Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 178
Filter
Add more filters

Publication year range
1.
Cell ; 186(23): 5068-5083.e23, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37804830

ABSTRACT

Metabolic reprogramming is a hallmark of cancer. However, mechanisms underlying metabolic reprogramming and how altered metabolism in turn enhances tumorigenicity are poorly understood. Here, we report that arginine levels are elevated in murine and patient hepatocellular carcinoma (HCC), despite reduced expression of arginine synthesis genes. Tumor cells accumulate high levels of arginine due to increased uptake and reduced arginine-to-polyamine conversion. Importantly, the high levels of arginine promote tumor formation via further metabolic reprogramming, including changes in glucose, amino acid, nucleotide, and fatty acid metabolism. Mechanistically, arginine binds RNA-binding motif protein 39 (RBM39) to control expression of metabolic genes. RBM39-mediated upregulation of asparagine synthesis leads to enhanced arginine uptake, creating a positive feedback loop to sustain high arginine levels and oncogenic metabolism. Thus, arginine is a second messenger-like molecule that reprograms metabolism to promote tumor growth.


Subject(s)
Arginine , Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Humans , Mice , Arginine/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Lipid Metabolism , Liver Neoplasms/metabolism
2.
Cell ; 185(11): 1814-1836, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35580586

ABSTRACT

The target of rapamycin (TOR), discovered 30 years ago, is a highly conserved serine/threonine protein kinase that plays a central role in regulating cell growth and metabolism. It is activated by nutrients, growth factors, and cellular energy. TOR forms two structurally and functionally distinct complexes, TORC1 and TORC2. TOR signaling activates cell growth, defined as an increase in biomass, by stimulating anabolic metabolism while inhibiting catabolic processes. With emphasis on mammalian TOR (mTOR), we comprehensively reviewed the literature and identified all reported direct substrates. In the context of recent structural information, we discuss how mTORC1 and mTORC2, despite having a common catalytic subunit, phosphorylate distinct substrates. We conclude that the two complexes recruit different substrates to phosphorylate a common, minimal motif.


Subject(s)
Multiprotein Complexes , TOR Serine-Threonine Kinases , Animals , Mammals/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Multiprotein Complexes/metabolism , Phosphorylation , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
4.
Mol Cell ; 82(22): 4246-4261.e11, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36400009

ABSTRACT

Acetyl-coenzyme A (acetyl-CoA) plays an important role in metabolism, gene expression, signaling, and other cellular processes via transfer of its acetyl group to proteins and metabolites. However, the synthesis and usage of acetyl-CoA in disease states such as cancer are poorly characterized. Here, we investigated global acetyl-CoA synthesis and protein acetylation in a mouse model and patient samples of hepatocellular carcinoma (HCC). Unexpectedly, we found that acetyl-CoA levels are decreased in HCC due to transcriptional downregulation of all six acetyl-CoA biosynthesis pathways. This led to hypo-acetylation specifically of non-histone proteins, including many enzymes in metabolic pathways. Importantly, repression of acetyl-CoA synthesis promoted oncogenic dedifferentiation and proliferation. Mechanistically, acetyl-CoA synthesis was repressed by the transcription factors TEAD2 and E2A, previously unknown to control acetyl-CoA synthesis. Knockdown of TEAD2 and E2A restored acetyl-CoA levels and inhibited tumor growth. Our findings causally link transcriptional reprogramming of acetyl-CoA metabolism, dedifferentiation, and cancer.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mice , Animals , Acetyl Coenzyme A/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Histones/metabolism , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Carcinogenesis/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
5.
Cell ; 156(4): 627-8, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-24529368

ABSTRACT

mTORC1 is activated primarily on the lysosome. Menon et al. and Demetriades et al. show that mTORC1 deactivation on the lysosome is determined by recruitment of its negative regulator, the tumor suppressor complex TSC1-TSC2. These reports highlight the importance of subcellular localization in the regulation of mTORC1.


Subject(s)
Amino Acids/metabolism , Insulin/metabolism , Lysosomes/metabolism , Multiprotein Complexes/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Humans , Mechanistic Target of Rapamycin Complex 1 , Tuberous Sclerosis Complex 2 Protein
6.
Cell ; 157(7): 1515-26, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24949965

ABSTRACT

The phenomenon of aging is an intrinsic feature of life. Accordingly, the possibility to manipulate it has fascinated humans likely since time immemorial. Recent evidence is shaping a picture where low caloric regimes and exercise may improve healthy senescence, and several pharmacological strategies have been suggested to counteract aging. Surprisingly, the most effective interventions proposed to date converge on only a few cellular processes, in particular nutrient signaling, mitochondrial efficiency, proteostasis, and autophagy. Here, we critically examine drugs and behaviors to which life- or healthspan-extending properties have been ascribed and discuss the underlying molecular mechanisms.


