Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.200
Filter
1.
Int J Mol Sci ; 25(16)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39201491

ABSTRACT

The analysis of protein fold usage, similar to codon usage, offers profound insights into the evolution of biological systems and the origins of modern proteomes. While previous studies have examined fold distribution in modern genomes, our study focuses on the comparative distribution and usage of protein folds in ribosomes across bacteria, archaea, and eukaryotes. We identify the prevalence of certain 'super-ribosome folds,' such as the OB fold in bacteria and the SH3 domain in archaea and eukaryotes. The observed protein fold distribution in the ribosomes announces the future power-law distribution where only a few folds are highly prevalent, and most are rare. Additionally, we highlight the presence of three copies of proto-Rossmann folds in ribosomes across all kingdoms, showing its ancient and fundamental role in ribosomal structure and function. Our study also explores early mechanisms of molecular convergence, where different protein folds bind equivalent ribosomal RNA structures in ribosomes across different kingdoms. This comparative analysis enhances our understanding of ribosomal evolution, particularly the distinct evolutionary paths of the large and small subunits, and underscores the complex interplay between RNA and protein components in the transition from the RNA world to modern cellular life. Transcending the concept of folds also makes it possible to group a large number of ribosomal proteins into five categories of urfolds or metafolds, which could attest to their ancestral character and common origins. This work also demonstrates that the gradual acquisition of extensions by simple but ordered folds constitutes an inexorable evolutionary mechanism. This observation supports the idea that simple but structured ribosomal proteins preceded the development of their disordered extensions.


Subject(s)
Archaea , Evolution, Molecular , Protein Folding , Ribosomal Proteins , Ribosomes , Ribosomes/metabolism , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Archaea/metabolism , Archaea/genetics , Eukaryota/metabolism , Eukaryota/genetics , Bacteria/metabolism , Bacteria/genetics , RNA, Ribosomal/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/chemistry
2.
Biomolecules ; 14(8)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39199362

ABSTRACT

Here we review the functions of ribosomal proteins (RPs) in the nucleolar stages of large ribosomal subunit assembly in the yeast Saccharomyces cerevisiae. We summarize the effects of depleting RPs on pre-rRNA processing and turnover, on the assembly of other RPs, and on the entry and exit of assembly factors (AFs). These results are interpreted in light of recent near-atomic-resolution cryo-EM structures of multiple assembly intermediates. Results are discussed with respect to each neighborhood of RPs and rRNA. We identify several key mechanisms related to RP behavior. Neighborhoods of RPs can assemble in one or more than one step. Entry of RPs can be triggered by molecular switches, in which an AF is replaced by an RP binding to the same site. To drive assembly forward, rRNA structure can be stabilized by RPs, including clamping rRNA structures or forming bridges between rRNA domains.


Subject(s)
RNA, Ribosomal , Ribosomal Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , RNA, Ribosomal/metabolism , RNA, Ribosomal/chemistry , RNA, Ribosomal/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Cell Nucleolus/metabolism
3.
Nat Commun ; 15(1): 5938, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025855

ABSTRACT

Numerous molecular machines are required to drive the central dogma of molecular biology. However, the means by which these numerous proteins emerged in the early evolutionary stage of life remains enigmatic. Many of them possess small ß-barrel folds with different topologies, represented by double-psi ß-barrels (DPBBs) conserved in DNA and RNA polymerases, and similar but topologically distinct six-stranded ß-barrel RIFT or five-stranded ß-barrel folds such as OB and SH3 in ribosomal proteins. Here, we discover that the previously reconstructed ancient DPBB sequence could also adopt a ß-barrel fold named Double-Zeta ß-barrel (DZBB), as a metamorphic protein. The DZBB fold is not found in any modern protein, although its structure shares similarities with RIFT and OB. Indeed, DZBB could be transformed into them through simple engineering experiments. Furthermore, the OB designs could be further converted into SH3 by circular-permutation as previously predicted. These results indicate that these ß-barrels diversified quickly from a common ancestor at the beginning of the central dogma evolution.


Subject(s)
DNA-Directed RNA Polymerases , Evolution, Molecular , Models, Molecular , Ribosomal Proteins , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/chemistry , Protein Folding , Amino Acid Sequence
4.
J Phys Chem B ; 128(29): 7002-7021, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39012038

ABSTRACT

Ribosomes bind to many metabolic enzymes and change their activity. A general mechanism for ribosome-mediated amplification of metabolic enzyme activity, RAMBO, was formulated and elucidated for the glycolytic enzyme triosephosphate isomerase, TPI. The RAMBO effect results from a ribosome-dependent electric field-substrate dipole interaction energy that can increase or decrease the ground state of the reactant and product to regulate catalytic rates. NMR spectroscopy was used to determine the interaction surface of TPI binding to ribosomes and to measure the corresponding kinetic rates in the absence and presence of intact ribosome particles. Chemical cross-linking and mass spectrometry revealed potential ribosomal protein binding partners of TPI. Structural results and related changes in TPI energetics and activity show that the interaction between TPI and ribosomal protein L11 mediate the RAMBO effect.


Subject(s)
Ribosomes , Triose-Phosphate Isomerase , Triose-Phosphate Isomerase/metabolism , Triose-Phosphate Isomerase/chemistry , Ribosomes/metabolism , Ribosomes/chemistry , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Kinetics , Electricity , Protein Binding
5.
Nature ; 632(8025): 656-663, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39048817

ABSTRACT

Dysregulated transcription due to disruption in histone lysine methylation dynamics is an established contributor to tumorigenesis1,2. However, whether analogous pathologic epigenetic mechanisms act directly on the ribosome to advance oncogenesis is unclear. Here we find that trimethylation of the core ribosomal protein L40 (rpL40) at lysine 22 (rpL40K22me3) by the lysine methyltransferase SMYD5 regulates mRNA translation output to promote malignant progression of gastric adenocarcinoma (GAC) with lethal peritoneal ascites. A biochemical-proteomics strategy identifies the monoubiquitin fusion protein partner rpL40 (ref. 3) as the principal physiological substrate of SMYD5 across diverse samples. Inhibiting the SMYD5-rpL40K22me3 axis in GAC cell lines reprogrammes protein synthesis to attenuate oncogenic gene expression signatures. SMYD5 and rpL40K22me3 are upregulated in samples from patients with GAC and negatively correlate with clinical outcomes. SMYD5 ablation in vivo in familial and sporadic mouse models of malignant GAC blocks metastatic disease, including peritoneal carcinomatosis. Suppressing SMYD5 methylation of rpL40 inhibits human cancer cell and patient-derived GAC xenograft growth and renders them hypersensitive to inhibitors of PI3K and mTOR. Finally, combining SMYD5 depletion with PI3K-mTOR inhibition and chimeric antigen receptor T cell administration cures an otherwise lethal in vivo mouse model of aggressive GAC-derived peritoneal carcinomatosis. Together, our work uncovers a ribosome-based epigenetic mechanism that facilitates the evolution of malignant GAC and proposes SMYD5 targeting as part of a potential combination therapy to treat this cancer.


Subject(s)
Methyltransferases , Ribosomal Proteins , Ribosomes , Stomach Neoplasms , Animals , Female , Humans , Mice , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Lysine/metabolism , Methylation/drug effects , Methyltransferases/antagonists & inhibitors , Methyltransferases/deficiency , Methyltransferases/metabolism , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Protein Biosynthesis , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Ribosomes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Nucleic Acids Res ; 52(15): 9028-9048, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39041433

