Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
JACS Au ; 3(7): 2025-2035, 2023 Jul 24.
Article in English | MEDLINE | ID: mdl-37502151

ABSTRACT

Carbene footprinting is a recently developed mass spectrometry-based chemical labeling technique that probes protein interactions and conformation. Here, we use the methodology to investigate binding interactions between the protease human Caspase-1 (C285A) and full-length human Gasdermin D (hGSDMD), which are important in inflammatory cell death. GSDMD is cleaved by Caspase-1, releasing its N-terminal domain which oligomerizes in the membrane to form large pores, resulting in lytic cell death. Regions of reduced carbene labeling (masking), caused by protein binding, were observed for each partner in the presence of the other and were consistent with hCaspase-1 exosite and active-site interactions. Most notably, the results showed direct occupancy of hCaspase-1 (C285A) active-site by hGSDMD for the first time. Differential carbene labeling of full-length hGSDMD and the pore-forming N-terminal domain assembled in liposomes showed masking of the latter, consistent with oligomeric assembly and insertion into the lipid bilayer. Interactions between Caspase-1 and the specific inhibitor VRT-043198 were also studied by this approach. In wild-type hCaspase-1, VRT-043198 modifies the active-site Cys285 through the formation of a S,O-hemiacetal. Here, we showed by carbene labeling that this inhibitor can noncovalently occupy the active site of a C285A mutant. These findings add considerably to our knowledge of the hCaspase-1-hGSDMD system.

2.
Nat Chem ; 14(12): 1375-1382, 2022 12.
Article in English | MEDLINE | ID: mdl-36357787

ABSTRACT

G-protein-coupled receptors signal through cognate G proteins. Despite the widespread importance of these receptors, their regulatory mechanisms for G-protein selectivity are not fully understood. Here we present a native mass spectrometry-based approach to interrogate both biased signalling and allosteric modulation of the ß1-adrenergic receptor in response to various ligands. By simultaneously capturing the effects of ligand binding and receptor coupling to different G proteins, we probed the relative importance of specific interactions with the receptor through systematic changes in 14 ligands, including isoprenaline derivatives, full and partial agonists, and antagonists. We observed enhanced dynamics of the intracellular loop 3 in the presence of isoprenaline, which is capable of acting as a biased agonist. We also show here that endogenous zinc ions augment the binding in receptor-Gs complexes and propose a zinc ion-binding hotspot at the TM5/TM6 intracellular interface of the receptor-Gs complex. Further interrogation led us to propose a mechanism in which zinc ions facilitate a structural transition of the intermediate complex towards the stable state.


Subject(s)
Receptors, Adrenergic, beta-2 , Receptors, G-Protein-Coupled , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/metabolism , Allosteric Regulation , Isoproterenol/pharmacology , Receptors, G-Protein-Coupled/metabolism , Ligands , GTP-Binding Proteins/metabolism , Ions , Mass Spectrometry , Zinc/metabolism
3.
mBio ; 12(5): e0178721, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34544275

ABSTRACT

Colicins are protein antibiotics deployed by Escherichia coli to eliminate competing strains. Colicins frequently exploit outer membrane (OM) nutrient transporters to penetrate the selectively permeable bacterial cell envelope. Here, by applying live-cell fluorescence imaging, we were able to monitor the entry of the pore-forming toxin colicin B (ColB) into E. coli and localize it within the periplasm. We further demonstrate that single-stranded DNA coupled to ColB can also be transported to the periplasm, emphasizing that the import routes of colicins can be exploited to carry large cargo molecules into bacteria. Moreover, we characterize the molecular mechanism of ColB association with its OM receptor FepA by applying a combination of photoactivated cross-linking, mass spectrometry, and structural modeling. We demonstrate that complex formation is coincident with large-scale conformational changes in the colicin. Thereafter, active transport of ColB through FepA involves the colicin taking the place of the N-terminal half of the plug domain that normally occludes this iron transporter. IMPORTANCE Decades of excessive use of readily available antibiotics has generated a global problem of antibiotic resistance and, hence, an urgent need for novel antibiotic solutions. Bacteriocins are protein-based antibiotics produced by bacteria to eliminate closely related competing bacterial strains. Bacteriocin toxins have evolved to bypass the complex cell envelope in order to kill bacterial cells. Here, we uncover the cellular penetration mechanism of a well-known but poorly understood bacteriocin called colicin B that is active against Escherichia coli. Moreover, we demonstrate that the colicin B-import pathway can be exploited to deliver conjugated DNA cargo into bacterial cells. Our work leads to a better understanding of the way bacteriocins, as potential alternative antibiotics, execute their mode of action as well as highlighting how they might even be exploited in the genomic manipulation of Gram-negative bacteria.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Biological Transport/drug effects , Carrier Proteins/metabolism , Colicins/pharmacology , DNA/metabolism , Iron/metabolism , Receptors, Cell Surface/metabolism , Anti-Bacterial Agents/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacteriocins/genetics , Carrier Proteins/genetics , Cell Membrane/metabolism , Colicins/chemistry , Colicins/genetics , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , Models, Molecular , Periplasm/metabolism , Periplasmic Proteins/metabolism , Protein Conformation , Protein Transport , Receptors, Cell Surface/genetics
4.
JACS Au ; 1(12): 2385-2393, 2021 Dec 27.
Article in English | MEDLINE | ID: mdl-34977906

ABSTRACT

In solution, the charge of a protein is intricately linked to its stability, but electrospray ionization distorts this connection, potentially limiting the ability of native mass spectrometry to inform about protein structure and dynamics. How the behavior of intact proteins in the gas phase depends on the presence and distribution of ionizable surface residues has been difficult to answer because multiple chargeable sites are present in virtually all proteins. Turning to protein engineering, we show that ionizable side chains are completely dispensable for charging under native conditions, but if present, they are preferential protonation sites. The absence of ionizable side chains results in identical charge state distributions under native-like and denaturing conditions, while coexisting conformers can be distinguished using ion mobility separation. An excess of ionizable side chains, on the other hand, effectively modulates protein ion stability. In fact, moving a single ionizable group can dramatically alter the gas-phase conformation of a protein ion. We conclude that although the sum of the charges is governed solely by Coulombic terms, their locations affect the stability of the protein in the gas phase.

5.
J Biol Chem ; 295(27): 9147-9156, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32398259

ABSTRACT

Colicins are Escherichia coli-specific bacteriocins that translocate across the outer bacterial membrane by a poorly understood mechanism. Group A colicins typically parasitize the proton-motive force-linked Tol system in the inner membrane via porins after first binding an outer membrane protein receptor. Recent studies have suggested that the pore-forming group A colicin N (ColN) instead uses lipopolysaccharide as a receptor. Contrary to this prevailing view, using diffusion-precipitation assays, native state MS, isothermal titration calorimetry, single-channel conductance measurements in planar lipid bilayers, and in vivo fluorescence imaging, we demonstrate here that ColN uses OmpF both as its receptor and translocator. This dual function is achieved by ColN having multiple distinct OmpF-binding sites, one located within its central globular domain and another within its disordered N terminus. We observed that the ColN globular domain associates with the extracellular surface of OmpF and that lipopolysaccharide (LPS) enhances this binding. Approximately 90 amino acids of ColN then translocate through the porin, enabling the ColN N terminus to localize within the lumen of an OmpF subunit from the periplasmic side of the membrane, a binding mode reminiscent of that observed for the nuclease colicin E9. We conclude that bifurcated engagement of porins is intrinsic to the import mechanism of group A colicins.


Subject(s)
Colicins/metabolism , Porins/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacteriocins/metabolism , Binding Sites/physiology , Diffusion , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Lipid Bilayers/metabolism , Lipopolysaccharides/metabolism , Membrane Transport Proteins/metabolism , Models, Molecular , Porins/genetics , Protein Binding/physiology , Protein Conformation , Protein Transport , Receptors, Cell Surface/metabolism
6.
Angew Chem Int Ed Engl ; 59(9): 3523-3528, 2020 02 24.
Article in English | MEDLINE | ID: mdl-31886601

ABSTRACT

Membrane proteins engage in a variety of contacts with their surrounding lipids, but distinguishing between specifically bound lipids, and non-specific, annular interactions is a challenging problem. Applying native mass spectrometry to three membrane protein complexes with different lipid-binding properties, we explore the ability of detergents to compete with lipids bound in different environments. We show that lipids in annular positions on the presenilin homologue protease are subject to constant exchange with detergent. By contrast, detergent-resistant lipids bound at the dimer interface in the leucine transporter show decreased koff rates in molecular dynamics simulations. Turning to the lipid flippase MurJ, we find that addition of the natural substrate lipid-II results in the formation of a 1:1 protein-lipid complex, where the lipid cannot be displaced by detergent from the highly protected active site. In summary, we distinguish annular from non-annular lipids based on their exchange rates in solution.


Subject(s)
Lipids/chemistry , Mass Spectrometry , Membrane Proteins/chemistry , Cardiolipins/chemistry , Cardiolipins/metabolism , Detergents/chemistry , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Proteins/metabolism , Methanomicrobiaceae/metabolism , Molecular Dynamics Simulation , Presenilins/chemistry , Presenilins/metabolism , Protein Binding
7.
Proc Natl Acad Sci U S A ; 115(26): 6691-6696, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29891712

ABSTRACT

Strong interactions between lipids and proteins occur primarily through association of charged headgroups and amino acid side chains, rendering the protonation status of both partners important. Here we use native mass spectrometry to explore lipid binding as a function of charge of the outer membrane porin F (OmpF). We find that binding of anionic phosphatidylglycerol (POPG) or zwitterionic phosphatidylcholine (POPC) to OmpF is sensitive to electrospray polarity while the effects of charge are less pronounced for other proteins in outer or mitochondrial membranes: the ferripyoverdine receptor (FpvA) or the voltage-dependent anion channel (VDAC). Only marginal charge-induced differences were observed for inner membrane proteins: the ammonia channel (AmtB) or the mechanosensitive channel. To understand these different sensitivities, we performed an extensive bioinformatics analysis of membrane protein structures and found that OmpF, and to a lesser extent FpvA and VDAC, have atypically high local densities of basic and acidic residues in their lipid headgroup-binding regions. Coarse-grained molecular dynamics simulations, in mixed lipid bilayers, further implicate changes in charge by demonstrating preferential binding of anionic POPG over zwitterionic POPC to protonated OmpF, an effect not observed to the same extent for AmtB. Moreover, electrophysiology and mass-spectrometry-based ligand-binding experiments, at low pH, show that POPG can maintain OmpF channels in open conformations for extended time periods. Since the outer membrane is composed almost entirely of anionic lipopolysaccharide, with similar headgroup properties to POPG, such anionic lipid binding could prevent closure of OmpF channels, thereby increasing access of antibiotics that use porin-mediated pathways.


Subject(s)
Phosphatidylcholines/metabolism , Phosphatidylglycerols/metabolism , Porins/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Cation Transport Proteins/chemistry , Cation Transport Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Hydrogen-Ion Concentration , Models, Chemical , Models, Molecular , Molecular Dynamics Simulation , Porins/chemistry , Protein Binding , Protein Conformation , Spectrometry, Mass, Electrospray Ionization , Voltage-Dependent Anion Channels/chemistry , Voltage-Dependent Anion Channels/metabolism , Voltage-Gated Sodium Channels/chemistry , Voltage-Gated Sodium Channels/metabolism
8.
Nat Protoc ; 13(5): 1106-1120, 2018 05.
Article in English | MEDLINE | ID: mdl-29700483

ABSTRACT

With the recent success in determining membrane protein structures, further detailed understanding of the identity and function of the bound lipidome is essential. Using an approach that combines high-energy native mass spectrometry (HE-nMS) and solution-phase lipid profiling, this protocol can be used to determine the identity of the endogenous lipids that directly interact with a protein. Furthermore, this method can identify systems in which such lipid binding has a major role in regulating the oligomeric assembly of membrane proteins. The protocol begins with recording of the native mass spectrum of the protein of interest, under successive delipidation conditions, to determine whether delipidation leads to disruption of the oligomeric state. Subsequently, we propose using a bipronged strategy: first, an HE-nMS platform is used that allows dissociation of the detergent micelle at the front end of the instrument. This allows for isolation of the protein-lipid complex at the quadrupole and successive fragmentation at the collision cell, which leads to identification of the bound lipid masses. Next, simultaneous coupling of this with in-solution LC-MS/MS-based identification of extracted lipids reveals the complete identity of the interacting lipidome that copurifies with the proteins. Assimilation of the results of these two sets of experiments divulges the complete identity of the set of lipids that directly interact with the membrane protein of interest, and can further delineate its role in maintaining the oligomeric state of the protein. The entire procedure takes 2 d to complete.


Subject(s)
Mass Spectrometry/methods , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Protein Binding , Protein Multimerization
9.
Nat Struct Mol Biol ; 25(3): 279-288, 2018 03.
Article in English | MEDLINE | ID: mdl-29434345

ABSTRACT

Cotranslational protein folding can facilitate rapid formation of functional structures. However, it can also cause premature assembly of protein complexes, if two interacting nascent chains are in close proximity. By analyzing known protein structures, we show that homomeric protein contacts are enriched toward the C termini of polypeptide chains across diverse proteomes. We hypothesize that this is the result of evolutionary constraints for folding to occur before assembly. Using high-throughput imaging of protein homomers in Escherichia coli and engineered protein constructs with N- and C-terminal oligomerization domains, we show that, indeed, proteins with C-terminal homomeric interface residues consistently assemble more efficiently than those with N-terminal interface residues. Using in vivo, in vitro and in silico experiments, we identify features that govern successful assembly of homomers, which have implications for protein design and expression optimization.


Subject(s)
Multiprotein Complexes/chemistry , Protein Biosynthesis , Protein Multimerization , Protein Subunits/biosynthesis , Evolution, Molecular , Models, Molecular , Molecular Chaperones/metabolism , Protein Domains , Protein Engineering , Protein Folding , Protein Subunits/chemistry , RNA, Messenger/metabolism , Ribosomes/metabolism , Solubility
10.
Angew Chem Int Ed Engl ; 56(46): 14463-14468, 2017 11 13.
Article in English | MEDLINE | ID: mdl-28884954

ABSTRACT

Mass spectrometry (MS) applications for intact protein complexes typically require electrospray (ES) ionization and have not been achieved via direct desorption from surfaces. Desorption ES ionization (DESI) MS has however transformed the study of tissue surfaces through release and characterisation of small molecules. Motivated by the desire to screen for ligand binding to intact protein complexes we report the development of a native DESI platform. By establishing conditions that preserve non-covalent interactions we exploit the surface to capture a rapid turnover enzyme-substrate complex and to optimise detergents for membrane protein study. We demonstrate binding of lipids and drugs to membrane proteins deposited on surfaces and selectivity from a mix of related agonists for specific binding to a GPCR. Overall therefore we introduce this native DESI platform with the potential for high-throughput ligand screening of some of the most challenging drug targets including GPCRs.


Subject(s)
Membrane Proteins/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Ligands , Lipid Metabolism , Membrane Proteins/metabolism , Molecular Weight , Pharmaceutical Preparations/metabolism , Protein Binding , Solubility , Surface Properties
11.
Angew Chem Int Ed Engl ; 56(47): 14873-14877, 2017 11 20.
Article in English | MEDLINE | ID: mdl-28960650

ABSTRACT

Mapping the interaction sites between membrane-spanning proteins is a key challenge in structural biology. In this study a carbene-footprinting approach was developed and applied to identify the interfacial sites of a trimeric, integral membrane protein, OmpF, solubilised in micelles. The diazirine-based footprinting probe is effectively sequestered by, and incorporated into, the micelles, thus leading to efficient labelling of the membrane-spanning regions of the protein upon irradiation at 349 nm. Areas associated with protein-protein interactions between the trimer subunits remained unlabelled, thus revealing their location.


Subject(s)
Membrane Proteins/chemistry , Methane/analogs & derivatives , Amino Acid Sequence , Binding Sites , Chromatography, Liquid , Detergents/chemistry , Diazomethane/chemistry , Methane/chemistry , Micelles , Oxidation-Reduction , Protein Multimerization , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
12.
Sci Adv ; 3(6): e1701016, 2017 06.
Article in English | MEDLINE | ID: mdl-28630934

ABSTRACT

G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors belong to the largest family of membrane-embedded cell surface proteins and are involved in a diverse array of physiological processes. Despite progress in the mass spectrometry of membrane protein complexes, G protein-coupled receptors have remained intractable because of their low yield and instability after extraction from cell membranes. We established conditions in the mass spectrometer that preserve noncovalent ligand binding to the human purinergic receptor P2Y1. Results established differing affinities for nucleotides and the drug MRS2500 and link antagonist binding with the absence of receptor phosphorylation. Overall, therefore, our results are consistent with drug binding, preventing the conformational changes that facilitate downstream signaling. More generally, we highlight opportunities for mass spectrometry to probe effects of ligand binding on G protein-coupled receptors.


Subject(s)
Ligands , Mass Spectrometry , Receptors, G-Protein-Coupled/chemistry , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Models, Molecular , Molecular Conformation , Phosphorylation , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Receptors, Purinergic P2Y1/chemistry , Receptors, Purinergic P2Y1/metabolism , Structure-Activity Relationship
13.
Structure ; 25(5): 773-782.e5, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28434916

ABSTRACT

Select lectins have powerful anti-viral properties that effectively neutralize HIV-1 by targeting the dense glycan shield on the virus. Here, we reveal the mechanism by which one of the most potent lectins, BanLec, achieves its inhibition. We identify that BanLec recognizes a subset of high-mannose glycans via bidentate interactions spanning the two binding sites present on each BanLec monomer that were previously considered separate carbohydrate recognition domains. We show that both sites are required for high-affinity glycan binding and virus neutralization. Unexpectedly we find that BanLec adopts a tetrameric stoichiometry in solution whereby the glycan-binding sites are positioned to optimally target glycosylated viral spikes. The tetrameric architecture, together with bidentate binding to individual glycans, leads to layers of multivalency that drive viral neutralization through enhanced avidity effects. These structural insights will prove useful in engineering successful lectin therapeutics targeting the dense glycan shield of HIV.


Subject(s)
Antiviral Agents/chemistry , Plant Lectins/chemistry , Polysaccharides/metabolism , Antiviral Agents/pharmacology , Binding Sites , HIV-1/chemistry , HIV-1/drug effects , Musa/chemistry , Plant Lectins/metabolism , Plant Lectins/pharmacology , Polysaccharides/chemistry , Protein Binding , Protein Multimerization
14.
Nature ; 541(7637): 421-424, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28077870

ABSTRACT

Oligomerization of membrane proteins in response to lipid binding has a critical role in many cell-signalling pathways but is often difficult to define or predict. Here we report the development of a mass spectrometry platform to determine simultaneously the presence of interfacial lipids and oligomeric stability and to uncover how lipids act as key regulators of membrane-protein association. Evaluation of oligomeric strength for a dataset of 125 α-helical oligomeric membrane proteins reveals an absence of interfacial lipids in the mass spectra of 12 membrane proteins with high oligomeric stability. For the bacterial homologue of the eukaryotic biogenic transporters (LeuT, one of the proteins with the lowest oligomeric stability), we found a precise cohort of lipids within the dimer interface. Delipidation, mutation of lipid-binding sites or expression in cardiolipin-deficient Escherichia coli abrogated dimer formation. Molecular dynamics simulation revealed that cardiolipin acts as a bidentate ligand, bridging across subunits. Subsequently, we show that for the Vibrio splendidus sugar transporter SemiSWEET, another protein with low oligomeric stability, cardiolipin shifts the equilibrium from monomer to functional dimer. We hypothesized that lipids are essential for dimerization of the Na+/H+ antiporter NhaA from E. coli, which has the lowest oligomeric strength, but not for the substantially more stable homologous Thermus thermophilus protein NapA. We found that lipid binding is obligatory for dimerization of NhaA, whereas NapA has adapted to form an interface that is stable without lipids. Overall, by correlating interfacial strength with the presence of interfacial lipids, we provide a rationale for understanding the role of lipids in both transient and stable interactions within a range of α-helical membrane proteins, including G-protein-coupled receptors.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Lipids/chemistry , Lipids/pharmacology , Mass Spectrometry/methods , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Protein Multimerization/drug effects , Binding Sites/genetics , Cardiolipins/chemistry , Cardiolipins/metabolism , Cardiolipins/pharmacology , Cell Membrane/chemistry , Cell Membrane/metabolism , Escherichia coli/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Ligands , Models, Molecular , Molecular Dynamics Simulation , Moritella/chemistry , Protein Stability/drug effects , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/metabolism , Thermodynamics , Thermus thermophilus/chemistry
15.
Nat Commun ; 7: 12194, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27432510

ABSTRACT

Redox-regulated effector systems that counteract oxidative stress are essential for all forms of life. Here we uncover a new paradigm for sensing oxidative stress centred on the hydrophobic core of a sensor protein. RsrA is an archetypal zinc-binding anti-sigma factor that responds to disulfide stress in the cytoplasm of Actinobacteria. We show that RsrA utilizes its hydrophobic core to bind the sigma factor σ(R) preventing its association with RNA polymerase, and that zinc plays a central role in maintaining this high-affinity complex. Oxidation of RsrA is limited by the rate of zinc release, which weakens the RsrA-σ(R) complex by accelerating its dissociation. The subsequent trigger disulfide, formed between specific combinations of RsrA's three zinc-binding cysteines, precipitates structural collapse to a compact state where all σ(R)-binding residues are sequestered back into its hydrophobic core, releasing σ(R) to activate transcription of anti-oxidant genes.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Hydrophobic and Hydrophilic Interactions , Oxidative Stress , Sigma Factor/antagonists & inhibitors , Amino Acid Sequence , Cysteine/metabolism , Kinetics , Magnetic Resonance Spectroscopy , Oxidation-Reduction , Zinc/metabolism
16.
Nat Commun ; 7: 11578, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27174498

ABSTRACT

The prolactin receptor is an archetype member of the class I cytokine receptor family, comprising receptors with fundamental functions in biology as well as key drug targets. Structurally, each of these receptors represent an intriguing diversity, providing an exceptionally challenging target for structural biology. Here, we access the molecular architecture of the monomeric human prolactin receptor by combining experimental and computational efforts. We solve the NMR structure of its transmembrane domain in micelles and collect structural data on overlapping fragments of the receptor with small-angle X-ray scattering, native mass spectrometry and NMR spectroscopy. Along with previously published data, these are integrated by molecular modelling to generate a full receptor structure. The result provides the first full view of a class I cytokine receptor, exemplifying the architecture of more than 40 different receptor chains, and reveals that the extracellular domain is merely the tip of a molecular iceberg.


Subject(s)
Crystallography, X-Ray/methods , Models, Molecular , Receptors, Prolactin/chemistry , Humans , Magnetic Resonance Spectroscopy/methods , Micelles , Protein Conformation, alpha-Helical , Protein Domains , Receptors, Prolactin/isolation & purification , Scattering, Small Angle
17.
J Am Soc Mass Spectrom ; 27(6): 1099-104, 2016 06.
Article in English | MEDLINE | ID: mdl-27106602

ABSTRACT

Membrane protein complexes are commonly introduced to the mass spectrometer solubilized in detergent micelles. The collisional activation used to remove the detergent, however, often causes protein unfolding and dissociation. As in the case for soluble proteins, electrospray in the positive ion mode is most commonly used for the study of membrane proteins. Here we show several distinct advantages of employing the negative ion mode. Negative polarity can yield lower average charge states for membrane proteins solubilized in saccharide detergents, with enhanced peak resolution and reduced adduct formation. Most importantly, we demonstrate that negative ion mode electrospray ionization (ESI) minimizes subunit dissociation in the gas phase, allowing access to biologically relevant oligomeric states. Together, these properties mean that intact membrane protein ions can be generated in a greater range of solubilizing detergents. The formation of negative ions, therefore, greatly expands the possibilities of using mass spectrometry on this intractable class of protein. Graphical Abstract ᅟ.


Subject(s)
Ions , Membrane Proteins/chemistry , Spectrometry, Mass, Electrospray Ionization , Detergents/chemistry , Micelles
18.
BMC Biol ; 14: 31, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27083547

ABSTRACT

BACKGROUND: Extracellular signal-regulated kinase 2 (ERK2) is an S/T kinase with more than 200 known substrates, and with critical roles in regulation of cell growth and differentiation and currently no membrane proteins have been linked to ERK2 scaffolding. METHODS AND RESULTS: Here, we identify the human Na(+)/H(+) exchanger 1 (hNHE1) as a membrane scaffold protein for ERK2 and show direct hNHE1-ERK1/2 interaction in cellular contexts. Using nuclear magnetic resonance (NMR) spectroscopy and immunofluorescence analysis we demonstrate that ERK2 scaffolding by hNHE1 occurs by one of three D-domains and by two non-canonical F-sites located in the disordered intracellular tail of hNHE1, mutation of which reduced cellular hNHE1-ERK1/2 co-localization, as well as reduced cellular ERK1/2 activation. Time-resolved NMR spectroscopy revealed that ERK2 phosphorylated the disordered tail of hNHE1 at six sites in vitro, in a distinct temporal order, with the phosphorylation rates at the individual sites being modulated by the docking sites in a distant dependent manner. CONCLUSIONS: This work characterizes a new type of scaffolding complex, which we term a "shuffle complex", between the disordered hNHE1-tail and ERK2, and provides a molecular mechanism for the important ERK2 scaffolding function of the membrane protein hNHE1, which regulates the phosphorylation of both hNHE1 and ERK2.


Subject(s)
Cation Transport Proteins/metabolism , Intrinsically Disordered Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Sequence , Cation Transport Proteins/chemistry , Cell Line , Enzyme Activation , Humans , Intrinsically Disordered Proteins/chemistry , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 3/chemistry , Mitogen-Activated Protein Kinase 3/metabolism , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Folding , Protein Interaction Maps , Protein Structure, Tertiary , Sodium-Hydrogen Exchanger 1 , Sodium-Hydrogen Exchangers/chemistry
19.
Nat Methods ; 13(4): 333-6, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26901650

ABSTRACT

Small molecules are known to stabilize membrane proteins and to modulate their function and oligomeric state, but such interactions are often hard to precisely define. Here we develop and apply a high-resolution, Orbitrap mass spectrometry-based method for analyzing intact membrane protein-ligand complexes. Using this platform, we resolve the complexity of multiple binding events, quantify small molecule binding and reveal selectivity for endogenous lipids that differ only in acyl chain length.


Subject(s)
Lipids/chemistry , Mass Spectrometry/methods , Membrane Proteins/metabolism , Peptide Fragments/metabolism , Small Molecule Libraries/metabolism , Humans , Models, Molecular , Protein Binding
20.
Chem Commun (Camb) ; 51(85): 15582-4, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26356172

ABSTRACT

Mass spectrometry of intact membrane protein complexes requires removal of the detergent micelle by collisional activation. We demonstrate that the necessary energy can be obtained by adjusting the degree of collisional cooling in the ion source. This enables us to extend the energy regime for dissociation of membrane protein complexes.


Subject(s)
Membrane Proteins/chemistry , Thermodynamics , Gases/chemistry , Mass Spectrometry , Protein Unfolding
SELECTION OF CITATIONS
SEARCH DETAIL
...