Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 225
Filter
1.
Nat Chem Biol ; 20(1): 83-92, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37857992

ABSTRACT

The inner mitochondrial membrane (IMM) generates power to drive cell function, and its dynamics control mitochondrial health and cellular homeostasis. Here, we describe the cell-permeant, lipid-like small molecule MAO-N3 and use it to assemble high-density environmentally sensitive (HIDE) probes that selectively label and image the IMM in live cells and multiple cell states. MAO-N3 pairs with strain-promoted azide-alkyne click chemistry-reactive fluorophores to support HIDE imaging using confocal, structured illumination, single-molecule localization and stimulated emission depletion microscopy, all with significantly improved resistance to photobleaching. These probes generate images with excellent spatial and temporal resolution, require no genetic manipulations, are non-toxic in model cell lines and primary cardiomyocytes (even under conditions that amplify the effects of mitochondrial toxins) and can visualize mitochondrial dynamics for 12.5 h. This probe will enable comprehensive studies of IMM dynamics with high temporal and spatial resolution.


Subject(s)
Fluorescent Dyes , Mitochondrial Membranes , Humans , HeLa Cells , Microscopy, Fluorescence/methods , Lipids , Monoamine Oxidase
2.
Nucleic Acids Res ; 52(3): 1027-1042, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38214230

ABSTRACT

The ribosome is a ribonucleoprotein complex found in all domains of life. Its role is to catalyze protein synthesis, the messenger RNA (mRNA)-templated formation of amide bonds between α-amino acid monomers. Amide bond formation occurs within a highly conserved region of the large ribosomal subunit known as the peptidyl transferase center (PTC). Here we describe the step-wise design and characterization of mini-PTC 1.1, a 284-nucleotide RNA that recapitulates many essential features of the Escherichia coli PTC. Mini-PTC 1.1 folds into a PTC-like structure under physiological conditions, even in the absence of r-proteins, and engages small molecule analogs of A- and P-site tRNAs. The sequence of mini-PTC 1.1 differs from the wild type E. coli ribosome at 12 nucleotides that were installed by a cohort of citizen scientists using the on-line video game Eterna. These base changes improve both the secondary structure and tertiary folding of mini-PTC 1.1 as well as its ability to bind small molecule substrate analogs. Here, the combined input from Eterna citizen-scientists and RNA structural analysis provides a robust workflow for the design of a minimal PTC that recapitulates many features of an intact ribosome.


Subject(s)
Escherichia coli , Ribosomes , Humans , Amides , Escherichia coli/genetics , Escherichia coli/metabolism , Peptidyl Transferases/genetics , Peptidyl Transferases/chemistry , Ribosomes/metabolism , RNA, Transfer/metabolism
3.
Proc Natl Acad Sci U S A ; 117(33): 19879-19887, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32747536

ABSTRACT

The ribosome translates the genetic code into proteins in all domains of life. Its size and complexity demand long-range interactions that regulate ribosome function. These interactions are largely unknown. Here, we apply a global coevolution method, statistical coupling analysis (SCA), to identify coevolving residue networks (sectors) within the 23S ribosomal RNA (rRNA) of the large ribosomal subunit. As in proteins, SCA reveals a hierarchical organization of evolutionary constraints with near-independent groups of nucleotides forming physically contiguous networks within the three-dimensional structure. Using a quantitative, continuous-culture-with-deep-sequencing assay, we confirm that the top two SCA-predicted sectors contribute to ribosome function. These sectors map to distinct ribosome activities, and their origins trace to phylogenetic divergences across all domains of life. These findings provide a foundation to map ribosome allostery, explore ribosome biogenesis, and engineer ribosomes for new functions. Despite differences in chemical structure, protein and RNA enzymes appear to share a common internal logic of interaction and assembly.


Subject(s)
Escherichia coli/genetics , RNA, Bacterial/chemistry , RNA, Ribosomal, 23S/chemistry , Ribosomes/genetics , Escherichia coli/chemistry , Escherichia coli/metabolism , Evolution, Molecular , Nucleic Acid Conformation , Phylogeny , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Ribosomal, 23S/genetics , RNA, Ribosomal, 23S/metabolism , Ribosomes/chemistry , Ribosomes/metabolism
4.
J Am Chem Soc ; 144(27): 12138-12146, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35776693

ABSTRACT

Electrical potential differences across lipid bilayers play foundational roles in cellular physiology. Plasma membrane voltage is the most widely studied; however, the bilayers of organelles like mitochondria, lysosomes, nuclei, and the endoplasmic reticulum (ER) also provide opportunities for ionic compartmentalization and the generation of transmembrane potentials. Unlike plasma membranes, organellar bilayers, cloistered within the cell, remain recalcitrant to traditional approaches like patch-clamp electrophysiology. To address the challenge of monitoring changes in organelle membrane potential, we describe the design, synthesis, and application of the LUnAR RhoVR (Ligation Unquenched for Activation and Redistribution Rhodamine-based Voltage Reporter) for optically monitoring membrane potential changes in the ER of living cells. We pair a tetrazine-quenched RhoVR for voltage sensing with a transcyclooctene (TCO)-conjugated ceramide (Cer-TCO) for targeting to the ER. Bright fluorescence is observed only at the coincidence of the LUnAR RhoVR and TCO in the ER, minimizing non-specific, off-target fluorescence. We show that the product of the LUnAR RhoVR and Cer-TCO is voltage-sensitive and that the LUnAR RhoVR can be targeted to an intact ER in living cells. Using the LUnAR RhoVR, we use two-color, ER-localized, fast voltage imaging coupled with cytosolic Ca2+ imaging to validate the electroneutrality of Ca2+ release from internal stores. Finally, we use the LUnAR RhoVR to directly visualize functional coupling between the plasma-ER membranes in patch clamped cell lines, providing the first direct evidence of the sign of the ER potential response to plasma membrane potential changes. We envision that the LUnAR RhoVR, along with other existing organelle-targeting TCO probes, could be applied widely for exploring organelle physiology.


Subject(s)
Fluorescent Dyes , Organelles , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Fluorescent Dyes/metabolism , Ionophores/metabolism , Lysosomes/metabolism , Membrane Potentials , Organelles/metabolism , Rhodamines/metabolism
5.
Nat Chem Biol ; 16(4): 408-414, 2020 04.
Article in English | MEDLINE | ID: mdl-32094922

ABSTRACT

We report new lipid-based, high-density, environmentally sensitive (HIDE) probes that accurately and selectively image endo-lysosomes and their dynamics at super-resolution for extended times. Treatment of live cells with the small molecules DiIC16TCO or DiIC16'TCO followed by in situ tetrazine ligation reaction with the silicon-rhodamine dye SiR-Tz generates the HIDE probes DiIC16-SiR and DiIC16'-SiR in the endo-lysosomal membrane. These new probes support the acquisition of super-resolution videos of organelle dynamics in primary cells for more than 7 min with no detectable change in endosome structure or function. Using DiIC16-SiR and DiIC16'-SiR, we describe direct evidence of endosome motility defects in cells from patients with Niemann-Pick Type-C disease. In wild-type fibroblasts, the probes reveal distinct but rare inter-endosome kiss-and-run events that cannot be observed using confocal methods. Our results shed new light on the role of NPC1 in organelle motility and cholesterol trafficking.


Subject(s)
Endosomes/metabolism , Lysosomes/metabolism , Microscopy, Fluorescence/methods , Biological Transport , Carbocyanines/chemistry , Carrier Proteins/metabolism , Cholesterol/metabolism , Fibroblasts/metabolism , Fluorescent Dyes , HeLa Cells , Humans , Membrane Glycoproteins/metabolism , Protein Transport
6.
Proc Natl Acad Sci U S A ; 116(2): 512-521, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30610181

ABSTRACT

Protein therapeutics represent a significant and growing component of the modern pharmacopeia, but their potential to treat human disease is limited because most proteins fail to traffic across biological membranes. Recently, we discovered a class of cell-permeant miniature proteins (CPMPs) containing a precisely defined, penta-arginine (penta-Arg) motif that traffics readily to the cytosol and nucleus of mammalian cells with efficiencies that rival those of hydrocarbon-stapled peptides active in animals and man. Like many cell-penetrating peptides (CPPs), CPMPs enter the endocytic pathway; the difference is that CPMPs containing a penta-Arg motif are released efficiently from endosomes, while other CPPs are not. Here, we seek to understand how CPMPs traffic from endosomes into the cytosol and what factors contribute to the efficiency of endosomal release. First, using two complementary cell-based assays, we exclude endosomal rupture as the primary means of endosomal escape. Next, using an RNA interference screen, fluorescence correlation spectroscopy, and confocal imaging, we identify VPS39-a gene encoding a subunit of the homotypic fusion and protein-sorting (HOPS) complex-as a critical determinant in the trafficking of CPMPs and hydrocarbon-stapled peptides to the cytosol. Although CPMPs neither inhibit nor activate HOPS function, HOPS activity is essential to efficiently deliver CPMPs to the cytosol. CPMPs localize within the lumen of Rab7+ and Lamp1+ endosomes and their transport requires HOPS activity. Overall, our results identify Lamp1+ late endosomes and lysosomes as portals for passing proteins into the cytosol and suggest that this environment is prerequisite for endosomal escape.


Subject(s)
Carrier Proteins/genetics , Cell-Penetrating Peptides , Endosomes/metabolism , Membrane Fusion/drug effects , Amino Acid Motifs , Autophagy-Related Proteins , Carrier Proteins/metabolism , Cell Line, Tumor , Cell-Penetrating Peptides/pharmacokinetics , Cell-Penetrating Peptides/pharmacology , Cytosol/metabolism , Endosomes/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Protein Transport/drug effects , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
7.
Biochemistry ; 60(7): 500-512, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33557518

ABSTRACT

We previously reported a family of hydrocarbon-stapled peptides designed to interact with the epidermal growth factor receptor (EGFR) juxtamembrane (JM) segment, blocking its ability to form a coiled coil dimer that is essential for receptor activation. These hydrocarbon-stapled peptides, most notably E1S, decreased the proliferation of cell lines that express wild-type EGFR (H2030 and A431) as well as those expressing the oncogenic mutants EGFR L858R (H3255) and L858R/T790M (H1975). Although our previous investigations provided evidence that E1S interacted with EGFR directly, the location and details of these interactions were not established. Here we apply biochemical and cross-linking mass spectrometry tools to better define the interactions between E1S and EGFR. Taken with previously reported structure-activity relationships, our results support a model in which E1S interacts simultaneously with both the JM and the C-lobe of the activator kinase, effectively displacing the JM of the receiver kinase. Our results also reveal potential interactions between E1S and the N-terminal region of the C-terminal tail. We propose a model in which E1S inhibits EGFR by both mimicking and inhibiting JM coiled coil formation. This model could be used to design novel, allosteric (and perhaps nonpeptidic) EGFR inhibitors.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Amino Acid Sequence/genetics , Cell Line, Tumor , Cell Membrane/metabolism , ErbB Receptors/genetics , Humans , Mutation , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
8.
Biochemistry ; 59(41): 3965-3972, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32941004

ABSTRACT

Mutations in the epidermal growth factor receptor (EGFR) extracellular domain (ECD) are implicated in the development of glioblastoma multiforme (GBM), which is a highly aggressive form of brain cancer. Of particular interest to GBM is the EGFR variant known as EGFRvIII, which is distinguished by an in-frame deletion of exons 2-7, which encode ECD residues 6-273. Included within the deleted region is an autoinhibitory tether, whose absence, alongside unique disulfide interactions within the truncated ECD, supports assembly of a constitutively active asymmetric kinase dimer. Previous studies have shown that the binding of growth factors to the ECD of wild-type EGFR leads to the formation of two distinct coiled coil dimers in the cytoplasmic juxtamembrane (JM) segment, whose identities correlate with the downstream phenotype. One coiled coil contains leucine residues at the interhelix interface (EGF-type), whereas the other contains charged and polar side chains (TGF-α-type). It has been proposed that growth-factor-dependent structural changes in the ECD and adjacent transmembrane helix are transduced into distinct JM coiled coils. Here, we show that, in the absence of this growth-factor-induced signal, the JM of EGFRvIII adopts both EGF-type and TGF-α-type structures, providing direct evidence for this hypothesis. These studies confirm that the signals that define JM coiled coil identity begin within the ECD, and support a model in which growth-factor-induced conformational changes are transmitted from the ECD through the transmembrane helix to favor different coiled coil isomers within the JM.


Subject(s)
ErbB Receptors/metabolism , ErbB Receptors/chemistry , ErbB Receptors/genetics , Exons/genetics , Humans , Mutation/genetics , Protein Domains , Signal Transduction/genetics , Signal Transduction/physiology
9.
Angew Chem Int Ed Engl ; 59(8): 3122-3126, 2020 02 17.
Article in English | MEDLINE | ID: mdl-31828898

ABSTRACT

By transplanting identity elements into E. coli tRNAfMet , we have engineered an orthogonal initiator tRNA (itRNATy2 ) that is a substrate for Methanocaldococcus jannaschii TyrRS. We demonstrate that itRNATy2 can initiate translation in vivo with aromatic non-canonical amino acids (ncAAs) bearing diverse sidechains. Although the initial system suffered from low yields, deleting redundant copies of tRNAfMet from the genome afforded an E. coli strain in which the efficiency of non-canonical initiation equals elongation. With this improved system we produced a protein containing two distinct ncAAs at the first and second positions, an initial step towards producing completely unnatural polypeptides in vivo. This work provides a valuable tool to synthetic biology and demonstrates remarkable versatility of the E. coli translational machinery for initiation with ncAAs in vivo.


Subject(s)
Amino Acids/metabolism , Protein Biosynthesis/physiology , Protein Engineering/methods , Humans
10.
Biochemistry ; 58(45): 4494-4504, 2019 11 12.
Article in English | MEDLINE | ID: mdl-31607123

ABSTRACT

Ribosome engineering has emerged as a promising field in synthetic biology, particularly concerning the production of new sequence-defined polymers. Mutant ribosomes have been developed that improve the incorporation of several nonstandard monomers including d-amino acids, dipeptides, and ß-amino acids into polypeptide chains. However, there remains little mechanistic understanding of how these ribosomes catalyze incorporation of these new substrates. Here, we probed the properties of a mutant ribosome-P7A7-evolved for better in vivo ß-amino acid incorporation through in vitro biochemistry and cryo-electron microscopy. Although P7A7 is a functional ribosome in vivo, it is inactive in vitro, and assembles poorly into 70S ribosome complexes. Structural characterization revealed large regions of disorder in the peptidyltransferase center and nearby features, suggesting a defect in assembly. Comparison of RNA helix and ribosomal protein occupancy with other assembly intermediates revealed that P7A7 is stalled at a late stage in ribosome assembly, explaining its weak activity. These results highlight the importance of ensuring efficient ribosome assembly during ribosome engineering toward new catalytic abilities.


Subject(s)
Amino Acids/genetics , Escherichia coli/genetics , Ribosomes/genetics , Amino Acids/chemistry , Escherichia coli/chemistry , Models, Molecular , Mutation , Peptides/chemistry , Peptides/genetics , Protein Biosynthesis , Ribosomes/chemistry
11.
Bioorg Med Chem ; 26(6): 1197-1202, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29150077

ABSTRACT

We have previously reported that miniature proteins containing a distinct array of 5 arginine residues on a folded α-helix - a penta-arg motif - traffic with high efficiency from endosomes into the cytosol and nucleus of mammalian cells. Here we evaluate whether a penta-arg motif can improve the intracellular trafficking of an otherwise impermeant hydrocarbon-stapled peptide, SAH-p53-4Rho. We prepared a panel of SAH-p53-4Rho variants containing penta-arg sequences with different spacings and axial arrangement and evaluated their overall uptake (as judged by flow cytometry) and their intracellular access (as determined by fluorescence correlation spectroscopy, FCS). One member of this panel reached the cytosol extremely well, matching the level achieved by SAH-p53-8Rho, a previously reported and highly permeant hydrocarbon-stapled peptide. Notably, we found no relationship between cellular uptake as judged by flow cytometry and cytosolic access as determined by FCS. This result reiterates that overall uptake and endosomal release represent fundamentally different biological processes. To determine cytosolic and/or nuclear access, one must measure concentration directly using a quantitative and non-amplified tool such as FCS. As has been observed for highly cell permeant miniature proteins such as ZF5.3, optimal penetration of hydrocarbon-stapled peptides into the cell cytosol results when the penta-arg motif is located within more (as opposed to less) structured regions.


Subject(s)
Arginine/chemistry , Cytosol/metabolism , Peptides/metabolism , Amino Acid Sequence , Cell Survival/drug effects , Flow Cytometry , HeLa Cells , Humans , Hydrocarbons/chemistry , Peptides/chemistry , Peptides/pharmacology , Spectrometry, Fluorescence
12.
Angew Chem Int Ed Engl ; 57(1): 37, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29235715

ABSTRACT

Ronald Breslow, Samuel Latham Mitchill Professor of Chemistry at Columbia University, passed away on October 25, 2017, at the age of 86. Breslow made remarkable contributions to the fields of physical-organic and bioorganic chemistry, including biological and biomimetic transformations, and the use of molecular recognition to control reaction selectivity.

13.
Biochemistry ; 56(39): 5194-5201, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28792749

ABSTRACT

Living cells are complex and dynamic assemblies that carefully sequester and orchestrate multiple diverse processes that enable growth, division, regulation, movement, and communication. Membrane-bound organelles such as the endoplasmic reticulum, mitochondria, plasma membrane, and others are integral to these processes, and their functions demand dynamic reorganization in both space and time. Visualizing these dynamics in live cells over long time periods demands probes that label discrete organelles specifically, at high density, and withstand long-term irradiation. Here we describe the evolution of our work on the development of a set of high-density environmentally sensitive (HIDE) membrane probes that enable long-term, live-cell nanoscopy of the dynamics of multiple organelles in live cells using single-molecule switching and stimulated emission depletion imaging modalities.


Subject(s)
Molecular Imaging/methods , Organelles/metabolism , HeLa Cells , Humans , Intracellular Membranes/metabolism , Signal-To-Noise Ratio , Time Factors
16.
Angew Chem Int Ed Engl ; 56(35): 10408-10412, 2017 08 21.
Article in English | MEDLINE | ID: mdl-28679029

ABSTRACT

Super-resolution imaging of live cells over extended time periods with high temporal resolution requires high-density labeling and extraordinary fluorophore photostability. Herein, we achieve this goal by combining the attributes of the high-density plasma membrane probe DiI-TCO and the photostable STED dye SiR-Tz. These components undergo rapid tetrazine ligation within the plasma membrane to generate the HIDE probe DiI-SiR. Using DiI-SiR, we visualized filopodia dynamics in HeLa cells over 25 min at 0.5 s temporal resolution, and visualized dynamic contact-mediated repulsion events in primary mouse hippocampal neurons over 9 min at 2 s temporal resolution. HIDE probes such as DiI-SiR are non-toxic and do not require transfection, and their apparent photostability significantly improves the ability to monitor dynamic processes in live cells at super-resolution over biologically relevant timescales.


Subject(s)
Cell Membrane/chemistry , Fluorescent Dyes/chemistry , Nanotechnology , Optical Imaging , HeLa Cells , Humans , Microscopy, Fluorescence , Molecular Structure , Tumor Cells, Cultured
18.
J Am Chem Soc ; 138(22): 7143-50, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27163487

ABSTRACT

Fluorogenic dyes such as FlAsH and ReAsH are used widely to localize, monitor, and characterize proteins and their assemblies in live cells. These bis-arsenical dyes can become fluorescent when bound to a protein containing four proximal Cys thiols-a tetracysteine (Cys4) motif. Yet the mechanism by which bis-arsenicals become fluorescent upon binding a Cys4 motif is unknown, and this nescience limits more widespread application of this tool. Here we probe the origins of ReAsH fluorogenicity using both computation and experiment. Our results support a model in which ReAsH fluorescence depends on the relative orientation of the aryl chromophore and the appended arsenic chelate: the fluorescence is rotamer-restricted. Our results do not support a model in which fluorogenicity arises from the relief of ring strain. The calculations identify those As-aryl rotamers that support fluorescence and those that do not and correlate well with prior experiments. The rotamer-restricted model we propose is supported further by biophysical studies: the excited-state fluorescence lifetime of a complex between ReAsH and a protein bearing a high-affinity Cys4 motif is longer than that of ReAsH-EDT2, and the fluorescence intensity of ReAsH-EDT2 increases in solvents of increasing viscosity. By providing a higher resolution view of the structural basis for fluorogenicity, these results provide a clear strategy for the design of more selective bis-arsenicals and better-optimized protein targets, with a concomitant improvement in the ability to characterize previously invisible protein conformational changes and assemblies in live cells.


Subject(s)
Arsenicals/chemistry , Cysteine/chemistry , Fluorescent Dyes/chemistry , Oxazines/chemistry , Proteins/chemistry , Binding Sites , Fluorescence , Models, Theoretical , Protein Binding , Solutions , Sulfhydryl Compounds/chemistry , Viscosity
19.
J Am Chem Soc ; 138(9): 3175-82, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26859619

ABSTRACT

We report a synthetic glycosylation reaction between sucrosyl acceptors and glycosyl fluoride donors to yield the derived trisaccharides. This reaction proceeds at room temperature in an aqueous solvent mixture. Calcium salts and a tertiary amine base promote the reaction with high site-selectivity for either the 3'-position or 1'-position of the fructofuranoside unit. Because nonenzymatic aqueous oligosaccharide syntheses are underdeveloped, mechanistic studies were carried out in order to identify the origin of the selectivity, which we hypothesized was related to the structure of the hydroxyl group array in sucrose. The solution conformation of various monodeoxysucrose analogs revealed the co-operative nature of the hydroxyl groups in mediating both this aqueous glycosyl bond-forming reaction and the site-selectivity at the same time.


Subject(s)
Calcium/chemistry , Methylamines/chemistry , Sucrose/chemistry , Trisaccharides/chemical synthesis , Glycosylation , Trisaccharides/chemistry
20.
J Am Chem Soc ; 138(16): 5194-7, 2016 04 27.
Article in English | MEDLINE | ID: mdl-27086674

ABSTRACT

It has recently been reported that ribosomes from erythromycin-resistant Escherichia coli strains, when isolated in S30 extracts and incubated with chemically mis-acylated tRNA, can incorporate certain ß-amino acids into full length DHFR in vitro. Here we report that wild-type E. coli EF-Tu and phenylalanyl-tRNA synthetase collaborate with these mutant ribosomes and others to incorporate ß(3)-Phe analogs into full length DHFR in vivo. E. coli harboring the most active mutant ribosomes are robust, with a doubling time only 14% longer than wild-type. These results reveal the unexpected tolerance of E. coli and its translation machinery to the ß(3)-amino acid backbone and should embolden in vivo selections for orthogonal translational machinery components that incorporate diverse ß-amino acids into proteins and peptides. E. coli harboring mutant ribosomes may possess the capacity to incorporate many non-natural, non-α-amino acids into proteins and other sequence-programmed polymeric materials.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Escherichia coli Proteins/metabolism , Peptide Elongation Factor Tu/metabolism , Phenylalanine/analogs & derivatives , Protein Engineering/methods , Amino Acyl-tRNA Synthetases/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Molecular Dynamics Simulation , Mutation , Phenylalanine/metabolism , Phenylalanine-tRNA Ligase/metabolism , RNA, Ribosomal, 23S , Substrate Specificity , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL