Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cell ; 162(1): 59-71, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26095252

ABSTRACT

eIF4E, the major cap-binding protein, has long been considered limiting for translating the mammalian genome. However, the eIF4E dose requirement at an organismal level remains unexplored. By generating an Eif4e haploinsufficient mouse, we found that a 50% reduction in eIF4E expression, while compatible with normal development and global protein synthesis, significantly impeded cellular transformation. Genome-wide translational profiling uncovered a translational program induced by oncogenic transformation and revealed a critical role for the dose of eIF4E, specifically in translating a network of mRNAs enriched for a unique 5' UTR signature. In particular, we demonstrate that the dose of eIF4E is essential for translating mRNAs that regulate reactive oxygen species, fueling transformation and cancer cell survival in vivo. Our findings indicate eIF4E is maintained at levels in excess for normal development that are hijacked by cancer cells to drive a translational program supporting tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic , Embryo, Mammalian/metabolism , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Dosage , 5' Untranslated Regions , Animals , Carcinogenesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Biosynthesis , Reactive Oxygen Species/metabolism
2.
Mol Cell ; 73(6): 1162-1173.e5, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30712990

ABSTRACT

The MHC class I antigen presentation system enables T cell immunosurveillance of cancers and viruses. A substantial fraction of the immunopeptidome derives from rapidly degraded nascent polypeptides (DRiPs). By knocking down each of the 80 ribosomal proteins, we identified proteins that modulate peptide generation without altering source protein expression. We show that 60S ribosomal proteins L6 (RPL6) and RPL28, which are adjacent on the ribosome, play opposite roles in generating an influenza A virus-encoded peptide. Depleting RPL6 decreases ubiquitin-dependent peptide presentation, whereas depleting RPL28 increases ubiquitin-dependent and -independent peptide presentation. 40S ribosomal protein S28 (RPS28) knockdown increases total peptide supply in uninfected cells by increasing DRiP synthesis from non-canonical translation of "untranslated" regions and non-AUG start codons and sensitizes tumor cells for T cell targeting. Our findings raise the possibility of modulating immunosurveillance by pharmaceutical targeting ribosomes.


Subject(s)
Antigen Presentation , Histocompatibility Antigens Class I/biosynthesis , Ribosomal Proteins/metabolism , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism , T-Lymphocytes/metabolism , Animals , Cell Line, Tumor , Coculture Techniques , HEK293 Cells , Histocompatibility Antigens Class I/immunology , Host-Pathogen Interactions , Humans , Immunologic Surveillance , Influenza A virus/immunology , Influenza A virus/pathogenicity , Melanoma/immunology , Melanoma/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Ribosomal Proteins/genetics , Ribosome Subunits, Large, Eukaryotic/genetics , Ribosome Subunits, Small, Eukaryotic/genetics , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/virology
3.
Front Oncol ; 13: 1206561, 2023.
Article in English | MEDLINE | ID: mdl-37601686

ABSTRACT

During their quest for growth, adaptation, and survival, cancer cells create a favorable environment through the manipulation of normal cellular mechanisms. They increase anabolic processes, including protein synthesis, to facilitate uncontrolled proliferation and deplete the tumor microenvironment of resources. As a dynamic adaptation to the self-imposed oncogenic stress, cancer cells promptly hijack translational control to alter gene expression. Rewiring the cellular proteome shifts the phenotypic balance between growth and adaptation to promote therapeutic resistance and cancer cell survival. The integrated stress response (ISR) is a key translational program activated by oncogenic stress that is utilized to fine-tune protein synthesis and adjust to environmental barriers. Here, we focus on the role of ISR signaling for driving cancer progression. We highlight mechanisms of regulation for distinct mRNA translation downstream of the ISR, expand on oncogenic signaling utilizing the ISR in response to environmental stresses, and pinpoint the impact this has for cancer cell plasticity during resistance to therapy. There is an ongoing need for innovative drug targets in cancer treatment, and modulating ISR activity may provide a unique avenue for clinical benefit.

4.
Cell Metab ; 35(3): 517-534.e8, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36804058

ABSTRACT

The efficacy of immunotherapy is limited by the paucity of T cells delivered and infiltrated into the tumors through aberrant tumor vasculature. Here, we report that phosphoglycerate dehydrogenase (PHGDH)-mediated endothelial cell (EC) metabolism fuels the formation of a hypoxic and immune-hostile vascular microenvironment, driving glioblastoma (GBM) resistance to chimeric antigen receptor (CAR)-T cell immunotherapy. Our metabolome and transcriptome analyses of human and mouse GBM tumors identify that PHGDH expression and serine metabolism are preferentially altered in tumor ECs. Tumor microenvironmental cues induce ATF4-mediated PHGDH expression in ECs, triggering a redox-dependent mechanism that regulates endothelial glycolysis and leads to EC overgrowth. Genetic PHGDH ablation in ECs prunes over-sprouting vasculature, abrogates intratumoral hypoxia, and improves T cell infiltration into the tumors. PHGDH inhibition activates anti-tumor T cell immunity and sensitizes GBM to CAR T therapy. Thus, reprogramming endothelial metabolism by targeting PHGDH may offer a unique opportunity to improve T cell-based immunotherapy.


Subject(s)
Glioblastoma , Receptors, Chimeric Antigen , Animals , Mice , Humans , Glioblastoma/therapy , Glioblastoma/metabolism , Phosphoglycerate Dehydrogenase/metabolism , Cell Line, Tumor , Immunotherapy, Adoptive , T-Lymphocytes/metabolism , Tumor Microenvironment
5.
J Biol Chem ; 286(8): 6791-800, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21177857

ABSTRACT

Protein synthesis is a key regulated cellular process that links nutrient availability and organismal growth. It has long been known that some cellular proteins continue to be synthesized under conditions where global translation is severely compromised. One prominent example is the selective translation of heat shock proteins (Hsps) under stress conditions. Although the transcriptional regulation of Hsp genes has been well established, neither the specific translation-promoting features nor the regulatory mechanism of the translation machinery have been clearly defined. Here we show that the stress-induced preferential translation of Hsp70 mRNA is negatively regulated by PI3K-mTORC1 signaling. Despite the transcriptional up-regulation, the translation of Hsp70 mRNA is deficient in cells lacking tuberous sclerosis complex 2. Conversely, Hsp70 synthesis is enhanced under the reduced PI3K-mTORC1 signaling. We found that the 5' UTR of Hsp70 mRNA contributes to cap-independent translation without exhibiting typical features of internal ribosome entry site. Our findings imply a plausible mechanism for how persistent PI3K-mTORC1 signaling favors the development of age-related pathologies by attenuating stress resistance.


Subject(s)
HSP70 Heat-Shock Proteins/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Protein Biosynthesis/physiology , Proteins/metabolism , RNA Caps/metabolism , Stress, Physiological/physiology , 5' Untranslated Regions , Animals , Cell Line , HSP70 Heat-Shock Proteins/genetics , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes , Phosphatidylinositol 3-Kinases/genetics , Proteins/genetics , RNA Caps/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases
6.
iScience ; 24(7): 102748, 2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34278258

ABSTRACT

The cap-binding protein eukaryotic initiation factor 4E (eIF4E) promotes translation of mRNAs associated with proliferation and survival and is an attractive target for cancer therapeutics. Here, we used Eif4e germline and conditional knockout models to assess the impact of reduced Eif4e gene dosage on B-cell leukemogenesis compared to effects on normal pre-B and mature B-cell function. Using a BCR-ABL-driven pre-B-cell leukemia model, we find that loss of one allele of Eif4e impairs transformation and reduces fitness in competition assays in vitro and in vivo. In contrast, reduced Eif4e gene dosage had no significant effect on development of pre-B and mature B cells or on survival or proliferation of non-transformed B lineage cells. These results demonstrate that inhibition of eIF4E could be a new therapeutic tool for pre-B-cell leukemia while preserving development and function of normal B cells.

7.
Nat Metab ; 3(2): 244-257, 2021 02.
Article in English | MEDLINE | ID: mdl-33619378

ABSTRACT

Obesity is a global epidemic leading to increased mortality and susceptibility to comorbidities, with few viable therapeutic interventions. A hallmark of disease progression is the ectopic deposition of lipids in the form of lipid droplets in vital organs such as the liver. However, the mechanisms underlying the dynamic storage and processing of lipids in peripheral organs remain an outstanding question. Here, we show an unexpected function for the major cap-binding protein, eIF4E, in high-fat-diet-induced obesity. In response to lipid overload, select networks of proteins involved in fat deposition are altered in eIF4E-deficient mice. Specifically, distinct messenger RNAs involved in lipid metabolic processing and storage pathways are enhanced at the translation level by eIF4E. Failure to translationally upregulate these mRNAs results in increased fatty acid oxidation, which enhances energy expenditure. We further show that inhibition of eIF4E phosphorylation genetically-and by a potent clinical compound-restrains weight gain following intake of a high-fat diet. Together, our study uncovers translational control of lipid processing as a driver of high-fat-diet-induced weight gain and provides a pharmacological target to treat obesity.


Subject(s)
Adipogenesis/genetics , Diet, High-Fat , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Lipid Metabolism/genetics , Obesity/genetics , Adipocytes/pathology , Animals , Energy Metabolism , Fatty Acids/metabolism , Lipid Droplets , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/pathology , Oxidation-Reduction , Phosphorylation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
9.
Nat Cell Biol ; 21(7): 889-899, 2019 07.
Article in English | MEDLINE | ID: mdl-31263264

ABSTRACT

The c-Myc oncogene drives malignant progression and induces robust anabolic and proliferative programmes leading to intrinsic stress. The mechanisms enabling adaptation to MYC-induced stress are not fully understood. Here we reveal an essential role for activating transcription factor 4 (ATF4) in survival following MYC activation. MYC upregulates ATF4 by activating general control nonderepressible 2 (GCN2) kinase through uncharged transfer RNAs. Subsequently, ATF4 co-occupies promoter regions of over 30 MYC-target genes, primarily those regulating amino acid and protein synthesis, including eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1), a negative regulator of translation. 4E-BP1 relieves MYC-induced proteotoxic stress and is essential to balance protein synthesis. 4E-BP1 activity is negatively regulated by mammalian target of rapamycin complex 1 (mTORC1)-dependent phosphorylation and inhibition of mTORC1 signalling rescues ATF4-deficient cells from MYC-induced endoplasmic reticulum stress. Acute deletion of ATF4 significantly delays MYC-driven tumour progression and increases survival in mouse models. Our results establish ATF4 as a cellular rheostat of MYC activity, which ensures that enhanced translation rates are compatible with survival and tumour progression.


Subject(s)
Activating Transcription Factor 4/genetics , Genes, myc/genetics , Transcriptional Activation/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Endoplasmic Reticulum Stress/genetics , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Transgenic , Phosphoproteins/genetics , Phosphorylation , Protein Biosynthesis/physiology , TOR Serine-Threonine Kinases/metabolism
10.
Cancer Cell ; 33(1): 91-107.e6, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29316436

ABSTRACT

KRAS is a regulator of the nutrient stress response in non-small-cell lung cancer (NSCLC). Induction of the ATF4 pathway during nutrient depletion requires AKT and NRF2 downstream of KRAS. The tumor suppressor KEAP1 strongly influences the outcome of activation of this pathway during nutrient stress; loss of KEAP1 in KRAS mutant cells leads to apoptosis. Through ATF4 regulation, KRAS alters amino acid uptake and asparagine biosynthesis. The ATF4 target asparagine synthetase (ASNS) contributes to apoptotic suppression, protein biosynthesis, and mTORC1 activation. Inhibition of AKT suppressed ASNS expression and, combined with depletion of extracellular asparagine, decreased tumor growth. Therefore, KRAS is important for the cellular response to nutrient stress, and ASNS represents a promising therapeutic target in KRAS mutant NSCLC.


Subject(s)
Activating Transcription Factor 4/metabolism , Asparaginase/pharmacology , Aspartate-Ammonia Ligase/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Homeostasis/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice
11.
Sci Transl Med ; 10(439)2018 05 02.
Article in English | MEDLINE | ID: mdl-29720449

ABSTRACT

Oncogenic lesions up-regulate bioenergetically demanding cellular processes, such as protein synthesis, to drive cancer cell growth and continued proliferation. However, the hijacking of these key processes by oncogenic pathways imposes onerous cell stress that must be mitigated by adaptive responses for cell survival. The mechanism by which these adaptive responses are established, their functional consequences for tumor development, and their implications for therapeutic interventions remain largely unknown. Using murine and humanized models of prostate cancer (PCa), we show that one of the three branches of the unfolded protein response is selectively activated in advanced PCa. This adaptive response activates the phosphorylation of the eukaryotic initiation factor 2-α (P-eIF2α) to reset global protein synthesis to a level that fosters aggressive tumor development and is a marker of poor patient survival upon the acquisition of multiple oncogenic lesions. Using patient-derived xenograft models and an inhibitor of P-eIF2α activity, ISRIB, our data show that targeting this adaptive brake for protein synthesis selectively triggers cytotoxicity against aggressive metastatic PCa, a disease for which presently there is no cure.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Prostatic Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Humans , Male , Mice , Prostatic Neoplasms/drug therapy , Unfolded Protein Response/drug effects , Unfolded Protein Response/physiology
13.
Clin Cancer Res ; 23(12): 3045-3052, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28007777

ABSTRACT

Purpose: mTOR regulates many normal physiological processes and when hyperactive can drive numerous cancers and human diseases. However, it is very challenging to detect and quantify mTOR signaling noninvasively in clinically relevant animal models of disease or man. We hypothesized that a nuclear imaging tool measuring intracellular mTOR activity could address this unmet need.Experimental Design: Although the biochemical activity of mTOR is not directly amenable to nuclear imaging probe development, we show that the transferrin receptor can be used to indirectly measure intracellular changes in mTOR activity.Results: After verifying that the uptake of radiolabeled transferrin (the soluble ligand of the transferrin receptor) is stimulated by active mTORC1 in vitro, we showed that 89Zr-labeled transferrin (Tf) can measure mTORC1 signaling dynamics in normal and cancerous mouse tissues with PET. Finally, we show that 89Zr-Tf can detect the upregulation of mTORC1 by tumor cells to escape the antitumor effects of a standard-of-care antiandrogen, which is to our knowledge the first example of applying PET to interrogate the biology of treatment resistant cancer.Conclusions: In summary, we have developed the first quantitative assay to provide a comprehensive measurement of mTOR signaling dynamics in vivo, in specific normal tissues, and during tumor development in genetically engineered animal models using a nuclear imaging tool that is readily translatable to man. Clin Cancer Res; 23(12); 3045-52. ©2016 AACR.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/isolation & purification , Molecular Imaging/methods , TOR Serine-Threonine Kinases/isolation & purification , Transferrin/chemistry , Animals , Cell Line, Tumor , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Positron-Emission Tomography , Radiopharmaceuticals/chemistry , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays , Zirconium/chemistry
14.
Nat Struct Mol Biol ; 22(5): 404-10, 2015 May.
Article in English | MEDLINE | ID: mdl-25866880

ABSTRACT

In response to stress, cells attenuate global protein synthesis but permit efficient translation of mRNAs encoding heat-shock proteins (HSPs). Although decades have passed since the first description of the heat-shock response, how cells achieve translational control of HSP synthesis remains enigmatic. Here we report an unexpected role for mitochondrial ribosomal protein L18 (MRPL18) in the mammalian cytosolic stress response. MRPL18 bears a downstream CUG start codon and generates a cytosolic isoform in a stress-dependent manner. Cytosolic MRPL18 incorporates into the 80S ribosome and facilitates ribosome engagement on mRNAs selected for translation during stress. MRPL18 knockdown has minimal effects on mitochondrial function but substantially dampens cytosolic HSP expression at the level of translation. Our results uncover a hitherto-uncharacterized stress-adaptation mechanism in mammalian cells, which involves formation of a 'hybrid' ribosome responsible for translational regulation during the cytosolic stress response.


Subject(s)
Heat-Shock Proteins/biosynthesis , Protein Biosynthesis/genetics , Ribosomal Proteins/genetics , Stress, Physiological/physiology , Cell Line, Tumor , Codon, Initiator/genetics , Gene Expression Regulation , HeLa Cells , Heat-Shock Response/genetics , Humans , Phosphorylation , Protein Isoforms/genetics , RNA Interference , RNA, Small Interfering , Ribosomal Proteins/metabolism , Ribosomes/genetics , eIF-2 Kinase/metabolism
15.
Sci Signal ; 6(271): ra24, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23592839

ABSTRACT

The discovery that rapamycin extends the life span of diverse organisms has triggered many studies aimed at identifying the underlying molecular mechanisms. Mammalian target of rapamycin complex 1 (mTORC1) regulates cell growth and may regulate organismal aging by controlling mRNA translation. However, how inhibiting mTORC1 and decreasing protein synthesis can extend life span remains an unresolved issue. We showed that constitutively active mTORC1 signaling increased general protein synthesis but unexpectedly reduced the quality of newly synthesized polypeptides. We demonstrated that constitutively active mTORC1 decreased translation fidelity by increasing the speed of ribosomal elongation. Conversely, rapamycin treatment restored the quality of newly synthesized polypeptides mainly by slowing the rate of ribosomal elongation. We also found distinct roles for mTORC1 downstream targets in maintaining protein homeostasis. Loss of S6 kinases, but not 4E-BP family proteins, which are both involved in regulation of translation, attenuated the effects of rapamycin on the quality of newly translated proteins. Our results reveal a mechanistic connection between mTORC1 and protein quality, highlighting the central role of nutrient signaling in growth and aging.


Subject(s)
Homeostasis/physiology , Multiprotein Complexes/metabolism , Protein Biosynthesis/physiology , Protein Stability , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , DNA Primers/genetics , Humans , Immunoblotting , Luciferases , Mechanistic Target of Rapamycin Complex 1 , Mice , Models, Biological , Reverse Transcriptase Polymerase Chain Reaction , Ribosomes/physiology
16.
Cell Cycle ; 10(12): 1940-7, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21555915

ABSTRACT

A proper balance between synthesis, maturation and degradation of cellular proteins is crucial for cells to maintain physiological functions. The costly process of protein synthesis is tightly coupled to energy status and nutrient levels by the mammalian target of rapamycin (mTOR), whereas the quality of newly synthesized polypeptides is largely maintained by molecular chaperones and the ubiquitin-proteasome system. There is a wealth of evidence indicating close ties between the nutrient signaling pathway and the intracellular stress response. Dysregulation of both systems has been implicated in aging and age-associated pathologies. In this review, we describe molecular mechanisms underlying the connection between mTOR and the chaperone network and discuss the importance of their functional interaction in growth and aging.


Subject(s)
Homeostasis , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Humans , Molecular Chaperones , Proteasome Endopeptidase Complex , Ribosomes
SELECTION OF CITATIONS
SEARCH DETAIL