Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Chemphyschem ; 23(13): e202200127, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35499980

ABSTRACT

Solid-state NMR (ssNMR) spectroscopy has emerged as the method of choice to analyze the structural dynamics of fibrillar, membrane-bound, and crystalline proteins that are recalcitrant to other structural techniques. Recently, 1 H detection under fast magic angle spinning and multiple acquisition ssNMR techniques have propelled the structural analysis of complex biomacromolecules. However, data acquisition and resonance-specific assignments remain a bottleneck for this technique. Here, we present a comprehensive multi-acquisition experiment (PHRONESIS) that simultaneously generates up to ten 3D 1 H-detected ssNMR spectra. PHRONESIS utilizes broadband transfer and selective pulses to drive multiple independent polarization pathways. High selectivity excitation and de-excitation of specific resonances were achieved by high-fidelity selective pulses that were designed using a combination of an evolutionary algorithm and artificial intelligence. We demonstrated the power of this approach with microcrystalline U-13 C,15 N GB1 protein, reaching 100 % of the resonance assignments using one data set of ten 3D experiments. The strategy outlined in this work opens up new avenues for implementing novel 1 H-detected multi-acquisition ssNMR experiments to speed up and expand the application to larger biomolecular systems.


Subject(s)
Artificial Intelligence , Proteins , Algorithms , Magnetic Resonance Spectroscopy/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Proteins/chemistry
2.
Structure ; 30(3): 360-370.e6, 2022 03 03.
Article in English | MEDLINE | ID: mdl-34875216

ABSTRACT

SERCA is a P-type ATPase embedded in the sarcoplasmic reticulum and plays a central role in muscle relaxation. SERCA's function is regulated by single-pass membrane proteins called regulins. Unlike other regulins, dwarf open reading frame (DWORF) expressed in cardiac muscle has a unique activating effect. Here, we determine the structure and topology of DWORF in lipid bilayers using a combination of oriented sample solid-state NMR spectroscopy and replica-averaged orientationally restrained molecular dynamics. We found that DWORF's structural topology consists of a dynamic N-terminal domain, an amphipathic juxtamembrane helix that crosses the lipid groups at an angle of 64°, and a transmembrane C-terminal helix with an angle of 32°. A kink induced by Pro15, unique to DWORF, separates the two helical domains. A single Pro15Ala mutant significantly decreases the kink and eliminates DWORF's activating effect on SERCA. Overall, our findings directly link DWORF's structural topology to its activating effect on SERCA.


Subject(s)
Calcium-Binding Proteins , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Lipid Bilayers/metabolism , Molecular Dynamics Simulation , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
3.
Sci Adv ; 7(48): eabi7154, 2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34826239

ABSTRACT

The sarcoplasmic reticulum (SR) Ca2+-ATPase (SERCA) plays a central role in muscle contractility and nonshivering thermogenesis. SERCA is regulated by sarcolipin (SLN), a single-pass membrane protein that uncouples Ca2+ transport from ATP hydrolysis, promoting futile enzymatic cycles and heat generation. The molecular determinants for regulating heat release by the SERCA/SLN complex are unclear. Using thermocalorimetry, chemical cross-linking, and solid-state NMR spectroscopy in oriented phospholipid bicelles, we show that SERCA's functional uncoupling and heat release rate are dictated by specific SERCA/SLN intramembrane interactions, with the carboxyl-terminal residues anchoring SLN to the SR membrane in an inhibitory topology. Systematic deletion of the carboxyl terminus does not prevent the SERCA/SLN complex formation but reduces uncoupling in a graded manner. These studies emphasize the critical role of lipids in defining the active topology of SLN and modulating the heat release rate by the SERCA/SLN complex, with implications in fat metabolism and basal metabolic rate.

4.
Elife ; 102021 05 12.
Article in English | MEDLINE | ID: mdl-33978571

ABSTRACT

Phospholamban (PLN) is a mini-membrane protein that directly controls the cardiac Ca2+-transport response to ß-adrenergic stimulation, thus modulating cardiac output during the fight-or-flight response. In the sarcoplasmic reticulum membrane, PLN binds to the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA), keeping this enzyme's function within a narrow physiological window. PLN phosphorylation by cAMP-dependent protein kinase A or increase in Ca2+ concentration reverses the inhibitory effects through an unknown mechanism. Using oriented-sample solid-state NMR spectroscopy and replica-averaged NMR-restrained structural refinement, we reveal that phosphorylation of PLN's cytoplasmic regulatory domain signals the disruption of several inhibitory contacts at the transmembrane binding interface of the SERCA-PLN complex that are propagated to the enzyme's active site, augmenting Ca2+ transport. Our findings address long-standing questions about SERCA regulation, epitomizing a signal transduction mechanism operated by posttranslationally modified bitopic membrane proteins.


Subject(s)
Allosteric Regulation , Calcium-Binding Proteins/chemistry , Phosphorylation , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Animals , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Escherichia coli , Magnetic Resonance Spectroscopy , Membrane Proteins/metabolism , Molecular Structure , Protein Conformation , Rabbits , Sarcoplasmic Reticulum , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction
5.
Acc Chem Res ; 54(6): 1430-1439, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33655754

ABSTRACT

Membrane proteins mediate a plethora of cellular functions and represent important targets for drug development. Unlike soluble proteins, membrane proteins require native-like environments to fold correctly and be active. Therefore, modern structural biology techniques have aimed to determine the structure and dynamics of these membrane proteins at physiological temperature and in liquid crystalline lipid bilayers. With the flourishing of new NMR methodologies and improvements in sample preparations, magic angle spinning (MAS) and oriented sample solid-state NMR (OS-ssNMR) spectroscopy of membrane proteins is experiencing a new renaissance. Born as antagonistic approaches, these techniques nowadays offer complementary information on the structural topology and dynamics of membrane proteins reconstituted in lipid membranes. By spinning biosolid samples at the magic angle (θ = 54.7°), MAS NMR experiments remove the intrinsic anisotropy of the NMR interactions, increasing spectral resolution. Internuclear spin interactions (spin exchange) are reintroduced by RF pulses, providing distances and torsion angles to determine secondary, tertiary, and quaternary structures of membrane proteins. OS-ssNMR, on the other hand, directly detects anisotropic NMR parameters such as dipolar couplings (DC) and anisotropic chemical shifts (CS), providing orientational constraints to determine the architecture (i.e., topology) of membrane proteins relative to the lipid membrane. Defining the orientation of membrane proteins and their interactions with lipid membranes is of paramount importance since lipid-protein interactions can shape membrane protein conformations and ultimately define their functional states.In this Account, we report selected studies from our group integrating MAS and OS-ssNMR techniques to give a comprehensive view of the biological processes occurring at cellular membranes. We focus on the main experiments for both techniques, with an emphasis on new implementation to increase both sensitivity and spectral resolution. We also describe how the structural constraints derived from both isotropic and anisotropic NMR parameters are integrated into dynamic structural modeling using replica-averaged orientational-restrained molecular dynamics simulations (RAOR-MD). We showcase small membrane proteins that are involved in Ca2+ transport and regulate cardiac and skeletal muscle contractility: phospholamban (PLN, 6 kDa), sarcolipin (SLN, 4 kDa), and DWORF (4 kDa). We summarize our results for the structures of these polypeptides free and in complex with the sarcoplasmic reticulum (SR) Ca2+-ATPase (SERCA, 110 kDa). Additionally, we illustrate the progress toward the determination of the structural topology of a six transmembrane protein associated with succinate and acetate transport (SatP, hexamer 120 kDa). From these examples, the integrated MAS and OS-ssNMR approach, in combination with modern computational methods, emerges as a way to overcome the challenges posed by studying large membrane protein systems.


Subject(s)
Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular , Lipid Bilayers/metabolism , Membrane Proteins/metabolism , Molecular Dynamics Simulation
6.
J Am Chem Soc ; 142(21): 9686-9699, 2020 05 27.
Article in English | MEDLINE | ID: mdl-32383602

ABSTRACT

Alpha synuclein (αS) oligomers are a key component of Lewy bodies implicated in Parkinson's disease (PD). Although primarily intracellular, extracellular αS exocytosed from neurons also contributes to PD pathogenesis through a prion-like transmission mechanism. Here, we show at progressive degrees of resolution that the most abundantly expressed extracellular protein, human serum albumin (HSA), inhibits αS oligomer (αSn) toxicity through a three-pronged mechanism. First, endogenous HSA targets αSn with sub-µM affinity via solvent-exposed hydrophobic sites, breaking the catalytic cycle that promotes αS self-association. Second, HSA remodels αS oligomers and high-MW fibrils into chimeric intermediates with reduced toxicity. Third, HSA unexpectedly suppresses membrane interactions with the N-terminal and central αS regions. Overall, our findings suggest that the extracellular proteostasis network may regulate αS cell-to-cell transmission not only by reducing the populations of membrane-binding competent αS oligomers but possibly also by shielding the membrane interface from residual toxic species.


Subject(s)
Molecular Chaperones/metabolism , Serum Albumin, Human/metabolism , alpha-Synuclein/metabolism , Cell Line, Tumor , Cell Survival , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Chaperones/chemistry , Serum Albumin, Human/chemistry , alpha-Synuclein/chemistry
7.
Biochim Biophys Acta Biomembr ; 1862(1): 183034, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31400305

ABSTRACT

Hematopoietic-substrate-1 associated protein X-1 (HAX-1) is a 279 amino acid protein expressed ubiquitously. In cardiac muscle, HAX-1 was found to modulate the sarcoendoplasmic reticulum calcium ATPase (SERCA) by shifting its apparent Ca2+ affinity (pCa). It has been hypothesized that HAX-1 binds phospholamban (PLN), enhancing its inhibitory function on SERCA. HAX-1 effects are reversed by cAMP-dependent protein kinase A that phosphorylates PLN at Ser16. To date, the molecular mechanisms for HAX-1 regulation of the SERCA/PLN complex are still unknown. Using enzymatic, in cell assays, circular dichroism, and NMR spectroscopy, we found that in the absence of a binding partner HAX-1 is essentially disordered and adopts a partial secondary structure upon interaction with lipid membranes. Also, HAX-1 interacts with the cytoplasmic region of monomeric and pentameric PLN as detected by NMR and in cell FRET assays, respectively. We propose that the regulation of the SERCA/PLN complex by HAX-1 is mediated by its interactions with lipid membranes, adding another layer of control in Ca2+ homeostatic balance in the heart muscle.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Calcium-Binding Proteins/metabolism , Calcium/metabolism , Cytoplasm/metabolism , Membrane Lipids/metabolism , Myocardium/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Animals , Calcium-Binding Proteins/ultrastructure , Humans , Intrinsically Disordered Proteins , Magnetic Resonance Spectroscopy , Protein Structure, Secondary , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
8.
Sci Rep ; 9(1): 8225, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31160739

ABSTRACT

2D homonuclear NMR spectroscopy is an essential technique to characterize small and large molecules, such as organic compounds, metabolites, and biomacromolecules at atomic resolution. However, for complex samples 2D homonuclear spectra display poor resolution, making spectral assignment very cumbersome. Here, we propose a new method that exploits the differential T2* relaxation times of individual resonances and resolves the 2D NMR peaks into pseudo-3D spectra, where time is the 3rd dimension. T2* weIghted DEconvolution or TIDE analyzes individual free induction decays (FIDs) and dissects them into sub-FIDs that are transformed into pseudo-3D spectra combining Fourier transformation and covariance NMR. TIDE achieves higher resolution and sensitivity for NMR spectra than classical covariance NMR reducing offset-dependent artifacts. We demonstrate the performance of TIDE for magic angle spinning (MAS) [13C,13C]-DARR NMR spectra of single- and multi-span membrane proteins embedded in lipid bilayers. Since TIDE is applicable to all type of homonuclear correlation experiments for liquid and solid samples, we anticipate that it will be a general method for processing NMR data of biomacromolecules, complex mixtures of metabolites as well as material samples.


Subject(s)
Algorithms , Magnetic Resonance Spectroscopy , Membrane Proteins/chemistry , Muscle Proteins/chemistry , Proteolipids/chemistry
9.
Sci Signal ; 12(580)2019 05 07.
Article in English | MEDLINE | ID: mdl-31064885

ABSTRACT

Bacterial adhesins mediate adhesion to substrates and biofilm formation. Adhesins of the LPXTG family are posttranslationally processed by the cell membrane-localized peptidase sortase A, which cleaves the LPXTG motif. This generates a short C-terminal peptide (C-pep) that remains in the cell membrane, whereas the mature adhesin is incorporated into the cell wall. Genes encoding adhesins of the oral bacterium Streptococcus gordonii were differentially expressed depending on whether the bacteria were isolated from saliva or dental plaque and appeared to be coordinately regulated. Deletion of sspA and sspB (sspAB), both of which encode LPXTG-containing adhesins, unexpectedly enhanced adhesion and biofilm formation. C-peps produced from a model LPXTG-containing adhesin localized to the cell membrane and bound to and inhibited the intramembrane sensor histidine kinase SGO_1180, thus preventing activation of the cognate response regulator SGO_1181. The absence of SspAB C-peps induced the expression of the scaCBA operon encoding the lipoprotein adhesin ScaA, which was sufficient to preserve and even enhance biofilm formation. This C-pep-driven regulatory circuit also exists in pathogenic streptococci and is likely conserved among Gram-positive bacteria. This quality control mechanism ensures that the bacteria can form biofilms under diverse environmental conditions and may play a role in optimizing adhesion and biofilm formation.


Subject(s)
Adhesins, Bacterial/metabolism , Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Membrane Glycoproteins/metabolism , Streptococcus gordonii/metabolism , Adhesins, Bacterial/genetics , Amino Acid Motifs/genetics , Amino Acid Sequence , Aminoacyltransferases/genetics , Bacterial Proteins/genetics , Biofilms , Cysteine Endopeptidases/genetics , Dental Plaque/microbiology , Gene Expression Regulation, Bacterial , Mutation , Peptide Fragments/genetics , Peptide Fragments/metabolism , Saliva/microbiology , Sequence Homology, Amino Acid , Streptococcus gordonii/genetics , Streptococcus gordonii/physiology
10.
Methods Enzymol ; 621: 281-304, 2019.
Article in English | MEDLINE | ID: mdl-31128784

ABSTRACT

Solid-state NMR (ssNMR) is an ideal tool to study structure and dynamics of membrane proteins in their native lipid environment. In principle, ssNMR has no size limitations. However, this feature is rarely exploited as large membrane proteins display severe resonance overlap. In addition, dismal yields from recombinant bacterial expression systems limit severely spectroscopic characterization of membrane proteins. For very large mammalian membrane proteins, extraction from the original organism remains the most viable approach. In this case, NMR-observable nuclei must be introduced post-translationally, but the approaches developed so far are rather scarce. Here, we detail the synthesis and engineering of a reactive 13C-ethylmethanethiosulfonate (13C-EMTS) reagent for the post-translational alkylation of cysteine sidechains of a 110kDa sarcoplasmic reticulum Ca2+-ATPase (SERCA) extracted from rabbit skeletal muscle tissue. When reconstituted into liposomes, it is possible to resolve the resonances of the engineered ethyl groups by magic-angle spinning (MAS) 2D [13C,13C]-DARR experiments. Notably, the ethyl-group modification does not perturb the function of SERCA, yielding well-resolved 13C-13C fingerprints that are used to image its structural states in the catalytic cycle and filtering out overwhelming naturally-abundant 13C nuclei signals arising from the enzyme and lipids. We anticipate that this approach will be used together with 19F NMR to monitor conformational transitions of enzymes and proteins that are difficult to express recombinantly.


Subject(s)
Cysteine/analysis , Membrane Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Animals , Calcium-Binding Proteins/chemistry , Humans , Isotope Labeling/methods , Models, Molecular , Muscle Proteins/chemistry , Protein Conformation , Proteolipids/chemistry , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry
11.
Anal Chem ; 91(12): 7698-7704, 2019 Jun 18.
Article in English | MEDLINE | ID: mdl-31120239

ABSTRACT

The Henderson equation is usually used to calculate liquid-junction potentials between miscible electrolyte solutions. However, the potentials of reference electrodes that comprise an electrolyte-filled nanoporous glass frit may also be affected by charge screening. As reported previously, when the Debye length approaches or surpasses the glass pore diameter, reference potentials depend on the composition of the bridge electrolyte, the pore size of the frit, and the concentration of electrolyte in the sample. We report here that stirring of samples may alter the reference potential as it affects the electrolyte concentration in the section of the nanoporous glass frit that is facing the sample solution. When the flow rate of bridge electrolyte into the sample is small, convective mass transport of sample into the nanoporous frit occurs. The depth of penetration into the frit is only a few nanometers but, despite the use of concentrated salt bridges, this is enough to affect the extent of electrostatic screening when samples of low ionic strength are measured. Mixing of sample and salt bridge solutions-and in particular penetration of sample components into the frit-was optically monitored by observation of a deeply colored Fe[(SCN)(H2O)5]2+ complex that formed in situ exclusively in the region where the sample and salt bridge mixed. Importantly, because flow through nanoporous frits is very slow, mass transport through these frits is dominated by diffusion. Consequently, over as little as 1 h, reference electrode frits with low flow rates become contaminated with sample components and undergo depletion of electrolyte within the frit to a depth of several millimeters, which can negatively affect subsequent experiments.

12.
Chem Commun (Camb) ; 55(39): 5551-5554, 2019 May 09.
Article in English | MEDLINE | ID: mdl-30972394

ABSTRACT

A supramolecular synthon approach was exploited to design amorphous solid dispersions (ASDs) of drugs containing an amino aromatic nitrogen moiety and a polyacrylic acid polymer. The interaction between a drug and polymer was confirmed by differential scanning calorimetry, spectroscopy (IR and 15N NMR), and X-ray crystallography. The interaction decreased the molecular mobility, conferred exceptional physical stability and enhanced the drug dissolution.

13.
Biochim Biophys Acta Biomembr ; 1860(6): 1335-1341, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29501609

ABSTRACT

Approximately, 70% of the Ca2+ ion transport into the sarcoplasmic reticulum is catalyzed by the sarcoplasmic reticulum Ca2+-ATPase (SERCA), whose activity is endogenously regulated by phospholamban (PLN). PLN comprises a TM inhibitory region and a cytoplasmic regulatory region that harbors a consensus sequence for cAMP-dependent protein kinase (PKA). The inhibitory region binds the ATPase, reducing its apparent Ca2+ binding affinity. ß-adrenergic stimulation activates PKA, which phosphorylates PLN at Ser 16, reversing its inhibitory function. Mutations and post-translational modifications of PLN may lead to dilated cardiomyopathy (DCM) and heart failure. PLN's cytoplasmic region interconverts between a membrane-associated T state and a membrane-detached R state. The importance of these structural transitions on SERCA regulation is emerging, but the effects of natural occurring mutations and their relevance to the progression of heart disease are unclear. Here we use solid-state NMR spectroscopy to investigate the structural dynamics of two lethal PLN mutations, R9C and R25C, which lead to DCM. We found that the R25C mutant enhances the dynamics of PLN and shifts the conformational equilibrium toward the R state confirmation, whereas the R9C mutant drives the amphipathic cytoplasmic domain toward the membrane-associate state, enriching the T state population. The changes in membrane interactions caused by these mutations may explain the aberrant regulation of SERCA.


Subject(s)
Amino Acid Substitution , Calcium-Binding Proteins/chemistry , Cardiomyopathy, Dilated/genetics , Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Mutation, Missense , Point Mutation , Amino Acid Sequence , Calcium-Binding Proteins/genetics , Consensus Sequence , Humans , Membrane Proteins/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Protein Domains , Protein Structure, Secondary , Recombinant Proteins/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
14.
ACS Chem Biol ; 11(2): 329-34, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26650884

ABSTRACT

To transmit signals across cellular compartments, many membrane-embedded enzymes undergo extensive conformational rearrangements. Monitoring these events in lipid bilayers by NMR at atomic resolution has been challenging due to the large size of these systems. It is further exacerbated for large mammalian proteins that are difficult to express and label with NMR-active isotopes. Here, we synthesized and engineered (13)C ethyl groups on native cysteines to map the structural transitions of the sarcoplasmic reticulum Ca(2+)-ATPase, a 110 kDa transmembrane enzyme that transports Ca(2+) into the sarcoplasmic reticulum. Using magic angle spinning NMR, we monitored the chemical shifts of the methylene and methyl groups of the derivatized cysteine residues along the major steps of the enzymatic cycle. The methylene chemical shifts are sensitive to the ATPase conformational changes induced upon nucleotide and Ca(2+) ion binding and are ideal probes for active and inactive states of the enzyme. This new approach is extendable to large mammalian enzymes and signaling proteins with native or engineered cysteine residues in their amino acid sequence.


Subject(s)
Lipid Bilayers/chemistry , Nuclear Magnetic Resonance, Biomolecular , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry , Animals , Binding Sites , Calcium/metabolism , Cysteine/analysis , Lipid Bilayers/metabolism , Models, Molecular , Protein Conformation , Rabbits , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
15.
Mol Pharm ; 12(9): 3339-50, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26070543

ABSTRACT

The effects of specific drug-polymer interactions (ionic or hydrogen-bonding) on the molecular mobility of model amorphous solid dispersions (ASDs) were investigated. ASDs of ketoconazole (KTZ), a weakly basic drug, with each of poly(acrylic acid) (PAA), poly(2-hydroxyethyl methacrylate) (PHEMA), and polyvinylpyrrolidone (PVP) were prepared. Drug-polymer interactions in the ASDs were evaluated by infrared and solid-state NMR, the molecular mobility quantified by dielectric spectroscopy, and crystallization onset monitored by differential scanning calorimetry (DSC) and variable temperature X-ray diffractometry (VTXRD). KTZ likely exhibited ionic interactions with PAA, hydrogen-bonding with PHEMA, and weaker dipole-dipole interactions with PVP. On the basis of dielectric spectroscopy, the α-relaxation times of the ASDs followed the order: PAA > PHEMA > PVP. In addition, the presence of ionic interactions also translated to a dramatic and disproportionate decrease in mobility as a function of polymer concentration. On the basis of both DSC and VTXRD, an increase in strength of interaction translated to higher crystallization onset temperature and a decrease in extent of crystallization. Stronger drug-polymer interactions, by reducing the molecular mobility, can potentially delay the crystallization onset temperature as well as crystallization extent.


Subject(s)
Acrylic Resins/chemistry , Ketoconazole/chemistry , Methacrylates/chemistry , Polymers/chemistry , Povidone/chemistry , Calorimetry, Differential Scanning , Crystallization , Drug Interactions , Drug Stability , Hydrogen Bonding , Phase Transition , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
16.
Structure ; 22(12): 1744-1753, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25456411

ABSTRACT

TLQP-21, a VGF-encoded peptide is emerging as a novel target for obesity-associated disorders. TLQP-21 is found in the sympathetic nerve terminals in the adipose tissue and targets the G-protein-coupled receptor complement-3a receptor1 (C3aR1). The mechanisms of TLQP-21-induced receptor activation remain unexplored. Here, we report that TLQP-21 is intrinsically disordered and undergoes a disorder-to-order transition, adopting an α-helical conformation upon targeting cells expressing the C3aR1. We determined that the hot spots for TLQP-21 are located at the C terminus, with mutations in the last four amino acids progressively reducing the bioactivity and, a single site mutation (R21A) or C-terminal amidation abolishing its function completely. Additionally, the human TLQP-21 sequence carrying a S20A substitution activates the human C3aR1 receptor with lower potency compared to the rodent sequence. These studies reveal the mechanism of action of TLQP-21 and provide molecular templates for designing agonists and antagonists to modulate C3aR1 functions.


Subject(s)
Peptide Fragments/metabolism , Receptors, Complement/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Female , Mice , Models, Molecular , Protein Binding , Protein Conformation , Rats , Rats, Wistar , Spleen/cytology , Spleen/metabolism
17.
Nat Commun ; 5: 3827, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24871041

ABSTRACT

α-synuclein (αS) is a protein involved in neurotransmitter release in presynaptic terminals, and whose aberrant aggregation is associated with Parkinson's disease. In dopaminergic neurons, αS exists in a tightly regulated equilibrium between water-soluble and membrane-associated forms. Here we use a combination of solid-state and solution NMR spectroscopy to characterize the conformations of αS bound to lipid membranes mimicking the composition and physical properties of synaptic vesicles. The study shows three αS regions possessing distinct structural and dynamical properties, including an N-terminal helical segment having a role of membrane anchor, an unstructured C-terminal region that is weakly associated with the membrane and a central region acting as a sensor of the lipid properties and determining the affinity of αS membrane binding. Taken together, our data define the nature of the interactions of αS with biological membranes and provide insights into their roles in the function of this protein and in the molecular processes leading to its aggregation.


Subject(s)
Cell Membrane/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Lipid Bilayers/chemistry , Lipids/chemistry , Magnetic Resonance Spectroscopy , Models, Biological , Protein Binding , Protein Structure, Secondary , Unilamellar Liposomes/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...