Subject(s)
Aging/drug effects , Animals , Antioxidants/administration & dosage , Autophagy , Diet , Exercise , Humans , Longevity/drug effects , Signal Transduction
7.
Mol Cell ; 81(10): 2057-2058, 2021 05 20.
Article in English | MEDLINE | ID: mdl-34019785

ABSTRACT

Cho et al. (2021) and Villa et al. (2021) demonstrate that mTORC1 stimulates m6A mRNA methylation via WTAP expression and SAM synthesis. Increased mRNA methylation in turn promotes cell growth by enhancing mRNA degradation or translation.


Subject(s)
RNA Stability , Writing , Mechanistic Target of Rapamycin Complex 1/genetics , Methylation , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Mol Cell ; 81(11): 2403-2416.e5, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33852892

ABSTRACT

The activation of cap-dependent translation in eukaryotes requires multisite, hierarchical phosphorylation of 4E-BP by the 1 MDa kinase mammalian target of rapamycin complex 1 (mTORC1). To resolve the mechanism of this hierarchical phosphorylation at the atomic level, we monitored by NMR spectroscopy the interaction of intrinsically disordered 4E binding protein isoform 1 (4E-BP1) with the mTORC1 subunit regulatory-associated protein of mTOR (Raptor). The N-terminal RAIP motif and the C-terminal TOR signaling (TOS) motif of 4E-BP1 bind separate sites in Raptor, resulting in avidity-based tethering of 4E-BP1. This tethering orients the flexible central region of 4E-BP1 toward the mTORC1 kinase site for phosphorylation. The structural constraints imposed by the two tethering interactions, combined with phosphorylation-induced conformational switching of 4E-BP1, explain the hierarchy of 4E-BP1 phosphorylation by mTORC1. Furthermore, we demonstrate that mTORC1 recognizes both free and eIF4E-bound 4E-BP1, allowing rapid phosphorylation of the entire 4E-BP1 pool and efficient activation of translation. Finally, our findings provide a mechanistic explanation for the differential rapamycin sensitivity of the 4E-BP1 phosphorylation sites.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Cell Cycle Proteins/chemistry , Eukaryotic Initiation Factor-4E/chemistry , Mechanistic Target of Rapamycin Complex 1/chemistry , Regulatory-Associated Protein of mTOR/chemistry , TOR Serine-Threonine Kinases/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chaetomium/chemistry , Chaetomium/genetics , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Kinetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Models, Molecular , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism , Signal Transduction , Structural Homology, Protein , Substrate Specificity , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
9.
Cell ; 154(4): 859-74, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23953116

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) controls growth and survival in response to metabolic cues. Oxidative stress affects mTORC1 via inhibitory and stimulatory inputs. Whereas downregulation of TSC1-TSC2 activates mTORC1 upon oxidative stress, the molecular mechanism of mTORC1 inhibition remains unknown. Here, we identify astrin as an essential negative mTORC1 regulator in the cellular stress response. Upon stress, astrin inhibits mTORC1 association and recruits the mTORC1 component raptor to stress granules (SGs), thereby preventing mTORC1-hyperactivation-induced apoptosis. In turn, balanced mTORC1 activity enables expression of stress factors. By identifying astrin as a direct molecular link between mTORC1, SG assembly, and the stress response, we establish a unifying model of mTORC1 inhibition and activation upon stress. Importantly, we show that in cancer cells, apoptosis suppression during stress depends on astrin. Being frequently upregulated in tumors, astrin is a potential clinically relevant target to sensitize tumors to apoptosis.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cytoplasmic Granules/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Oxidative Stress , Regulatory-Associated Protein of mTOR
10.
EMBO J ; 42(5): e112344, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36691768

ABSTRACT

Target of rapamycin complex 1 (TORC1) promotes biogenesis and inhibits the degradation of ribosomes in response to nutrient availability. To ensure a basal supply of ribosomes, cells are known to preserve a small pool of dormant ribosomes under nutrient-limited conditions. However, the regulation of these dormant ribosomes is poorly characterized. Here, we show that upon inhibition of yeast TORC1 by rapamycin or nitrogen starvation, the ribosome preservation factor Stm1 mediates the formation of nontranslating, dormant 80S ribosomes. Furthermore, Stm1-bound 80S ribosomes are protected from proteasomal degradation. Upon nutrient replenishment, TORC1 directly phosphorylates and inhibits Stm1 to reactivate translation. Finally, we find that SERBP1, a mammalian ortholog of Stm1, is likewise required for the formation of dormant 80S ribosomes upon mTORC1 inhibition in mammalian cells. These data suggest that TORC1 regulates ribosomal dormancy in an evolutionarily conserved manner by directly targeting a ribosome preservation factor.


Subject(s)
Saccharomyces cerevisiae Proteins , Animals , Mammals , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
11.
Immunity ; 48(3): 542-555.e6, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29523440

ABSTRACT

Glycolysis is linked to the rapid response of memory CD8+ T cells, but the molecular and subcellular structural elements enabling enhanced glucose metabolism in nascent activated memory CD8+ T cells are unknown. We found that rapid activation of protein kinase B (PKB or AKT) by mammalian target of rapamycin complex 2 (mTORC2) led to inhibition of glycogen synthase kinase 3ß (GSK3ß) at mitochondria-endoplasmic reticulum (ER) junctions. This enabled recruitment of hexokinase I (HK-I) to the voltage-dependent anion channel (VDAC) on mitochondria. Binding of HK-I to VDAC promoted respiration by facilitating metabolite flux into mitochondria. Glucose tracing pinpointed pyruvate oxidation in mitochondria, which was the metabolic requirement for rapid generation of interferon-γ (IFN-γ) in memory T cells. Subcellular organization of mTORC2-AKT-GSK3ß at mitochondria-ER contact sites, promoting HK-I recruitment to VDAC, thus underpins the metabolic reprogramming needed for memory CD8+ T cells to rapidly acquire effector function.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Endoplasmic Reticulum/metabolism , Energy Metabolism , Immunologic Memory , Mitochondria/metabolism , Signal Transduction , Cell Respiration , Endoplasmic Reticulum/ultrastructure , Glycogen Synthase Kinase 3 beta/metabolism , Glycolysis , Intracellular Membranes/metabolism , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 2/metabolism , Mitochondria/ultrastructure , Models, Biological , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/deficiency
12.
Proc Natl Acad Sci U S A ; 121(1): e2315242121, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38154064

ABSTRACT

High-risk neuroblastoma (NB) is a significant clinical challenge. MYCN and Anaplastic Lymphoma Kinase (ALK), which are often involved in high-risk NB, lead to increased replication stress in cancer cells, suggesting therapeutic strategies. We previously identified an ATR (ataxia telangiectasia and Rad3-related)/ALK inhibitor (ATRi/ALKi) combination as such a strategy in two independent genetically modified mouse NB models. Here, we identify an underlying molecular mechanism, in which ALK signaling leads to phosphorylation of ATR and CHK1, supporting an effective DNA damage response. The importance of ALK inhibition is supported by mouse data, in which ATRi monotreatment resulted in a robust initial response, but subsequent relapse, in contrast to a 14-d ALKi/ATRi combination treatment that resulted in a robust and sustained response. Finally, we show that the remarkable response to the 14-d combined ATR/ALK inhibition protocol reflects a robust differentiation response, reprogramming tumor cells to a neuronal/Schwann cell lineage identity. Our results identify an ability of ATR inhibition to promote NB differentiation and underscore the importance of further exploring combined ALK/ATR inhibition in NB, particularly in high-risk patient groups with oncogene-induced replication stress.


Subject(s)
Neuroblastoma , Receptor Protein-Tyrosine Kinases , Humans , Mice , Animals , Anaplastic Lymphoma Kinase/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Cell Proliferation , Cell Line, Tumor , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Neuroblastoma/pathology , DNA Repair , DNA Damage , Ataxia Telangiectasia Mutated Proteins/genetics
13.
Nat Rev Mol Cell Biol ; 15(3): 155-62, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24556838

ABSTRACT

More than 20 years after its discovery, our understanding of target of rapamycin (TOR) signalling continues to grow. Recent global 'omics' studies have revealed physiological roles of mammalian TOR (mTOR) in protein, nucleotide and lipid synthesis. Furthermore, emerging evidence provides new insight into the control of mTOR by other pathways such as Hippo, WNT and Notch signalling. Together, this progress has expanded the list of downstream effectors and upstream regulators of mTOR signalling.


Subject(s)
Models, Biological , Signal Transduction , TOR Serine-Threonine Kinases/physiology , Animals , Antibiotics, Antineoplastic/pharmacology , Humans , Signal Transduction/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism
14.
Cell ; 144(5): 757-68, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21376236

ABSTRACT

The target of rapamycin (TOR) is a highly conserved protein kinase and a central controller of growth. Mammalian TOR complex 2 (mTORC2) regulates AGC kinase family members and is implicated in various disorders, including cancer and diabetes. Here, we investigated the upstream regulation of mTORC2. A genetic screen in yeast and subsequent studies in mammalian cells revealed that ribosomes, but not protein synthesis, are required for mTORC2 signaling. Active mTORC2 was physically associated with the ribosome, and insulin-stimulated PI3K signaling promoted mTORC2-ribosome binding, suggesting that ribosomes activate mTORC2 directly. Findings with melanoma and colon cancer cells suggest that mTORC2-ribosome association is important in oncogenic PI3K signaling. Thus, TORC2-ribosome interaction is a likely conserved mechanism of TORC2 activation that is physiologically relevant in both normal and cancer cells. As ribosome content determines growth capacity of a cell, this mechanism of TORC2 regulation ensures that TORC2 is active only in growing cells.


Subject(s)
Multiprotein Complexes/metabolism , Ribosomes/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , HeLa Cells , Humans , Insulin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Saccharomyces cerevisiae/metabolism
15.
Cell ; 136(3): 399-400, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19203575

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1), which promotes cell growth, is regulated by specific nutrients such as the amino acid leucine. In this issue, Nicklin et al. (2009) describe a mechanism by which glutamine facilitates the uptake of leucine, leading to mTORC1 activation.


Subject(s)
Glutamine/metabolism , Protein Kinases/metabolism , Animals , Autophagy , Humans , Large Neutral Amino Acid-Transporter 1/metabolism , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Proteins , TOR Serine-Threonine Kinases , Transcription Factors/metabolism
16.
Nature ; 555(7698): 678-682, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29562234

ABSTRACT

Histidine phosphorylation, the so-called hidden phosphoproteome, is a poorly characterized post-translational modification of proteins. Here we describe a role of histidine phosphorylation in tumorigenesis. Proteomic analysis of 12 tumours from an mTOR-driven hepatocellular carcinoma mouse model revealed that NME1 and NME2, the only known mammalian histidine kinases, were upregulated. Conversely, expression of the putative histidine phosphatase LHPP was downregulated specifically in the tumours. We demonstrate that LHPP is indeed a protein histidine phosphatase. Consistent with these observations, global histidine phosphorylation was significantly upregulated in the liver tumours. Sustained, hepatic expression of LHPP in the hepatocellular carcinoma mouse model reduced tumour burden and prevented the loss of liver function. Finally, in patients with hepatocellular carcinoma, low expression of LHPP correlated with increased tumour severity and reduced overall survival. Thus, LHPP is a protein histidine phosphatase and tumour suppressor, suggesting that deregulated histidine phosphorylation is oncogenic.


Subject(s)
Histidine/metabolism , Inorganic Pyrophosphatase/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Animals , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Humans , Inorganic Pyrophosphatase/deficiency , Inorganic Pyrophosphatase/genetics , Male , Mice , Phosphorylation , Proteomics , Survival Analysis , TOR Serine-Threonine Kinases/metabolism , Tumor Burden , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
17.
Am J Physiol Endocrinol Metab ; 324(6): E589-E598, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37166264

ABSTRACT

Mammalian target of rapamycin complex 2 (mTORC2) is a protein kinase complex that plays an important role in energy homeostasis. Loss of adipose mTORC2 reduces lipogenic enzyme expression and de novo lipogenesis in adipose tissue. Adipose-specific mTORC2 knockout mice also display triglyceride accumulation in the liver. However, the mechanism and physiological role of hepatic triglyceride accumulation upon loss of adipose mTORC2 are unknown. Here, we show that loss of adipose mTORC2 increases the expression of de novo lipogenic enzymes in the liver, thereby causing accumulation of hepatic triglyceride and hypertriglyceridemia. Simultaneous inhibition of lipogenic enzymes in adipose tissue and liver by ablating mTORC2 in both tissues prevented accumulation of hepatic triglycerides and hypertriglyceridemia. However, loss of adipose and hepatic mTORC2 caused severe insulin resistance and glucose intolerance. Thus our findings suggest that increased hepatic lipogenesis is a compensatory mechanism to cope with loss of lipogenesis in adipose tissue, and further suggest that mTORC2 in adipose tissue and liver plays a crucial role in maintaining whole body energy homeostasis.NEW & NOTEWORTHY Loss of adipose and hepatic mTORC2 causes diabetes.


Subject(s)
Hypertriglyceridemia , Liver , Mice , Animals , Liver/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Lipogenesis/genetics , Obesity/metabolism , Glucose/metabolism , Homeostasis , Hypertriglyceridemia/complications , Hypertriglyceridemia/metabolism , Triglycerides/metabolism , Mammals/metabolism
18.
PLoS Comput Biol ; 18(9): e1009767, 2022 09.
Article in English | MEDLINE | ID: mdl-36067230

ABSTRACT

Comprehensive molecular characterization of cancer subtypes is essential for predicting clinical outcomes and searching for personalized treatments. We present bnClustOmics, a statistical model and computational tool for multi-omics unsupervised clustering, which serves a dual purpose: Clustering patient samples based on a Bayesian network mixture model and learning the networks of omics variables representing these clusters. The discovered networks encode interactions among all omics variables and provide a molecular characterization of each patient subgroup. We conducted simulation studies that demonstrated the advantages of our approach compared to other clustering methods in the case where the generative model is a mixture of Bayesian networks. We applied bnClustOmics to a hepatocellular carcinoma (HCC) dataset comprising genome (mutation and copy number), transcriptome, proteome, and phosphoproteome data. We identified three main HCC subtypes together with molecular characteristics, some of which are associated with survival even when adjusting for the clinical stage. Cluster-specific networks shed light on the links between genotypes and molecular phenotypes of samples within their respective clusters and suggest targets for personalized treatments.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Bayes Theorem , Carcinoma, Hepatocellular/genetics , Cluster Analysis , Humans , Liver Neoplasms/genetics , Proteome , Transcriptome
19.
Proc Natl Acad Sci U S A ; 117(3): 1524-1532, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31919282

ABSTRACT

Loss of the tumor suppressor tuberous sclerosis complex 1 (Tsc1) in the liver promotes gluconeogenesis and glucose intolerance. We asked whether this could be attributed to aberrant expression of small RNAs. We performed small-RNA sequencing on liver of Tsc1-knockout mice, and found that miRNAs of the delta-like homolog 1 (Dlk1)-deiodinase iodothyronine type III (Dio3) locus are up-regulated in an mTORC1-dependent manner. Sustained mTORC1 signaling during development prevented CpG methylation and silencing of the Dlk1-Dio3 locus, thereby increasing miRNA transcription. Deletion of miRNAs encoded by the Dlk1-Dio3 locus reduced gluconeogenesis, glucose intolerance, and fasting blood glucose levels. Thus, miRNAs contribute to the metabolic effects observed upon loss of TSC1 and hyperactivation of mTORC1 in the liver. Furthermore, we show that miRNA is a downstream effector of hyperactive mTORC1 signaling.


Subject(s)
Calcium-Binding Proteins/metabolism , Gluconeogenesis/physiology , Iodide Peroxidase/metabolism , MicroRNAs/metabolism , Tuberous Sclerosis Complex 1 Protein/metabolism , Up-Regulation , Animals , Calcium-Binding Proteins/genetics , Genetic Loci , Genomic Imprinting , Gluconeogenesis/genetics , Iodide Peroxidase/genetics , Liver/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , MicroRNAs/genetics , Sequence Analysis , Signal Transduction , Transcriptome , Tuberous Sclerosis Complex 1 Protein/genetics
20.
Am J Physiol Endocrinol Metab ; 323(2): E133-E144, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35723227

ABSTRACT

Mammalian target of rapamycin (mTOR) kinase is an essential hub where nutrients and growth factors converge to control cellular metabolism. mTOR interacts with different accessory proteins to form complexes 1 and 2 (mTORC), and each complex has different intracellular targets. Although mTORC1's role in ß-cells has been extensively studied, less is known about mTORC2's function in ß-cells. Here, we show that mice with constitutive and inducible ß-cell-specific deletion of RICTOR (ßRicKO and ißRicKO mice, respectively) are glucose intolerant due to impaired insulin secretion when glucose is injected intraperitoneally. Decreased insulin secretion in ßRicKO islets was caused by abnormal actin polymerization. Interestingly, when glucose was administered orally, no difference in glucose homeostasis and insulin secretion were observed, suggesting that incretins are counteracting the mTORC2 deficiency. Mechanistically, glucagon-like peptide-1 (GLP-1), but not gastric inhibitory polypeptide (GIP), rescued insulin secretion in vivo and in vitro by improving actin polymerization in ßRicKO islets. In conclusion, mTORC2 regulates glucose-stimulated insulin secretion by promoting actin filament remodeling.NEW & NOTEWORTHY The current studies uncover a novel mechanism linking mTORC2 signaling to glucose-stimulated insulin secretion by modulation of the actin filaments. This work also underscores the important role of GLP-1 in rescuing defects in insulin secretion by modulating actin polymerization and suggests that this effect is independent of mTORC2 signaling.


Subject(s)
Actins , Insulin , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Mammals/metabolism , Mechanistic Target of Rapamycin Complex 2 , Mice , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL