Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters










Publication year range
2.
Adv Sci (Weinh) ; 11(11): e2307930, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38164822

ABSTRACT

5-Methylcytosine (5mC) is the central epigenetic mark of mammalian DNA, and plays fundamental roles in chromatin regulation. 5mC is dynamically read and translated into regulatory outputs by methyl-CpG-binding domain (MBD) proteins. These multidomain readers recognize 5mC via an MBD domain, and undergo additional domain-dependent interactions with multiple additional chromatin components. However, studying this dynamic process is limited by a lack of methods to conditionally control the 5mC affinity of MBD readers in cells. Light-control of MBD association to chromatin by genetically encoding a photocaged serine at the MBD-DNA interface is reported. The authors study the association of MBD1 to mouse pericentromeres, dependent on its CxxC3 and transcriptional repressor domains (TRD) which interact with unmethylated CpG and heterochromatin-associated complexes, respectively. Both domains significantly modulate association kinetics, arguing for a model in which the CxxC3 delays methylation responses of MBD1 by holding it at unmethylated loci, whereas the TRD promotes responses by aiding heterochromatin association is studied. Their approach offers otherwise inaccessible kinetic insights into the domain-specific regulation of a central MBD reader, and sets the basis for further unravelling how the integration of MBDs into complex heterochromatin interaction networks control the kinetics of 5mC reading and translation into altered chromatin states.


Subject(s)
Chromatin , DNA-Binding Proteins , Animals , Mice , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , 5-Methylcytosine , Heterochromatin , DNA Methylation , Transcription Factors/genetics , DNA/metabolism , Mammals/metabolism
3.
Angew Chem Int Ed Engl ; 63(17): e202318837, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38284298

ABSTRACT

Mammalian genomes are regulated by epigenetic cytosine (C) modifications in palindromic CpG dyads. Including canonical cytosine 5-methylation (mC), a total of four different 5-modifications can theoretically co-exist in the two strands of a CpG, giving rise to a complex array of combinatorial marks with unique regulatory potentials. While tailored readers for individual marks could serve as versatile tools to study their functions, it has been unclear whether a natural protein scaffold would allow selective recognition of marks that vastly differ from canonical, symmetrically methylated CpGs. We conduct directed evolution experiments to generate readers of 5-carboxylcytosine (caC) dyads based on the methyl-CpG-binding domain (MBD), the widely conserved natural reader of mC. Despite the stark steric and chemical differences to mC, we discover highly selective, low nanomolar binders of symmetric and asymmetric caC-dyads. Together with mutational and modelling studies, our findings reveal a striking evolutionary flexibility of the MBD scaffold, allowing it to completely abandon its conserved mC recognition mode in favour of noncanonical dyad recognition, highlighting its potential for epigenetic reader design.


Subject(s)
Cytosine , Cytosine/analogs & derivatives , DNA Methylation , Animals , CpG Islands , Cytosine/chemistry , Epigenesis, Genetic , Mammals/metabolism
4.
Nucleic Acids Res ; 51(12): 6495-6506, 2023 07 07.
Article in English | MEDLINE | ID: mdl-36919612

ABSTRACT

5-methylcytosine (mC) and its TET-oxidized derivatives exist in CpG dyads of mammalian DNA and regulate cell fate, but how their individual combinations in the two strands of a CpG act as distinct regulatory signals is poorly understood. Readers that selectively recognize such novel 'CpG duplex marks' could be versatile tools for studying their biological functions, but their design represents an unprecedented selectivity challenge. By mutational studies, NMR relaxation, and MD simulations, we here show that the selectivity of the first designer reader for an oxidized CpG duplex mark hinges on precisely tempered conformational plasticity of the scaffold adopted during directed evolution. Our observations reveal the critical aspect of defined motional features in this novel reader for affinity and specificity in the DNA/protein interaction, providing unexpected prospects for further design progress in this novel area of DNA recognition.


Subject(s)
5-Methylcytosine , DNA , Epigenesis, Genetic , Animals , CpG Islands/genetics , DNA/chemistry , DNA Methylation , Epigenomics , Mammals/metabolism , Molecular Conformation
5.
ACS Chem Biol ; 18(2): 230-236, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36693632

ABSTRACT

Transcription-activator-like effectors (TALEs) are programmable DNA binding proteins that can be used for sequence-specific, imaging-based analysis of cellular 5-methylcytosine. However, this has so far been limited to highly repetitive satellite DNA. To expand this approach to the analysis of coding single gene loci, we here explore a number of signal amplification strategies for increasing imaging sensitivity with TALEs. We develop a straightforward amplification protocol and employ it to target the MUC4 gene, which features only a small cluster of repeat sequences. This offers high sensitivity imaging of MUC4, and in costaining experiments with pairs of one TALE selective for unmethylated cytosine and one universal control TALE enables analyzing methylation changes in the target independently of changes in target accessibility. These advancements offer prospects for 5-methylcytosine analysis at coding, nonrepetitive gene loci by the use of designed TALE probe collections.


Subject(s)
5-Methylcytosine , Transcription Activator-Like Effectors , Transcription Activator-Like Effectors/genetics , 5-Methylcytosine/metabolism , DNA/genetics , DNA/metabolism , Repetitive Sequences, Nucleic Acid , DNA-Binding Proteins/metabolism
6.
RSC Med Chem ; 13(12): 1540-1548, 2022 Dec 14.
Article in English | MEDLINE | ID: mdl-36545435

ABSTRACT

Ten-eleven translocation dioxygenases (TETs) are the erasers of 5-methylcytosine (mC), the central epigenetic regulator of mammalian DNA. TETs convert mC to three oxidized derivatives with unique physicochemical properties and inherent regulatory potential, and it initializes active demethylation by the base excision repair pathway. Potent small molecule inhibitors would be useful tools to study TET functions by conditional control. To facilitate the discovery of such tools, we here report a high-throughput screening pipeline and its application to screen and validate 31.5k compounds for inhibition of TET2. Using a homogenous fluorescence assay, we discover a novel quinoline-based scaffold that we further validate with an orthogonal semi-high throughput MALDI-MS assay for direct monitoring of substrate turnover. Structure-activity relationship (SAR) studies involving >20 derivatives of this scaffold led to the identification of optimized inhibitors, and together with computational studies suggested a plausible model for its mode of action.

7.
ACS Chem Biol ; 17(7): 1844-1852, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35709470

ABSTRACT

Methyl-CpG binding domain (MBD) proteins and ten-eleven-translocation (TET) dioxygenases are the readers and erasers of 5-methylcytosine (5mC), the central epigenetic mark of mammalian DNA. We employ light-activatable human TET1 controlled by a genetically encoded photocaged serine to enable in vivo kinetic studies of their interplay at the common substrate methylated cytosine-guanine (mCpG). We identify the multidomain reader MBD1 to negatively regulate TET1-catalyzed 5mC oxidation kinetics via its mCpG-binding MBD domain. However, we also identify the third Cys-x-x-Cys (CXXC3) domain of MBD1 to promote oxidation kinetics by TET1, dependent on its ability to bind nonmethylated CpG, the final product of TET-mediated mCpG oxidation and active demethylation. In contrast, we do not observe differences in TET1 regulation for MBD1 variants with or without the transcriptional repressor domain. Our approach reveals a complex, domain-dependent interplay of these readers and erasers of 5mC with different domain-specific contributions of MBD1 to the overall kinetics of TET1-catalyzed global 5mC oxidation kinetics that contribute to a better understanding of dynamic methylome shaping.


Subject(s)
5-Methylcytosine , Dioxygenases , 5-Methylcytosine/metabolism , Animals , Cytosine/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , Humans , Kinetics , Mammals/metabolism , Mixed Function Oxygenases/metabolism , Oxidation-Reduction , Proto-Oncogene Proteins/metabolism , Reading , Transcription Factors/metabolism
8.
J Am Chem Soc ; 144(7): 2987-2993, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35157801

ABSTRACT

5-Methylcytosine (mC) and 5-hydroxymethylcytosine (hmC), the two main epigenetic modifications of mammalian DNA, exist in symmetric and asymmetric combinations in the two strands of CpG dyads. However, revealing such combinations in single DNA duplexes is a significant challenge. Here, we evolve methyl-CpG-binding domains (MBDs) derived from MeCP2 by bacterial cell surface display, resulting in the first affinity probes for hmC/mC CpGs. One mutant has low nanomolar affinity for a single hmC/mC CpG, discriminates against all 14 other modified CpG dyads, and rivals the selectivity of wild-type MeCP2. Structural studies indicate that this protein has a conserved scaffold and recognizes hmC and mC with two dedicated sets of residues. The mutant allows us to selectively address and enrich hmC/mC-containing DNA fragments from genomic DNA backgrounds. We anticipate that this novel probe will be a versatile tool to unravel the function of hmC/mC marks in diverse aspects of chromatin biology.


Subject(s)
5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/chemistry , DNA/isolation & purification , Methyl-CpG-Binding Protein 2/chemistry , Peptide Fragments/chemistry , DNA/chemistry , DNA Methylation , Directed Molecular Evolution , HEK293 Cells , Humans , Methyl-CpG-Binding Protein 2/genetics , Peptide Fragments/genetics , Protein Domains
9.
Cell Death Dis ; 12(6): 530, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34031359

ABSTRACT

Non-coding RNA from pericentromeric satellite repeats are involved in stress-dependent splicing processes, maintenance of heterochromatin, and are required to protect genome stability. Here we show that the long non-coding satellite III RNA (SatIII) generates resistance against the topoisomerase IIa (TOP2A) inhibitor etoposide in lung cancer. Because heat shock conditions (HS) protect cells against the toxicity of etoposide, and SatIII is significantly induced under HS, we hypothesized that the protective effect could be traced back to SatIII. Using genome methylation profiles of patient-derived xenograft mouse models we show that the epigenetic modification of the SatIII DNA locus and the resulting SatIII expression predict chemotherapy resistance. In response to stress, SatIII recruits TOP2A to nuclear stress bodies, which protects TOP2A from a complex formation with etoposide and results in decreased DNA damage after treatment. We show that BRD4 inhibitors reduce the expression of SatIII, restoring etoposide sensitivity.


Subject(s)
Drug Resistance, Neoplasm/genetics , Etoposide/therapeutic use , RNA, Long Noncoding/physiology , Animals , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Centromere/genetics , Centromere/metabolism , DNA Methylation/physiology , DNA Topoisomerases, Type II/drug effects , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , HeLa Cells , Humans , Male , Mice, Inbred NOD , Mice, SCID , Poly-ADP-Ribose Binding Proteins/drug effects , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , RNA, Long Noncoding/genetics , Transcription Factors/antagonists & inhibitors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Angew Chem Int Ed Engl ; 60(24): 13507-13512, 2021 06 07.
Article in English | MEDLINE | ID: mdl-33826797

ABSTRACT

5-Methylcytosine (5mC), the central epigenetic mark of mammalian DNA, plays fundamental roles in chromatin regulation. 5mC is written onto genomes by DNA methyltransferases (DNMT), and perturbation of this process is an early event in carcinogenesis. However, studying 5mC functions is limited by the inability to control individual DNMTs with spatiotemporal resolution in vivo. We report light-control of DNMT catalysis by genetically encoding a photocaged cysteine as a catalytic residue. This enables translation of inactive DNMTs, their rapid activation by light-decaging, and subsequent monitoring of de novo DNA methylation. We provide insights into how cancer-related DNMT mutations alter de novo methylation in vivo, and demonstrate local and tuneable cytosine methylation by light-controlled DNMTs fused to a programmable transcription activator-like effector domain targeting pericentromeric satellite-3 DNA. We further study early events of transcriptome alterations upon DNMT-catalyzed cytosine methylation. Our study sets a basis to dissect the order and kinetics of diverse chromatin-associated events triggered by normal and aberrant DNA methylation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/radiation effects , Light , 5-Methylcytosine/metabolism , Biocatalysis , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA (Cytosine-5-)-Methyltransferases/genetics , HEK293 Cells , Humans , Mutation , Transcriptome/radiation effects
11.
Curr Opin Chem Biol ; 63: 1-10, 2021 08.
Article in English | MEDLINE | ID: mdl-33588304

ABSTRACT

Modifications of the cytosine 5-position are dynamic epigenetic marks of mammalian DNA with important regulatory roles in development and disease. Unraveling biological functions of such modified nucleobases is tightly connected with the potential of available methods for their analysis. Whereas genome-wide nucleobase quantification and mapping are first-line analyses, targeted analyses move into focus the more genomic sites with high biological significance are identified. We here review recent developments in an emerging field that addresses such targeted analyses via probes that combine a programmable, sequence-specific DNA-binding domain with the ability to directly recognize or cross-link an epigenetically modified nucleobase of interest. We highlight how such probes offer simple, high-resolution nucleobase analyses in vitro and enable in situ correlations between a nucleobase and other chromatin regulatory elements at user-defined loci on the single-cell level by imaging.


Subject(s)
5-Methylcytosine/chemistry , DNA/chemistry , Epigenesis, Genetic/genetics , Binding Sites , Chromatin/chemistry , Cross-Linking Reagents/chemistry , DNA Methylation , DNA-Directed DNA Polymerase/metabolism , Fluorescent Dyes/chemistry , Genomics , Humans , Molecular Conformation , Molecular Imaging , Optical Imaging , Single-Cell Analysis
12.
Int J Mol Sci ; 22(4)2021 Feb 04.
Article in English | MEDLINE | ID: mdl-33557445

ABSTRACT

The authors wish to make the following two corrections to this paper [...].

13.
Chembiochem ; 22(4): 645-651, 2021 02 15.
Article in English | MEDLINE | ID: mdl-32991020

ABSTRACT

Transcription-activator-like effectors (TALEs) are repeat-based, programmable DNA-binding proteins that can be engineered to recognize sequences of canonical and epigenetically modified nucleobases. Fluorescent TALEs can be used for the imaging-based analysis of cellular 5-methylcytosine (5 mC) in repetitive DNA sequences. This is based on recording fluorescence ratios from cell co-stains with two TALEs: an analytical TALE targeting the cytosine (C) position of interest through a C-selective repeat that is blocked by 5 mC, and a control TALE targeting the position with a universal repeat that binds both C and 5 mC. To enhance this approach, we report herein the development of novel 5 mC-selective repeats and their integration into TALEs that can replace universal TALEs in imaging-based 5 mC analysis, resulting in a methylation-dependent response of both TALEs. We screened a library of size-reduced repeats and identified several 5 mC binders. Compared to the 5 mC-binding repeat of natural TALEs and to the universal repeat, two repeats containing aromatic residues showed enhancement of 5 mC binding and selectivity in cellular transcription activation and electromobility shift assays, respectively. In co-stains of cellular SATIII DNA with a corresponding C-selective TALE, this selectivity results in a positive methylation response of the new TALE, offering perspectives for studying 5 mC functions in chromatin regulation by in situ imaging with increased dynamic range.


Subject(s)
5-Methylcytosine/analysis , DNA Methylation , Image Processing, Computer-Assisted/methods , Molecular Probes/metabolism , Repetitive Sequences, Nucleic Acid , Transcription Activator-Like Effectors/metabolism , Genetic Engineering , HEK293 Cells , Humans , Molecular Probes/chemistry , Transcription Activator-Like Effectors/chemistry
14.
Methods Mol Biol ; 2198: 381-399, 2021.
Article in English | MEDLINE | ID: mdl-32822046

ABSTRACT

Transcription-activator like effectors (TALEs) are DNA-binding proteins used for genome targeting. TALEs contain a central domain of concatenated repeats, of which each selectively recognizes one nucleobase at the DNA major groove. Based on this simple and predictable interaction with little context dependence, TALEs offer programmable targeting of user-defined DNA sequences. Since many epigenetic DNA modifications protrude into the DNA major groove, natural and engineered TALE repeats can provide "epigenetic" selectivity, making TALEs a flexible platform to design probes for the analysis of epigenetic DNA modifications. Here, we describe guidelines for the design of TALE proteins with selectivity for epigenetic cytosine 5-modifications, the validation of their interaction with modified DNA nucleobases, and their employment in affinity enrichment assays. These techniques enable quantification of epigenetic nucleobases in user-defined genomic DNA sequences with nucleotide and strand resolution.


Subject(s)
Epigenomics/methods , Transcription Activator-Like Effectors/chemical synthesis , Transcription Activator-Like Effectors/metabolism , 5-Methylcytosine/chemistry , Animals , Cytosine/metabolism , DNA/chemistry , DNA Methylation/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic/genetics , Genome/genetics , Humans , Transcription Activator-Like Effectors/genetics
15.
J Am Chem Soc ; 142(16): 7289-7294, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32286069

ABSTRACT

Ten-eleven-translocation (TET) dioxygenases catalyze the oxidation of 5-methylcytosine (5mC), the central epigenetic regulator of mammalian DNA. This activity dynamically reshapes the epigenome and transcriptome by depositing oxidized 5mC derivatives and initiating active DNA demethylation. However, studying this dynamic is hampered by the inability to selectively activate individual TETs with temporal control in cells. We report activation of TETs in mammalian cells by incorporation of genetically encoded 4,5-dimethoxy-2-nitrobenzyl-l-serine as a transient active-site block, and its subsequent deprotection with light. Our approach enables precise insights into the impact of cancer-associated TET2 mutations on the kinetics of TET2 catalysis in vivo, and allows time-resolved monitoring of target gene activation and transcriptome reorganization. This sets a basis for dissecting the order and kinetics of chromatin-associated events triggered by TET catalysis, ranging from DNA demethylation to chromatin and transcription regulation.


Subject(s)
5-Methylcytosine/metabolism , Dioxygenases/metabolism , Humans , Oxidation-Reduction , Transcriptome
16.
Sci Rep ; 10(1): 4053, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32132616

ABSTRACT

5-Methylcytosine (mC) exists in CpG dinucleotides of mammalian DNA and plays key roles in chromatin regulation during development and disease. As a main regulatory pathway, fully methylated CpG are recognized by methyl-CpG-binding domain (MBD) proteins that act in concert with chromatin remodelers, histone deacetylases and methyltransferases to trigger transcriptional downregulation. In turn, MBD mutations can alter CpG binding, and in case of the MBD protein MeCP2 can cause the neurological disorder Rett syndrome (RTT). An additional layer of complexity in CpG recognition is added by ten-eleven-translocation (TET) dioxygenases that oxidize mC to 5-hydroxymethyl-, 5-formyl- and 5-carboxylcytosine, giving rise to fifteen possible combinations of cytosine modifications in the two CpG strands. We report a comprehensive, comparative interaction analysis of the human MBD proteins MeCP2, MBD1, MBD2, MBD3, and MBD4 with all CpG combinations and observe individual preferences of each MBD for distinct combinations. In addition, we profile four MeCP2 RTT mutants and reveal that although interactions to methylated CpGs are similarly affected by the mutations, interactions to oxidized mC combinations are differentially affected. These findings argue for a complex interplay between local TET activity/processivity and CpG recognition by MBDs, with potential consequences for the transcriptional landscape in normal and RTT states.


Subject(s)
CpG Islands , Cytosine/analogs & derivatives , Cytosine/chemistry , Dinucleotide Repeats , Methyl-CpG-Binding Protein 2/chemistry , Rett Syndrome , Cytosine/metabolism , Humans , Methyl-CpG-Binding Protein 2/metabolism
17.
Angew Chem Int Ed Engl ; 59(23): 8927-8931, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32167219

ABSTRACT

We report programmable receptors for the imaging-based analysis of 5-methylcytosine (5mC) in user-defined DNA sequences of single cells. Using fluorescent transcription-activator-like effectors (TALEs) that can recognize sequences of canonical and epigenetic nucleobases through selective repeats, we imaged cellular SATIII DNA, the origin of nuclear stress bodies (nSB). We achieve high nucleobase selectivity of natural repeats in imaging and demonstrate universal nucleobase binding by an engineered repeat. We use TALE pairs differing in only one such repeat in co-stains to detect 5mC in SATIII sequences with nucleotide resolution independently of differences in target accessibility. Further, we directly correlate the presence of heat shock factor 1 with 5mC at its recognition sequence, revealing a potential function of 5mC in its recruitment as initial step of nSB formation. This opens a new avenue for studying 5mC functions in chromatin regulation in situ with nucleotide, locus, and cell resolution.


Subject(s)
5-Methylcytosine/metabolism , Genomics , Molecular Imaging , Nucleotides/metabolism , HeLa Cells , Humans , Single-Cell Analysis
18.
Phys Chem Chem Phys ; 22(9): 4875-4879, 2020 Mar 07.
Article in English | MEDLINE | ID: mdl-32072999

ABSTRACT

Structural studies on proteins directly in their native environment are required for a comprehensive understanding of their function. Electron paramagnetic resonance (EPR) spectroscopy and in particular double electron-electron resonance (DEER) distance determination are suited to investigate spin-labeled proteins directly in the cell. The combination of intracellular bioorthogonal labeling with in-cell DEER measurements does not require additional purification or delivery steps of spin-labeled protein to the cells. In this study, we express eGFP in E. coli and use copper-catalyzed azide-alkyne cycloaddition (CuAAC) for the site-directed spin labeling of the protein in vivo, followed by in-cell EPR distance determination. Inter-spin distance measurements of spin-labeled eGFP agree with in vitro measurements and calculations based on the rotamer library of the spin label.


Subject(s)
Electron Spin Resonance Spectroscopy , Escherichia coli/metabolism , Green Fluorescent Proteins/chemistry , Alkynes/chemistry , Azides/chemistry , Catalysis , Copper/chemistry , Cycloaddition Reaction , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Nitrogen Oxides/chemistry , Spin Labels
19.
Chem Sci ; 11(46): 12506-12511, 2020 Oct 20.
Article in English | MEDLINE | ID: mdl-34123231

ABSTRACT

Enrichment of chromatin segments from specific genomic loci of living cells is an important goal in chromatin biology, since it enables establishing local molecular compositions as the basis of locus function. A central enrichment strategy relies on the expression of DNA-binding domains that selectively interact with a local target sequence followed by fixation and isolation of the associated chromatin segment. The efficiency and selectivity of this approach critically depend on the employed enrichment tag and the strategy used for its introduction into the DNA-binding domain or close-by proteins. We here report chromatin enrichment by expressing programmable transcription-activator-like effectors (TALEs) bearing single strained alkynes or alkenes introduced via genetic code expansion. This enables in situ biotinylation at a defined TALE site via strain-promoted inverse electron demand Diels Alder cycloadditions for single-step, high affinity enrichment. By targeting human pericentromeric SATIII repeats, the origin of nuclear stress bodies, we demonstrate enrichment of SATIII DNA and SATIII-associated proteins, and identify factors enriched during heat stress.

20.
Chembiochem ; 21(7): 958-962, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31657498

ABSTRACT

Electron paramagnetic resonance (EPR) spectroscopy in combination with site-directed spin labeling (SDSL) is a powerful tool in protein structural research. Nitroxides are highly suitable spin labeling reagents, but suffer from limited stability, particularly in the cellular environment. Herein we present the synthesis of a maleimide- and an azide-modified tetraethyl-shielded isoindoline-based nitroxide (M- and Az-TEIO) for labeling of cysteines or the noncanonical amino acid para-ethynyl-l-phenylalanine (pENF). We demonstrate the high stability of TEIO site-specifically attached to the protein thioredoxin (TRX) against reduction in prokaryotic and eukaryotic environments, and conduct double electron-electron resonance (DEER) measurements. We further generate a rotamer library for the new residue pENF-Az-TEIO that affords a distance distribution that is in agreement with the measured distribution.


Subject(s)
Alkynes/chemistry , Amino Acids/chemistry , Cysteine/chemistry , Nitrogen Oxides/chemistry , Azides/chemistry , Electron Spin Resonance Spectroscopy , Isoindoles/chemistry , Spin Labels , Thioredoxins/chemistry , Thioredoxins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...