Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 389
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
Cell ; 166(4): 963-976, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27477511

ABSTRACT

Pancreatic cancer is a deadly malignancy that lacks effective therapeutics. We previously reported that oncogenic Kras induced the redox master regulator Nfe2l2/Nrf2 to stimulate pancreatic and lung cancer initiation. Here, we show that NRF2 is necessary to maintain pancreatic cancer proliferation by regulating mRNA translation. Specifically, loss of NRF2 led to defects in autocrine epidermal growth factor receptor (EGFR) signaling and oxidation of specific translational regulatory proteins, resulting in impaired cap-dependent and cap-independent mRNA translation in pancreatic cancer cells. Combined targeting of the EGFR effector AKT and the glutathione antioxidant pathway mimicked Nrf2 ablation to potently inhibit pancreatic cancer ex vivo and in vivo, representing a promising synthetic lethal strategy for treating the disease.


Subject(s)
NF-E2-Related Factor 2/metabolism , Pancreatic Neoplasms/metabolism , Protein Biosynthesis , Animals , Autocrine Communication , Cysteine/metabolism , Glutathione/metabolism , Humans , Mice , Organoids/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction
3.
Nat Rev Mol Cell Biol ; 19(12): 791-807, 2018 12.
Article in English | MEDLINE | ID: mdl-30038383

ABSTRACT

Advances in sequencing and high-throughput techniques have provided an unprecedented opportunity to interrogate human diseases on a genome-wide scale. The list of disease-causing mutations is expanding rapidly, and mutations affecting mRNA translation are no exception. Translation (protein synthesis) is one of the most complex processes in the cell. The orchestrated action of ribosomes, tRNAs and numerous translation factors decodes the information contained in mRNA into a polypeptide chain. The intricate nature of this process renders it susceptible to deregulation at multiple levels. In this Review, we summarize current evidence of translation deregulation in human diseases other than cancer. We discuss translation-related diseases on the basis of the molecular aberration that underpins their pathogenesis (including tRNA dysfunction, ribosomopathies, deregulation of the integrated stress response and deregulation of the mTOR pathway) and describe how deregulation of translation generates the phenotypic variability observed in these disorders.


Subject(s)
Disease/genetics , Protein Biosynthesis/genetics , Protein Biosynthesis/physiology , Animals , Biological Variation, Population/genetics , Humans , Peptide Initiation Factors/genetics , RNA, Messenger/genetics , RNA, Transfer/genetics , Ribosomes/genetics , Stress, Physiological/genetics , TOR Serine-Threonine Kinases/genetics
4.
Nat Immunol ; 18(9): 1046-1057, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28714979

ABSTRACT

Translation is a critical process in protein synthesis, but translational regulation in antigen-specific T cells in vivo has not been well defined. Here we have characterized the translatome of virus-specific CD8+ effector T cells (Teff cells) during acute infection of mice with lymphocytic choriomeningitis virus (LCMV). Antigen-specific T cells exerted dynamic translational control of gene expression that correlated with cell proliferation and stimulation via the T cell antigen receptor (TCR). The translation of mRNAs that encode translation machinery, including ribosomal proteins, was upregulated during the T cell clonal-expansion phase, followed by inhibition of the translation of those transcripts when the CD8+ Teff cells stopped dividing just before the contraction phase. That translational suppression was more pronounced in terminal effector cells than in memory precursor cells and was regulated by antigenic stimulation and signals from the kinase mTOR. Our studies show that translation of transcripts encoding ribosomal proteins is regulated during the differentiation of CD8+ Teff cells and might have a role in fate 'decisions' involved in the formation of memory cells.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Protein Biosynthesis/immunology , Animals , Arenaviridae Infections/genetics , Arenaviridae Infections/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/genetics , Flow Cytometry , Gene Expression Regulation , Immunologic Memory/immunology , Interferon-gamma/immunology , Lymphocytic choriomeningitis virus , Mice , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell/immunology , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , TOR Serine-Threonine Kinases/immunology
5.
Cell ; 158(6): 1238-1239, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215483

ABSTRACT

Transfer RNAs (tRNAs) deliver amino acids to the ribosome during mRNA translation. Gingold et al. now provide evidence that alterations in the cellular tRNA repertoire are tightly coordinated with changes in mRNA expression. These changes in the tRNA repertoire dictate translational programs that distinguish differentiating from proliferating cells.


Subject(s)
Cell Differentiation , Cell Proliferation , Protein Biosynthesis , RNA, Transfer/genetics , Humans
6.
Cell ; 157(1): 26-40, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24679524

ABSTRACT

The subcellular position of a protein is a key determinant of its function. Mounting evidence indicates that RNA localization, where specific mRNAs are transported subcellularly and subsequently translated in response to localized signals, is an evolutionarily conserved mechanism to control protein localization. On-site synthesis confers novel signaling properties to a protein and helps to maintain local proteome homeostasis. Local translation plays particularly important roles in distal neuronal compartments, and dysregulated RNA localization and translation cause defects in neuronal wiring and survival. Here, we discuss key findings in this area and possible implications of this adaptable and swift mechanism for spatial control of gene function.


Subject(s)
Protein Biosynthesis , Proteins/genetics , Proteins/metabolism , RNA Transport , RNA, Messenger/metabolism , Animals , Humans , Nervous System Diseases/embryology , Nervous System Diseases/metabolism , Proteins/chemistry
7.
Mol Cell ; 81(2): 398-407.e4, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33340489

ABSTRACT

Mechanistic target of rapamycin complex 1 (mTORC1) controls cell growth and proliferation by sensing fluctuations in environmental cues such as nutrients, growth factors, and energy levels. The Rag GTPases (Rags) serve as a critical module that signals amino acid (AA) availability to modulate mTORC1 localization and activity. Recent studies have demonstrated how AAs regulate mTORC1 activity through Rags. Here, we uncover an unconventional pathway that activates mTORC1 in response to variations in threonine (Thr) levels via mitochondrial threonyl-tRNA synthetase TARS2. TARS2 interacts with inactive Rags, particularly GTP-RagC, leading to increased GTP loading of RagA. mTORC1 activity in cells lacking TARS2 is resistant to Thr repletion, showing that TARS2 is necessary for Thr-dependent mTORC1 activation. The requirement of TARS2, but not cytoplasmic threonyl-tRNA synthetase TARS, for this effect demonstrates an additional layer of complexity in the regulation of mTORC1 activity.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/genetics , Mitochondria/metabolism , Monomeric GTP-Binding Proteins/genetics , Threonine-tRNA Ligase/genetics , Threonine/metabolism , Gene Expression Regulation , Guanosine Diphosphate/metabolism , Guanosine Triphosphate/metabolism , HEK293 Cells , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Monomeric GTP-Binding Proteins/metabolism , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism , Signal Transduction , Threonine-tRNA Ligase/antagonists & inhibitors , Threonine-tRNA Ligase/metabolism
8.
Mol Cell ; 81(6): 1187-1199.e5, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33581076

ABSTRACT

Type I interferons (IFNs) are critical cytokines in the host defense against invading pathogens. Sustained production of IFNs, however, is detrimental to the host, as it provokes autoimmune diseases. Thus, the expression of IFNs is tightly controlled. We report that the mRNA 5' cap-binding protein 4EHP plays a key role in regulating type I IFN concomitant with controlling virus replication, both in vitro and in vivo. Mechanistically, 4EHP suppresses IFN-ß production by effecting the miR-34a-induced translational silencing of Ifnb1 mRNA. miR-34a is upregulated by both RNA virus infection and IFN-ß induction, prompting a negative feedback regulatory mechanism that represses IFN-ß expression via 4EHP. These findings demonstrate the direct involvement of 4EHP in virus-induced host response, underscoring a critical translational silencing mechanism mediated by 4EHP and miR-34a to impede sustained IFN production. This study highlights an intrinsic regulatory function for miRNA and the translation machinery in maintaining host homeostasis.


Subject(s)
Eukaryotic Initiation Factor-4E/immunology , Immunity, Innate , MicroRNAs/immunology , Protein Biosynthesis/immunology , RNA Virus Infections/immunology , RNA Viruses/immunology , Animals , Eukaryotic Initiation Factor-4E/genetics , HEK293 Cells , Humans , Interferon-beta/genetics , Interferon-beta/immunology , Mice , Mice, Transgenic , MicroRNAs/genetics , RNA Virus Infections/genetics , RNA Viruses/genetics
9.
Nat Immunol ; 17(5): 514-522, 2016 May.
Article in English | MEDLINE | ID: mdl-27043414

ABSTRACT

Cytosolic DNA-mediated activation of the transcription factor IRF3 is a key event in host antiviral responses. Here we found that infection with DNA viruses induced interaction of the metabolic checkpoint kinase mTOR downstream effector and kinase S6K1 and the signaling adaptor STING in a manner dependent on the DNA sensor cGAS. We further demonstrated that the kinase domain, but not the kinase function, of S6K1 was required for the S6K1-STING interaction and that the TBK1 critically promoted this process. The formation of a tripartite S6K1-STING-TBK1 complex was necessary for the activation of IRF3, and disruption of this signaling axis impaired the early-phase expression of IRF3 target genes and the induction of T cell responses and mucosal antiviral immunity. Thus, our results have uncovered a fundamental regulatory mechanism for the activation of IRF3 in the cytosolic DNA pathway.


Subject(s)
DNA/immunology , Interferon Regulatory Factor-3/immunology , Membrane Proteins/immunology , Ribosomal Protein S6 Kinases, 90-kDa/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Cytosol/immunology , Cytosol/metabolism , Cytosol/virology , DNA/genetics , DNA/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , HEK293 Cells , Herpes Simplex/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/physiology , Humans , Immunization/methods , Immunoblotting , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Nucleotidyltransferases/metabolism , Ovalbumin/genetics , Ovalbumin/immunology , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
10.
Mol Cell ; 77(6): 1176-1192.e16, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31999954

ABSTRACT

Microexons represent the most highly conserved class of alternative splicing, yet their functions are poorly understood. Here, we focus on closely related neuronal microexons overlapping prion-like domains in the translation initiation factors, eIF4G1 and eIF4G3, the splicing of which is activity dependent and frequently disrupted in autism. CRISPR-Cas9 deletion of these microexons selectively upregulates synaptic proteins that control neuronal activity and plasticity and further triggers a gene expression program mirroring that of activated neurons. Mice lacking the Eif4g1 microexon display social behavior, learning, and memory deficits, accompanied by altered hippocampal synaptic plasticity. We provide evidence that the eIF4G microexons function as a translational brake by causing ribosome stalling, through their propensity to promote the coalescence of cytoplasmic granule components associated with translation repression, including the fragile X mental retardation protein FMRP. The results thus reveal an autism-disrupted mechanism by which alternative splicing specializes neuronal translation to control higher order cognitive functioning.


Subject(s)
Autistic Disorder/physiopathology , Cognitive Dysfunction/pathology , Eukaryotic Initiation Factor-4G/physiology , Exons/genetics , Fragile X Mental Retardation Protein/metabolism , Neuroblastoma/pathology , Neurons/pathology , Animals , Behavior, Animal , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Fragile X Mental Retardation Protein/genetics , Male , Mice , Mice, Inbred C57BL , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neurogenesis , Neurons/metabolism , Protein Biosynthesis , RNA Splicing , Tumor Cells, Cultured
11.
Nature ; 590(7845): 315-319, 2021 02.
Article in English | MEDLINE | ID: mdl-33328636

ABSTRACT

Effective pharmacotherapy for major depressive disorder remains a major challenge, as more than 30% of patients are resistant to the first line of treatment (selective serotonin reuptake inhibitors)1. Sub-anaesthetic doses of ketamine, a non-competitive N-methyl-D-aspartate receptor antagonist2,3, provide rapid and long-lasting antidepressant effects in these patients4-6, but the molecular mechanism of these effects remains unclear7,8. Ketamine has been proposed to exert its antidepressant effects through its metabolite (2R,6R)-hydroxynorketamine ((2R,6R)-HNK)9. The antidepressant effects of ketamine and (2R,6R)-HNK in rodents require activation of the mTORC1 kinase10,11. mTORC1 controls various neuronal functions12, particularly through cap-dependent initiation of mRNA translation via the phosphorylation and inactivation of eukaryotic initiation factor 4E-binding proteins (4E-BPs)13. Here we show that 4E-BP1 and 4E-BP2 are key effectors of the antidepressant activity of ketamine and (2R,6R)-HNK, and that ketamine-induced hippocampal synaptic plasticity depends on 4E-BP2 and, to a lesser extent, 4E-BP1. It has been hypothesized that ketamine activates mTORC1-4E-BP signalling in pyramidal excitatory cells of the cortex8,14. To test this hypothesis, we studied the behavioural response to ketamine and (2R,6R)-HNK in mice lacking 4E-BPs in either excitatory or inhibitory neurons. The antidepressant activity of the drugs is mediated by 4E-BP2 in excitatory neurons, and 4E-BP1 and 4E-BP2 in inhibitory neurons. Notably, genetic deletion of 4E-BP2 in inhibitory neurons induced a reduction in baseline immobility in the forced swim test, mimicking an antidepressant effect. Deletion of 4E-BP2 specifically in inhibitory neurons also prevented the ketamine-induced increase in hippocampal excitatory neurotransmission, and this effect concurred with the inability of ketamine to induce a long-lasting decrease in inhibitory neurotransmission. Overall, our data show that 4E-BPs are central to the antidepressant activity of ketamine.


Subject(s)
Antidepressive Agents/pharmacology , Eukaryotic Initiation Factor-4E/metabolism , Ketamine/pharmacology , Neurons/drug effects , Neurons/metabolism , Protein Biosynthesis/drug effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Depressive Disorder, Major/drug therapy , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/drug effects , Interneurons/metabolism , Ketamine/analogs & derivatives , Ketamine/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mutation , Neural Inhibition/drug effects , Neural Inhibition/genetics , Neurons/classification , Neurons/cytology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Synaptic Transmission/drug effects
12.
Nat Immunol ; 15(6): 503-11, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24840981

ABSTRACT

Selective translational control of gene expression is emerging as a principal mechanism for the regulation of protein abundance that determines a variety of functions in both the adaptive immune system and the innate immune system. The translation-initiation factor eIF4E acts as a node for such regulation, but non-eIF4E mechanisms are also prevalent. Studies of 'translatomes' (genome-wide pools of translated mRNA) have facilitated mechanistic discoveries by identifying key regulatory components, including transcription factors, that are under translational control. Here we review the current knowledge on mechanisms that regulate translation and thereby modulate immunological function. We further describe approaches for measuring and analyzing translatomes and how such powerful tools can facilitate future insights on the role of translational control in the immune system.


Subject(s)
Gene Expression Regulation/genetics , Immune System/immunology , Protein Biosynthesis/genetics , Transcription, Genetic , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Cell Cycle Proteins , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4E/immunology , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/genetics , Phosphoproteins/genetics , Phosphoproteins/immunology , Protein Biosynthesis/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/genetics
13.
Cell ; 145(3): 333-4, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21529706

ABSTRACT

The ribosome plays a universally conserved role in catalyzing protein synthesis. Kondrashov et al. (2011) now provide evidence that the loss of function of ribosomal protein L38 in mice leads to a selective reduction in the translation of Hox mRNAs, thus suggesting that ribosomal proteins play a critical role during embryonic development.

14.
Nature ; 586(7829): 412-416, 2020 10.
Article in English | MEDLINE | ID: mdl-33029011

ABSTRACT

An important tenet of learning and memory is the notion of a molecular switch that promotes the formation of long-term memory1-4. The regulation of proteostasis is a critical and rate-limiting step in the consolidation of new memories5-10. One of the most effective and prevalent ways to enhance memory is by regulating the synthesis of proteins controlled by the translation initiation factor eIF211. Phosphorylation of the α-subunit of eIF2 (p-eIF2α), the central component of the integrated stress response (ISR), impairs long-term memory formation in rodents and birds11-13. By contrast, inhibiting the ISR by mutating the eIF2α phosphorylation site, genetically11 and pharmacologically inhibiting the ISR kinases14-17, or mimicking reduced p-eIF2α with the ISR inhibitor ISRIB11, enhances long-term memory in health and disease18. Here we used molecular genetics to dissect the neuronal circuits by which the ISR gates cognitive processing. We found that learning reduces eIF2α phosphorylation in hippocampal excitatory neurons and a subset of hippocampal inhibitory neurons (those that express somatostatin, but not parvalbumin). Moreover, ablation of p-eIF2α in either excitatory or somatostatin-expressing (but not parvalbumin-expressing) inhibitory neurons increased general mRNA translation, bolstered synaptic plasticity and enhanced long-term memory. Thus, eIF2α-dependent mRNA translation controls memory consolidation via autonomous mechanisms in excitatory and somatostatin-expressing inhibitory neurons.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Hippocampus/cytology , Memory Consolidation , Neurons/metabolism , Somatostatin/metabolism , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/physiology , Eukaryotic Initiation Factor-2/deficiency , Eukaryotic Initiation Factor-2/genetics , Excitatory Postsynaptic Potentials , Hippocampus/physiology , Long-Term Potentiation , Male , Memory, Long-Term , Mice , Mice, Inbred C57BL , Neural Inhibition , Neuronal Plasticity , Parvalbumins , Phosphorylation , Pyramidal Cells/physiology , Synaptic Transmission
15.
Nucleic Acids Res ; 52(3): 1064-1079, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38038264

ABSTRACT

mRNA translation is a fundamental process for life. Selection of the translation initiation site (TIS) is crucial, as it establishes the correct open reading frame for mRNA decoding. Studies in vertebrate mRNAs discovered that a purine at -3 and a G at +4 (where A of the AUG initiator codon is numbered + 1), promote TIS recognition. However, the TIS context in other eukaryotes has been poorly experimentally analyzed. We analyzed in vitro the influence of the -3, -2, -1 and + 4 positions of the TIS context in rabbit, Drosophila, wheat, and yeast. We observed that -3A conferred the best translational efficiency across these species. However, we found variability at the + 4 position for optimal translation. In addition, the Kozak motif that was defined from mammalian cells was only weakly predictive for wheat and essentially non-predictive for yeast. We discovered eight conserved sequences that significantly disfavored translation. Due to the big differences in translational efficiency observed among weak TIS context sequences, we define a novel category that we termed 'barren AUG context sequences (BACS)', which represent sequences disfavoring translation. Analysis of mRNA-ribosomal complexes structures provided insights into the function of BACS. The gene ontology of the BACS-containing mRNAs is presented.


Subject(s)
Codon, Initiator , Conserved Sequence , Protein Biosynthesis , Animals , Rabbits , Codon, Initiator/genetics , Mammals/genetics , Peptide Chain Initiation, Translational , RNA, Messenger/metabolism , Yeasts , Eukaryota/genetics , Eukaryota/metabolism
16.
Proc Natl Acad Sci U S A ; 120(25): e2300008120, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37307456

ABSTRACT

mRNA translation initiation plays a critical role in learning and memory. The eIF4F complex, composed of the cap-binding protein eIF4E, ATP-dependent RNA helicase eIF4A, and scaffolding protein eIF4G, is a pivotal factor in the mRNA translation initiation process. eIF4G1, the major paralogue of the three eIF4G family members, is indispensable for development, but its function in learning and memory is unknown. To study the role of eIF4G1 in cognition, we used an eIF4G1 haploinsufficient (eIF4G1-1D) mouse model. The axonal arborization of eIF4G1-1D primary hippocampal neurons was significantly disrupted, and the mice displayed impairment in hippocampus-dependent learning and memory. Translatome analysis showed that the translation of mRNAs encoding proteins of the mitochondrial oxidative phosphorylation (OXPHOS) system was decreased in the eIF4G1-1D brain, and OXPHOS was decreased in eIF4G1-silenced cells. Thus, eIF4G1-mediated mRNA translation is crucial for optimal cognitive function, which is dependent on OXPHOS and neuronal morphogenesis.


Subject(s)
Eukaryotic Initiation Factor-4G , Oxidative Phosphorylation , Animals , Mice , RNA, Messenger , Peptide Chain Initiation, Translational , Morphogenesis , DNA Helicases
17.
Proc Natl Acad Sci U S A ; 120(49): e2308671120, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38015848

ABSTRACT

Activation of neuronal protein synthesis upon learning is critical for the formation of long-term memory. Here, we report that learning in the contextual fear conditioning paradigm engenders a decrease in eIF2α (eukaryotic translation initiation factor 2) phosphorylation in astrocytes in the hippocampal CA1 region, which promotes protein synthesis. Genetic reduction of eIF2α phosphorylation in hippocampal astrocytes enhanced contextual and spatial memory and lowered the threshold for the induction of long-lasting plasticity by modulating synaptic transmission. Thus, learning-induced dephosphorylation of eIF2α in astrocytes bolsters hippocampal synaptic plasticity and consolidation of long-term memories.


