Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Biochem Biophys Res Commun ; 738: 150544, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39163817

ABSTRACT

GLUT7 is a Class II glucose transporter predominantly expressed at the apical membrane of enterocytes in the small intestine. Here, we report the cryo-EM structure of nanodisc-reconstituted human GLUT7 in the apo state at 3.3 Å resolution. Our atomic model reveals a typical major facilitator superfamily fold, with the substrate-binding site open to the extracellular side of the membrane. Despite the nearly identical conformation to its closest family member, rat GLUT5, our structure unveils distinct features of the substrate-binding cavity that may influence substrate specificity and binding mode. A homology model of the inward-open human GLUT7 indicates that similar to other members of the GLUT family, it may undergo a global rocker-switch-like reorientation of the transmembrane bundles to facilitate substrate translocation across the membrane. Our work enhances the current structural understanding of the GLUT family, and lays a foundation for rational design of regulators of GLUTs and other sugar transporters.

2.
Proc Natl Acad Sci U S A ; 116(36): 17786-17791, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31371498

ABSTRACT

Antibodies are indispensable tools in protein engineering and structural biology. Antibodies suitable for structural studies should recognize the 3-dimensional (3D) conformations of target proteins. Generating such antibodies and characterizing their complexes with antigens take a significant amount of time and effort. Here, we show that we can expand the application of well-characterized antibodies by "transplanting" the epitopes that they recognize to proteins with completely different structures and sequences. Previously, several antibodies have been shown to recognize the alpha-helical conformation of antigenic peptides. We demonstrate that these antibodies can be made to bind to a variety of unrelated "off-target" proteins by modifying amino acids in the preexisting alpha helices of such proteins. Using X-ray crystallography, we determined the structures of the engineered protein-antibody complexes. All of the antibodies bound to the epitope-transplanted proteins, forming accurately predictable structures. Furthermore, we showed that binding of these antihelix antibodies to the engineered target proteins can modulate their catalytic activities by trapping them in selected functional states. Our method is simple and efficient, and it will have applications in protein X-ray crystallography, electron microscopy, and nanotechnology.


Subject(s)
Epitopes/chemistry , Proteins/chemistry , Single-Chain Antibodies/chemistry , Crystallography, X-Ray , Humans , Protein Conformation, alpha-Helical
3.
Drug Dev Res ; 83(3): 783-799, 2022 05.
Article in English | MEDLINE | ID: mdl-35040501

ABSTRACT

m6 A RNA methyltransferase (METTL3-14) catalyzes the methylation of adenosine in mRNA and plays important roles in mRNA functions, and it has been implicated in the progression of multiple cancers, including acute myeloid leukemia (AML). In this study, we describe the discovery of the first allosteric inhibitor of the METTL3-14 complex based on structure-activity relationship (SAR) and optimization studies of the hit compound, 4-[2-[5-chloro-1-(diphenylmethyl)-2-methyl-1H-indol-3-yl]-ethoxy]benzoic acid (CDIBA). Compound 43n was optimized throughout the modifications of 4 different regions of the structure, and it displayed potent enzyme inhibitory activity of the METTL3-14 complex (IC50  = 2.81 µM) and an antiproliferative effect in the AML cell lines by suppressing the m6 A level of mRNA. The inhibition mechanism and binding mode of 43n were based on the interaction of the reversible and noncompetitive inhibitory profile at the allosteric site along with selectivity for the METTL3-14 complex relative to each subunit enzyme or truncated complex enzyme.


Subject(s)
Enzyme Inhibitors , Leukemia, Myeloid, Acute , Methyltransferases , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Indoles/pharmacology , Methyltransferases/antagonists & inhibitors , Methyltransferases/chemistry , Methyltransferases/genetics , Methyltransferases/metabolism , RNA/chemistry , RNA/metabolism , RNA, Messenger/metabolism
4.
J Struct Biol ; 213(1): 107700, 2021 03.
Article in English | MEDLINE | ID: mdl-33545350

ABSTRACT

In fungi the ß-class of carbonic anhydrases (ß-CAs) are zinc metalloenzymes that are essential for growth, survival, differentiation, and virulence. Aspergillus fumigatus is the most important pathogen responsible for invasive aspergillosis and possesses two major ß-CAs, CafA and CafB. Recently we reported the biochemical characterization and 1.8 Å crystal structure of CafA. Here, we report a crystallographic analysis of CafB revealing the mechanism of enzyme catalysis and establish the relationship of this enzyme to other ß-CAs. While CafA has a typical open conformation, CafB, when exposed to acidic pH and/or an oxidative environment, has a novel type of active site in which a disulfide bond is formed between two zinc-ligating cysteines, expelling the zinc ion and stabilizing the inactive form of the enzyme. Based on the structural data, we generated an oxidation-resistant mutant (Y159A) of CafB. The crystal structure of the mutant under reducing conditions retains a catalytic zinc at the expected position, tetrahedrally coordinated by three residues (C57, H113 and C116) and an aspartic acid (D59), and replacing the zinc-bound water molecule in the closed form. Furthermore, the active site of CafB crystals grown under zinc-limiting conditions has a novel conformation in which the solvent-exposed catalytic cysteine (C116) is flipped out of the metal coordination sphere, facilitating release of the zinc ion. Taken together, our results suggest that A. fumigatus use sophisticated activity-inhibiting strategies to enhance its survival during infection.


Subject(s)
Aspergillus fumigatus/metabolism , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Catalysis , Catalytic Domain/physiology , Crystallography, X-Ray/methods , Kinetics , Zinc/metabolism
5.
J Struct Biol ; 208(1): 61-68, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31376470

ABSTRACT

The ß-carbonic anhydrases (ß-CAs) are widely distributed zinc-metalloenzymes that play essential roles in growth, survival, development and virulence in fungi. The majority of filamentous ascomycetes possess multiple ß-CA isoforms among which major and minor forms have been characterized. We examined the catalytic behavior of the two minor ß-CAs, CafC and CafD, of Aspergillus fumigatus, and found that both enzymes exhibited low CO2 hydration activities. To understand the structural basis of their low activities, we performed X-ray crystallographic and site-directed mutagenesis studies. Both enzymes exist as homodimers. Like other Type-I ß-CAs, the CafC active site has an "open" conformation in which the zinc ion is tetrahedrally coordinated by three residues (C36, H88 and C91) and a water molecule. However, L25 and L78 on the rim of the catalytic entry site protrude into the active site cleft, partially occluding access to it. Single (L25G or L78G) and double mutants provided evidence that widening the entrance to the active site greatly accelerates catalytic activity. By contrast, CafD has a typical Type-II "closed" conformation in which the zinc-bound water molecule is replaced by aspartic acid (D36). The most likely explanation for this result is that an arginine that is largely conserved within the ß-CA family is replaced by glycine (G38), so that D36 cannot undergo a conformational change by forming a D-R pair that creates the space for a zinc-bound water molecule and switches the enzyme to the active form. The CafD structure also reveals the presence of a "non-catalytic" zinc ion in the dimer interface, which may contribute to stabilizing the dimeric assembly.


Subject(s)
Aspergillus fumigatus/metabolism , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Catalysis , Catalytic Domain , Protein Binding , Zinc/metabolism
6.
Biochem Biophys Res Commun ; 503(4): 2899-2905, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30126635

ABSTRACT

Glial fibrillary acidic protein (GFAP) is a homopolymeric type III intermediate filament (IF) that plays essential roles in cell migration, mitosis, development, and signaling in astrocytes and a specific type of glial cells. Its overexpression and genetic mutations lead to abnormal IF networks and accumulation of Rosenthal fibers, which results in the fatal neurodegenerative disorder Alexander disease. Herein, we present the first crystal structure of human GFAP spanning the central coiled-coil 1B domain at 2.5 Šresolution. The domain forms a tetramer comprising two equivalent parallel coiled-coil dimers that pack together in an antiparallel manner. Its assembly is stabilized by extensive networks of intermolecular hydrogen bonds, salt bridges, and hydrophobic interactions. Furthermore, mapping of the GFAP mutations associated with Alexander disease reveals that most involve residues buried in the core of the interface, and are likely to disrupt the intermolecular interactions and/or introduce steric clashes, thereby decreasing GFAP solubility and promoting aggregation. Based on our structural analysis and previous biochemical studies, we propose that GFAP assembles in the A11 mode in which coiled-coil 1B dimers lie in close axial proximity in an antiparallel fashion to provide a stable tetrameric platform for the organization of the GFAP filament.


Subject(s)
Glial Fibrillary Acidic Protein/chemistry , Alexander Disease/genetics , Crystallography, X-Ray , Humans , Intermediate Filaments/chemistry , Protein Conformation , Protein Domains , Protein Multimerization , Protein Stability
7.
Biochem Biophys Res Commun ; 495(1): 1201-1207, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29180014

ABSTRACT

Prompt removal of misfolded membrane proteins and misassembled membrane protein complexes is essential for membrane homeostasis. However, the elimination of these toxic proteins from the hydrophobic membrane environment has high energetic barriers. The transmembrane protein, FtsH, is the only known ATP-dependent protease responsible for this task. The mechanisms by which FtsH recognizes, unfolds, translocates, and proteolyzes its substrates remain unclear. The structure and function of the ATPase and protease domains of FtsH have been previously characterized while the role of the FtsH periplasmic domain has not clearly identified. Here, we report the 1.5-1.95 Å resolution crystal structures of the Thermotoga maritima FtsH periplasmic domain (tmPD) and describe the dynamic features of tmPD oligomerization.


Subject(s)
ATP-Dependent Proteases/chemistry , ATP-Dependent Proteases/ultrastructure , Peptide Hydrolases/chemistry , Peptide Hydrolases/ultrastructure , Protein Multimerization , Thermotoga maritima/enzymology , Binding Sites , Computer Simulation , Enzyme Activation , Models, Chemical , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Structure-Activity Relationship
8.
Immunity ; 31(6): 873-84, 2009 Dec 18.
Article in English | MEDLINE | ID: mdl-19931471

ABSTRACT

Toll-like receptor 2 (TLR2) initiates potent immune responses by recognizing diacylated and triacylated lipopeptides. Its ligand specificity is controlled by whether it heterodimerizes with TLR1 or TLR6. We have determined the crystal structures of TLR2-TLR6-diacylated lipopeptide, TLR2-lipoteichoic acid, and TLR2-PE-DTPA complexes. PE-DTPA, 1,2-dimyristoyl-sn-glycero-3-phosphoethanolamine-N-diethylenetriaminepentaacetic acid, is a synthetic phospholipid derivative. Two major factors contribute to the ligand specificity of TLR2-TLR1 or TLR2-TLR6 heterodimers. First, the lipid channel of TLR6 is blocked by two phenylalanines. Simultaneous mutation of these phenylalanines made TLR2-TLR6 fully responsive not only to diacylated but also to triacylated lipopeptides. Second, the hydrophobic dimerization interface of TLR2-TLR6 is increased by 80%, which compensates for the lack of amide lipid interaction between the lipopeptide and TLR2-TLR6. The structures of the TLR2-lipoteichoic acid and the TLR2-PE-DTPA complexes demonstrate that a precise interaction pattern of the head group is essential for a robust immune response by TLR2 heterodimers.


Subject(s)
Lipopeptides/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 6/immunology , Acylation , Animals , Binding Sites , Crystallography, X-Ray , Hagfishes , Humans , Ligands , Lipopeptides/chemistry , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Mice , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/immunology , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Teichoic Acids/chemistry , Teichoic Acids/immunology , Teichoic Acids/metabolism , Toll-Like Receptor 1/chemistry , Toll-Like Receptor 1/immunology , Toll-Like Receptor 2/chemistry , Toll-Like Receptor 6/chemistry
9.
Nature ; 490(7421): 566-9, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-23000902

ABSTRACT

P-glycoprotein (P-gp) is an ATP-binding cassette transporter that confers multidrug resistance in cancer cells. It also affects the absorption, distribution and clearance of cancer-unrelated drugs and xenobiotics. For these reasons, the structure and function of P-gp have been studied extensively for decades. Here we present biochemical characterization of P-gp from Caenorhabditis elegans and its crystal structure at a resolution of 3.4 ångströms. We find that the apparent affinities of P-gp for anticancer drugs actinomycin D and paclitaxel are approximately 4,000 and 100 times higher, respectively, in the membrane bilayer than in detergent. This affinity enhancement highlights the importance of membrane partitioning when a drug accesses the transporter in the membrane. Furthermore, the transporter in the crystal structure opens its drug pathway at the level of the membrane's inner leaflet. In the helices flanking the opening to the membrane, we observe extended loops that may mediate drug binding, function as hinges to gate the pathway or both. We also find that the interface between the transmembrane and nucleotide-binding domains, which couples ATP hydrolysis to transport, contains a ball-and-socket joint and salt bridges similar to the ATP-binding cassette importers, suggesting that ATP-binding cassette exporters and importers may use similar mechanisms to achieve alternating access for transport. Finally, a model of human P-gp derived from the structure of C. elegans P-gp not only is compatible with decades of biochemical analysis, but also helps to explain perplexing functional data regarding the Phe335Ala mutant. These results increase our understanding of the structure and function of this important molecule.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Caenorhabditis elegans/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Crystallography, X-Ray , Dactinomycin/metabolism , Humans , Hydrolysis , Lipid Bilayers/metabolism , Models, Biological , Models, Molecular , Paclitaxel/metabolism , Protein Structure, Tertiary , Structural Homology, Protein , Structure-Activity Relationship
10.
Biochem Biophys Res Commun ; 486(2): 470-475, 2017 04 29.
Article in English | MEDLINE | ID: mdl-28315686

ABSTRACT

Anti-bacterial and anti-viral neuraminidase agents inhibit neuraminidase activity catalyzing the hydrolysis of terminal N-acetylneuraminic acid (Neu5Ac) from glycoconjugates and help to prevent the host pathogenesis that lead to fatal infectious diseases including influenza, bacteremia, sepsis, and cholera. Emerging antibiotic and drug resistances to commonly used anti-neuraminidase agents such as oseltamivir (Tamiflu) and zanamivir (Relenza) have highlighted the need to develop new anti-neuraminidase drugs. We obtained a serendipitous complex crystal of the catalytic domain of Clostridium perfringens neuraminidase (CpNanICD) with 2-(cyclohexylamino)ethanesulfonic acid (CHES) as a buffer. Here, we report the crystal structure of CpNanICD in complex with CHES at 1.24 Å resolution. Amphipathic CHES binds to the catalytic site of CpNanICD similar to the substrate (Neu5Ac) binding site. The 2-aminoethanesulfonic acid moiety and cyclohexyl groups of CHES interact with the cluster of three arginine residues and with the hydrophobic pocket of the CpNanICD catalytic site. In addition, a structural comparison with other bacterial and human neuraminidases suggests that CHES could serve as a scaffold for the development of new anti-neuraminidase agents targeting CpNanI.


Subject(s)
Bacterial Proteins/chemistry , Clostridium perfringens/chemistry , Enzyme Inhibitors/chemistry , Neuraminidase/chemistry , Taurine/analogs & derivatives , Amino Acid Motifs , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Cloning, Molecular , Clostridium perfringens/enzymology , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Models, Molecular , Neuraminidase/antagonists & inhibitors , Neuraminidase/genetics , Neuraminidase/metabolism , Protein Domains , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structural Homology, Protein , Taurine/chemistry
11.
Immunity ; 29(2): 182-91, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18701082

ABSTRACT

Toll-like receptors (TLRs) play central roles in the innate immune response by recognizing conserved structural patterns in diverse microbial molecules. Here, we discuss ligand binding and activation mechanisms of the TLR family. Hydrophobic ligands of TLR1, TLR2, and TLR4 interact with internal protein pockets. In contrast, dsRNA, a hydrophilic ligand, interacts with the solvent-exposed surface of TLR3. Binding of agonistic ligands, lipopeptides or dsRNA, induces dimerization of the ectodomains of the various TLRs, forming dimers that are strikingly similar in shape. In these "m"-shaped complexes, the C termini of the extracellular domains of the TLRs converge in the middle. This observation suggests the hypothesis that dimerization of the extracellular domains forces the intracellular TIR domains to dimerize, and this initiates signaling by recruiting intracellular adaptor proteins.


Subject(s)
Lymphocyte Antigen 96/metabolism , Protein Structure, Tertiary , Toll-Like Receptors/chemistry , Toll-Like Receptors/metabolism , Animals , Binding Sites , Dimerization , Hydrophobic and Hydrophilic Interactions , Ligands , Lipopolysaccharide Receptors/immunology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Lipopolysaccharides/metabolism , Lymphocyte Antigen 96/immunology , Receptors, Interleukin-1/metabolism
12.
Commun Biol ; 7(1): 672, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822018

ABSTRACT

ATP-binding cassette transporter B6 (ABCB6), a protein essential for heme biosynthesis in mitochondria, also functions as a heavy metal efflux pump. Here, we present cryo-electron microscopy structures of human ABCB6 bound to a cadmium Cd(II) ion in the presence of antioxidant thiol peptides glutathione (GSH) and phytochelatin 2 (PC2) at resolutions of 3.2 and 3.1 Å, respectively. The overall folding of the two structures resembles the inward-facing apo state but with less separation between the two halves of the transporter. Two GSH molecules are symmetrically bound to the Cd(II) ion in a bent conformation, with the central cysteine protruding towards the metal. The N-terminal glutamate and C-terminal glycine of GSH do not directly interact with Cd(II) but contribute to neutralizing positive charges of the binding cavity by forming hydrogen bonds and van der Waals interactions with nearby residues. In the presence of PC2, Cd(II) binding to ABCB6 is similar to that observed with GSH, except that two cysteine residues of each PC2 molecule participate in Cd(II) coordination to form a tetrathiolate. Structural comparison of human ABCB6 and its homologous Atm-type transporters indicate that their distinct substrate specificity might be attributed to variations in the capping residues situated at the top of the substrate-binding cavity.


Subject(s)
ATP-Binding Cassette Transporters , Humans , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/metabolism , ATP-Binding Cassette Transporters/ultrastructure , Binding Sites , Cadmium/metabolism , Cadmium/chemistry , Cryoelectron Microscopy , Glutathione/metabolism , Glutathione/chemistry , Models, Molecular , Phytochelatins/metabolism , Phytochelatins/chemistry , Protein Binding , Protein Conformation
13.
J Med Chem ; 67(16): 14443-14465, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39102524

ABSTRACT

The P2X3 receptor (P2X3R), an ATP-gated cation channel predominantly expressed in C- and Aδ-primary afferent neurons, has been proposed as a drug target for neurological inflammatory diseases, e.g., neuropathic pain, and chronic cough. Aiming to develop novel, selective P2X3R antagonists, tetrazolopyrimidine-based hit compound 9 was optimized through structure-activity relationship studies by modifying the tetrazole core as well as side chain substituents. The optimized antagonist 26a, featuring a cyclopropane-substituted triazolopyrimidine core, displayed potent P2X3R-antagonistic activity (IC50 = 54.9 nM), 20-fold selectivity versus the heteromeric P2X2/3R, and high selectivity versus other P2XR subtypes. Noncompetitive P2X3R blockade was experimentally confirmed by calcium influx assays. Cryo-electron microscopy revealed that 26a stabilizes the P2X3R in its desensitized state, acting as a molecular barrier to prevent ions from accessing the central pore. In vivo studies in a rat neuropathic pain model (spinal nerve ligation) showed dose-dependent antiallodynic effects of 26a, thus presenting a novel, promising lead structure.


Subject(s)
Cryoelectron Microscopy , Purinergic P2X Receptor Antagonists , Pyrimidines , Receptors, Purinergic P2X3 , Triazoles , Animals , Purinergic P2X Receptor Antagonists/pharmacology , Purinergic P2X Receptor Antagonists/chemistry , Purinergic P2X Receptor Antagonists/chemical synthesis , Structure-Activity Relationship , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Rats , Receptors, Purinergic P2X3/metabolism , Humans , Triazoles/pharmacology , Triazoles/chemistry , Triazoles/chemical synthesis , Allosteric Site , Male , Neuralgia/drug therapy , Drug Discovery , Rats, Sprague-Dawley
14.
Life Sci Alliance ; 7(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39089879

ABSTRACT

The large-conductance calcium-activated potassium (BKCa) channel, which is crucial for urinary bladder smooth muscle relaxation, is a potential target for overactive bladder treatment. Our prior work unveiled CTIBD as a promising BKCa channel activator, altering V 1/2 and G max This study investigates CTIBD's activation mechanism, revealing its independence from the Ca2+ and membrane voltage sensing of the BKCa channel. Cryo-electron microscopy disclosed that two CTIBD molecules bind to hydrophobic regions on the extracellular side of the lipid bilayer. Key residues (W22, W203, and F266) are important for CTIBD binding, and their replacement with alanine reduces CTIBD-mediated channel activation. The triple-mutant (W22A/W203A/F266A) channel showed the smallest V 1/2 shift with a minimal impact on activation and deactivation kinetics by CTIBD. At the single-channel level, CTIBD treatment was much less effective at increasing P o in the triple mutant, mainly because of a drastically increased dissociation rate compared with the WT. These findings highlight CTIBD's mechanism, offering crucial insights for developing small-molecule treatments for BKCa-related pathophysiological conditions.


Subject(s)
Chloride Channel Agonists , Cryoelectron Microscopy , Large-Conductance Calcium-Activated Potassium Channels , Animals , Humans , Binding Sites , Calcium/metabolism , HEK293 Cells , Ion Channel Gating , Kinetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/agonists , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Large-Conductance Calcium-Activated Potassium Channels/agonists , Large-Conductance Calcium-Activated Potassium Channels/chemistry , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Lipid Bilayers/metabolism , Mutation , Protein Binding , Chloride Channel Agonists/chemistry , Chloride Channel Agonists/pharmacology
15.
iScience ; 26(12): 108386, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38025788

ABSTRACT

The endoplasmic reticulum (ER) consists of the nuclear envelope and a connected peripheral network of tubules and interspersed sheets. The structure of ER tubules is generated and maintained by various proteins, including reticulons, DP1/Yop1p, atlastins, and lunapark. Reticulons and DP1/Yop1p stabilize the high membrane curvature of ER tubules, and atlastins mediate homotypic membrane fusion between ER tubules; however, the exact role of lunapark remains poorly characterized. Here, using isolated yeast ER microsomes and reconstituted proteoliposomes, we directly examined the function of the yeast lunapark Lnp1p for yeast atlastin Sey1p-mediated ER fusion and found that Lnp1p inhibits Sey1p-driven membrane fusion. Furthermore, by using a newly developed assay for monitoring trans-Sey1p complex assembly, a prerequisite for ER fusion, we found that assembly of trans-Sey1p complexes was increased by the deletion of LNP1 and decreased by the overexpression of Lnp1p, indicating that Lnp1p inhibits Sey1p-mediated fusion by interfering with assembly of trans-Sey1p complexes.

16.
Commun Biol ; 6(1): 960, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37735522

ABSTRACT

Human ATP-binding cassette transporter subfamily B6 (ABCB6) is a mitochondrial ATP-driven pump that translocates porphyrins from the cytoplasm into mitochondria for heme biosynthesis. Within the transport pathway, a conserved aromatic residue W546 located in each monomer plays a pivotal role in stabilizing the occluded conformation via π-stacking interactions. Herein, we employed cryo-electron microscopy to investigate the structural consequences of a single W546A mutation in ABCB6, both in detergent micelles and nanodiscs. The results demonstrate that the W546A mutation alters the conformational dynamics of detergent-purified ABCB6, leading to entrapment of the transporter in an outward-facing transient state. However, in the nanodisc system, we observed a direct interaction between the transporter and a phospholipid molecule that compensates for the absence of the W546 residue, thereby facilitating the normal conformational transition of the transporter toward the occluded state following ATP hydrolysis. The findings also reveal that adoption of the outward-facing conformation causes charge repulsion between ABCB6 and the bound substrate, and rearrangement of key interacting residues at the substrate-binding site. Consequently, the affinity for the substrate is significantly reduced, facilitating its release from the transporter.


Subject(s)
Detergents , Porphyrins , Humans , Cryoelectron Microscopy , ATP-Binding Cassette Transporters/genetics , Membrane Transport Proteins , Adenosine Triphosphate
17.
Eur J Med Chem ; 260: 115720, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37633203

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease (Mpro) has been targeted for the development of anti-SARS-CoV-2 agents against COVID-19 infection because Mpro processes essential viral polyproteins and plays a key role in SARS-CoV-2 replication. In this study, we report the development of novel SARS-CoV-2 Mpro inhibitors derived from carmofur, a previously identified compound that has shown moderate potency as a covalent inhibitor of SARS-CoV-2 Mpro. To employ a structure-guided drug design strategy, a putative intact binding mode of carmofur at catalytic active site of Mpro was initially predicted by docking simulation. Based on the predicted binding mode, a series of carmofur derivatives aiming to occupy the Mpro substrate binding regions were investigated for structure-activity relationship analysis. As a result, an indole-based derivative, speculated to interact with the S4 binding pocket, 21b (IC50 = 1.5 ± 0.1 µM) was discovered. Its structure was further modified and evaluated in silico by combining docking simulation, free energy perturbation calculation and subpocket interaction analysis to optimize the interactions at the S2 and S4 binding pockets. Among the newly designed novel derivatives, 21h and 21i showed the best inhibitory potencies against Mpro with IC50 values of 0.35 and 0.37 µM, respectively. Moreover, their antiviral activities were confirmed with EC50 values of 20-30 µM in the SARS-CoV-2-infected cell-based assay, suggesting that these novel Mpro inhibitors could be applied as potential lead compounds for the development of substantial anti-SARS-CoV-2 agents.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Antiviral Agents/pharmacology , Fluorouracil
18.
Pharmaceuticals (Basel) ; 15(4)2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35455436

ABSTRACT

N6A-methyladenosine (m6A) post-transcriptional modification, the most abundant internal RNA modification, is catalyzed by the METTL3-14 methyltransferase complex. Recently, attention has been drawn to the METTL3-14 complex regarding its significant roles in the pathogenesis of acute myeloid leukemia (AML), attracting the potential of novel therapeutic targets for the disease. Herein, we report the identification and characterization of eltrombopag as a selective allosteric inhibitor of the METTL3-14 complex. Eltrombopag exhibited selective inhibitory activity in the most active catalytic form of the METTL3-14 complex by direct binding, and the mechanism of inhibition was confirmed as a noncompetitive inhibition by interacting at a putative allosteric binding site in METTL3, which was predicted by cavity search and molecular docking studies. At a cellular level, eltrombopag displayed anti-proliferative effects in the relevant AML cell line, MOLM-13, in correlation with a reduction in m6A levels. Molecular mechanism studies of eltrombopag using m6A-seq analysis provided further evidence of its cellular function by determining the hypomethylation of leukemogenic genes in eltrombopag-treated MOLM-13 cells and the overlapping of the pattern with those of METTL3-knockdown MOLM-13 cells. In conclusion, eltrombopag was first disclosed as a functional METTL3-14 allosteric inhibitor in AML cells, which could be utilized for the further development of novel anti-AML therapy.

19.
Nat Commun ; 13(1): 5851, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36195619

ABSTRACT

TAPL is a lysosomal ATP-binding cassette transporter that translocates a broad spectrum of polypeptides from the cytoplasm into the lysosomal lumen. Here we report that, in addition to its well-known role as a peptide translocator, TAPL exhibits an ATP-dependent phosphatidylserine floppase activity that is the possible cause of its high basal ATPase activity and of the lack of coupling between ATP hydrolysis and peptide efflux. We also present the cryo-EM structures of mouse TAPL complexed with (i) phospholipid, (ii) cholesteryl hemisuccinate (CHS) and 9-mer peptide, and (iii) ADP·BeF3. The inward-facing structure reveals that F449 protrudes into the cylindrical transport pathway and divides it into a large hydrophilic central cavity and a sizable hydrophobic upper cavity. In the structure, the peptide binds to TAPL in horizontally-stretched fashion within the central cavity, while lipid molecules plug vertically into the upper cavity. Together, our results suggest that TAPL uses different mechanisms to function as a peptide translocase and a phosphatidylserine floppase.


Subject(s)
Peptides , Phosphatidylserines , ATP-Binding Cassette Transporters/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Animals , Lysosomes/metabolism , Mice , Peptides/chemistry , Phosphatidylserines/metabolism
20.
Eur J Hum Genet ; 30(6): 687-694, 2022 06.
Article in English | MEDLINE | ID: mdl-35246666

ABSTRACT

Alexander disease (AxD) is a neurodegenerative astrogliopathy caused by mutation in the glial fibrillary acidic protein (GFAP) gene. A 42-year-old Korean man presented with temporary gait disturbance and psychiatric regression after a minor head trauma in the absence of bulbar symptoms and signs. Magnetic resonance images of the brain and spinal cord showed significant atrophy of the medulla oblongata and the entire spinal cord as well as contrast-enhanced T2 hypointensity in the basal ganglia. DNA sequencing revealed a novel 33-bp in-frame deletion mutation (p.Glu138_Leu148del) within the 1B rod domain of GFAP, which was predicted to be deleterious by PROVEAN analysis. To test whether the deletion mutant is disease-causing, we performed in vitro GFAP assembly and sedimentation assays, and GFAP aggregation assays in human adrenal carcinoma SW13 (Vim-) cells and rat primary astrocytes. All the assays revealed that GFAP p.Glu138_Leu148del is aggregation prone. Based on these findings, we diagnosed the patient with Type II AxD. This is a report that demonstrates the pathogenicity of InDel mutation of GFAP through functional studies. This patient's atypical presentation as well as the discrepancy between clinical symptoms and radiologic findings may extend the scope of AxD.


Subject(s)
Alexander Disease , Alexander Disease/diagnosis , Alexander Disease/genetics , Alexander Disease/pathology , Animals , Brain/metabolism , Glial Fibrillary Acidic Protein/genetics , Humans , Mutation , Phenotype , Rats
SELECTION OF CITATIONS
SEARCH DETAIL