Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Mol Pharm ; 21(5): 2272-2283, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38607681

ABSTRACT

Over the years, there has been significant interest in PEGylated lipid-based nanocarriers within the drug delivery field. The inevitable interplay between the nanocarriers and plasma protein plays a pivotal role in their in vivo biological fate. Understanding the factors influencing lipid-based nanocarrier and protein corona interactions is of paramount importance in the design and clinical translation of these nanocarriers. Herein, discoid-shaped lipid nanodiscs (sNDs) composed of different phospholipids with varied lipid tails and head groups were fabricated. We investigated the impact of phospholipid components on the interaction between sNDs and serum proteins, particle stability, and biodistribution. The results showed that all of these lipid nanodiscs remained stable over a 15 day storage period, while their stability in the blood serum demonstrated significant differences. The sND composed of POPG exhibited the least stability due to its potent complement activation capability, resulting in rapid blood clearance. Furthermore, a negative correlation between the complement activation capability and serum stability was identified. Pharmacokinetic and biodistribution experiments indicated that phospholipid composition did not influence the capability of sNDs to evade the accelerated blood clearance phenomenon. Complement deposition on the sND was inversely associated with the area under the curve. Additionally, all lipid nanodiscs exhibited dominant adsorption of apolipoprotein. Remarkably, the POPC-based lipid nanodisc displayed a significantly higher deposition of apolipoprotein E, contributing to an obvious brain distribution, which provides a promising tool for brain-targeted drug delivery.


Subject(s)
Nanoparticles , Phospholipids , Protein Corona , Protein Corona/chemistry , Animals , Phospholipids/chemistry , Tissue Distribution , Mice , Nanoparticles/chemistry , Drug Carriers/chemistry , Nanostructures/chemistry , Male , Complement Activation/drug effects , Lipids/chemistry , Drug Delivery Systems/methods , Blood Proteins/metabolism , Blood Proteins/chemistry
2.
Biochim Biophys Acta Biomembr ; 1866(1): 184230, 2024 01.
Article in English | MEDLINE | ID: mdl-37704040

ABSTRACT

Nanodiscs are binary discoidal complexes of a phospholipid bilayer circumscribed by belt-like helical scaffold proteins. Using coarse-grained and all-atom molecular dynamics simulations, we explore the stability, size, and structure of nanodiscs formed between the N-terminal domain of apolipoprotein E3 (apoE3-NT) and variable number of 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) molecules. We study both parallel and antiparallel double-belt configurations, consisting of four proteins per nanodisc. Our simulations predict nanodiscs containing between 240 and 420 DMPC molecules to be stable. The antiparallel configurations exhibit an average of 1.6 times more amino acid interactions between protein chains and 2 times more ionic contacts, compared to the parallel configuration. With one exception, DMPC order parameters are consistently larger in the antiparallel configuration than in the parallel one. In most cases, the root mean square deviation of the positions of the protein backbone atoms is smaller in the antiparallel configuration. We further report nanodisc size, thickness, radius of gyration, and solvent accessible surface area. Combining all investigated parameters, we hypothesize the antiparallel protein configuration leading to more stable and more rigid nanodiscs than the parallel one.


Subject(s)
Lipid Bilayers , Molecular Dynamics Simulation , Apolipoprotein E3 , Lipid Bilayers/chemistry , Dimyristoylphosphatidylcholine/chemistry , Phospholipids/chemistry , Proteins
3.
Biomolecules ; 12(8)2022 08 04.
Article in English | MEDLINE | ID: mdl-36008970

ABSTRACT

Atomic-resolution structural studies of membrane-associated proteins and peptides in a membrane environment are important to fully understand their biological function and the roles played by them in the pathology of many diseases. However, the complexity of the cell membrane has severely limited the application of commonly used biophysical and biochemical techniques. Recent advancements in NMR spectroscopy and cryoEM approaches and the development of novel membrane mimetics have overcome some of the major challenges in this area. For example, the development of a variety of lipid-nanodiscs has enabled stable reconstitution and structural and functional studies of membrane proteins. In particular, the ability of synthetic amphipathic polymers to isolate membrane proteins directly from the cell membrane, along with the associated membrane components such as lipids, without the use of a detergent, has opened new avenues to study the structure and function of membrane proteins using a variety of biophysical and biological approaches. This review article is focused on covering the various polymers and approaches developed and their applications for the functional reconstitution and structural investigation of membrane proteins. The unique advantages and limitations of the use of synthetic polymers are also discussed.


Subject(s)
Membrane Proteins , Nanostructures , Lipid Bilayers/chemistry , Membrane Proteins/metabolism , Membranes , Nanostructures/chemistry , Polymers/chemistry
4.
Anal Biochem ; 639: 114521, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34906540

ABSTRACT

NMR is a powerful tool for characterizing intermolecular interactions at atomic resolution. However, the nature of the complex interactions of membrane-binding proteins makes it difficult to elucidate the interaction mechanisms. Here, we demonstrated that structural and thermodynamic analyses using solution NMR spectroscopy and isothermal titration calorimetry (ITC) can clearly detect a specific interaction between the pleckstrin homology (PH) domain of ceramide transport protein (CERT) and phosphatidylinositol 4-monophosphate (PI4P) embedded in the lipid nanodisc, and distinguish the specific interaction from nonspecific interactions with the bulk surface of the lipid nanodisc. This NMR-ITC hybrid strategy provides detailed characterization of protein-lipid membrane interactions.


Subject(s)
Lipid Bilayers/metabolism , Magnetic Resonance Spectroscopy/methods , Phosphatidylinositol Phosphates/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Calorimetry/instrumentation , Calorimetry/methods , Humans , Lipid Bilayers/chemistry , Magnetic Resonance Spectroscopy/instrumentation , Molecular Dynamics Simulation , Nanostructures/chemistry , Phosphatidylinositol Phosphates/chemistry , Protein Binding , Protein Domains , Protein Serine-Threonine Kinases/chemistry , Titrimetry/instrumentation , Titrimetry/methods
5.
J Biol Chem ; 297(2): 100964, 2021 08.
Article in English | MEDLINE | ID: mdl-34270960

ABSTRACT

Vacuolar H+-ATPases (V-ATPases) are large, multisubunit proton pumps that acidify the lumen of organelles in virtually every eukaryotic cell and in specialized acid-secreting animal cells, the enzyme pumps protons into the extracellular space. In higher organisms, most of the subunits are expressed as multiple isoforms, with some enriched in specific compartments or tissues and others expressed ubiquitously. In mammals, subunit a is expressed as four isoforms (a1-4) that target the enzyme to distinct biological membranes. Mutations in a isoforms are known to give rise to tissue-specific disease, and some a isoforms are upregulated and mislocalized to the plasma membrane in invasive cancers. However, isoform complexity and low abundance greatly complicate purification of active human V-ATPase, a prerequisite for developing isoform-specific therapeutics. Here, we report the purification of an active human V-ATPase in native lipid nanodiscs from a cell line stably expressing affinity-tagged a isoform 4 (a4). We find that exogenous expression of this single subunit in HEK293F cells permits assembly of a functional V-ATPase by incorporation of endogenous subunits. The ATPase activity of the preparation is >95% sensitive to concanamycin A, indicating that the lipid nanodisc-reconstituted enzyme is functionally coupled. Moreover, this strategy permits purification of the enzyme's isolated membrane subcomplex together with biosynthetic assembly factors coiled-coil domain-containing protein 115, transmembrane protein 199, and vacuolar H+-ATPase assembly integral membrane protein 21. Our work thus lays the groundwork for biochemical characterization of active human V-ATPase in an a subunit isoform-specific manner and establishes a platform for the study of the assembly and regulation of the human holoenzyme.