ABSTRACT

Increasingly many studies reveal how ribosome composition can be tuned to optimally translate the transcriptome of individual cell types. In this study, we investigated the expression pattern, structure within the ribosome and effect on protein synthesis of the ribosomal protein paralog 39L (RPL39L). With a novel mass spectrometric approach we revealed the expression of RPL39L protein beyond mouse germ cells, in human pluripotent cells, cancer cell lines and tissue samples. We generated RPL39L knock-out mouse embryonic stem cell (mESC) lines and demonstrated that RPL39L impacts the dynamics of translation, to support the pluripotency and differentiation, spontaneous and along the germ cell lineage. Most differences in protein abundance between WT and RPL39L KO lines were explained by widespread autophagy. By CryoEM analysis of purified RPL39 and RPL39L-containing ribosomes we found that, unlike RPL39, RPL39L has two distinct conformations in the exposed segment of the nascent peptide exit tunnel, creating a distinct hydrophobic patch that has been predicted to support the efficient co-translational folding of alpha helices. Our study shows that ribosomal protein paralogs provide switchable modular components that can tune translation to the protein production needs of individual cell types.


Subject(s)
Protein Biosynthesis , Protein Folding , Ribosomal Proteins , Ribosomes , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/chemistry , Animals , Mice , Humans , Ribosomes/metabolism , Protein Conformation, alpha-Helical , Mice, Knockout , Mouse Embryonic Stem Cells/metabolism , Cell Differentiation/genetics
7.
J Biol Chem ; 300(8): 107519, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38950860

ABSTRACT

The mitochondrial ribosome (mitoribosome) is responsible for the synthesis of key oxidative phosphorylation subunits encoded by the mitochondrial genome. Defects in mitoribosomal function therefore can have serious consequences for the bioenergetic capacity of the cell. Mutation of the conserved mitoribosomal mL44 protein has been directly linked to childhood cardiomyopathy and progressive neurophysiology issues. To further explore the functional significance of the mL44 protein in supporting mitochondrial protein synthesis, we have performed a mutagenesis study of the yeast mL44 homolog, the MrpL3/mL44 protein. We specifically investigated the conserved hydrophobic pocket region of the MrpL3/mL44 protein, where the known disease-related residue in the human mL44 protein (L156R) is located. While our findings identify a number of residues in this region critical for MrpL3/mL44's ability to support the assembly of translationally active mitoribosomes, the introduction of the disease-related mutation into the equivalent position in the yeast protein (residue A186) was found to not have a major impact on function. The human and yeast mL44 proteins share many similarities in sequence and structure; however results presented here indicate that these two proteins have diverged somewhat in evolution. Finally, we observed that mutation of the MrpL3/mL44 does not impact the translation of all mitochondrial encoded proteins equally, suggesting the mitochondrial translation system may exhibit a transcript hierarchy and prioritization.


Subject(s)
Mitochondrial Proteins , Mitochondrial Ribosomes , Protein Biosynthesis , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/chemistry , Humans , Mitochondrial Ribosomes/metabolism , Mitochondrial Ribosomes/chemistry , Hydrophobic and Hydrophilic Interactions , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/chemistry , Mitochondria/metabolism , Mitochondria/genetics
8.
Int J Biol Macromol ; 277(Pt 3): 134213, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39069039

ABSTRACT

African swine fever virus (ASFV) severely threatens the global economy and food security. ASFV encodes >150 genes, but the functions of most of them have yet to be characterized in detail. Here we explored the function of the ASFV CP312R gene and found that CP312R plays an essential role in ASFV replication. Knockout of the CP312R gene terminated viral replication and CP312R knockdown substantially suppressed ASFV infection in vitro. Furthermore, we resolved the crystal structure of pCP312R to 2.3 Å resolution and found that pCP312R has the potential to bind nucleic acids. LC-MS analysis and co-immunoprecipitation assay revealed that pCP312R interacts with RPS27A, a component of the 40S ribosomal subunit. Confocal microscopy showed the interaction between pCP312R and RPS27A leaded to a modification in the subcellular localization of this host protein, which suppresses host protein translation. Renilla-Glo luciferase assay and Ribopuromycylation analysis evidenced that knockout of RPS27A completely aborted the shutoff activity of pCP312R, and trans-complementation of RPS27A recovered pCP312R shutoff activity in RPS27A-knockout cells. Our findings shed light on the function of ASFV CP312R gene in virus infection, which triggers inhibition of host protein synthesis.


Subject(s)
African Swine Fever Virus , Protein Biosynthesis , Ribosomal Proteins , Viral Proteins , Virus Replication , African Swine Fever Virus/metabolism , African Swine Fever Virus/genetics , Animals , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/chemistry , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/chemistry , Swine , Host-Pathogen Interactions , Protein Binding , Chlorocebus aethiops , Vero Cells , African Swine Fever/virology , African Swine Fever/metabolism
9.
Biomolecules ; 14(7)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39062596

ABSTRACT

Ribosomes are not totally globular machines. Instead, they comprise prominent structural protrusions and a myriad of tentacle-like projections, which are frequently made up of ribosomal RNA expansion segments and N- or C-terminal extensions of ribosomal proteins. This is more evident in higher eukaryotic ribosomes. One of the most characteristic protrusions, present in small ribosomal subunits in all three domains of life, is the so-called beak, which is relevant for the function and regulation of the ribosome's activities. During evolution, the beak has transitioned from an all ribosomal RNA structure (helix h33 in 16S rRNA) in bacteria, to an arrangement formed by three ribosomal proteins, eS10, eS12 and eS31, and a smaller h33 ribosomal RNA in eukaryotes. In this review, we describe the different structural and functional properties of the eukaryotic beak. We discuss the state-of-the-art concerning its composition and functional significance, including other processes apparently not related to translation, and the dynamics of its assembly in yeast and human cells. Moreover, we outline the current view about the relevance of the beak's components in human diseases, especially in ribosomopathies and cancer.


Subject(s)
Ribosomes , Humans , Ribosomes/metabolism , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Eukaryota/metabolism , RNA, Ribosomal/metabolism , RNA, Ribosomal/chemistry , RNA, Ribosomal/genetics , Animals
10.
Elife ; 132024 Jun 18.
Article in English | MEDLINE | ID: mdl-38896445

ABSTRACT

The protein translocon at the endoplasmic reticulum comprises the Sec61 translocation channel and numerous accessory factors that collectively facilitate the biogenesis of secretory and membrane proteins. Here, we leveraged recent advances in cryo-electron microscopy (cryo-EM) and structure prediction to derive insights into several novel configurations of the ribosome-translocon complex. We show how a transmembrane domain (TMD) in a looped configuration passes through the Sec61 lateral gate during membrane insertion; how a nascent chain can bind and constrain the conformation of ribosomal protein uL22; and how the translocon-associated protein (TRAP) complex can adjust its position during different stages of protein biogenesis. Most unexpectedly, we find that a large proportion of translocon complexes contains RAMP4 intercalated into Sec61's lateral gate, widening Sec61's central pore and contributing to its hydrophilic interior. These structures lead to mechanistic hypotheses for translocon function and highlight a remarkably plastic machinery whose conformations and composition adjust dynamically to its diverse range of substrates.


Subject(s)
Cryoelectron Microscopy , Ribosomes , SEC Translocation Channels , Ribosomes/metabolism , Ribosomes/chemistry , Ribosomes/ultrastructure , SEC Translocation Channels/metabolism , SEC Translocation Channels/chemistry , Endoplasmic Reticulum/metabolism , Protein Conformation , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Humans , Models, Molecular , Protein Transport , Membrane Proteins/metabolism , Membrane Proteins/chemistry
11.
Nucleic Acids Res ; 52(13): 7947-7960, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38842932

ABSTRACT

Ribosome biogenesis is a highly regulated cellular process that involves the control of numerous assembly factors. The small protein YjgA has been reported to play a role in the late stages of 50S assembly. However, the precise molecular mechanism underlying its function remains unclear. In this study, cryo-electron microscopy (cryo-EM) structures revealed that depletion of YjgA or its N-terminal loop in Escherichia coli both lead to the accumulation of immature 50S particles with structural abnormalities mainly in peptidyl transferase center (PTC) and H68/69 region. CryoDRGN analysis uncovered 8 and 6 distinct conformations of pre50S for ΔyjgA and YjgA-ΔNloop, respectively. These conformations highlighted the role of the N-terminal loop of YjgA in integrating uL16 and stabilizing H89 in PTC, which was further verified by the pull-down assays of YjgA and its mutants with uL16. Together with the function of undocking H68 through the binding of its C-terminal CTLH-like domain to the base of the L1 stalk, YjgA facilitates the maturation of PTC. This study identified critical domains of YjgA contributing to 50S assembly efficiency, providing a comprehensive understanding of the dual roles of YjgA in accelerating ribosome biogenesis and expanding our knowledge of the intricate processes governing cellular protein synthesis.


Subject(s)
Cryoelectron Microscopy , Escherichia coli Proteins , Escherichia coli , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Peptidyl Transferases/metabolism , Peptidyl Transferases/genetics , Ribosomes/metabolism , Ribosomes/genetics , Ribosome Subunits, Large, Bacterial/metabolism , Ribosome Subunits, Large, Bacterial/genetics , Ribosome Subunits, Large, Bacterial/chemistry , Models, Molecular , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/chemistry , Protein Binding
12.
Biophys Chem ; 312: 107273, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38850843

ABSTRACT

Bacillus anthracis, the causative agent of anthrax, poses a substantial threat to public health and national security, and is recognized as a potential bioweapon due to its capacity to form resilient spores with enduring viability. Inhalation or ingestion of even minute quantities of aerosolized spores can lead to widespread illness and fatalities, underscoring the formidable lethality of the bacterium. With an untreated mortality rate of 100%, Bacillus anthracis is a disconcerting candidate for bioterrorism. In response to this critical scenario, we employed state-of-the-art computational tools to conceive and characterize flexible RNA aptamer therapeutics tailored for anthrax. The foundational structure of the flexible RNA aptamers was designed by removing the C2'-C3' in each nucleotide unit. Leveraging the crystal structure of Bacillus anthracis ribosomal protein S8 complexed with an RNA aptamer, we explored the structural, dynamic, and energetic aspects of the modified RNA aptamer - S8 protein complexes through extensive all-atom explicit-solvent molecular dynamics simulations (400 ns, 3 replicas each), followed by drawing comparisons to the control system. Our findings demonstrate the enhanced binding competencies of the flexible RNA aptamers to the S8 protein via better shape complementarity and improved H-bond network compared to the control RNA aptamer. This research offers valuable insights into the development of RNA aptamer therapeutics targeting Bacillus anthracis, paving the way for innovative strategies to mitigate the impact of this formidable pathogen.


Subject(s)
Aptamers, Nucleotide , Bacillus anthracis , Molecular Dynamics Simulation , Ribosomal Proteins , Bacillus anthracis/chemistry , Bacillus anthracis/metabolism , Bacillus anthracis/drug effects , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacology , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Ribosomal Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/antagonists & inhibitors
13.
Annu Rev Biochem ; 93(1): 189-210, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38768392

ABSTRACT

During the last ten years, developments in cryo-electron microscopy have transformed our understanding of eukaryotic ribosome assembly. As a result, the field has advanced from a list of the vast array of ribosome assembly factors toward an emerging molecular movie in which individual frames are represented by structures of stable ribosome assembly intermediates with complementary biochemical and genetic data. In this review, we discuss the mechanisms driving the assembly of yeast and human small and large ribosomal subunits. A particular emphasis is placed on the most recent findings that illustrate key concepts of ribosome assembly, such as folding of preribosomal RNA, the enforced chronology of assembly, enzyme-mediated irreversible transitions, and proofreading of preribosomal particles.


Subject(s)
Cryoelectron Microscopy , Ribosomal Proteins , Ribosomes , Humans , Ribosomes/metabolism , Ribosomes/ultrastructure , Ribosomes/chemistry , Ribosomes/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , RNA, Ribosomal/metabolism , RNA, Ribosomal/chemistry , RNA, Ribosomal/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Models, Molecular , Eukaryotic Cells/metabolism , Eukaryotic Cells/ultrastructure , RNA Folding , Ribosome Subunits, Small, Eukaryotic/metabolism , Ribosome Subunits, Small, Eukaryotic/chemistry , Ribosome Subunits, Small, Eukaryotic/genetics , Ribosome Subunits, Small, Eukaryotic/ultrastructure , Animals
14.
Sci Rep ; 14(1): 12324, 2024 05 29.
Article in English | MEDLINE | ID: mdl-38811604

ABSTRACT

In order to become bioactive, proteins must be translated and protected from aggregation during biosynthesis. The ribosome and molecular chaperones play a key role in this process. Ribosome-bound nascent chains (RNCs) of intrinsically disordered proteins and RNCs bearing a signal/arrest sequence are known to interact with ribosomal proteins. However, in the case of RNCs bearing foldable protein sequences, not much information is available on these interactions. Here, via a combination of chemical crosslinking and time-resolved fluorescence-anisotropy, we find that nascent chains of the foldable globin apoHmp1-140 interact with ribosomal protein L23 and have a freely-tumbling non-interacting N-terminal compact region comprising 63-94 residues. Longer RNCs (apoHmp1-189) also interact with an additional yet unidentified ribosomal protein, as well as with chaperones. Surprisingly, the apparent strength of RNC/r-protein interactions does not depend on nascent-chain sequence. Overall, foldable nascent chains establish and expand interactions with selected ribosomal proteins and chaperones, as they get longer. These data are significant because they reveal the interplay between independent conformational sampling and nascent-protein interactions with the ribosomal surface.


Subject(s)
Protein Folding , Ribosomal Proteins , Ribosomes , Ribosomes/metabolism , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Protein Binding , Molecular Chaperones/metabolism , Molecular Chaperones/chemistry , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/chemistry , Protein Biosynthesis , Models, Molecular , Protein Conformation , Humans
15.
Nat Commun ; 15(1): 4272, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769321

ABSTRACT

The mitoribosome translates mitochondrial mRNAs and regulates energy conversion that is a signature of aerobic life forms. We present a 2.2 Å resolution structure of human mitoribosome together with validated mitoribosomal RNA (rRNA) modifications, including aminoacylated CP-tRNAVal. The structure shows how mitoribosomal proteins stabilise binding of mRNA and tRNA helping to align it in the decoding center, whereas the GDP-bound mS29 stabilizes intersubunit communication. Comparison between different states, with respect to tRNA position, allowed us to characterize a non-canonical L1 stalk, and molecular dynamics simulations revealed how it facilitates tRNA transitions in a way that does not require interactions with rRNA. We also report functionally important polyamines that are depleted when cells are subjected to an antibiotic treatment. The structural, biochemical, and computational data illuminate the principal functional components of the translation mechanism in mitochondria and provide a description of the structure and function of the human mitoribosome.


Subject(s)
Mitochondrial Ribosomes , RNA, Transfer , Humans , RNA, Transfer/metabolism , RNA, Transfer/chemistry , RNA, Transfer/genetics , Mitochondrial Ribosomes/metabolism , Mitochondrial Ribosomes/chemistry , Ligands , Molecular Dynamics Simulation , RNA, Messenger/metabolism , RNA, Messenger/genetics , Mitochondria/metabolism , RNA, Ribosomal/metabolism , RNA, Ribosomal/chemistry , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Guanosine Diphosphate/metabolism , Polyamines/metabolism , Polyamines/chemistry , Protein Binding
16.
Biomol Biomed ; 24(5): 1117-1132, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-38606907

ABSTRACT

The 67 kDa laminin receptor (67LR) was identified as the first laminin receptor shown to be involved in the carcinogenesis of various cancers, including colorectal cancer. While the exact composition of this 67 kDa receptor remains unknown, it has been reported to be formed by the 37 kDa ribosomal protein SA (RPSA) covalently attached to another unidentified protein. The goal of this study was to clarify the molecular structure of 67LR to enhance our understanding of its role in malignancies. Using cell fractionation of colorectal cancer cells, the 67 kDa immunoreactive protein corresponding to 67LR was found in the soluble protein fraction, while some of the 37 kDa RPSA exhibited plasma membrane-like properties. Proteomic analysis of the 67 kDa fraction revealed the absence of RPSA but identified the ß-galactosidase-related 67 kDa elastin-binding protein (67EBP), another laminin binding receptor which presents amino acid sequence similarities that can explain the immune cross reactivity with RPSA. The downregulation of ß-galactosidase through short hairpin RNA (shRNA) led to a reduction in both 67LR and 67EBP immunoreactive proteins, confirming the misidentification of 67LR and 67EBP in colorectal cancer cells. Based on these findings, we propose to redefine the 67LR as the RPSA-containing laminin receptor (RCLR) to avoid confusion with the 67EBP.


Subject(s)
Colorectal Neoplasms , Receptors, Laminin , Ribosomal Proteins , Humans , Receptors, Laminin/metabolism , Receptors, Laminin/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Cell Line, Tumor , beta-Galactosidase/metabolism , beta-Galactosidase/genetics
17.
Structure ; 32(5): 562-574.e3, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38458197

ABSTRACT

Plants being sessile organisms exhibit unique features in ribosomes, which might aid in rapid gene expression and regulation in response to varying environmental conditions. Here, we present high-resolution structures of the 60S and 80S ribosomes from wheat, a monocot staple crop plant (Triticum aestivum). While plant ribosomes have unique plant-specific rRNA modification (Cm1847) in the peptide exit tunnel (PET), the zinc-finger motif in eL34 is absent, and uL4 is extended, making an exclusive interaction network. We note differences in the eL15-helix 11 (25S) interaction, eL6-ES7 assembly, and certain rRNA chemical modifications between monocot and dicot ribosomes. In eukaryotes, we observe highly conserved rRNA modification (Gm75) in 5.8S rRNA and a flipped base (G1506) in PET. These features are likely involved in sensing or stabilizing nascent chain. Finally, we discuss the importance of the universal conservation of three consecutive rRNA modifications in all ribosomes for their interaction with A-site aminoacyl-tRNA.


Subject(s)
Cryoelectron Microscopy , Models, Molecular , RNA, Ribosomal , Ribosomes , Triticum , Triticum/metabolism , Triticum/genetics , Ribosomes/metabolism , RNA, Ribosomal/metabolism , RNA, Ribosomal/chemistry , Ribosomal Proteins/metabolism , Ribosomal Proteins/chemistry , Plant Proteins/metabolism , Plant Proteins/chemistry , Plant Proteins/genetics , Binding Sites , Nucleic Acid Conformation
18.
Cell ; 187(5): 1314-1314.e1, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38428399

ABSTRACT

Ribosome production is essential for cell growth. Approximately 200 assembly factors drive this complicated pathway that starts in the nucleolus and ends in the cytoplasm. A large number of structural snapshots of the pre-60S pathway have revealed the principles behind large subunit synthesis. To view this SnapShot, open or download the PDF.


Subject(s)
Cell Nucleolus , Eukaryotic Cells , Ribosomes , Cell Nucleolus/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/chemistry , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosomes/metabolism , Eukaryotic Cells/chemistry , Eukaryotic Cells/cytology , Eukaryotic Cells/metabolism
19.
Biosystems ; 238: 105196, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38537772

ABSTRACT

Protein domain repeats are known to arise due to tandem duplications of internal genes. However, the understanding of the underlying mechanisms of this process is incomplete. The goal of this work was to investigate the mechanism of occurrence of repeat expansion based on studying the sequences of 1324 rpsA genes of bacterial S1 ribosomal proteins containing different numbers of S1 structural domains. The rpsA gene encodes ribosomal S1 protein, which is essential for cell viability as it interacts with both mRNA and proteins. Gene ontology (GO) analysis of S1 domains in ribosomal S1 proteins revealed that bacterial protein sequences in S1 mainly have 3 types of molecular functions: RNA binding activity, nucleic acid activity, and ribosome structural component. Our results show that the maximum value of rpsA gene identity for full-length proteins was found for S1 proteins containing six structural domains (58%). Analysis of consensus sequences showed that parts of the rpsA gene encoding separate S1 domains have no a strictly repetitive structure between groups containing different numbers of S1 domains. At the same time, gene regions encoding some conserved residues that form the RNA-binding site remain conserved. The detected phylogenetic similarity suggests that the proposed fold of the rpsA translation initiation region of Escherichia coli has functional value and is important for translational control of rpsA gene expression in other bacterial phyla, but not only in gamma Proteobacteria.


Subject(s)
Bacteria , Ribosomal Proteins , Base Sequence , Phylogeny , Base Composition , Ribosomal Proteins/genetics , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Bacteria/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , RNA/metabolism , Structure-Activity Relationship
20.
Nature ; 626(8001): 1125-1132, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38355796

ABSTRACT

To conserve energy during starvation and stress, many organisms use hibernation factor proteins to inhibit protein synthesis and protect their ribosomes from damage1,2. In bacteria, two families of hibernation factors have been described, but the low conservation of these proteins and the huge diversity of species, habitats and environmental stressors have confounded their discovery3-6. Here, by combining cryogenic electron microscopy, genetics and biochemistry, we identify Balon, a new hibernation factor in the cold-adapted bacterium Psychrobacter urativorans. We show that Balon is a distant homologue of the archaeo-eukaryotic translation factor aeRF1 and is found in 20% of representative bacteria. During cold shock or stationary phase, Balon occupies the ribosomal A site in both vacant and actively translating ribosomes in complex with EF-Tu, highlighting an unexpected role for EF-Tu in the cellular stress response. Unlike typical A-site substrates, Balon binds to ribosomes in an mRNA-independent manner, initiating a new mode of ribosome hibernation that can commence while ribosomes are still engaged in protein synthesis. Our work suggests that Balon-EF-Tu-regulated ribosome hibernation is a ubiquitous bacterial stress-response mechanism, and we demonstrate that putative Balon homologues in Mycobacteria bind to ribosomes in a similar fashion. This finding calls for a revision of the current model of ribosome hibernation inferred from common model organisms and holds numerous implications for how we understand and study ribosome hibernation.


Subject(s)
Bacterial Proteins , Cold-Shock Response , Peptide Termination Factors , Protein Biosynthesis , Psychrobacter , Ribosomal Proteins , Ribosomes , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Peptide Elongation Factor Tu/chemistry , Peptide Elongation Factor Tu/metabolism , Peptide Elongation Factor Tu/ultrastructure , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/ultrastructure , Ribosomes/chemistry , Ribosomes/metabolism , Ribosomes/ultrastructure , Psychrobacter/chemistry , Psychrobacter/genetics , Psychrobacter/metabolism , Psychrobacter/ultrastructure , Cryoelectron Microscopy , Peptide Termination Factors/chemistry , Peptide Termination Factors/genetics , Peptide Termination Factors/metabolism , Peptide Termination Factors/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL