Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 585
Filter
1.
Clin Chim Acta ; 561: 119765, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38852790

ABSTRACT

BACKGROUND AND AIMS: Cystic fibrosis (CF) is an autosomal recessive disease caused by mutations to the CF transmembrane conductance regulator (CFTR). Symptoms and severity of the disease can be quite variable suggesting modifier genes play an important role. MATERIALS AND METHODS: Exome sequencing was performed on six individuals carrying homozygous deltaF508 for CFTR genotype but present with rapidly progressing CF (RPCF). Data was analyzed using an unbiased genome-wide genetic burden test against 3076 controls. Single cell RNA sequencing data from LungMAP was utilized to evaluate unique and co-expression of candidate genes, and structural modeling to evaluate the deleterious effects of identified candidate variants. RESULTS: We have identified solute carrier family 26 member 9 (SLC26A9) as a modifier gene to be associated with RPCF. Two rare missense SLC26A9 variants were discovered in three of six individuals deemed to have RPCF: c.229G > A; p.G77S (present in two patients), and c.1885C > T; p.P629S. Co-expression of SLC26A9 and CFTR mRNA is limited across different lung cell types, with the highest level of co-expression seen in human (6.3 %) and mouse (9.0 %) alveolar type 2 (AT2) cells. Structural modeling suggests deleterious effects of these mutations as they are in critical protein domains which might affect the anion transport capability of SLC26A9. CONCLUSION: The enrichment of rare and potentially deleterious SLC26A9 mutations in patients with RPCF suggests SLC26A9 may act as an alternative anion transporter in CF and is a modifier gene associated with this lung phenotype.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Homozygote , Mutation , Sulfate Transporters , Humans , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Sulfate Transporters/genetics , Sulfate Transporters/chemistry , Sulfate Transporters/metabolism , Female , Male , Antiporters/genetics , Antiporters/chemistry , Animals , Mice
2.
Biophys J ; 123(12): 1751-1762, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38773769

ABSTRACT

The anion exchanger solute carrier family 26 (SLC26)A9, consisting of the transmembrane (TM) domain and the cytoplasmic STAS domain, plays an essential role in regulating chloride transport across cell membranes. Recent studies have indicated that C-terminal helices block the entrance of the putative ion transport pathway. However, the precise functions of the STAS domain and C-terminal helix, as well as the underlying molecular mechanisms governing the transport process, remain poorly understood. In this study, we performed molecular dynamics simulations of three distinct models of human SLC26A9, full-length, STAS domain removal (ΔSTAS), and C-terminus removal (ΔC), to investigate their conformational dynamics and ion-binding properties. Stable binding of ions to the binding sites was exclusively observed in the ΔC model in these simulations. Comparing the full-length and ΔC simulations, the ΔC model displayed enhanced motion of the STAS domain. Furthermore, comparing the ΔSTAS and ΔC simulations, the ΔSTAS simulation failed to exhibit stable ion bindings to the sites despite the absence of the C-terminus blocking the ion transmission pathway in both systems. These results suggest that the removal of the C-terminus not only unblocks the access of ions to the permeation pathway but also triggers STAS domain motion, gating the TM domain to promote ions' entry into their binding site. Further analysis revealed that the asymmetric motion of the STAS domain leads to the expansion of the ion permeation pathway within the TM domain, resulting in the stiffening of the flexible TM12 helix near the ion-binding site. This structural change in the TM12 helix stabilizes chloride ion binding, which is essential for SLC26A9's alternate-access mechanism. Overall, our study provides new insights into the molecular mechanisms of SLC26A9 transport and may pave the way for the development of novel treatments for diseases associated with dysregulated ion transport.


Subject(s)
Antiporters , Chlorides , Molecular Dynamics Simulation , Protein Domains , Sulfate Transporters , Sulfate Transporters/metabolism , Sulfate Transporters/chemistry , Sulfate Transporters/genetics , Humans , Chlorides/metabolism , Antiporters/chemistry , Antiporters/metabolism , Antiporters/genetics , Ion Transport , Binding Sites , Protein Binding
3.
Nat Commun ; 15(1): 4537, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806470

ABSTRACT

The multidrug efflux transporter EmrE from Escherichia coli requires anionic residues in the substrate binding pocket for coupling drug transport with the proton motive force. Here, we show how protonation of a single membrane embedded glutamate residue (Glu14) within the homodimer of EmrE modulates the structure and dynamics in an allosteric manner using NMR spectroscopy. The structure of EmrE in the Glu14 protonated state displays a partially occluded conformation that is inaccessible for drug binding by the presence of aromatic residues in the binding pocket. Deprotonation of a single Glu14 residue in one monomer induces an equilibrium shift toward the open state by altering its side chain position and that of a nearby tryptophan residue. This structural change promotes an open conformation that facilitates drug binding through a conformational selection mechanism and increases the binding affinity by approximately 2000-fold. The prevalence of proton-coupled exchange in efflux systems suggests a mechanism that may be shared in other antiporters where acid/base chemistry modulates access of drugs to the substrate binding pocket.


Subject(s)
Antiporters , Escherichia coli Proteins , Escherichia coli , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Antiporters/metabolism , Antiporters/chemistry , Antiporters/genetics , Binding Sites , Protein Binding , Protons , Protein Conformation , Magnetic Resonance Spectroscopy , Glutamic Acid/metabolism , Glutamic Acid/chemistry , Models, Molecular
4.
J Biol Chem ; 300(5): 107261, 2024 May.
Article in English | MEDLINE | ID: mdl-38582450

ABSTRACT

Mammalian SLC26 proteins are membrane-based anion transporters that belong to the large SLC26/SulP family, and many of their variants are associated with hereditary diseases. Recent structural studies revealed a strikingly similar homodimeric molecular architecture for several SLC26 members, implying a shared molecular principle. Now a new question emerges as to how these structurally similar proteins execute diverse physiological functions. In this study, we sought to identify the common versus distinct molecular mechanism among the SLC26 proteins using both naturally occurring and artificial missense changes introduced to SLC26A4, SLC26A5, and SLC26A9. We found: (i) the basic residue at the anion binding site is essential for both anion antiport of SLC26A4 and motor functions of SLC26A5, and its conversion to a nonpolar residue is crucial but not sufficient for the fast uncoupled anion transport in SLC26A9; (ii) the conserved polar residues in the N- and C-terminal cytosolic domains are likely involved in dynamic hydrogen-bonding networks and are essential for anion antiport of SLC26A4 but not for motor (SLC26A5) and uncoupled anion transport (SLC26A9) functions; (iii) the hydrophobic interaction between each protomer's last transmembrane helices, TM14, is not of functional significance in SLC26A9 but crucial for the functions of SLC26A4 and SLC26A5, likely contributing to optimally orient the axis of the relative movements of the core domain with respect to the gate domains within the cell membrane. These findings advance our understanding of the molecular mechanisms underlying the diverse physiological roles of the SLC26 family of proteins.


Subject(s)
Antiporters , Sulfate Transporters , Animals , Humans , Antiporters/metabolism , Antiporters/genetics , Antiporters/chemistry , Binding Sites , HEK293 Cells , Hydrogen Bonding , Models, Molecular , Mutation, Missense , Protein Domains , Protein Structure, Quaternary , Protein Structure, Tertiary , Sulfate Transporters/metabolism , Sulfate Transporters/genetics , Sulfate Transporters/chemistry , Protein Multimerization , Protein Structure, Secondary
5.
J Phys Chem B ; 128(11): 2697-2706, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38447081

ABSTRACT

CLCF fluoride/proton antiporters move fluoride ions out of bacterial cells, leading to fluoride resistance in these bacteria. However, many details about their operating mechanisms remain unclear. Here, we report a combined quantum-mechanical/molecular-mechanical (QM/MM) study of a CLCF homologue from Enterococci casseliflavus (Eca), in accord with the previously proposed windmill mechanism. Our multiscale modeling sheds light on two critical steps in the transport cycle: (i) the external gating residue E118 pushing a fluoride in the external binding site into the extracellular vestibule and (ii) an incoming fluoride reconquering the external binding site by forcing out E118. Both steps feature competitions for the external binding site between the negatively charged carboxylate of E118 and the fluoride. Remarkably, the displaced E118 by fluoride accepts a proton from the nearby R117, initiating the next transport cycle. We also demonstrate the importance of accurate quantum descriptions of fluoride solvation. Our results provide clues to the mysterious E318 residue near the central binding site, suggesting that the transport activities are unlikely to be disrupted by the glutamate interacting with a well-solvated fluoride at the central binding site. This differs significantly from the structurally similar CLC chloride/proton antiporters, where a fluoride trapped deep in the hydrophobic pore causes the transporter to be locked down. A free-energy barrier of 10-15 kcal/mol was estimated via umbrella sampling for a fluoride ion traveling through the pore to repopulate the external binding site.


Subject(s)
Antiporters , Protons , Antiporters/chemistry , Antiporters/metabolism , Fluorides/chemistry , Models, Molecular , Membrane Transport Proteins/metabolism , Chlorides/chemistry , Chloride Channels/chemistry , Chloride Channels/metabolism , Ion Transport
6.
Nat Struct Mol Biol ; 31(4): 644-656, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38279055

ABSTRACT

CLCs are dimeric chloride channels and anion/proton exchangers that regulate processes such as muscle contraction and endo-lysosome acidification. Common gating controls their activity; its closure simultaneously silences both protomers, and its opening allows them to independently transport ions. Mutations affecting common gating in human CLCs cause dominant genetic disorders. The structural rearrangements underlying common gating are unknown. Here, using single-particle cryo-electron microscopy, we show that the prototypical Escherichia coli CLC-ec1 undergoes large-scale rearrangements in activating conditions. The slow, pH-dependent remodeling of the dimer interface leads to the concerted opening of the intracellular H+ pathways and is required for transport. The more frequent formation of short water wires in the open H+ pathway enables Cl- pore openings. Mutations at disease-causing sites favor CLC-ec1 activation and accelerate common gate opening in the human CLC-7 exchanger. We suggest that the pH activation mechanism of CLC-ec1 is related to the common gating of CLC-7.


Subject(s)
Escherichia coli Proteins , Protons , Humans , Cryoelectron Microscopy , Ions/metabolism , Chloride Channels/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Hydrogen-Ion Concentration , Antiporters/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism
7.
J Microbiol Biotechnol ; 33(7): 857-863, 2023 Jul 28.
Article in English | MEDLINE | ID: mdl-37100762

ABSTRACT

Pathogenic bacteria that colonize the human intestinal tract have evolved strategies to overcome acidic conditions when they pass through the gastrointestinal tract. Amino acid-mediated acid resistance systems are effective survival strategies in a stomach that is full of amino acid substrate. The amino acid antiporter, amino acid decarboxylase, and ClC chloride antiporter are all engaged in these systems, and each one plays a role in protecting against or adapting to the acidic environment. The ClC chloride antiporter, a member of the ClC channel family, eliminates negatively charged intracellular chloride ions to avoid inner membrane hyperpolarization as an electrical shunt of the acid resistance system. In this review, we will discuss the structure and function of the prokaryotic ClC chloride antiporter of amino acid-mediated acid resistance system.


Subject(s)
Antiporters , Chlorides , Humans , Chlorides/metabolism , Antiporters/chemistry , Antiporters/metabolism , Amino Acids , Gram-Negative Bacteria/metabolism , Chloride Channels/chemistry , Chloride Channels/metabolism
8.
Acta Biochim Biophys Sin (Shanghai) ; 55(4): 683-690, 2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37097058

ABSTRACT

MgtE is a Mg 2+-selective channel regulated by the intracellular Mg 2+ concentration. MgtE family proteins are highly conserved in all domains of life and contribute to cellular Mg 2+ homeostasis. In humans, mutations in the SLC41 proteins, the eukaryotic counterparts of the bacterial MgtE, are known to be associated with various diseases. The first MgtE structure from a thermophilic bacterium, Thermus thermophilus, revealed that MgtE forms a homodimer consisting of transmembrane and cytoplasmic domains with a plug helix connecting the two and that the cytoplasmic domain possesses multiple Mg 2+ binding sites. Structural and electrophysiological analyses revealed that the dissociation of Mg 2+ ions from the cytoplasmic domain induces structural changes in the cytoplasmic domain, leading to channel opening. Thus, previous works showed the importance of MgtE cytoplasmic Mg 2+ binding sites. Nevertheless, due to the limited structural information on MgtE from different species, the conservation and diversity of the cytoplasmic Mg 2+ binding site in MgtE family proteins remain unclear. Here, we report crystal structures of the Mg 2+-bound MgtE cytoplasmic domains from two different bacterial species, Chryseobacterium hispalense and Clostridiales bacterium, and identify multiple Mg 2+ binding sites, including ones that were not observed in the previous MgtE structure. These structures reveal the conservation and diversity of the cytoplasmic Mg 2+ binding site in the MgtE family proteins.


Subject(s)
Antiporters , Bacterial Proteins , Humans , Antiporters/chemistry , Antiporters/genetics , Antiporters/metabolism , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Models, Molecular , X-Rays , Thermus thermophilus
9.
J Chem Inf Model ; 63(8): 2445-2455, 2023 04 24.
Article in English | MEDLINE | ID: mdl-37053383

ABSTRACT

Fluoride is a natural antibiotic abundantly present in the environment and, in micromolar concentrations, is able to inhibit enzymes necessary for bacteria to survive. However, as is the case with many antibiotics, bacteria have evolved resistance methods, including through the use of recently discovered membrane proteins. One such protein is the CLCF F-/H+ antiporter protein, a member of the CLC superfamily of anion-transport proteins. Though previous studies have examined this F- transporter, many questions are still left unanswered. To reveal details of the transport mechanism used by CLCF, we have employed molecular dynamics simulations and umbrella sampling calculations. Our results have led to several discoveries, including the mechanism of proton import and how it is able to aid in the fluoride export. Additionally, we have determined the role of the previously identified residues Glu118, Glu318, Met79, and Tyr396. This work is among the first studies of the CLCF F-/H+ antiporter and is the first computational investigation to model the full transport process, proposing a mechanism which couples the F- export with the H+ import.


Subject(s)
Antiporters , Fluorides , Antiporters/chemistry , Antiporters/metabolism , Fluorides/chemistry , Protons , Ion Transport , Membrane Transport Proteins/metabolism
10.
Eur J Med Chem ; 249: 115149, 2023 Mar 05.
Article in English | MEDLINE | ID: mdl-36724632

ABSTRACT

The anion exchanger protein SLC26A3 (down-regulated in adenoma, DRA) is expressed in the luminal membrane of intestinal epithelial cells in colon, where it facilitates the absorption of Cl- and oxalate. We previously identified a 4,8-dimethylcoumarin class of SLC26A3 inhibitors that act from the SLC26A3 cytoplasmic surface, and demonstrated their efficacy in mouse models of constipation and hyperoxaluria. Here, screening of 50,000 new compounds and 1740 chemical analogs of active compounds from the primary screen produced five novel classes of SLC26A3-selective inhibitors (1,3-dioxoisoindoline-amides; N-(5-sulfamoyl-1,3,4-thiadiazol-2-yl)acetamides; thiazolo-pyrimidin-5-ones; 3-carboxy-2-phenylbenzofurans and benzoxazin-4-ones) with IC50 down to 100 nM. Kinetic washout and onset of action studies revealed an extracellular site of action for the thiazolo-pyrimidin-5-one and 3-carboxy-2-phenylbenzofuran inhibitors. Molecular docking computations revealed putative binding sites for these inhibitors. In a loperamide model of constipation in mice, orally administered 7-(2-chloro-phenoxymethyl)-3-phenyl-thiazolo [3,2-a]pyrimidin-5-one (3a) significantly increased stool weight, pellet number and water content. SLC26A3 inhibitors with an extracellular site of action offer the possibility of creating non-absorbable, luminally acting inhibitors with minimal systemic exposure following oral administration. Our findings also suggest that inhibitors of related SLC26 anion transporters with an extracellular site of action might be identified for pharmacological modulation of selected epithelial ion transport processes.


Subject(s)
Antiporters , Constipation , Mice , Animals , Antiporters/chemistry , Antiporters/metabolism , Antiporters/pharmacology , Molecular Docking Simulation , Biological Transport , Anions , Chlorides/metabolism , Sulfate Transporters/metabolism
11.
J Biol Chem ; 299(2): 102805, 2023 02.
Article in English | MEDLINE | ID: mdl-36529287

ABSTRACT

EmrE, a small multidrug resistance transporter from Escherichia coli, confers broad-spectrum resistance to polyaromatic cations and quaternary ammonium compounds. Previous transport assays demonstrate that EmrE transports a +1 and a +2 substrate with the same stoichiometry of two protons:one cationic substrate. This suggests that EmrE substrate binding capacity is limited to neutralization of the two essential glutamates, E14A and E14B (one from each subunit in the antiparallel homodimer), in the primary binding site. Here, we explicitly test this hypothesis, since EmrE has repeatedly broken expectations for membrane protein structure and transport mechanism. We previously showed that EmrE can bind a +1 cationic substrate and proton simultaneously, with cationic substrate strongly associated with one E14 residue, whereas the other remains accessible to bind and transport a proton. Here, we demonstrate that EmrE can bind a +2 cation substrate and a proton simultaneously using NMR pH titrations of EmrE saturated with divalent substrates, for a net +1 charge in the transport pore. Furthermore, we find that EmrE can alternate access and transport a +2 substrate and proton at the same time. Together, these results lead us to conclude that E14 charge neutralization does not limit the binding and transport capacity of EmrE.


Subject(s)
Antiporters , Catalytic Domain , Escherichia coli Proteins , Escherichia coli , Glutamates , Static Electricity , Antiporters/chemistry , Antiporters/metabolism , Escherichia coli/chemistry , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Glutamates/chemistry , Glutamates/metabolism , Protons , Substrate Specificity , Protein Binding , Nuclear Magnetic Resonance, Biomolecular , Hydrogen-Ion Concentration , Drug Resistance, Multiple, Bacterial , Ion Transport
12.
Nat Commun ; 13(1): 7655, 2022 Dec 10.
Article in English | MEDLINE | ID: mdl-36496486

ABSTRACT

Small multidrug resistance (SMR) transporters contribute to antibiotic resistance through proton-coupled efflux of toxic compounds. Previous biophysical studies of the E. coli SMR transporter EmrE suggest that it should also be able to perform proton/toxin symport or uniport, leading to toxin susceptibility rather than resistance in vivo. Here we show EmrE does confer susceptibility to several previously uncharacterized small-molecule substrates in E. coli, including harmane. In vitro electrophysiology assays demonstrate that harmane binding triggers uncoupled proton flux through EmrE. Assays in E. coli are consistent with EmrE-mediated dissipation of the transmembrane pH gradient as the mechanism underlying the in vivo phenotype of harmane susceptibility. Furthermore, checkerboard assays show this alternative EmrE transport mode can synergize with some existing antibiotics, such as kanamycin. These results demonstrate that it is possible to not just inhibit multidrug efflux, but to activate alternative transport modes detrimental to bacteria, suggesting a strategy to address antibiotic resistance.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Antiporters/chemistry , Protons , Drug Resistance, Multiple , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 119(34): e2206129119, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35969794

ABSTRACT

The Amino Acid-Polyamine-Organocation (APC) transporter GadC contributes to the survival of pathogenic bacteria under extreme acid stress by exchanging extracellular glutamate for intracellular γ-aminobutyric acid (GABA). Its structure, determined in an inward-facing conformation at alkaline pH, consists of the canonical LeuT-fold with a conserved five-helix inverted repeat, thereby resembling functionally divergent transporters such as the serotonin transporter SERT and the glucose-sodium symporter SGLT1. However, despite this structural similarity, it is unclear if the conformational dynamics of antiporters such as GadC follow the blueprint of these or other LeuT-fold transporters. Here, we used double electron-electron resonance (DEER) spectroscopy to monitor the conformational dynamics of GadC in lipid bilayers in response to acidification and substrate binding. To guide experimental design and facilitate the interpretation of the DEER data, we generated an ensemble of structural models in multiple conformations using a recently introduced modification of AlphaFold2 . Our experimental results reveal acid-induced conformational changes that dislodge the Cterminus from the permeation pathway coupled with rearrangement of helices that enables isomerization between inward- and outward-facing states. The substrate glutamate, but not GABA, modulates the dynamics of an extracellular thin gate without shifting the equilibrium between inward- and outward-facing conformations. In addition to introducing an integrated methodology for probing transporter conformational dynamics, the congruence of the DEER data with patterns of structural rearrangements deduced from ensembles of AlphaFold2 models illuminates the conformational cycle of GadC underpinning transport and exposes yet another example of the divergence between the dynamics of different families in the LeuT-fold.


Subject(s)
Antiporters , Bacterial Proteins , Membrane Proteins , Protein Conformation , Antiporters/chemistry , Bacterial Proteins/chemistry , Electron Spin Resonance Spectroscopy , Glutamates , Hydrogen-Ion Concentration , Membrane Proteins/chemistry , Models, Molecular , Molecular Dynamics Simulation , gamma-Aminobutyric Acid
14.
J Biochem ; 172(4): 217-224, 2022 Sep 30.
Article in English | MEDLINE | ID: mdl-35818339

ABSTRACT

The aspartate:alanine exchanger family of membrane transporters includes industrially important transporters such as succinate exporter and glutamate exporter. No high-resolution structure is available from this family so far, and the transport mechanism of these transporters also remains unclear. In the present study, we focus on the oligomeric status of the aspartate:alanine antiporter (AspT) of Tetragenococcus halophilus, which is the prototype of this family. To investigate the oligomeric structure of AspT, we established a system that produces high yields of highly purified AspT and determined the oligomeric structure of AspT by analysis with size exclusion chromatography coupled with multi-angle light scattering and blue native PAGE and by comparison of the wild-type AspT with a single-cysteine mutant that forms spontaneous inter-molecular thiol crosslinking. All the results consistently support the notion that AspT is a homodimer in solutions and in membranes.


Subject(s)
Alanine , Aspartic Acid , Alanine/chemistry , Antiporters/chemistry , Aspartic Acid/chemistry , Cysteine , Enterococcaceae , Glutamates , Membrane Transport Proteins , Succinates
15.
J Mol Biol ; 434(19): 167746, 2022 10 15.
Article in English | MEDLINE | ID: mdl-35843285

ABSTRACT

Found in all domains of life, transporters belonging to the LeuT-fold class mediate the import and exchange of hydrophilic and charged compounds such as amino acids, metals, and sugar molecules. Nearly two decades of investigations on the eponymous bacterial transporter LeuT have yielded a library of high-resolution snapshots of its conformational cycle linked by solution-state experimental data obtained from multiple techniques. In parallel, its topology has been observed in symporters and antiporters characterized by a spectrum of substrate specificities and coupled to gradients of distinct ions. Here we review and compare mechanistic models of transport for LeuT, its well-studied homologs, as well as functionally distant members of the fold, emphasizing the commonalities and divergences in alternating access and the corresponding energy landscapes. Our integrated summary illustrates how fold conservation, a hallmark of the LeuT fold, coincides with divergent choreographies of alternating access that nevertheless capitalize on recurrent structural motifs. In addition, it highlights the knowledge gap that hinders the leveraging of the current body of research into detailed mechanisms of transport for this important class of membrane proteins.


Subject(s)
Antiporters , Bacterial Proteins , Leucine , Symporters , Antiporters/chemistry , Bacterial Proteins/chemistry , Biological Transport , Leucine/metabolism , Protein Folding , Symporters/chemistry
16.
J Chem Phys ; 156(8): 085102, 2022 Feb 28.
Article in English | MEDLINE | ID: mdl-35232188

ABSTRACT

Successful functioning of biological cells relies on efficient translocation of different materials across cellular membranes. An important part of this transportation system is membrane channels that are known as antiporters and symporters. They exploit the energy stored as a trans-membrane gradient of one type of molecules to transport the other types of molecules against their gradients. For symporters, the directions of both fluxes for driving and driven species coincide, while for antiporters, the fluxes move in opposite directions. There are surprising experimental observations that despite differing only by the direction of transport fluxes, the molecular mechanisms of translocation adopted by antiporters and symporters seem to be drastically different. We present chemical-kinetic models to quantitatively investigate this phenomenon. Our theoretical approach allows us to explain why antiporters mostly utilize a single-site transportation when only one molecule of any type might be associated with the channel. At the same time, the transport in symporters requires two molecules of different types to be simultaneously associated with the channel. In addition, we investigate the kinetic constraints and efficiency of symporters and compare them with the same properties of antiporters. Our theoretical analysis clarifies some important physical-chemical features of cellular trans-membrane transport.


Subject(s)
Antiporters , Symporters , Antiporters/chemistry , Antiporters/metabolism , Biological Transport , Biological Transport, Active , Models, Theoretical , Symporters/metabolism
17.
Nat Struct Mol Biol ; 29(2): 108-120, 2022 02.
Article in English | MEDLINE | ID: mdl-35173351

ABSTRACT

The Na+/H+ exchanger SLC9B2, also known as NHA2, correlates with the long-sought-after Na+/Li+ exchanger linked to the pathogenesis of diabetes mellitus and essential hypertension in humans. Despite the functional importance of NHA2, structural information and the molecular basis for its ion-exchange mechanism have been lacking. Here we report the cryo-EM structures of bison NHA2 in detergent and in nanodiscs, at 3.0 and 3.5 Å resolution, respectively. The bison NHA2 structure, together with solid-state membrane-based electrophysiology, establishes the molecular basis for electroneutral ion exchange. NHA2 consists of 14 transmembrane (TM) segments, rather than the 13 TMs previously observed in mammalian Na+/H+ exchangers (NHEs) and related bacterial antiporters. The additional N-terminal helix in NHA2 forms a unique homodimer interface with a large intracellular gap between the protomers, which closes in the presence of phosphoinositol lipids. We propose that the additional N-terminal helix has evolved as a lipid-mediated remodeling switch for the regulation of NHA2 activity.


Subject(s)
Sodium-Hydrogen Exchangers/chemistry , Sodium-Hydrogen Exchangers/metabolism , Amino Acid Sequence , Animals , Antiporters/chemistry , Antiporters/genetics , Antiporters/metabolism , Binding Sites , Bison/genetics , Bison/metabolism , Cryoelectron Microscopy , Humans , Lipid Metabolism , Mass Spectrometry , Models, Molecular , Molecular Dynamics Simulation , Nanostructures/chemistry , Nanostructures/ultrastructure , Protein Multimerization , Proteolipids/chemistry , Proteolipids/metabolism , Sodium-Hydrogen Exchangers/genetics , Static Electricity
18.
BMC Plant Biol ; 21(1): 595, 2021 Dec 16.
Article in English | MEDLINE | ID: mdl-34915842

ABSTRACT

Sweet potato (Ipomoea batatas (L.) Lam.) is a good source of carbohydrates, an excellent raw material for starch-based industries, and a strong candidate for biofuel production due to its high starch content. However, the molecular basis of starch biosynthesis and accumulation in sweet potato is still insufficiently understood. Glucose-6-phosphate/phosphate translocators (GPTs) mediate the import of glucose-6-phosphate (Glc6P) into plastids for starch synthesis. Here, we report the isolation of a GPT-encoding gene, IbG6PPT1, from sweet potato and the identification of two additional IbG6PPT1 gene copies in the sweet potato genome. IbG6PPT1 encodes a chloroplast membrane-localized GPT belonging to the GPT1 group and highly expressed in storage root of sweet potato. Heterologous expression of IbG6PPT1 resulted in increased starch content in the leaves, root tips, and seeds and soluble sugar in seeds of Arabidopsis thaliana, but a reduction in soluble sugar in the leaves. These findings suggested that IbG6PPT1 might play a critical role in the distribution of carbon sources in source and sink and the accumulation of carbohydrates in storage tissues and would be a good candidate gene for controlling critical starch properties in sweet potato.


Subject(s)
Antiporters/isolation & purification , Glucose-6-Phosphate/metabolism , Ipomoea batatas/chemistry , Monosaccharide Transport Proteins/isolation & purification , Plant Proteins/isolation & purification , Antiporters/chemistry , Antiporters/genetics , Antiporters/metabolism , Chloroplasts/chemistry , Cloning, Molecular , Genes, Plant , Ipomoea batatas/genetics , Ipomoea batatas/metabolism , Models, Molecular , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/metabolism , Plant Roots/metabolism , Protein Conformation , Starch/metabolism , Sugars/metabolism
19.
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
20.
Molecules ; 26(22)2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34834047

ABSTRACT

Intracellular transport of chloride by members of the CLC transporter family involves a coupled exchange between a Cl- anion and a proton (H+), which makes the transport function dependent on ambient pH. Transport activity peaks at pH 4.5 and stalls at neutral pH. However, a structure of the WT protein at acidic pH is not available, making it difficult to assess the global conformational rearrangements that support a pH-dependent gating mechanism. To enable modeling of the CLC-ec1 dimer at acidic pH, we have applied molecular dynamics simulations (MD) featuring a new force field modification scheme-termed an Equilibrium constant pH approach (ECpH). The ECpH method utilizes linear interpolation between the force field parameters of protonated and deprotonated states of titratable residues to achieve a representation of pH-dependence in a narrow range of physiological pH values. Simulations of the CLC-ec1 dimer at neutral and acidic pH comparing ECpH-MD to canonical MD, in which the pH-dependent protonation is represented by a binary scheme, substantiates the better agreement of the conformational changes and the final model with experimental data from NMR, cross-link and AFM studies, and reveals structural elements that support the gate-opening at pH 4.5, including the key glutamates Gluin and Gluex.


Subject(s)
Antiporters/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Hydrogen-Ion Concentration , Microscopy, Atomic Force , Molecular Dynamics Simulation , Protein Conformation , Protons
SELECTION OF CITATIONS
SEARCH DETAIL