Subject(s)
Vacuolar Proton-Translocating ATPases , Biological Transport , Cell Membrane/metabolism , Humans , Saccharomyces cerevisiae/metabolism
6.
Structure ; 29(11): 1286-1294.e6, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34174188

ABSTRACT

The 12-transmembrane protein Patched (Ptc1) acts as a suppressor for Hedgehog (Hh) signaling by depleting sterols in the cytoplasmic membrane leaflet that are required for the activation of downstream regulators. The positive modulator Hh inhibits Ptc1's transporter function by binding to Ptc1 and its co-receptors, which are locally concentrated in invaginated microdomains known as caveolae. Here, we reconstitute the mouse Ptc1 into lipid nanodiscs and determine its structure using single-particle cryoelectron microscopy. The structure is overall similar to those in amphipol and detergents but displays various conformational differences in the transmembrane region. Although most particles show monomers, we observe Ptc1 dimers with distinct interaction patterns and different membrane curvatures, some of which are reminiscent of caveolae. We find that an extramembranous "hand-shake" region rich in hydrophobic and aromatic residues mediates inter-Ptc1 interactions under different membrane curvatures. Our data provide a plausible framework for Ptc1 clustering in the highly curved caveolae.


Subject(s)
Caveolae/metabolism , Cell Membrane/metabolism , Hedgehog Proteins/metabolism , Lipids , Patched-1 Receptor/metabolism , Animals , Cryoelectron Microscopy , Mice , Signal Transduction/physiology
7.
ACS Appl Bio Mater ; 4(6): 4760-4768, 2021 06 21.
Article in English | MEDLINE | ID: mdl-35007026

ABSTRACT

The study of membrane proteins remains challenging, especially in a native membrane environment. Recently, major progress has been made using maleic acid copolymers, such as styrene maleic acid, to purify membrane proteins and study them directly with native lipids associated with the membrane. Additional maleic acid copolymers, such as diisobutylene maleic acid (DIBMA) membrane-mimetic systems, are being developed and found to have improved spectroscopic properties and pH stability. We studied DIBMA and its lipid particles in solution to better understand its assembly, without and with the lipids, to provide an insight regarding how to use it in solution for better membrane extraction. Using small-angle neutron and X-ray scattering (SANS/SAXS), we show that DIBMA organizes into structures of different size scales at various concentrations and ionic strengths. The polymer performed reasonably well under most solvent conditions except in very low concentrations and high-salt conditions that could result in limited interaction with lipids. To explore DIBMA lipid particles as a suitable membrane-mimetic system for neutron scattering studies of membrane proteins, we measured and determined the contrast-matching point of DIBMA to be ∼12% (v/v) D2O - similar to that of most protiated lipid molecules but distinct from that of regular protiated proteins - providing a natural contrast for separating their neutron scattering signals. Using SANS contrast variation, we demonstrated that the scattering from the whole lipid particle can be annihilated. Further, we determined that a well-defined lipid nanodisc structure with DIBMA was contrast-matched. These results demonstrate that the DIBMA lipid particle is an outstanding "stealth" membrane-mimetic for membrane proteins. The results provide a structural framework for understanding the organization and assembly process of the polymer itself and the lipid molecules. Such an understanding is imperative for structural techniques such as cryo-electron microscopy, nuclear magnetic resonance, small-angle scattering, and other biophysical techniques.


Subject(s)
Alkenes/chemistry , Dimyristoylphosphatidylcholine/chemistry , Lipid Bilayers/chemistry , Maleates/chemistry , Membrane Proteins/chemistry , Polymers/chemistry , Biomedical Research , Biomimetics , Hydrogen-Ion Concentration , Osmolar Concentration
8.
J Chem Inf Model ; 60(5): 2448-2457, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32163280

ABSTRACT

A giant technological leap in the field of cryo-electron microscopy (cryo-EM) has assured the achievement of near-atomic resolution structures of biological macromolecules. As a recognition of this accomplishment, the Nobel Prize in Chemistry was awarded in 2017 to Jacques Dubochet, Joachim Frank, and Richard Henderson, the pioneers in the field of cryo-EM. Currently, the technique has become the method of choice for structural analysis of heterogeneous and intrinsically dynamic biological complexes. In the past few years, the most prolific branch of cryo-EM, single particle analysis, has revolutionized the structural biology field and structural investigation of membrane proteins, in particular. To achieve high-resolution structures of macromolecules in noncrystalline specimens, from sample and grid preparation to image acquisition, image data processing, and analysis of 3D maps, methodological advances in each of the steps play critical roles. In this Review, we discuss two areas in single particle cryo-EM, the remarkable developments in sample preparation strategies, particularly for membrane proteins, and breakthroughs in methodologies for molecular model building on the high-resolution 3D density maps that brought promises to resolve high-resolution (<4 Å) structures of biological macromolecules.


Subject(s)
Membrane Proteins , Single Molecule Imaging , Cryoelectron Microscopy , Macromolecular Substances , Models, Molecular
9.
Colloids Surf B Biointerfaces ; 186: 110680, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31835183

ABSTRACT

The integral membrane protein, bacteriorhodopsin (BR) was encapsulated in sol-gel derived porous silica gel monoliths in native purple membrane (BR-PM) and synthetic lipid nanodisc (BR nanodisc) environments. BR nanodiscs were synthesized by solubilizing purple membrane in discoidal phospholipid bilayer stabilized by amphipathic Styrene-Maleic Acid (SMA) copolymer. UV-vis absorbance spectroscopy and dynamic-light scattering indicated the formation of BR monomers solubilized in lipid nanodiscs 10.2 ± 0.7 nm in average diameter. Fluorescence and absorbance spectroscopic techniques were utilized to probe conformational, environmental, and rotational changes associated with the tryptophan residues and the covalently-bound retinal moiety of BR upon entrapment in the silica matrix. We show that the immobilized BR in both membrane environments retained its bound retinal cofactor and the ability of the cofactor to undergo conformational changes upon light illumination necessary for BR's activity as a proton transporter. For purple membrane fragments, the results indicated that the local pH in the pores around BR after encapsulation was important for its stability at temperatures higher than 50 °C. Under the same buffering conditions, retinal was released from silica-encapsulated BR-PM and BR nanodiscs beginning at 80 °C (without a conformational change) and 50 °C (with a conformational change), respectively, reflecting differences in protein-protein (trimeric vs. monomeric) and protein-lipid interactions.


Subject(s)
Bacteriorhodopsins/chemistry , Lipids/chemistry , Nanostructures/chemistry , Purple Membrane/chemistry , Silicon Dioxide/chemistry , Gels/chemistry , Particle Size , Surface Properties
10.
J Biol Chem ; 294(16): 6439-6449, 2019 04 19.
Article in English | MEDLINE | ID: mdl-30792311

ABSTRACT

The vacuolar H+-ATPase (V-ATPase; V1Vo-ATPase) is an ATP-dependent proton pump that acidifies subcellular compartments in all eukaryotic organisms. V-ATPase activity is regulated by reversible disassembly into autoinhibited V1-ATPase and Vo proton channel subcomplexes, a process that is poorly understood on the molecular level. V-ATPase is a rotary motor, and recent structural analyses have revealed different rotary states for disassembled V1 and Vo, a mismatch that is likely responsible for their inability to reconstitute into holo V-ATPase in vitro Here, using the model organism Saccharomyces cerevisiae, we show that a key impediment for binding of V1 to Vo is the conformation of the inhibitory C-terminal domain of subunit H (HCT). Using biolayer interferometry and biochemical analyses of purified mutant V1-ATPase and Vo proton channel reconstituted into vacuolar lipid-containing nanodiscs, we further demonstrate that disruption of HCT's V1-binding site facilitates assembly of a functionally coupled and stable V1Vo-ATPase. Unlike WT, this mutant enzyme was resistant to MgATP hydrolysis-induced dissociation, further highlighting HCT's role in the mechanism of V-ATPase regulation. Our findings provide key insight into the molecular events underlying regulation of V-ATPase activity by reversible disassembly.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Vacuolar Proton-Translocating ATPases/chemistry , Mutation , Protein Domains , Protein Structure, Quaternary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
11.
Biochim Biophys Acta Biomembr ; 1861(4): 787-797, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30639287

ABSTRACT

CEACAM1, a homotypic transmembrane receptor with 12 or 72 amino acid cytosolic domain isoforms, is converted from inactive cis-dimers to active trans-dimers by calcium-calmodulin (Ca2+/CaM). Previously, the weak binding of Ca2+/CaM to the human 12 AA cytosolic domain was studied using C-terminal anchored peptides. We now show the binding of 15N labeled Phe-454 cytosolic domain peptides in solution or membrane anchored using NMR demonstrates a significant role for the lipid bilayer. Although binding is increased by the mutation Phe454Ala, this mutation was previously shown to abrogate actin binding. On the other hand, Ca2+/CaM binding is abrogated by phosphorylation of nearby Thr-457, a post-translation modification required for actin binding and subsequent in vitro lumen formation. Binding of Ca2+/CaM to a membrane proximal peptide from the long 72 AA cytosolic domain anchored to lipid nanodiscs was very weak compared to lipid free conditions, suggesting membrane specific effects between the two isoforms. NMR analysis of 15N labeled Ca2+/CaM with unlabeled peptides showed the C-lobe of Ca2+/CaM is involved in peptide interactions, and hydrophobic residues such as Met-109, Val-142 and Met-144 play important roles in binding peptide. This information was incorporated into transmembrane models of CEACAM1 binding to Ca2+/CaM. The lack of Ca2+/CaM binding to phosphorylated Thr-457, a residue we have previously shown to be phosphorylated by CaMK2D, also dependent on Ca2+/CaM, suggests stepwise binding of the cytosolic domain first to Ca2+/CaM and then to actin.


Subject(s)
Antigens, CD/chemistry , Calcium/chemistry , Calmodulin/chemistry , Cell Adhesion Molecules/chemistry , Models, Molecular , Nanostructures/chemistry , Peptides/chemistry , Antigens, CD/genetics , Calmodulin/genetics , Cell Adhesion Molecules/genetics , Humans , Nanostructures/ultrastructure , Nuclear Magnetic Resonance, Biomolecular , Peptides/genetics
12.
Life (Basel) ; 8(4)2018 Nov 02.
Article in English | MEDLINE | ID: mdl-30400226

ABSTRACT

Membrane proteins are important drug targets which play a pivotal role in various cellular activities. However, unlike cytosolic proteins, most of them are difficult-to-express proteins. In this study, to synthesize and produce sufficient quantities of membrane proteins for functional and structural analysis, we used a bottom-up approach in a reconstituted cell-free synthesis system, the PURE system, supplemented with artificial lipid mimetics or micelles. Membrane proteins were synthesized by the cell-free system and integrated into lipid bilayers co-translationally. Membrane proteins such as the G-protein coupled receptors were expressed in the PURE system and a productivity ranging from 0.04 to 0.1 mg per mL of reaction was achieved with a correct secondary structure as predicted by circular dichroism spectrum. In addition, a ligand binding constant of 27.8 nM in lipid nanodisc and 39.4 nM in micelle was obtained by surface plasmon resonance and the membrane protein localization was confirmed by confocal microscopy in giant unilamellar vesicles. We found that our method is a promising approach to study the different classes of membrane proteins in their native-like artificial lipid bilayer environment for functional and structural studies.

13.
Elife ; 72018 08 15.
Article in English | MEDLINE | ID: mdl-30109985

ABSTRACT

Voltage-activated potassium (Kv) channels open to conduct K+ ions in response to membrane depolarization, and subsequently enter non-conducting states through distinct mechanisms of inactivation. X-ray structures of detergent-solubilized Kv channels appear to have captured an open state even though a non-conducting C-type inactivated state would predominate in membranes in the absence of a transmembrane voltage. However, structures for a voltage-activated ion channel in a lipid bilayer environment have not yet been reported. Here we report the structure of the Kv1.2-2.1 paddle chimera channel reconstituted into lipid nanodiscs using single-particle cryo-electron microscopy. At a resolution of ~3 Å for the cytosolic domain and ~4 Å for the transmembrane domain, the structure determined in nanodiscs is similar to the previously determined X-ray structure. Our findings show that large differences in structure between detergent and lipid bilayer environments are unlikely, and enable us to propose possible structural mechanisms for C-type inactivation.


Subject(s)
Kv1.2 Potassium Channel/ultrastructure , Lipid Bilayers/chemistry , Nanocomposites/ultrastructure , Shab Potassium Channels/ultrastructure , Animals , Cryoelectron Microscopy , Crystallography, X-Ray , Ion Channel Gating , Kv1.2 Potassium Channel/chemistry , Nanocomposites/chemistry , Potassium/chemistry , Protein Conformation , Rats , Shab Potassium Channels/chemistry
14.
Elife ; 72018 07 20.
Article in English | MEDLINE | ID: mdl-30028291

ABSTRACT

Cyclic nucleotide-modulated channels have important roles in visual signal transduction and pacemaking. Binding of cyclic nucleotides (cAMP/cGMP) elicits diverse functional responses in different channels within the family despite their high sequence and structure homology. The molecular mechanisms responsible for ligand discrimination and gating are unknown due to lack of correspondence between structural information and functional states. Using single particle cryo-electron microscopy and single-channel recording, we assigned functional states to high-resolution structures of SthK, a prokaryotic cyclic nucleotide-gated channel. The structures for apo, cAMP-bound, and cGMP-bound SthK in lipid nanodiscs, correspond to no, moderate, and low single-channel activity, respectively, consistent with the observation that all structures are in resting, closed states. The similarity between apo and ligand-bound structures indicates that ligand-binding domains are strongly coupled to pore and SthK gates in an allosteric, concerted fashion. The different orientations of cAMP and cGMP in the 'resting' and 'activated' structures suggest a mechanism for ligand discrimination.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide-Gated Cation Channels/chemistry , Cyclic Nucleotide-Gated Cation Channels/metabolism , Cryoelectron Microscopy , Models, Molecular , Protein Binding , Protein Conformation , Spirochaeta/enzymology
15.
Mol Cell ; 69(6): 993-1004.e3, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29526695

ABSTRACT

The molecular mechanism of transmembrane proton translocation in rotary motor ATPases is not fully understood. Here, we report the 3.5-Å resolution cryoEM structure of the lipid nanodisc-reconstituted Vo proton channel of the yeast vacuolar H+-ATPase, captured in a physiologically relevant, autoinhibited state. The resulting atomic model provides structural detail for the amino acids that constitute the proton pathway at the interface of the proteolipid ring and subunit a. Based on the structure and previous mutagenesis studies, we propose the chemical basis of transmembrane proton transport. Moreover, we discovered that the C terminus of the assembly factor Voa1 is an integral component of mature Vo. Voa1's C-terminal transmembrane α helix is bound inside the proteolipid ring, where it contributes to the stability of the complex. Our structure rationalizes possible mechanisms by which mutations in human Vo can result in disease phenotypes and may thus provide new avenues for therapeutic interventions.


Subject(s)
Cryoelectron Microscopy , Nanoparticles , Saccharomyces cerevisiae Proteins/ultrastructure , Saccharomyces cerevisiae/enzymology , Vacuolar Proton-Translocating ATPases/ultrastructure , Genotype , Humans , Membrane Lipids/chemistry , Models, Molecular , Mutation , Phenotype , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Subunits , Protons , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
16.
J Biomol NMR ; 70(1): 21-31, 2018 01.
Article in English | MEDLINE | ID: mdl-29168021

ABSTRACT

NMR spectroscopy of membrane proteins involved in electron transport is difficult due to the presence of both the lipids and paramagnetic centers. Here we report the solution NMR study of the NADPH-cytochrome P450 oxidoreductase (POR) in its reduced and oxidized states. We interrogate POR, first, in its truncated soluble form (70 kDa), which is followed by experiments with the full-length protein incorporated in a lipid nanodisc (240 kDa). To overcome paramagnetic relaxation in the reduced state of POR as well as the signal broadening due to its high molecular weight, we utilized the methyl-TROSY approach. Extrinsic 13C-methyl groups were introduced by modifying the engineered surface-exposed cysteines with methyl-methanethiosulfonate. Chemical shift dispersion of the resonances from different sites in POR was sufficient to monitor differential effects of the reduction-oxidation process and conformation changes in the POR structure related to its function. Despite the high molecular weight of the POR-nanodisc complex, the surface-localized 13C-methyl probes were sufficiently mobile to allow for signal detection at 600 MHz without perdeuteration. This work demonstrates a potential of the solution methyl-TROSY in analysis of structure, dynamics, and function of POR, which may also be applicable to similar paramagnetic and flexible membrane proteins.


Subject(s)
Membrane Proteins/chemistry , NADPH-Ferrihemoprotein Reductase/chemistry , Carbon Isotopes , Lipids , Membrane Proteins/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism , Oxidation-Reduction , Protein Binding , Protein Conformation , Solubility , Structure-Activity Relationship
17.
Biochim Biophys Acta Biomembr ; 1859(10): 2096-2105, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28782501

ABSTRACT

Lipid nanodisc, a model membrane platform originally synthesized for study of membrane proteins, has recently been used as the carrier to deliver amphiphilic drugs into target tumor cells. However, the central question of how cells interact with such emerging nanomaterials remains unclear and deserves our research for both improving the delivery efficiency and reducing the side effect. In this work, a binary lipid nanodisc is designed as the minimum model to investigate its interactions with plasma membranes by using the dissipative particle dynamics method. Three typical interaction pathways, including the membrane attachment with lipid domain exchange of nanodiscs, the partial membrane wrapping with nanodisc vesiculation, and the receptor-mediated endocytosis, are discovered. For the first pathway, the boundary normal lipids acting as ligands diffuse along the nanodisc rim to gather at the membrane interface, repelling the central bola lipids to reach a stable membrane attachment. If bola lipids are positioned at the periphery and act as ligands, they diffuse to form a large aggregate being wrapped by the membrane, leaving the normal lipids exposed on the membrane exterior by assembling into a vesicle. Finally, by setting both central normal lipids and boundary bola lipids as ligands, the receptor-mediated endocytosis occurs via both deformation and self-rotation of the nanodiscs. All above pathways for soft lipid nanodiscs are quite different from those for rigid nanoparticles, which may provide useful guidelines for design of soft lipid nanodiscs in widespread biomedical applications.


Subject(s)
Cell Membrane/metabolism , Lipid Bilayers/metabolism , Lipids/physiology , Endocytosis/physiology , Membrane Proteins/metabolism , Models, Molecular , Nanoparticles/chemistry , Nanostructures/chemistry
18.
ACS Nano ; 11(4): 3651-3661, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28291322

ABSTRACT

Gold nanoparticles (AuNPs) exhibit strong fluorescent and electromagnetic properties, which can be enhanced upon clustering and used in therapeutic, imaging, and sensing applications. A combination of gold nanoparticles with lipid nanodiscs can be attractive for AuNP self-assembly and useful in biomedical applications. Using molecular dynamics simulations we show that lipid nanodiscs can serve as templates for AuNP clustering into rings and string-like structures. We demonstrate that equilibrium encapsulation of 1 nm hydrophobically modified AuNPs into lipid nanodiscs composed of a mixture of dipalmitoylphosphatidylcholine (DPPC) and dihexanoylphosphatidylcholine (DHPC) lipids occurs at the rim and results in formation of a ring of gold. The interior of the nanodisc is inaccessible to AuNPs due to the DPPC liquid crystalline order. With temperature increase the lipid order diminishes, initiating the nanodisc transformation into a vesicle, upon which encapsulated AuNPs cluster into a close-packed string or nanoring, thereby stalling the vesiculation process at a "round vase" or cup-like stage depending on the AuNP concentration. In contrast, encapsulation of AuNPs by an equilibrium lipid vesicle results in its deformation with randomly clustered AuNPs, in agreement with experimental observations. We characterize the AuNP cluster size and surface-to-surface pair distribution, both of which impact the AuNP luminescent properties. We investigate the effect of alkane tether length on the nanodisc stability and AuNP clustering inside the nanodiscs and vesicles. Our results show that lipid nanodiscs can enhance gold cluster formation, which can be further exploited in imaging applications.

19.
Structure ; 25(2): 384-392, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28089451

ABSTRACT

For some membrane proteins, detergent-mediated solubilization compromises protein stability and functionality, often impairing biophysical and structural analyses. Hence, membrane-protein structure determination is a continuing bottleneck in the field of protein crystallography. Here, as an alternative to approaches mediated by conventional detergents, we report the crystallogenesis of a recombinantly produced membrane protein that never left a lipid bilayer environment. We used styrene-maleic acid (SMA) copolymers to solubilize lipid-embedded proteins into SMA nanodiscs, purified these discs by affinity and size-exclusion chromatography, and transferred proteins into the lipidic cubic phase (LCP) for in meso crystallization. The 2.0-Å structure of an α-helical seven-transmembrane microbial rhodopsin thus obtained is of high quality and virtually identical to the 2.2-Å structure obtained from traditional detergent-based purification and subsequent LCP crystallization.


Subject(s)
Bacteriorhodopsins/chemistry , Crystallography, X-Ray/methods , Halobacteriaceae/chemistry , Maleates/chemistry , Membrane Proteins/chemistry , Nanostructures/chemistry , Polystyrenes/chemistry , Bacteriorhodopsins/genetics , Bacteriorhodopsins/metabolism , Cloning, Molecular , Crystallization , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Conformation, alpha-Helical , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solubility
20.
J Biol Chem ; 292(5): 1749-1761, 2017 02 03.
Article in English | MEDLINE | ID: mdl-27965356

ABSTRACT

Eukaryotic vacuolar H+-ATPase (V-ATPase) is a multisubunit enzyme complex that acidifies subcellular organelles and the extracellular space. V-ATPase consists of soluble V1-ATPase and membrane-integral Vo proton channel sectors. To investigate the mechanism of V-ATPase regulation by reversible disassembly, we recently determined a cryo-EM reconstruction of yeast Vo The structure indicated that, when V1 is released from Vo, the N-terminal cytoplasmic domain of subunit a (aNT) changes conformation to bind rotor subunit d However, insufficient resolution precluded a precise definition of the aNT-d interface. Here we reconstituted Vo into lipid nanodiscs for single-particle EM. 3D reconstructions calculated at ∼15-Šresolution revealed two sites of contact between aNT and d that are mediated by highly conserved charged residues. Alanine mutagenesis of some of these residues disrupted the aNT-d interaction, as shown by isothermal titration calorimetry and gel filtration of recombinant subunits. A recent cryo-EM study of holo V-ATPase revealed three major conformations corresponding to three rotational states of the central rotor of the enzyme. Comparison of the three V-ATPase conformations with the structure of nanodisc-bound Vo revealed that Vo is halted in rotational state 3. Combined with our prior work that showed autoinhibited V1-ATPase to be arrested in state 2, we propose a model in which the conformational mismatch between free V1 and Vo functions to prevent unintended reassembly of holo V-ATPase when activity is not needed.


Subject(s)
Membrane Lipids/chemistry , Nanostructures/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Vacuolar Proton-Translocating ATPases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...