, Skeletal ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Sirolimus ,TOR Serine-Threonine Kinases ,RPTOR ,Ribosomal Protein S6 Kinases, 70-kDa ,Regulatory-Associated Protein of mTOR ,Cell Biology ,Phosphoproteins ,Molecular biology ,Anti-Bacterial Agents ,Cell biology ,Mice, Inbred C57BL ,Amino Acid Substitution ,Stress, Mechanical ,Signal transduction ,Protein Binding ,Signal Transduction ,medicine.drug - Abstract
The activation of mTOR signaling is necessary for mechanically-induced changes in skeletal muscle mass, but the mechanisms that regulate the mechanical activation of mTOR signaling remain poorly defined. In this study, we set out to determine if changes in the phosphorylation of Raptor contribute to the mechanical activation of mTOR. To accomplish this goal, mouse skeletal muscles were subjected to mechanical stimulation via a bout of eccentric contractions (EC). Using mass spectrometry and Western blot analysis, we found that ECs induced an increase in Raptor S696, T706, and S863 phosphorylation, and this effect was not inhibited by rapamycin. This observation suggested that changes in Raptor phosphorylation might be an upstream event in the pathway through which mechanical stimuli activate mTOR. To test this, we employed a phospho-defective mutant of Raptor (S696A/T706A/S863A) and found that the EC-induced activation of mTOR signaling was significantly blunted in muscles expressing this mutant. Furthermore, mutation of the three phosphorylation sites altered the interactions of Raptor with PRAS40 and p70(S6k), and it also prevented the EC-induced dissociation of Raptor from p70(S6k). Combined, these results suggest that changes in the phosphorylation of Raptor play an important role in the pathway through which mechanical stimuli activate mTOR signaling.
- Published
- 2014
44. Activation of AMPK protects against hydrogen peroxide-induced osteoblast apoptosis through autophagy induction and NADPH maintenance: New implications for osteonecrosis treatment?
- Author
-
Lun-qing Zhu, Chang She, Xiao-dong Wang, Qi Rong Dong, and Yun-fang Zhen
- Subjects
Programmed cell death ,Apoptosis ,mTORC1 ,AMP-Activated Protein Kinases ,Mice ,chemistry.chemical_compound ,AMP-activated protein kinase ,Autophagy ,Animals ,Humans ,Protein Kinase Inhibitors ,Cell Line, Transformed ,chemistry.chemical_classification ,Reactive oxygen species ,Osteoblasts ,biology ,Osteonecrosis ,AMPK ,Hydrogen Peroxide ,Cell Biology ,Cell biology ,Enzyme Activation ,Oxidative Stress ,HEK293 Cells ,chemistry ,Cytoprotection ,biology.protein ,NADP ,Nicotinamide adenine dinucleotide phosphate - Abstract
Elevated hydrogen peroxide (H2O2) causes osteoblast dysfunction and apoptosis, serving as an important contributor to the development of osteonecrosis. Here we aimed to understand the role of AMP-activated protein kinase (AMPK) in the process. We observed a high level of AMPK activation in surgery isolated patients' osteonecrosis tissues. In cultured osteoblastoma MG63 cells, H2O2 stimulation induced significant AMPK activation, oxidative stress, cell death and apoptosis. Inhibition of AMPK by its inhibitor (compound C) or by shRNA-mediated knockdown dramatically enhanced H2O2-induced MG63 cell apoptosis, while over-expression of AMPK in HEK-293 cells alleviated H2O2-induced cell damage. These results confirmed that H2O2-activated AMPK is pro-cell survival. We observed that H2O2 induced protective autophagy in MG63 cells, and AMPK-dependent Ulk1 activation and mTORC1 (mTOR complex 1) inactivation might involve autophagy activation. Further, AMPK activation inhibited H2O2-induced oxidative stress, probably through inhibiting NADPH (nicotinamide adenine dinucleotide phosphate) depletion, since more NADPH depletion and oxidative stress were induced by H2O2 in AMPK deficient MG63 cells. Finally, we observed a significant AMPK activation in H2O2-treated primary cultured and transformed (MC3T3-E1) osteoblasts, and AMPK inhibitor compound C enhanced death by H2O2 in these cells. Based on these results, we concluded that H2O2-induced AMPK activation is pro-survival and anti-apoptosis in osteoblasts. Autophagy induction and NADPH maintenance are involved in AMPK-mediated pro-survival effects. AMPK might represent a novel molecular target for osteonecrosis treatment.
- Published
- 2014
45. MicroRNA-451 regulates AMPK/mTORC1 signaling and fascin1 expression in HT-29 colorectal cancer
- Author
-
Xiao-Yang Wu, Min-Bin Chen, Jun-Yi Han, Wei Shen, Mu-Xin Wei, Pei-Hua Lu, Chen Li, and Jian Wang
- Subjects
P70-S6 Kinase 1 ,mTORC1 ,AMP-Activated Protein Kinases ,Mechanistic Target of Rapamycin Complex 1 ,Models, Biological ,Cell Movement ,microRNA ,Humans ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Gene knockdown ,Chemistry ,TOR Serine-Threonine Kinases ,Microfilament Proteins ,AMPK ,Cell migration ,Cell Biology ,Ribonucleotides ,Aminoimidazole Carboxamide ,Cell biology ,Enzyme Activation ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,HEK293 Cells ,Gene Knockdown Techniques ,Multiprotein Complexes ,Cancer cell ,Cancer research ,RNA Interference ,biological phenomena, cell phenomena, and immunity ,Carrier Proteins ,Colorectal Neoplasms ,HT29 Cells ,Signal Transduction - Abstract
The earlier studies have shown that Fascin1 (FSCN1), the actin bundling protein, is over-expressed in colorectal cancers, and is associated with cancer cell progression. Here, we aimed to understand the molecular mechanisms regulating FSCN1 expression by focusing on mammalian target of rapamycin (mTOR) signaling and its regulator microRNA-451. We found that microRNA-451 was over-expressed in multiple colorectal cancer tissues, and its expression was correlated with mTOR complex 1 (mTORC1) activity and FSCN1 expression. In cultured colorectal cancer HT-29 cells, knockdown of FSCN1 by RNAi inhibited cell migration and proliferation. Activation of mTORC1 was required for FSCN1 expression, HT-29 cell migration and proliferation, as RAD001 and rapamycin, two mTORC1 inhibitors, suppressed FSCN1 expression, HT-29 cell migration and proliferation. Meanwhile, forced activation of AMP-activated protein kinase (AMPK), the negative regulator of mTORC1, by its activators or by the genetic mutation, inhibited mTORC1 activation, FSCN1 expression, cell migration and proliferation. In HT-29 cells, we found that over-expression of microRNA-451 inhibited AMPK activation, causing mTORC1 over-activation and FSCN1 up-regulation, cells were with high migration ability and proliferation rate. Significantly, these effects by microRNA-451 were largely inhibited by mTORC1 inhibitors or the AMPK activator AICAR. On the other hand, knockdown of miRNA-451 by the treatment of HT-29 cells with miRNA-451 antagomir inhibited mTORC1 activation and FSCN1 expression. The proliferation and migration of HT-29 cells after miRNA-45 knockdown were also inhibited. Our results suggested that the over-expressed microRNA-451 in colon cancer cells might inhibit AMPK to activate mTORC1, which mediates FSCN1 expression and cancer cell progression.
- Published
- 2014
46. Inhibition of 4E-BP1 phosphorylation promotes tubular cell escaping from G2/M arrest and ameliorates kidney fibrosis
- Author
-
Mingjie Wang, Jianzhong Li, Chunsun Dai, Xian Xue, Yan Liang, Jiafa Ren, Xiaoli Sun, Wei Wei, and Yuan Gui
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Aristolochic acid ,Mitosis ,Apoptosis ,Cell Cycle Proteins ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Kidney ,Nephropathy ,Histones ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Phosphorylation ,Fibroblast ,Adaptor Proteins, Signal Transducing ,urogenital system ,Cell growth ,Cell Cycle ,Gene Expression Regulation, Developmental ,Epithelial Cells ,Cell Biology ,Transfection ,medicine.disease ,Fibrosis ,Cell biology ,Kidney Tubules ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Aristolochic Acids ,Cell Division ,Signal Transduction - Abstract
Upon occurrence of kidney injury, tubular cells arrested in G2/M stage may promote interstitial fibroblast activation and kidney fibrosis through producing large amounts of pro-fibrotic cytokines. MTORC1 signaling is essential for controlling cell growth, however, the role and mechanisms for mTORC1 in regulating tubular cell cycle progression during kidney fibrosis are not clear. Here we reported that p-S6 abundance was increased at 15 min, reached peak at 1 h and declined from 3 h to 24 h, while the abundance of p-4E-BP1 and p-Histone H3 was increased from 15 min to 24 h in tubular epithelial cells at the similar pattern after serum stimulation. The phosphorylation of 4E-BP1 was prohibited in NRK-52E cells by the transfection of 4E-BP1 plasmid with four phospho-sites mutation (4E-BP1A4). 4E-BP1A4 transfection led to less G2/M cell arrest as well as the production of pro-fibrotic cytokine and extracellular matrix in NRK-52E cells. In addition, aristolochic acid (AA)-induced tubular cell G2/M arrest induced by treatment was also largely attenuated in NRK-52E cells transfected with 4E-BP1A4. In mouse kidneys with UUO nephropathy, p-4E-BP1 abundance was markedly elevated in the mitotic tubular cells. Therefore, these data indicates that suppressing 4E-BP1 phosphorylation may inhibit tubular cell G2/M-arrest and kidney fibrosis.
- Published
- 2019
47. Targeting AMP-activated protein kinase (AMPK) for treatment of autosomal dominant polycystic kidney disease.
- Author
-
Song, Xuewen, Tsakiridis, Evelyn, Steinberg, Gregory R., and Pei, York
- Subjects
- *
POLYCYSTIC kidney disease , *MTOR protein , *PROTEIN kinases , *CYSTIC kidney disease , *ENERGY metabolism - Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenetic kidney disease worldwide and an important cause of chronic kidney disease. Multiple experimental studies have highlighted the role of increased mammalian target of rapamycin complex 1 (mTORC1) and reduced AMP-activated protein kinase (AMPK) signaling in modulating cyst growth in ADPKD. Notably, mTORC1 and AMPK are two diametrically opposing sensors of energy metabolism which regulate cell growth and proliferation. Although pharmacological mTORC1 inhibition was highly effective in experimental studies of ADPKD, clinical trials of mTORC1 inhibitors showed a lack of efficacy with low-dose treatment and poor tolerability with high-dose treatment. Therapeutic AMPK activation has been shown to attenuate cystic kidney disease severity in Pkd1 mutant animal models by improving mitochondrial biogenesis and reducing tissue inflammation. This review summarizes the current knowledge on the function of AMPK as a regulator of cellular energy metabolism and how AMPK activation by pharmacological and non-pharmacological means can potentially be exploited to treat ADPKD in the clinical settings. • Metabolic reprogramming has emerged as an important pathogenic mechanism in ADPKD. • AMPK is a key sensor of energy metabolism that regulates cell growth and proliferation. • Increased aerobic glycolysis, reduced mitochondrial biogenesis and AMPK activity are key pathogenic features in ADPKD. • Therapeutic AMPK activation may slow ADPKD by improving energy metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
48. The rationale of targeting mammalian target of rapamycin for ischemic stroke
- Author
-
Hui-Hua Li, Zhao Zhong Chong, and Qingqiang Yao
- Subjects
Brain Infarction ,Sirolimus ,Neurogenesis ,TOR Serine-Threonine Kinases ,RPTOR ,Brain ,Neovascularization, Physiologic ,P70-S6 Kinase 1 ,Cell Biology ,mTORC1 ,Biology ,mTORC2 ,Brain Ischemia ,Cell biology ,Cancer research ,biology.protein ,Animals ,Humans ,MLST8 ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Signal Transduction ,RHEB - Abstract
Given the current limitation of therapeutic approach for ischemic stroke, a leading cause of disability and mortality in the developed countries, to develop new therapeutic strategies for this devastating disease is urgently necessary. As a serine/threonine kinase, mammalian target of rapamycin (mTOR) activation can mediate broad biological activities that include protein synthesis, cytoskeleton organization, and cell survival. mTOR functions through mTORC1 and mTORC2 complexes and their multiple downstream substrates, such as eukaryotic initiation factor 4E-binding protein 1, p70 ribosomal S6 kinase, sterol regulatory element-binding protein 1, hypoxia inducible factor-1, and signal transducer and activator transcription 3, Yin Ying 1, Akt, protein kinase c-alpha, Rho GTPase, serum-and gucocorticoid-induced protein kinase 1, etc. Specially, the role of mTOR in the central nervous system has been attracting considerable attention. Based on the ability of mTOR to prevent neuronal apoptosis, inhibit autophagic cell death, promote neurogenesis, and improve angiogenesis, mTOR may acquire the capability of limiting the ischemic neuronal death and promoting the neurological recovery. Consequently, to regulate the activity of mTOR holds a potential as a novel therapeutic strategy for ischemic stroke.
- Published
- 2013
49. Post-translational regulation of mTOR complex 1 in hypoxia and reoxygenation
- Author
-
Thilo Hagen and Chia Yee Tan
- Subjects
Regulator ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Tuberous Sclerosis Complex 1 Protein ,Cell Line ,Transcription (biology) ,Proto-Oncogene Proteins ,Tuberous Sclerosis Complex 2 Protein ,Humans ,Post-translational regulation ,RNA, Small Interfering ,Transcription factor ,Cell growth ,TOR Serine-Threonine Kinases ,Tumor Suppressor Proteins ,HEK 293 cells ,Membrane Proteins ,Cell Biology ,HCT116 Cells ,Hypoxia-Inducible Factor 1, alpha Subunit ,Cell Hypoxia ,Cell biology ,HEK293 Cells ,Biochemistry ,Multiprotein Complexes ,MCF-7 Cells ,RNA Interference ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Signal Transduction ,Transcription Factors - Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is a key regulator of cell growth and proliferation in response to various upstream signals. Hypoxia has been shown to exert a strong inhibitory effect on mTORC1 activity. Various mechanisms involving gene transcription have been proposed to mediate the effect of hypoxia on mTORC1 activity. Here we show that oxygen concentrations regulate mTORC1 activity in a highly dynamic manner. The rapid response of mTORC1 to changes in oxygen concentrations was not mediated by the HIF transcription factor or its transcriptional targets, REDD1 and BNIP3. Interestingly, we observed that the rapid response of mTORC1 activity to changes in oxygen concentrations is independent of transcription and new protein synthesis. This suggests a post-translational regulation mTORC1 activity in hypoxia and reoxygenation. We also provide evidence that hypoxia does not regulate mTORC1 via the TSC1/2 or Ragulator pathways but directly at the level of mTORC1. In conclusion, our results suggest that mTORC1 can respond rapidly to changes in oxygen concentrations via a post-translational mechanism that may involve a heme containing protein.
- Published
- 2013
50. ERK1/2-dependent activation of mTOR/mTORC1/p70S6K regulates thrombin-induced RPE cell proliferation
- Author
-
Ana María López-Colomé, Irene Lee-Rivera, Alejandro Parrales, and Edith López
- Subjects
Mechanistic Target of Rapamycin Complex 2 ,Retinal Pigment Epithelium ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Protein Serine-Threonine Kinases ,Biology ,mTORC2 ,Cell Line ,Phosphatidylinositol 3-Kinases ,Cyclin D1 ,Thrombin ,medicine ,Animals ,Phosphorylation ,RNA, Small Interfering ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,Cell growth ,TOR Serine-Threonine Kinases ,Intracellular Signaling Peptides and Proteins ,Pyruvate Dehydrogenase Acetyl-Transferring Kinase ,Ribosomal Protein S6 Kinases, 70-kDa ,Regulatory-Associated Protein of mTOR ,Cell Biology ,Phosphoproteins ,eye diseases ,Rats ,Cell biology ,Rapamycin-Insensitive Companion of mTOR Protein ,Multiprotein Complexes ,biology.protein ,Cancer research ,RNA Interference ,sense organs ,Carrier Proteins ,Proto-Oncogene Proteins c-akt ,Platelet-derived growth factor receptor ,Signal Transduction ,medicine.drug - Abstract
Epithelial-mesenchymal transition (EMT), proliferation and migration of RPE cells characterize the development of proliferative vitreoretinopathy (PVR) and other fibro-proliferative eye diseases leading to blindness. A common event in these pathologies is the alteration of the BRB which allows the interaction of RPE cells with thrombin, a pro-inflammatory protease contained in serum. Thrombin promotion of cytoskeletal reorganization, proliferation, and migration has been reported in different cell types, although the molecular mechanisms involved in these processes remain poorly understood. Our previous work demonstrated that thrombin promotes RPE cell proliferation, cytoskeletal remodeling and migration, hallmark processes in the development of PVR. Thrombin induction of RPE cell proliferation requires PI3K, PDK1, and Akt/PKB (Akt) signaling leading to cyclin D1 gene expression. Since Akt functions as an upstream activator of mechanistic target of rapamycin complex 1 (mTORC1) and is also a downstream target for mTORC2, the aim of this work was to determine whether mTOR is involved in thrombin-induced RPE cell proliferation by regulating cyclin D1 expression in immortalized rat RPE-J cell line. Results demonstrate that thrombin-induced cyclin D1 expression and cell proliferation require Akt-independent phosphorylation/activation of mTOR at Ser 2448 mediated by PI3K/PKC-ζ/ERK1/2 signaling, concomitant to Akt-dependent activation of p70S6K carried by mTORC1.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.