Subject(s)
Astrocytes , Long-Term Potentiation , Long-Term Potentiation/physiology , Neuronal Plasticity/genetics , Hippocampus/physiology , Protein Biosynthesis , CA1 Region, Hippocampal , Memory, Long-Term/physiology
18.
Annu Rev Biochem ; 79: 351-79, 2010.
Article in English | MEDLINE | ID: mdl-20533884

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs that extensively regulate gene expression in animals, plants, and protozoa. miRNAs function posttranscriptionally by usually base-pairing to the mRNA 3'-untranslated regions to repress protein synthesis by mechanisms that are not fully understood. In this review, we describe principles of miRNA-mRNA interactions and proteins that interact with miRNAs and function in miRNA-mediated repression. We discuss the multiple, often contradictory, mechanisms that miRNAs have been reported to use, which cause translational repression and mRNA decay. We also address the issue of cellular localization of miRNA-mediated events and a role for RNA-binding proteins in activation or relief of miRNA repression.


Subject(s)
MicroRNAs/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Animals , Gene Expression Regulation , Humans , MicroRNAs/chemistry , RNA Stability , RNA-Binding Proteins/metabolism
19.
J Cell Sci ; 136(19)2023 10 01.
Article in English | MEDLINE | ID: mdl-37732428

ABSTRACT

Viruses use microRNAs (miRNAs) to impair the host antiviral response and facilitate viral infection by expressing their own miRNAs or co-opting cellular miRNAs. miRNAs inhibit translation initiation of their target mRNAs by recruiting the GIGYF2-4EHP (or EIF4E2) translation repressor complex to the mRNA 5'-cap structure. We recently reported that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-encoded non-structural protein 2 (NSP2) interacts with GIGYF2. This interaction is critical for blocking translation of the Ifnb1 mRNA that encodes the cytokine interferon ß, and thereby impairs the host antiviral response. However, it is not known whether NSP2 also affects miRNA-mediated silencing. Here, we demonstrate the pervasive augmentation of miRNA-mediated translational repression of cellular mRNAs by NSP2. We show that NSP2 interacts with argonaute 2 (AGO2), the core component of the miRNA-induced silencing complex (miRISC), via GIGYF2 and enhances the translational repression mediated by natural miRNA-binding sites in the 3' untranslated region of cellular mRNAs. Our data reveal an additional layer of the complex mechanism by which SARS-CoV-2 and likely other coronaviruses manipulate the host gene expression program by co-opting the host miRNA-mediated silencing machinery.


Subject(s)
COVID-19 , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , COVID-19/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Antiviral Agents
20.
Cell ; 140(3): 338-48, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-20144759

ABSTRACT

As chronic inflammation is a hallmark of obesity, pathways that integrate nutrient- and pathogen sensing pathways are of great interest in understanding the mechanisms of insulin resistance, type 2 diabetes, and other chronic metabolic pathologies. Here, we provide evidence that double-stranded RNA-dependent protein kinase (PKR) can respond to nutrient signals as well as endoplasmic reticulum (ER) stress and coordinate the activity of other critical inflammatory kinases such as the c-Jun N-terminal kinase (JNK) to regulate insulin action and metabolism. PKR also directly targets and modifies insulin receptor substrate and hence integrates nutrients and insulin action with a defined pathogen response system. Dietary and genetic obesity features marked activation of PKR in adipose and liver tissues and absence of PKR alleviates metabolic deterioration due to nutrient or energy excess in mice. These findings demonstrate PKR as a critical component of an inflammatory complex that responds to nutrients and organelle dysfunction.


Subject(s)
Metabolic Diseases/metabolism , eIF-2 Kinase/metabolism , Animals , Female , Humans , Insulin Receptor Substrate Proteins/metabolism , MAP Kinase Kinase 4/metabolism , Male , Mice , eIF-2 Kinase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL