Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Structure ; 32(7): 866-877.e4, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38626766

ABSTRACT

Trafficking receptors control protein localization through the recognition of specific signal sequences that specify unique cellular locations. Differences in luminal pH are important for the vectorial trafficking of cargo receptors. The KDEL receptor is responsible for maintaining the integrity of the ER by retrieving luminally localized folding chaperones in a pH-dependent mechanism. Structural studies have revealed the end states of KDEL receptor activation and the mechanism of selective cargo binding. However, precisely how the KDEL receptor responds to changes in luminal pH remains unclear. To explain the mechanism of pH sensing, we combine analysis of X-ray crystal structures of the KDEL receptor at neutral and acidic pH with advanced computational methods and cell-based assays. We show a critical role for ordered water molecules that allows us to infer a direct connection between protonation in different cellular compartments and the consequent changes in the affinity of the receptor for cargo.


Subject(s)
Receptors, Peptide , Hydrogen-Ion Concentration , Humans , Receptors, Peptide/metabolism , Receptors, Peptide/chemistry , Receptors, Peptide/genetics , Crystallography, X-Ray , Protein Transport , Protein Binding , Models, Molecular , Endoplasmic Reticulum/metabolism , Binding Sites
2.
Nat Struct Mol Biol ; 30(11): 1786-1793, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37482561

ABSTRACT

In mammals, the kidney plays an essential role in maintaining blood homeostasis through the selective uptake, retention or elimination of toxins, drugs and metabolites. Organic anion transporters (OATs) are responsible for the recognition of metabolites and toxins in the nephron and their eventual urinary excretion. Inhibition of OATs is used therapeutically to improve drug efficacy and reduce nephrotoxicity. The founding member of the renal organic anion transporter family, OAT1 (also known as SLC22A6), uses the export of α-ketoglutarate (α-KG), a key intermediate in the Krebs cycle, to drive selective transport and is allosterically regulated by intracellular chloride. However, the mechanisms linking metabolite cycling, drug transport and intracellular chloride remain obscure. Here, we present cryogenic-electron microscopy structures of OAT1 bound to α-KG, the antiviral tenofovir and clinical inhibitor probenecid, used in the treatment of Gout. Complementary in vivo cellular assays explain the molecular basis for α-KG driven drug elimination and the allosteric regulation of organic anion transport in the kidney by chloride.


Subject(s)
Chlorides , Organic Anion Transport Protein 1 , Animals , Organic Anion Transport Protein 1/metabolism , Chlorides/metabolism , Kidney/metabolism , Biological Transport , Anions/metabolism , Ketoglutaric Acids/metabolism , Mammals/metabolism
3.
Nat Commun ; 13(1): 4845, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35977944

ABSTRACT

Amino acid transporters play a key role controlling the flow of nutrients across the lysosomal membrane and regulating metabolism in the cell. Mutations in the gene encoding the transporter cystinosin result in cystinosis, an autosomal recessive metabolic disorder characterised by the accumulation of cystine crystals in the lysosome. Cystinosin is a member of the PQ-loop family of solute carrier (SLC) transporters and uses the proton gradient to drive cystine export into the cytoplasm. However, the molecular basis for cystinosin function remains elusive, hampering efforts to develop novel treatments for cystinosis and understand the mechanisms of ion driven transport in the PQ-loop family. To address these questions, we present the crystal structures of cystinosin from Arabidopsis thaliana in both apo and cystine bound states. Using a combination of in vitro and in vivo based assays, we establish a mechanism for cystine recognition and proton coupled transport. Mutational mapping and functional characterisation of human cystinosin further provide a framework for understanding the molecular impact of disease-causing mutations.


Subject(s)
Amino Acid Transport Systems, Neutral , Cystinosis , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Biological Transport , Cystine/metabolism , Cystinosis/genetics , Humans , Lysosomes/metabolism , Protons
4.
Mol Cell ; 82(5): 920-932.e7, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35245456

ABSTRACT

IDO1 oxidizes tryptophan (TRP) to generate kynurenine (KYN), the substrate for 1-carbon and NAD metabolism, and is implicated in pro-cancer pathophysiology and infection biology. However, the mechanistic relationships between IDO1 in amino acid depletion versus product generation have remained a longstanding mystery. We found an unrecognized link between IDO1 and cell survival mediated by KYN that serves as the source for molecules that inhibit ferroptotic cell death. We show that this effect requires KYN export from IDO1-expressing cells, which is then available for non-IDO1-expressing cells via SLC7A11, the central transporter involved in ferroptosis suppression. Whether inside the "producer" IDO1+ cell or the "receiver" cell, KYN is converted into downstream metabolites, suppressing ferroptosis by ROS scavenging and activating an NRF2-dependent, AHR-independent cell-protective pathway, including SLC7A11, propagating anti-ferroptotic signaling. IDO1, therefore, controls a multi-pronged protection pathway from ferroptotic cell death, underscoring the need to re-evaluate the use of IDO1 inhibitors in cancer treatment.


Subject(s)
Amino Acid Transport System y+ , Ferroptosis , Kynurenine , Neoplasms , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Kynurenine/metabolism , Kynurenine/pharmacology , Neoplasms/metabolism , Signal Transduction , Tryptophan/metabolism
5.
Nat Commun ; 12(1): 7147, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34880232

ABSTRACT

Cysteine plays an essential role in cellular redox homoeostasis as a key constituent of the tripeptide glutathione (GSH). A rate limiting step in cellular GSH synthesis is the availability of cysteine. However, circulating cysteine exists in the blood as the oxidised di-peptide cystine, requiring specialised transport systems for its import into the cell. System xc- is a dedicated cystine transporter, importing cystine in exchange for intracellular glutamate. To counteract elevated levels of reactive oxygen species in cancerous cells system xc- is frequently upregulated, making it an attractive target for anticancer therapies. However, the molecular basis for ligand recognition remains elusive, hampering efforts to specifically target this transport system. Here we present the cryo-EM structure of system xc- in both the apo and glutamate bound states. Structural comparisons reveal an allosteric mechanism for ligand discrimination, supported by molecular dynamics and cell-based assays, establishing a mechanism for cystine transport in human cells.


Subject(s)
Antiporters/chemistry , Antiporters/metabolism , Cystine/metabolism , Glutamic Acid/metabolism , Glutathione/biosynthesis , Amino Acid Transport System y+/chemistry , Amino Acid Transport System y+/metabolism , Antiporters/genetics , Biochemistry , Cryoelectron Microscopy , Cysteine/metabolism , Fusion Regulatory Protein 1, Heavy Chain/chemistry , Fusion Regulatory Protein 1, Heavy Chain/metabolism , HEK293 Cells , Humans , Neoplasms , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Up-Regulation
6.
Sci Adv ; 7(35)2021 08.
Article in English | MEDLINE | ID: mdl-34433568

ABSTRACT

The SLC15 family of proton-coupled solute carriers PepT1 and PepT2 play a central role in human physiology as the principal route for acquiring and retaining dietary nitrogen. A remarkable feature of the SLC15 family is their extreme substrate promiscuity, which has enabled the targeting of these transporters for the improvement of oral bioavailability for several prodrug molecules. Although recent structural and biochemical studies on bacterial homologs have identified conserved sites of proton and peptide binding, the mechanism of peptide capture and ligand promiscuity remains unclear for mammalian family members. Here, we present the cryo-electron microscopy structure of the outward open conformation of the rat peptide transporter PepT2 in complex with an inhibitory nanobody. Our structure, combined with molecular dynamics simulations and biochemical and cell-based assays, establishes a framework for understanding peptide and prodrug recognition within this pharmaceutically important transporter family.


Subject(s)
Prodrugs , Symporters , Animals , Cryoelectron Microscopy , Mammals/metabolism , Membrane Transport Proteins/metabolism , Peptide Transporter 1/chemistry , Peptides/metabolism , Protons , Rats
7.
Structure ; 29(10): 1182-1191.e4, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34242558

ABSTRACT

Tuberculosis (TB) is the leading cause of death from a single infectious agent and in 2019 an estimated 10 million people worldwide contracted the disease. Although treatments for TB exist, continual emergence of drug-resistant variants necessitates urgent development of novel antituberculars. An important new target is the lipid transporter MmpL3, which is required for construction of the unique cell envelope that shields Mycobacterium tuberculosis (Mtb) from the immune system. However, a structural understanding of the mutations in Mtb MmpL3 that confer resistance to the many preclinical leads is lacking, hampering efforts to circumvent resistance mechanisms. Here, we present the cryoelectron microscopy structure of Mtb MmpL3 and use it to comprehensively analyze the mutational landscape of drug resistance. Our data provide a rational explanation for resistance variants local to the central drug binding site, and also highlight a potential alternative route to resistance operating within the periplasmic domain.


Subject(s)
Bacterial Proteins/chemistry , Drug Resistance, Bacterial , Membrane Transport Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cryoelectron Microscopy , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mutation
8.
Elife ; 102021 06 17.
Article in English | MEDLINE | ID: mdl-34137369

ABSTRACT

ER proteins of widely differing abundance are retrieved from the Golgi by the KDEL-receptor. Abundant ER proteins tend to have KDEL rather than HDEL signals, whereas ADEL and DDEL are not used in most organisms. Here, we explore the mechanism of selective retrieval signal capture by the KDEL-receptor and how HDEL binds with 10-fold higher affinity than KDEL. Our results show the carboxyl-terminus of the retrieval signal moves along a ladder of arginine residues as it enters the binding pocket of the receptor. Gatekeeper residues D50 and E117 at the entrance of this pocket exclude ADEL and DDEL sequences. D50N/E117Q mutation of human KDEL-receptors changes the selectivity to ADEL and DDEL. However, further analysis of HDEL, KDEL, and RDEL-bound receptor structures shows that affinity differences are explained by interactions between the variable -4 H/K/R position of the signal and W120, rather than D50 or E117. Together, these findings explain KDEL-receptor selectivity, and how signal variants increase dynamic range to support efficient ER retrieval of low and high abundance proteins.


Subject(s)
Endoplasmic Reticulum/metabolism , Receptors, Peptide , Golgi Apparatus/metabolism , Humans , Mutation/genetics , Protein Sorting Signals/genetics , Protein Transport/genetics , Receptors, Peptide/chemistry , Receptors, Peptide/genetics , Receptors, Peptide/metabolism
9.
Nature ; 595(7865): 130-134, 2021 07.
Article in English | MEDLINE | ID: mdl-34040256

ABSTRACT

Folates (also known as vitamin B9) have a critical role in cellular metabolism as the starting point in the synthesis of nucleic acids, amino acids and the universal methylating agent S-adenylsmethionine1,2. Folate deficiency is associated with a number of developmental, immune and neurological disorders3-5. Mammals cannot synthesize folates de novo; several systems have therefore evolved to take up folates from the diet and distribute them within the body3,6. The proton-coupled folate transporter (PCFT) (also known as SLC46A1) mediates folate uptake across the intestinal brush border membrane and the choroid plexus4,7, and is an important route for the delivery of antifolate drugs in cancer chemotherapy8-10. How PCFT recognizes folates or antifolate agents is currently unclear. Here we present cryo-electron microscopy structures of PCFT in a substrate-free state and in complex with a new-generation antifolate drug (pemetrexed). Our results provide a structural basis for understanding antifolate recognition and provide insights into the pH-regulated mechanism of folate transport mediated by PCFT.


Subject(s)
Cryoelectron Microscopy , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/metabolism , Pemetrexed/chemistry , Pemetrexed/metabolism , Proton-Coupled Folate Transporter/chemistry , Proton-Coupled Folate Transporter/metabolism , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Biological Transport , Humans , Models, Molecular , Proton-Coupled Folate Transporter/ultrastructure , Protons
10.
Nat Commun ; 10(1): 4657, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31604945

ABSTRACT

Nucleotide sugars are the activated form of monosaccharides used by glycosyltransferases during glycosylation. In eukaryotes the SLC35 family of solute carriers are responsible for their selective uptake into the Endoplasmic Reticulum or Golgi apparatus. The structure of the yeast GDP-mannose transporter, Vrg4, revealed a requirement for short chain lipids and a marked difference in transport rate between the nucleotide sugar and nucleoside monophosphate, suggesting a complex network of regulatory elements control transport into these organelles. Here we report the crystal structure of the GMP bound complex of Vrg4, revealing the molecular basis for GMP recognition and transport. Molecular dynamics, combined with biochemical analysis, reveal a lipid mediated dimer interface and mechanism for coordinating structural rearrangements during transport. Together these results provide further insight into how SLC35 family transporters function within the secretory pathway and sheds light onto the role that membrane lipids play in regulating transport across the membrane.


Subject(s)
Guanosine Monophosphate/chemistry , Lipid Bilayers/metabolism , Membrane Transport Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Binding Sites , Guanosine Monophosphate/metabolism , Membrane Transport Proteins/metabolism , Models, Chemical , Protein Domains , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity
11.
Curr Opin Struct Biol ; 57: 127-134, 2019 08.
Article in English | MEDLINE | ID: mdl-30999236

ABSTRACT

The Golgi apparatus plays a central role in the secretory pathway as a hub for posttranslational modification, protein sorting and quality control. To date, there is little structural or biochemical information concerning the function of transporters that reside within this organelle. The SLC35 family of nucleotide sugar transporters link the synthesis of activated sugar molecules and sulfate in the cytoplasm, with the luminal transferases that catalyse their attachment to proteins and lipids during glycosylation and sulfation. A recent crystal structure of the GDP-mannose transporter has revealed key sequence motifs that direct ligand recognition and transport. Further biochemical studies unexpectedly found a requirement for short chain lipids in activating the transporter, suggesting a possible route for transport regulation within the Golgi.


Subject(s)
Golgi Apparatus/metabolism , Membrane Transport Proteins/metabolism , Nucleotides/metabolism , Sugars/metabolism , Animals , Humans , Membrane Transport Proteins/chemistry
12.
Science ; 363(6431): 1103-1107, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30846601

ABSTRACT

Selective export and retrieval of proteins between the endoplasmic reticulum (ER) and Golgi apparatus is indispensable for eukaryotic cell function. An essential step in the retrieval of ER luminal proteins from the Golgi is the pH-dependent recognition of a carboxyl-terminal Lys-Asp-Glu-Leu (KDEL) signal by the KDEL receptor. Here, we present crystal structures of the chicken KDEL receptor in the apo ER state, KDEL-bound Golgi state, and in complex with an antagonistic synthetic nanobody (sybody). These structures show a transporter-like architecture that undergoes conformational changes upon KDEL binding and reveal a pH-dependent interaction network crucial for recognition of the carboxyl terminus of the KDEL signal. Complementary in vitro binding and in vivo cell localization data explain how these features create a pH-dependent retrieval system in the secretory pathway.


Subject(s)
Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Receptors, Peptide/chemistry , Animals , Chickens , Crystallography, X-Ray , Hydrogen-Ion Concentration , Mice , Protein Conformation , Receptors, Peptide/metabolism
13.
Nat Commun ; 9(1): 550, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416041

ABSTRACT

Amino acids play essential roles in cell biology as regulators of metabolic pathways. Arginine in particular is a major signalling molecule inside the cell, being a precursor for both l-ornithine and nitric oxide (NO) synthesis and a key regulator of the mTORC1 pathway. In mammals, cellular arginine availability is determined by members of the solute carrier (SLC) 7 family of cationic amino acid transporters. Whereas CAT-1 functions to supply cationic amino acids for cellular metabolism, CAT-2A and -2B are required for macrophage activation and play important roles in regulating inflammation. Here, we present the crystal structure of a close homologue of the mammalian CAT transporters that reveals how these proteins specifically recognise arginine. Our structural and functional data provide a model for cationic amino acid transport in mammalian cells and reveals mechanistic insights into proton-coupled, sodium-independent amino acid transport in the wider APC superfamily.


Subject(s)
Amino Acid Transport Systems, Basic/chemistry , Amino Acid Transport Systems, Basic/metabolism , Animals , Arginine/metabolism , Crystallization , Crystallography, X-Ray , Nitric Oxide/biosynthesis , Ornithine/biosynthesis
14.
Nature ; 551(7681): 521-524, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29143814

ABSTRACT

Glycosylation is a fundamental cellular process that, in eukaryotes, occurs in the lumen of both the Golgi apparatus and the endoplasmic reticulum. Nucleotide sugar transporters (NSTs) are an essential component of the glycosylation pathway, providing the diverse range of substrates required for the glycosyltransferases. NSTs are linked to several developmental and immune disorders in humans, and in pathogenic microbes they have an important role in virulence. How NSTs recognize and transport activated monosaccharides, however, is currently unclear. Here we present the crystal structure of an NST, the GDP-mannose transporter Vrg4, in both the substrate-free and the bound states. A hitherto unobserved requirement of short-chain lipids in activating the transporter supports a model for regulation within the highly dynamic membranes of the Golgi apparatus. Our results provide a structural basis for understanding nucleotide sugar recognition, and provide insights into the transport and regulatory mechanism of this family of intracellular transporters.


Subject(s)
Golgi Apparatus/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Monosaccharides/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/cytology , Biological Transport , Crystallography, X-Ray , Glycosylation , Structure-Activity Relationship , Substrate Specificity
15.
Proc Natl Acad Sci U S A ; 114(50): 13182-13187, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29180426

ABSTRACT

POT transporters represent an evolutionarily well-conserved family of proton-coupled transport systems in biology. An unusual feature of the family is their ability to couple the transport of chemically diverse ligands to an inwardly directed proton electrochemical gradient. For example, in mammals, fungi, and bacteria they are predominantly peptide transporters, whereas in plants the family has diverged to recognize nitrate, plant defense compounds, and hormones. Although recent structural and biochemical studies have identified conserved sites of proton binding, the mechanism through which transport is coupled to proton movement remains enigmatic. Here we show that different POT transporters operate through distinct proton-coupled mechanisms through changes in the extracellular gate. A high-resolution crystal structure reveals the presence of ordered water molecules within the peptide binding site. Multiscale molecular dynamics simulations confirm proton transport occurs through these waters via Grotthuss shuttling and reveal that proton binding to the extracellular side of the transporter facilitates a reorientation from an inward- to outward-facing state. Together these results demonstrate that within the POT family multiple mechanisms of proton coupling have likely evolved in conjunction with variation of the extracellular gate.


Subject(s)
Bacterial Proteins/chemistry , Membrane Transport Proteins/chemistry , Peptides/metabolism , Protons , Bacterial Proteins/metabolism , Binding Sites , Membrane Transport Proteins/metabolism , Molecular Dynamics Simulation , Protein Binding , Xanthomonas/chemistry , Xanthomonas/metabolism
16.
Adv Exp Med Biol ; 922: 61-72, 2016.
Article in English | MEDLINE | ID: mdl-27553235

ABSTRACT

Alpha helical membrane proteins are the targets for many pharmaceutical drugs and play important roles in physiology and disease processes. In recent years, substantial progress has been made in determining their atomic structure using X-ray crystallography. However, a major bottleneck still remains; the identification of conditions that give crystals that are suitable for structure determination. Over the past 10 years we have been analysing the crystallisation conditions reported for alpha helical membrane proteins with the aim to facilitate a rational approach to the design and implementation of successful crystallisation screens. The result has been the development of MemGold, MemGold2 and the additive screen MemAdvantage. The associated analysis, summarised and updated in this chapter, has revealed a number of surprisingly successfully strategies for crystallisation and detergent selection.


Subject(s)
Membrane Proteins/chemistry , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Buffers , Chemical Precipitation , Crystallization , Crystallography, X-Ray , Detergents/pharmacology , Humans , Hydrogen-Ion Concentration , Membrane Proteins/isolation & purification , Protein Structure, Secondary , Salts/chemistry
17.
Cell Chem Biol ; 23(2): 299-309, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-27028887

ABSTRACT

Membrane transporters are critical modulators of drug pharmacokinetics, efficacy, and safety. One example is the proton-dependent oligopeptide transporter PepT1, also known as SLC15A1, which is responsible for the uptake of the ?-lactam antibiotics and various peptide-based prodrugs. In this study, we modeled the binding of various peptides to a bacterial homolog, PepT(St), and evaluated a range of computational methods for predicting the free energy of binding. Our results show that a hybrid approach (endpoint methods to classify peptides into good and poor binders and a theoretically exact method for refinement) is able to accurately predict affinities, which we validated using proteoliposome transport assays. Applying the method to a homology model of PepT1 suggests that the approach requires a high-quality structure to be accurate. Our study provides a blueprint for extending these computational methodologies to other pharmaceutically important transporter families.


Subject(s)
Anti-Bacterial Agents/metabolism , Oligopeptides/metabolism , Prodrugs/metabolism , Symporters/metabolism , Anti-Bacterial Agents/chemistry , Humans , Ligands , Molecular Dynamics Simulation , Oligopeptides/chemistry , Peptide Transporter 1 , Prodrugs/chemistry , Symporters/chemistry , Thermodynamics
18.
Structure ; 23(10): 1889-1899, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26320580

ABSTRACT

Mammals obtain nitrogen via the uptake of di- and tri-peptides in the gastrointestinal tract through the action of PepT1 and PepT2, which are members of the POT family of proton-coupled oligopeptide transporters. PepT1 and PepT2 also play an important role in drug transport in the human body. Recent crystal structures of bacterial homologs revealed a conserved peptide-binding site and mechanism of transport. However, a key structural difference exists between bacterial and mammalian homologs with only the latter containing a large extracellular domain, the function of which is currently unknown. Here, we present the crystal structure of the extracellular domain from both PepT1 and PepT2 that reveal two immunoglobulin-like folds connected in tandem, providing structural insight into mammalian peptide transport. Functional and biophysical studies demonstrate that these domains interact with the intestinal protease trypsin, suggesting a role in clustering proteolytic activity to the site of peptide transport in eukaryotic cells.


Subject(s)
Oligopeptides/chemistry , Symporters/chemistry , Trypsin/chemistry , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Oligopeptides/chemical synthesis , Peptide Transporter 1 , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Symporters/genetics , Symporters/metabolism , Trypsin/genetics , Trypsin/metabolism
19.
Structure ; 23(7): 1350-61, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26073602

ABSTRACT

There has been exponential growth in the number of membrane protein structures determined. Nevertheless, these structures are usually resolved in the absence of their lipid environment. Coarse-grained molecular dynamics (CGMD) simulations enable insertion of membrane proteins into explicit models of lipid bilayers. We have automated the CGMD methodology, enabling membrane protein structures to be identified upon their release into the PDB and embedded into a membrane. The simulations are analyzed for protein-lipid interactions, identifying lipid binding sites, and revealing local bilayer deformations plus molecular access pathways within the membrane. The coarse-grained models of membrane protein/bilayer complexes are transformed to atomistic resolution for further analysis and simulation. Using this automated simulation pipeline, we have analyzed a number of recently determined membrane protein structures to predict their locations within a membrane, their lipid/protein interactions, and the functional implications of an enhanced understanding of the local membrane environment of each protein.


Subject(s)
Lipid Bilayers/chemistry , Membrane Proteins/chemistry , Molecular Dynamics Simulation , Binding Sites , Hydrophobic and Hydrophilic Interactions , Protein Conformation
20.
Elife ; 32014 Dec 02.
Article in English | MEDLINE | ID: mdl-25457052

ABSTRACT

Peptide transport plays an important role in cellular homeostasis as a key route for nitrogen acquisition in mammalian cells. PepT1 and PepT2, the mammalian proton coupled peptide transporters (POTs), function to assimilate and retain diet-derived peptides and play important roles in drug pharmacokinetics. A key characteristic of the POT family is the mechanism of peptide selectivity, with members able to recognise and transport >8000 different peptides. In this study, we present thermodynamic evidence that in the bacterial POT family transporter PepTSt, from Streptococcus thermophilus, at least two alternative transport mechanisms operate to move peptides into the cell. Whilst tri-peptides are transported with a proton:peptide stoichiometry of 3:1, di-peptides are co-transported with either 4 or 5 protons. This is the first thermodynamic study of proton:peptide stoichiometry in the POT family and reveals that secondary active transporters can evolve different coupling mechanisms to accommodate and transport chemically and physically diverse ligands across the membrane.


Subject(s)
Membrane Transport Proteins/metabolism , Thermodynamics , Bacterial Proteins/metabolism , Protein Transport , Streptococcus thermophilus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...