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










Publication year range
1.
Chem Asian J ; 18(8): e202300029, 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-36808824

ABSTRACT

We have proposed a new method for the exploration of organic functional molecules, using an exhaustive molecular generator combined without combinatorial explosion and electronic state predicted by machine learning and adapted for developing n-type organic semiconductor molecules for field-effect transistors. Our method first enumerates skeletal structures as much as possible and next generates fused ring structures using substitution operations for atomic nodes and bond edges. We have succeeded in generating more than 4.8 million molecules. We calculated the electron affinity (EA) of about 51 thousand molecules with DFT calculation and trained the graph neural networks to estimate EA values of generated molecules. Finally, we obtained the 727 thousand molecules as candidates that satisfy EA values over 3 eV. The number of these possible candidate molecules is far beyond what we have been able to propose based on our knowledge and experience in synthetic chemistry, indicating a wide diversity of organic molecules.

2.
Sci Adv ; 8(34): eabp9011, 2022 Aug 26.
Article in English | MEDLINE | ID: mdl-36001659

ABSTRACT

Site-2 proteases are a conserved family of intramembrane proteases that cleave transmembrane substrates to regulate signal transduction and maintain proteostasis. Here, we elucidated crystal structures of inhibitor-bound forms of bacterial site-2 proteases including Escherichia coli RseP. Structure-based chemical modification and cross-linking experiments indicated that the RseP domains surrounding the active center undergo conformational changes to expose the substrate-binding site, suggesting that RseP has a gating mechanism to regulate substrate entry. Furthermore, mutational analysis suggests that a conserved electrostatic linkage between the transmembrane and peripheral membrane-associated domains mediates the conformational changes. In vivo cleavage assays also support that the substrate transmembrane helix is unwound by strand addition to the intramembrane ß sheet of RseP and is clamped by a conserved asparagine residue at the active center for efficient cleavage. This mechanism underlying the substrate binding, i.e., unwinding and clamping, appears common across distinct families of intramembrane proteases that cleave transmembrane segments.

3.
PLoS One ; 17(2): e0263296, 2022.
Article in English | MEDLINE | ID: mdl-35113924

ABSTRACT

G-protein-coupled receptors (GPCR) are a family of membrane receptors that play important roles in the regulation of various physiological phenomena. LPA receptors (LPA1-6) are members of the class A GPCRs, which transduce a lysophosphatidic acid (LPA) signal across the cell membrane and evoke various responses, including cellular survival, proliferation, differentiation, and migration. The crystal structure of LPA6 revealed a gap between its transmembrane helices (TMs), which is opened toward the membrane side. This led to the proposal of the "lateral access model," in which its lipophilic ligand directly enters the binding pocket through the gap structure at the membrane. In this study, we performed molecular dynamics (MD) simulations and Markov state model (MSM) analyses of LPA6 and LPA, to elucidate the long timescale dynamics of the ligand binding process. The results from the 71.4-µs MD simulation suggested that the flexibility of the TMs constituting the gap structure enables the lateral entrance of the ligand, and the key interactions between the receptor and ligand facilitate the transition state of the ligand binding process.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Receptors, Lysophosphatidic Acid/chemistry , Cell Membrane/metabolism , Computer Simulation , Crystallography, X-Ray , HEK293 Cells , Humans , Ligands , Markov Chains , Molecular Dynamics Simulation , Protein Binding , Signal Transduction , Transforming Growth Factor alpha/chemistry
4.
Elife ; 102021 03 23.
Article in English | MEDLINE | ID: mdl-33752801

ABSTRACT

Channelrhodopsins (ChRs) are microbial light-gated ion channels utilized in optogenetics to control neural activity with light . Light absorption causes retinal chromophore isomerization and subsequent protein conformational changes visualized as optically distinguished intermediates, coupled with channel opening and closing. However, the detailed molecular events underlying channel gating remain unknown. We performed time-resolved serial femtosecond crystallographic analyses of ChR by using an X-ray free electron laser, which revealed conformational changes following photoactivation. The isomerized retinal adopts a twisted conformation and shifts toward the putative internal proton donor residues, consequently inducing an outward shift of TM3, as well as a local deformation in TM7. These early conformational changes in the pore-forming helices should be the triggers that lead to opening of the ion conducting pore.


Subject(s)
Algal Proteins/genetics , Channelrhodopsins/genetics , Chlamydomonas reinhardtii/genetics , Algal Proteins/chemistry , Algal Proteins/metabolism , Amino Acid Sequence , Channelrhodopsins/chemistry , Channelrhodopsins/metabolism , Chlamydomonas reinhardtii/metabolism , Crystallography , Isomerism , Protein Conformation , Protein Structure, Secondary , Sequence Alignment
5.
Protein Sci ; 29(12): 2398-2407, 2020 12.
Article in English | MEDLINE | ID: mdl-33016372

ABSTRACT

System xc - is an amino acid antiporter that imports L-cystine into cells and exports intracellular L-glutamate, at a 1:1 ratio. As L-cystine is an essential precursor for glutathione synthesis, system xc - supports tumor cell growth through glutathione-based oxidative stress resistance and is considered as a potential therapeutic target for cancer treatment. System xc - consists of two subunits, the light chain subunit SLC7A11 (xCT) and the heavy chain subunit SLC3A2 (also known as CD98hc or 4F2hc), which are linked by a conserved disulfide bridge. Although the recent structures of another SLC7 member, L-type amino acid transporter 1 (LAT1) in complex with CD98hc, have provided the structural basis toward understanding the amino acid transport mechanism, the detailed molecular mechanism of xCT remains unknown. To revealthe molecular mechanism, we performed single-particle analyses of the xCT-CD98hc complex. As wild-type xCT-CD98hc displayed poor stability and could not be purified to homogeneity, we applied a consensus mutagenesis approach to xCT. The consensus mutated construct exhibited increased stability as compared to the wild-type, and enabled the cryoelectron microscopy (cryo-EM) map to be obtained at 6.2 Å resolution by single-particle analysis. The cryo-EM map revealed sufficient electron density to assign secondary structures. In the xCT structure, the hash and arm domains are well resolved, whereas the bundle domain shows some flexibility. CD98hc is positioned next to the xCT transmembrane domain. This study provides the structural basis of xCT, and our consensus-based strategy could represent a good choice toward solving unstable protein structures.


Subject(s)
Amino Acid Transport System y+/chemistry , Amino Acid Transport System y+/ultrastructure , Cryoelectron Microscopy , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Animals , Fusion Regulatory Protein 1, Heavy Chain/chemistry , Fusion Regulatory Protein 1, Heavy Chain/genetics , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Fusion Regulatory Protein 1, Heavy Chain/ultrastructure , HEK293 Cells , Humans , Mutagenesis , Protein Domains , Protein Stability , Protein Structure, Secondary , Sf9 Cells , Spodoptera
6.
Nat Commun ; 11(1): 1606, 2020 03 30.
Article in English | MEDLINE | ID: mdl-32231207

ABSTRACT

Tetraspanins play critical roles in various physiological processes, ranging from cell adhesion to virus infection. The members of the tetraspanin family have four membrane-spanning domains and short and large extracellular loops, and associate with a broad range of other functional proteins to exert cellular functions. Here we report the crystal structure of CD9 and the cryo-electron microscopic structure of CD9 in complex with its single membrane-spanning partner protein, EWI-2. The reversed cone-like molecular shape of CD9 generates membrane curvature in the crystalline lipid layers, which explains the CD9 localization in regions with high membrane curvature and its implications in membrane remodeling. The molecular interaction between CD9 and EWI-2 is mainly mediated through the small residues in the transmembrane region and protein/lipid interactions, whereas the fertilization assay revealed the critical involvement of the LEL region in the sperm-egg fusion, indicating the different dependency of each binding domain for other partner proteins.


Subject(s)
Tetraspanin 29/chemistry , Tetraspanin 29/physiology , Animals , Antigens, CD/chemistry , Cell Adhesion/physiology , Cryoelectron Microscopy , Crystallography, X-Ray , Female , Fertilization/physiology , Humans , Male , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Mice, Knockout , Models, Molecular , Protein Conformation , Tetraspanin 29/genetics
7.
Structure ; 26(9): 1284-1296.e4, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30196811

ABSTRACT

Secondary active transporters translocate their substrates using the electrochemical potentials of other chemicals and undergo large-scale conformational changes. Despite extensive structural studies, the atomic details of the transport mechanism still remain elusive. We performed a series of all-atom molecular dynamics simulations of the triose-phosphate/phosphate translocator (TPT), which exports organic phosphates in the chloroplast stroma in strict counter exchange with inorganic phosphate (Pi). Biased sampling methods, including the string method and umbrella sampling, successfully reproduced the conformational changes between the inward- and outward-facing states, along with the substrate binding. The free energy landscape of this entire TPT transition pathway demonstrated the alternating access and substrate translocation mechanisms, which revealed that Pi is relayed by positively charged residues along the transition pathway. Furthermore, the conserved Glu207 functions as a "molecular switch", linking the local substrate binding and the global conformational transition. Our results provide atomic-detailed insights into the substrate transport mechanism of the antiporter.


Subject(s)
Algal Proteins/chemistry , Algal Proteins/metabolism , Rhodophyta/metabolism , Biological Transport , Computer Simulation , Hydrogen Bonding , Molecular Dynamics Simulation , Phosphates/metabolism , Protein Folding , Rhodophyta/chemistry
8.
Biophys J ; 115(7): 1281-1291, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30236783

ABSTRACT

Channelrhodopsins (ChRs) are microbial light-gated ion channels with a retinal chromophore and are widely utilized in optogenetics to precisely control neuronal activity with light. Despite increasing understanding of their structures and photoactivation kinetics, the atomistic mechanism of light gating and ion conduction remains elusive. Here, we present an atomic structural model of a chimeric ChR in a precursor state of the channel opening determined by an accurate hybrid molecular simulation technique and a statistical theory of internal water distribution. The photoactivated structure features extensive tilt of the chromophore accompanied by redistribution of water molecules in its binding pocket, which is absent in previously known photoactivated structures of analogous photoreceptors, and widely agrees with structural and spectroscopic experimental evidence of ChRs. The atomistic model manifests a photoactivated ion-conduction pathway that is markedly different from a previously proposed one and successfully explains experimentally observed mutagenic effects on key channel properties.


Subject(s)
Channelrhodopsins/chemistry , Channelrhodopsins/metabolism , Ion Channel Gating/radiation effects , Light , Models, Molecular , Protein Conformation/radiation effects , Thermodynamics
9.
Nat Plants ; 3(10): 825-832, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28970497

ABSTRACT

The triose-phosphate/phosphate translocator (TPT) catalyses the strict 1:1 exchange of triose-phosphate, 3-phosphoglycerate and inorganic phosphate across the chloroplast envelope, and plays crucial roles in photosynthesis. Despite rigorous study for more than 40 years, the molecular mechanism of TPT is poorly understood because of the lack of structural information. Here we report crystal structures of TPT bound to two different substrates, 3-phosphoglycerate and inorganic phosphate, in occluded conformations. The structures reveal that TPT adopts a 10-transmembrane drug/metabolite transporter fold. Both substrates are bound within the same central pocket, where conserved lysine, arginine and tyrosine residues recognize the shared phosphate group. A structural comparison with the outward-open conformation of the bacterial drug/metabolite transporter suggests a rocker-switch motion of helix bundles, and molecular dynamics simulations support a model in which this rocker-switch motion is tightly coupled to the substrate binding, to ensure strict 1:1 exchange. These results reveal the unique mechanism of sugar phosphate/phosphate exchange by TPT.


Subject(s)
Phosphate Transport Proteins/chemistry , Phosphate Transport Proteins/metabolism , Plant Proteins/chemistry , Plant Proteins/metabolism , Arabidopsis/metabolism , Glyceric Acids/chemistry , Glyceric Acids/metabolism , Models, Molecular , Phosphates/chemistry , Phosphates/metabolism , Protein Conformation , Rhodophyta/metabolism , Structure-Activity Relationship , Substrate Specificity
10.
Nat Commun ; 8(1): 876, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29026074

ABSTRACT

P2X receptors are non-selective cation channels gated by extracellular ATP, and the P2X7 receptor subtype plays a crucial role in the immune and nervous systems. Altered expression and dysfunctions of P2X7 receptors caused by genetic deletions, mutations, and polymorphic variations have been linked to various diseases, such as rheumatoid arthritis and hypertension. Despite the availability of crystal structures of P2X receptors, the mechanism of competitive antagonist action for P2X receptors remains controversial. Here, we determine the crystal structure of the chicken P2X7 receptor in complex with the competitive P2X antagonist, TNP-ATP. The structure reveals an expanded, incompletely activated conformation of the channel, and identified the unique recognition manner of TNP-ATP, which is distinct from that observed in the previously determined human P2X3 receptor structure. A structure-based computational analysis furnishes mechanistic insights into the TNP-ATP-dependent inhibition. Our work provides structural insights into the functional mechanism of the P2X competitive antagonist.P2X receptors are nonselective cation channels that are gated by extracellular ATP. Here the authors present the crystal structure of chicken P2X7 with its bound competitive antagonist TNP-ATP and give mechanistic insights into TNP-ATP dependent inhibition through further computational analysis and electrophysiology measurements.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Receptors, Purinergic P2X7/chemistry , Adenosine Triphosphate/chemistry , Animals , Binding Sites , Chickens , Computational Biology , Crystallography, X-Ray , Models, Molecular , Protein Structure, Tertiary , Purinergic P2X Receptor Antagonists , Structure-Activity Relationship
11.
Cell Res ; 26(12): 1288-1301, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27909292

ABSTRACT

Ca2+ release from the sarcoplasmic reticulum (SR) and endoplasmic reticulum (ER) is crucial for muscle contraction, cell growth, apoptosis, learning and memory. The trimeric intracellular cation (TRIC) channels were recently identified as cation channels balancing the SR and ER membrane potentials, and are implicated in Ca2+ signaling and homeostasis. Here we present the crystal structures of prokaryotic TRIC channels in the closed state and structure-based functional analyses of prokaryotic and eukaryotic TRIC channels. Each trimer subunit consists of seven transmembrane (TM) helices with two inverted repeated regions. The electrophysiological, biochemical and biophysical analyses revealed that TRIC channels possess an ion-conducting pore within each subunit, and that the trimer formation contributes to the stability of the protein. The symmetrically related TM2 and TM5 helices are kinked at the conserved glycine clusters, and these kinks are important for the channel activity. Furthermore, the kinks of the TM2 and TM5 helices generate lateral fenestrations at each subunit interface. Unexpectedly, these lateral fenestrations are occupied with lipid molecules. This study provides the structural and functional framework for the molecular mechanism of this ion channel superfamily.


Subject(s)
Archaeal Proteins/chemistry , Bacterial Proteins/chemistry , Ion Channels/chemistry , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Ion Channels/genetics , Ion Channels/metabolism , Microscopy, Fluorescence , Patch-Clamp Techniques , Potassium Chloride/pharmacology , Protein Multimerization , Protein Stability , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Rhodobacter sphaeroides/metabolism , Sulfolobus solfataricus/metabolism , Temperature , Yeasts/drug effects , Yeasts/metabolism
12.
Nature ; 534(7607): 417-20, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27281193

ABSTRACT

The drug/metabolite transporter (DMT) superfamily is a large group of membrane transporters ubiquitously found in eukaryotes, bacteria and archaea, and includes exporters for a remarkably wide range of substrates, such as toxic compounds and metabolites. YddG is a bacterial DMT protein that expels aromatic amino acids and exogenous toxic compounds, thereby contributing to cellular homeostasis. Here we present structural and functional analyses of YddG. Using liposome-based analyses, we show that Escherichia coli and Starkeya novella YddG export various amino acids. The crystal structure of S. novella YddG at 2.4 Å resolution reveals a new membrane transporter topology, with ten transmembrane segments in an outward-facing state. The overall structure is basket-shaped, with a large substrate-binding cavity at the centre of the molecule, and is composed of inverted structural repeats related by two-fold pseudo-symmetry. On the basis of this intramolecular symmetry, we propose a structural model for the inward-facing state and a mechanism of the conformational change for substrate transport, which we confirmed by biochemical analyses. These findings provide a structural basis for the mechanism of transport of DMT superfamily proteins.


Subject(s)
Amino Acid Transport Systems, Neutral/chemistry , Amino Acid Transport Systems, Neutral/metabolism , Amino Acids/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Alphaproteobacteria/chemistry , Alphaproteobacteria/metabolism , Biological Transport , Crystallography, X-Ray , Escherichia coli/chemistry , Escherichia coli/metabolism , Liposomes/chemistry , Liposomes/metabolism , Models, Molecular , Protein Conformation , Structure-Activity Relationship
13.
Cell Rep ; 14(4): 932-944, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26804916

ABSTRACT

P2X receptors are trimeric ATP-gated cation channels involved in physiological processes ranging widely from neurotransmission to pain and taste signal transduction. The modulation of the channel gating, including that by divalent cations, contributes to these diverse physiological functions of P2X receptors. Here, we report the crystal structure of an invertebrate P2X receptor from the Gulf Coast tick Amblyomma maculatum in the presence of ATP and Zn(2+) ion, together with electrophysiological and computational analyses. The structure revealed two distinct metal binding sites, M1 and M2, in the extracellular region. The M1 site, located at the trimer interface, is responsible for Zn(2+) potentiation by facilitating the structural change of the extracellular domain for pore opening. In contrast, the M2 site, coupled with the ATP binding site, might contribute to regulation by Mg(2+). Overall, our work provides structural insights into the divalent cation modulations of P2X receptors.


Subject(s)
Arthropod Proteins/chemistry , Receptors, Purinergic P2X/chemistry , Adenosine Triphosphate/pharmacology , Amino Acid Sequence , Animals , Arthropod Proteins/metabolism , Magnesium/pharmacology , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Binding , Receptors, Purinergic P2X/metabolism , Ticks , Xenopus , Zinc/pharmacology
14.
Cell Rep ; 13(8): 1561-8, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26586438

ABSTRACT

The bacterial SecYEG translocon functions as a conserved protein-conducting channel. Conformational transitions of SecYEG allow protein translocation across the membrane without perturbation of membrane permeability. Here, we report the crystal structures of intact SecYEG at 2.7-Å resolution and of peptide-bound SecYEG at 3.6-Å resolution. The higher-resolution structure revealed that the cytoplasmic loop of SecG covers the hourglass-shaped channel, which was confirmed to also occur in the membrane by disulfide bond formation analysis and molecular dynamics simulation. The cytoplasmic loop may be involved in protein translocation. In addition, the previously unknown peptide-bound crystal structure of SecYEG implies that interactions between the cytoplasmic side of SecY and signal peptides are related to lateral gate opening at the first step of protein translocation. These SecYEG structures therefore provide a number of structural insights into the Sec machinery for further study.


Subject(s)
Escherichia coli Proteins/chemistry , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Protein Transport/physiology , Crystallography, X-Ray/methods , Cytoplasm/metabolism , Escherichia coli Proteins/metabolism , Protein Binding/physiology , Protein Sorting Signals/physiology , Protein Structure, Tertiary , SEC Translocation Channels
15.
PLoS One ; 10(6): e0131094, 2015.
Article in English | MEDLINE | ID: mdl-26114863

ABSTRACT

Channelrhodopsin (ChR) is a light-gated cation channel that responds to blue light. Since ChR can be readily expressed in specific neurons to precisely control their activities by light, it has become a powerful tool in neuroscience. Although the recently solved crystal structure of a chimeric ChR, C1C2, provided the structural basis for ChR, our understanding of the molecular mechanism of ChR still remains limited. Here we performed electrophysiological analyses and all-atom molecular dynamics (MD) simulations, to investigate the importance of the intracellular and central constrictions of the ion conducting pore observed in the crystal structure of C1C2. Our electrophysiological analysis revealed that two glutamate residues, Glu122 and Glu129, in the intracellular and central constrictions, respectively, should be deprotonated in the photocycle. The simulation results suggested that the deprotonation of Glu129 in the central constriction leads to ion leakage in the ground state, and implied that the protonation of Glu129 is important for preventing ion leakage in the ground state. Moreover, we modeled the 13-cis retinal bound; i.e., activated C1C2, and performed MD simulations to investigate the conformational changes in the early stage of the photocycle. Our simulations suggested that retinal photoisomerization induces the conformational change toward channel opening, including the movements of TM6, TM7 and TM2. These insights into the dynamics of the ground states and the early photocycle stages enhance our understanding of the channel function of ChR.


Subject(s)
Ion Channel Gating , Molecular Dynamics Simulation , Retinaldehyde/metabolism , Rhodopsin/chemistry , Rhodopsin/metabolism , Bacteriorhodopsins/chemistry , Bacteriorhodopsins/metabolism , Crystallography, X-Ray , Diterpenes , Electrophysiology , Glutamine/chemistry , Glutamine/genetics , HEK293 Cells , Humans , Models, Molecular , Protein Interaction Domains and Motifs , Retinaldehyde/chemistry
16.
Nature ; 521(7550): 48-53, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25849775

ABSTRACT

Krokinobacter eikastus rhodopsin 2 (KR2) is the first light-driven Na(+) pump discovered, and is viewed as a potential next-generation optogenetics tool. Since the positively charged Schiff base proton, located within the ion-conducting pathway of all light-driven ion pumps, was thought to prohibit the transport of a non-proton cation, the discovery of KR2 raised the question of how it achieves Na(+) transport. Here we present crystal structures of KR2 under neutral and acidic conditions, which represent the resting and M-like intermediate states, respectively. Structural and spectroscopic analyses revealed the gating mechanism, whereby the flipping of Asp116 sequesters the Schiff base proton from the conducting pathway to facilitate Na(+) transport. Together with the structure-based engineering of the first light-driven K(+) pumps, electrophysiological assays in mammalian neurons and behavioural assays in a nematode, our studies reveal the molecular basis for light-driven non-proton cation pumps and thus provide a framework that may advance the development of next-generation optogenetics.


Subject(s)
Flavobacteriaceae/chemistry , Ion Pumps/chemistry , Ion Pumps/radiation effects , Light , Rhodopsin/chemistry , Rhodopsin/radiation effects , Sodium/metabolism , Binding Sites , Crystallography, X-Ray , Hydrogen-Ion Concentration , Ion Pumps/genetics , Ion Pumps/metabolism , Ion Transport/genetics , Ion Transport/radiation effects , Models, Biological , Models, Molecular , Mutagenesis/genetics , Optogenetics , Potassium/metabolism , Protein Conformation , Protein Engineering , Retinaldehyde/chemistry , Retinaldehyde/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism , Schiff Bases , Structure-Activity Relationship
17.
Nature ; 509(7501): 516-20, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24739968

ABSTRACT

Newly synthesized membrane proteins must be accurately inserted into the membrane, folded and assembled for proper functioning. The protein YidC inserts its substrates into the membrane, thereby facilitating membrane protein assembly in bacteria; the homologous proteins Oxa1 and Alb3 have the same function in mitochondria and chloroplasts, respectively. In the bacterial cytoplasmic membrane, YidC functions as an independent insertase and a membrane chaperone in cooperation with the translocon SecYEG. Here we present the crystal structure of YidC from Bacillus halodurans, at 2.4 Å resolution. The structure reveals a novel fold, in which five conserved transmembrane helices form a positively charged hydrophilic groove that is open towards both the lipid bilayer and the cytoplasm but closed on the extracellular side. Structure-based in vivo analyses reveal that a conserved arginine residue in the groove is important for the insertion of membrane proteins by YidC. We propose an insertion mechanism for single-spanning membrane proteins, in which the hydrophilic environment generated by the groove recruits the extracellular regions of substrates into the low-dielectric environment of the membrane.


Subject(s)
Bacillus/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Arginine/metabolism , Cell Membrane/chemistry , Conserved Sequence , Crystallography, X-Ray , Hydrophobic and Hydrophilic Interactions , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Protein Folding , Static Electricity , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...