Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
2.
J Cell Biol ; 221(2)2022 02 07.
Article in English | MEDLINE | ID: mdl-35024764

ABSTRACT

The repertoire of extratranslational functions of components of the protein synthesis apparatus is expanding to include control of key cell signaling networks. However, very little is known about noncanonical functions of members of the protein synthesis machinery in regulating cellular mechanics. We demonstrate that the eukaryotic initiation factor 6 (eIF6) modulates cellular mechanobiology. eIF6-depleted endothelial cells, under basal conditions, exhibit unchanged nascent protein synthesis, polysome profiles, and cytoskeleton protein expression, with minimal effects on ribosomal biogenesis. In contrast, using traction force and atomic force microscopy, we show that loss of eIF6 leads to reduced stiffness and force generation accompanied by cytoskeletal and focal adhesion defects. Mechanistically, we show that eIF6 is required for the correct spatial mechanoactivation of ERK1/2 via stabilization of an eIF6-RACK1-ERK1/2-FAK mechanocomplex, which is necessary for force-induced remodeling. These results reveal an extratranslational function for eIF6 and a novel paradigm for how mechanotransduction, the cellular cytoskeleton, and protein translation constituents are linked.


Subject(s)
Endothelial Cells/metabolism , Mechanotransduction, Cellular , Peptide Initiation Factors/metabolism , Animals , Biomechanical Phenomena , Cattle , Cytoskeleton/metabolism , Focal Adhesions/metabolism , HEK293 Cells , Humans , MAP Kinase Signaling System , Mice , Protein Biosynthesis , Ribosomes/metabolism
3.
Sci Adv ; 7(28)2021 Jul.
Article in English | MEDLINE | ID: mdl-34244146

ABSTRACT

The response of endothelial cells to mechanical forces is a critical determinant of vascular health. Vascular pathologies, such as atherosclerosis, characterized by abnormal mechanical forces are frequently accompanied by endothelial-to-mesenchymal transition (EndMT). However, how forces affect the mechanotransduction pathways controlling cellular plasticity, inflammation, and, ultimately, vessel pathology is poorly understood. Here, we identify a mechanoreceptor that is sui generis for EndMT and unveil a molecular Alk5-Shc pathway that leads to EndMT and atherosclerosis. Depletion of Alk5 abrogates shear stress-induced EndMT responses, and genetic targeting of endothelial Shc reduces EndMT and atherosclerosis in areas of disturbed flow. Tensional force and reconstitution experiments reveal a mechanosensory function for Alk5 in EndMT signaling that is unique and independent of other mechanosensors. Our findings are of fundamental importance for understanding how mechanical forces regulate biochemical signaling, cell plasticity, and vascular disease.

4.
Cells ; 9(3)2020 03 07.
Article in English | MEDLINE | ID: mdl-32156009

ABSTRACT

Mechanical forces acting on biological systems, at both the macroscopic and microscopic levels, play an important part in shaping cellular phenotypes. There is a growing realization that biomolecules that respond to force directly applied to them, or via mechano-sensitive signalling pathways, can produce profound changes to not only transcriptional pathways, but also in protein translation. Forces naturally occurring at the molecular level can impact the rate at which the bacterial ribosome translates messenger RNA (mRNA) transcripts and influence processes such as co-translational folding of a nascent protein as it exits the ribosome. In eukaryotes, force can also be transduced at the cellular level by the cytoskeleton, the cell's internal filamentous network. The cytoskeleton closely associates with components of the translational machinery such as ribosomes and elongation factors and, as such, is a crucial determinant of localized protein translation. In this review we will give (1) a brief overview of protein translation in bacteria and eukaryotes and then discuss (2) how mechanical forces are directly involved with ribosomes during active protein synthesis and (3) how eukaryotic ribosomes and other protein translation machinery intimately associates with the mechanosensitive cytoskeleton network.


Subject(s)
Eukaryotic Cells/metabolism , Protein Biosynthesis/physiology , Proteins/metabolism , RNA, Messenger/metabolism , Ribosomes/metabolism , Humans , Phenotype , RNA, Messenger/genetics , Ribosomes/genetics
5.
Front Cell Dev Biol ; 8: 34, 2020.
Article in English | MEDLINE | ID: mdl-32083081

ABSTRACT

The cardiovascular system can sense and adapt to changes in mechanical stimuli by remodeling the physical properties of the heart and blood vessels in order to maintain homeostasis. Imbalances in mechanical forces and/or impaired sensing are now not only implicated but are, in some cases, considered to be drivers for the development and progression of cardiovascular disease. There is now growing evidence to highlight the role of mechanical forces in the regulation of protein translation pathways. The canonical mechanism of protein synthesis typically involves transcription and translation. Protein translation occurs globally throughout the cell to maintain general function but localized protein synthesis allows for precise spatiotemporal control of protein translation. This Review will cover studies on the role of biomechanical stress -induced translational control in the heart (often in the context of physiological and pathological hypertrophy). We will also discuss the much less studied effects of mechanical forces in regulating protein translation in the vasculature. Understanding how the mechanical environment influences protein translational mechanisms in the cardiovascular system, will help to inform disease pathogenesis and potential areas of therapeutic intervention.

6.
Nature ; 578(7794): 290-295, 2020 02.
Article in English | MEDLINE | ID: mdl-32025034

ABSTRACT

Shear stress on arteries produced by blood flow is important for vascular development and homeostasis but can also initiate atherosclerosis1. Endothelial cells that line the vasculature use molecular mechanosensors to directly detect shear stress profiles that will ultimately lead to atheroprotective or atherogenic responses2. Plexins are key cell-surface receptors of the semaphorin family of cell-guidance signalling proteins and can regulate cellular patterning by modulating the cytoskeleton and focal adhesion structures3-5. However, a role for plexin proteins in mechanotransduction has not been examined. Here we show that plexin D1 (PLXND1) has a role in mechanosensation and mechanically induced disease pathogenesis. PLXND1 is required for the response of endothelial cells to shear stress in vitro and in vivo and regulates the site-specific distribution of atherosclerotic lesions. In endothelial cells, PLXND1 is a direct force sensor and forms a mechanocomplex with neuropilin-1 and VEGFR2 that is necessary and sufficient for conferring mechanosensitivity upstream of the junctional complex and integrins. PLXND1 achieves its binary functions as either a ligand or a force receptor by adopting two distinct molecular conformations. Our results establish a previously undescribed mechanosensor in endothelial cells that regulates cardiovascular pathophysiology, and provide a mechanism by which a single receptor can exhibit a binary biochemical nature.


Subject(s)
Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mechanotransduction, Cellular , Membrane Glycoproteins/metabolism , Stress, Mechanical , Animals , Atherosclerosis/metabolism , Female , Integrins/metabolism , Mice , Neuropilin-1/metabolism , Pliability , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
7.
Sci Rep ; 7: 41223, 2017 01 25.
Article in English | MEDLINE | ID: mdl-28120882

ABSTRACT

Fibronectin (FN) assembly and fibrillogenesis are critically important in both development and the adult organism, but their importance in vascular functions is not fully understood. Here we identify a novel pathway by which haemodynamic forces regulate FN assembly and fibrillogenesis during vascular remodelling. Induction of disturbed shear stress in vivo and in vitro resulted in complex FN fibril assembly that was dependent on the mechanosensor PECAM. Loss of PECAM also inhibited the cell-intrinsic ability to remodel FN. Gain- and loss-of-function experiments revealed that PECAM-dependent RhoA activation is required for FN assembly. Furthermore, PECAM-/- mice exhibited reduced levels of active ß1 integrin that were responsible for reduced RhoA activation and downstream FN assembly. These data identify a new pathway by which endothelial mechanotransduction regulates FN assembly and flow-mediated vascular remodelling.


Subject(s)
Carotid Arteries/metabolism , Fibronectins/metabolism , Hemodynamics , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Animals , Carotid Arteries/pathology , Carotid Arteries/physiology , Cattle , Cells, Cultured , Integrin beta1/metabolism , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vascular Remodeling , rhoA GTP-Binding Protein/metabolism
8.
PLoS One ; 9(10): e109325, 2014.
Article in English | MEDLINE | ID: mdl-25296172

ABSTRACT

A naturally-occurring fragment of tyrosyl-tRNA synthetase (TyrRS) has been shown in higher eukaryotes to 'moonlight' as a pro-angiogenic cytokine in addition to its primary role in protein translation. Pro-angiogenic cytokines have previously been proposed to be promising therapeutic mechanisms for the treatment of myocardial infarction. Here, we show that systemic delivery of the natural fragment of TyRS, mini-TyrRS, improves heart function in mice after myocardial infarction. This improvement is associated with reduced formation of scar tissue, increased angiogenesis of cardiac capillaries, recruitment of c-kitpos cells and proliferation of myocardial fibroblasts. This work demonstrates that mini-TyrRS has beneficial effects on cardiac repair and regeneration and offers support for the notion that elucidation of the ever expanding repertoire of noncanonical functions of aminoacyl tRNA synthetases offers unique opportunities for development of novel therapeutics.


Subject(s)
Amino Acyl-tRNA Synthetases/chemistry , Heart/drug effects , Heart/physiopathology , Myocardial Infarction/drug therapy , Myocardial Infarction/physiopathology , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Animals , Apoptosis/drug effects , Biological Products/pharmacology , Biological Products/therapeutic use , Capillaries/drug effects , Capillaries/physiopathology , Cell Proliferation/drug effects , Fibroblasts/drug effects , Fibroblasts/pathology , Fibrosis , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Neovascularization, Physiologic/drug effects , Peptide Fragments/therapeutic use , Proto-Oncogene Proteins c-kit/metabolism
9.
Nat Commun ; 5: 3984, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24917553

ABSTRACT

Endothelial cells (ECs) lining blood vessels express many mechanosensors, including platelet endothelial cell adhesion molecule-1 (PECAM-1), that convert mechanical force into biochemical signals. While it is accepted that mechanical stresses and the mechanical properties of ECs regulate vessel health, the relationship between force and biological response remains elusive. Here we show that ECs integrate mechanical forces and extracellular matrix (ECM) cues to modulate their own mechanical properties. We demonstrate that the ECM influences EC response to tension on PECAM-1. ECs adherent on collagen display divergent stiffening and focal adhesion growth compared with ECs on fibronectin. This is because of protein kinase A (PKA)-dependent serine phosphorylation and inactivation of RhoA. PKA signalling regulates focal adhesion dynamics and EC compliance in response to shear stress in vitro and in vivo. Our study identifies an ECM-specific, mechanosensitive signalling pathway that regulates EC compliance and may serve as an atheroprotective mechanism that maintains blood vessel integrity in vivo.


Subject(s)
Aorta/physiology , Endothelium, Vascular/physiology , Extracellular Matrix/physiology , Hemodynamics , Animals , Aorta/cytology , Aorta/enzymology , Aorta/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Endothelium, Vascular/metabolism , Extracellular Matrix/enzymology , Extracellular Matrix/metabolism , Focal Adhesions , Male , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , rhoA GTP-Binding Protein/metabolism
10.
Nat Commun ; 4: 1417, 2013.
Article in English | MEDLINE | ID: mdl-23361008

ABSTRACT

Leucyl-tRNA synthetases (LeuRSs) have an essential role in translation and are promising targets for antibiotic development. Agrocin 84 is a LeuRS inhibitor produced by the biocontrol agent Agrobacterium radiobacter K84 that targets pathogenic strains of A. tumefaciens, the causative agent of plant tumours. Agrocin 84 acts as a molecular Trojan horse and is processed inside the pathogen into a toxic moiety (TM84). Here we show using crystal structure, thermodynamic and kinetic analyses, that this natural antibiotic employs a unique and previously undescribed mechanism to inhibit LeuRS. TM84 requires tRNA(Leu) for tight binding to the LeuRS synthetic active site, unlike any previously reported inhibitors. TM84 traps the enzyme-tRNA complex in a novel 'aminoacylation-like' conformation, forming novel interactions with the KMSKS loop and the tRNA 3'-end. Our findings reveal an intriguing tRNA-dependent inhibition mechanism that may confer a distinct evolutionary advantage in vivo and inform future rational antibiotic design.


Subject(s)
Adenine Nucleotides/pharmacology , Agrobacterium tumefaciens/enzymology , Biological Control Agents , Leucine-tRNA Ligase/antagonists & inhibitors , Plant Tumors/microbiology , RNA, Plant/metabolism , RNA, Transfer/metabolism , Adenine Nucleotides/chemistry , Agrobacterium tumefaciens/drug effects , Aminoacylation/drug effects , Calorimetry , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Leucine-tRNA Ligase/chemistry , Leucine-tRNA Ligase/metabolism , Models, Molecular , Protein Binding/drug effects , Protein Conformation , Protein Structure, Tertiary , RNA, Plant/chemistry , RNA, Transfer/chemistry
11.
J Biol Chem ; 285(49): 37944-52, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-20876530

ABSTRACT

The heptapeptide-nucleotide microcin C (McC) is a potent inhibitor of enteric bacteria growth. Inside a sensitive cell, McC is processed by aminopeptidases, which release a nonhydrolyzable aspartyl-adenylate, a strong inhibitor of aspartyl-tRNA synthetase. The mccABCDE operon is sufficient for McC production and resistance of the producing cell to McC. An additional gene, mccF, which is adjacent to but not part of the mccABCDE operon, also provides resistance to exogenous McC. MccF is similar to Escherichia coli LdcA, an L,D-carboxypeptidase whose substrate is monomeric murotetrapeptide L-Ala-D-Glu-meso-A(2)pm-D-Ala or its UDP-activated murein precursor. The mechanism by which MccF provides McC resistance remained unknown. Here, we show that MccF detoxifies both intact and processed McC by cleaving an amide bond between the C-terminal aspartate and the nucleotide moiety. MccF also cleaves the same bond in nonhydrolyzable aminoacyl sulfamoyl adenosines containing aspartyl, glutamyl, and, to a lesser extent, seryl aminoacyl moieties but is ineffective against other aminoacyl adenylates.


Subject(s)
Bacteriocins/pharmacology , Drug Resistance, Bacterial/physiology , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Peptide Hydrolases/metabolism , Aspartate-tRNA Ligase/antagonists & inhibitors , Bacteriocins/metabolism , Drug Resistance, Bacterial/drug effects , Enzyme Inhibitors/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Operon/physiology , Peptide Hydrolases/genetics , Peptidoglycan/genetics , Peptidoglycan/metabolism
12.
Vascul Pharmacol ; 52(1-2): 21-6, 2010.
Article in English | MEDLINE | ID: mdl-19962454

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) are enzymes that join amino acids to tRNAs. Although they are housekeeping enzymes essential for protein synthesis, aaRSs are now known to participate in a wide variety of functions, including transcription, translation, splicing, inflammation, angiogenesis and apoptosis. In eukaryotes, the functional expansion of aaRSs is closely linked to evolutionary advantages conferred by recruitment into protein complexes as well as various structural adaptations. The elucidation and understanding of the diverse functions of aaRSs is a major goal of current and future research. These investigations will undoubtedly provide some of the most fundamental understanding of how and possibly why synthetases became so tightly involved in such a vast array of cell signaling pathways.


Subject(s)
Amino Acyl-tRNA Synthetases/physiology , Protein Biosynthesis/physiology , RNA, Transfer, Amino Acid-Specific/physiology , Signal Transduction/physiology , Animals , Humans
13.
Proc Natl Acad Sci U S A ; 103(23): 8846-51, 2006 Jun 06.
Article in English | MEDLINE | ID: mdl-16731618

ABSTRACT

Agrobacterium radiobacter K84, used worldwide to biocontrol crown gall disease caused by Agrobacterium tumefaciens, produces an antiagrobacterial compound called agrocin 84. We report the nucleotide sequence of pAgK84, a 44.42-kb plasmid coding for production of this disubstituted adenine nucleotide antibiotic. pAgK84 encodes 36 ORFs, 17 of which (agn) code for synthesis of or immunity to agrocin 84. Two genes, agnB2 and agnA, encode aminoacyl tRNA synthetase homologues. We have shown that the toxic moiety of agrocin 84 inhibits cellular leucyl-tRNA synthetases and AgnB2, which confers immunity to the antibiotic, is a resistant form of this enzyme. AgnA, a truncated homologue of asparaginyl tRNA synthetase could catalyze the phosphoramidate bond between a precursor of the methyl pentanamide side group and the nucleotide. We propose previously undescribed chemistry, catalyzed by AgnB1, to generate the precursor necessary for this phosphoramidate linkage. AgnC7 is related to ribonucleotide reductases and could generate the 3'-deoxyarabinose moiety of the nucleoside. Bioinformatics suggest that agnC3, agnC4, and agnC6 contribute to maturation of the methyl pentanamide, whereas agnC2 may produce the glucofuranose side group bound to the adenine ring. AgnG is related to bacterial exporters. An agnG mutant accumulated agrocin 84 intracellularly but did not export the antibiotic. pAgK84 is transmissible and encodes genes for conjugative DNA processing but lacks a type IV secretion system, suggesting that pAgK84 transfers by mobilization. By sequence analysis, the deletion engineered into pAgK1026 removed the oriT and essential tra genes, confirming the enhanced environmental safety of this modified form of pAgK84.


Subject(s)
Adenine Nucleotides/biosynthesis , Adenine Nucleotides/pharmacology , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/pharmacology , Plant Tumors/microbiology , Adenine Nucleotides/chemistry , Adenine Nucleotides/metabolism , Anti-Bacterial Agents/metabolism , Base Sequence , Conjugation, Genetic , DNA Replication/genetics , Molecular Sequence Data , Mutation/genetics , Pest Control, Biological , Physical Chromosome Mapping , Rhizobium/cytology
14.
Science ; 309(5740): 1533, 2005 Sep 02.
Article in English | MEDLINE | ID: mdl-16141066

ABSTRACT

Crops can be devastated by pathogenic strains of Agrobacterium tumefaciens that cause crown gall tumors. This devastation can be prevented by the nonpathogenic biocontrol agent A. radiobacter K84, which prevents disease by production of the "Trojan horse" toxin agrocin 84, which is specifically imported into tumorgenic A. tumefaciens strains to cause cell death. We demonstrate that this biocontrol agent targets A. tumefaciens leucyl-tRNA synthetase (LeuRS), an essential enzyme for cell viability, while the agent itself survives by having a second, self-protective copy of the synthetase. In principle, this strategy from nature could be applied to other crop diseases by direct intervention.


Subject(s)
Adenine Nucleotides/toxicity , Agrobacterium tumefaciens/enzymology , Bacteriocins/toxicity , Leucine-tRNA Ligase/antagonists & inhibitors , Plant Tumors , Adenine Nucleotides/chemistry , Adenine Nucleotides/metabolism , Agrobacterium tumefaciens/metabolism , Bacteriocins/chemistry , Bacteriocins/metabolism
15.
Proc Natl Acad Sci U S A ; 102(12): 4264-9, 2005 Mar 22.
Article in English | MEDLINE | ID: mdl-15767583

ABSTRACT

The enzyme YkvM from Bacillus subtilis was identified previously along with three other enzymes (YkvJKL) in a bioinformatics search for enzymes involved in the biosynthesis of queuosine, a 7-deazaguanine modified nucleoside found in tRNA(GUN) of Bacteria and Eukarya. Genetic analysis of ykvJKLM mutants in Acinetobacter confirmed that each was essential for queuosine biosynthesis, and the genes were renamed queCDEF. QueF exhibits significant homology to the type I GTP cyclohydrolases characterized by FolE. Given that GTP is the precursor to queuosine and that a cyclohydrolase-like reaction was postulated as the initial step in queuosine biosynthesis, QueF was proposed to be the putative cyclohydrolase-like enzyme responsible for this reaction. We have cloned the queF genes from B. subtilis and Escherichia coli and characterized the recombinant enzymes. Contrary to the predictions based on sequence analysis, we discovered that the enzymes, in fact, catalyze a mechanistically unrelated reaction, the NADPH-dependent reduction of 7-cyano-7-deazaguanineto7-aminomethyl-7-deazaguanine, a late step in the biosynthesis of queuosine. We report here in vitro and in vivo studies that demonstrate this catalytic activity, as well as preliminary biochemical and bioinformatics analysis that provide insight into the structure of this family of enzymes.


Subject(s)
Bacillus subtilis/enzymology , Escherichia coli/enzymology , GTP Cyclohydrolase/chemistry , GTP Cyclohydrolase/metabolism , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Acinetobacter/enzymology , Acinetobacter/genetics , Amino Acid Sequence , Bacillus subtilis/genetics , Base Sequence , DNA, Bacterial/genetics , Escherichia coli/genetics , GTP Cyclohydrolase/genetics , Genes, Bacterial , Molecular Sequence Data , Nucleoside Q/biosynthesis , Oxidoreductases/genetics , Protein Folding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid
16.
J Biol Chem ; 280(4): 2405-8, 2005 Jan 28.
Article in English | MEDLINE | ID: mdl-15579907

ABSTRACT

A natural fragment of an enzyme that catalyzes the first step of protein synthesis-human tryptophanyl-tRNA synthetase (T2-TrpRS) has potent anti-angiogenic activity. A cellular receptor through which T2-TrpRS exerts its anti-angiogenic activity has not previously been identified. Here T2-TrpRS was shown to bind at intercellular junctions of endothelial cells (ECs). Using genetic knock-outs, binding was established to depend on VE-cadherin, a calcium-dependent adhesion molecule, which is selectively expressed in ECs, concentrated at adherens junctions, and is essential for normal vascular development. In contrast, T2-TrpRS binding to EC junctions was not dependent on platelet endothelial cell adhesion molecule type-1, another adhesion molecule found at EC junctions. Pull-down assays confirmed direct complex formation between T2-TrpRS and VE-cadherin. Binding of T2-TrpRS inhibited VEGF-induced ERK activation and EC migration. Thus, a VE-cadherin-dependent pathway is proposed to link T2-TrpRS to inhibition of new blood vessel formation.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Angiogenesis Inhibitors/pharmacology , Cadherins/physiology , Cytokines/metabolism , Tryptophan-tRNA Ligase/metabolism , Animals , Antigens, CD , Aorta/cytology , Blotting, Western , Cattle , Cell Movement , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gap Junctions , Green Fluorescent Proteins/metabolism , Immunoprecipitation , Microscopy, Confocal , Microscopy, Fluorescence , Neovascularization, Pathologic , Protein Binding , Recombinant Proteins/chemistry , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
17.
J Biol Chem ; 279(8): 6280-5, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14660578

ABSTRACT

Queuosine (Q) is a hypermodified 7-deazaguanosine nucleoside located in the anticodon wobble position of four amino acid-specific tRNAs. In bacteria, Q is produced de novo from GTP via the 7-deazaguanosine precursor preQ1 (7-aminoethyl 7-deazaguanine) by an uncharacterized pathway. PreQ1 is subsequently transferred to its specific tRNA by a tRNA-guanine transglycosylase (TGT) and then further modified in situ to produce Q. Here we use comparative genomics to implicate four gene families (best exemplified by the B. subtilis operon ykvJKLM) as candidates in the preQ1 biosynthetic pathway. Deletions were constructed in genes for each of the four orthologs in Acinetobacter. High pressure liquid chromatography analysis showed the Q nucleoside was absent from the tRNAs of each of four deletion strains. Electrospray ionization mass spectrometry confirmed the absence of Q in each mutant strain. Finally, introduction of the Bacillus subtilis ykvJKLM operon in trans complemented the Q deficiency of the two deletion mutants that were tested. Thus, the products of these four genes (named queC, -D, -E, and -F) are essential for the Q biosynthetic pathway.


Subject(s)
Genes, Bacterial , Nucleoside Q/biosynthesis , Nucleoside Q/chemistry , Acinetobacter/genetics , Alleles , Bacillus subtilis/genetics , Catalysis , Chromatography, High Pressure Liquid , DNA/chemistry , Gene Deletion , Models, Chemical , Multigene Family , Mutation , Operon , RNA, Transfer/metabolism , Spectrometry, Mass, Electrospray Ionization , Time Factors
18.
Nature ; 420(6917): 841-4, 2002.
Article in English | MEDLINE | ID: mdl-12490955

ABSTRACT

RNA molecules are thought to have been prominent in the early history of life on Earth because of their ability both to encode genetic information and to exhibit catalytic function. The modern genetic alphabet relies on two sets of complementary base pairs to store genetic information. However, owing to the chemical instability of cytosine, which readily deaminates to uracil, a primitive genetic system composed of the bases A, U, G and C may have been difficult to establish. It has been suggested that the first genetic material instead contained only a single base-pairing unit. Here we show that binary informational macromolecules, containing only two different nucleotide subunits, can act as catalysts. In vitro evolution was used to obtain ligase ribozymes composed of only 2,6-diaminopurine and uracil nucleotides, which catalyse the template-directed joining of two RNA molecules, one bearing a 5'-triphosphate and the other a 3'-hydroxyl. The active conformation of the fastest isolated ribozyme had a catalytic rate that was about 36,000-fold faster than the uncatalysed rate of reaction. This ribozyme is specific for the formation of biologically relevant 3',5'-phosphodiester linkages.


Subject(s)
2-Aminopurine/analogs & derivatives , 2-Aminopurine/metabolism , Nucleotides/metabolism , RNA, Catalytic/genetics , RNA, Catalytic/metabolism , Uracil/metabolism , Base Sequence , Binding Sites , Catalysis , Directed Molecular Evolution , Evolution, Molecular , Kinetics , Ligases/chemistry , Ligases/genetics , Ligases/metabolism , Molecular Sequence Data , Nucleic Acid Conformation , Nucleotides/genetics , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/metabolism , RNA/genetics , RNA/metabolism , RNA, Catalytic/chemistry , Substrate Specificity , Templates, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...