Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Anal Chem ; 96(19): 7373-7379, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38696819

ABSTRACT

Cross-linking mass spectrometry (XL-MS) has evolved into a pivotal technique for probing protein interactions. This study describes the implementation of Parallel Accumulation-Serial Fragmentation (PASEF) on timsTOF instruments, enhancing the detection and analysis of protein interactions by XL-MS. Addressing the challenges in XL-MS, such as the interpretation of complex spectra, low abundant cross-linked peptides, and a data acquisition bias, our current study integrates a peptide-centric approach for the analysis of XL-MS data and presents the foundation for integrating data-independent acquisition (DIA) in XL-MS with a vendor-neutral and open-source platform. A novel workflow is described for processing data-dependent acquisition (DDA) of PASEF-derived information. For this, software by Bruker Daltonics is used, enabling the conversion of these data into a format that is compatible with MeroX and Skyline software tools. Our approach significantly improves the identification of cross-linked products from complex mixtures, allowing the XL-MS community to overcome current analytical limitations.


Subject(s)
Cross-Linking Reagents , Mass Spectrometry , Software , Workflow , Cross-Linking Reagents/chemistry , Peptides/chemistry , Peptides/analysis , Humans
2.
Protein Sci ; 32(9): e4753, 2023 09.
Article in English | MEDLINE | ID: mdl-37572332

ABSTRACT

Within the cell, the trace element molybdenum (Mo) is only biologically active when complexed either within the nitrogenase-specific FeMo cofactor or within the molybdenum cofactor (Moco). Moco consists of an organic part, called molybdopterin (MPT) and an inorganic part, that is, the Mo-center. The enzyme which catalyzes the Mo-center formation is the molybdenum insertase (Mo-insertase). Mo-insertases consist of two functional domains called G- and E-domain. The G-domain catalyzes the formation of adenylated MPT (MPT-AMP), which is the substrate for the E-domain, that catalyzes the actual molybdate insertion reaction. Though the functions of E- and G-domain have been elucidated to great structural and mechanistic detail, their combined function is poorly characterized. In this work, we describe a structural model of the eukaryotic Mo-insertase Cnx1 complex that was generated based on cross-linking mass spectrometry combined with computational modeling. We revealed Cnx1 to form an asymmetric hexameric complex which allows the E- and G-domain active sites to align in a catalytic productive orientation toward each other.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Metalloproteins , Arabidopsis Proteins/chemistry , Calnexin/chemistry , Calnexin/metabolism , Arabidopsis/chemistry , Molybdenum/metabolism , Coenzymes/chemistry , Metalloproteins/chemistry , Pteridines/chemistry
3.
Protein J ; 42(3): 219-228, 2023 06.
Article in English | MEDLINE | ID: mdl-37233895

ABSTRACT

Metamorphic, or fold-switching, proteins feature different folds that are physiologically relevant. The human chemokine XCL1 (or Lymphotactin) is a metamorphic protein that features two native states, an [Formula: see text] and an all[Formula: see text] fold, which have similar stability at physiological condition. Here, extended molecular dynamics (MD) simulations, principal component analysis of atomic fluctuations and thermodynamic modeling based on both the configurational volume and free energy landscape, are used to obtain a detailed characterization of the conformational thermodynamics of human Lymphotactin and of one of its ancestors (as was previously obtained by genetic reconstruction). Comparison of our computational results with the available experimental data show that the MD-based thermodynamics can explain the experimentally observed variation of the conformational equilibrium between the two proteins. In particular, our computational data provide an interpretation of the thermodynamic evolution in this protein, revealing the relevance of the configurational entropy and of the shape of the free energy landscape within the essential space (i.e., the space defined by the generalized internal coordinates providing the largest, typically non-Gaussian, structural fluctuations).


Subject(s)
Lymphokines , Sialoglycoproteins , Humans , Thermodynamics , Lymphokines/chemistry , Lymphokines/metabolism , Sialoglycoproteins/chemistry , Sialoglycoproteins/metabolism , Molecular Dynamics Simulation
4.
Sci Rep ; 13(1): 8497, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37231156

ABSTRACT

The tetrameric tumor suppressor p53 represents a great challenge for 3D-structural analysis due to its high degree of intrinsic disorder (ca. 40%). We aim to shed light on the structural and functional roles of p53's C-terminal region in full-length, wild-type human p53 tetramer and their importance for DNA binding. For this, we employed complementary techniques of structural mass spectrometry (MS) in an integrated approach with computational modeling. Our results show no major conformational differences in p53 between DNA-bound and DNA-free states, but reveal a substantial compaction of p53's C-terminal region. This supports the proposed mechanism of unspecific DNA binding to the C-terminal region of p53 prior to transcription initiation by specific DNA binding to the core domain of p53. The synergies between complementary structural MS techniques and computational modeling as pursued in our integrative approach is envisioned to serve as general strategy for studying intrinsically disordered proteins (IDPs) and intrinsically disordered region (IDRs).


Subject(s)
Intrinsically Disordered Proteins , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Computer Simulation , Intrinsically Disordered Proteins/chemistry , DNA/metabolism , Mass Spectrometry , Protein Binding
5.
Angew Chem Int Ed Engl ; 61(46): e202205726, 2022 11 14.
Article in English | MEDLINE | ID: mdl-36115020

ABSTRACT

α-Synuclein (α-syn) is an intrinsically disordered protein (IDP) that undergoes liquid-liquid phase separation (LLPS), fibrillation, and forms insoluble intracellular Lewy bodies in neurons, which are the hallmark of Parkinson's Disease (PD). Neurotoxicity precedes the formation of aggregates and might be related to α-syn LLPS. The molecular mechanisms underlying the early stages of LLPS are still elusive. To obtain structural insights into α-syn upon LLPS, we take advantage of cross-linking/mass spectrometry (XL-MS) and introduce an innovative approach, termed COMPASS (COMPetitive PAiring StatisticS). In this work, we show that the conformational ensemble of α-syn shifts from a "hairpin-like" structure towards more "elongated" conformational states upon LLPS. We obtain insights into the critical initial stages of LLPS and establish a novel mass spectrometry-based approach that will aid to solve open questions in LLPS structural biology.


Subject(s)
Intrinsically Disordered Proteins , Parkinson Disease , Humans , alpha-Synuclein/chemistry , Parkinson Disease/metabolism , Intrinsically Disordered Proteins/chemistry , Neurons/metabolism , Molecular Conformation
6.
Chembiochem ; 23(11): e202100665, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35333001

ABSTRACT

The tumor suppressor protein p53 is a transcription factor that is referred to as the "guardian of the genome" and plays an important role in cancer development. p53 is active as a homotetramer; the S100ß homodimer binds to the intrinsically disordered C-terminus of p53 affecting its transcriptional activity. The p53/S100ß complex is regarded as highly promising therapeutic target in cancer. It has been suggested that S100ß exerts its oncogenic effects by altering the p53 oligomeric state. Our aim was to study the structures and oligomerization behavior of different p53/S100ß complexes by ESI-MS, XL-MS, and SPR. Wild-type p53 and single amino acid variants, representing different oligomeric states of p53 were individually investigated regarding their binding behavior towards S100ß. The stoichiometry of the different p53/S100ß complexes were determined by ESI-MS showing that tetrameric, dimeric, and monomeric p53 variants all bind to an S100ß dimer. In addition, XL-MS revealed the topologies of the p53/S100ß complexes to be independent of p53's oligomeric state. With SPR, the thermodynamic parameters were determined for S100ß binding to tetrameric, dimeric, or monomeric p53 variants. Our data prove that the S100ß homodimer binds to different oligomeric states of p53 with similar binding affinities. This emphasizes the need for alternative explanations to describe the molecular mechanisms underlying p53/S100ß interaction.


Subject(s)
Neoplasms , Tumor Suppressor Protein p53 , Humans , Protein Binding , S100 Calcium Binding Protein beta Subunit , Transcription Factors/metabolism , Tumor Suppressor Protein p53/chemistry
7.
Int J Biol Macromol ; 182: 502-511, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33848543

ABSTRACT

High-resolution structural data of complexes between antibodies and membrane receptors still represent a demanding task. In this study, we used complementary sets of experimental data to obtain a structural model of the complex formed by the human epidermal growth factor receptor 2 (HER2) and its specific nanobody A10. First we identified by NMR the residues that bind or rearrange as a consequence of the complex formation. In parallel, the complex was cross-linked, digested and the resulting peptides were characterized by mass-spectrometry to define maximal distance restraints between HER2 and A10 amino acids in their complex. These independent datasets guided a docking process, refined by molecular dynamics simulations, to develop a model of the complex and estimate per-residue free-energy contributions. Such a model explains the experimental data and identifies a second, non-canonical paratope, located in the region opposite to the conventional nanobody paratope, formed by the hypervariable loop regions LH1 and LH3. Both paratopes contributed substantially to the overall affinity by binding to independent HER2 epitopes. Nanobody mutants with substitution of key interaction residues, as indicated by the model, possess significantly lower affinity for HER2. This is the first described case of a "natural" biparatopic nanobody, directly selected by in-vitro panning.


Subject(s)
Binding Sites, Antibody , Receptor, ErbB-2/chemistry , Single-Chain Antibodies/chemistry , Humans , Molecular Docking Simulation , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/immunology , Protein Binding , Receptor, ErbB-2/immunology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology
8.
J Mol Biol ; 433(10): 166947, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33744315

ABSTRACT

The rod-outer-segment guanylyl cyclase 1 (ROS-GC1) is a key transmembrane protein for retinal phototransduction. Mutations of ROS-GC1 correlate with different retinal diseases that often lead to blindness. No structural data are available for ROS-GC1 so far. We performed a 3D-structural analysis of native ROS-GC1 from bovine retina by cross-linking/mass spectrometry (XL-MS) and computational modeling. Absolute quantification and activity measurements of native ROS-GC1 were performed by MS-based assays directly in bovine retina samples. Our data present the first 3D-structural analysis of active, full-length ROS-GC1 derived from bovine retina. We propose a novel domain organization for the intracellular domain ROS-GC1. Our XL-MS data of native ROS-GC1 from rod-outer-segment preparations of bovine retina agree with a dimeric architecture. Our integrated approach can serve as a blueprint for conducting 3D-structural studies of membrane proteins in their native environment.


Subject(s)
Cyclic GMP/chemistry , Guanylate Cyclase/chemistry , Peptides/metabolism , Receptors, Cell Surface/chemistry , Rod Cell Outer Segment/chemistry , Amino Acid Motifs , Animals , Binding Sites , Cattle , Cloning, Molecular , Cross-Linking Reagents/chemistry , Cyclic GMP/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , HEK293 Cells , Humans , Mass Spectrometry/methods , Models, Molecular , Peptides/chemical synthesis , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Rod Cell Outer Segment/metabolism , Succinimides/chemistry
10.
Sci Rep ; 10(1): 12618, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32724211

ABSTRACT

The ribosome is not only a highly complex molecular machine that translates the genetic information into proteins, but also an exceptional specimen for testing and optimizing cross-linking/mass spectrometry (XL-MS) workflows. Due to its high abundance, ribosomal proteins are frequently identified in proteome-wide XL-MS studies of cells or cell extracts. Here, we performed in-depth cross-linking of the E. coli ribosome using the amine-reactive cross-linker disuccinimidyl diacetic urea (DSAU). We analyzed 143 E. coli ribosomal structures, mapping a total of 10,771 intramolecular distances for 126 cross-link-pairs and 3,405 intermolecular distances for 97 protein pairs. Remarkably, 44% of intermolecular cross-links covered regions that have not been resolved in any high-resolution E. coli ribosome structure and point to a plasticity of cross-linked regions. We systematically characterized all cross-links and discovered flexible regions, conformational changes, and stoichiometric variations in bound ribosomal proteins, and ultimately remodeled 2,057 residues (15,794 atoms) in total. Our working model explains more than 95% of all cross-links, resulting in an optimized E. coli ribosome structure based on the cross-linking data obtained. Our study might serve as benchmark for conducting biochemical experiments on newly modeled protein regions, guided by XL-MS. Data are available via ProteomeXchange with identifier PXD018935.


Subject(s)
Cross-Linking Reagents/chemistry , Mass Spectrometry , Molecular Conformation , Ribosomes/chemistry , Catalytic Domain , Cryoelectron Microscopy , Escherichia coli/metabolism , Models, Molecular , Molecular Chaperones/chemistry , Pliability , Protein Binding , RNA, Messenger/chemistry , Ribosomal Proteins/chemistry , Rotation , X-Rays
11.
Nat Commun ; 11(1): 2277, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32385295

ABSTRACT

Cullin RING-type E3 ubiquitin ligases SCFTIR1/AFB1-5 and their AUX/IAA targets perceive the phytohormone auxin. The F-box protein TIR1 binds a surface-exposed degron in AUX/IAAs promoting their ubiquitylation and rapid auxin-regulated proteasomal degradation. Here, by adopting biochemical, structural proteomics and in vivo approaches we unveil how flexibility in AUX/IAAs and regions in TIR1 affect their conformational ensemble allowing surface accessibility of degrons. We resolve TIR1·auxin·IAA7 and TIR1·auxin·IAA12 complex topology, and show that flexible intrinsically disordered regions (IDRs) in the degron's vicinity, cooperatively position AUX/IAAs on TIR1. We identify essential residues at the TIR1 N- and C-termini, which provide non-native interaction interfaces with IDRs and the folded PB1 domain of AUX/IAAs. We thereby establish a role for IDRs in modulating auxin receptor assemblies. By securing AUX/IAAs on two opposite surfaces of TIR1, IDR diversity supports locally tailored positioning for targeted ubiquitylation, and might provide conformational flexibility for a multiplicity of functional states.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Indoleacetic Acids/metabolism , Intrinsically Disordered Proteins/metabolism , Proteolysis , Receptors, Cell Surface/metabolism , Arabidopsis Proteins/chemistry , Binding Sites , Intrinsically Disordered Proteins/chemistry , Molecular Dynamics Simulation , Mutation/genetics , Phylogeny , Protein Domains , Ubiquitination
12.
J Am Soc Mass Spectrom ; 31(2): 183-189, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-32031397

ABSTRACT

Previous studies have shown the benefits of the amine-reactive, CID-MS/MS-cleavable cross-linker disuccinimidyl dibutyric urea (DSBU) for structural proteomics studies via cross-linking/MS (XL-MS). To further facilitate the automation of XL-MS experiments, we synthesized a deuterated (D12) version of the DSBU cross-linker combining the advantages of MS-cleavable linkers and isotope labeling. The rationale of conducting XL-MS with a mixture of unlabeled and stable isotope-labeled DSBU is to obtain characteristic mass differences at the MS level indicating cross-linked species. These cross-linked species can then be selected for fragmentation by collisional activation. At the MS/MS level, the characteristic 26-u doublets arising from cleavage of the central urea group in DSBU confirm the amino acid sequences of cross-linked peptides as well as the exact cross-linking sites. D12-labeled DSBU was tested on three systems with increasing complexity: (i) bovine serum albumin as purified protein, (ii) Escherichia coli ribosome as large, multimeric protein assembly, and (iii) Drosophila embryo extract as complete proteome. We demonstrate the benefits arising from the use of isotope-labeled DSBU for an automated assignment of cross-linked products. Combining isotope labeling and MS cleavability in one cross-linker resulted in higher cross-link identification numbers especially for highly complex protein mixtures.


Subject(s)
Cross-Linking Reagents/chemistry , Proteins/chemistry , Succinimides/chemistry , Tandem Mass Spectrometry/methods , Urea/chemistry , Deuterium/chemistry , Isotope Labeling/methods , Protein Conformation , Proteins/analysis
13.
Curr Opin Biotechnol ; 63: 48-53, 2020 06.
Article in English | MEDLINE | ID: mdl-31891863

ABSTRACT

Just recently, chemical cross-linking combined with mass spectrometry (XL-MS) has emerged as valuable tool to study protein interaction networks on the system-wide level. The current challenges in XL-MS are to develop robust workflows enabling a comprehensive capture of dynamic biological assemblies in their native environment in a routine manner. In this review, we will highlight both the latest technological developments as well as selected applications of XL-MS for investigating protein networks in cells, organisms, and tissue. In addition, different bioinformatics tools for data analysis will be presented. In light of these exciting new developments, XL-MS can be expected to become one of the most versatile techniques to address important biological questions in a timely manner.


Subject(s)
Protein Interaction Maps , Proteins , Computational Biology , Cross-Linking Reagents , Mass Spectrometry , Proteins/metabolism
14.
J Mass Spectrom ; 55(1): e4449, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31820512

ABSTRACT

Chemical cross-linking combined with mass spectrometry (XL-MS) and computational modeling has evolved as an alternative method to derive protein 3D structures and to map protein interaction networks. Special focus has been laid recently on the development and application of cross-linkers that are cleavable by collisional activation as they yield distinct signatures in tandem mass spectra. Building on our experiences with cross-linkers containing an MS-labile urea group, we now present the biuret-based, CID-MS/MS-cleavable cross-linker imidodicarbonyl diimidazole (IDDI) and demonstrate its applicability for protein cross-linking studies based on the four model peptides angiotensin II, MRFA, substance P, and thymopentin.


Subject(s)
Biuret/analogs & derivatives , Biuret/chemistry , Cross-Linking Reagents/chemistry , Peptides/chemistry , Angiotensin II/chemistry , Chromatography, High Pressure Liquid , Imidazoles/chemistry , Proof of Concept Study , Protein Conformation , Substance P/chemistry , Tandem Mass Spectrometry , Thymopentin/chemistry
15.
Sci Rep ; 9(1): 15058, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31636333

ABSTRACT

Calcium- and Integrin-Binding protein 2 (CIB2) is a small and ubiquitously expressed protein with largely unknown biological function but ascertained role in hearing physiology and disease. Recent studies found that CIB2 binds Ca2+ with moderate affinity and dimerizes under conditions mimicking the physiological ones. Here we provided new lines of evidence on CIB2 oligomeric state and the mechanism of interaction with the α7B integrin target. Based on a combination of native mass spectrometry, chemical cross-linking/mass spectrometry, analytical gel filtration, dynamic light scattering and molecular dynamics simulations we conclude that CIB2 is monomeric under all tested conditions and presents uncommon hydrodynamic properties, most likely due to the high content of hydrophobic solvent accessible surface. Surface plasmon resonance shows that the interaction with α7B occurs with relatively low affinity and is limited to the cytosolic region proximal to the membrane, being kinetically favored in the presence of physiological Mg2+ and in the absence of Ca2+. Although CIB2 binds to an α7B peptide in a 1:1 stoichiometry, the formation of the complex might induce binding of another CIB2 molecule.


Subject(s)
Calcium-Binding Proteins/metabolism , Hydrodynamics , Protein Multimerization , Animals , Calcium/metabolism , Calcium-Binding Proteins/chemistry , Calmodulin/metabolism , Cattle , Cell Membrane/metabolism , Dynamic Light Scattering , Humans , Integrins , Magnesium/metabolism , Models, Molecular , Molecular Weight , Peptides/metabolism , Protein Binding , Protein Stability , Protein Structure, Tertiary , Proteolysis , Structural Homology, Protein
16.
Anal Chem ; 91(15): 10236-10244, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31283178

ABSTRACT

We present a cross-linking/mass spectrometry workflow for performing proteome-wide cross-linking analyses within 1 week. The workflow is based on the commercially available mass spectrometry-cleavable cross-linker disuccinimidyl dibutyric urea and can be employed by every lab having access to a mass spectrometer with tandem mass spectrometry capabilities. We provide an updated version 2.0 of the freeware software tool MeroX, available at www.StavroX.com , that allows us to conduct fully automated and reliable studies delivering insights into protein-protein interaction networks and protein conformations at the proteome level. We exemplify our optimized workflow for mapping protein-protein interaction networks in Drosophila melanogaster embryos on a system-wide level. From cross-linked Drosophila embryo extracts, we detected 29931 cross-link spectrum matches corresponding to 7436 unique cross-linked residues in biological triplicate experiments at a 1% false discovery rate. Among these, 1611 interprotein cross-linking sites were identified and yielded valuable information about protein-protein interactions. The 5825 remaining intraprotein cross-links yield information about the conformational landscape of proteins in their cellular environment.


Subject(s)
Cross-Linking Reagents/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Proteome/analysis , Animals , Protein Interaction Mapping , Software
17.
Anal Chem ; 91(11): 6953-6961, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31045356

ABSTRACT

The number of publications in the field of chemical cross-linking combined with mass spectrometry (XL-MS) to derive constraints for protein three-dimensional structure modeling and to probe protein-protein interactions has increased during the last years. As the technique is now becoming routine for in vitro and in vivo applications in proteomics and structural biology there is a pressing need to define protocols as well as data analysis and reporting formats. Such consensus formats should become accepted in the field and be shown to lead to reproducible results. This first, community-based harmonization study on XL-MS is based on the results of 32 groups participating worldwide. The aim of this paper is to summarize the status quo of XL-MS and to compare and evaluate existing cross-linking strategies. Our study therefore builds the framework for establishing best practice guidelines to conduct cross-linking experiments, perform data analysis, and define reporting formats with the ultimate goal of assisting scientists to generate accurate and reproducible XL-MS results.


Subject(s)
Cross-Linking Reagents/chemistry , Mass Spectrometry/methods , Serum Albumin, Bovine/analysis , Serum Albumin, Bovine/chemistry , Laboratories , Mass Spectrometry/instrumentation , Reproducibility of Results
18.
J Am Soc Mass Spectrom ; 30(1): 139-148, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29679287

ABSTRACT

Cleavable cross-linkers are gaining increasing importance for chemical cross-linking/mass spectrometry (MS) as they permit a reliable and automated data analysis in structural studies of proteins and protein assemblies. Here, we introduce 1,3-diallylurea (DAU) as the first CID-MS/MS-cleavable, photo-thiol-reactive cross-linker. DAU is a commercially available, inexpensive reagent that efficiently undergoes an anti-Markovnikov hydrothiolation with cysteine residues in the presence of a radical initiator upon UV-A irradiation. Radical cysteine cross-linking proceeds via an orthogonal "click reaction" and yields stable alkyl sulfide products. DAU reacts at physiological pH and cross-linking reactions with peptides, and proteins can be performed at temperatures as low as 4 °C. The central urea bond is efficiently cleaved upon collisional activation during tandem MS experiments generating characteristic product ions. This improves the reliability of automated cross-link identification. Different radical initiators have been screened for the cross-linking reaction of DAU using the thiol-containing compounds cysteine and glutathione. Our concept has also been exemplified for the biologically relevant proteins bMunc13-2 and retinal guanylyl cyclase-activating protein-2. Graphical abstract ᅟ.


Subject(s)
Allyl Compounds/pharmacology , Cross-Linking Reagents/chemistry , Proteins/chemistry , Sulfhydryl Compounds/chemistry , Urea/analogs & derivatives , Urea/pharmacology , Cysteine/chemistry , Glutathione/chemistry , Guanylate Cyclase-Activating Proteins/chemistry , Hydrogen-Ion Concentration , Nerve Tissue Proteins/chemistry , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
19.
Mass Spectrom Rev ; 38(2): 187-201, 2019 03.
Article in English | MEDLINE | ID: mdl-29660147

ABSTRACT

Free radical-initiated peptide sequencing (FRIPS) has recently been introduced as an analytical strategy to create peptide radical ions in a predictable and effective way by collisional activation of specifically modified peptides ions. FRIPS is based on the unimolecular dissociation of open-shell ions and yields fragments that resemble those obtained by electron capture dissociation (ECD) or electron transfer dissociation (ETD). In this review article, we describe the fundamentals of FRIPS and highlight its fruitful combination with chemical cross-linking/mass spectrometry (MS) as a highly promising option to derive complementary structural information of peptides and proteins. FRIPS does not only yield exhaustive sequence information of cross-linked peptides, but also defines the exact cross-linking sites of the connected peptides. The development of more advanced FRIPS cross-linkers that extend the FRIPS-based cross-linking/MS approach to the study of large protein assemblies and protein interaction networks can be eagerly anticipated.


Subject(s)
Cross-Linking Reagents/chemistry , Free Radicals/chemistry , Peptides/chemistry , Proteins/chemistry , Sequence Analysis, Protein/methods , Tandem Mass Spectrometry/methods , Animals , Humans , Protein Conformation
20.
Chem Sci ; 9(43): 8282-8290, 2018 Nov 21.
Article in English | MEDLINE | ID: mdl-30542577

ABSTRACT

Functionality of enzymes is strongly related to water dynamic processes. The control of the redox potential for metallo-enzymes is intimately linked to the mediation of water molecules in the first and second coordination spheres. Here, we report a unique example of supramolecular control of the redox properties of a biomimetic monocopper complex by water molecules. It is shown that the copper complex based on a calix[6]arene covalently capped with a tetradentate [tris(2-methylpyridyl)amine] (tmpa) core, embedding the metal ion in a hydrophobic cavity, can exist in three different states. The first system displays a totally irreversible redox behaviour. It corresponds to the reduction of the 5-coordinate mono-aqua-CuII complex, which is the thermodynamic species in the +II state. The second system is detected at a high redox potential. It is ascribed to an "empty cavity" or "water-free" state, where the CuI ion sits in a 4-coordinate trigonal environment provided by the tmpa cap. This complex is the thermodynamic species in the +I state under "dry conditions". Surprisingly, a third redox system appears as the water concentration is increased. Under water-saturation conditions, it displays a pseudo-reversible behaviour at a low scan rate at the mid-point from the water-free and aqua species. This third system is not observed with the Cu-tmpa complex deprived of a cavity. In the calix[6]cavity environment, it is ascribed to a species where a pair of water molecules is hosted by the calixarene cavity. A molecular mechanism for the CuII/CuI redox process with an interplay of (H2O) x (x = 0, 1, 2) hosting is proposed on the basis of computational studies. Such an unusual behaviour is ascribed to the unexpected stabilization of the CuI state by inclusion of the pair of water molecules. This phenomenon strongly evidences the drastic influence of the interaction between water molecules and a hydrophobic cavity on controlling the thermodynamics and kinetics of the CuII/CuI electron transfer process.

SELECTION OF CITATIONS
SEARCH DETAIL
...