Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Nat Chem Biol ; 17(10): 1065-1074, 2021 10.
Article in English | MEDLINE | ID: mdl-34168367

ABSTRACT

The clinical benefits of pan-mTOR active-site inhibitors are limited by toxicity and relief of feedback inhibition of receptor expression. To address these limitations, we designed a series of compounds that selectively inhibit mTORC1 and not mTORC2. These 'bi-steric inhibitors' comprise a rapamycin-like core moiety covalently linked to an mTOR active-site inhibitor. Structural modification of these components modulated their affinities for their binding sites on mTOR and the selectivity of the bi-steric compound. mTORC1-selective compounds potently inhibited 4EBP1 phosphorylation and caused regressions of breast cancer xenografts. Inhibition of 4EBP1 phosphorylation was sufficient to block cancer cell growth and was necessary for maximal antitumor activity. At mTORC1-selective doses, these compounds do not alter glucose tolerance, nor do they relieve AKT-dependent feedback inhibition of HER3. Thus, in preclinical models, selective inhibitors of mTORC1 potently inhibit tumor growth while causing less toxicity and receptor reactivation as compared to pan-mTOR inhibitors.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Breast Neoplasms/drug therapy , Cell Line, Tumor , Female , Gene Expression Regulation/drug effects , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Structure-Activity Relationship
2.
PLoS Biol ; 17(12): e3000535, 2019 12.
Article in English | MEDLINE | ID: mdl-31800587

ABSTRACT

The mechanisms that govern organelle adaptation and remodelling remain poorly defined. The endo-lysosomal system degrades cargo from various routes, including endocytosis, phagocytosis, and autophagy. For phagocytes, endosomes and lysosomes (endo-lysosomes) are kingpin organelles because they are essential to kill pathogens and process and present antigens. During phagocyte activation, endo-lysosomes undergo a morphological transformation, going from a collection of dozens of globular structures to a tubular network in a process that requires the phosphatidylinositol-3-kinase-AKT-mechanistic target of rapamycin (mTOR) signalling pathway. Here, we show that the endo-lysosomal system undergoes an expansion in volume and holding capacity during phagocyte activation within 2 h of lipopolysaccharides (LPS) stimulation. Endo-lysosomal expansion was paralleled by an increase in lysosomal protein levels, but this was unexpectedly largely independent of the transcription factor EB (TFEB) and transcription factor E3 (TFE3), which are known to scale up lysosome biogenesis. Instead, we demonstrate a hitherto unappreciated mechanism of acute organelle expansion via mTOR Complex 1 (mTORC1)-dependent increase in translation, which appears to be mediated by both S6Ks and 4E-BPs. Moreover, we show that stimulation of RAW 264.7 macrophage cell line with LPS alters translation of a subset but not all of mRNAs encoding endo-lysosomal proteins, thereby suggesting that endo-lysosome expansion is accompanied by functional remodelling. Importantly, mTORC1-dependent increase in translation activity was necessary for efficient and rapid antigen presentation by dendritic cells. Collectively, we identified a previously unknown and functionally relevant mechanism for endo-lysosome expansion that relies on mTORC1-dependent translation to stimulate endo-lysosome biogenesis in response to an infection signal.


Subject(s)
Antigen Presentation/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Lysosomes/metabolism , Phagocytes/metabolism , Animals , Autophagy , Endosomes/drug effects , Endosomes/metabolism , Female , Lipopolysaccharides/pharmacology , Lysosomes/drug effects , Macrophage Activation , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Phagocytes/drug effects , Phagocytosis , Phosphatidylinositol 3-Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
3.
Genome Res ; 26(5): 636-48, 2016 05.
Article in English | MEDLINE | ID: mdl-26984228

ABSTRACT

The diversity of MTOR-regulated mRNA translation remains unresolved. Whereas ribosome-profiling suggested that MTOR almost exclusively stimulates translation of the TOP (terminal oligopyrimidine motif) and TOP-like mRNAs, polysome-profiling indicated that MTOR also modulates translation of mRNAs without the 5' TOP motif (non-TOP mRNAs). We demonstrate that in ribosome-profiling studies, detection of MTOR-dependent changes in non-TOP mRNA translation was obscured by low sensitivity and methodology biases. Transcription start site profiling using nano-cap analysis of gene expression (nanoCAGE) revealed that not only do many MTOR-sensitive mRNAs lack the 5' TOP motif but that 5' UTR features distinguish two functionally and translationally distinct subsets of MTOR-sensitive mRNAs: (1) mRNAs with short 5' UTRs enriched for mitochondrial functions, which require EIF4E but are less EIF4A1-sensitive; and (2) long 5' UTR mRNAs encoding proliferation- and survival-promoting proteins, which are both EIF4E- and EIF4A1-sensitive. Selective inhibition of translation of mRNAs harboring long 5' UTRs via EIF4A1 suppression leads to sustained expression of proteins involved in respiration but concomitant loss of those protecting mitochondrial structural integrity, resulting in apoptosis. Conversely, simultaneous suppression of translation of both long and short 5' UTR mRNAs by MTOR inhibitors results in metabolic dormancy and a predominantly cytostatic effect. Thus, 5' UTR features define different modes of MTOR-sensitive translation of functionally distinct subsets of mRNAs, which may explain the diverse impact of MTOR and EIF4A inhibitors on neoplastic cells.


Subject(s)
5' Untranslated Regions/physiology , Eukaryotic Initiation Factor-4E/metabolism , Protein Biosynthesis/physiology , TOR Serine-Threonine Kinases/metabolism , Apoptosis/physiology , Female , Humans , MCF-7 Cells
6.
Cell Metab ; 28(6): 817-832.e8, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30244971

ABSTRACT

There is increasing interest in therapeutically exploiting metabolic differences between normal and cancer cells. We show that kinase inhibitors (KIs) and biguanides synergistically and selectively target a variety of cancer cells. Synthesis of non-essential amino acids (NEAAs) aspartate, asparagine, and serine, as well as glutamine metabolism, are major determinants of the efficacy of KI/biguanide combinations. The mTORC1/4E-BP axis regulates aspartate, asparagine, and serine synthesis by modulating mRNA translation, while ablation of 4E-BP1/2 substantially decreases sensitivity of breast cancer and melanoma cells to KI/biguanide combinations. Efficacy of the KI/biguanide combinations is also determined by HIF-1α-dependent perturbations in glutamine metabolism, which were observed in VHL-deficient renal cancer cells. This suggests that cancer cells display metabolic plasticity by engaging non-redundant adaptive mechanisms, which allows them to survive therapeutic insults that target cancer metabolism.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Amino Acids/metabolism , Animals , Biguanides/pharmacology , Cell Cycle Proteins , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , K562 Cells , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Xenograft Model Antitumor Assays
7.
Nat Commun ; 7: 11127, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-27040916

ABSTRACT

Ternary complex (TC) and eIF4F complex assembly are the two major rate-limiting steps in translation initiation regulated by eIF2α phosphorylation and the mTOR/4E-BP pathway, respectively. How TC and eIF4F assembly are coordinated, however, remains largely unknown. We show that mTOR suppresses translation of mRNAs activated under short-term stress wherein TC recycling is attenuated by eIF2α phosphorylation. During acute nutrient or growth factor stimulation, mTORC1 induces eIF2ß phosphorylation and recruitment of NCK1 to eIF2, decreases eIF2α phosphorylation and bolsters TC recycling. Accordingly, eIF2ß mediates the effect of mTORC1 on protein synthesis and proliferation. In addition, we demonstrate a formerly undocumented role for CK2 in regulation of translation initiation, whereby CK2 stimulates phosphorylation of eIF2ß and simultaneously bolsters eIF4F complex assembly via the mTORC1/4E-BP pathway. These findings imply a previously unrecognized mode of translation regulation, whereby mTORC1 and CK2 coordinate TC and eIF4F complex assembly to stimulate cell proliferation.


Subject(s)
Casein Kinase II/physiology , Eukaryotic Initiation Factor-4F/metabolism , Multiprotein Complexes/physiology , TOR Serine-Threonine Kinases/physiology , Ternary Complex Factors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Casein Kinase II/genetics , Casein Kinase II/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2/physiology , Gene Expression Regulation , HEK293 Cells , Humans , MCF-7 Cells , Mechanistic Target of Rapamycin Complex 1 , Models, Genetic , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Oncogene Proteins/metabolism , Peptide Chain Initiation, Translational , Phosphorylation , Signal Transduction , Stress, Physiological , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
8.
J Vis Exp ; (87)2014 May 17.
Article in English | MEDLINE | ID: mdl-24893926

ABSTRACT

mRNA translation plays a central role in the regulation of gene expression and represents the most energy consuming process in mammalian cells. Accordingly, dysregulation of mRNA translation is considered to play a major role in a variety of pathological states including cancer. Ribosomes also host chaperones, which facilitate folding of nascent polypeptides, thereby modulating function and stability of newly synthesized polypeptides. In addition, emerging data indicate that ribosomes serve as a platform for a repertoire of signaling molecules, which are implicated in a variety of post-translational modifications of newly synthesized polypeptides as they emerge from the ribosome, and/or components of translational machinery. Herein, a well-established method of ribosome fractionation using sucrose density gradient centrifugation is described. In conjunction with the in-house developed "anota" algorithm this method allows direct determination of differential translation of individual mRNAs on a genome-wide scale. Moreover, this versatile protocol can be used for a variety of biochemical studies aiming to dissect the function of ribosome-associated protein complexes, including those that play a central role in folding and degradation of newly synthesized polypeptides.


Subject(s)
Centrifugation, Density Gradient/methods , Genome-Wide Association Study/methods , Polyribosomes/chemistry , Protein Biosynthesis , Humans , MCF-7 Cells , Polyribosomes/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Sucrose/chemistry
9.
Mol Cell Biol ; 33(13): 2510-26, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23608534

ABSTRACT

Folding of newly synthesized polypeptides (NSPs) into functional proteins is a highly regulated process. Rigorous quality control ensures that NSPs attain their native fold during or shortly after completion of translation. Nonetheless, signaling pathways that govern the degradation of NSPs in mammals remain elusive. We demonstrate that the stress-induced c-Jun N-terminal kinase (JNK) is recruited to ribosomes by the receptor for activated protein C kinase 1 (RACK1). RACK1 is an integral component of the 40S ribosome and an adaptor for protein kinases. Ribosome-associated JNK phosphorylates the eukaryotic translation elongation factor 1A isoform 2 (eEF1A2) on serines 205 and 358 to promote degradation of NSPs by the proteasome. These findings establish a role for a RACK1/JNK/eEF1A2 complex in the quality control of NSPs in response to stress.


Subject(s)
GTP-Binding Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neoplasm Proteins/metabolism , Peptide Elongation Factor 1/metabolism , Peptides/metabolism , Receptors, Cell Surface/metabolism , Animals , Base Sequence , Cell Line , GTP-Binding Proteins/genetics , Humans , JNK Mitogen-Activated Protein Kinases/genetics , MAP Kinase Kinase 7/genetics , MAP Kinase Kinase 7/metabolism , Molecular Sequence Data , Neoplasm Proteins/genetics , Peptide Elongation Factor 1/genetics , Phosphorylation , Polyribosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Receptors for Activated C Kinase , Receptors, Cell Surface/genetics , Ribosomes/metabolism , Serine/metabolism , Signal Transduction
10.
Cell Metab ; 18(5): 698-711, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24206664

ABSTRACT

mRNA translation is thought to be the most energy-consuming process in the cell. Translation and energy metabolism are dysregulated in a variety of diseases including cancer, diabetes, and heart disease. However, the mechanisms that coordinate translation and energy metabolism in mammals remain largely unknown. The mechanistic/mammalian target of rapamycin complex 1 (mTORC1) stimulates mRNA translation and other anabolic processes. We demonstrate that mTORC1 controls mitochondrial activity and biogenesis by selectively promoting translation of nucleus-encoded mitochondria-related mRNAs via inhibition of the eukaryotic translation initiation factor 4E (eIF4E)-binding proteins (4E-BPs). Stimulating the translation of nucleus-encoded mitochondria-related mRNAs engenders an increase in ATP production capacity, a required energy source for translation. These findings establish a feed-forward loop that links mRNA translation to oxidative phosphorylation, thereby providing a key mechanism linking aberrant mTOR signaling to conditions of abnormal cellular energy metabolism such as neoplasia and insulin resistance.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation , Mitochondria/metabolism , Mitochondrial Turnover , Multiprotein Complexes/metabolism , Phosphoproteins/metabolism , Protein Biosynthesis , TOR Serine-Threonine Kinases/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Autophagy/genetics , Cell Cycle Proteins , Cell Nucleus/metabolism , Cell Respiration , DNA, Mitochondrial/biosynthesis , DNA-Binding Proteins/metabolism , Genome, Human/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mitochondrial Proteins/metabolism , Models, Biological , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomal Protein S6 Kinases/metabolism , Transcription Factors/metabolism
11.
Nat Med ; 16(10): 1157-60, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20871611

ABSTRACT

Migraine with aura is a common, debilitating, recurrent headache disorder associated with transient and reversible focal neurological symptoms. A role has been suggested for the two-pore domain (K2P) potassium channel, TWIK-related spinal cord potassium channel (TRESK, encoded by KCNK18), in pain pathways and general anaesthesia. We therefore examined whether TRESK is involved in migraine by screening the KCNK18 gene in subjects diagnosed with migraine. Here we report a frameshift mutation, F139WfsX24, which segregates perfectly with typical migraine with aura in a large pedigree. We also identified prominent TRESK expression in migraine-salient areas such as the trigeminal ganglion. Functional characterization of this mutation demonstrates that it causes a complete loss of TRESK function and that the mutant subunit suppresses wild-type channel function through a dominant-negative effect, thus explaining the dominant penetrance of this allele. These results therefore support a role for TRESK in the pathogenesis of typical migraine with aura and further support the role of this channel as a potential therapeutic target.


Subject(s)
Migraine with Aura/genetics , Mutation , Potassium Channels/genetics , Animals , Genetic Linkage , Humans , Mice , Polymorphism, Single Nucleotide , Potassium Channels/physiology
SELECTION OF CITATIONS
SEARCH DETAIL