Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 228
Filter
Add more filters

Publication year range
1.
Cell ; 180(3): 411-426.e16, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31928844

ABSTRACT

Stress granules are condensates of non-translating mRNAs and proteins involved in the stress response and neurodegenerative diseases. Stress granules form in part through intermolecular RNA-RNA interactions, and to better understand how RNA-based condensation occurs, we demonstrate that RNA is effectively recruited to the surfaces of RNA or RNP condensates in vitro. We demonstrate that, through ATP-dependent RNA binding, the DEAD-box protein eIF4A reduces RNA condensation in vitro and limits stress granule formation in cells. This defines a function for eIF4A to limit intermolecular RNA-RNA interactions in cells. These results establish an important role for eIF4A, and potentially other DEAD-box proteins, as ATP-dependent RNA chaperones that limit the condensation of RNA, analogous to the function of proteins like HSP70 in combatting protein aggregates.


Subject(s)
DEAD-box RNA Helicases/metabolism , Eukaryotic Initiation Factor-4A/metabolism , Eukaryotic Initiation Factor-4F/metabolism , RNA Helicases/metabolism , RNA, Fungal/metabolism , Ribonucleoproteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphate/metabolism , HeLa Cells , Humans , Microscopy, Confocal , Protein Binding , RNA, Fungal/isolation & purification , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Time-Lapse Imaging
2.
Annu Rev Biochem ; 88: 307-335, 2019 06 20.
Article in English | MEDLINE | ID: mdl-31220979

ABSTRACT

The stage at which ribosomes are recruited to messenger RNAs (mRNAs) is an elaborate and highly regulated phase of protein synthesis. Upon completion of this step, a ribosome is positioned at an appropriate initiation codon and primed to synthesize the encoded polypeptide product. In most circumstances, this step commits the ribosome to translate the mRNA. We summarize the knowledge regarding the initiation factors implicated in this activity as well as review different mechanisms by which this process is conducted.


Subject(s)
Eukaryota/metabolism , Peptide Chain Initiation, Translational , Peptide Initiation Factors/metabolism , RNA, Messenger/metabolism , Ribosomes/metabolism , Eukaryota/genetics , Humans
3.
Proc Natl Acad Sci U S A ; 121(4): e2318093121, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38232291

ABSTRACT

In this study, we aimed to address the current limitations of therapies for macro-metastatic triple-negative breast cancer (TNBC) and provide a therapeutic lead that overcomes the high degree of heterogeneity associated with this disease. Specifically, we focused on well-documented but clinically underexploited cancer-fueling perturbations in mRNA translation as a potential therapeutic vulnerability. We therefore developed an orally bioavailable rocaglate-based molecule, MG-002, which hinders ribosome recruitment and scanning via unscheduled and non-productive RNA clamping by the eukaryotic translation initiation factor (eIF) 4A RNA helicase. We demonstrate that MG-002 potently inhibits mRNA translation and primary TNBC tumor growth without causing overt toxicity in mice. Importantly, given that metastatic spread is a major cause of mortality in TNBC, we show that MG-002 attenuates metastasis in pre-clinical models. We report on MG-002, a rocaglate that shows superior properties relative to existing eIF4A inhibitors in pre-clinical models. Our study also paves the way for future clinical trials exploring the potential of MG-002 in TNBC and other oncological indications.


Subject(s)
RNA Helicases , Triple Negative Breast Neoplasms , Humans , Animals , Mice , RNA Helicases/genetics , RNA Helicases/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Protein Biosynthesis , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Ribosomes/metabolism
4.
Nature ; 586(7829): 407-411, 2020 10.
Article in English | MEDLINE | ID: mdl-33029009

ABSTRACT

To survive in a dynamic environment, animals need to identify and appropriately respond to stimuli that signal danger1. Survival also depends on suppressing the threat-response during a stimulus that predicts the absence of threat (safety)2-5. An understanding of the biological substrates of emotional memories during a task in which animals learn to flexibly execute defensive responses to a threat-predictive cue and a safety cue is critical for developing treatments for memory disorders such as post-traumatic stress disorder5. The centrolateral amygdala is an important node in the neuronal circuit that mediates defensive responses6-9, and a key brain area for processing and storing threat memories. Here we applied intersectional chemogenetic strategies to inhibitory neurons in the centrolateral amygdala of mice to block cell-type-specific translation programs that are sensitive to depletion of eukaryotic initiation factor 4E (eIF4E) and phosphorylation of eukaryotic initiation factor 2α (p-eIF2α). We show that de novo translation in somatostatin-expressing inhibitory neurons in the centrolateral amygdala is necessary for the long-term storage of conditioned-threat responses, whereas de novo translation in protein kinase Cδ-expressing inhibitory neurons in the centrolateral amygdala is necessary for the inhibition of a conditioned response to a safety cue. Our results provide insight into the role of de novo protein synthesis in distinct inhibitory neuron populations in the centrolateral amygdala during the consolidation of long-term memories.


Subject(s)
Amygdala/cytology , Amygdala/physiology , Emotions , Memory/physiology , Neural Inhibition , Neurons/physiology , Animals , Conditioning, Psychological , Cues , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Fear/physiology , Female , Heterotrimeric GTP-Binding Proteins/metabolism , Male , Mice , Protein Biosynthesis , RNA Caps/genetics , RNA Caps/metabolism , Signal Transduction , Somatostatin/metabolism
5.
RNA ; 29(6): 826-835, 2023 06.
Article in English | MEDLINE | ID: mdl-36882295

ABSTRACT

Inhibition of eukaryotic translation initiation through unscheduled RNA clamping of the DEAD-box (DDX) RNA helicases eIF4A1 and eIF4A2 has been documented for pateamine A (PatA) and rocaglates-two structurally different classes of compounds that share overlapping binding sites on eIF4A. Clamping of eIF4A to RNA causes steric blocks that interfere with ribosome binding and scanning, rationalizing the potency of these molecules since not all eIF4A molecules need to be engaged to elicit a biological effect. In addition to targeting translation, PatA and analogs have also been shown to target the eIF4A homolog, eIF4A3-a helicase necessary for exon junction complex (EJC) formation. EJCs are deposited on mRNAs upstream of exon-exon junctions and, when present downstream from premature termination codons (PTCs), participate in nonsense-mediated decay (NMD), a quality control mechanism aimed at preventing the production of dominant-negative or gain-of-function polypeptides from faulty mRNA transcripts. We find that rocaglates can also interact with eIF4A3 to induce RNA clamping. Rocaglates also inhibit EJC-dependent NMD in mammalian cells, but this does not appear to be due to induced eIF4A3-RNA clamping, but rather a secondary consequence of translation inhibition incurred by clamping eIF4A1 and eIF4A2 to mRNA.


Subject(s)
Nonsense Mediated mRNA Decay , RNA , Animals , RNA/metabolism , RNA, Messenger/metabolism , Codon, Nonsense , Exons , Eukaryotic Initiation Factor-4A/chemistry , Mammals/genetics
6.
Proc Natl Acad Sci U S A ; 119(32): e2204539119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35878012

ABSTRACT

Viruses evade the innate immune response by suppressing the production or activity of cytokines such as type I interferons (IFNs). Here we report the discovery of a mechanism by which the SARS-CoV-2 virus coopts an intrinsic cellular machinery to suppress the production of the key immunostimulatory cytokine IFN-ß. We reveal that the SARS-CoV-2 encoded nonstructural protein 2 (NSP2) directly interacts with the cellular GIGYF2 protein. This interaction enhances the binding of GIGYF2 to the mRNA cap-binding protein 4EHP, thereby repressing the translation of the Ifnb1 mRNA. Depletion of GIGYF2 or 4EHP significantly enhances IFN-ß production, which inhibits SARS-CoV-2 replication. Our findings reveal a target for rescuing the antiviral innate immune response to SARS-CoV-2 and other RNA viruses.


Subject(s)
COVID-19 , Carrier Proteins , Interferon Type I , Viral Nonstructural Proteins , COVID-19/genetics , Carrier Proteins/metabolism , Cell Line , Eukaryotic Initiation Factor-4E/metabolism , Humans , Immunity, Innate , Interferon Type I/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , SARS-CoV-2 , Viral Nonstructural Proteins/metabolism , Virus Replication
7.
Immunity ; 42(1): 41-54, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25607458

ABSTRACT

Naive T cells undergo metabolic reprogramming to support the increased energetic and biosynthetic demands of effector T cell function. However, how nutrient availability influences T cell metabolism and function remains poorly understood. Here we report plasticity in effector T cell metabolism in response to changing nutrient availability. Activated T cells were found to possess a glucose-sensitive metabolic checkpoint controlled by the energy sensor AMP-activated protein kinase (AMPK) that regulated mRNA translation and glutamine-dependent mitochondrial metabolism to maintain T cell bioenergetics and viability. T cells lacking AMPKα1 displayed reduced mitochondrial bioenergetics and cellular ATP in response to glucose limitation in vitro or pathogenic challenge in vivo. Finally, we demonstrated that AMPKα1 is essential for T helper 1 (Th1) and Th17 cell development and primary T cell responses to viral and bacterial infections in vivo. Our data highlight AMPK-dependent regulation of metabolic homeostasis as a key regulator of T cell-mediated adaptive immunity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/metabolism , AMP-Activated Protein Kinases/genetics , Adaptation, Physiological/immunology , Animals , Cells, Cultured , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Energy Metabolism , Glucose/metabolism , Glutamine/metabolism , Humans , Immunomodulation , Lymphocyte Activation/genetics , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Protein Biosynthesis/genetics
8.
RNA ; 27(6): 676-682, 2021 06.
Article in English | MEDLINE | ID: mdl-33762403

ABSTRACT

Circular (circ) RNA expression vectors are used as a method of identifying and characterizing RNA sequences that harbor internal ribosome entry site (IRES) activity. During the course of developing a vector series tailored for IRES discovery, we found evidence for the occurrence of trans-spliced mRNAs arising when sequences with promoter activity were embedded between the upstream CTD and downstream NTD exons of the pre-mRNA. These trans-spliced products regenerate the same open reading frame expected from a circRNA and can lead to false-positive signals in screens relying on circRNA expression vectors for IRES discovery. Our results caution against interpretations of IRES activity solely based on results obtained from circRNA expression vectors.


Subject(s)
Internal Ribosome Entry Sites , RNA, Circular/metabolism , Trans-Splicing , Animals , Gene Expression , Genetic Vectors/genetics , Humans , Promoter Regions, Genetic , RNA Precursors/genetics
9.
Biochem Soc Trans ; 51(1): 113-124, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36661272

ABSTRACT

Fundamental studies unraveled the role of eukaryotic initiation factor (eIF) 4E in mRNA translation and its control. Under physiological conditions, regulation of translation by eIF4E is essential to cellular homeostasis. Under stress, gene flow information is parsed by eIF4E to support adaptive mechanisms that favor cell survival. Dysregulated eIF4E activity fuels tumor formation and progression and modulates response to therapy. Thus, there has been heightened interest in understanding eIF4E function in controlling gene expression as well as developing strategies to block its activity to treat disease.


Subject(s)
Eukaryotic Initiation Factor-4E , Neoplasms , Humans , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/metabolism , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , Phosphorylation
10.
RNA ; 26(5): 541-549, 2020 05.
Article in English | MEDLINE | ID: mdl-32014999

ABSTRACT

The PI3K/Akt/mTOR kinase pathway is extensively deregulated in human cancers. One critical node under regulation of this signaling axis is eukaryotic initiation factor (eIF) 4F, a complex involved in the control of translation initiation rates. eIF4F-dependent addictions arise during tumor initiation and maintenance due to increased eIF4F activity-generally in response to elevated PI3K/Akt/mTOR signaling flux. There is thus much interest in exploring eIF4F as a small molecule target for the development of new anticancer drugs. The DEAD-box RNA helicase, eIF4A, is an essential subunit of eIF4F, and several potent small molecules (rocaglates, hippuristanol, pateamine A) affecting its activity have been identified and shown to demonstrate anticancer activity in vitro and in vivo in preclinical models. Recently, a number of new small molecules have been reported as having the capacity to target and inhibit eIF4A. Here, we undertook a comparative analysis of their biological activity and specificity relative to the eIF4A inhibitor, hippuristanol.


Subject(s)
Antineoplastic Agents/chemistry , Eukaryotic Initiation Factor-4A/chemistry , Neoplasms/drug therapy , Small Molecule Libraries/chemistry , Sterols/chemistry , Antineoplastic Agents/pharmacology , Benzofurans/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Epoxy Compounds/chemistry , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4F/antagonists & inhibitors , Eukaryotic Initiation Factor-4F/chemistry , Humans , Macrolides/chemistry , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-akt/genetics , Small Molecule Libraries/pharmacology , Sterols/pharmacology , TOR Serine-Threonine Kinases/genetics , Thiazoles/chemistry
11.
Mol Cell ; 56(1): 79-89, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25280105

ABSTRACT

In animals, key functions of microRNA-induced silencing complex (miRISC) are translational repression and deadenylation followed by mRNA decay. While miRISC represses translation initiation, it is poorly understood how miRISC exerts this function. Here we assessed the effect of miRISC on synergistic recruitment of translation initiation factors to target mRNAs by using direct biochemical assays. We show that miRISC promotes eIF4AI and eIF4AII release from target mRNAs prior to dissociation of eIF4E and eIF4G in a deadenylation-independent manner. Strikingly, miRISC-induced release of eIF4AI and eIF4AII from target mRNAs and miRISC-induced inhibition of cap-dependent translation can both be counteracted by the RNA-binding protein HuD via a direct interaction of HuD with eIF4A. Furthermore, the pharmacological eIF4A inhibitor silvestrol, which locks eIF4A on mRNAs, conferred resistance to miRNA-mediated translational repression. In summary, we propose that both eIF4AI and eIF4AII are functionally important targets in miRISC-mediated translation control.


Subject(s)
Eukaryotic Initiation Factor-4A/metabolism , MicroRNAs/physiology , Models, Genetic , RNA, Messenger/metabolism , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/genetics , HEK293 Cells , Humans , RNA-Induced Silencing Complex/physiology , Transcription Initiation, Genetic , Triterpenes/pharmacology
12.
Cell Mol Life Sci ; 78(19-20): 6709-6719, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34559254

ABSTRACT

Eukaryotic initiation factor (eIF) 4F plays a central role in the ribosome recruitment phase of cap-dependent translation. This heterotrimeric complex consists of a cap binding subunit (eIF4E), a DEAD-box RNA helicase (eIF4A), and a large bridging protein (eIF4G). In mammalian cells, there are two genes encoding eIF4A (eIF4A1 and eIF4A2) and eIF4G (eIF4G1 and eIF4G3) paralogs that can assemble into eIF4F complexes. To query the essential nature of the eIF4F subunits in normal development, we used CRISPR/Cas9 to generate mouse strains with targeted ablation of each gene encoding the different eIF4F subunits. We find that Eif4e, Eif4g1, and Eif4a1 are essential for viability in the mouse, whereas Eif4g3 and Eif4a2 are not. However, Eif4g3 and Eif4a2 do play essential roles in spermatogenesis. Crossing of these strains to the lymphoma-prone Eµ-Myc mouse model revealed that heterozygosity at the Eif4e or Eif4a1 loci significantly delayed tumor onset. Lastly, tumors derived from Eif4e∆38 fs/+/Eµ-Myc or Eif4a1∆5 fs/+/Eµ-Myc mice show increased sensitivity to the chemotherapeutic agent doxorubicin, in vivo. Our study reveals that eIF4A2 and eIF4G3 play non-essential roles in gene expression regulation during embryogenesis; whereas reductions in eIF4E or eIF4A1 levels are protective against tumor development in a murine Myc-driven lymphoma setting.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Eukaryotic Initiation Factor-4F/genetics , Animals , Female , Gene Expression Regulation/genetics , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Subunits/genetics , Spermatogenesis/genetics
13.
Nucleic Acids Res ; 48(15): 8562-8575, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32749456

ABSTRACT

Eukaryotic cellular mRNAs possess a 5' cap structure (m7GpppN) which plays a critical role in translation initiation mediated by eukaryotic initiation factor (eIF) 4F. The heterotrimeric eIF4F complex possesses several activities imparted by its subunits that include cap recognition (by eIF4E), RNA unwinding (eIF4A), and factor/ribosome recruitment (eIF4G). Mammalian cells have paralogs of all three eIF4F subunits and it remains an open question as to whether these all can participate in the process of ribosome recruitment. To query the activities of the eIF4F subunits in translation initiation, we adopted an RNA-tethering assay in which select subunits are recruited to a specific address on a reporter mRNA template. We find that all eIF4F subunits can participate in the initiation process. Based on eIF4G:eIF4A structural information, we also designed obligate dimer pairs to probe the activity of all combinations of eIF4G and eIF4A paralogs. We demonstrate that both eIF4GI and eIF4GII can associate with either eIF4A1 or eIF4A2 to recruit ribosomes to mRNA templates. In combination with eIF4E and eIF4E3, our results indicate the presence of up to eight eIF4F complexes that can operate in translation initiation.


Subject(s)
Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4F/genetics , Eukaryotic Initiation Factor-4G/genetics , Protein Biosynthesis , Amino Acid Sequence/genetics , Animals , Eukaryotic Initiation Factor-4E/chemistry , Eukaryotic Initiation Factor-4F/chemistry , HEK293 Cells , Humans , Mice , Protein Binding/genetics , RNA Cap Analogs/genetics , RNA Caps/genetics , RNA, Messenger/genetics , Ribosomes/genetics
14.
Nucleic Acids Res ; 48(9): 4643-4657, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32282904

ABSTRACT

We report on the synthesis of siRNAs containing both 2'-5'- and 3'-5'-internucleotide linkages and their effects on siRNA structure, function, and interaction with RNAi proteins. Screening of these siRNAs against their corresponding mRNA targets showed that 2'-5' linkages were well tolerated in the sense strand, but only at a few positions in the antisense strand. Extensive modification of the antisense strand minimally affected 5'-phosphorylation of the siRNA by kinases, however, it negatively affected siRNA loading into human AGO2. Modelling and molecular dynamics simulations were fully consistent with these findings. Furthermore, our studies indicated that the presence of a single 5'p-rN1-(2'-5')-N2 unit in the antisense strand does not alter the 'clover leaf' bend and sugar puckers that are critical for anchoring the 5'-phosphate to Ago 2 MID domain. Importantly, 2'-5'-linkages had the added benefit of abrogating immune-stimulatory activity of siRNAs. Together, these results demonstrate that 2'-5'/3'-5'-modified siRNAs, when properly designed, can offer an efficient new class of siRNAs with diminished immune-stimulatory responses.


Subject(s)
RNA Interference , RNA, Small Interfering/chemistry , Argonaute Proteins/metabolism , Carbohydrate Conformation , HeLa Cells , Humans , Luciferases, Firefly/genetics , Molecular Dynamics Simulation , Nucleic Acid Conformation , RNA, Small Interfering/chemical synthesis , RNA, Small Interfering/immunology , Tumor Suppressor Protein p53/genetics
15.
Nucleic Acids Res ; 48(17): 9521-9537, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32766783

ABSTRACT

Hippuristanol (Hipp) is a natural product that selectively inhibits protein synthesis by targeting eukaryotic initiation factor (eIF) 4A, a DEAD-box RNA helicase required for ribosome recruitment to mRNA templates. Hipp binds to the carboxyl-terminal domain of eIF4A, locks it in a closed conformation, and inhibits its RNA binding. The dependencies of mRNAs for eIF4A during initiation is contingent on the degree of secondary structure within their 5' leader region. Interest in targeting eIF4A therapeutically in cancer and viral-infected settings stems from the dependencies that certain cellular (e.g. pro-oncogenic, pro-survival) and viral mRNAs show towards eIF4A. Using a CRISPR/Cas9-based variomics screen, we identify functional EIF4A1 Hipp-resistant alleles, which in turn allowed us to link the translation-inhibitory and cytotoxic properties of Hipp to eIF4A1 target engagement. Genome-wide translational profiling in the absence or presence of Hipp were undertaken and our validation studies provided insight into the structure-activity relationships of eIF4A-dependent mRNAs. We find that mRNA 5' leader length, overall secondary structure and cytosine content are defining features of Hipp-dependent mRNAs.


Subject(s)
5' Untranslated Regions , Drug Resistance, Neoplasm/genetics , Eukaryotic Initiation Factor-4A/genetics , Sterols/pharmacology , CRISPR-Cas Systems , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mutation , Ribosomes/genetics , Ribosomes/metabolism
16.
Proc Natl Acad Sci U S A ; 116(8): 2925-2934, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718434

ABSTRACT

Phosphatases of regenerating liver (PRL-1, PRL-2, and PRL-3, also known as PTP4A1, PTP4A2, and PTP4A3) control magnesium homeostasis through an association with the CNNM magnesium transport regulators. Although high PRL levels have been linked to cancer progression, regulation of their expression is poorly understood. Here we show that modulating intracellular magnesium levels correlates with a rapid change of PRL expression by a mechanism involving its 5'UTR mRNA region. Mutations or CRISPR-Cas9 targeting of the conserved upstream ORF present in the mRNA leader derepress PRL protein synthesis and attenuate the translational response to magnesium levels. Mechanistically, magnesium depletion reduces intracellular ATP but up-regulates PRL protein expression via activation of the AMPK/mTORC2 pathway, which controls cellular energy status. Hence, altered PRL-2 expression leads to metabolic reprogramming of the cells. These findings uncover a magnesium-sensitive mechanism controlling PRL expression, which plays a role in cellular bioenergetics.


Subject(s)
Cellular Reprogramming/genetics , Energy Metabolism/genetics , Neoplasms/genetics , Protein Tyrosine Phosphatases/genetics , AMP-Activated Protein Kinase Kinases , CRISPR-Cas Systems , Cation Transport Proteins , Cell Cycle Proteins/genetics , Cyclins/genetics , Gene Expression Regulation, Neoplastic , Humans , Liver Regeneration/genetics , MCF-7 Cells , Magnesium/metabolism , Mechanistic Target of Rapamycin Complex 2/genetics , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Neoplasms/pathology , Protein Kinases/genetics
17.
Proc Natl Acad Sci U S A ; 116(16): 7857-7866, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30940748

ABSTRACT

Protein synthesis is essential for cell growth, proliferation, and survival. Protein synthesis is a tightly regulated process that involves multiple mechanisms. Deregulation of protein synthesis is considered as a key factor in the development and progression of a number of diseases, such as cancer. Here we show that the dynamic modification of proteins by O-linked ß-N-acetyl-glucosamine (O-GlcNAcylation) regulates translation initiation by modifying core initiation factors eIF4A and eIF4G, respectively. Mechanistically, site-specific O-GlcNAcylation of eIF4A on Ser322/323 disrupts the formation of the translation initiation complex by perturbing its interaction with eIF4G. In addition, O-GlcNAcylation inhibits the duplex unwinding activity of eIF4A, leading to impaired protein synthesis, and decreased cell proliferation. In contrast, site-specific O-GlcNAcylation of eIF4G on Ser61 promotes its interaction with poly(A)-binding protein (PABP) and poly(A) mRNA. Depletion of eIF4G O-GlcNAcylation results in inhibition of protein synthesis, cell proliferation, and soft agar colony formation. The differential glycosylation of eIF4A and eIF4G appears to be regulated in the initiation complex to fine-tune protein synthesis. Our study thus expands the current understanding of protein synthesis, and adds another dimension of complexity to translational control of cellular proteins.


Subject(s)
Glycosylation , Peptide Chain Initiation, Translational , Cell Line, Tumor , Eukaryotic Initiation Factor-4G/chemistry , Eukaryotic Initiation Factor-4G/metabolism , Humans , Models, Molecular , Neoplasms/chemistry , Neoplasms/metabolism , Poly(A)-Binding Proteins/chemistry , Poly(A)-Binding Proteins/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism
18.
Genes Dev ; 28(1): 85-97, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24395249

ABSTRACT

Genetically engineered mouse models (GEMMs) have greatly expanded our knowledge of pancreatic ductal adenocarcinoma (PDAC) and serve as a critical tool to identify and evaluate new treatment strategies. However, the cost and time required to generate conventional pancreatic cancer GEMMs limits their use for investigating novel genetic interactions in tumor development and maintenance. To address this problem, we developed flexible embryonic stem cell (ESC)-based GEMMs that facilitate the rapid generation of genetically defined multiallelic chimeric mice without further strain intercrossing. The ESCs harbor a latent Kras mutant (a nearly ubiquitous feature of pancreatic cancer), a homing cassette, and other genetic elements needed for rapid insertion and conditional expression of tetracycline-controlled transgenes, including fluorescence-coupled shRNAs capable of efficiently silencing gene function by RNAi. This system produces a disease that recapitulates the progression of pancreatic cancer in human patients and enables the study and visualization of the impact of gene perturbation at any stage of pancreas cancer progression. We describe the use of this approach to dissect temporal roles for the tumor suppressor Pten and the oncogene c-Myc in pancreatic cancer development and maintenance.


Subject(s)
Disease Models, Animal , Embryonic Stem Cells , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Animals , Animals, Genetically Modified , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Reproducibility of Results
19.
J Cell Mol Med ; 25(14): 7089-7094, 2021 07.
Article in English | MEDLINE | ID: mdl-34114734

ABSTRACT

MYSM1 is a chromatin-binding protein, widely investigated for its functions in haematopoiesis in human and mouse; however, its role in haematologic malignancies remains unexplored. Here, we investigate the cross-talk between MYSM1 and oncogenic cMYC in the transcriptional regulation of genes encoding ribosomal proteins, and the implications of these mechanisms for cMYC-driven carcinogenesis. We demonstrate that in cMYC-driven B cell lymphoma in mouse models, MYSM1-loss represses ribosomal protein gene expression and protein synthesis. Importantly, the loss of MYSM1 also strongly inhibits cMYC oncogenic activity and protects against B cell lymphoma onset and progression in the mouse models. This advances the understanding of the molecular and transcriptional mechanisms of lymphomagenesis, and suggests MYSM1 as a possible drug target for cMYC-driven malignancies.


Subject(s)
Lymphoma, B-Cell/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Trans-Activators/deficiency , Ubiquitin-Specific Proteases/deficiency , Animals , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Mice , Proto-Oncogene Proteins c-myc/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism
20.
J Pathol ; 252(1): 77-87, 2020 09.
Article in English | MEDLINE | ID: mdl-32558936

ABSTRACT

Atypical teratoid rhabdoid tumor (ATRT) is a fatal pediatric malignancy of the central neural system lacking effective treatment options. It belongs to the rhabdoid tumor family and is usually caused by biallelic inactivation of SMARCB1, encoding a key subunit of SWI/SNF chromatin remodeling complexes. Previous studies proposed that SMARCB1 loss drives rhabdoid tumor by promoting cell cycle through activating transcription of cyclin D1 while suppressing p16. However, low cyclin D1 protein expression is observed in most ATRT patient tumors. The underlying mechanism and therapeutic implication of this molecular trait remain unknown. Here, we show that SMARCB1 loss in ATRT leads to the reduction of cyclin D1 expression by upregulating MIR17HG, a microRNA (miRNA) cluster known to generate multiple miRNAs targeting CCND1. Furthermore, we find that this cyclin D1 deficiency in ATRT results in marked in vitro and in vivo sensitivity to the CDK4/6 inhibitor palbociclib as a single agent. Our study identifies a novel genetic interaction between SMARCB1 and MIR17HG in regulating cyclin D1 in ATRT and suggests a rationale to treat ATRT patients with FDA-approved CDK4/6 inhibitors. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Proteins/genetics , Rhabdoid Tumor/genetics , SMARCB1 Protein/genetics , Teratoma/genetics , Cell Line, Tumor , Cell Survival , Cyclin D1/metabolism , Humans , Proteins/metabolism , Rhabdoid Tumor/metabolism , Rhabdoid Tumor/pathology , SMARCB1 Protein/metabolism , Teratoma/metabolism , Teratoma/pathology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL