Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 169(6): 1051-1065.e18, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575669

ABSTRACT

During eukaryotic evolution, ribosomes have considerably increased in size, forming a surface-exposed ribosomal RNA (rRNA) shell of unknown function, which may create an interface for yet uncharacterized interacting proteins. To investigate such protein interactions, we establish a ribosome affinity purification method that unexpectedly identifies hundreds of ribosome-associated proteins (RAPs) from categories including metabolism and cell cycle, as well as RNA- and protein-modifying enzymes that functionally diversify mammalian ribosomes. By further characterizing RAPs, we discover the presence of ufmylation, a metazoan-specific post-translational modification (PTM), on ribosomes and define its direct substrates. Moreover, we show that the metabolic enzyme, pyruvate kinase muscle (PKM), interacts with sub-pools of endoplasmic reticulum (ER)-associated ribosomes, exerting a non-canonical function as an RNA-binding protein in the translation of ER-destined mRNAs. Therefore, RAPs interconnect one of life's most ancient molecular machines with diverse cellular processes, providing an additional layer of regulatory potential to protein expression.


Subject(s)
Ribosomes/chemistry , Ribosomes/metabolism , Animals , Carrier Proteins/metabolism , Embryonic Stem Cells/metabolism , Endoplasmic Reticulum/metabolism , Mass Spectrometry , Membrane Proteins/metabolism , Mice , Protein Biosynthesis , Protein Interaction Mapping , Protein Processing, Post-Translational , Ribosomal Proteins/metabolism , Thyroid Hormones/metabolism , Ubiquitin-Protein Ligases/metabolism , Thyroid Hormone-Binding Proteins
2.
Mol Cell ; 84(18): 3545-3563.e25, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39260367

ABSTRACT

Ribosomes are emerging as direct regulators of gene expression, with ribosome-associated proteins (RAPs) allowing ribosomes to modulate translation. Nevertheless, a lack of technologies to enrich RAPs across sample types has prevented systematic analysis of RAP identities, dynamics, and functions. We have developed a label-free methodology called RAPIDASH to enrich ribosomes and RAPs from any sample. We applied RAPIDASH to mouse embryonic tissues and identified hundreds of potential RAPs, including Dhx30 and Llph, two forebrain RAPs important for neurodevelopment. We identified a critical role of LLPH in neural development linked to the translation of genes with long coding sequences. In addition, we showed that RAPIDASH can identify ribosome changes in cancer cells. Finally, we characterized ribosome composition remodeling during immune cell activation and observed extensive changes post-stimulation. RAPIDASH has therefore enabled the discovery of RAPs in multiple cell types, tissues, and stimuli and is adaptable to characterize ribosome remodeling in several contexts.


Subject(s)
Macrophages , Ribosomal Proteins , Ribosomes , Animals , Ribosomes/metabolism , Ribosomes/genetics , Mice , Humans , Macrophages/metabolism , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , Protein Biosynthesis , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Gene Expression Regulation, Developmental , Cell Line, Tumor , Mice, Inbred C57BL
3.
Nat Rev Mol Cell Biol ; 19(3): 158-174, 2018 03.
Article in English | MEDLINE | ID: mdl-29165424

ABSTRACT

RNA molecules can fold into intricate shapes that can provide an additional layer of control of gene expression beyond that of their sequence. In this Review, we discuss the current mechanistic understanding of structures in 5' untranslated regions (UTRs) of eukaryotic mRNAs and the emerging methodologies used to explore them. These structures may regulate cap-dependent translation initiation through helicase-mediated remodelling of RNA structures and higher-order RNA interactions, as well as cap-independent translation initiation through internal ribosome entry sites (IRESs), mRNA modifications and other specialized translation pathways. We discuss known 5' UTR RNA structures and how new structure probing technologies coupled with prospective validation, particularly compensatory mutagenesis, are likely to identify classes of structured RNA elements that shape post-transcriptional control of gene expression and the development of multicellular organisms.


Subject(s)
5' Untranslated Regions , RNA, Messenger/genetics , Animals , Eukaryotic Initiation Factor-3/metabolism , Eukaryotic Initiation Factor-4F/metabolism , G-Quadruplexes , Humans , Internal Ribosome Entry Sites , Models, Biological , Models, Molecular , Peptide Chain Initiation, Translational , Protein Biosynthesis , RNA Folding , RNA Helicases/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Ribosomes/genetics , Ribosomes/metabolism
4.
Nat Rev Mol Cell Biol ; 19(10): 673, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30111874

ABSTRACT

The legend of Figure 1 has been modified to remove misleading referencing to evolution. The title of the legend has been modified from 'Evolutionary expansion of eukaryotic 5' UTR lengths' to 'Interspecies variation in 5' UTR lengths'; the first sentence of the legend has been modified from 'The length of 5' untranslated regions (UTRs) has increased in eukaryotes during evolution…' to 'The length of 5' untranslated regions (UTRs) varies in eukaryotes…'. The changes have been made in the HTML and PDF versions of the manuscript.

5.
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
6.
Mol Cell ; 82(12): 2179-2184, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35714581

ABSTRACT

The concept of specialized ribosomes has garnered equal amounts of interest and skepticism since it was first introduced. We ask researchers in the field to provide their perspective on the topic and weigh in on the evidence (or lack thereof) and what the future may bring.


Subject(s)
Protein Biosynthesis , Ribosomes , Ribosomes/genetics , Ribosomes/metabolism
7.
Mol Cell ; 82(13): 2370-2384.e10, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35512709

ABSTRACT

The p53 transcription factor drives anti-proliferative gene expression programs in response to diverse stressors, including DNA damage and oncogenic signaling. Here, we seek to uncover new mechanisms through which p53 regulates gene expression using tandem affinity purification/mass spectrometry to identify p53-interacting proteins. This approach identified METTL3, an m6A RNA-methyltransferase complex (MTC) constituent, as a p53 interactor. We find that METTL3 promotes p53 protein stabilization and target gene expression in response to DNA damage and oncogenic signals, by both catalytic activity-dependent and independent mechanisms. METTL3 also enhances p53 tumor suppressor activity in in vivo mouse cancer models and human cancer cells. Notably, METTL3 only promotes tumor suppression in the context of intact p53. Analysis of human cancer genome data further supports the notion that the MTC reinforces p53 function in human cancer. Together, these studies reveal a fundamental role for METTL3 in amplifying p53 signaling in response to cellular stress.


Subject(s)
Methyltransferases , Tumor Suppressor Protein p53 , Animals , Carcinogenesis , Methyltransferases/metabolism , Mice , RNA , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics
8.
Nature ; 620(7972): 163-171, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37495694

ABSTRACT

An outstanding mystery in biology is why some species, such as the axolotl, can regenerate tissues whereas mammals cannot1. Here, we demonstrate that rapid activation of protein synthesis is a unique feature of the injury response critical for limb regeneration in the axolotl (Ambystoma mexicanum). By applying polysome sequencing, we identify hundreds of transcripts, including antioxidants and ribosome components that are selectively activated at the level of translation from pre-existing messenger RNAs in response to injury. By contrast, protein synthesis is not activated in response to non-regenerative digit amputation in the mouse. We identify the mTORC1 pathway as a key upstream signal that mediates tissue regeneration and translational control in the axolotl. We discover unique expansions in mTOR protein sequence among urodele amphibians. By engineering an axolotl mTOR (axmTOR) in human cells, we show that these changes create a hypersensitive kinase that allows axolotls to maintain this pathway in a highly labile state primed for rapid activation. This change renders axolotl mTOR more sensitive to nutrient sensing, and inhibition of amino acid transport is sufficient to inhibit tissue regeneration. Together, these findings highlight the unanticipated impact of the translatome on orchestrating the early steps of wound healing in a highly regenerative species and provide a missing link in our understanding of vertebrate regenerative potential.


Subject(s)
Ambystoma mexicanum , Biological Evolution , Protein Biosynthesis , Regeneration , TOR Serine-Threonine Kinases , Animals , Humans , Mice , Ambystoma mexicanum/physiology , Amino Acid Sequence , Extremities/physiology , Regeneration/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/metabolism , Wound Healing , Mechanistic Target of Rapamycin Complex 1/metabolism , Species Specificity , Antioxidants/metabolism , Nutrients/metabolism , Polyribosomes/genetics , Polyribosomes/metabolism
9.
Annu Rev Cell Dev Biol ; 31: 31-54, 2015.
Article in English | MEDLINE | ID: mdl-26443190

ABSTRACT

A central question in cell and developmental biology is how the information encoded in the genome is differentially interpreted to generate a diverse array of cell types. A growing body of research on posttranscriptional gene regulation is revealing that both global protein synthesis rates and the translation of specific mRNAs are highly specialized in different cell types. How this exquisite translational regulation is achieved is the focus of this review. Two levels of regulation are discussed: the translation machinery and cis-acting elements within mRNAs. Recent evidence shows that the ribosome itself directs how the genome is translated in time and space and reveals surprising functional specificity in individual components of the core translation machinery. We are also just beginning to appreciate the rich regulatory information embedded in the untranslated regions of mRNAs, which direct the selective translation of transcripts. These hidden RNA regulons may interface with a myriad of RNA-binding proteins and specialized translation machinery to provide an additional layer of regulation to how transcripts are spatiotemporally expressed. Understanding this largely unexplored world of translational codes hardwired in the core translation machinery is an exciting new research frontier fundamental to our understanding of gene regulation, organismal development, and evolution.


Subject(s)
Genome/genetics , Protein Biosynthesis/genetics , RNA/genetics , Regulon/genetics , Ribosomes/genetics , Animals , Gene Expression Regulation/genetics , Humans , RNA, Messenger/genetics , RNA-Binding Proteins/genetics
10.
Mol Cell ; 80(6): 980-995.e13, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33202249

ABSTRACT

Ribosomes have been suggested to directly control gene regulation, but regulatory roles for ribosomal RNA (rRNA) remain largely unexplored. Expansion segments (ESs) consist of multitudes of tentacle-like rRNA structures extending from the core ribosome in eukaryotes. ESs are remarkably variable in sequence and size across eukaryotic evolution with largely unknown functions. In characterizing ribosome binding to a regulatory element within a Homeobox (Hox) 5' UTR, we identify a modular stem-loop within this element that binds to a single ES, ES9S. Engineering chimeric, "humanized" yeast ribosomes for ES9S reveals that an evolutionary change in the sequence of ES9S endows species-specific binding of Hoxa9 mRNA to the ribosome. Genome editing to site-specifically disrupt the Hoxa9-ES9S interaction demonstrates the functional importance for such selective mRNA-rRNA binding in translation control. Together, these studies unravel unexpected gene regulation directly mediated by rRNA and how ribosome evolution drives translation of critical developmental regulators.


Subject(s)
Homeodomain Proteins/genetics , Protein Biosynthesis/genetics , RNA, Ribosomal/ultrastructure , Ribosomes/genetics , 5' Untranslated Regions/genetics , Gene Expression Regulation/genetics , Genes, Homeobox/genetics , Homeodomain Proteins/ultrastructure , Nucleic Acid Conformation , RNA, Messenger/genetics , RNA, Ribosomal/genetics , Ribosomes/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/ultrastructure , Species Specificity
11.
Cell ; 145(3): 383-397, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21529712

ABSTRACT

Historically, the ribosome has been viewed as a complex ribozyme with constitutive rather than regulatory capacity in mRNA translation. Here we identify mutations of the Ribosomal Protein L38 (Rpl38) gene in mice exhibiting surprising tissue-specific patterning defects, including pronounced homeotic transformations of the axial skeleton. In Rpl38 mutant embryos, global protein synthesis is unchanged; however the translation of a select subset of Homeobox mRNAs is perturbed. Our data reveal that RPL38 facilitates 80S complex formation on these mRNAs as a regulatory component of the ribosome to confer transcript-specific translational control. We further show that Rpl38 expression is markedly enriched in regions of the embryo where loss-of-function phenotypes occur. Unexpectedly, a ribosomal protein (RP) expression screen reveals dynamic regulation of individual RPs within the vertebrate embryo. Collectively, these findings suggest that RP activity may be highly regulated to impart a new layer of specificity in the control of gene expression and mammalian development.


Subject(s)
Body Patterning , Bone Diseases, Developmental/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Protein Biosynthesis , Ribosomal Proteins/metabolism , Animals , Humans , Mice , Mice, Inbred C57BL , Mutation , Organogenesis , RNA, Messenger/metabolism , Ribosomal Proteins/genetics , Ribosomes/metabolism , Tail/abnormalities
12.
Mol Cell ; 71(3): 364-374, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30075139

ABSTRACT

The ribosome has recently transitioned from being viewed as a passive, indiscriminate machine to a more dynamic macromolecular complex with specialized roles in the cell. Here, we discuss the historical milestones from the discovery of the ribosome itself to how this ancient machinery has gained newfound appreciation as a more regulatory participant in the central dogma of gene expression. The first emerging examples of direct changes in ribosome composition at the RNA and protein level, coupled with an increased awareness of the role individual ribosomal components play in the translation of specific mRNAs, is opening a new field of study centered on ribosome-mediated control of gene regulation. In this Perspective, we discuss our current understanding of the known functions for ribosome heterogeneity, including specialized translation of individual transcripts, and its implications for the regulation and expression of key gene regulatory networks. In addition, we suggest what the crucial next steps are to ascertain the extent of ribosome heterogeneity and specialization and its importance for regulation of the proteome within subcellular space, across different cell types, and during multi-cellular organismal development.


Subject(s)
Ribosomes/metabolism , Ribosomes/physiology , Animals , Gene Expression Regulation , Gene Regulatory Networks , Humans , Internal Ribosome Entry Sites/physiology , Protein Biosynthesis , RNA/metabolism , RNA, Messenger/metabolism , Ribosomal Proteins/metabolism
13.
Mol Cell ; 72(6): 1013-1020.e6, 2018 12 20.
Article in English | MEDLINE | ID: mdl-30576652

ABSTRACT

Expansion segments (ESs) are enigmatic insertions within the eukaryotic ribosome, the longest of which resemble tentacle-like extensions that vary in length and sequence across evolution, with a largely unknown function. By selectively engineering rRNA in yeast, we find that one of the largest ESs, ES27L, has an unexpected function in translation fidelity. Ribosomes harboring a deletion in the distal portion of ES27L have increased amino acid misincorporation, as well as readthrough and frameshifting errors. By employing quantitative mass spectrometry, we further find that ES27L acts as an RNA scaffold to facilitate binding of a conserved enzyme, methionine amino peptidase (MetAP). We show that MetAP unexpectedly controls the accuracy of ribosome decoding, which is coupled to an increase in its enzymatic function through its interaction with ES27L. These findings reveal that variable ESs of the ribosome serve important functional roles and act as platforms for the binding of proteins that modulate translation across evolution.


Subject(s)
Caulobacter crescentus/metabolism , Mouse Embryonic Stem Cells/metabolism , RNA, Bacterial/metabolism , RNA, Fungal/metabolism , RNA, Ribosomal/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Aminopeptidases/metabolism , Animals , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Binding Sites , Caulobacter crescentus/genetics , Cell Line , Fungal Proteins/biosynthesis , Fungal Proteins/genetics , Mice , Nucleic Acid Conformation , Protein Binding , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Fungal/chemistry , RNA, Fungal/genetics , RNA, Ribosomal/chemistry , RNA, Ribosomal/genetics , Ribosomes/chemistry , Ribosomes/genetics , Saccharomyces cerevisiae/genetics , Structure-Activity Relationship
14.
Mol Cell ; 67(1): 71-83.e7, 2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28625553

ABSTRACT

Emerging studies have linked the ribosome to more selective control of gene regulation. However, an outstanding question is whether ribosome heterogeneity at the level of core ribosomal proteins (RPs) exists and enables ribosomes to preferentially translate specific mRNAs genome-wide. Here, we measured the absolute abundance of RPs in translating ribosomes and profiled transcripts that are enriched or depleted from select subsets of ribosomes within embryonic stem cells. We find that heterogeneity in RP composition endows ribosomes with differential selectivity for translating subpools of transcripts, including those controlling metabolism, cell cycle, and development. As an example, mRNAs enriched in binding to RPL10A/uL1-containing ribosomes are shown to require RPL10A/uL1 for their efficient translation. Within several of these transcripts, this level of regulation is mediated, at least in part, by internal ribosome entry sites. Together, these results reveal a critical functional link between ribosome heterogeneity and the post-transcriptional circuitry of gene expression.


Subject(s)
Embryonic Stem Cells/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Internal Ribosome Entry Sites , Protein Interaction Maps , RNA Interference , RNA, Messenger/genetics , Ribosomal Proteins/genetics , Ribosomes/genetics , Transcriptome , Transfection
15.
Nat Rev Genet ; 19(7): 431-452, 2018 07.
Article in English | MEDLINE | ID: mdl-29725087

ABSTRACT

Regulation of mRNA translation offers the opportunity to diversify the expression and abundance of proteins made from individual gene products in cells, tissues and organisms. Emerging evidence has highlighted variation in the composition and activity of several large, highly conserved translation complexes as a means to differentially control gene expression. Heterogeneity and specialized functions of individual components of the ribosome and of the translation initiation factor complexes eIF3 and eIF4F, which are required for recruitment of the ribosome to the mRNA 5' untranslated region, have been identified. In this Review, we summarize the evidence for selective mRNA translation by components of these macromolecular complexes as a means to dynamically control the translation of the proteome in time and space. We further discuss the implications of this form of gene expression regulation for a growing number of human genetic disorders associated with mutations in the translation machinery.


Subject(s)
5' Untranslated Regions/physiology , Eukaryotic Initiation Factor-3/metabolism , Eukaryotic Initiation Factor-4F/metabolism , Gene Expression Regulation/physiology , Peptide Chain Initiation, Translational/physiology , Ribosomes/metabolism , Animals , Humans , Ribosomes/genetics
16.
Nat Rev Mol Cell Biol ; 13(6): 355-69, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22617470

ABSTRACT

Historically, the ribosome has been viewed as a complex ribozyme with constitutive rather than intrinsic regulatory capacity in mRNA translation. However, emerging studies reveal that ribosome activity may be highly regulated. Heterogeneity in ribosome composition resulting from differential expression and post-translational modifications of ribosomal proteins, ribosomal RNA (rRNA) diversity and the activity of ribosome-associated factors may generate 'specialized ribosomes' that have a substantial impact on how the genomic template is translated into functional proteins. Moreover, constitutive components of the ribosome may also exert more specialized activities by virtue of their interactions with specific mRNA regulatory elements such as internal ribosome entry sites (IRESs) or upstream open reading frames (uORFs). Here we discuss the hypothesis that intrinsic regulation by the ribosome acts to selectively translate subsets of mRNAs harbouring unique cis-regulatory elements, thereby introducing an additional level of regulation in gene expression and the life of an organism.


Subject(s)
Cell Biology , Gene Expression Regulation , Ribosomes/metabolism , Animals , Humans , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Haematologica ; 108(5): 1222-1231, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36384250

ABSTRACT

Diamond-Blackfan anemia (DBA) is a ribosomopathy that is characterized by macrocytic anemia, congenital malformations, and early onset during childhood. Genetic studies have demonstrated that most patients carry mutations in one of the 20 related genes, most of which encode ribosomal proteins (RP). Treatment of DBA includes corticosteroid therapy, chronic red blood cell transfusion, and other forms of immunosuppression. Currently, hematopoietic stem cell transplantation is the only cure for DBA. Interestingly, spontaneous remissions occur in 10-20% of transfusion-dependent DBA patients. However, there is no consistent association between specific mutations and clinical manifestations. In the past decades, researchers have made significant progress in understanding the pathogenesis of DBA, but it remains unclear how the ubiquitous RP haploinsufficiency causes the erythroid-specific defect in hematopoiesis in DBA patients, and why there is a difference in penetrance and spontaneous remission among individuals who carry identical mutations. In this paper, we provide a comprehensive review of the development of DBA animal models and discuss the future research directions for these important experimental systems.


Subject(s)
Anemia, Diamond-Blackfan , Animals , Anemia, Diamond-Blackfan/genetics , Ribosomal Proteins/genetics , Mutation , Models, Animal , Hematopoiesis
19.
Nat Rev Mol Cell Biol ; 16(5): 268, 2015 May.
Article in English | MEDLINE | ID: mdl-25907611
20.
Nature ; 517(7532): 33-8, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25409156

ABSTRACT

Emerging evidence suggests that the ribosome has a regulatory function in directing how the genome is translated in time and space. However, how this regulation is encoded in the messenger RNA sequence remains largely unknown. Here we uncover unique RNA regulons embedded in homeobox (Hox) 5' untranslated regions (UTRs) that confer ribosome-mediated control of gene expression. These structured RNA elements, resembling viral internal ribosome entry sites (IRESs), are found in subsets of Hox mRNAs. They facilitate ribosome recruitment and require the ribosomal protein RPL38 for their activity. Despite numerous layers of Hox gene regulation, these IRES elements are essential for converting Hox transcripts into proteins to pattern the mammalian body plan. This specialized mode of IRES-dependent translation is enabled by an additional regulatory element that we term the translation inhibitory element (TIE), which blocks cap-dependent translation of transcripts. Together, these data uncover a new paradigm for ribosome-mediated control of gene expression and organismal development.


Subject(s)
5' Untranslated Regions/genetics , Gene Expression Regulation/genetics , Genes, Homeobox/genetics , Regulatory Sequences, Ribonucleic Acid/genetics , Ribosomes/metabolism , Animals , Bone and Bones/embryology , Bone and Bones/metabolism , Cell Line , Conserved Sequence , Evolution, Molecular , Mice , Molecular Sequence Data , Protein Biosynthesis/genetics , RNA Caps/metabolism , Ribosomal Proteins/metabolism , Ribosomes/chemistry , Substrate Specificity , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL