Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
Cell Chem Biol ; 26(9): 1203-1213.e13, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31231029

ABSTRACT

The mechanistic target of rapamycin (mTOR) is a central regulator of cellular metabolic processes. Dysregulation of this kinase complex can result in a variety of human diseases. Rapamycin and its analogs target mTORC1 directly; however, chronic treatment in certain cell types and in vivo results in the inhibition of both mTORC1 and mTORC2. We have developed a high-throughput cell-based screen for the detection of phosphorylated forms of the mTORC1 (4E-BP1, S6K1) and mTORC2 (Akt) substrates and have identified and characterized a chemical scaffold that demonstrates a profile consistent with the selective inhibition of mTORC1. Stable isotope labeling of amino acids in cell culture-based proteomic target identification revealed that class I glucose transporters were the primary target for these compounds yielding potent inhibition of glucose uptake and, as a result, selective inhibition of mTORC1. The link between the glucose uptake and selective mTORC1 inhibition are discussed in the context of a yet-to-be discovered glucose sensor.


Subject(s)
Glucose Transport Proteins, Facilitative/drug effects , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Sirolimus/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical/methods , Glucose/metabolism , High-Throughput Screening Assays/methods , Humans , Mechanistic Target of Rapamycin Complex 2/drug effects , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred C57BL , Multiprotein Complexes/metabolism , Phosphorylation , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Sirolimus/analogs & derivatives , Sirolimus/metabolism , Transcription Factors/metabolism
3.
Nat Commun ; 9(1): 548, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416044

ABSTRACT

The small G-protein Rheb activates the mechanistic target of rapamycin complex 1 (mTORC1) in response to growth factor signals. mTORC1 is a master regulator of cellular growth and metabolism; aberrant mTORC1 signaling is associated with fibrotic, metabolic, and neurodegenerative diseases, cancers, and rare disorders. Point mutations in the Rheb switch II domain impair its ability to activate mTORC1. Here, we report the discovery of a small molecule (NR1) that binds Rheb in the switch II domain and selectively blocks mTORC1 signaling. NR1 potently inhibits mTORC1 driven phosphorylation of ribosomal protein S6 kinase beta-1 (S6K1) but does not inhibit phosphorylation of AKT or ERK. In contrast to rapamycin, NR1 does not cause inhibition of mTORC2 upon prolonged treatment. Furthermore, NR1 potently and selectively inhibits mTORC1 in mouse kidney and muscle in vivo. The data presented herein suggest that pharmacological inhibition of Rheb is an effective approach for selective inhibition of mTORC1 with therapeutic potential.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Ras Homolog Enriched in Brain Protein/antagonists & inhibitors , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Animals , Cell Line, Tumor , Crystallography, X-Ray , HEK293 Cells , Humans , Jurkat Cells , MCF-7 Cells , Male , Mice, Inbred C57BL , Molecular Structure , Phosphorylation/drug effects , Ras Homolog Enriched in Brain Protein/genetics , Ras Homolog Enriched in Brain Protein/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Small Molecule Libraries/chemistry
4.
Cancer Res ; 76(24): 7168-7180, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27913436

ABSTRACT

Aberrant activation of the PI3K/mTOR pathway is a common feature of many cancers and an attractive target for therapy, but resistance inevitably evolves as is the case for any cancer cell-targeted therapy. In animal tumor models, chronic inhibition of PI3K/mTOR initially inhibits tumor growth, but over time, tumor cells escape inhibition. In this study, we identified a context-dependent mechanism of escape whereby tumor cells upregulated the proto-oncogene transcriptional regulators c-MYC and YAP1. This mechanism was dependent on both constitutive ERK activity as well as inhibition of the stress kinase p38. Inhibition of p38 relieved proliferation arrest and allowed upregulation of MYC and YAP through stabilization of CREB. These data provide new insights into cellular signaling mechanisms that influence resistance to PI3K/mTOR inhibitors. Furthermore, they suggest that therapies that inactivate YAP or MYC or augment p38 activity could enhance the efficacy of PI3K/mTOR inhibitors. Cancer Res; 76(24); 7168-80. ©2016 AACR.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Drug Resistance, Neoplasm/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Neoplasms, Experimental/pathology , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Fluorescent Antibody Technique , Heterografts , Humans , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred NOD , Microscopy, Confocal , Neoplasms, Experimental/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transcription Factors , YAP-Signaling Proteins
5.
Science ; 341(6144): 1236566, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23888043

ABSTRACT

The mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) protein kinase promotes growth and is the target of rapamycin, a clinically useful drug that also prolongs life span in model organisms. A persistent mystery is why the phosphorylation of many bona fide mTORC1 substrates is resistant to rapamycin. We find that the in vitro kinase activity of mTORC1 toward peptides encompassing established phosphorylation sites varies widely and correlates strongly with the resistance of the sites to rapamycin, as well as to nutrient and growth factor starvation within cells. Slight modifications of the sites were sufficient to alter mTORC1 activity toward them in vitro and to cause concomitant changes within cells in their sensitivity to rapamycin and starvation. Thus, the intrinsic capacity of a phosphorylation site to serve as an mTORC1 substrate, a property we call substrate quality, is a major determinant of its sensitivity to modulators of the pathway. Our results reveal a mechanism through which mTORC1 effectors can respond differentially to the same signals.


Subject(s)
Peptides/metabolism , Proteins/chemistry , Proteins/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/chemistry , TOR Serine-Threonine Kinases/metabolism , Amino Acid Motifs , Amino Acids/metabolism , Animals , Cell Line , Culture Media , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Naphthyridines/pharmacology , Peptides/chemistry , Phosphorylation , Proteins/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors
6.
Cancer Res ; 73(8): 2574-86, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23436801

ABSTRACT

mTOR is a highly conserved serine/threonine protein kinase that serves as a central regulator of cell growth, survival, and autophagy. Deregulation of the PI3K/Akt/mTOR signaling pathway occurs commonly in cancer and numerous inhibitors targeting the ATP-binding site of these kinases are currently undergoing clinical evaluation. Here, we report the characterization of Torin2, a second-generation ATP-competitive inhibitor that is potent and selective for mTOR with a superior pharmacokinetic profile to previous inhibitors. Torin2 inhibited mTORC1-dependent T389 phosphorylation on S6K (RPS6KB1) with an EC(50) of 250 pmol/L with approximately 800-fold selectivity for cellular mTOR versus phosphoinositide 3-kinase (PI3K). Torin2 also exhibited potent biochemical and cellular activity against phosphatidylinositol-3 kinase-like kinase (PIKK) family kinases including ATM (EC(50), 28 nmol/L), ATR (EC(50), 35 nmol/L), and DNA-PK (EC(50), 118 nmol/L; PRKDC), the inhibition of which sensitized cells to Irradiation. Similar to the earlier generation compound Torin1 and in contrast to other reported mTOR inhibitors, Torin2 inhibited mTOR kinase and mTORC1 signaling activities in a sustained manner suggestive of a slow dissociation from the kinase. Cancer cell treatment with Torin2 for 24 hours resulted in a prolonged block in negative feedback and consequent T308 phosphorylation on Akt. These effects were associated with strong growth inhibition in vitro. Single-agent treatment with Torin2 in vivo did not yield significant efficacy against KRAS-driven lung tumors, but the combination of Torin2 with mitogen-activated protein/extracellular signal-regulated kinase (MEK) inhibitor AZD6244 yielded a significant growth inhibition. Taken together, our findings establish Torin2 as a strong candidate for clinical evaluation in a broad number of oncologic settings where mTOR signaling has a pathogenic role.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Cycle Proteins/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Naphthyridines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins , Autophagy/drug effects , Benzimidazoles/pharmacology , Binding, Competitive , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Synergism , Humans , Kinetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Naphthyridines/administration & dosage , Naphthyridines/chemistry , Protein Binding , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays , ras Proteins/genetics
7.
J Biol Chem ; 287(13): 9742-9752, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22223645

ABSTRACT

An intensive recent effort to develop ATP-competitive mTOR inhibitors has resulted in several potent and selective molecules such as Torin1, PP242, KU63794, and WYE354. These inhibitors are being widely used as pharmacological probes of mTOR-dependent biology. To determine the potency and specificity of these agents, we have undertaken a systematic kinome-wide effort to profile their selectivity and potency using chemical proteomics and assays for enzymatic activity, protein binding, and disruption of cellular signaling. Enzymatic and cellular assays revealed that all four compounds are potent inhibitors of mTORC1 and mTORC2, with Torin1 exhibiting ∼20-fold greater potency for inhibition of Thr-389 phosphorylation on S6 kinases (EC(50) = 2 nM) relative to other inhibitors. In vitro biochemical profiling at 10 µM revealed binding of PP242 to numerous kinases, although WYE354 and KU63794 bound only to p38 kinases and PI3K isoforms and Torin1 to ataxia telangiectasia mutated, ATM and Rad3-related protein, and DNA-PK. Analysis of these protein targets in cellular assays did not reveal any off-target activities for Torin1, WYE354, and KU63794 at concentrations below 1 µM but did show that PP242 efficiently inhibited the RET receptor (EC(50), 42 nM) and JAK1/2/3 kinases (EC(50), 780 nM). In addition, Torin1 displayed unusually slow kinetics for inhibition of the mTORC1/2 complex, a property likely to contribute to the pharmacology of this inhibitor. Our results demonstrated that, with the exception of PP242, available ATP-competitive compounds are highly selective mTOR inhibitors when applied to cells at concentrations below 1 µM and that the compounds may represent a starting point for medicinal chemistry efforts aimed at developing inhibitors of other PI3K kinase-related kinases.


Subject(s)
Enzyme Inhibitors/pharmacokinetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Adenosine Triphosphate , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proteins/antagonists & inhibitors , Proteins/metabolism , Proteomics/methods , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Methods Mol Biol ; 821: 447-60, 2012.
Article in English | MEDLINE | ID: mdl-22125084

ABSTRACT

The mammalian Target of Rapamycin (mTOR)-mediated signaling transduction pathway has been observed to be deregulated in a wide variety of cancer and metabolic diseases. Despite extensive clinical development efforts, the well-known allosteric mTOR inhibitor rapamycin and structurally related rapalogs have failed to show significant single-agent antitumor efficacy in most types of cancer. This limited clinical success may be due to the inability of the rapalogs to maintain a complete blockade mTOR-mediated signaling. Therefore, numerous efforts have been initiated to develop ATP-competitive mTOR inhibitors that would block both mTORC1 and mTORC2 complex activity. Here, we describe our experimental approaches to develop Torin1 using a medium throughput cell-based screening assay and structure-guided drug design.


Subject(s)
Adenosine Triphosphate/metabolism , Drug Design , High-Throughput Screening Assays/methods , Naphthyridines/chemistry , Protein Kinase Inhibitors/chemistry , Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Models, Molecular , Molecular Structure , Multiprotein Complexes , Naphthyridines/pharmacology , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
9.
Cell ; 146(3): 408-20, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21816276

ABSTRACT

The nutrient- and growth factor-responsive kinase mTOR complex 1 (mTORC1) regulates many processes that control growth, including protein synthesis, autophagy, and lipogenesis. Through unknown mechanisms, mTORC1 promotes the function of SREBP, a master regulator of lipo- and sterolgenic gene transcription. Here, we demonstrate that mTORC1 regulates SREBP by controlling the nuclear entry of lipin 1, a phosphatidic acid phosphatase. Dephosphorylated, nuclear, catalytically active lipin 1 promotes nuclear remodeling and mediates the effects of mTORC1 on SREBP target gene, SREBP promoter activity, and nuclear SREBP protein abundance. Inhibition of mTORC1 in the liver significantly impairs SREBP function and makes mice resistant, in a lipin 1-dependent fashion, to the hepatic steatosis and hypercholesterolemia induced by a high-fat and -cholesterol diet. These findings establish lipin 1 as a key component of the mTORC1-SREBP pathway.


Subject(s)
Nuclear Proteins/metabolism , Proteins/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 2/metabolism , Animals , Humans , Lipid Metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Phosphatidate Phosphatase , TOR Serine-Threonine Kinases
10.
Science ; 332(6035): 1317-22, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21659604

ABSTRACT

The mammalian target of rapamycin (mTOR) protein kinase is a master growth promoter that nucleates two complexes, mTORC1 and mTORC2. Despite the diverse processes controlled by mTOR, few substrates are known. We defined the mTOR-regulated phosphoproteome by quantitative mass spectrometry and characterized the primary sequence motif specificity of mTOR using positional scanning peptide libraries. We found that the phosphorylation response to insulin is largely mTOR dependent and that mTOR exhibits a unique preference for proline, hydrophobic, and aromatic residues at the +1 position. The adaptor protein Grb10 was identified as an mTORC1 substrate that mediates the inhibition of phosphoinositide 3-kinase typical of cells lacking tuberous sclerosis complex 2 (TSC2), a tumor suppressor and negative regulator of mTORC1. Our work clarifies how mTORC1 inhibits growth factor signaling and opens new areas of investigation in mTOR biology.


Subject(s)
GRB10 Adaptor Protein/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Proteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Humans , Insulin/metabolism , Mass Spectrometry , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Naphthyridines/pharmacology , Phosphoproteins/metabolism , Phosphorylation , Proteome/metabolism , Sirolimus/pharmacology
11.
Bioorg Med Chem Lett ; 21(13): 4036-40, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21621413

ABSTRACT

Starting from small molecule mTOR inhibitor Torin1, replacement of the piperazine ring with a phenyl ring resulted in a new series of mTOR inhibitors (as exemplified by 10) that showed superior potency and selectivity for mTOR, along with significantly improved mouse liver microsome stability and a longer in vivo half-life.


Subject(s)
Drug Discovery , Microsomes, Liver/drug effects , Naphthyridines/chemical synthesis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cells, Cultured , Drug Stability , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Mice , Molecular Structure , Naphthyridines/chemistry , Naphthyridines/pharmacology
12.
J Med Chem ; 54(5): 1473-80, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21322566

ABSTRACT

The mTOR mediated PI3K/AKT/mTOR signal transduction pathway has been demonstrated to play a key role in a broad spectrum of cancers. Starting from the mTOR selective inhibitor 1 (Torin1), a focused medicinal chemistry effort led to the discovery of an improved mTOR inhibitor 3 (Torin2), which possesses an EC(50) of 0.25 nM for inhibiting cellular mTOR activity. Compound 3 exhibited 800-fold selectivity over PI3K (EC(50): 200 nM) and over 100-fold binding selectivity relative to 440 other protein kinases. Compound 3 has significantly improved bioavailability (54%), metabolic stability, and plasma exposure relative to compound 1.


Subject(s)
Aminopyridines/chemical synthesis , Antineoplastic Agents/chemical synthesis , Naphthyridines/chemical synthesis , TOR Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Aminopyridines/pharmacokinetics , Aminopyridines/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biological Availability , Drug Stability , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Models, Molecular , Naphthyridines/pharmacokinetics , Naphthyridines/pharmacology , Structure-Activity Relationship
13.
J Med Chem ; 53(19): 7146-55, 2010 Oct 14.
Article in English | MEDLINE | ID: mdl-20860370

ABSTRACT

The mTOR protein is a master regulator of cell growth and proliferation, and inhibitors of its kinase activity have the potential to become new class of anticancer drugs. Starting from quinoline 1, which was identified in a biochemical mTOR assay, we developed a tricyclic benzonaphthyridinone inhibitor 37 (Torin1), which inhibited phosphorylation of mTORC1 and mTORC2 substrates in cells at concentrations of 2 and 10 nM, respectively. Moreover, Torin1 exhibits 1000-fold selectivity for mTOR over PI3K (EC(50) = 1800 nM) and exhibits 100-fold binding selectivity relative to 450 other protein kinases. Torin1 was efficacious at a dose of 20 mg/kg in a U87MG xenograft model and demonstrated good pharmacodynamic inhibition of downstream effectors of mTOR in tumor and peripheral tissues. These results demonstrate that Torin1 is a useful probe of mTOR-dependent phenomena and that benzonaphthridinones represent a promising scaffold for the further development of mTOR-specific inhibitors with the potential for clinical utility.


Subject(s)
Antineoplastic Agents/chemical synthesis , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Naphthyridines/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Humans , In Vitro Techniques , Male , Mice , Mice, Nude , Microsomes, Liver/metabolism , Models, Molecular , Naphthyridines/pharmacokinetics , Naphthyridines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Structure-Activity Relationship , TOR Serine-Threonine Kinases , Xenograft Model Antitumor Assays
14.
Anal Chem ; 82(15): 6643-51, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20614870

ABSTRACT

Here we report the use of capillary isoelectric focusing under native conditions for the separation of protein complex isoforms and subcomplexes. Using biologically relevant HIS-tag and FLAG-tag purified protein complexes, we demonstrate the separations of protein complex isoforms of the mammalian target of rapamycin complex (mTORC1 and 2) and the subcomplexes and different phosphorylation states of the Dam1 complex. The high efficiency capillary isoelectric focusing separation allowed for resolution of protein complexes and subcomplexes similar in size and biochemical composition. By performing separations with native buffers and reduced temperature (15 degrees C) we were able to maintain the complex integrity of the more thermolabile mTORC2 during isoelectric focusing and detection (<45 min). Increasing the separation temperature allowed us to monitor dissociation of the Dam1 complex into its subcomplexes (25 degrees C) and eventually its individual protein components (30 degrees C). The separation of two different phosphorylation states of the Dam1 complex, generated from an in vitro kinase assay with Mps1 kinase, was straightforward due to the large pI shift upon multiple phosphorylation events. The separation of the protein complex isoforms of mTORC, on the other hand, required the addition of a small pI range (4-6.5) of ampholytes to improve resolution and stability of the complexes. We show that native capillary isoelectric focusing is a powerful method for the difficult separations of large, similar, unstable protein complexes. This method shows potential for differentiation of protein complex isoform and subcomplex compositions, post-translational modifications, architectures, stabilities, equilibria, and relative abundances under biologically relevant conditions.


Subject(s)
Isoelectric Focusing/methods , Multiprotein Complexes/isolation & purification , Buffers , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/isolation & purification , Cell Line , Histidine/chemistry , Humans , Hydrogen-Ion Concentration , Mechanistic Target of Rapamycin Complex 1 , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/isolation & purification , Multiprotein Complexes/chemistry , Oligopeptides/chemistry , Peptides/chemistry , Phosphorylation , Protein Isoforms/chemistry , Protein Isoforms/isolation & purification , Protein Subunits/chemistry , Protein Subunits/isolation & purification , Proteins , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/isolation & purification , TOR Serine-Threonine Kinases , Temperature , Transcription Factors/chemistry , Transcription Factors/isolation & purification , Viscosity
15.
Mol Cell ; 38(5): 768-74, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20542007

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) regulates cell growth in response to the nutrient and energy status of the cell, and its deregulation is common in human cancers. Little is known about the overall architecture and subunit organization of this essential signaling complex. We have determined the three-dimensional (3D) structure of the fully assembled human mTORC1 by cryo-electron microscopy (cryo-EM). Our analyses reveal that mTORC1 is an obligate dimer with an overall rhomboid shape and a central cavity. The dimeric interfaces are formed by interlocking interactions between the mTOR and raptor subunits. Extended incubation with FKBP12-rapamycin compromises the structural integrity of mTORC1 in a stepwise manner, leading us to propose a model in which rapamycin inhibits mTORC1-mediated phosphorylation of 4E-BP1 and S6K1 through different mechanisms.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Sirolimus/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Antibiotics, Antineoplastic/chemistry , Cell Line , Computer Simulation , Cryoelectron Microscopy , Humans , Mechanistic Target of Rapamycin Complex 1 , Models, Molecular , Multiprotein Complexes , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Proteins/chemistry , Proteins/metabolism , Regulatory-Associated Protein of mTOR , Sirolimus/chemistry , TOR Serine-Threonine Kinases , Tacrolimus Binding Protein 1A/chemistry , Tacrolimus Binding Protein 1A/metabolism , Transcription Factors/genetics
16.
Cell ; 137(5): 873-86, 2009 May 29.
Article in English | MEDLINE | ID: mdl-19446321

ABSTRACT

The mTORC1 and mTORC2 pathways regulate cell growth, proliferation, and survival. We identify DEPTOR as an mTOR-interacting protein whose expression is negatively regulated by mTORC1 and mTORC2. Loss of DEPTOR activates S6K1, Akt, and SGK1, promotes cell growth and survival, and activates mTORC1 and mTORC2 kinase activities. DEPTOR overexpression suppresses S6K1 but, by relieving feedback inhibition from mTORC1 to PI3K signaling, activates Akt. Consistent with many human cancers having activated mTORC1 and mTORC2 pathways, DEPTOR expression is low in most cancers. Surprisingly, DEPTOR is highly overexpressed in a subset of multiple myelomas harboring cyclin D1/D3 or c-MAF/MAFB translocations. In these cells, high DEPTOR expression is necessary to maintain PI3K and Akt activation and a reduction in DEPTOR levels leads to apoptosis. Thus, we identify a novel mTOR-interacting protein whose deregulated overexpression in multiple myeloma cells represents a mechanism for activating PI3K/Akt signaling and promoting cell survival.


Subject(s)
Cell Survival , Multiple Myeloma/metabolism , Protein Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Cell Line , Cyclin D1/metabolism , Cyclin D3 , Cyclins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
17.
J Biol Chem ; 284(12): 8023-32, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19150980

ABSTRACT

The mammalian target of rapamycin (mTOR) kinase is the catalytic subunit of two functionally distinct complexes, mTORC1 and mTORC2, that coordinately promote cell growth, proliferation, and survival. Rapamycin is a potent allosteric mTORC1 inhibitor with clinical applications as an immunosuppressant and anti-cancer agent. Here we find that Torin1, a highly potent and selective ATP-competitive mTOR inhibitor that directly inhibits both complexes, impairs cell growth and proliferation to a far greater degree than rapamycin. Surprisingly, these effects are independent of mTORC2 inhibition and are instead because of suppression of rapamycin-resistant functions of mTORC1 that are necessary for cap-dependent translation and suppression of autophagy. These effects are at least partly mediated by mTORC1-dependent and rapamycin-resistant phosphorylation of 4E-BP1. Our findings challenge the assumption that rapamycin completely inhibits mTORC1 and indicate that direct inhibitors of mTORC1 kinase activity may be more successful than rapamycin at inhibiting tumors that depend on mTORC1.


Subject(s)
Adenosine Triphosphate/metabolism , Antibiotics, Antineoplastic/pharmacology , Carrier Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Multienzyme Complexes/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sirolimus/pharmacology , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Autophagy/drug effects , Cell Cycle Proteins , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Eukaryotic Initiation Factors , Immunosuppressive Agents/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multienzyme Complexes/antagonists & inhibitors , Multiprotein Complexes , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Proteins , RNA Caps/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/antagonists & inhibitors
18.
Mol Cell ; 25(6): 903-15, 2007 Mar 23.
Article in English | MEDLINE | ID: mdl-17386266

ABSTRACT

The heterotrimeric mTORC1 protein kinase nucleates a signaling network that promotes cell growth in response to insulin and becomes constitutively active in cells missing the TSC1 or TSC2 tumor suppressors. Insulin stimulates the phosphorylation of S6K1, an mTORC1 substrate, but it is not known how mTORC1 kinase activity is regulated. We identify PRAS40 as a raptor-interacting protein that binds to mTORC1 in insulin-deprived cells and whose in vitro interaction with mTORC1 is disrupted by high salt concentrations. PRAS40 inhibits cell growth, S6K1 phosphorylation, and rheb-induced activation of the mTORC1 pathway, and in vitro it prevents the great increase in mTORC1 kinase activity induced by rheb1-GTP. Insulin stimulates Akt/PKB-mediated phosphorylation of PRAS40, which prevents its inhibition of mTORC1 in cells and in vitro. We propose that the relative strengths of the rheb- and PRAS40-mediated inputs to mTORC1 set overall pathway activity and that insulin activates mTORC1 through the coordinated regulation of both.


Subject(s)
Insulin/physiology , Phosphoproteins/metabolism , Protein Kinase Inhibitors/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins , Cell Line , Enzyme Inhibitors/metabolism , Humans , Kinetics , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Phosphorylation , Proteins , TOR Serine-Threonine Kinases
19.
Biochemistry ; 46(1): 145-55, 2007 Jan 09.
Article in English | MEDLINE | ID: mdl-17198384

ABSTRACT

The structure of tryptophan 2,3-dioxygenase (TDO) from Ralstonia metallidurans was determined at 2.4 A. TDO catalyzes the irreversible oxidation of l-tryptophan to N-formyl kynurenine, which is the initial step in tryptophan catabolism. TDO is a heme-containing enzyme and is highly specific for its substrate l-tryptophan. The structure is a tetramer with a heme cofactor bound at each active site. The monomeric fold, as well as the heme binding site, is similar to that of the large domain of indoleamine 2,3-dioxygenase, an enzyme that catalyzes the same reaction except with a broader substrate tolerance. Modeling of the putative (S)-tryptophan hydroperoxide intermediate into the active site, as well as substrate analogue and mutagenesis studies, are consistent with a Criegee mechanism for the reaction.


Subject(s)
Heme/metabolism , Quinolinic Acid/metabolism , Tryptophan Oxygenase/chemistry , Tryptophan Oxygenase/metabolism , Tryptophan/metabolism , Binding Sites , Crystallography, X-Ray , Glycine/genetics , Glycine/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Folding , Ralstonia/classification , Ralstonia/enzymology , Structure-Activity Relationship , Substrate Specificity
20.
J Am Chem Soc ; 128(7): 2346-55, 2006 Feb 22.
Article in English | MEDLINE | ID: mdl-16478190

ABSTRACT

D(2)O-grown crystals of yeast zinc porphyrin substituted cytochrome c peroxidase (ZnCcP) in complex with yeast iso-1-cytochrome c (yCc) diffract to higher resolution (1.7 A) and pack differently than H(2)O-grown crystals (2.4-3.0 A). Two ZnCcP's bind the same yCc (porphyrin-to-porphyrin separations of 19 and 29 A), with one ZnCcP interacting through the same interface found in the H(2)O crystals. The triplet excited-state of at least one of the two unique ZnCcP's is quenched by electron transfer (ET) to Fe(III)yCc (k(e) = 220 s(-1)). Measurement of thermal recombination ET between Fe(II)yCc and ZnCcP+ in the D(2)O-treated crystals has both slow and fast components that differ by 2 orders of magnitude (k(eb)(1) = 2200 s(-1), k(eb)(2) = 30 s(-1)). Back ET in H(2)O-grown crystals is too fast for observation, but soaking H(2)O-grown crystals in D(2)O for hours generates slower back ET, with kinetics similar to those of the D(2)O-grown crystals (k(eb)(1) = 7000 s(-1), k(eb)(2) = 100 s(-1)). Protein-film voltammetry of yCc adsorbed to mixed alkanethiol monolayers on gold electrodes shows slower ET for D(2)O-grown yCc films than for H(2)O-grown films (k(H) = 800 s(-1); k(D) = 540 s(-1) at 20 degrees C). Soaking H(2)O- or D(2)O-grown films in the counter solvent produces an immediate inverse isotope effect that diminishes over hours until the ET rate reaches that found in the counter solvent. Thus, D(2)O substitution perturbs interactions and ET between yCc and either CcP or electrode films. The effects derive from slow exchanging protons or solvent molecules that in the crystal produce only small structural changes.


Subject(s)
Cytochrome-c Peroxidase/chemistry , Cytochromes c/chemistry , Metalloporphyrins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Zinc/chemistry , Crystallization , Crystallography, X-Ray , Deuterium Oxide/chemistry , Electrochemistry , Electrodes , Kinetics , Models, Molecular , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...