Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 632
Filter
1.
PLoS One ; 19(6): e0304512, 2024.
Article in English | MEDLINE | ID: mdl-38829838

ABSTRACT

The Organic Cation Transporter Novel 1 (OCTN1), also known as SLC22A4, is widely expressed in various human tissues, and involved in numerous physiological and pathological processes remains. It facilitates the transport of organic cations, zwitterions, with selectivity for positively charged solutes. Ergothioneine, an antioxidant compound, and acetylcholine (Ach) are among its substrates. Given the lack of experimentally solved structures of this protein, this study aimed at generating a reliable 3D model of OCTN1 to shed light on its substrate-binding preferences and the role of sodium in substrate recognition and transport. A chimeric model was built by grafting the large extracellular loop 1 (EL1) from an AlphaFold-generated model onto a homology model. Molecular dynamics simulations revealed domain-specific mobility, with EL1 exhibiting the highest impact on overall stability. Molecular docking simulations identified cytarabine and verapamil as highest affinity ligands, consistent with their known inhibitory effects on OCTN1. Furthermore, MM/GBSA analysis allowed the categorization of substrates into weak, good, and strong binders, with molecular weight strongly correlating with binding affinity to the recognition site. Key recognition residues, including Tyr211, Glu381, and Arg469, were identified through interaction analysis. Ach demonstrated a low interaction energy, supporting the hypothesis of its one-directional transport towards to outside of the membrane. Regarding the role of sodium, our model suggested the involvement of Glu381 in sodium binding. Molecular dynamics simulations of systems at increasing levels of Na+ concentrations revealed increased sodium occupancy around Glu381, supporting experimental data associating Na+ concentration to molecule transport. In conclusion, this study provides valuable insights into the 3D structure of OCTN1, its substrate-binding preferences, and the role of sodium in the recognition. These findings contribute to the understanding of OCTN1 involvement in various physiological and pathological processes and may have implications for drug development and disease management.


Subject(s)
Molecular Docking Simulation , Molecular Dynamics Simulation , Organic Cation Transport Proteins , Humans , Organic Cation Transport Proteins/chemistry , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Symporters/chemistry , Symporters/metabolism , Binding Sites , Protein Binding , Ergothioneine/chemistry , Ergothioneine/metabolism , Sodium/metabolism , Sodium/chemistry , Computer Simulation , Acetylcholine/metabolism , Acetylcholine/chemistry , Ligands
3.
Biophys J ; 123(10): 1195-1210, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38544409

ABSTRACT

Human Na+ taurocholate co-transporting protein (hNTCP) is a key bile salt transporter to maintain enterohepatic circulation and is responsible for the recognition of hepatitis B and D viruses. Despite landmark cryoelectron microscopy studies revealing open-pore and inward-facing states of hNTCP stabilized by antibodies, the transport mechanism remains largely unknown. To address this knowledge gap, we used molecular dynamics and enhanced sampling metadynamics simulations to elucidate the intrinsic mechanism of hNTCP-mediated taurocholate acid (TCA) transport driven by Na+ binding. We uncovered three TCA-binding modes, including one that closely matched the limited cryoelectron microscopy density observed in the open-pore hNTCP. We also captured several key hNTCP conformations in the substrate transport cycle, particularly including an outward-facing, substrate-bound state. Furthermore, we provided thermodynamic evidence supporting that changes in the Na+-binding state drive the TCA transport by exploiting the amphiphilic nature of the substrate and modulating the protein environment, thereby enabling the TCA molecule to flip through. Understanding these mechanistic details of Na+-driven bile acid transport may aid in the development of hNTCP-targeted therapies for liver diseases.


Subject(s)
Molecular Dynamics Simulation , Organic Anion Transporters, Sodium-Dependent , Sodium , Symporters , Taurocholic Acid , Symporters/metabolism , Symporters/chemistry , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters, Sodium-Dependent/chemistry , Humans , Sodium/metabolism , Taurocholic Acid/metabolism , Biological Transport , Bile Acids and Salts/metabolism , Thermodynamics
4.
Hum Mol Genet ; 32(9): 1552-1564, 2023 04 20.
Article in English | MEDLINE | ID: mdl-36611016

ABSTRACT

Congenital myasthenic syndrome (CMS) is a heterogeneous condition associated with 34 different genes, including SLC5A7, which encodes the high-affinity choline transporter 1 (CHT1). CHT1 is expressed in presynaptic neurons of the neuromuscular junction where it uses the inward sodium gradient to reuptake choline. Biallelic CHT1 mutations often lead to neonatal lethality, and less commonly to non-lethal motor weakness and developmental delays. Here, we report detailed biochemical characterization of two novel mutations in CHT1, p.I294T and p.D349N, which we identified in an 11-year-old patient with a history of neonatal respiratory distress, and subsequent hypotonia and global developmental delay. Heterologous expression of each CHT1 mutant in human embryonic kidney cells showed two different mechanisms of reduced protein function. The p.I294T CHT1 mutant transporter function was detectable, but its abundance and half-life were significantly reduced. In contrast, the p.D349N CHT1 mutant was abundantly expressed at the cell membrane, but transporter function was absent. The residual function of the p.I294T CHT1 mutant may explain the non-lethal form of CMS in this patient, and the divergent mechanisms of reduced CHT1 function that we identified may guide future functional studies of the CHT1 myasthenic syndrome. Based on these in vitro studies that provided a diagnosis, treatment with cholinesterase inhibitor together with physical and occupational therapy significantly improved the patient's strength and quality of life.


Subject(s)
Mutant Proteins , Mutation , Myasthenic Syndromes, Congenital , Symporters , Myasthenic Syndromes, Congenital/drug therapy , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/metabolism , Myasthenic Syndromes, Congenital/rehabilitation , Humans , Male , Child , HEK293 Cells , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Half-Life , Cell Membrane/metabolism , Protein Transport , Staurosporine/pharmacology , Pyridostigmine Bromide/therapeutic use , Quality of Life , Symporters/chemistry , Symporters/genetics , Symporters/metabolism
5.
Biochemistry ; 62(1): 118-133, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36516499

ABSTRACT

Osmosensing transporter ProP forestalls cellular dehydration by detecting environments with high osmotic pressure and mediating the accumulation of organic osmolytes by bacterial cells. It is composed of 12 transmembrane helices with cytoplasmic N- and C-termini. In Escherichia coli, dimers form when the C-terminal domains of ProP molecules form homodimeric, antiparallel, α-helical coiled coils. No dominant negative effect was detected when inactive and active ProP molecules formed heterodimers in vivo. Purification of ProP in detergent dodecylmaltoside yielded monomers, which were functional after reconstitution in proteoliposomes. With other evidence, this suggests that ProP monomers function independently whether in the monomeric or dimeric state. Amino acid replacements that disrupted or reversed the coiled coil did not prevent in vivo dimerization of ProP detected with a bacterial two-hybrid system. Maleimide labeling detected no osmolality-dependent variation in the reactivities of cysteine residues introduced to transmembrane helix (TM) XII. In contrast, coarse-grained molecular dynamic simulations detected deformation of the lipid around TMs III and VI, on the lipid-exposed protein surface opposite to TM XII. This suggests that the dimer interface of ProP includes the surfaces of TMs III and VI, not of TM XII as previously suggested by crosslinking data. Homology modeling suggested that coiled-coil formation and dimerization via such an interface are not mutually exclusive. In previous work, alterations to the C-terminal coiled coil blocked co-localization of ProP with phospholipid cardiolipin at E. coli cell poles. Thus, dimerization may contribute to ProP targeting, adjust its lipid environment, and hence indirectly modify its osmotic stress response.


Subject(s)
Escherichia coli Proteins , Symporters , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Amino Acid Sequence , Dimerization , Symporters/chemistry , Membrane Transport Proteins/metabolism , Phospholipids/metabolism
6.
Nature ; 612(7941): 795-801, 2022 12.
Article in English | MEDLINE | ID: mdl-36517601

ABSTRACT

The sodium/iodide symporter (NIS) is the essential plasma membrane protein that mediates active iodide (I-) transport into the thyroid gland, the first step in the biosynthesis of the thyroid hormones-the master regulators of intermediary metabolism. NIS couples the inward translocation of I- against its electrochemical gradient to the inward transport of Na+ down its electrochemical gradient1,2. For nearly 50 years before its molecular identification3, NIS was the molecule at the centre of the single most effective internal radiation cancer therapy: radioiodide (131I-) treatment for thyroid cancer2. Mutations in NIS cause congenital hypothyroidism, which must be treated immediately after birth to prevent stunted growth and cognitive deficiency2. Here we report three structures of rat NIS, determined by single-particle cryo-electron microscopy: one with no substrates bound; one with two Na+ and one I- bound; and one with one Na+ and the oxyanion perrhenate bound. Structural analyses, functional characterization and computational studies show the substrate-binding sites and key residues for transport activity. Our results yield insights into how NIS selects, couples and translocates anions-thereby establishing a framework for understanding NIS function-and how it transports different substrates with different stoichiometries and releases substrates from its substrate-binding cavity into the cytosol.


Subject(s)
Iodides , Sodium , Symporters , Animals , Rats , Cryoelectron Microscopy , Iodides/metabolism , Sodium/metabolism , Symporters/chemistry , Symporters/metabolism , Symporters/ultrastructure , Binding Sites , Substrate Specificity , Ion Transport
7.
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
8.
Nat Struct Mol Biol ; 29(6): 604-612, 2022 06.
Article in English | MEDLINE | ID: mdl-35710838

ABSTRACT

Brain development and function require uptake of essential omega-3 fatty acids in the form of lysophosphatidylcholine via major-facilitator superfamily transporter MFSD2A, a potential pharmaceutical target to modulate blood-brain barrier (BBB) permeability. MFSD2A is also the receptor of endogenous retroviral envelope syncytin-2 (SYNC2) in human placenta, where it mediates cell-cell fusion and formation of the maternal-fetal interface. Here, we report a cryo-electron microscopy structure of the human MFSD2A-SYNC2 complex that reveals a large hydrophobic cavity in the transporter C-terminal domain to occlude long aliphatic chains. The transporter architecture suggests an alternating-access transport mechanism for lipid substrates in mammalian MFS transporters. SYNC2 establishes an extensive binding interface with MFSD2A, and a SYNC2-soluble fragment acts as a long-sought-after inhibitor of MFSD2A transport. Our work uncovers molecular mechanisms important to brain and placenta development and function, and SYNC2-mediated inhibition of MFSD2A transport suggests strategies to aid delivery of therapeutic macromolecules across the BBB.


Subject(s)
Pregnancy Proteins/chemistry , Symporters/chemistry , Animals , Brain/metabolism , Cryoelectron Microscopy , Female , Humans , Lysophosphatidylcholines/chemistry , Lysophosphatidylcholines/metabolism , Mammals/metabolism , Membrane Transport Proteins/metabolism , Pregnancy , Pregnancy Proteins/metabolism , Symporters/metabolism
9.
Proc Natl Acad Sci U S A ; 119(27): e2109083119, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35759661

ABSTRACT

Cation-chloride cotransporters (CCCs) catalyze electroneutral symport of Cl- with Na+ and/or K+ across membranes. CCCs are fundamental in cell volume homeostasis, transepithelia ion movement, maintenance of intracellular Cl- concentration, and neuronal excitability. Here, we present a cryoelectron microscopy structure of human K+-Cl- cotransporter (KCC)1 bound with the VU0463271 inhibitor in an outward-open state. In contrast to many other amino acid-polyamine-organocation transporter cousins, our first outward-open CCC structure reveals that opening the KCC1 extracellular ion permeation path does not involve hinge-bending motions of the transmembrane (TM) 1 and TM6 half-helices. Instead, rocking of TM3 and TM8, together with displacements of TM4, TM9, and a conserved intracellular loop 1 helix, underlie alternate opening and closing of extracellular and cytoplasmic vestibules. We show that KCC1 intriguingly exists in one of two distinct dimeric states via different intersubunit interfaces. Our studies provide a blueprint for understanding the mechanisms of CCCs and their inhibition by small molecule compounds.


Subject(s)
Solute Carrier Family 12, Member 4 , Symporters , Cations/metabolism , Chlorides/metabolism , Cryoelectron Microscopy , Humans , Ion Transport , Protein Conformation, alpha-Helical , Solute Carrier Family 12, Member 4/chemistry , Symporters/antagonists & inhibitors , Symporters/chemistry , K Cl- Cotransporters
10.
Nature ; 606(7916): 1021-1026, 2022 06.
Article in English | MEDLINE | ID: mdl-35580629

ABSTRACT

Chronic infection with hepatitis B virus (HBV) affects more than 290 million people worldwide, is a major cause of cirrhosis and hepatocellular carcinoma, and results in an estimated 820,000 deaths annually1,2. For HBV infection to be established, a molecular interaction is required between the large glycoproteins of the virus envelope (known as LHBs) and the host entry receptor sodium taurocholate co-transporting polypeptide (NTCP), a sodium-dependent bile acid transporter from the blood to hepatocytes3. However, the molecular basis for the virus-transporter interaction is poorly understood. Here we report the cryo-electron microscopy structures of human, bovine and rat NTCPs in the apo state, which reveal the presence of a tunnel across the membrane and a possible transport route for the substrate. Moreover, the cryo-electron microscopy structure of human NTCP in the presence of the myristoylated preS1 domain of LHBs, together with mutation and transport assays, suggest a binding mode in which preS1 and the substrate compete for the extracellular opening of the tunnel in NTCP. Our preS1 domain interaction analysis enables a mechanistic interpretation of naturally occurring HBV-insusceptible mutations in human NTCP. Together, our findings provide a structural framework for HBV recognition and a mechanistic understanding of sodium-dependent bile acid translocation by mammalian NTCPs.


Subject(s)
Cryoelectron Microscopy , Hepatitis B virus , Organic Anion Transporters, Sodium-Dependent , Receptors, Virus , Symporters , Animals , Apoproteins/chemistry , Apoproteins/genetics , Apoproteins/metabolism , Apoproteins/ultrastructure , Cattle , Hepatitis B virus/metabolism , Hepatocytes/metabolism , Humans , Mutation , Organic Anion Transporters, Sodium-Dependent/chemistry , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters, Sodium-Dependent/ultrastructure , Rats , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Receptors, Virus/ultrastructure , Sodium/metabolism , Symporters/chemistry , Symporters/genetics , Symporters/metabolism , Symporters/ultrastructure
11.
Nature ; 606(7916): 1027-1031, 2022 06.
Article in English | MEDLINE | ID: mdl-35580630

ABSTRACT

Around 250 million people are infected with hepatitis B virus (HBV) worldwide1, and 15 million may also carry the satellite virus hepatitis D virus (HDV), which confers even greater risk of severe liver disease2. The HBV receptor has been identified as sodium taurocholate co-transporting polypeptide (NTCP), which interacts directly with the first 48 amino acid residues of the N-myristoylated N-terminal preS1 domain of the viral large protein3. Despite the pressing need for therapeutic agents to counter HBV, the structure of NTCP remains unsolved. This 349-residue protein is closely related to human apical sodium-dependent bile acid transporter (ASBT), another member of the solute carrier family SLC10. Crystal structures have been reported of similar bile acid transporters from bacteria4,5, and these models are believed to resemble closely both NTCP and ASBT. Here we have used cryo-electron microscopy to solve the structure of NTCP bound to an antibody, clearly showing that the transporter has no equivalent of the first transmembrane helix found in other SLC10 proteins, and that the N terminus is exposed on the extracellular face. Comparison of our structure with those of related proteins indicates a common mechanism of bile acid transport, but the NTCP structure displays an additional pocket formed by residues that are known to interact with preS1, presenting new opportunities for structure-based drug design.


Subject(s)
Bile Acids and Salts , Cryoelectron Microscopy , Hepatitis B virus , Organic Anion Transporters, Sodium-Dependent , Receptors, Virus , Symporters , Antibodies , Bile Acids and Salts/metabolism , Hepatitis B virus/metabolism , Hepatocytes/metabolism , Humans , Organic Anion Transporters, Sodium-Dependent/chemistry , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters, Sodium-Dependent/ultrastructure , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Receptors, Virus/ultrastructure , Symporters/chemistry , Symporters/metabolism , Symporters/ultrastructure
12.
Nature ; 606(7916): 1015-1020, 2022 06.
Article in English | MEDLINE | ID: mdl-35545671

ABSTRACT

The liver takes up bile salts from blood to generate bile, enabling absorption of lipophilic nutrients and excretion of metabolites and drugs1. Human Na+-taurocholate co-transporting polypeptide (NTCP) is the main bile salt uptake system in liver. NTCP is also the cellular entry receptor of human hepatitis B and D viruses2,3 (HBV/HDV), and has emerged as an important target for antiviral drugs4. However, the molecular mechanisms underlying NTCP transport and viral receptor functions remain incompletely understood. Here we present cryo-electron microscopy structures of human NTCP in complexes with nanobodies, revealing key conformations of its transport cycle. NTCP undergoes a conformational transition opening a wide transmembrane pore that serves as the transport pathway for bile salts, and exposes key determinant residues for HBV/HDV binding to the outside of the cell. A nanobody that stabilizes pore closure and inward-facing states impairs recognition of the HBV/HDV receptor-binding domain preS1, demonstrating binding selectivity of the viruses for open-to-outside over inward-facing conformations of the NTCP transport cycle. These results provide molecular insights into NTCP 'gated-pore' transport and HBV/HDV receptor recognition mechanisms, and are expected to help with development of liver disease therapies targeting NTCP.


Subject(s)
Bile Acids and Salts , Cryoelectron Microscopy , Liver , Organic Anion Transporters, Sodium-Dependent , Sodium , Symporters , Bile/metabolism , Bile Acids and Salts/metabolism , Hepatitis B virus/metabolism , Hepatitis Delta Virus/metabolism , Hepatocytes/metabolism , Humans , Liver/metabolism , Organic Anion Transporters, Sodium-Dependent/chemistry , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters, Sodium-Dependent/ultrastructure , Protein Conformation , Receptors, Virus/metabolism , Single-Domain Antibodies , Sodium/metabolism , Symporters/chemistry , Symporters/metabolism , Symporters/ultrastructure , Virus Internalization
13.
J Mol Biol ; 434(12): 167598, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35461877

ABSTRACT

Cation selectivity and coupling are important attributes of cation-coupled symporters. Salmonella typhimurium melibiose permease (MelBSt) catalyzes the co-transport of galactosides with cations (H+, Li+, or Na+). 3-D crystal structures of MelBSt have revealed the molecular recognition for sugar substrates, but the cation binding and coupling mechanisms have not been defined to atomic levels. In its human homolog MFSD2A, a lethal mutation was mapped at its Na+-binding pocket; however, none of the structures in this subfamily resolved its cation binding. In this study, molecular dynamics simulations reveal the binding interactions of Na+ and Li+ with MelBSt. Interestingly, Thr121, the lethal mutation position in MFSD2A, forms stable interaction with Na+ but is at a distance from Li+. Most mutations among 11 single-site Thr121 mutants of MelBSt exhibited little effects on the galactoside binding, but largely altered the cation selectivity with severe inhibitions on Na+ binding. Few mutants (Pro and Ala) completely lost the Na+ binding and Na+-coupled transport, but their Li+ or H+ modes of activity were largely retained. It can be concluded that Thr121 is necessary for Na+ binding, but not required for the binding of H+ or Li+, so a subset of the Na+-binding pocket is enough for Li+ binding. In addition, the protein stability for some mutants can be only retained in the presence of Li+, but not by Na+ due to the lack of affinity. This finding, together with other identified thermostable mutants, supports that the charge balance of the cation-binding site plays an important role in MelBSt protein stability.


Subject(s)
Bacterial Proteins , Salmonella typhimurium , Symporters , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cations/metabolism , Humans , Lithium/metabolism , Melibiose/metabolism , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics , Sodium/metabolism , Symporters/chemistry , Symporters/genetics , Symporters/metabolism
14.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35055100

ABSTRACT

The Novel Organic Cation Transporter, OCTN1, is the first member of the OCTN subfamily; it belongs to the wider Solute Carrier family SLC22, which counts many members including cation and anion organic transporters. The tertiary structure has not been resolved for any cation organic transporter. The functional role of OCNT1 is still not well assessed despite the many functional studies so far conducted. The lack of a definitive identification of OCTN1 function can be attributed to the different experimental systems and methodologies adopted for studying each of the proposed ligands. Apart from the contradictory data, the international scientific community agrees on a role of OCTN1 in protecting cells and tissues from oxidative and/or inflammatory damage. Moreover, the involvement of this transporter in drug interactions and delivery has been well clarified, even though the exact profile of the transported/interacting molecules is still somehow confusing. Therefore, OCTN1 continues to be a hot topic in terms of its functional role and structure. This review focuses on the most recent advances on OCTN1 in terms of functional aspects, physiological roles, substrate specificity, drug interactions, tissue expression, and relationships with pathology.


Subject(s)
Biomarkers , Disease Susceptibility , Drug Interactions , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Symporters/genetics , Symporters/metabolism , Acetylation , Animals , Binding Sites , Biological Transport , Ergothioneine/metabolism , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Models, Molecular , Molecular Conformation , Organ Specificity , Organic Cation Transport Proteins/chemistry , Protein Binding , Structure-Activity Relationship , Symporters/chemistry
15.
J Mol Biol ; 434(2): 167356, 2022 01 30.
Article in English | MEDLINE | ID: mdl-34780780

ABSTRACT

The crucial function of neurotransmitter:sodium symporters (NSS) in facilitating the reuptake of neurotransmitters into neuronal cells makes them attractive drug targets for treating multiple mental diseases. Due to the challenges in working with eukaryotic NSS proteins, LeuT, a prokaryotic amino acid transporter, has served as a model protein for studying structure-function relationships of NSS family proteins. With hydrogen-deuterium exchange mass spectrometry (HDX-MS), slow unfolding/refolding kinetics were identified in multiple regions of LeuT, suggesting that substrate translocation involves cooperative fluctuations of helical stretches. Earlier work has solely been performed at non-native temperatures (25 °C) for LeuT, which is evolutionarily adapted to function at high temperatures (85 - 95 °C). To address the effect of temperature on LeuT dynamics, we have performed HDX-MS experiments at elevated temperatures (45 °C and 60 °C). At these elevated temperatures, multiple regions in LeuT exhibited increased dynamics compared to 25 °C. Interestingly, coordinated slow unfolding/refolding of key regions could still be observed, though considerably faster. We have further investigated the conformational impact of binding the efficiently transported substrate alanine (Ala) relative to the much slower transported substrate leucine (Leu). Comparing the HDX of the Ala-bound versus Leu-bound state of LeuT, we observe distinct differences that could explain the faster transport rate (kcat) of Ala relative to Leu. Importantly, slow unfolding/refolding dynamics could still be observed in regions of Ala-bound LeuT . Overall, our work brings new insights into the conformational dynamics of LeuT and provides a better understanding of the transport mechanism of LeuT and possibly other transporters bearing the LeuT fold.


Subject(s)
Molecular Conformation , Neurotransmitter Agents , Symporters/chemistry , Temperature , Kinetics , Membrane Proteins , Molecular Dynamics Simulation , Pharmaceutical Preparations , Plasma Membrane Neurotransmitter Transport Proteins/chemistry , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Protein Conformation , Sodium
16.
Nature ; 596(7872): 444-448, 2021 08.
Article in English | MEDLINE | ID: mdl-34349262

ABSTRACT

MFSD2A is a sodium-dependent lysophosphatidylcholine symporter that is responsible for the uptake of docosahexaenoic acid into the brain1,2, which is crucial for the development and performance of the brain3. Mutations that affect MFSD2A cause microcephaly syndromes4,5. The ability of MFSD2A to transport lipid is also a key mechanism that underlies its function as an inhibitor of transcytosis to regulate the blood-brain barrier6,7. Thus, MFSD2A represents an attractive target for modulating the permeability of the blood-brain barrier for drug delivery. Here we report the cryo-electron microscopy structure of mouse MFSD2A. Our structure defines the architecture of this important transporter, reveals its unique extracellular domain and uncovers its substrate-binding cavity. The structure-together with our functional studies and molecular dynamics simulations-identifies a conserved sodium-binding site, reveals a potential lipid entry pathway and helps to rationalize MFSD2A mutations that underlie microcephaly syndromes. These results shed light on the critical lipid transport function of MFSD2A and provide a framework to aid in the design of specific modulators for therapeutic purposes.


Subject(s)
Blood-Brain Barrier/metabolism , Lipid Metabolism , Symporters/chemistry , Symporters/metabolism , Animals , Binding Sites , Biological Transport , HEK293 Cells , Humans , Mice , Models, Molecular , Molecular Dynamics Simulation , Mutation , Protein Domains , Sodium/metabolism , Symporters/genetics , Symporters/ultrastructure
17.
Commun Biol ; 4(1): 931, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34341464

ABSTRACT

Major facilitator superfamily_2 transporters are widely found from bacteria to mammals. The melibiose transporter MelB, which catalyzes melibiose symport with either Na+, Li+, or H+, is a prototype of the Na+-coupled MFS transporters, but its sugar recognition mechanism has been a long-unsolved puzzle. Two high-resolution X-ray crystal structures of a Salmonella typhimurium MelB mutant with a bound ligand, either nitrophenyl-α-D-galactoside or dodecyl-ß-D-melibioside, were refined to a resolution of 3.05 or 3.15 Å, respectively. In the substrate-binding site, the interaction of both galactosyl moieties on the two ligands with MelBSt are virturally same, so the sugar specificity determinant pocket can be recognized, and hence the molecular recognition mechanism for sugar binding in MelB has been deciphered. The conserved cation-binding pocket is also proposed, which directly connects to the sugar specificity pocket. These key structural findings have laid a solid foundation for our understanding of the cooperative binding and symport mechanisms in Na+-coupled MFS transporters, including eukaryotic transporters such as MFSD2A.


Subject(s)
Bacterial Proteins/chemistry , Crystallography, X-Ray , Melibiose/metabolism , Salmonella typhimurium/metabolism , Symporters/chemistry
18.
FASEB J ; 35(8): e21681, 2021 08.
Article in English | MEDLINE | ID: mdl-34196428

ABSTRACT

The sodium/iodide symporter (NIS) expresses at the basolateral plasma membrane of the thyroid follicular cell and mediates iodide accumulation required for normal thyroid hormonogenesis. Loss-of-function NIS variants cause congenital hypothyroidism due to impaired iodide accumulation in thyroid follicular cells underscoring the significance of NIS for thyroid physiology. Here we report novel findings derived from the thorough characterization of the nonsense NIS mutant p.R636* NIS-leading to a truncated protein missing the last eight amino acids-identified in twins with congenital hypothyroidism. R636* NIS is severely mislocalized into intracellular vesicular compartments due to the lack of a conserved carboxy-terminal type 1 PDZ-binding motif. As a result, R636* NIS is barely targeted to the plasma membrane and therefore iodide transport is reduced. Deletion of the PDZ-binding motif causes NIS accumulation into late endosomes and lysosomes. Using PDZ domain arrays, we revealed that the PDZ-domain containing protein SCRIB binds to the carboxy-terminus of NIS by a PDZ-PDZ interaction. Furthermore, in CRISPR/Cas9-based SCRIB deficient cells, NIS expression at the basolateral plasma membrane is compromised, leading to NIS localization into intracellular vesicular compartments. We conclude that the PDZ-binding motif is a plasma membrane retention signal that participates in the polarized expression of NIS by selectively interacting with the PDZ-domain containing protein SCRIB, thus retaining the transporter at the basolateral plasma membrane. Our data provide insights into the molecular mechanisms that regulate NIS expression at the plasma membrane, a topic of great interest in the thyroid cancer field considering the relevance of NIS-mediated radioactive iodide therapy for differentiated thyroid carcinoma.


Subject(s)
Membrane Proteins/metabolism , Symporters/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Codon, Nonsense , Congenital Hypothyroidism/genetics , Congenital Hypothyroidism/metabolism , Conserved Sequence , Dogs , Endosomes/metabolism , HEK293 Cells , Humans , Lysosomes/metabolism , Madin Darby Canine Kidney Cells , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Mutagenesis, Site-Directed , PDZ Domains/genetics , Protein Structure, Secondary , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Symporters/chemistry , Symporters/genetics , Thyroid Gland/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
19.
Nature ; 595(7866): 315-319, 2021 07.
Article in English | MEDLINE | ID: mdl-34135507

ABSTRACT

Docosahexaenoic acid is an omega-3 fatty acid that is essential for neurological development and function, and it is supplied to the brain and eyes predominantly from dietary sources1-6. This nutrient is transported across the blood-brain and blood-retina barriers in the form of lysophosphatidylcholine by major facilitator superfamily domain containing 2A (MFSD2A) in a Na+-dependent manner7,8. Here we present the structure of MFSD2A determined using single-particle cryo-electron microscopy, which reveals twelve transmembrane helices that are separated into two pseudosymmetric domains. The transporter is in an inward-facing conformation and features a large amphipathic cavity that contains the Na+-binding site and a bound lysolipid substrate, which we confirmed using native mass spectrometry. Together with our functional analyses and molecular dynamics simulations, this structure reveals details of how MFSD2A interacts with substrates and how Na+-dependent conformational changes allow for the release of these substrates into the membrane through a lateral gate. Our work provides insights into the molecular mechanism by which this atypical major facility superfamily transporter mediates the uptake of lysolipids into the brain, and has the potential to aid in the delivery of neurotherapeutic agents.


Subject(s)
Biological Transport , Blood-Brain Barrier/metabolism , Cryoelectron Microscopy , Fatty Acids, Omega-3/metabolism , Symporters/chemistry , Symporters/metabolism , Animals , Binding Sites , Chickens , Fatty Acids, Omega-3/chemistry , Mass Spectrometry , Models, Molecular , Molecular Dynamics Simulation , Protein Domains , Sodium/metabolism , Symporters/ultrastructure
20.
Mol Genet Genomic Med ; 9(8): e1740, 2021 08.
Article in English | MEDLINE | ID: mdl-34192422

ABSTRACT

BACKGROUND: Diarrhea is a common occurrence in children below the age of 5 years. In chronic cases, it induces malnutrition that severely stunts growth. Bile acid diarrhea (BAD), caused by malabsorption of bile acid (BA), is a rare form of chronic diarrhea seldom observed in pediatric patients. Here, we present a clinical report on a novel case of chronic BAD, with severe stunting in an infant, induced by a homozygous mutation of SLC10A2. METHODS: We performed DNA extraction, whole-exome sequencing analysis, and mutation analysis of SLC10A2 to obtain genetic data on the patient. We subsequently analyzed the patient's clinical and genetic data. RESULTS: The patient's clinical manifestations were chronic diarrhea with increased BAs in the feces and extreme stunting, which was diagnosed as BAD. A homozygous mutation of SLC10A2 at the c.313T>C (rs201206937) site was detected. CONCLUSION: Our report reveals the youngest case illustrating the characteristics of BAD induced by genetic variant at 313T>C, and the second case entailing a clear association between a SLC10A2 genetic mutation and the onset of BAD. Our findings expand the mutant spectrum of the SLC10A2 gene and contribute to the refinement of the genotype-phenotype mapping of severe stunting induced by pediatric BAD. Moreover, they highlight the value of molecular genetic screening for diagnosing BAD in young patients.


Subject(s)
Diarrhea/genetics , Growth Disorders/genetics , Organic Anion Transporters, Sodium-Dependent/genetics , Symporters/genetics , Bile Acids and Salts/metabolism , Diarrhea/pathology , Female , Growth Disorders/pathology , Homozygote , Humans , Infant , Mutation , Organic Anion Transporters, Sodium-Dependent/chemistry , Organic Anion Transporters, Sodium-Dependent/metabolism , Protein Stability , Symporters/chemistry , Symporters/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...